157 results on '"Gabriella Campadelli-Fiume"'
Search Results
2. Innovative retargeted oncolytic herpesvirus against nectin4-positive cancers
- Author
-
Andrea Vannini, Federico Parenti, Cristina Forghieri, Catia Barboni, Anna Zaghini, Gabriella Campadelli-Fiume, and Tatiana Gianni
- Subjects
oncolytic virus ,oncolytic herpes simplex virus ,retargeting ,nectin4 ,anti-cancer vaccine ,immunotherapy ,Biology (General) ,QH301-705.5 - Abstract
Nectin4 is a recently discovered tumor associated antigen expressed in cancers that constitute relevant unmet clinical needs, including the undruggable triple negative breast cancer, pancreatic ductal carcinoma, bladder/urothelial cancer, cervical cancer, lung carcinoma and melanoma. So far, only one nectin4-specific drug—Enfortumab Vedotin—has been approved and the clinical trials that test novel therapeutics are only five. Here we engineered R-421, an innovative retargeted onco-immunotherapeutic herpesvirus highly specific for nectin4 and unable to infect through the natural herpes receptors, nectin1 or herpesvirus entry mediator. In vitro, R-421 infected and killed human nectin4-positive malignant cells and spared normal cells, e.g., human fibroblasts. Importantly from a safety viewpoint, R-421 failed to infect malignant cells that do not harbor nectin4 gene amplification/overexpression, whose expression level was moderate-to-low. In essence, there was a net threshold value below which cells were spared from infection, irrespective of whether they were malignant or normal; the only cells that R-421 targeted were the malignant overexpressing ones. In vivo, R-421 decreased or abolished the growth of murine tumors made transgenic for human nectin4 and conferred sensitivity to immune checkpoint inhibitors in combination therapies. Its efficacy was augmented by the cyclophosphamide immunomodulator and decreased by depletion of CD8-positive lymphocytes, arguing that it was in part T cell-mediated. R-421 elicited in-situ vaccination that protected from distant challenge tumors. This study provides proof-of-principle specificity and efficacy data justifying nectin4-retargeted onco-immunotherapeutic herpesvirus as an innovative approach against a number of difficult-to-drug clinical indications.
- Published
- 2023
- Full Text
- View/download PDF
3. Retargeted and Multi-cytokine-Armed Herpes Virus Is a Potent Cancer Endovaccine for Local and Systemic Anti-tumor Treatment
- Author
-
Maria De Lucia, Gabriella Cotugno, Veronica Bignone, Irene Garzia, Linda Nocchi, Francesca Langone, Biljana Petrovic, Emanuele Sasso, Simona Pepe, Guendalina Froechlich, Chiara Gentile, Nicola Zambrano, Gabriella Campadelli-Fiume, Alfredo Nicosia, Elisa Scarselli, and Anna Morena D’Alise
- Subjects
oncolytic virus ,endovaccine ,cancer ,cytokines ,immune checkpoint ,retargeted Herpes virus ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Oncolytic viruses (OVs) are novel anti-tumor agents with the ability to selectively infect and kill tumor cells while sparing normal tissue. Beyond tumor cytolysis, OVs are capable of priming an anti-tumor immune response via lysis and cross-presentation of locally expressed endogenous tumor antigens, acting as an “endovaccine.” The effectiveness of OVs, similar to other immunotherapies, can be hampered by an immunosuppressive tumor microenvironment. In this study, we modified a previously generated oncolytic herpes simplex virus (oHSV) retargeted to the human HER2 (hHER2) tumor molecule and encoding murine interleukin-12 (mIL-12), by insertion of a second immunomodulatory molecule, murine granulocyte-macrophage colony-stimulating factor (mGM-CSF), to maximize therapeutic efficacy. We assessed the efficacy of this double-armed virus (R-123) compared to singly expressing GM-CSF and IL-12 oHSVs in tumor-bearing mice. While monotherapies were poorly effective, combination with α-PD1 enhanced the anti-tumor response, with the highest efficacy of 100% response rate achieved by the combination of R-123 and α-PD1. Efficacy was T cell-dependent, and the induced immunity was long lasting and able to reject a second contralateral tumor. Importantly, systemic delivery of R-123 combined with α-PD1 was effective in inhibiting the development of tumor metastasis. As such, this approach could have a significant therapeutic impact paving the way for further development of this platform in cancer immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
