1
|
Nishimae F, Sakurai F, Ono R, Onishi R, Takayama K, Mizuguchi H. A dopamine antagonist, domperidone enhances the replication of an oncolytic adenovirus in human tumour cells. J Gen Virol 2022; 103. [PMID: 35731650 DOI: 10.1099/jgv.0.001752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oncolytic adenoviruses (OAds) have attracted much attention as novel anticancer agents. Numerous studies have examined the antitumour effects of combinational use of an OAd and anticancer agents; however, few chemical compounds enhancing OAd infection have been reported. In this study, we screened a food and drug administration (FDA)-approved drug library containing 1134 small chemical compounds to identify chemical compounds that enhance OAd replication in human tumour cells. We found that domperidone, a dopamine D2 receptor antagonist, significantly enhanced the replication of an OAd in human tumour cells, including human pancreatic tumour cells, by two-fivefold, resulting in improvement of OAd-mediated tumour cell killing activities. The E1A mRNA levels were significantly increased in domperidone-pre-treated cells following OAd infection, which contributed to the promotion of OAd replication. However, mRNA levels of the dopamine D2 receptor (DRD2), which is known to be a target molecule of domperidone, were undetectable in most of the tumour cells by real-time reverse transcription (RT)-PCR analysis, indicating that domperidone promoted OAd replication by acting on a molecule other than DRD2. This study provides important clues for the improvement of OAd-mediated cancer therapy.
Collapse
Affiliation(s)
- Fumitaka Nishimae
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Ryosuke Ono
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Rika Onishi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kosuke Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| |
Collapse
|
2
|
Trivedi LU, Femnou Mbuntum L, Halm EA, Mansi I. Is Statin Use Associated With Risk of Thyroid Diseases? Results of a Retrospective Cohort Study. Ann Pharmacother 2021; 55:1110-1119. [PMID: 33412925 DOI: 10.1177/1060028020986552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Given the ubiquity of statin use and prevalence of thyroid diseases, such as thyroid cancer, hyperthyroidism, and thyroiditis, understanding their association deserves further attention. OBJECTIVE To examine the association between statin use and thyroid cancer, thyrotoxicosis, goiter, and thyroiditis. METHODS Using Tricare data, 2 propensity score (PS)-matched cohorts of statin users and nonusers were formed: (1) a PS-matched general cohort (all patients aged 30-85 years) and (2) a PS-matched healthy cohort (excluded patients with cardiovascular diseases or severe comorbidities). Outcomes were thyroid cancer, thyrotoxicosis, goiter, and thyroiditis. Odds ratios (ORs) and 95% CIs of outcomes were estimated using conditional regression analysis. RESULTS Of 43 438 patients, the PS-matched general cohort matched 6342 statin users to 6342 nonusers. The OR of thyroid cancer was 0.62 (95% CI = 0.39-0.996). There was no significant difference between statin users and nonusers in risk of thyrotoxicosis (OR = 0.88; 95% CI = 0.71-1.09), goiter (OR = 0.9; 95% CI = 0.77-1.03), or thyroiditis (OR = 0.78; 95% CI = 0.53-1.15). In the PS-matched healthy cohort (3351 statin users to 3351 nonusers), there was no difference between statin users and nonusers in any outcome. Limitations of the study include its retrospective observational design and use of administrative codes in outcomes ascertainment. CONCLUSION AND RELEVANCE This study did not demonstrate any association of statins with harmful effects on thyroid diseases, which offers assurance to clinicians and patients. Furthermore, statin use appears to be associated with a decreased risk of thyroid cancer, but more studies are needed.
Collapse
Affiliation(s)
| | | | - Ethan A Halm
- University of Texas Southwestern, Dallas, TX, USA
| | - Ishak Mansi
- University of Texas Southwestern, Dallas, TX, USA.,VA North Texas Health System, Dallas, TX, USA
| |
Collapse
|
3
|
Malfitano AM, Di Somma S, Iannuzzi CA, Pentimalli F, Portella G. Virotherapy: From single agents to combinatorial treatments. Biochem Pharmacol 2020; 177:113986. [PMID: 32330494 DOI: 10.1016/j.bcp.2020.113986] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
Virotherpay is emerging as a promising strategy against cancer, and three oncolytic viruses (OVs) have gained approval in different countries for the treatment of several cancer types. Beyond the capability to selectively infect, replicate and lyse cancer cells, OVs act through a multitude of events, including modification of the tumour micro/macro-environment as well as a complex modulation of the anti-tumour immune response by activation of danger signals and immunogenic cell death pathways. Most OVs show limited effects, depending on the viral platform and the interactions with the host. OVs used as monotherapy only in a minority of patients elicited a full response. Better outcomes were obtained using OVs in combination with other treatments, such as immune therapy or chemotherapy, suggesting that the full potential of OVs can be unleashed in combination with other treatment modalities. Here, we report the main described combination of OVs with conventional chemotherapeutic agents: platinum salts, mitotic inhibitors, anthracyclines and other antibiotics, anti-metabolites, alkylating agents and topoisomerase inhibitors. Additionally, our work provides an overview of OV combination with targeted therapies: histone deacetylase inhibitors, kinase inhibitors, monoclonal antibodies, inhibitors of DNA repair, inhibitors of the proteasome complex and statins that demonstrated enhanced OV anti-neoplastic activity. Although further studies are required to assess the best combinations to translate the results in the clinic, it is clear that combined therapies, acting with complementary mechanisms of action might be useful to target cancer lesions resistant to currently available treatments.
Collapse
Affiliation(s)
- Anna Maria Malfitano
- Dipartimento di Scienze Mediche Traslazionali, Università Federico II Napoli, Italy
| | - Sarah Di Somma
- Dipartimento di Scienze Mediche Traslazionali, Università Federico II Napoli, Italy
| | | | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Giuseppe Portella
- Dipartimento di Scienze Mediche Traslazionali, Università Federico II Napoli, Italy.
| |
Collapse
|
4
|
Cisplatin Relocalizes RNA Binding Protein HuR and Enhances the Oncolytic Activity of E4orf6 Deleted Adenovirus. Cancers (Basel) 2020; 12:cancers12040809. [PMID: 32230919 PMCID: PMC7226092 DOI: 10.3390/cancers12040809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/14/2020] [Accepted: 03/24/2020] [Indexed: 11/24/2022] Open
Abstract
The combination of adenoviruses and chemotherapy agents is a novel approach for human cancer therapeutics. A meticulous analysis between adenovirus and chemotherapeutic agents can help to design an effective anticancer therapy. Human antigen R (HuR) is an RNA binding protein that binds to the AU-rich element (ARE) of specific mRNA and is involved in the export and stabilization of ARE-mRNA. Our recent report unveiled that the E4orf6 gene deleted oncolytic adenovirus (dl355) replicated for certain types of cancers where ARE-mRNA is stabilized. This study aimed to investigate whether a combined treatment of dl355 and Cis-diamminedichloroplatinum (CDDP) can have a synergistic cell-killing effect on cancer cells. We confirmed the effect of CDDP in nucleocytoplasmic HuR shuttling. In vitro and in vivo experiments showed the enhancement of cancer cell death by apoptosis induction and a significant reduction in tumor growth following combination treatment. These results suggested that combination therapy exerted a synergistic antitumor activity by upregulation of CDDP induced cytoplasmic HuR, which led to ARE mRNA stabilization and increased virus proliferation. Besides, the enhanced cell-killing effect was due to the activation of the intrinsic apoptotic pathway. Therefore, the combined treatment of CDDP and dl355 could represent a rational approach for cancer therapy.
