1
|
Falanga AP, Lupia A, Tripodi L, Morgillo CM, Moraca F, Roviello GN, Catalanotti B, Amato J, Pastore L, Cerullo V, D'Errico S, Piccialli G, Oliviero G, Borbone N. Exploring the DNA 2-PNA heterotriplex formation in targeting the Bcl-2 gene promoter: A structural insight by physico-chemical and microsecond-scale MD investigation. Heliyon 2024; 10:e24599. [PMID: 38317891 PMCID: PMC10839560 DOI: 10.1016/j.heliyon.2024.e24599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Peptide Nucleic Acids (PNAs) represent a promising tool for gene modulation in anticancer treatment. The uncharged peptidyl backbone and the resistance to chemical and enzymatic degradation make PNAs highly advantageous to form stable hybrid complexes with complementary DNA and RNA strands, providing higher stability than the corresponding natural analogues. Our and other groups' research has successfully shown that tailored PNA sequences can effectively downregulate the expression of human oncogenes using antigene, antisense, or anti-miRNA approaches. Specifically, we identified a seven bases-long PNA sequence, complementary to the longer loop of the main G-quadruplex structure formed by the bcl2midG4 promoter sequence, capable of downregulating the expression of the antiapoptotic Bcl-2 protein and enhancing the anticancer activity of an oncolytic adenovirus. Here, we extended the length of the PNA probe with the aim of including the double-stranded Bcl-2 promoter among the targets of the PNA probe. Our investigation primarily focused on the structural aspects of the resulting DNA2-PNA heterotriplex that were determined by employing conventional and accelerated microsecond-scale molecular dynamics simulations and chemical-physical analysis. Additionally, we conducted preliminary biological experiments using cytotoxicity assays on human A549 and MDA-MB-436 adenocarcinoma cell lines, employing the oncolytic adenovirus delivery strategy.
Collapse
Affiliation(s)
- Andrea P. Falanga
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Antonio Lupia
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Lorella Tripodi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore S.c.a.r.l., Naples, 80145, Italy
| | - Carmine M. Morgillo
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Federica Moraca
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Giovanni N. Roviello
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale Delle Ricerche, Naples, 80131, Italy
| | - Bruno Catalanotti
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Jussara Amato
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Lucio Pastore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore S.c.a.r.l., Naples, 80145, Italy
| | - Vincenzo Cerullo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
- ImmunoViroTherapy Lab (IVT), Drug Research Program (DRP), Faculty of Pharmacy, University of Helsinki, 00100, Helsinki, Finland
| | - Stefano D'Errico
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Gennaro Piccialli
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Giorgia Oliviero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| | - Nicola Borbone
- Dipartimento di Farmacia, Università Degli Studi di Napoli Federico II, Naples, 80131, Italy
| |
Collapse
|
2
|
Hashemi Goradel N, Nemati M, Bakhshandeh A, Arashkia A, Negahdari B. Nanovaccines for cancer immunotherapy: Focusing on complex formation between adjuvant and antigen. Int Immunopharmacol 2023; 117:109887. [PMID: 36841155 DOI: 10.1016/j.intimp.2023.109887] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 02/27/2023]
Abstract
As an interesting cancer immunotherapy approach, cancer vaccines have been developed to deliver tumor antigens and adjuvants to antigen-presenting cells (APCs). Although the safety and easy production shifted the vaccine designing platforms toward the subunit vaccines, their efficacy is limited due to inefficient vaccine delivery. Nanotechnology-based vaccines, called nanovaccines, address the delivery limitations through co-delivery of antigens and adjuvants into lymphoid organs and APCs and their intracellular release, leading to cross-presentation of antigens and induction of potent anti-tumor immune responses. Although the nanovaccines, either as encapsulating agents or biomimetic nanoparticles, exert the desired anti-tumor activities, there is evidence that the mixing formulation to form nanocomplexes between antigens and adjuvants based on the electrostatic interactions provokes high levels of immune responses owing to Ags' availability and faster release. Here, we summarized the various platforms for developing cancer vaccines and the advantages of using delivery systems. The cancer nanovaccines, including nanoparticle-based and biomimetic-based nanovaccines, are discussed in detail. Finally, we focused on the nanocomplexes formation between antigens and adjuvants as promising cancer nanovaccine platforms.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Bakhshandeh
- Department of Industrial Engineering and Management Systems, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Kuryk L, Møller ASW. Next generation oncolytic viruses expressing PADI1 and TIMP2 exhibit anti-tumor activity against melanoma in nude and humanized mouse models. Mol Ther Oncolytics 2023; 28:158-170. [PMID: 36816748 PMCID: PMC9922816 DOI: 10.1016/j.omto.2023.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Immunotherapy of metastatic melanoma (MM) has vastly improved the longevity of only a minority of patients. To broaden the repertoire of agents against MM, we investigated the effectiveness of locally interrupting tumor blood endothelial cell proliferation and angiogenesis, arginine deprivation, or both on the growth of melanoma by constructing and characterizing the effectiveness of four oncolytic adenoviruses. ONCOS-207 (which expressed tissue inhibitor of metalloprotease type 2 [TIMP2]), ONCOS-209 (which expressed peptidyl arginine deiminase [PADI1]), and ONCOS-210 and ONCOS-212 (which expressed both TIMP2 and PADI1) exhibited oncolytic activity against four melanoma cell lines in vitro. ONCOS-212 treatments significantly inhibited tumor growth in an A2058 tumor model in nude mice compared with vehicle control. The inhibitory effects of the two transgenes of ONCOS-212 on tumor growth appeared to be synergistic. These viruses also significantly inhibited tumor growth in a humanized NOG model of melanoma (A2058 xenograft). All viruses significantly increased the percentage of activated CD8+ T cells in the tumor-infiltrating lymphocytes. The abscopal effect of ONCOS-212 treatments in the A2058 tumor challenge model in hNOG mice supports the hypothesis that the human immune response contributes to the anti-tumor activity of ONCOS-212. These results support the further development of ONCOS-212 for cancer treatment.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Targovax ASA, Clinical Science, Vollsveien 19, NO-1366 Lysaker Oslo, Norway,National Institute of Public Health NIH – National Research Institute, Department of Virology, Chocimska 24, 00-791 Warsaw, Poland,Corresponding author: Lukasz Kuryk, National Institute of Public Health NIH – National Research Institute, Department of Virology, Chocimska 24, 00-791 Warsaw, Poland.
| | | |
Collapse
|
4
|
Saini U, Smith BQ, Dorayappan KDP, Yoo JY, Maxwell GL, Kaur B, Konishi I, O’Malley D, Cohn DE, Selvendiran K. Targeting TMEM205 mediated drug resistance in ovarian clear cell carcinoma using oncolytic virus. J Ovarian Res 2022; 15:130. [PMID: 36476493 PMCID: PMC9730683 DOI: 10.1186/s13048-022-01054-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 10/17/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) accounts for approximately 8-10% of epithelial ovarian cancers in the United States. Although it is rare, OCCC usually presents with treatment challenges and the overall prognosis is far worse than high grade serous ovarian cancer HGSOC. The objective of this study was to examine the therapeutic relevance of combining oncolytic virus with cisplatin for ovarian cancer clear cell carcinoma (OCCC). RESULTS We identified that TMEM205, a recently discovered transmembrane protein, contributes to chemoresistance in OCCC cells via the exosomal pathway. Mechanistically, TMEM205 undergoes ligand-independent constitutive endocytosis and co-localizes with Rab11 to contribute to the late recycling endosomes in a clathrin-independent manner. Further, we observed that oncolytic virus (oHSV) pretreatment followed by treatment with cisplatin decreases TMEM205 expression and sensitizes cells to cisplatin in a synergistic manner in OCCC cells. TMEM205 interacts with glycoprotein-C of oHSV post-infection; both of these proteins undergo ubiquitination and ultimately get shuttled outside the cell via exosomes. Thus, we demonstrate the mechanotransduction pathway of TMEM205-mediated chemoresistance along with targeting this pathway using oHSV and cisplatin as a powerful therapeutic strategy for OCCC. oHSV combination with cisplatin inhibits OCCC tumor growth in vivo in immunodeficient and immunocompetent mice models. CONCLUSION Our results suggest that the combination of oHSV and cisplatin in immunocompetent as well as immune deficient OCCC tumor bearing mice reduces overall tumor burden as well as metastatic disease thereby providing survival benefit. Additionally, the detection of TMEM205 in exosomal cargo early in OCCC development has potential to be exploited as a biomarker.