4. Towards a Precision Medicine Approach and In Situ Vaccination against Prostate Cancer by PSMA-Retargeted oHSV
- Author
-
Andrea Vannini, Federico Parenti, Daniela Bressanin, Catia Barboni, Anna Zaghini, Gabriella Campadelli-Fiume, and Tatiana Gianni
- Subjects
oncolytic virus ,oncolytic herpes simplex virus ,retargeting ,PSMA ,prostate cancer ,vaccination ,Microbiology ,QR1-502 - Abstract
Prostate specific membrane antigen (PSMA) is a specific high frequency cell surface marker of prostate cancers. Theranostic approaches targeting PSMA show no major adverse effects and rule out off-tumor toxicity. A PSMA-retargeted oHSV (R-405) was generated which both infected and was cytotoxic exclusively for PSMA-positive cells, including human prostate cancer LNCaP and 22Rv1 cells, and spared PSMA-negative cells. R-405 in vivo efficacy against LLC1-PSMA and Renca-PSMA tumors consisted of inhibiting primary tumor growth, establishing long-term T immune response, immune heating of the microenvironment, de-repression of the anti-tumor immune phenotype, and sensitization to checkpoint blockade. The in situ vaccination protected from distant challenge tumors, both PSMA-positive and PSMA-negative, implying that it was addressed also to LLC1 tumor antigens. PSMA-retargeted oHSVs are a precision medicine tool worth being additionally investigated in the immunotherapeutic and in situ vaccination landscape against prostate cancers.
- Published
- 2021
- Full Text
- View/download PDF
5. Genotype of Immunologically Hot or Cold Tumors Determines the Antitumor Immune Response and Efficacy by Fully Virulent Retargeted oHSV
- Author
-
Tatiana Gianni, Valerio Leoni, Mara Sanapo, Federico Parenti, Daniela Bressanin, Catia Barboni, Anna Zaghini, Gabriella Campadelli-Fiume, and Andrea Vannini
- Subjects
oncolytic virus ,oncolytic herpes simplex virus ,retargeting ,HER2 ,tumor genotype ,vaccination ,Microbiology ,QR1-502 - Abstract
We report on the efficacy of the non-attenuated HER2-retargeted oHSV named R-337 against the immunologically hot CT26-HER2 tumor, and an insight into the basis of the immune protection. Preliminarily, we conducted an RNA immune profiling and immune cell content characterization of CT26-HER2 tumor in comparison to the immunologically cold LLC1-HER2 tumor. CT26-HER2 tumor was implanted into HER2-transgenic BALB/c mice. Hallmarks of R-337 effects were the protection from primary tumor, long-term adaptive vaccination directed to both HER2 and CT26-wt cell neoantigens. The latter effect differentiated R-337 from OncoVEXGM-CSF. As to the basis of the immune protection, R-337 orchestrated several changes to the tumor immune profile, which cumulatively reversed the immunosuppression typical of this tumor (graphical abstract). Thus, Ido1 (inhibitor of T cell anticancer immunity) levels and T regulatory cell infiltration were decreased; Cd40 and Cd27 co-immunostimulatory markers were increased; the IFNγ cascade was activated. Of note was the dampening of IFN-I response, which we attribute to the fact that R-337 is fully equipped with genes that contrast the host innate response. The IFN-I shut-down likely favored viral replication and the expression of the mIL-12 payload, which, in turn, boosted the antitumor response. The results call for a characterization of tumor immune markers to employ oncolytic herpesviruses more precisely.