Collapse
|
5
|
Virotherapy as a Potential Therapeutic Approach for the Treatment of Aggressive Thyroid Cancer. Cancers (Basel) 2019; 11:cancers11101532. [PMID: 31636245 PMCID: PMC6826611 DOI: 10.3390/cancers11101532] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Virotherapy is a novel cancer treatment based on oncolytic viruses (OVs), which selectively infect and lyse cancer cells, without harming normal cells or tissues. Several viruses, either naturally occurring or developed through genetic engineering, are currently under investigation in clinical studies. Emerging reports suggesting the immune-stimulatory property of OVs against tumor cells further support the clinical use of OVs for the treatment of lesions lacking effective therapies. Poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC), have a poor prognosis and limited treatment options. Therefore, several groups investigated the therapeutic potential of OVs in PDTC/ATC models producing experimental data sustaining the potential clinical efficacy of OVs in these cancer models. Moreover, the presence of an immunosuppressive microenvironment further supports the potential use of OVs in ATC. In this review, we present the results of the studies evaluating the efficacy of OVs alone or in combination with other treatment options. In particular, their potential therapeutic combination with multiple kinases inhibitors (MKIs) or immune checkpoint inhibitors are discussed.
Collapse
|
6
|
Di Somma S, Iannuzzi CA, Passaro C, Forte IM, Iannone R, Gigantino V, Indovina P, Botti G, Giordano A, Formisano P, Portella G, Malfitano AM, Pentimalli F. The Oncolytic Virus dl922-947 Triggers Immunogenic Cell Death in Mesothelioma and Reduces Xenograft Growth. Front Oncol 2019; 9:564. [PMID: 31355131 PMCID: PMC6639422 DOI: 10.3389/fonc.2019.00564] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/10/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Malignant pleural mesothelioma (MPM) is an aggressive cancer associated with asbestos exposure that urgently requires effective therapeutic strategies. Current treatments are unable to increase significantly patient survival, which is often limited to <1 year from diagnosis. Virotherapy, based on the use of oncolytic viruses that exert anti-cancer effects by direct cell lysis and through the induction of anti-tumor immune response, represents an alternative therapeutic option for rare tumors with limited life expectancy. In this study, we propose the use of the adenovirus dl922-947, engineered to allow selective replication in cancer cells, to counteract MPM. Methods: We performed a thorough preclinical assessment of dl922-947 effects in a set of MPM cell lines and xenografts. Cytotoxicity of dl922-947 alone and in combination assays was evaluated by sulforhodamine B assay. Cell cycle, calreticulin expression, and high mobility group box protein 1 (HMGB1) secretion were determined by flow cytometry, whereas ATP content was determined by a luminescence-based bioassay. The modulation of angiogenic factors in MPM-infected cells was evaluated through ELISA. Results: We found that dl922-947 infection exhibits cytotoxic effects in MPM cell lines, affecting cell viability, cell cycle progression, and regulating main hallmarks of immunogenic cell death inducing calreticulin surface exposure, HMGB1 and ATP release. Our results also suggest that dl922-947 may affect angiogenic signals by regulation of VEGF-A and IL-8 secretion. Furthermore, dl922-947 shows anti-tumor efficacy in murine xenograft models reducing tumor growth and enhancing survival. Finally, the combination with cisplatin potentiated the cytotoxic effect of dl922-947. Conclusions: Overall our data identify virotherapy, based on the use of dl922-947, as a new possible therapeutic strategy against MPM, which could be used alone, in combination with standard chemotherapy drugs, as shown here, or other approaches also aimed at enhancing the antitumoral immune response elicited by the virus.
Collapse
Affiliation(s)
- Sarah Di Somma
- Dipartimento Scienze Mediche Traslazionali, Università di Napoli "Federico II", Naples, Italy
| | | | - Carmela Passaro
- Dipartimento Scienze Mediche Traslazionali, Università di Napoli "Federico II", Naples, Italy
| | - Iris Maria Forte
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Raffaella Iannone
- Dipartimento Scienze Mediche Traslazionali, Università di Napoli "Federico II", Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Paola Indovina
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Antonio Giordano
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA, United States.,Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Pietro Formisano
- Dipartimento Scienze Mediche Traslazionali, Università di Napoli "Federico II", Naples, Italy
| | - Giuseppe Portella
- Dipartimento Scienze Mediche Traslazionali, Università di Napoli "Federico II", Naples, Italy
| | - Anna Maria Malfitano
- Dipartimento Scienze Mediche Traslazionali, Università di Napoli "Federico II", Naples, Italy
| | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
7
|
Miyagawa Y, Araki K, Yamashita T, Tanaka S, Tanaka Y, Tomifuji M, Ueda Y, Yonemitsu Y, Shimada H, Shiotani A. Induction of cell fusion/apoptosis in anaplastic thyroid carcinoma in orthotopic mouse model by urokinase‐specific oncolytic Sendai virus. Head Neck 2019; 41:2873-2882. [DOI: 10.1002/hed.25769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yoshihiro Miyagawa
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Koji Araki
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Taku Yamashita
- Department of Otolaryngology ‐ Head and Neck SurgeryKitasato University School of Medicine Sagamihara Japan
| | - Shingo Tanaka
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Yuya Tanaka
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Masayuki Tomifuji
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| | - Yasuji Ueda
- Section of Gene Medicine, R&D CenterID Pharma Co., Ltd. Tokyo Japan
| | - Yoshikazu Yonemitsu
- R&D Laboratory for Innovative Biotherapeutics Science, Graduate School of Pharmaceutical SciencesKyushu University Fukuoka Japan
| | - Hideaki Shimada
- Department of SurgeryToho University School of Medicine Tokyo Japan
| | - Akihiro Shiotani
- Department of Otolaryngology ‐ Head and Neck SurgeryNational Defense Medical College Saitama Japan
| |
Collapse
|
8
|
Passaro C, Somma SD, Malfitano AM, Portella G. Oncolytic virotherapy for anaplastic and poorly differentiated thyroid cancer: a promise or a clinical reality? INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2018. [DOI: 10.2217/ije-2017-0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses (OVs) selectively infect and lyse cancer cells. A direct lytic effect of OVs has been theorized in the initial studies; however, the antineoplastic effect of OVs is also due to the induction of an immune response against cancer cells. Anaplastic thyroid cancer is one of the most aggressive human malignancies with a short survival time of about 6–12 months from the diagnosis. The lack of effective therapies has prompted to investigate the efficacy of OVs in anaplastic thyroid carcinoma. Different OVs have been tested in preclinical studies, either as single agents or in combinatorial treatments. In this review, the results of these studies are summarized and future perspective discussed.