Collapse
Affiliation(s)
- Uksha Saini
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Brentley Q. Smith
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Kalpana Deepa Priya Dorayappan
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Ji Young Yoo
- grid.267308.80000 0000 9206 2401Department of Neurosurgery, University of Texas, Health Science Center, Houston, USA
| | - G. Larry Maxwell
- grid.414629.c0000 0004 0401 0871Inova Women’s Service Line and the Inova Schar Cancer Institute, Falls Church, VA USA
| | - Balveen Kaur
- grid.267308.80000 0000 9206 2401Department of Neurosurgery, University of Texas, Health Science Center, Houston, USA
| | - Ikuo Konishi
- grid.258799.80000 0004 0372 2033Division of GYN/ONC, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - David O’Malley
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - David E. Cohn
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
5
|
Achmad H, Saleh Ibrahim Y, Mohammed Al-Taee M, Gabr GA, Waheed Riaz M, Hamoud Alshahrani S, Alexis Ramírez-Coronel A, Turki Jalil A, Setia Budi H, Sawitri W, Elena Stanislavovna M, Gupta J. Nanovaccines in cancer immunotherapy: Focusing on dendritic cell targeting. Int Immunopharmacol 2022; 113:109434. [DOI: 10.1016/j.intimp.2022.109434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
|
6
|
Wang J, Zuo S, Zhang Y, Li S, Shi Y, Du T, Han J, Jin N, Li Y, Li X. Recombinant Oncolytic Adenovirus Combined with Cyclophosphamide Induces Synergy in the Treatment of Breast Cancer in vitro and in vivo. Cancer Manag Res 2022; 14:2749-2761. [PMID: 36133740 PMCID: PMC9484773 DOI: 10.2147/cmar.s373271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/08/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose Oncolytic virus therapy has gradually become an integral approach in cancer treatment. We explored the therapeutic effects of the combination of a dual cancer-selective anti-tumor recombinant adenovirus (Ad-Apoptin-hTERTp-E1a) and cyclophosphamide on breast cancer cells. Methods The inhibition of MCF-7 and MDA-MB-231 breast cancer cells by Ad-Apoptin-hTERTp-E1a (Ad-VT), cyclophosphamide, and Ad-VT + Cyclophosphamide was investigated using the CCK-8 assay. The combination index (CI) was calculated using CalcuSyn software to determine the best combination based on the inhibition rates of the different treatment combinations. The CCK-8 assay and crystal violet staining were used to detect the cytotoxicity of the combined Ad-VT and cyclophosphamide in breast cancer cells and breast epithelial cells. Subsequently, Hoechst staining, annexin V flow cytometry, and JC-1 staining were used to analyze the inhibitory pathway of Ad-VT plus cyclophosphamide on breast cancer cells. Cell migration and invasion of breast cancer cells were assessed using the cell-scratch and Transwell assays. The anti-tumor effects of different treatment groups in a tumor-bearing nude mouse model also were analyzed. Results The treatment combination of Ad-VT (40 MOI) and cyclophosphamide (400 µM) significantly inhibited MCF-7 and MDA-MB-231 cells and reduced the toxicity of cyclophosphamide in normal cells. Ad-VT primarily induced breast cancer cell apoptosis through the endogenous apoptotic pathway. Apoptosis was significantly increased after treatment with Ad-VT plus cyclophosphamide. The combination significantly inhibited the migration and invasion of MCF-7 and MDA-MB-231 cells. The in vivo experiments demonstrated that exposure to Ad-VT plus cyclophosphamide significantly inhibited tumor growth and extended the survival time of the nude mice. Conclusion Ad-VT plus cyclophosphamide reduced toxicity and exhibited increased efficacy in treating breast cancer cells.
Collapse
Affiliation(s)
- Jing Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Shuting Zuo
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Yan Zhang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Shanzhi Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Ying Shi
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Tonghua Du
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, 130000, People's Republic of China
| | - Jicheng Han
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Ningyi Jin
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, People's Republic of China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China
| | - Xiao Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130117, People's Republic of China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, People's Republic of China
| |
Collapse
|
7
|
Kuryk L, Rodella G, Staniszewska M, Pancer KW, Wieczorek M, Salmaso S, Caliceti P, Garofalo M. Novel Insights Into Mesothelioma Therapy: Emerging Avenues and Future Prospects. Front Oncol 2022; 12:916839. [PMID: 35785199 PMCID: PMC9247278 DOI: 10.3389/fonc.2022.916839] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022] Open
Abstract
Malignant mesothelioma is a rare and aggressive cancer that develops in the thin layer surrounding the mesothelium and is mainly caused by asbestos exposure. Despite improvements in patient prognosis with conventional cancer treatments, such as surgery, chemotherapy, and radiotherapy, there are still no curative treatment modalities for advanced disease. In recent years, new therapeutic avenues have been explored. Improved understanding of the mechanisms underlying the dynamic tumor interaction with the immune system has led to the development of immunotherapeutic approaches. Numerous recent clinical trials have shown a desire to develop more effective treatments that can be used to fight against the disease. Immune checkpoint inhibitors, oncolytic adenoviruses, and their combination represent a promising strategy that can be used to synergistically overcome immunosuppression in the mesothelioma tumor microenvironment. This review provides a synthesized overview of the current state of knowledge on new therapeutic options for mesothelioma with a focus on the results of clinical trials conducted in the field.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health National Institute of Hygiene (NIH)—National Institute of Research, Warsaw, Poland
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Giulia Rodella
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Katarzyna Wanda Pancer
- Department of Virology, National Institute of Public Health National Institute of Hygiene (NIH)—National Institute of Research, Warsaw, Poland
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health National Institute of Hygiene (NIH)—National Institute of Research, Warsaw, Poland
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
8
|
Xu W, Fang F, Wang Y, Qin L, Han Y, Huang Y, Li B, Liu Y, Wang Z. Co-overexpression of TRAIL and Smac sensitizes MDA-MB-231 cells to radiation through apoptosis depending on mitochondrial pathway. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:37-48. [PMID: 35006369 DOI: 10.1007/s00411-021-00961-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Pro-apoptosis in cancer cells has been proposed as a beneficial therapeutic strategy for potentiating the anticancer effects of radiotherapy. TNF-related apoptosis inducing ligand (TRAIL) and Second mitochondria derived activator of caspase (Smac) can induce cell apoptosis. Herein, we designed a conditionally replicating adenoviral co-overexpression vector of TRAIL and Smac regulated by the Egr1 promoter, in which hTERT, E1A-E1B and E1B55K genes were inserted to achieve enhanced tumor targeting characteristics. After breast cancer MDA-MB-231 cells were infected and irradiated, cellular proliferation and colony formation were measured, apoptotic rate was detected by FCM after AnnexinV-FITC/PI staining. To explore the molecular mechanisms of apoptosis, mRNA and protein levels of TRAIL, Smac, Cytochrome c (Cyt c), death receptor 5 (DR5), caspase-8, -9 and -3 were measured by quantitative real-time PCR, ELISA and Western blot, and caspase-3 activity was detected using caspase-3 activity kits. The results showed that TRAIL and/or Smac overexpression enhanced proliferation inhibition and radio-sensitivity through apoptosis. In addition, the combination of IR and overexpression of TRAIL and/or Smac can activate more apoptosis in tumor cells, and the transcriptional levels and protein expressions of Cyt c, DR5, caspase-8, -9 and -3 had similar regularity with apoptotic changes, indicating the molecular mechanisms of TRAIL and Smac involves the mitochondrial pathway. Our findings may have implications for novel radiotherapy plans for breast tumor treatment.
Collapse
Affiliation(s)
- Weiqiang Xu
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Fang Fang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yuexuan Wang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Lijing Qin
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yu Han
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yuwei Huang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Bin Li
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yang Liu
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China.
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
9
|
Heterologous administration of HPV16 E7 epitope-loaded nanocomplexes inhibits tumor growth in mouse model. Int Immunopharmacol 2021; 101:108298. [PMID: 34739928 DOI: 10.1016/j.intimp.2021.108298] [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] [Received: 05/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/02/2023]
Abstract
The nanostructured complexes can result in enhanced vaccine efficacy by facilitating the distribution and uptake of antigens by antigen-presenting cells (APCs), thereby stimulating immune responses. Here, we hypothesized that either directly coating of nanoadjuvants including aluminum phosphate (AlPO4) and adenovirus (Ad) with a modified HPV16 E7 MHC-I specific epitope, RAHYNIVTF49-57, or mixing the CpG oligodeoxynucleotide (CpG-ODN) with the cationic epitope to form nanocomlexes, and their combinational therapy would enhance their anti-tumor effects in a TC-1 mouse model. The positively-charged HPV16 E7 epitope was attracted to the oppositely-charged adjuvants by electrostatic interaction to generate epitope/adjuvant nanocomplexes. We showed that coating the nanosized adjuvants with the cationic epitope increased the particles' surface charge without significant change in their size. We then tested the cellular immunogenicity and therapeutic efficacy of nanocomplexes by measuring IL-10 and IFN-γ production, the expression of CD107a as a marker of CTL response, and tumor growth inhibition. The nanocomplexes were administered either in homologous or heterologous prime-boost regimens, and heterologous immunizations including Ad/Pep-CpG/Pep, CpG/Pep-Ad/Pep, Ad/Pep-Alum/Pep, and Alum/Pep-Ad/Pep induced significantly higher levels of IL-10, IFN-γ, and CD107a-expressing CD8 T cells compared with homologous administrations. Furthermore, the tumor growth was significantly suppressed in mice receiving nanostructured complexes in the heterologous immunizations. Our study highlights the potential of the heterologous prime-boost administration of the epitope-coated nanostructures as an effective immunization strategy.