- Published
- 2021
- Full Text
- View/download PDF
6. Specificity, Safety, Efficacy of EGFRvIII-Retargeted Oncolytic HSV for Xenotransplanted Human Glioblastoma
- Author
-
Irene Appolloni, Francesco Alessandrini, Laura Menotti, Elisa Avitabile, Daniela Marubbi, Noemi Piga, Davide Ceresa, Francesca Piaggio, Gabriella Campadelli-Fiume, and Paolo Malatesta
- Subjects
oncolytic virotherapy ,glioblastoma stem cells ,glioblastoma initiating cells ,EGFRvIII ,oncolytic HSV ,Microbiology ,QR1-502 - Abstract
Glioblastoma is a lethal primary brain tumor lacking effective therapy. The secluded onset site, combined with the infiltrative properties of this tumor, require novel targeted therapies. In this scenario, the use of oncolytic viruses retargeted to glioblastoma cells and able to spread across the tumor cells represent an intriguing treatment strategy. Here, we tested the specificity, safety and efficacy of R-613, the first oncolytic HSV fully retargeted to EGFRvIII, a variant of the epidermal growth factor receptor carrying a mutation typically found in glioblastoma. An early treatment with R-613 on orthotopically transplanted EGFRvIII-expressing human glioblastoma significantly increased the median survival time of mice. In this setting, the growth of human glioblastoma xenotransplants was monitored by a secreted luciferase reporter and showed that R-613 is able to substantially delay the development of the tumor masses. When administered as late treatment to a well-established glioblastomas, R-613 appeared to be less effective. Notably the uninfected tumor cells derived from the explanted tumor masses were still susceptible to R-613 infection ex vivo, thus suggesting that multiple treatments could enhance R-613 therapeutic efficacy, making R-613 a promising oncolytic HSV candidate for glioblastoma treatment.
- Published
- 2021
- Full Text
- View/download PDF
7. A fully-virulent retargeted oncolytic HSV armed with IL-12 elicits local immunity and vaccine therapy towards distant tumors.
- Author
-
Valerio Leoni, Andrea Vannini, Valentina Gatta, Julie Rambaldi, Mara Sanapo, Catia Barboni, Anna Zaghini, Patrizia Nanni, Pier-Luigi Lollini, Costanza Casiraghi, and Gabriella Campadelli-Fiume
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Oncolytic herpes simplex viruses (oHSVs) showed efficacy in clinical trials and practice. Most of them gain cancer-specificity from deletions/mutations in genes that counteract the host response, and grow selectively in cancer cells defective in anti-viral response. Because of the deletions/mutations, they are frequently attenuated or over-attenuated. We developed next-generation oHSVs, which carry no deletion/mutation, gain cancer-specificity from specific retargeting to tumor cell receptors-e.g. HER2 (human epidermal growth factor receptor 2)-hence are fully-virulent in the targeted cancer cells. The type of immunotherapy they elicit was not predictable, since non-attenuated HSVs induce and then dampen the innate response, whereas deleted/attenuated viruses fail to contrast it, and since the retargeted oHSVs replicate efficiently in tumor cells, but spare other cells in the tumor. We report on the first efficacy study of HER2-retargeted, fully-virulent oHSVs in immunocompetent mice. Their safety profile was very high. Both the unarmed R-LM113 and the IL-12-armed R-115 inhibited the growth of the primary HER2-Lewis lung carcinoma-1 (HER2-LLC1) tumor, R-115 being constantly more efficacious. All the mice that did not die because of the primary treated tumors, were protected from the growth of contralateral untreated tumors. The long-term survivors were protected from a second contralateral tumor, providing additional evidence for an abscopal immunotherapeutic effect. Analysis of the local response highlighted that particularly R-115 unleashed the immunosuppressive tumor microenvironment, i.e. induced immunomodulatory cytokines, including IFNγ, T-bet which promoted Th1 polarization. Some of the tumor infiltrating cells, e.g. CD4+, CD335+ cells were increased in the tumors of all responders mice, irrespective of which virus was employed, whereas CD8+, Foxp3+, CD141+ were increased and CD11b+ cells were decreased preferentially in R-115-treated mice. The durable response included a breakage of tolerance towards both HER2 and the wt tumor cells, and underscored a systemic immunotherapeutic vaccine response.