Collapse
Affiliation(s)
- Carmela Passaro
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Di Somma
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Anna Maria Malfitano
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Giuseppe Portella
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
| |
Collapse
|
9
|
Jiang K, Song C, Kong L, Hu L, Lin G, Ye T, Yao G, Wang Y, Chen H, Cheng W, Barr MP, Liu Q, Zhang G, Ding C, Meng S. Recombinant oncolytic Newcastle disease virus displays antitumor activities in anaplastic thyroid cancer cells. BMC Cancer 2018; 18:746. [PMID: 30021550 PMCID: PMC6052588 DOI: 10.1186/s12885-018-4522-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/18/2018] [Indexed: 12/31/2022] Open
Abstract
Background Anaplastic thyroid cancer (ATC) is one of the most aggressive of all solid tumors for which no effective therapies are currently available. Oncolytic Newcastle disease virus (NDV) has shown the potential to induce oncolytic cell death in a variety of cancer cells of diverse origins. However, whether oncolytic NDV displays antitumor effects in ATC remains to be investigated. We have previously shown that the oncolytic NDV strain FMW (NDV/FMW) induces oncolytic cell death in several cancer types. In the present study, we investigated the oncolytic effects of NDV/FMW in ATC. Methods In this study, a recombinant NDV expressing green fluorescent protein (GFP) was generated using an NDV reverse genetics system. The resulting virus was named after rFMW/GFP and the GFP expression in infected cells was demonstrated by direct fluorescence and immunoblotting. Viral replication was evaluated by end-point dilution assay in DF-1 cell lines. Oncolytic effects were examined by biochemical and morphological experiments in cultural ATC cells and in mouse models. Results rFMW/GFP replicated robustly in ATC cells as did its parent virus (NDV/FMW) while the expression of GFP protein was detected in lungs and spleen of mice intravenously injected with rFMW/GFP. We further showed that rFMW/GFP infection substantially increased early and late apoptosis in the ATC cell lines, THJ-16 T and THJ-29 T and increased caspase-3 processing and Poly (ADP-ribose) polymerase (PARP) cleavage in ATC cells as assessed by immunoblotting. In addition, rFMW/GFP induced lyses of spheroids derived from ATC cells in three-dimensional (3D) cultures. We further demonstrated that rFMW/GFP infection resulted in the activation of p38 MAPK signaling, but not Erk1/2 or JNK, in THJ-16 T and THJ-29 T cells. Notably, inhibition of p38 MAPK activity by SB203580 decreased rFMW/GFP-induced cleavage of caspase-3 and PARP in THJ-16 T and THJ-29 T cells. Finally, both rFMW/GFP and its parent virus inhibited tumor growth in mice bearing THJ-16 T derived tumors. Conclusion Taken together, these data indicate that both the recombinant reporter virus rFMW/GFP and its parent virus NDV/FMW, display oncolytic activities in ATC cells in vitro and in vivo and suggest that oncolytic NDV may have potential as a novel therapeutic strategy for ATC. Electronic supplementary material The online version of this article (10.1186/s12885-018-4522-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ke Jiang
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue Road, Shanghai, 200241, China
| | - Lingkai Kong
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Lulu Hu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Guibin Lin
- Laboratory Center, The Third People's Hospital of Huizhou, Affiliated Hospital Guangzhou Medical University, Huizhou, 516002, China
| | - Tian Ye
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Gang Yao
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Yupeng Wang
- Department of Dermatology of First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116021, China
| | - Haibo Chen
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Wei Cheng
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Martin P Barr
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences St. James's Hospital and Trinity College Dublin, Dublin, Ireland
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China
| | - Guirong Zhang
- Central laboratory, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, 44 Xiaoheyan Road, Shenyang, 110042, China.
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue Road, Shanghai, 200241, China.
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Room 415, 9 Lvshun Road South, Dalian, 116044, China.
| |
Collapse
|
10
|
Li L, You LS, Mao LP, Jin SH, Chen XH, Qian WB. Combing oncolytic adenovirus expressing Beclin-1 with chemotherapy agent doxorubicin synergistically enhances cytotoxicity in human CML cells in vitro. Acta Pharmacol Sin 2018; 39:251-260. [PMID: 28905936 DOI: 10.1038/aps.2017.100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/09/2017] [Indexed: 02/07/2023]
Abstract
Cancer virotherapy provides a new strategy to treat cancer that can directly kill cancer cells by oncolysis. Insertion of therapeutic genes into the genome of a modified adenovirus, thereby creating a so-called gene-virotherapy that shares the advantages of gene therapy and virotherapy. In this study we investigated whether a strategy that combines the oncolytic effects of an adenoviral vector with the simultaneous expression of the autophagy gene Beclin-1 offered a therapeutic advantage for chronic myeloid leukemia (CML) cells with resistance to chemotherapy and evaluated the synergistic effects of SG511-BECN and doxorubicin (Dox) in human CML cells in vitro. Oncolytic virus SG511-BECN was constructed through introducing the Beclin-1 gene into the oncolytic adenoviral backbone. SG511-BECN displayed significantly improved antileukemia activity on multidrug-resistant CML cell line K562/A02, which was mediated via induction of autophagic cell death. Furthermore, Dox could synergize with SG511-BECN to kill the CML cells by improving the infectious efficiency of the oncolytic adenovirus without causing significant damage to normal human mononuclear cells. The results demonstrate that targeting the autophagic cell death pathway and combination of a chemotherapy agent with oncolytic adenovirus may be a novel strategy for the treatment of leukemia with chemotherapy resistance.
Collapse
|
11
|
Synergistic Anti-tumour Effects of Quercetin and Oncolytic Adenovirus expressing TRAIL in Human Hepatocellular Carcinoma. Sci Rep 2018; 8:2182. [PMID: 29391509 PMCID: PMC5794998 DOI: 10.1038/s41598-018-20213-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/16/2018] [Indexed: 01/13/2023] Open
Abstract
The combination of oncolytic adenoviruses and specific chemotherapy agents is fast emerging as a novel therapeutic approach for resistan the patocellular carcinoma (HCC) cells. A detailed analysis of the network between adenovirus and chemotherapeutic agents can help design an effective strategy to combat HCC. We sought to investigate whether a combined treatment of ZD55-TRAIL and quercetin can have an enhanced cell-killing effect on HCC cells. In-vitro experiments showed that quercetin can enhance ZD55-TRAIL mediated growth inhibition and apoptosis in HCC cells. In addition, we showed that quercetin reduced ZD55-TRAIL mediated NF-κB activation and down-regulated its downstream targets, which in turn promoted the pro-apoptotic action of ZD55-TRAIL. Furthermore, in-vivo experiments in mice injected with HuH-7 cells resulted in significantly greater reduction in tumour growth and volume following combined ZD55-TRAIL and quercetin treatment. In conclusion, we demonstrated that quercetin could sensitize human HCC cells to apoptosis via ZD55-TRAIL in-vitro and in-vivo and presented ZD55-TRAIL and quercetin combination as a suitable anti-HCC therapy.
Collapse
|
12
|
Ge Y, Lei W, Ma Y, Wang Y, Wei B, Chen X, Ru G, He X, Mou X, Wang S. Synergistic antitumor effects of CDK inhibitor SNS‑032 and an oncolytic adenovirus co‑expressing TRAIL and Smac in pancreatic cancer. Mol Med Rep 2017; 15:3521-3528. [PMID: 28440486 PMCID: PMC5436152 DOI: 10.3892/mmr.2017.6472] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/07/2017] [Indexed: 12/19/2022] Open
Abstract
Gene therapy using oncolytic adenoviruses is a novel approach for human cancer therapeutics. The current study aimed to investigate whether the combined use of an adenovirus expressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and second mitochondria-derived activator of caspase (Smac) upon caspase activation (ZD55-TRAIL-IETD-Smac) and cyclin-dependent kinase (CDK) inhibitor SNS-032 will synergistically reinforce their anti-pancreatic cancer activities. The experiments in vitro demonstrated that SNS-032 enhances ZD55-TRAIL-IETD-Smac-induced apoptosis and causes marked pancreatic cancer cell death. Western blot assays suggested that the SNS-032 intensified ZD55-TRAIL-IETD-Smac-induced apoptosis of pancreatic cancer cells by affecting anti-apoptotic signaling elements, including CDK-2, CDK-9, Mcl-1 and XIAP. Additionally, animal experiments further confirmed that the combination of SNS-032 and ZD55-TRAIL-IETD-Smac significantly inhibited the growth of BxPC-3 pancreatic tumor xenografts. In conclusion, the present study demonstrated that SNS-032 sensitizes human pancreatic cancer cells to ZD55-TRAIL-IETD-Smac-induced cell death in vitro and in vivo. These findings indicate that combined treatment with SNS-032 and ZD55-TRAIL-IETD-Smac could represent a rational approach for anti-pancreatic cancer therapy.