Collapse
|
10
|
Darvishi B, Dinarvand R, Mohammadpour H, Kamarul T, Sharifi AM. Dual l-Carnosine/ Aloe vera Nanophytosomes with Synergistically Enhanced Protective Effects against Methylglyoxal-Induced Angiogenesis Impairment. Mol Pharm 2021; 18:3302-3325. [PMID: 34297586 DOI: 10.1021/acs.molpharmaceut.1c00248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Microvascular complications are among the major outcomes of patients with type II diabetes mellitus, which are the consequences of impaired physiological functioning of small blood vessels and angiogenic responses in these patients. Overproduction and accumulation of methylglyoxal (MGO), a highly reactive dicarbonyl byproduct of glycolysis pathway, has been acclaimed as the main inducer of impaired angiogenic responses and microvascular dysfunction in diabetic patients with uncontrolled hyperglycemia. Hence, an effective approach to overcome diabetes-associated microvascular complications is to neutralize the deleterious activity of enhanced the concentration of MGO in the body. Owing to the glycation inhibitory activity of Aloe vera whole extract, and capability of l-carnosine, an endogenous dipeptide, in attenuating MGO's destructive activity, we examined whether application of a combination of l-carnosine and A. vera could be an effective way of synergistically weakening this reactive dicarbonyl's impaired angiogenic effects. Additionally, overcoming the poor cellular uptake and internalization of l-carnosine and A. vera, a nanophytosomal formulation of the physical mixture of two compounds was also established. Although l-carnosine and A. vera at whole studied combination ratios could synergistically enhance viability of human umbilical vein endothelial cells (HUVECs) treated with MGO, the 25:1 w/w ratio was the most effective one among the others (27 ± 0.5% compared to 12 ± 0.3 to 18 ± 0.4%; F (4, 15) = 183.9, P < 0.0001). Developing dual nanophytosomes of l-carnosine/A. vera (25:1) combination ratio, we demonstrated superiority of the nanophytosomal formulation in protecting HUVECs against MGO-induced toxicity following a 24-72 h incubation period (17.3, 15.8, and 12.4% respectively). Moreover, 500 μg/mL concentration of dual l-carnosine/A. vera nanophytosomes exhibited a superior free radical scavenging potency (63 ± 4 RFU vs 83 ± 5 RFU; F (5, 12) = 54.81, P < 0.0001) and nitric oxide synthesizing capacity (26.11 ± 0.19 vs 5.1 ± 0.33; F (5, 12) = 2537, P < 0.0001) compared to their physical combination counterpart. Similarly, 500 μg/mL dual l-carnosine/A. vera nanophytosome-treated HUVECs demonstrated a superior tube formation capacity (15 ± 3 vs 2 ± 0.3; F (5, 12) = 30.87, P < 0.001), wound scratch healing capability (4.92 ± 0.3 vs 3.07 ± 0.3 mm/h; F (5, 12) = 39.21, P < 0.0001), and transwell migration (586 ± 32 vs 394 ± 18; F (5, 12) = 231.8, P < 0.001) and invasion (172 ± 9 vs 115 ± 5; F (5, 12) = 581.1, P < 0.0001) activities compared to the physical combination treated ones. Further confirming the proangiogenic activity of the dual l-carnosine/A. vera nanophytosomes, a significant shift toward expression of proangiogenic genes including HIF-1α, VEGFA, bFGF, KDR, and Ang II was reported in treated HUVECs. Overall, dual l-carnosine/A. vera nanophytosomes could be a potential candidate for attenuating type II DM-associated microvascular complications with an impaired angiogenesis background.
Collapse
Affiliation(s)
- Behrad Darvishi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran.,Razi Drug Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Hadiseh Mohammadpour
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran 14155-5583, Iran
| | - Tunku Kamarul
- Tissue Engineering Group, (NOCERAL), Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ali Mohammad Sharifi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran.,Razi Drug Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran.,Tissue Engineering Group, (NOCERAL), Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.,Stem cell and Regenerative Medicine research center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| |
Collapse
|
11
|
Garofalo M, Bellato F, Magliocca S, Malfanti A, Kuryk L, Rinner B, Negro S, Salmaso S, Caliceti P, Mastrotto F. Polymer Coated Oncolytic Adenovirus to Selectively Target Hepatocellular Carcinoma Cells. Pharmaceutics 2021; 13:pharmaceutics13070949. [PMID: 34202714 PMCID: PMC8309094 DOI: 10.3390/pharmaceutics13070949] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 01/15/2023] Open
Abstract
Despite significant advances in chemotherapy, the overall prognosis of hepatocellular carcinoma (HCC) remains extremely poor. HCC targeting strategies were combined with the tumor cell cytotoxicity of oncolytic viruses (OVs) to develop a more efficient and selective therapeutic system. OVs were coated with a polygalactosyl-b-agmatyl diblock copolymer (Gal32-b-Agm29), with high affinity for the asialoglycoprotein receptor (ASGPR) expressed on the liver cell surface, exploiting the electrostatic interaction of the positively charged agmatine block with the negatively charged adenoviral capsid surface. The polymer coating altered the viral particle diameter (from 192 to 287 nm) and zeta-potential (from -24.7 to 23.3 mV) while hiding the peculiar icosahedral symmetrical OV structure, as observed by TEM. Coated OVs showed high potential therapeutic value on the human hepatoma cell line HepG2 (cytotoxicity of 72.4% ± 4.96), expressing a high level of ASGPRs, while a lower effect was attained with ASPGR-negative A549 cell line (cytotoxicity of 54.4% ± 1.59). Conversely, naked OVs showed very similar effects in both tested cell lines. Gal32-b-Agm29 OV coating enhanced the infectivity and immunogenic cell death program in HepG2 cells as compared to the naked OV. This strategy provides a rationale for future studies utilizing oncolytic viruses complexed with polymers toward effective treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (F.B.); (S.M.); (S.S.); (P.C.)
- Correspondence: (M.G.); (F.M.); Tel.: +39-04-9827-5710 (M.G.); +39-04-9827-5708 (F.M.)
| | - Federica Bellato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (F.B.); (S.M.); (S.S.); (P.C.)
| | - Salvatore Magliocca
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (F.B.); (S.M.); (S.S.); (P.C.)
| | - Alessio Malfanti
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1 73.12, 1200 Brussels, Belgium;
| | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health—National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland;
- Clinical Science, Targovax Oy, Saukonpaadenranta 2, 00180 Helsinki, Finland
| | - Beate Rinner
- Division of Biomedical Research, Medical University of Graz, Roseggerweg 48, 8036 Graz, Austria;
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (F.B.); (S.M.); (S.S.); (P.C.)
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (F.B.); (S.M.); (S.S.); (P.C.)
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (F.B.); (S.M.); (S.S.); (P.C.)
- Correspondence: (M.G.); (F.M.); Tel.: +39-04-9827-5710 (M.G.); +39-04-9827-5708 (F.M.)
| |
Collapse
|
12
|
Kana SI, Essani K. Immuno-Oncolytic Viruses: Emerging Options in the Treatment of Colorectal Cancer. Mol Diagn Ther 2021; 25:301-313. [PMID: 33713031 DOI: 10.1007/s40291-021-00517-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 12/18/2022]
Abstract
Colorectal cancer is the third most common neoplasm in the world and the third leading cause of cancer-related deaths in the USA. A safer and more effective therapeutic intervention against this malignant carcinoma is called for given the limitations and toxicities associated with the currently available treatment modalities. Immuno-oncolytic or oncolytic virotherapy, the use of viruses to selectively or preferentially kill cancer cells, has emerged as a potential anticancer treatment modality. Oncolytic viruses act as double-edged swords against the tumors through the direct cytolysis of cancer cells and the induction of antitumor immunity. A number of such viruses have been tested against colorectal cancer, in both preclinical and clinical settings, and many have produced promising results. Oncolytic virotherapy has also shown synergistic antitumor efficacy in combination with conventional treatment regimens. In this review, we describe the status of this therapeutic approach against colorectal cancer at both preclinical and clinical levels. Successes with and the challenges of using oncolytic viruses, both as monotherapy and in combination therapy, are also highlighted.