- Published
- 2018
- Full Text
- View/download PDF
8. Insertion of a ligand to HER2 in gB retargets HSV tropism and obviates the need for activation of the other entry glycoproteins.
- Author
-
Biljana Petrovic, Tatiana Gianni, Valentina Gatta, and Gabriella Campadelli-Fiume
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Herpes simplex virus (HSV) entry into the cells requires glycoproteins gD, gH/gL and gB, activated in a cascade fashion by conformational modifications induced by cognate receptors and intermolecular signaling. The receptors are nectin1 and HVEM (Herpes virus entry mediator) for gD, and αvβ6 or αvβ8 integrin for gH. In earlier work, insertion of a single chain antibody (scFv) to the cancer receptor HER2 (human epidermal growth factor receptor 2) in gD, or in gH, resulted in HSVs specifically retargeted to the HER2-positive cancer cells, hence in highly specific non-attenuated oncolytic agents. Here, the scFv to HER2 was inserted in gB (gBHER2). The insertion re-targeted the virus tropism to the HER2-positive cancer cells. This was unexpected since gB is known to be a fusogenic glycoprotein, not a tropism determinant. The gB-retargeted recombinant offered the possibility to investigate how HER2 mediated entry. In contrast to wt-gB, the activation of the chimeric gBHER2 did not require the activation of the gD and of gH/gL by their respective receptors. Furthermore, a soluble form of HER2 could replace the membrane-bound HER2 in mediating virus entry, hinting that HER2 acted by inducing conformational changes to the chimeric gB. This study shows that (i) gB can be modified and become the major determinant of HSV tropism; (ii) the chimeric gBHER2 bypasses the requirement for receptor-mediated activation of other essential entry glycoproteins.
- Published
- 2017
- Full Text
- View/download PDF
9. HSV as A Platform for the Generation of Retargeted, Armed, and Reporter-Expressing Oncolytic Viruses
- Author
-
Laura Menotti, Elisa Avitabile, Valentina Gatta, Paolo Malatesta, Biljana Petrovic, and Gabriella Campadelli-Fiume
- Subjects
retargeting ,oncolytic herpesvirus ,PSMA ,EGFR ,EGFRvIII ,mIL12 ,Gaussia Luciferase ,HER2 ,Microbiology ,QR1-502 - Abstract
Previously, we engineered oncolytic herpes simplex viruses (o-HSVs) retargeted to the HER2 (epidermal growth factor receptor 2) tumor cell specific receptor by the insertion of a single chain antibody (scFv) to HER2 in gD, gH, or gB. Here, the insertion of scFvs to three additional cancer targets—EGFR (epidermal growth factor receptor), EGFRvIII, and PSMA (prostate specific membrane antigen)—in gD Δ6–38 enabled the generation of specifically retargeted o-HSVs. Viable recombinants resulted from the insertion of an scFv in place of aa 6–38, but not in place of aa 61–218. Hence, only the gD N-terminus accepted all tested scFv inserts. Additionally, the insertion of mIL12 in the US1-US2 intergenic region of the HER2- or EGFRvIII-retargeted o-HSVs, and the further insertion of Gaussia Luciferase, gave rise to viable recombinants capable of secreting the cytokine and the reporter. Lastly, we engineered two known mutations in gB; they increased the ability of an HER2-retargeted recombinant to spread among murine cells. Altogether, current data show that the o-HSV carrying the aa 6–38 deletion in gD serves as a platform for the specific retargeting of o-HSV tropism to a number of human cancer targets, and the retargeted o-HSVs serve as simultaneous vectors for two molecules.