Collapse
Affiliation(s)
- Yun Ge
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Wen Lei
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Buyun Wei
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Xiaoyi Chen
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Guoqing Ru
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xianglei He
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
13
|
Wang S, Shu J, Chen L, Chen X, Zhao J, Li S, Mou X, Tong X. Synergistic suppression effect on tumor growth of ovarian cancer by combining cisplatin with a manganese superoxide dismutase-armed oncolytic adenovirus. Onco Targets Ther 2016; 9:6381-6388. [PMID: 27799786 PMCID: PMC5074737 DOI: 10.2147/ott.s113014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Gene therapy on the basis of oncolytic adenovirus is a novel approach for human cancer therapeutics. We aim to investigate whether it will synergistically reinforce their antiovarian cancer activities when the combined use of ZD55-manganese superoxide dismutase (MnSOD) and cisplatin was performed. The experiments in vitro showed that ZD55-MnSOD enhances cisplatin-induced apoptosis and causes remarkable ovarian cancer cell death. Apoptosis induction by treatment with ZD55-MnSOD and/or cisplatin was detected in SKOV-3 by apoptotic cell staining, flow cytometry, and western blot analysis. In addition, the cytotoxicity caused by ZD55-MnSOD to normal cells was examined by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay and western blot analysis. Animal experiment further confirmed that combination of ZD55-MnSOD and cisplatin achieved significant inhibition of SKOV-3 ovarian tumor xenografted growth. In summary, we have demonstrated that ZD55-MnSOD can sensitize human ovarian cancer cells to cisplatin-induced cell death and apoptosis in vitro and in vivo. These findings indicate that the combined treatment with ZD55-MnSOD and cisplatin could represent a rational approach for antiovarian cancer therapy.
Collapse
Affiliation(s)
- Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| | - Jing Shu
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital
| | - Li Chen
- Department of Obstetrics and Gynecology, Hangzhou Red Cross Hospital, Hangzhou, People's Republic of China
| | - Xiaopan Chen
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital
| | - Jianhong Zhao
- Department of Obstetrics and Gynecology, Hangzhou Red Cross Hospital, Hangzhou, People's Republic of China
| | - Shuangshuang Li
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| | - Xiangmin Tong
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| |
Collapse
|
14
|
Passaro C, Borriello F, Vastolo V, Di Somma S, Scamardella E, Gigantino V, Franco R, Marone G, Portella G. The oncolytic virus dl922-947 reduces IL-8/CXCL8 and MCP-1/CCL2 expression and impairs angiogenesis and macrophage infiltration in anaplastic thyroid carcinoma. Oncotarget 2016; 7:1500-15. [PMID: 26625205 PMCID: PMC4811476 DOI: 10.18632/oncotarget.6430] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/15/2015] [Indexed: 01/11/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is one of the most aggressive human solid tumor and current treatments are ineffective in increasing patients' survival. Thus, the development of new therapeutic approaches for ATC is needed. We have previously shown that the oncolytic adenovirus dl922-947 induces ATC cell death in vitro and tumor regression in vivo. However, the impact of dl922-947 on the pro-tumorigenic ATC microenvironment is still unknown. Since viruses are able to regulate cytokine and chemokine production from infected cells, we sought to investigate whether dl922-947 virotherapy has such effect on ATC cells, thereby modulating ATC microenvironment. dl922-947 decreased IL-8/CXCL8 and MCP-1/CCL2 production by the ATC cell lines 8505-c and BHT101-5. These results correlated with dl922-947-mediated reduction of NF-κB p65 binding to IL8 promoter in 8505-c and BHT101-5 cells and CCL2 promoter in 8505-c cells. IL-8 stimulates cancer cell proliferation, survival and invasion, and also angiogenesis. dl922-947-mediated reduction of IL-8 impaired ATC cell motility in vitro and ATC-induced angiogenesis in vitro and in vivo. We also show that dl922-947-mediated reduction of the monocyte-attracting chemokine CCL2 decreased monocyte chemotaxis in vitro and tumor macrophage density in vivo. Interestingly, dl922-947 treatment induced the switch of tumor macrophages toward a pro-inflammatory M1 phenotype, likely by increasing the expression of the pro-inflammatory cytokine interferon-γ. Altogether, we demonstrate that dl922-947 treatment re-shape the pro-tumorigenic ATC microenvironment by modulating cancer-cell intrinsic factors and the immune response. An in-depth knowledge of dl922-947-mediated effects on ATC microenvironment may help to refine ATC virotherapy in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Carmela Passaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Francesco Borriello
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Viviana Vastolo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Sarah Di Somma
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Eloise Scamardella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Vincenzo Gigantino
- CNR Institute of Experimental Endocrinology and Oncology “G. Salvatore”, Naples, Italy
| | - Renato Franco
- Experimental Oncology, IRCCS Fondazione Pascale, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- CNR Institute of Experimental Endocrinology and Oncology “G. Salvatore”, Naples, Italy
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
15
|
Passaro C, Portella G. Oncolytic virotherapy for thyroid cancer: will it translate to the clinic? INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2015. [DOI: 10.2217/ije.14.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Carmela Passaro
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Giuseppe Portella
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Naples, Italy
| |
Collapse
|
16
|
Abstract
Glioblastoma Multiforme (GBM) is a rapidly progressing brain tumor. Despite the relatively low percentage of cancer patients with glioma diagnoses, recent statistics indicate that the number of glioma patients may have increased over the past decade. Current therapeutic options for glioma patients include tumor resection, chemotherapy, and concomitant radiation therapy with an average survival of approximately 16 months. The rapid progression of gliomas has spurred the development of novel treatment options, such as cancer gene therapy and oncolytic virotherapy. Preclinical testing of oncolytic adenoviruses using glioma models revealed both positive and negative sides of the virotherapy approach. Here we present a detailed overview of the glioma virotherapy field and discuss auxiliary therapeutic strategies with the potential for augmenting clinical efficacy of GBM virotherapy treatment.