Collapse
Affiliation(s)
- Sadia Islam Kana
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008-5410, USA
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008-5410, USA.
| |
Collapse
|
13
|
Yong T, Li X, Wei Z, Gan L, Yang X. Extracellular vesicles-based drug delivery systems for cancer immunotherapy. J Control Release 2020; 328:562-574. [DOI: 10.1016/j.jconrel.2020.09.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
|
14
|
Kuryk L, Bertinato L, Staniszewska M, Pancer K, Wieczorek M, Salmaso S, Caliceti P, Garofalo M. From Conventional Therapies to Immunotherapy: Melanoma Treatment in Review. Cancers (Basel) 2020; 12:cancers12103057. [PMID: 33092131 PMCID: PMC7589099 DOI: 10.3390/cancers12103057] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Here, we review the current state of knowledge in the field of cancer immunotherapy, focusing on the scientific rationale for the use of oncolytic viruses, checkpoint inhibitors and their combination to combat melanomas. Attention is also given to the immunological aspects of cancer therapy and the shift from conventional therapy towards immunotherapy. This review brings together information on how immunotherapy can be applied to support other cancer therapies in order to maximize the efficacy of melanoma treatment and improve clinical outcomes. Abstract In this review, we discuss the use of oncolytic viruses and checkpoint inhibitors in cancer immunotherapy in melanoma, with a particular focus on combinatory therapies. Oncolytic viruses are promising and novel anti-cancer agents, currently under investigation in many clinical trials both as monotherapy and in combination with other therapeutics. They have shown the ability to exhibit synergistic anticancer activity with checkpoint inhibitors, chemotherapy, radiotherapy. A coupling between oncolytic viruses and checkpoint inhibitors is a well-accepted strategy for future cancer therapies. However, eradicating advanced cancers and tailoring the immune response for complete tumor clearance is an ongoing problem. Despite current advances in cancer research, monotherapy has shown limited efficacy against solid tumors. Therefore, current improvements in virus targeting, genetic modification, enhanced immunogenicity, improved oncolytic properties and combination strategies have a potential to widen the applications of immuno-oncology (IO) in cancer treatment. Here, we summarize the strategy of combinatory therapy with an oncolytic vector to combat melanoma and highlight the need to optimize current practices and improve clinical outcomes.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
- Clinical Science, Targovax Oy, Saukonpaadenranta 2, 00180 Helsinki, Finland
- Correspondence: (L.K.); (M.G.)
| | - Laura Bertinato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Monika Staniszewska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
- Correspondence: (L.K.); (M.G.)
| |
Collapse
|
15
|
Tanaka KI, Kawahara M. Carnosine and Lung Disease. Curr Med Chem 2020; 27:1714-1725. [PMID: 31309876 DOI: 10.2174/0929867326666190712140545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 05/24/2019] [Accepted: 06/28/2019] [Indexed: 12/22/2022]
Abstract
Carnosine (β-alanyl-L-histidine) is a small dipeptide with numerous activities, including antioxidant effects, metal ion chelation, proton buffering capacity, and inhibitory effects on protein carbonylation and glycation. Carnosine has been mostly studied in organs where it is abundant, including skeletal muscle, cerebral cortex, kidney, spleen, and plasma. Recently, the effect of supplementation with carnosine has been studied in organs with low levels of carnosine, such as the lung, in animal models of influenza virus or lipopolysaccharide-induced acute lung injury and pulmonary fibrosis. Among the known protective effects of carnosine, its antioxidant effect has attracted increasing attention for potential use in treating lung disease. In this review, we describe the in vitro and in vivo biological and physiological actions of carnosine. We also report our recent study and discuss the roles of carnosine or its related compounds in organs where carnosine is present in only small amounts (especially the lung) and its protective mechanisms.
Collapse
Affiliation(s)
- Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| |
Collapse
|
16
|
Chmielewska K, Dzierzbicka K, Inkielewicz-Stępniak I, Przybyłowska M. Therapeutic Potential of Carnosine and Its Derivatives in the Treatment of Human Diseases. Chem Res Toxicol 2020; 33:1561-1578. [PMID: 32202758 DOI: 10.1021/acs.chemrestox.0c00010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Despite significant progress in the pathogenesis, diagnosis, treatment, and prevention of cancer and neurodegenerative diseases, their occurrence and mortality are still high around the world. The resistance of cancer cells to the drugs remains a significant problem in oncology today, while in the case of neuro-degenerative diseases, therapies reversing the process are still yet to be found. Furthermore, it is important to seek new chemotherapeutics reversing side effects of currently used drugs or helping them perform their function to inhibit progression of the disease. Carnosine, a dipeptide constisting of β-alanine and l-histidine, has a variety of functions to mention: antioxidant, antiglycation, and reducing the toxicity of metal ions. It has therefore been proposed to act as a therapeutic agent for many pathological states. The aim of this paper was to find if carnosine and its derivatives can be helpful in treating various diseases. Literature search presented in this review includes review and original papers found in SciFinder, PubMed, and Google Scholar. Searches were based on substantial keywords concerning therapeutic usage of carnosine and its derivatives in several diseases including neurodegenerative disorders and cancer. In this paper, we review articles and find that carnosine and its derivatives are potential therapeutic agents in many diseases including cancer, neurodegenerative diseases, diabetes, and schizophrenia. Carnosine and its derivatives can be used in treating neurodegenerative diseases, cancer, diabetes, or schizophrenia, although their usage is limited. Therefore, there's an urge to synthesize and analyze new substances, overcoming the limitation of carnosine itself.
Collapse
Affiliation(s)
- Klaudia Chmielewska
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk 80-233, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk 80-233, Poland
| | - Iwona Inkielewicz-Stępniak
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Gdansk 80-211, Poland
| | - Maja Przybyłowska
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk 80-233, Poland
| |
Collapse
|
17
|
Garofalo M, Grazioso G, Cavalli A, Sgrignani J. How Computational Chemistry and Drug Delivery Techniques Can Support the Development of New Anticancer Drugs. Molecules 2020; 25:E1756. [PMID: 32290224 PMCID: PMC7180704 DOI: 10.3390/molecules25071756] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 01/17/2023] Open
Abstract
The early and late development of new anticancer drugs, small molecules or peptides can be slowed down by some issues such as poor selectivity for the target or poor ADME properties. Computer-aided drug design (CADD) and target drug delivery (TDD) techniques, although apparently far from each other, are two research fields that can give a significant contribution to overcome these problems. Their combination may provide mechanistic understanding resulting in a synergy that makes possible the rational design of novel anticancer based therapies. Herein, we aim to discuss selected applications, some also from our research experience, in the fields of anticancer small organic drugs and peptides.
Collapse
Affiliation(s)
- Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Giovanni Grazioso
- Department of Pharmaceutical Sciences, University of Milano, 20133 Milan, Italy
| | - Andrea Cavalli
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| |
Collapse
|
18
|
Wang J, Li Y, Li S, Yao W, Liu X, Zhu Y, Li W, Sun L, Jin N, Li X. Anti-tumor Synergistic Effect of a Dual Cancer-Specific Recombinant Adenovirus and Paclitaxel on Breast Cancer. Front Oncol 2020; 10:244. [PMID: 32269962 PMCID: PMC7109281 DOI: 10.3389/fonc.2020.00244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/13/2020] [Indexed: 12/24/2022] Open
Abstract
This study aimed at investigating the anticancer potential of the recombinant adenovirus Ad-apoptin-hTERTp-E1a (Ad-VT) and its synergistic combination with paclitaxel (PTX) in breast cancer treatment. First, we used the Calcusyn software to analyze the synergy between the Ad-VT and paclitaxel, and to determine the final drug concentration. Second, we used crystal violet staining and WST-1 assays to analyze the inhibitory effect of Ad-VT and paclitaxel combination treatment on MCF-7, MDA-MB-231, and MCF-10A cells. Subsequently, we used Hoechst, Annexin V, JC-1 staining to analyze the inhibition pathway of drugs on breast cancer cells. We also used Transwell assays to analyze the cell migration and invasion of MCF-7 and MDA-MB-231 cells. The pGL4.51 plasmid was used to transfect and to generate MDA-MB-231 cells, that stably express luciferase (MDA-MB-231-LUC). The in vivo tumor inhibition effect of Ad-VT and paclitaxel combination treatment was subsequently confirmed using a tumor-bearing nude mouse model. This combination treatment can increase the inhibition of breast cancer cells and reduce paclitaxel toxicity. Ad-VT had a strong apoptosis-inducing effect on MCF-7 and MDA-MB-231 cells, that was mainly mediated through the mitochondrial apoptotic pathway. The combination of Ad-VT and paclitaxel could significantly increase the inhibition of breast cancer cell migration and invasion. Combination of Ad-VT and paclitaxel can inhibit tumor growth and reduce toxicity in vivo. Ad-VT can also inhibit the growth of breast cancer cells and promote their apoptosis. Meanwhile, when it is combined with paclitaxel, Ad-VT could play a significant role in a synergistic tumor inhibition.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yiquan Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Shanzhi Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Wei Yao
- Center for Disease Control and Prevention, Agency for Offices Administration, Central Military Commission, Beijing, China
| | - Xing Liu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yilong Zhu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Wenjie Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Liankun Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Ningyi Jin
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xiao Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
19
|
Liu X, Yang Z, Li Y, Zhu Y, Li W, Li S, Wang J, Cui Y, Shang C, Liu Z, Song G, Li C, Li X, Shao G, Jin N. Chemovirotherapy of Lung Squamous Cell Carcinoma by Combining Oncolytic Adenovirus With Gemcitabine. Front Oncol 2020; 10:229. [PMID: 32158698 PMCID: PMC7052302 DOI: 10.3389/fonc.2020.00229] [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: 11/02/2019] [Accepted: 02/10/2020] [Indexed: 12/28/2022] Open
Abstract
Oncolytic virotherapy is emerging as an important agent in cancer treatment. In a previous study, we designed and constructed Ad-Apoptin-hTERTp-E1a (Ad-VT), a dual cancer-selective anti-tumor recombinant adenovirus. In this study, crystal violet staining and WST-1 assays showed that Ad-VT has a significant tumor killing effect in a time and dose dependent manner. The combination of Ad-VT (10 MOI) and gemcitabine (10 nM) significantly inhibited NCI-H226 cells, but did not increase the killing effect of gemcitabine on human normal bronchial epithelial cells BEAS-2B. Hoechst, JC-1 and Annexin V experiments demonstrated that the combination of Ad-VT and gemcitabine mainly inhibited NCI-H226 cell proliferation by inducing apoptosis (mitochondrial pathway). The combination also significantly inhibited the migration and invasion abilities of NCI-H226 cells. In vivo, Ad-VT in combination with low-dose gemcitabine could effectively inhibit tumor growth and prolong survival of mice. Ad-VT has the characteristics of tumor-selective replication and killing, in vitro and in vivo. The combined application of Ad-VT and gemcitabine has a synergistic effect, which can increase the anti-tumor effect and reduce the toxicity of chemotherapy drugs, indicating that Ad-VT has a potential clinical value in the treatment of lung squamous cell carcinoma.