- Published
- 2018
- Full Text
- View/download PDF
10. The Engineering of a Novel Ligand in gH Confers to HSV an Expanded Tropism Independent of gD Activation by Its Receptors.
- Author
-
Valentina Gatta, Biljana Petrovic, and Gabriella Campadelli-Fiume
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Herpes simplex virus (HSV) enters cells by means of four essential glycoproteins - gD, gH/gL, gB, activated in a cascade fashion by gD binding to one of its receptors, nectin1 and HVEM. We report that the engineering in gH of a heterologous ligand - a single-chain antibody (scFv) to the cancer-specific HER2 receptor - expands the HSV tropism to cells which express HER2 as the sole receptor. The significance of this finding is twofold. It impacts on our understanding of HSV entry mechanism and the design of retargeted oncolytic-HSVs. Specifically, entry of the recombinant viruses carrying the scFv-HER2-gH chimera into HER2+ cells occurred in the absence of gD receptors, or upon deletion of key residues in gD that constitute the nectin1/HVEM binding sites. In essence, the scFv in gH substituted for gD-mediated activation and rendered a functional gD non-essential for entry via HER2. The activation of the gH moiety in the chimera was carried out by the scFv in cis, not in trans as it occurs with wt-gD. With respect to the design of oncolytic-HSVs, previous retargeting strategies were based exclusively on insertion in gD of ligands to cancer-specific receptors. The current findings show that (i) gH accepts a heterologous ligand. The viruses retargeted via gH (ii) do not require the gD-dependent activation, and (iii) replicate and kill cells at high efficiency. Thus, gH represents an additional tool for the design of fully-virulent oncolytic-HSVs retargeted to cancer receptors and detargeted from gD receptors.
- Published
- 2015
- Full Text
- View/download PDF
11. The epithelial αvβ3-integrin boosts the MYD88-dependent TLR2 signaling in response to viral and bacterial components.
- Author
-
Tatiana Gianni and Gabriella Campadelli-Fiume
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
TLR2 is a cell surface receptor which elicits an immediate response to a wide repertoire of bacteria and viruses. Its response is usually thought to be proinflammatory rather than an antiviral. In monocytic cells TLR2 cooperates with coreceptors, e.g. CD14, CD36 and αMβ2-integrin. In an earlier work we showed that αvβ3-integrin acts in concert with TLR2 to elicit an innate response to HSV, and to lipopolysaccharide. This response is characterized by production of IFN-α and -β, a specific set of cytokines, and NF-κB activation. We investigated the basis of the cooperation between αvβ3-integrin and TLR2. We report that β3-integrin participates by signaling through Y residues located in the C-tail, known to be involved in signaling activity. αvβ3-integrin boosts the MYD88-dependent TLR2 signaling and IRAK4 phosphorylation in 293T and in epithelial, keratinocytic and neuronal cell lines. The replication of ICP0minus HSV is greatly enhanced by DN versions of MYD88, of Akt - a hub of this pathway, or by β3integrin-silencing. αvβ3-integrin enables the recruitment of TLR2, MAL, MYD88 at lipid rafts, the platforms from where the signaling starts. The PAMP of the HSV-induced innate response is the gH/gL virion glycoprotein, which interacts with αvβ3-integrin and TLR2 independently one of the other, and cross-links the two receptors. Given the preferential distribution of αvβ3-integrin to epithelial cells, we propose that αvβ3-integrin serves as coreceptor of TLR2 in these cells. The results open the possibility that TLR2 makes use of coreceptors in a variety of cells to broaden its spectrum of activity and tissue specificity.