Collapse
Affiliation(s)
- I.V. Ulasov
- Swedish Medical Center, Center for Advanced Brain Tumor Treatment, 550 17th Avenue, James Tower, Suite 570, Seattle, WA 98122, USA
- Institute of Experimental Diagnostic and Biotherapy, N.N. Blokhin Cancer Research Center (RONC), Moscow 115478, Russia
- Corresponding author. Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, 550 17th Avenue, James Tower, Suite 570, Seattle, WA 98122, USA. Tel.: +1 206 991 2053; fax: +1 206 834 2608.
| | - A.V. Borovjagin
- Institute of Oral Health Research, University of Alabama at Birmingham School of Dentistry, 1919 7th Ave South, Birmingham, AL, 35294, USA
| | - B.A. Schroeder
- Michigan State University College of Medicine, Grand Rapids, MI, 49503, USA
| | - A.Y. Baryshnikov
- Institute of Experimental Diagnostic and Biotherapy, N.N. Blokhin Cancer Research Center (RONC), Moscow 115478, Russia
| |
Collapse
|
17
|
Passaro C, Volpe M, Botta G, Scamardella E, Perruolo G, Gillespie D, Libertini S, Portella G. PARP inhibitor olaparib increases the oncolytic activity of dl922-947 in in vitro and in vivo model of anaplastic thyroid carcinoma. Mol Oncol 2014; 9:78-92. [PMID: 25139258 DOI: 10.1016/j.molonc.2014.07.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 07/25/2014] [Accepted: 07/27/2014] [Indexed: 02/06/2023] Open
Abstract
PARP inhibitors are mostly effective as anticancer drugs in association with DNA damaging agents. We have previously shown that the oncolytic adenovirus dl922-947 induces extensive DNA damage, therefore we hypothesized a synergistic antitumoral effect of the PARP inhibitor olaparib in association with dl922-947. Anaplastic thyroid carcinoma was chosen as model since it is a particularly aggressive tumor and, because of its localized growth, it is suitable for intratumoral treatment with oncolytic viruses. Here, we show that dl922-947 infection induces PARP activation, and we confirm in vitro and in vivo that PARP inhibition increases dl922-947 replication and oncolytic activity. In vitro, the combination with olaparib exacerbates the appearance of cell death markers, such as Annexin V positivity, caspase 3 cleavage, cytochrome C release and propidium iodide permeability. In vivo, we also observed a better viral distribution upon PARP inhibition. Changes in CD31 levels suggest a direct effect of olaparib on tumor vascularization and on the viral distribution within the tumor mass. The observation that PARP inhibition enhances the effects of dl922-947 is highly promising not only for the treatment of anaplastic thyroid carcinoma but, in general, for the treatment of other tumors that could benefit from the use of oncolytic viruses.
Collapse
Affiliation(s)
- Carmela Passaro
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Massimiliano Volpe
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Ginevra Botta
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Eloise Scamardella
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Giuseppe Perruolo
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - David Gillespie
- The Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Silvana Libertini
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy; The Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow G61 1BD, UK.
| | - Giuseppe Portella
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Napoli, Italy.
| |
Collapse
|
18
|
Bressy C, Benihoud K. Association of oncolytic adenoviruses with chemotherapies: an overview and future directions. Biochem Pharmacol 2014; 90:97-106. [PMID: 24832861 DOI: 10.1016/j.bcp.2014.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/03/2014] [Accepted: 05/05/2014] [Indexed: 12/12/2022]
Abstract
Oncolytic adenoviruses have been used in different preclinical and clinical studies, showing their capacity to kill tumor cells without major adverse events. However, these studies also underline the limitations of this approach. The efficacy of oncolytic adenoviruses is hampered by their limited ability to transduce some tumor types, their lack of selectivity, and their poor dissemination within tumors. In addition, the host immune response may limit oncolytic adenovirus efficacy. Combining oncolytic adenoviruses with chemotherapeutics constitutes an appealing strategy to increase their potency. The first part of this review describes the molecular basis of oncolytic adenoviruses, their use in preclinical studies and clinical trials, their limitations, and strategies to circumvent these limitations. The second part will focus on studies combining oncolytic adenoviruses with chemotherapeutic drugs, including standard chemotherapeutic drugs, molecularly targeted drugs, and other drugs that have been combined with oncolytic adenoviruses. Finally, based on these studies, we describe future directions and general rules that could be followed to identify chemotherapeutic drugs displaying additive/synergistic effects when combined with oncolytic adenoviruses.
Collapse
Affiliation(s)
- Christian Bressy
- CNRS UMR 8203, Vectorologie et thérapeutiques anti-cancéreuses, Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif Cedex, France; Univ Paris-Sud, 15 rue Georges Clémenceau, 91405 Orsay Cedex, France
| | - Karim Benihoud
- CNRS UMR 8203, Vectorologie et thérapeutiques anti-cancéreuses, Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif Cedex, France; Univ Paris-Sud, 15 rue Georges Clémenceau, 91405 Orsay Cedex, France.
| |
Collapse
|
19
|
Passaro C, Abagnale A, Libertini S, Volpe M, Botta G, Cella L, Pacelli R, Halldèn G, Gillespie D, Portella G. Ionizing radiation enhances dl922-947-mediated cell death of anaplastic thyroid carcinoma cells. Endocr Relat Cancer 2013; 20:633-47. [PMID: 23839822 DOI: 10.1530/erc-13-0001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
dl922-947 is an oncolytic adenovirus potentially suitable for the treatment of aggressive localized tumors, such as anaplastic thyroid carcinoma (ATC). In this study, we have analyzed the effects of dl922-947 in combination with ionizing radiations, testing different schedules of administration and observing synergistic effects only when ATC cells were irradiated 24 h prior to viral infection. Cells undergoing combined treatment exhibited a marked increase in cell death and viral replication, suggesting that irradiation blocks cells in a more permissive state for viral life cycle. We also show that dl922-947 triggers a DNA damage response, characterized by mobilization of the MRN complex (composed by Mre11-Rad50-Nbs1), accumulation of γH2AX, and activation of the checkpoint kinases ataxia telangiectasia mutated (ATM) and Chk1. Based on these observations, we speculate that the DNA damage response acts as a cellular protective mechanism to hinder viral infection and replication. To confirm this hypothesis, we demonstrate that the ATM inhibitor KU55933 increased the oncolytic activity of dl922-947 and its replication. Finally, we validate the potential therapeutic use of this approach by showing in vivo that the combined treatment slows tumor xenograft growth more potently than either irradiation or infection alone.
Collapse
Affiliation(s)
- Carmela Passaro
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Facoltà di Medicina e Chirurgia, Università di Napoli Federico II, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Xu J, Mo Y, Wang X, Liu J, Zhang X, Wang J, Hu L, Yang C, Chen L, Wang Y. Conditionally replicative adenovirus-based mda-7/IL-24 expression enhances sensitivity of colon cancer cells to 5-fluorouracil and doxorubicin. J Gastroenterol 2013; 48:203-13. [PMID: 22820863 DOI: 10.1007/s00535-012-0623-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 05/28/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND Multiple drug resistance (MDR) greatly limits the efficacy of chemotherapy for colon cancer. An adenovirus armed with Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24; abbreviated to 'IL-24' here) was shown to reverse the MDR of colon cancer cells to oxaliplatin and doxorubicin. However, the relatively low expression level of IL-24 mediated by a replication-deficient adenoviral vector hindered its clinical application. METHODS To enhance IL-24-dependentreversion of the MDR phenotype, we utilized a conditionally replicative adenoviral vector, AdBB-IL24, to express IL-24 at a high level for more efficient MDR reversion. RESULTS An enzyme-linked immunosorbent assay (ELISA) suggested conditionally replicative adenoviral vector-mediated IL-24 expression was elevated in comparison with that of a replication-deficient adenoviral vector, Ad-IL24. AdBB-IL24 was shown to reverse MDR in colon cancer cells more potently than Ad-IL24. The AdBB-IL24-induced MDR reversion was linked to reduced P-glycoprotein (Pgp) and breast cancer resistance protein 1 (BCRP1) expression. Consistently, 5-fluorouracil and doxorubicin induced more apoptosis in AdBB-IL24-infected colon cancer cells compared with that in the Ad-IL24-infected cells. A cell viability assay showed that AdBB-IL24 could enhance the growth-inhibitory effect of 5-fluorouracil and doxorubicin on colon cancer cells more effectively than Ad-IL24 in vitro. In a mouse model, we also found that the combination of 5-fluorouracil and doxorubicin with AdBB-IL24 completely inhibited the growth of colon cancer cells. CONCLUSION We here provide evidence supporting conditionally replicative adenoviral vector-based gene therapy as a powerful strategy to enhance mda7/IL-24-dependent MDR reversion of colon cancer cells.