Collapse
Affiliation(s)
- Xing Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Zhiguang Yang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Yiquan Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Yilong Zhu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Wenjie Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shanzhi Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yingli Cui
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Department of Oncology Gynecology, The First Hospital of Jilin University, Changchun, China
| | - Chao Shang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Zirui Liu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Gaojie Song
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Ce Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Xiao Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Guoguang Shao
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China
| | - Ningyi Jin
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
20
|
Bellier J, Nokin MJ, Caprasse M, Tiamiou A, Blomme A, Scheijen JL, Koopmansch B, MacKay GM, Chiavarina B, Costanza B, Rademaker G, Durieux F, Agirman F, Maloujahmoum N, Cusumano PG, Lovinfosse P, Leung HY, Lambert F, Bours V, Schalkwijk CG, Hustinx R, Peulen O, Castronovo V, Bellahcène A. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab. Cell Rep 2020; 30:1400-1416.e6. [PMID: 32023458 DOI: 10.1016/j.celrep.2020.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 11/10/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
The use of cetuximab anti-epidermal growth factor receptor (anti-EGFR) antibodies has opened the era of targeted and personalized therapy in colorectal cancer (CRC). Poor response rates have been unequivocally shown in mutant KRAS and are even observed in a majority of wild-type KRAS tumors. Therefore, patient selection based on mutational profiling remains problematic. We previously identified methylglyoxal (MGO), a by-product of glycolysis, as a metabolite promoting tumor growth and metastasis. Mutant KRAS cells under MGO stress show AKT-dependent survival when compared with wild-type KRAS isogenic CRC cells. MGO induces AKT activation through phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin 2 (mTORC2) and Hsp27 regulation. Importantly, the sole induction of MGO stress in sensitive wild-type KRAS cells renders them resistant to cetuximab. MGO scavengers inhibit AKT and resensitize KRAS-mutated CRC cells to cetuximab in vivo. This study establishes a link between MGO and AKT activation and pinpoints this oncometabolite as a potential target to tackle EGFR-targeted therapy resistance in CRC.
Collapse
Affiliation(s)
- Justine Bellier
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Marie-Julie Nokin
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Maurine Caprasse
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Assia Tiamiou
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Jean L Scheijen
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands
| | | | | | - Barbara Chiavarina
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Brunella Costanza
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Gilles Rademaker
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Florence Durieux
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Ferman Agirman
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Naïma Maloujahmoum
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Pino G Cusumano
- Department of Senology, Liège University Hospital, University of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Oncology Imaging Division, Liège University Hospital, University of Liège, Liège, Belgium
| | - Hing Y Leung
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom; Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Frédéric Lambert
- Department of Human Genetics, Liège University Hospital, Liege, Belgium
| | - Vincent Bours
- Department of Human Genetics, Liège University Hospital, Liege, Belgium
| | - Casper G Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands
| | - Roland Hustinx
- Oncology Imaging Division, Liège University Hospital, University of Liège, Liège, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Liège, Belgium.
| |
Collapse
|
21
|
Kuryk L, Møller ASW. Chimeric oncolytic Ad5/3 virus replicates and lyses ovarian cancer cells through desmoglein-2 cell entry receptor. J Med Virol 2020; 92:1309-1315. [PMID: 31944306 PMCID: PMC7496614 DOI: 10.1002/jmv.25677] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/05/2020] [Indexed: 12/30/2022]
Abstract
Despite new therapies, the estimated 229 875 women living with ovarian cancer have a 5‐year survival rate of 47.6%. This cavity‐localized cancer lends itself to local administration of modalities, such as the oncolytic adenovirus (Ad) Ad5/3‐D24‐granulocyte‐macrophage colony‐stimulating factor virus (ONCOS‐102). Its repeated administration to a patient with chemotherapy‐refractory ovarian cancer induced CD8+ antitumor immune responses with the overall survival reaching 40 months. Here we probe the dominant receptor used by ONCOS‐102 in four established epithelial ovarian cancer cell lines. Ad3 can use the desmoglein‐2 (DSG2) and CD46 receptors on susceptible cells. DSG2 was nearly absent in A2780 cells but was expressed in more than 90% of OAW42, OVCAR3, and OV‐90 cells. After 96 hours, ONCOS‐102 treatment showed significant oncolytic activity (≧50%) in OAW42, OVCAR3, and OV‐90 cells, but minimal activity in A2780 cells, suggesting DSG2 as the dominant receptor for ONCOS‐102. Furthermore, retrospective analyses of phase I clinical trial of ONCOS‐102 treatment of 12 patients with varied tumors indicated a correlation between viral genomes in blood and DSG2 RNA expression. These data support the role of DSG2 expression on cancer cells in virus infectivity and the continued development of ONCOS‐102 for ovarian cancer treatment. These data support the role of DSG2 expression on cancer cells in virus infectivity and the continued development of ONCOS‐102 for ovarian cancer treatment.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Clinical Science, Targovax Oy, Helsinki, Finland.,Department of Virology, National Institute of Public Health - National Institute of Hygiene, Warsaw, Poland
| | | |
Collapse
|
22
|
Garofalo M, Villa A, Crescenti D, Marzagalli M, Kuryk L, Limonta P, Mazzaferro V, Ciana P. Heterologous and cross-species tropism of cancer-derived extracellular vesicles. Theranostics 2019; 9:5681-5693. [PMID: 31534511 PMCID: PMC6735396 DOI: 10.7150/thno.34824] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring cargo delivery vesicles that have recently received considerable attention for their roles in intercellular communication in many physiological and pathological processes, including tumourigenesis. EVs generated by different tissues demonstrated specific homing: in particular, cancer-derived EVs showed a selective tropism for the tumor tissue from which the vesicles originated. For this property, EVs have been proposed as drug delivery tools for anti-cancer therapies, although the limited knowledge about their in vivo tropism hinders their therapeutic applications. The current study aimed to characterize the targeting properties of cancer-derived EVs in vitro and their biodistribution in vivo, by using an imaging approach. Methods: EVs were generated from: i) murine lung (LL/2) and colon (MC-38) cancer lines, ii) human lung cancer cell line (A549) and iii) human liver biopsy samples from healthy individuals. EVs were loaded with fluorescent dyes alone or in combination with a biopharmaceutical agent, the oncolytic adenovirus (OV), characterized for charge and size and tested for their activity in cancer cell lines. Finally, optical imaging was extensively applied to study in vivo and ex vivo the biodistribution of EVs originated from different sources in different mouse models of cancer, including xenograft, syngeneic graft and the MMTV-NeuT genetically modified animal. Results: We initially demonstrated that even loading EVs even with a large biopharmaceutical oncolytic viruses (OVs) did not significantly change their charge and dimension properties, while increasing their anti-neoplastic activity compared to the virus or EVs alone. Interestingly, this activity was observed even if the EVs derived from lung cancer were applied to colon carcinoma cell lines and vice versa, suggesting that the EV uptake occurred in vitro without any specificity for the cancer cells from which the vesicles originated. When administered i.v (intravenously) to the mouse models of cancer, the tumour-derived EVs, but not the EVs derived from a healthy tissue, demonstrated a selective accumulation of the fluorescence at the tumour site 24 h after injection; adding OVs to the formulation did not change the tumour-specific tropism of the EVs also in vivo. Most interestingly, the in vivo experiments confirmed the in vitro observation of the generalized tropism of tumour-derived EVs for any neoplastic tissue, independent of the tumour type or even the species originating the vesicles. Conclusions: Taken together, our in vitro and in vivo data demonstrate for the first time a heterologous, cross-species tumour-tropism for cancer-derived EVs. This finding challenges our current view on the homing properties of EVs and opens new avenues for the selective delivery of diagnostic/therapeutic agents to solid tumours.
