1
|
Repositioning of Old Drugs for Novel Cancer Therapies: Continuous Therapeutic Perfusion of Aspirin and Oseltamivir Phosphate with Gemcitabine Treatment Disables Tumor Progression, Chemoresistance, and Metastases. Cancers (Basel) 2022; 14:cancers14153595. [PMID: 35892853 PMCID: PMC9331689 DOI: 10.3390/cancers14153595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Repositioning old drugs in combination with clinical standard chemotherapeutics opens a promising clinical treatment approach for patients with pancreatic cancer. This report presents a therapeutic repositioning of continuous perfusion of aspirin and oseltamivir phosphate in combination with gemcitabine treatment as an effective treatment option for pancreatic cancer. The data suggest that repositioning these drugs with continuous perfusion with gemcitabine disables chemoresistance, tumor progression, EMT program, cancer stem cells, and metastases in a preclinical mouse model of human pancreatic cancer. These promising results warrant additional investigation to assess the potential of translating into the clinical setting to improve the cancer patient prognosis for an otherwise fatal disease. Abstract Metastatic pancreatic cancer has an invariably fatal outcome, with an estimated median progression-free survival of approximately six months employing our best combination chemotherapeutic regimens. Once drug resistance develops, manifested by increased primary tumor size and new and growing metastases, patients often die rapidly from their disease. Emerging evidence indicates that chemotherapy may contribute to the development of drug resistance through the upregulation of epithelial–mesenchymal transition (EMT) pathways and subsequent cancer stem cell (CSC) enrichment. Neuraminidase-1 (Neu-1) regulates the activation of several receptor tyrosine kinases implicated in EMT induction, angiogenesis, and cellular proliferation. Here, continuous therapeutic targeting of Neu-1 using parenteral perfusion of oseltamivir phosphate (OP) and aspirin (ASA) with gemcitabine (GEM) treatment significantly disrupts tumor progression, critical compensatory signaling mechanisms, EMT program, CSC, and metastases in a preclinical mouse model of human pancreatic cancer. ASA- and OP-treated xenotumors significantly inhibited the metastatic potential when transferred into animals.
Collapse
|
3
|
Izzo F, Ionna F, Granata V, Albino V, Patrone R, Longo F, Guida A, Delrio P, Rega D, Scala D, Pezzuto R, Fusco R, Di Bernardo E, D’Alessio V, Grassi R, Contartese D, Palaia R. New Deployable Expandable Electrodes in the Electroporation Treatment in a Pig Model: A Feasibility and Usability Preliminary Study. Cancers (Basel) 2020; 12:cancers12020515. [PMID: 32102182 PMCID: PMC7072261 DOI: 10.3390/cancers12020515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023] Open
Abstract
The aim of the study is to evaluate the usability aspects of new deployable, expandable, electrode prototypes, in terms of suitability solutions for laparoscopic applications on the liver, endoscopic trans-oral and trans-anal procedures, electroporation segmentation in several steps, mechanical functionality (flexibility, penetrability), visibility of the electrode under instrumental guidance, compatibility of the electrode with laparoscopic/endoscopic accesses, surgical instruments, and procedural room and safety compatibility. The electroporation was performed on an animal model (Sus Scrofa Large White 60 kg) both in laparoscopy and endoscopy, under ultrasound guidance, and in open surgery. Electrodes without divergence, with needles coming out straight, parallel to each other, and electrodes with peripheral needles (four needles), diverging from the electrode shaft axis (electrode with non-zero divergence) have been tested. To cause an evaluable necrosis effect, the number of electrical pulses was increased to induce immediate liver cell death. Histological samples were analyzed by staining with Haematoxylin/Eosin or by immunohistochemical staining to confirm complete necrosis. The prototypes of expandable electrodes, tested in laparoscopy and endoscopy and in open surgery, respectively, are suitable in terms of usability, electroporation segmentation in several steps, mechanical functionality (flexibility, penetrability), visibility under instrumental guidance, compatibility with laparoscopic/endoscopic accesses, surgical instruments and procedural room safety, patient safety (no bleeding and/or perforation), and treatment efficacy (adequate ablated volume). Electroporation treatment using new deployable expandable electrode prototypes is safe and feasible. Moreover, electrode configurations allow for a gradual increase in the ablated area in consecutive steps, as confirmed by histology and immunohistochemistry.