Collapse
Affiliation(s)
- Jing Xu
- Department of Hepatobiliary Surgery, The First People's Hospital of Yunnan Province, Kunhua Hospital Affiliated to Kunming Medical College, Kunming, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Halldén G, Portella G. Oncolytic virotherapy with modified adenoviruses and novel therapeutic targets. Expert Opin Ther Targets 2012; 16:945-58. [PMID: 22880939 DOI: 10.1517/14728222.2012.712962] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Numerous oncolytic viral mutants derived from a variety of strains have antitumor efficacy with limited or no toxicity to normal tissue. While all modes of administration were determined to be safe in patients with solid cancers refractory to current standard of care, this therapeutic approach requires further improvements to achieve definite efficacy. AREAS COVERED We review the most promising clinical developments with several oncolytic viruses. The focus is on preclinical and clinical findings with replication-selective adenoviral mutants including ONYX-015, H101 and Ad5ΔCR mutants that, to date, are the most studied oncolytic viruses. Cellular pathways reported to play a role in virus-induced cell killing are reviewed as potential targets for the development of more effective combinatorial therapies. EXPERT OPINION The most promising clinical outcomes for metastatic cancers have been reported for oncolytic vaccinia and herpes virus mutants expressing the cytokine GMCSF. However, highly efficacious and selective adenoviral mutants have been developed that interact synergistically with cytotoxic drugs in model systems. We anticipate that by delineating the cellular targets for synergistic cancer cell killing in response to adenoviral mutants and drugs such as apoptosis and autophagy signaling, greatly improved anticancer therapies will result in the near future.
Collapse
Affiliation(s)
- Gunnel Halldén
- Queen Mary University of London, Barts Cancer Institute, Centre for Molecular Oncology, London, UK
| | | |
Collapse
|
22
|
Preclinical efficacy of the oncolytic measles virus expressing the sodium iodide symporter in iodine non-avid anaplastic thyroid cancer: a novel therapeutic agent allowing noninvasive imaging and radioiodine therapy. Cancer Gene Ther 2012; 19:659-65. [PMID: 22790962 DOI: 10.1038/cgt.2012.47] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anaplastic thyroid cancer is an extremely aggressive disease resistant to radioiodine treatment because of loss of sodium iodide symporter (NIS) expression. To enhance prognosis of this fatal cancer, we validated the preclinical efficacy of measles virus (MV)-NIS, the vaccine strain of the oncolytic MV (MV-Edm), modified to include the NIS gene. Western blotting analysis confirmed that a panel of eight anaplastic thyroid cancer (ATC)-derived cell lines do not express NIS protein, but do express CD46, the MV receptor. In vitro cell death assays and in vivo xenograft studies demonstrate the oncolytic efficacy of MV-NIS in BHT-101 and KTC-3, ATC-derived cell lines. Radioactive iodine uptake along with single-photon emission computed tomography (SPECT)-computed tomography imaging of KTC-3 xenografts after (99)Tc(m) administration confirmed NIS expression in vitro and in vivo, respectively, after virus treatment. Adjuvant administration of RAI, to MV-NIS-treated KTC-3 tumors showed a trend for increased tumor cell killing. As current treatment for ATC is only palliative, and MV-NIS is currently Food and Drug Administration approved for human clinical trials in myeloma, our data indicate that targeting ATC with MV-NIS could prove to be a novel therapeutic strategy for effective treatment of iodine-resistant ATC and will expedite its testing in clinical trials for this aggressive disease.
Collapse
|
23
|
Botta G, Passaro C, Libertini S, Abagnale A, Barbato S, Maione AS, Hallden G, Beguinot F, Formisano P, Portella G. Inhibition of autophagy enhances the effects of E1A-defective oncolytic adenovirus dl922-947 against glioma cells in vitro and in vivo. Hum Gene Ther 2012; 23:623-34. [PMID: 22475378 DOI: 10.1089/hum.2011.120] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Oncolytic viruses represent a novel therapeutic approach for aggressive tumors, such as glioblastoma multiforme, which are resistant to available treatments. Autophagy has been observed in cells infected with oncolytic viruses; however, its role in cell death/survival is unclear. To elucidate the potential therapeutic use of autophagy modulators in association with viral therapy, we analyzed autophagy induction in human glioma cell lines U373MG and U87MG infected with the oncolytic adenovirus dl922-947. dl922-947 infection triggered an autophagic cellular response, as shown by the development of acidic vesicular organelles, LC3-I→LC3-II conversion, and reduction of p62 levels. However, on infection, the Akt/mTOR/p70s6k pathway, which negatively regulates autophagy, was activated, whereas the ERK1/2 pathway, a positive regulator of autophagy, was inhibited. Accordingly, MEK inhibition by PD98059 sensitized glioma cells to dl922-947 effects, whereas autophagy induction by rapamycin protected cells from dl922-947-induced death. Treatment with two inhibitors of autophagy, chloroquine and 3-methyladenine, increased the cytotoxic effects of dl922-947 in vitro. In vivo, the growth of U87MG-induced xenografts was further reduced by adding chloroquine to the dl922-947 treatment. In conclusion, autophagy acts as a survival response in glioma cells infected with dl922-947, thus suggesting autophagy inhibitors as adjuvant/neoadjuvant drugs in oncolytic virus-based treatments.
Collapse
Affiliation(s)
- Ginevra Botta
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università Federico II, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Dhurandhar EJ, Dubuisson O, Mashtalir N, Krishnapuram R, Hegde V, Dhurandhar NV. E4orf1: a novel ligand that improves glucose disposal in cell culture. PLoS One 2011; 6:e23394. [PMID: 21886789 PMCID: PMC3160302 DOI: 10.1371/journal.pone.0023394] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 07/14/2011] [Indexed: 12/31/2022] Open
Abstract
Reducing dietary fat intake and excess adiposity, the cornerstones of behavioral treatment of insulin resistance (IR), are marginally successful over the long term. Ad36, a human adenovirus, offers a template to improve IR, independent of dietary fat intake or adiposity. Ad36 increases cellular glucose uptake via a Ras-mediated activation of phosphatidyl inositol 3-kinase(PI3K), and improves hyperglycemia in mice, despite a high-fat diet and without reducing adiposity. Ex-vivo studies suggest that Ad36 improves hyperglycemia in mice by increasing glucose uptake by adipose tissue and skeletal muscle, and by reducing hepatic glucose output. It is impractical to use Ad36 for therapeutic action. Instead, we investigated if the E4orf1 protein of Ad36, mediates its anti-hyperglycemic action. Such a candidate protein may offer an attractive template for therapeutic development. Experiment-1 determined that Ad36 'requires' E4orf1 protein to up-regulate cellular glucose uptake. Ad36 significantly increased glucose uptake in 3T3-L1 preadipocytes, which was abrogated by knocking down E4orf1 with siRNA. Experiment-2 identified E4orf1 as 'sufficient' to up-regulate glucose uptake. 3T3-L1 cells that inducibly express E4orf1, increased glucose uptake in an induction-dependent manner, compared to null vector control cells. E4orf1 up-regulated PI3K pathway and increased abundance of Ras--the obligatory molecule in Ad36-induced glucose uptake. Experiment-3: Signaling studies of cells transiently transfected with E4orf1 or a null vector, revealed that E4orf1 may activate Ras/PI3K pathway by binding to Drosophila discs-large (Dlg1) protein. E4orf1 activated total Ras and, particularly the H-Ras isoform. By mutating the PDZ domain binding motif (PBM) of E4orf1, Experiment-4 showed that E4orf1 requires its PBM to increase Ras activation or glucose uptake. Experiment-5: In-vitro, a transient transfection by E4orf1 significantly increased glucose uptake in preadipocytes, adipocytes, or myoblasts, and reduced glucose output by hepatocytes. Thus, the highly attractive anti-hyperglycemic effect of Ad36 is mirrored by E4orf1 protein, which may offer a novel ligand to develop anti-hyperglycemic drugs.