Collapse
Affiliation(s)
- Mariangela Garofalo
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Alessandro Villa
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Daniela Crescenti
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Lukasz Kuryk
- Targovax Oy, Clinical Science, Helsinki, Finland
- National Institute of Public Health - National Institute of Hygiene, Department of Virology, Warsaw, Poland
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Vincenzo Mazzaferro
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
- Istituto Nazionale Tumori Fondazione IRCCS, Milan, Italy
| | - Paolo Ciana
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| |
Collapse
|
23
|
Kuryk L, Møller ASW, Jaderberg M. Abscopal effect when combining oncolytic adenovirus and checkpoint inhibitor in a humanized NOG mouse model of melanoma. J Med Virol 2019; 91:1702-1706. [PMID: 31081549 PMCID: PMC6771875 DOI: 10.1002/jmv.25501] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022]
Abstract
Melanoma, an immunogenic tumor, is the first indication where oncolytic viruses are now becoming part of clinical practice. ONCOS‐102, a transgened adenovirus, has shown to act as a primer of relevant tumor targeting immune cells both in preclinical and clinical melanoma studies. Strategies to augment its effectiveness warrant investigation. Combination therapy of ONCOS‐102 with the checkpoint inhibitor (CPI) pembrolizumab was evaluated in a quasi‐human animal model, the humanized NOG mouse model. A dosing schedule of the combination, beginning the CPI concurrently with the oncolytic viral therapy and continuing the CPI treatment, appeared to induce an abscopal effect in untreated tumor lesions. Concurrent combination therapy with checkpoint inhibitors may improve the induction of antitumor immune responses of ONCOS‐102. The combinatory therapy of ONCOS‐102 and CPI, appeared to induce an abscopal effect in untreated tumor lesions. The data support the development of ONCOS‐102 with checkpoint inhibitors for the treatment of malignant cancer diseases.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Targovax Oy, Clinical Science, Helsinki, Finland.,Department of Virology, National Institute of Public Health - National Institute of Hygiene, Warsaw, Poland
| | | | | |
Collapse
|
24
|
Guerrero-Fonseca CA, López-Baquero MA, Bedoya-Rodríguez AA. Virus oncolíticos: un arma contra el cáncer. REVISTA DE LA FACULTAD DE MEDICINA 2019. [DOI: 10.15446/revfacmed.v67n2.68347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introducción. Los virus oncolíticos son virus atenuados, mutados o que por naturaleza se dirigen y matan específicamente células tumorales, sin afectar a las células normales. La administración intratumoral del virus ofrece la oportunidad de tratar el tumor primario pero no focos metastásicos, los cuales pueden ser alcanzados mediante la administración intravenosa. Sin embargo, su eficiencia puede disminuir por la presencia de una respuesta inmunológica preexistente en los sujetos tratados.Objetivo. Exponer las técnicas utilizadas para envolver y transportar los virus con el fin de eludir el sistema inmunológico antes de que el virus llegue al tumor.Materiales y métodos. Se realizó una búsqueda narrativa de la literatura original y de revisión en las bases de datos PubMed, JSTOR y EBSCO sobre métodos o técnicas utilizadas para el tratamiento del cáncer mediante el uso de virus oncolíticos.Resultados. La formación de nanocomplejos entre los virus oncolíticos y biopolímeros —ya sea mediante la unión química o mediante la unión a través de interacciones electrostáticas o el uso de micropartículas, células transportadoras, liposomas, ultrasonido o terapias combinadas– es eficaz para evitar la respuesta inmunológica del huésped contra el virus.Conclusión. Para evitar la respuesta inmunológica del huésped contra los virus oncolíticos se han desarrollo diversos métodos que permiten la liberación controlada y especifica de los mismos. Sin embargo, debido a la diversidad de los virus, se debe tener en cuenta que la eficacia de los métodos de protección y transporte depende de las características bioquímicas tanto del biomaterial como del virus.
Collapse
|
25
|
Bellier J, Nokin MJ, Lardé E, Karoyan P, Peulen O, Castronovo V, Bellahcène A. Methylglyoxal, a potent inducer of AGEs, connects between diabetes and cancer. Diabetes Res Clin Pract 2019; 148:200-211. [PMID: 30664892 DOI: 10.1016/j.diabres.2019.01.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/04/2019] [Indexed: 02/08/2023]
Abstract
Diabetes is one of the most frequent diseases throughout the world and its incidence is predicted to exponentially progress in the future. This metabolic disorder is associated with major complications such as neuropathy, retinopathy, atherosclerosis, and diabetic nephropathy, the severity of which correlates with hyperglycemia, suggesting that they are triggered by high glucose condition. Reducing sugars and reactive carbonyl species such as methylglyoxal (MGO) lead to glycation of proteins, lipids and DNA and the gradual accumulation of advanced glycation end products (AGEs) in cells and tissues. While AGEs are clearly implicated in the pathogenesis of diabetes complications, their potential involvement during malignant tumor development, progression and resistance to therapy is an emerging concept. Meta-analysis studies established that patients with diabetes are at higher risk of developing cancer and show a higher mortality rate than cancer patients free of diabetes. In this review, we highlight the potential connection between hyperglycemia-associated AGEs formation on the one hand and the recent evidence of pro-tumoral effects of MGO stress on the other hand. We also discuss the marked interest in anti-glycation compounds in view of their strategic use to treat diabetic complications but also to protect against augmented cancer risk in patients with diabetes.
Collapse
Affiliation(s)
- Justine Bellier
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Belgium
| | - Marie-Julie Nokin
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Belgium
| | - Eva Lardé
- Laboratoire des Biomolécules, UMR 7203, Sorbonne Université, Paris, France
| | - Philippe Karoyan
- Laboratoire des Biomolécules, UMR 7203, Sorbonne Université, Paris, France
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Belgium.
| |
Collapse
|
26
|
Optimization of Early Steps in Oncolytic Adenovirus ONCOS-401 Production in T-175 and HYPERFlasks. Int J Mol Sci 2019; 20:ijms20030621. [PMID: 30709038 PMCID: PMC6387112 DOI: 10.3390/ijms20030621] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Oncolytic adenoviruses can trigger lysis of tumor cells, induce an antitumor immune response, bypass classical chemotherapeutic resistance strategies of tumors, and provide opportunities for combination strategies. A major challenge is the development of scalable production methods for viral seed stocks and sufficient quantities of clinical grade viruses. Because of promising clinical signals in a compassionate use program (Advanced Therapy Access Program) which supported further development, we chose the oncolytic adenovirus ONCOS-401 as a testbed for a new approach to scale up. We found that the best viral production conditions in both T-175 flasks and HYPERFlasks included A549 cells grown to 220,000 cells/cm² (80% confluency), with ONCOS-401 infection at 30 multiplicity of infection (MOI), and an incubation period of 66 h. The Lysis A harvesting method with benzonase provided the highest viral yield from both T-175 and HYPERFlasks (10,887 ± 100 and 14,559 ± 802 infectious viral particles/cell, respectively). T-175 flasks and HYPERFlasks produced up to 2.1 × 10⁸ ± 0.2 and 1.75 × 10⁸ ± 0.08 infectious particles of ONCOS-401 per cm² of surface area, respectively. Our findings suggest a suitable stepwise process that can be applied to optimizing the initial production of other oncolytic viruses.
Collapse
|
27
|
Extracellular vesicles enhance the targeted delivery of immunogenic oncolytic adenovirus and paclitaxel in immunocompetent mice. J Control Release 2019; 294:165-175. [DOI: 10.1016/j.jconrel.2018.12.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/30/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
|
28
|
Goradel NH, Mohajel N, Malekshahi ZV, Jahangiri S, Najafi M, Farhood B, Mortezaee K, Negahdari B, Arashkia A. Oncolytic adenovirus: A tool for cancer therapy in combination with other therapeutic approaches. J Cell Physiol 2018; 234:8636-8646. [PMID: 30515798 DOI: 10.1002/jcp.27850] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/08/2018] [Indexed: 12/17/2022]
Abstract
Cancer therapy using oncolytic viruses is an emerging area, in which viruses are engineered to selectively propagate in tumor tissues without affecting healthy cells. Because of the advantages that adenoviruses (Ads) have over other viruses, they are more considered. To achieve tumor selectivity, two main modifications on Ads genome have been applied: small deletions and insertion of tissue- or tumor-specific promoters. Despite oncolytic adenoviruses ability in tumor cell lysis and immune responses stimulation, to further increase their antitumor effects, genomic modifications have been carried out including insertion of checkpoint inhibitors and antigenic or immunostimulatory molecules into the adenovirus genome and combination with dendritic cells and chemotherapeutic agents. This study reviews oncolytic adenoviruses structures, their antitumor efficacy in combination with other therapeutic strategies, and finally challenges around this treatment approach.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasir Mohajel
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Jahangiri
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
29
|
NF-κB Signaling in Targeting Tumor Cells by Oncolytic Viruses-Therapeutic Perspectives. Cancers (Basel) 2018; 10:cancers10110426. [PMID: 30413032 PMCID: PMC6265863 DOI: 10.3390/cancers10110426] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/04/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, oncolytic virotherapy became a promising therapeutic approach, leading to the introduction of a novel generation of anticancer drugs. However, despite evoking an antitumor response, introducing an oncolytic virus (OV) to the patient is still inefficient to overcome both tumor protective mechanisms and the limitation of viral replication by the host. In cancer treatment, nuclear factor (NF)-κB has been extensively studied among important therapeutic targets. The pleiotropic nature of NF-κB transcription factor includes its involvement in immunity and tumorigenesis. Therefore, in many types of cancer, aberrant activation of NF-κB can be observed. At the same time, the activity of NF-κB can be modified by OVs, which trigger an immune response and modulate NF-κB signaling. Due to the limitation of a monotherapy exploiting OVs only, the antitumor effect can be enhanced by combining OV with NF-κB-modulating drugs. This review describes the influence of OVs on NF-κB activation in tumor cells showing NF-κB signaling as an important aspect, which should be taken into consideration when targeting tumor cells by OVs.