Collapse
Affiliation(s)
- Francesco Izzo
- Division of Surgical Oncology, Hepatobiliary Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (V.A.); (R.P.)
- Correspondence:
| | - Franco Ionna
- Division of Surgical Oncology, Maxillo-Facial Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (F.I.); (F.L.); (A.G.)
| | - Vincenza Granata
- Division of Radiodiagnostic, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy;
| | - Vittorio Albino
- Division of Surgical Oncology, Hepatobiliary Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (V.A.); (R.P.)
| | - Renato Patrone
- Division of General and Oncologic Surgery, Department of Cardiothoracic Sciences, UNIVERSITA’ DEGLI STUDI DELLA CAMPANIA LUIGI VANVITELLI, NAPOLI, ITALIA, 80131 Naples, Italy;
| | - Francesco Longo
- Division of Surgical Oncology, Maxillo-Facial Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (F.I.); (F.L.); (A.G.)
| | - Agostino Guida
- Division of Surgical Oncology, Maxillo-Facial Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (F.I.); (F.L.); (A.G.)
| | - Paolo Delrio
- Division of Surgical Oncology, Colo-Rectal Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (P.D.); (D.R.); (D.S.); (R.P.)
| | - Daniela Rega
- Division of Surgical Oncology, Colo-Rectal Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (P.D.); (D.R.); (D.S.); (R.P.)
| | - Dario Scala
- Division of Surgical Oncology, Colo-Rectal Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (P.D.); (D.R.); (D.S.); (R.P.)
| | - Roberto Pezzuto
- Division of Surgical Oncology, Colo-Rectal Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (P.D.); (D.R.); (D.S.); (R.P.)
| | - Roberta Fusco
- Research & Development Division, Igea SpA, Via Casarea 65, Casalnuovo di Napoli, 80013 Naples, Italy; (R.F.); (E.D.B.); (V.D.)
| | - Elio Di Bernardo
- Research & Development Division, Igea SpA, Via Casarea 65, Casalnuovo di Napoli, 80013 Naples, Italy; (R.F.); (E.D.B.); (V.D.)
| | - Valeria D’Alessio
- Research & Development Division, Igea SpA, Via Casarea 65, Casalnuovo di Napoli, 80013 Naples, Italy; (R.F.); (E.D.B.); (V.D.)
| | - Roberto Grassi
- Division of Radiodiagnostic, UNIVERSITA’ DEGLI STUDI DELLA CAMPANIA LUIGI VANVITELLI, NAPOLI, ITALIA, Via Miraglia, 80143 Naples, Italy;
| | - Deyanira Contartese
- Laboratory Preclinical and Surgical Studies, IRCCS–ISTITUTO ORTOPEDICO RIZZOLI, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Raffaele Palaia
- Division of Surgical Oncology, Hepatobiliary Unit, ISTITUTO NAZIONALE TUMORI–IRCCS-FONDAZIONE G. PASCALE, NAPOLI, ITALIA, Via Mariano Semmola, 80131 Naples, Italy; (V.A.); (R.P.)
| |
Collapse
|
4
|
Haxho F, Neufeld RJ, Szewczuk MR. Neuraminidase-1: a novel therapeutic target in multistage tumorigenesis. Oncotarget 2018; 7:40860-40881. [PMID: 27029067 PMCID: PMC5130050 DOI: 10.18632/oncotarget.8396] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/18/2016] [Indexed: 12/15/2022] Open
Abstract
Several of the growth factors and their receptor tyrosine kinases (RTK) such as epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), nerve growth factor (NGF) and insulin are promising candidate targets for cancer therapy. Indeed, tyrosine kinase inhibitors (TKI) have been developed to target these growth factors and their receptors, and have demonstrated dramatic initial responses in cancer therapy. Yet, most patients ultimately develop TKI drug resistance and relapse. It is essential in the clinical setting that the targeted therapies are to circumvent multistage tumorigenesis, including genetic mutations at the different growth factor receptors, tumor neovascularization, chemoresistance of tumors, immune-mediated tumorigenesis and the development of tissue invasion and metastasis. Here, we identify a novel receptor signaling platform linked to EGF, NGF, insulin and TOLL-like receptor (TLR) activations, all of which are known to play major roles in tumorigenesis. The importance of these findings signify an innovative and promising entirely new targeted therapy for cancer. The role of mammalian neuraminidase-1 (Neu1) in complex with matrix metalloproteinase-9 and G protein-coupled receptor tethered to RTKs and TLRs is identified as a major target in multistage tumorigenesis. Evidence exposing the link connecting growth factor-binding and immune-mediated tumorigenesis to this novel receptor-signaling paradigm will be reviewed in its current relationship to cancer.