Collapse
Affiliation(s)
- Emily J. Dhurandhar
- Infections and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Olga Dubuisson
- Infections and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Nazar Mashtalir
- Infections and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Rashmi Krishnapuram
- Infections and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Vijay Hegde
- Infections and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| | - Nikhil V. Dhurandhar
- Infections and Obesity Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
25
|
Bhattacharyya M, Francis J, Eddouadi A, Lemoine NR, Halldén G. An oncolytic adenovirus defective in pRb-binding (dl922-947) can efficiently eliminate pancreatic cancer cells and tumors in vivo in combination with 5-FU or gemcitabine. Cancer Gene Ther 2011; 18:734-43. [PMID: 21836633 DOI: 10.1038/cgt.2011.45] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pancreatic adenocarcinoma has a poor prognosis and frequently develops resistance to standard chemotherapeutics. Oncolytic adenoviruses represent a promising approach to overcome treatment resistance. The replication-selective dl922-947 adenovirus, defective in pRb binding, targets cancers with deregulated cell cycle control, such as the majority of pancreatic tumors. Cell killing efficacy was higher for dl922-947 than for adenovirus type 5 (Ad5) and the clinically approved dl1520 in pancreatic cancer cells with K-ras, p16 and p53 mutations. Combinations of dl922-947 and 5-fluorouracil or gemcitabine (2'2'-difluoro-2-deoxytidine) resulted in strong synergistic cell killing in Suit-2 and the highly drug- and virus-resistant Hs766T cells. Viral uptake increased in response to drugs, but was independent of the expression levels of the viral attachment receptor coxsackie and adenovirus receptor (CAR), whereas expression levels of the internalization receptors α(v)β(3)- and α(v)β(5)-integrins were increased. Early viral E1A expression was potently induced with drugs contributing to the synergistic effects. The dl922-947 mutant was more efficacious than Ad5 in vivo in Hs766T and Suit-2 xenograft models. In combination with gemcitabine, median survival was further prolonged. We demonstrate that dl922-947 is highly efficacious in pancreatic cancers and conclude that oncolytic adenoviruses harboring the E1ACR2 deletion have great potential for development into future clinical candidates for pancreatic cancer.
Collapse
Affiliation(s)
- M Bhattacharyya
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | | | | | | |
Collapse
|
26
|
Botta G, Perruolo G, Libertini S, Cassese A, Abagnale A, Beguinot F, Formisano P, Portella G. PED/PEA-15 modulates coxsackievirus-adenovirus receptor expression and adenoviral infectivity via ERK-mediated signals in glioma cells. Hum Gene Ther 2010; 21:1067-76. [PMID: 20406097 DOI: 10.1089/hum.2009.181] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive human brain tumor, and is highly resistant to chemo- and radiotherapy. Selectively replicating oncolytic viruses represent a novel approach for the treatment of neoplastic diseases. Coxsackievirus-adenovirus receptor (CAR) is the primary receptor for adenoviruses, and loss or reduction of CAR greatly decreases adenoviral entry. Understanding the mechanisms regulating CAR expression and localization will contribute to increase the efficacy of oncolytic adenoviruses. Two glioma cell lines (U343MG and U373MG) were infected with the oncolytic adenovirus dl922-947. U373MG cells were more susceptible to cell death after viral infection, compared with U343MG cells. The enhanced sensitivity was paralleled by increased adenoviral entry and CAR mRNA and protein levels in U373MG cells. In addition, U373MG cells displayed a decreased ERK1/2 (extracellular signal-regulated kinase-1/2) nuclear-to-cytosolic ratio, compared with U343MG cells. Intracellular content of PED/PEA-15, an ERK1/2-interacting protein, was also augmented in these cells. Both ERK2 overexpression and genetic silencing of PED/PEA-15 by antisense oligonucleotides increased ERK nuclear accumulation and reduced CAR expression and adenoviral entry. Our data indicate that dl922-947 could represent an useful tool for the treatment of GBM and that PED/PEA-15 modulates CAR expression and adenoviral entry, by sequestering ERK1/2.
Collapse
Affiliation(s)
- Ginevra Botta
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università Federico II, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Jakobisiak M, Golab J. Statins can modulate effectiveness of antitumor therapeutic modalities. Med Res Rev 2010; 30:102-35. [PMID: 19526461 DOI: 10.1002/med.20162] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Despite significant, frequently very strong, antiproliferative and tumoricidal effects of statins demonstrated in vitro, their antitumor effects in animal models are modest, and their efficacy in clinical trials has not been proven. As such, statins seem unlikely to be ever regarded as antitumor agents. However, statins are regularly taken by many elderly cancer patients for the prevention of cardiovascular events. Owing to their pleiotropic effects in normal and tumor cells, statins interact in various ways with many antitumor treatment modalities, either potentiating or diminishing their effectiveness. Elucidation of these interactions might affect the choice of treatment to be planned in cancer patients as some combinations might be contraindicated, whereas others might elicit potentiated antitumor effects but at a cost of increased general toxicity. Some other combinations might induce either comparable or even stronger antitumor effects, but with a beneficial concomitant reduction of specific side effects. Most of the studies reviewed in this article have been carried in vitro or in experimental tumor models, but clinical relevance of the findings is also discussed.
Collapse
Affiliation(s)
- Marek Jakobisiak
- Department of Immunology, Center of Biostructure Research, The Medical University of Warsaw, Warsaw, Poland.
| | | |
Collapse
|
28
|
GIAGINIS CONSTANTINOS, ZARROS APOSTOLOS, ALEXANDROU PARASKEVI, KLIJANIENKO JERZY, DELLADETSIMA IOANNA, THEOCHARIS STAMATIOS. Evaluation of coxsackievirus and adenovirus receptor expression in human benign and malignant thyroid lesions. APMIS 2010; 118:210-21. [DOI: 10.1111/j.1600-0463.2009.02582.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Meng HT, Li L, Liu H, Wang Y, Li GC, Qian WB. Homoharringtonine acts synergistically with SG235-TRAIL, a conditionally replicating adenovirus, in human leukemia cell lines. Acta Pharmacol Sin 2009; 30:1529-36. [PMID: 19820719 DOI: 10.1038/aps.2009.147] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIM To investigate the synergistic effects of SG235-TRAIL, a novel oncolytic adenovirus expressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and homoharringtonine (HHT) in human leukemia cell lines. METHODS The combined effect of SG235-TRAIL and HHT was assessed using a crystal violet assay and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, followed by combination index analysis. Cell apoptosis was measured using flow cytometry combined with fluorescein-isothiocyanate-Annexin V staining. The activation of caspase pathway and the expression of Bcl-2 family proteins, TRAIL, and E1A were examined using Western blotting. RESULTS HHT synergized the cytotoxicity of SG235-TRAIL against leukemia cell lines Kasumi-1, KG-1, HL-60, and U937, concomitantly with increased apoptosis and enhanced activity of caspase-3 and -9. The combination therapy resulted in significantly lower levels of Bcl-2, Mcl-1, and Bid compared to treatment of cells with either HHT or SG235-TRAIL alone, suggesting that HHT sensitizes leukemia cells to SG235-TRAIL virus through alteration of anti-apoptotic signaling elements. Importantly, HHT combined with SG235-TRAIL did not show significant cytotoxicity to normal human mononuclear cells and mesenchymal stem cells. CONCLUSION Combining oncolytic adenovirus SG235-TRAIL and HHT synergistically enhances cytotoxicity in leukemia cells in vitro, suggesting that the combination therapy could represent a rational approach for the treatment of leukemia.