Collapse
|
30
|
Kuryk L, Møller ASW, Jaderberg M. Combination of immunogenic oncolytic adenovirus ONCOS-102 with anti-PD-1 pembrolizumab exhibits synergistic antitumor effect in humanized A2058 melanoma huNOG mouse model. Oncoimmunology 2018; 8:e1532763. [PMID: 30713786 PMCID: PMC6343802 DOI: 10.1080/2162402x.2018.1532763] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 01/15/2023] Open
Abstract
Malignant melanoma is an aggressive type of skin cancer whose incidence is increasing globally. Although surgery is effective in early stage melanoma, patients with advanced melanoma only have a 20% 5-year survival rate. Hence, combinations of existing and new immunotherapy technologies and immunotherapeutic agents are being evaluated. ONCOS-102 is an oncolytic adenovirus armed with human GM-CSF and an Ad5/3 chimeric capsid. It has shown to be well tolerated in phase I study (NCT01598129) wherein it induced antitumor immunity, infiltration of CD8 + T cells to tumors, and up-regulation of PD-L1. We propose that ONCOS-102 could serve as an immunosensitizer in combination therapies with checkpoint inhibitors. In this preclinical study, we investigated the cytotoxicity of ONCOS-102 and pembrolizumab, an anti-PD-1 antibody, in four human melanoma cell lines, A375, A2058, SK-Mel-2 and SK-Mel-28. Humanized mice engrafted with A2058 melanoma cells showed significant tumor volume reduction after ONCOS-102 treatment. Combination of pembrolizumab with ONCOS-102 reduced tumor volume to an even greater extent, while pembrolizumab (200 µg, or 400 µg) did not show any therapeutic benefit by itself. Body weight loss, and metastasis were not significantly affected by any treatment. These data support the scientific rationale for the ongoing clinical study of combination therapy of ONCOS-102 and pembrolizumab for the treatment of melanoma (NCT03003676).
Collapse
Affiliation(s)
- Lukasz Kuryk
- Clinical Science, Targovax Oy, Helsinki, Finland.,Department of Virology, National Institute of Public Health - National Institute of Hygiene, Warsaw, Poland
| | | | | |
Collapse
|
31
|
Pol JG, Lévesque S, Workenhe ST, Gujar S, Le Boeuf F, Clements DR, Fahrner JE, Fend L, Bell JC, Mossman KL, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Oncolytic viro-immunotherapy of hematologic and solid tumors. Oncoimmunology 2018; 7:e1503032. [PMID: 30524901 PMCID: PMC6279343 DOI: 10.1080/2162402x.2018.1503032] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/15/2018] [Indexed: 02/08/2023] Open
Abstract
Oncolytic viruses selectively target and kill cancer cells in an immunogenic fashion, thus supporting the establishment of therapeutically relevant tumor-specific immune responses. In 2015, the US Food and Drug Administration (FDA) approved the oncolytic herpes simplex virus T-VEC for use in advanced melanoma patients. Since then, a plethora of trials has been initiated to assess the safety and efficacy of multiple oncolytic viruses in patients affected with various malignancies. Here, we summarize recent preclinical and clinical progress in the field of oncolytic virotherapy.
Collapse
Affiliation(s)
- Jonathan G. Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Sarah Lévesque
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Samuel T. Workenhe
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, NS, Canada
- Department of Biology, Dalhousie University, NS, Canada
- Centre for Innovative and Collaborative Health Sciences Research, Quality and System Performance, IWK Health Centre, Halifax, NS, Canada
| | - Fabrice Le Boeuf
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | - Jean-Eudes Fahrner
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Transgene S.A., Illkirch-Graffenstaden, France
| | | | - John C. Bell
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Karen L. Mossman
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
32
|
Kuryk L, Møller ASW, Garofalo M, Cerullo V, Pesonen S, Alemany R, Jaderberg M. Antitumor-specific T-cell responses induced by oncolytic adenovirus ONCOS-102 (AdV5/3-D24-GM-CSF) in peritoneal mesothelioma mouse model. J Med Virol 2018; 90:1669-1673. [PMID: 29797583 PMCID: PMC6120454 DOI: 10.1002/jmv.25229] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/16/2018] [Indexed: 12/12/2022]
Abstract
Oncolytic adenoviral immunotherapy activates the innate immune system with subsequent induction of adaptive tumor‐specific immune responses to fight cancer. Hence, oncolytic viruses do not only eradicate cancer cells by direct lysis, but also generate antitumor immune response, allowing for long‐lasting cancer control and tumor reduction. Their therapeutic effect can be further enhanced by arming the oncolytic adenovirus with costimulatory transgenes and/or coadministration with other antitumor therapies. ONCOS‐102 has already been found to be well tolerated and efficacious against some types of treatment‐refractory tumors, including mesothelin‐positive ovarian cancer (NCT01598129). It induced local and systemic CD8+ T‐cell immunity and upregulated programmed death ligand 1. These results strongly advocate the use of ONCOS‐102 in combination with other therapeutic strategies in advanced and refractory tumors, especially those expressing the mesothelin antigen. The in vivo work presented herein describes the ability of the oncolytic adenovirus ONCOS‐102 to induce mesothelin‐specific T‐cells after the administration of the virus in bagg albino (BALB/c) mice with mesothelin‐positive tumors. We also demonstrate the effectiveness of the interferon‐γ the enzyme‐linked immunospot (ELISPOT) assay to detect the induction of T‐cells recognizing mesothelin, hexon, and E1A antigens in ONCOS‐102‐treated mesothelioma‐bearing BALB/c mice. Thus, the ELISPOT assay could be useful to monitor the progress of therapy with ONCOS‐102.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Clinical Science, Targovax Oy, Helsinki, Finland.,Department of Virology, National Institute of Public Health-National Institute of Hygiene, Warsaw, Poland.,Drug Research Program, ImmunoVirothearpy Lab, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Mariangela Garofalo
- Drug Research Program, ImmunoVirothearpy Lab, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Drug Research Program, ImmunoVirothearpy Lab, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Sari Pesonen
- Department of Clinical Science, Targovax Oy, Helsinki, Finland
| | - Ramon Alemany
- Catalan Institute of Oncology, IDIBELL, Barcelona, Spain
| | | |
Collapse
|
33
|
Garofalo M, Saari H, Somersalo P, Crescenti D, Kuryk L, Aksela L, Capasso C, Madetoja M, Koskinen K, Oksanen T, Mäkitie A, Jalasvuori M, Cerullo V, Ciana P, Yliperttula M. Antitumor effect of oncolytic virus and paclitaxel encapsulated in extracellular vesicles for lung cancer treatment. J Control Release 2018; 283:223-234. [PMID: 29864473 DOI: 10.1016/j.jconrel.2018.05.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023]
Abstract
Standard of care for cancer is commonly a combination of surgery with radiotherapy or chemoradiotherapy. However, in some advanced cancer patients this approach might still remaininefficient and may cause many side effects, including severe complications and even death. Oncolytic viruses exhibit different anti-cancer mechanisms compared with conventional therapies, allowing the possibility for improved effect in cancer therapy. Chemotherapeutics combined with oncolytic viruses exhibit stronger cytotoxic responses and oncolysis. Here, we have investigated the systemic delivery of the oncolytic adenovirus and paclitaxel encapsulated in extracellular vesicles (EV) formulation that, in vitro, significantly increased the transduction ratio and the infectious titer when compared with the virus and paclitaxel alone. We demonstrated that the obtained EV formulation reduced the in vivo tumor growth in animal xenograft model of human lung cancer. Indeed, we found that combined treatment of oncolytic adenovirus and paclitaxel encapsulated in EV has enhanced anticancer effects both in vitro and in vivo in lung cancer models. Transcriptomic comparison carried out on the explanted xenografts from the different treatment groups revealed that only 5.3% of the differentially expressed genes were overlapping indicating that a de novo genetic program is triggered by the presence of the encapsulated paclitaxel: this novel genetic program might be responsible of the observed enhanced antitumor effect. Our work provides a promising approach combining anticancer drugs and viral therapies by intravenous EV delivery as a strategy for the lung cancer treatment.