Collapse
Affiliation(s)
- Fiona Haxho
- Departments of Biomedical and Molecular Sciences, Kingston, Ontario, Canada
| | - Ronald J Neufeld
- Department of Chemical Engineering, Queen's University, Kingston, Ontario, Canada
| | - Myron R Szewczuk
- Departments of Biomedical and Molecular Sciences, Kingston, Ontario, Canada
| |
Collapse
|
5
|
Lafitte M, Rousseau B, Moranvillier I, Taillepierre M, Peuchant E, Guyonnet-Dupérat V, Bedel A, Dubus P, de Verneuil H, Moreau-Gaudry F, Dabernat S. In vivo gene transfer targeting in pancreatic adenocarcinoma with cell surface antigens. Mol Cancer 2012; 11:81. [PMID: 23088623 PMCID: PMC3546072 DOI: 10.1186/1476-4598-11-81] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 08/16/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma is a deadly malignancy resistant to current therapies. It is critical to test new strategies, including tumor-targeted delivery of therapeutic agents. This study tested the possibility to target the transfer of a suicide gene in tumor cells using an oncotropic lentiviral vector. RESULTS Three cell surface markers were evaluated to target the transduction of cells by lentiviruses pseudotyped with a modified glycoprotein from Sindbis virus. Only Mucin-4 and the Claudin-18 proteins were found efficient for targeted lentivirus transductions in vitro. In subcutaneous xenografts of human pancreatic cancer cells models, Claudin-18 failed to achieve efficient gene transfer but Mucin-4 was found very potent. Human pancreatic tumor cells were modified to express a fluorescent protein detectable in live animals by bioimaging, to perform a direct non invasive and costless follow up of the tumor growth. Targeted gene transfer of a bicistronic transgene bearing a luciferase gene and the herpes simplex virus thymidine kinase gene into orthotopic grafts was carried out with Mucin-4 oncotropic lentiviruses. By contrast to the broad tropism VSV-G carrying lentivirus, this oncotropic lentivirus was found to transduce specifically tumor cells, sparing normal pancreatic cells in vivo. Transduced cells disappeared after ganciclovir treatment while the orthotopic tumor growth was slowed down. CONCLUSION This work considered for the first time three aspect of pancreatic adenocarcinoma targeted therapy. First, lentiviral transduction of human pancreatic tumor cells was possible when cells were grafted orthotopically. Second, we used a system targeting the tumor cells with cell surface antigens and sparing the normal cells. Finally, the TK/GCV anticancer system showed promising results in vivo. Importantly, the approach presented here appeared to be a safer, much more specific and an as efficient way to perform gene delivery in pancreatic tumors, in comparison with a broad tropism lentivirus. This study will be useful in future designing of targeted therapies for pancreatic cancer.
Collapse
|
6
|
An RNA aptamer that specifically binds pancreatic adenocarcinoma up-regulated factor inhibits migration and growth of pancreatic cancer cells. Cancer Lett 2011; 313:76-83. [PMID: 21963224 DOI: 10.1016/j.canlet.2011.08.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/23/2011] [Accepted: 08/23/2011] [Indexed: 01/16/2023]
Abstract
Previously, we reported that a novel secretory protein, pancreatic adenocarcinoma up-regulated factor (PAUF), which is highly expressed in pancreatic cancer and mediates the growth and metastasis of pancreatic cancer cells. In this study, we generated and characterized a 2'-fluoropyrimidine modified RNA aptamer (P12FR2) directed against human PAUF. P12FR2 binds specifically to human PAUF with an estimated apparent K(D) of 77nM. P12FR2 aptamer inhibits PAUF-induced migration of PANC-1, human pancreatic cancer cells, in a wound healing assay. Moreover, intraperitoneal injection of P12FR2 decreased tumor growth by about 60% in an in vivo xenograft model with CFPAC-1 pancreatic cancer cells, without causing a loss of weight in the treated mice. Taken together, we propose here that PAUF-specific RNA aptamer, P12FR2, has the potential to be effective in the therapy of human pancreatic cancer.
Collapse
|