Collapse
|
30
|
Abstract
Anaplastic thyroid cancer (ATC) is a rare malignancy. While external beam radiation therapy has improved locoregional control, the median survival of approximately 4 months has not changed in more than half a century due to uncontrolled systemic metastases. The objective of this study was to review the literature in order to identify potential new strategies for treating this highly lethal cancer. PubMed searches were the principal source of articles reviewed. The molecular pathogenesis of ATC includes mutations in BRAF, RAS, catenin (cadherin-associated protein), beta 1, PIK3CA, TP53, AXIN1, PTEN, and APC genes, and chromosomal abnormalities are common. Several microarray studies have identified genes and pathways preferentially affected, and dysregulated microRNA profiles differ from differentiated thyroid cancers. Numerous proteins involving transcription factors, signaling pathways, mitosis, proliferation, cell cycle, apoptosis, adhesion, migration, epigenetics, and protein degradation are affected. A variety of agents have been successful in controlling ATC cell growth both in vitro and in nude mice xenografts. While many of these new compounds are in cancer clinical trials, there are few studies being conducted in ATC. With the recent increased knowledge of the many critical genes and proteins affected in ATC, and the extensive array of targeted therapies being developed for cancer patients, there are new opportunities to design clinical trials based upon tumor molecular profiling and preclinical studies of potentially synergistic combinatorial novel therapies.
Collapse
Affiliation(s)
- Robert C Smallridge
- Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida 32224, USA.
| | | | | |
Collapse
|
31
|
Libertini S, Iacuzzo I, Perruolo G, Scala S, Ieranò C, Franco R, Hallden G, Portella G. Bevacizumab increases viral distribution in human anaplastic thyroid carcinoma xenografts and enhances the effects of E1A-defective adenovirus dl922-947. Clin Cancer Res 2008; 14:6505-14. [PMID: 18927290 DOI: 10.1158/1078-0432.ccr-08-0200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Anaplastic thyroid carcinoma is a prime target for innovative therapy because it represents one of the most lethal human neoplasms and is refractory to conventional treatments such as chemotherapy and radiotherapy. We have evaluated a novel therapeutic approach based on the oncolytic replication-selective adenovirus dl922-947. EXPERIMENTAL DESIGN The antitumor efficacies of the E1ADeltaCR2 (dl922-947) and DeltaE1B55K (dl1520) mutants were compared in human thyroid anaplastic carcinoma cells in culture and in xenografts in vivo. To enhance the effects of dl922-947, anaplastic thyroid carcinoma tumor xenografts were treated with dl922-947 in combination with bevacizumab. RESULTS We showed that the efficacy of dl922-947 exceeded that of dl1520 in all tested anaplastic thyroid carcinoma cells in vitro and in vivo. Furthermore, bevacizumab in combination with dl922-947 significantly reduced tumor growth compared with single treatments alone. Bevacizumab treatment significantly improved viral distribution in neoplastic tissues. CONCLUSIONS Our data showed that dl922-947 had a higher oncolytic activity compared with dl1520 in anaplastic thyroid carcinoma cell lines and might represent a better option for virotherapy of anaplastic thyroid carcinoma. Moreover, bevacizumab increased the oncolytic effects of dl922-947 by enhancing viral distribution in tumors. The results described herein encourage the use of the dl922-947 virus in combination with bevacizumab.
Collapse
Affiliation(s)
- Silvana Libertini
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Universita di Napoli, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lovastatin induces apoptosis of k-ras-transformed thyroid cells via inhibition of ras farnesylation and by modulating redox state. J Mol Med (Berl) 2008; 86:1341-51. [DOI: 10.1007/s00109-008-0396-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 08/01/2008] [Accepted: 08/04/2008] [Indexed: 01/29/2023]
|
33
|
Rogers PM, Mashtalir N, Rathod MA, Dubuisson O, Wang Z, Dasuri K, Babin S, Gupta A, Markward N, Cefalu WT, Dhurandhar NV. Metabolically favorable remodeling of human adipose tissue by human adenovirus type 36. Diabetes 2008; 57:2321-31. [PMID: 18599527 PMCID: PMC2518483 DOI: 10.2337/db07-1311] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Accepted: 06/11/2008] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Experimental infection of rats with human adenovirus type 36 (Ad-36) promotes adipogenesis and improves insulin sensitivity in a manner reminiscent of the pharmacologic effect of thiozolinediones. To exploit the potential of the viral proteins as a therapeutic target for treating insulin resistance, this study investigated the ability of Ad-36 to induce metabolically favorable changes in human adipose tissue. RESEARCH DESIGN AND METHODS We determined whether Ad-36 increases glucose uptake in human adipose tissue explants. Cell-signaling pathways targeted by Ad-36 to increase glucose uptake were determined in the explants and human adipose-derived stem cells. Ad-2, a nonadipogenic human adenovirus, was used as a negative control. As a proof of concept, nondiabetic and diabetic subjects were screened for the presence of Ad-36 antibodies to ascertain if natural Ad-36 infection predicted improved glycemic control. RESULTS Ad-36 increased glucose uptake by adipose tissue explants obtained from nondiabetic and diabetic subjects. Without insulin stimulation, Ad-36 upregulated expressions of several proadipogenic genes, adiponectin, and fatty acid synthase and reduced the expression of inflammatory cytokine macrophage chemoattractant protein-1 in a phosphotidylinositol 3-kinase (PI3K)-dependent manner. In turn, the activation of PI3K by Ad-36 was independent of insulin receptor signaling but dependent on Ras signaling recruited by Ad-36. Ad-2 was nonadipogenic and did not increase glucose uptake. Natural Ad-36 infection in nondiabetic and diabetic subjects was associated with significantly lower fasting glucose levels and A1C, respectively. CONCLUSIONS Ad-36 proteins may provide novel therapeutic targets that remodel human adipose tissue to a more metabolically favorable profile.
Collapse
Affiliation(s)
- Pamela M. Rogers
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Nazar Mashtalir
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Miloni A. Rathod
- Department of Nutrition, Wayne State University, Detroit, Michigan
| | - Olga Dubuisson
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Zhong Wang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Kumar Dasuri
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Scott Babin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Alok Gupta
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Nathan Markward
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - William T. Cefalu
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Nikhil V. Dhurandhar
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| |
Collapse
|
34
|
Bifulco M. Therapeutic potential of statins in thyroid proliferative disease. ACTA ACUST UNITED AC 2008; 4:242-3. [DOI: 10.1038/ncpendmet0782] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2007] [Accepted: 12/17/2007] [Indexed: 11/09/2022]
|