Collapse
Affiliation(s)
- M Garofalo
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland; Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, Milan 20133, Italy.
| | - H Saari
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland
| | - P Somersalo
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland; Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, Milan 20133, Italy
| | - D Crescenti
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, Milan 20133, Italy
| | - L Kuryk
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland; National Institute of Public Health - National Institute of Hygiene, Department of Virology, 24 Chocimska str, 00-791 Warsaw, Poland; Targovax Oy, R&D, Clinical Science, R&D, Saukonpaadenranta 2, 00180 Helsinki, Finland
| | - L Aksela
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland
| | - C Capasso
- Laboratory of ImmunoViroTherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland
| | - M Madetoja
- Made Consulting, Tykistökatu 4 B, FI-20520 Turku, Finland
| | - K Koskinen
- Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Survontie 9C, 40500, Finland
| | - T Oksanen
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland
| | - A Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, P.O.Box 263, FI_00029 HUS, Helsinki, Finland
| | - M Jalasvuori
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland; Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Survontie 9C, 40500, Finland
| | - V Cerullo
- Laboratory of ImmunoViroTherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland
| | - P Ciana
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, Milan 20133, Italy
| | - M Yliperttula
- Division of Pharmaceutical Biosciences and Centre for Drug Research, University of Helsinki, Viikinkaari 5, Helsinki 00790, Finland.
| |
Collapse
|
34
|
Pang T, Wang X, Gao J, Chen W, Shen XJ, Nie MM, Luo T, Yin K, Fang G, Wang KX, Xue XC. Fiber-modified hexon-chimeric oncolytic adenovirus targeting cancer associated fibroblasts inhibits tumor growth in gastric carcinoma. Oncotarget 2017; 8:76468-76478. [PMID: 29100326 PMCID: PMC5652720 DOI: 10.18632/oncotarget.20273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 07/11/2017] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE To evaluate the effects of fiber-modified hexon-chimeric recombinant oncolytic adenovirus targeting cancer associated fibroblasts (CAFs) on the gastric CAFs and the transplantation tumor mice model of gastric carcinoma (GC). RESULTS Compared with BJ cells and GPFs, the reproduction and infectivity of P9, P9-4C or GP adenoviruses were markedly higher in gastric CAFs. In addition, P9, P9-4C or GP had a significantly relatively more killing effect on gastric CAFs compared with GPFs, and have less oncolytic effect in BJ cells. Furthermore, in transplantation tumor mice model of GC we found significantly higher hexon protein expression in tumor tissues, more decreasing tumor growth and increasing inhibitory rates after treatment of P9, P9-4C or GP adenoviruses compared with Ad adenovirus. MATERIALS AND METHODS Based on the construction of the recombinant oncolytic adenoviruses pRCAdHVR48-SDF1p-Ad/EGFP (Ad, as control) with the E1A gene transcription regulated by stromal-derived factor 1 (SDF1) promoter and the hexon replaced by hexon-chimeric (H5HVR48) gene, three fiber-modified hexon-chimeric oncolytic adenovirus through the modification fiber protein by insertion of different short peptides specifically binding to fibroblast activation protein (FAP), including pRCAdHVR48-SDF1p-FAP-P9/EGFP (P9), pRCAdHVR48-SDF1p-FAP-P9-4C/EGFP (P9-4C), pRCAdHVR48-SDF1p-FAP-GP/EGFP (GP), and their corresponding replication-defective adenovirus in parallel were reconstructed. Then the reproduction, infectivity and killing ability of the four above recombinant adenoviruses were evaluated in gastric CAFs compared with gastric para-mucosa fibroblasts (GPFs) and neonatal human foreskin fibroblasts (BJ). Furthermore, transplantation tumor mice model of GC was established, and then treated by the four above recombinant adenoviruses. Tumor size and tumor growth inhibitory rates were calculated, and histomorphology by HE staining and hexon expressions by immunohistochemistry were evaluated in tumor tissues. CONCLUSIONS The fiber-modified hexon-chimeric recombinant oncolytic adenovirus targeting CAFs can relatively specifically kill gastric CAFs and inhibit GC cells growth in vivo.
Collapse
Affiliation(s)
- Tao Pang
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Xinghua Wang
- Department of Microbiology, Second Military Medical University, ShangHai, China
| | - Jun Gao
- Department of Gastroenterology, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Wei Chen
- Department of Cardiology, ChangZheng Hospital, Second Military Medical University, ShangHai, China
| | - Xiao Jun Shen
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Ming Ming Nie
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Tianhang Luo
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Guoen Fang
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Kai Xuan Wang
- Department of Gastroenterology, ChangHai Hospital, Second Military Medical University, ShangHai, China
| | - Xu Chao Xue
- Department of Gastrointestinal Surgery, ChangHai Hospital, Second Military Medical University, ShangHai, China
| |
Collapse
|
35
|
Howells A, Marelli G, Lemoine NR, Wang Y. Oncolytic Viruses-Interaction of Virus and Tumor Cells in the Battle to Eliminate Cancer. Front Oncol 2017; 7:195. [PMID: 28944214 PMCID: PMC5596080 DOI: 10.3389/fonc.2017.00195] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022] Open
Abstract
Oncolytic viruses (OVs) are an emerging treatment option for many cancer types and have recently been the focus of extensive research aiming to develop their therapeutic potential. The ultimate aim is to design a virus which can effectively replicate within the host, specifically target and lyse tumor cells and induce robust, long lasting tumor-specific immunity. There are a number of viruses which are either naturally tumor-selective or can be modified to specifically target and eliminate tumor cells. This means they are able to infect only tumor cells and healthy tissue remains unharmed. This specificity is imperative in order to reduce the side effects of oncolytic virotherapy. These viruses can also be modified by various methods including insertion and deletion of specific genes with the aim of improving their efficacy and safety profiles. In this review, we have provided an overview of the various virus species currently being investigated for their oncolytic potential and the positive and negative effects of a multitude of modifications used to increase their infectivity, anti-tumor immunity, and treatment safety, in particular focusing on the interaction of tumor cells and OVs.
Collapse
Affiliation(s)
- Anwen Howells
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Giulia Marelli
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Kuryk L, Haavisto E, Garofalo M, Capasso C, Hirvinen M, Pesonen S, Ranki T, Vassilev L, Cerullo V. Synergistic anti-tumor efficacy of immunogenic adenovirus ONCOS-102 (Ad5/3-D24-GM-CSF) and standard of care chemotherapy in preclinical mesothelioma model. Int J Cancer 2016; 139:1883-93. [PMID: 27287512 DOI: 10.1002/ijc.30228] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/01/2016] [Accepted: 05/30/2016] [Indexed: 11/08/2022]
Abstract
Malignant mesothelioma (MM) is a rare cancer type caused mainly by asbestos exposure. The median overall survival time of a mesothelioma cancer patient is less than 1-year from diagnosis. Currently there are no curative treatment modalities for malignant mesothelioma, however treatments such as surgery, chemotherapy and radiotherapy can help to improve patient prognosis and increase life expectancy. Pemetrexed-Cisplatin is the only standard of care (SoC) chemotherapy for malignant mesothelioma, but the median PFS/OS (progression-free survival/overall survival) from the initiation of treatment is only up to 12 months. Therefore, new treatment strategies against malignant mesothelioma are in high demand. ONCOS-102 is a dual targeting, chimeric oncolytic adenovirus, coding for human GM-CSF. The safety and immune activating properties of ONCOS-102 have already been assessed in phase 1 study (NCT01598129). In this preclinical study, we evaluated the antineoplastic activity of combination treatment with SoC chemotherapy (Pemetrexed, Cisplatin, Carboplatin) and ONCOS-102 in xenograft BALB/c model of human malignant mesothelioma. We demonstrated that ONCOS-102 is able to induce immunogenic cell death of human mesothelioma cell lines in vitro and showed anti-tumor activity in the treatment of refractory H226 malignant pleural mesothelioma (MPM) xenograft model. While chemotherapy alone showed no anti-tumor activity in the mesothelioma mouse model, ONCOS-102 was able to slow down tumor growth. Interestingly, a synergistic anti-tumor effect was seen when ONCOS-102 was combined with chemotherapy regimens. These findings give a rationale for the clinical testing of ONCOS-102 in combination with first-line chemotherapy in patients suffering from malignant mesothelioma.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Targovax Oy, Saukonpaadenranta 2, Helsinki, Finland.,Laboratory of ImmunoViroTherapy, Division of Pharmaceutical Biosciences and Centre for Drug Research (CDR), University of Helsinki, Viikinkaari 5, Helsinki, 00790, Finland.,Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24 Str, Warsaw, 00-791, Poland
| | | | - Mariangela Garofalo
- Laboratory of ImmunoViroTherapy, Division of Pharmaceutical Biosciences and Centre for Drug Research (CDR), University of Helsinki, Viikinkaari 5, Helsinki, 00790, Finland
| | - Cristian Capasso
- Laboratory of ImmunoViroTherapy, Division of Pharmaceutical Biosciences and Centre for Drug Research (CDR), University of Helsinki, Viikinkaari 5, Helsinki, 00790, Finland
| | - Mari Hirvinen
- Laboratory of ImmunoViroTherapy, Division of Pharmaceutical Biosciences and Centre for Drug Research (CDR), University of Helsinki, Viikinkaari 5, Helsinki, 00790, Finland
| | - Sari Pesonen
- Targovax Oy, Saukonpaadenranta 2, Helsinki, Finland
| | - Tuuli Ranki
- Targovax Oy, Saukonpaadenranta 2, Helsinki, Finland
| | - Lotta Vassilev
- Oncos Therapeutics Oy, Saukonpaadenranta 2, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of ImmunoViroTherapy, Division of Pharmaceutical Biosciences and Centre for Drug Research (CDR), University of Helsinki, Viikinkaari 5, Helsinki, 00790, Finland
| |
Collapse
|
37
|
Accardo A, Del Zoppo L, Morelli G, Condorelli DF, Barresi V, Musso N, Spampinato G, Bellia F, Tabbì G, Rizzarelli E. Liposome antibody–ionophore conjugate antiproliferative activity increases by cellular metallostasis alteration. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00461j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Carnosine derivative containing liposomes functionalized with the Fab' fragment of Trastuzumab were synthesized.
Collapse
|