- Published
- 2014
- Full Text
- View/download PDF
12. Human Herpesvirus 6: An Emerging Pathogen
- Author
-
Gabriella Campadelli-Fiume, Prisco Mirandola, and Laura Menotti
- Subjects
Italy ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Infections with human herpesvirus 6 (HHV-6), a ß-herpesvirus of which two variant groups (A and B) are recognized, is very common, approaching 100% in seroprevalence. Primary infection with HHV-6B causes roseola infantum or exanthem subitum, a common childhood disease that resolves spontaneously. After primary infection, the virus replicates in the salivary glands and is shed in saliva, the recognized route of transmission for variant B strains; it remains latent in lymphocytes and monocytes and persists at low levels in cells and tissues. Not usually associated with disease in the immunocompetent, HHV-6 infection is a major cause of opportunistic viral infections in the immunosuppressed, typically AIDS patients and transplant recipients, in whom HHV-6 infection/reactivation may culminate in rejection of transplanted organs and death. Other opportunistic viruses, human cytomegalovirus and HHV-7, also infect or reactivate in persons at risk. Another disease whose pathogenesis may be correlated with HHV-6 is multiple sclerosis. Data in favor of and against the correlation are discussed.
- Published
- 1999
- Full Text
- View/download PDF
13. Preclinical therapy of disseminated HER-2⁺ ovarian and breast carcinomas with a HER-2-retargeted oncolytic herpesvirus.
- Author
-
Patrizia Nanni, Valentina Gatta, Laura Menotti, Carla De Giovanni, Marianna Ianzano, Arianna Palladini, Valentina Grosso, Massimiliano Dall'ora, Stefania Croci, Giordano Nicoletti, Lorena Landuzzi, Manuela Iezzi, Gabriella Campadelli-Fiume, and Pier-Luigi Lollini
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Oncolytic viruses aim to specifically kill tumor cells. A major challenge is the effective targeting of disseminated tumors in vivo. We retargeted herpes simplex virus (HSV) tropism to HER-2 oncoprotein p185, overexpressed in ovary and breast cancers. The HER-2-retargeted R-LM249 exclusively infects and kills tumor cells expressing high levels of human HER-2. Here, we assessed the efficacy of systemically i.p. delivered R-LM249 against disseminated tumors in mouse models that recapitulate tumor spread to the peritoneum in women. The human ovarian carcinoma SK-OV-3 cells implanted intraperitoneally (i.p.) in immunodeficient Rag2⁻/⁻;Il2rg⁻/⁻ mice gave rise to a progressive peritoneal carcinomatosis which mimics the fatal condition in advanced human patients. I.p. administration of R-LM249 strongly inhibited carcinomatosis, resulting in 60% of mice free from peritoneal diffusion, and 95% reduction in the total weight of neoplastic nodules. Intraperitoneal metastases are a common outcome in breast cancer: i.p. administration of R-LM249 strongly inhibited the growth of ovarian metastases of HER-2+ MDA-MB-453 breast cells. Brain metastases were also reduced. Cumulatively, upon i.p. administration the HER-2-redirected oncolytic HSV effectively reduced the growth of ovarian and breast carcinoma disseminated to the peritoneal cavity.
- Published
- 2013
- Full Text
- View/download PDF
14. αvβ6- and αvβ8-integrins serve as interchangeable receptors for HSV gH/gL to promote endocytosis and activation of membrane fusion.
- Author
-
Tatiana Gianni, Stefano Salvioli, Liudmila S Chesnokova, Lindsey M Hutt-Fletcher, and Gabriella Campadelli-Fiume
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Herpes simplex virus (HSV)--and herpesviruses in general--encode for a multipartite entry/fusion apparatus. In HSV it consists of the HSV-specific glycoprotein D (gD), and three additional glycoproteins, gH/gL and gB, conserved across the Herpesviridae family and responsible for the execution of fusion. According to the current model, upon receptor binding, gD propagates the activation to gH/gL and to gB in a cascade fashion. Questions remain about how the cascade of activation is controlled and how it is synchronized with virion endocytosis, to avoid premature activation and exhaustion of the glycoproteins. We considered the possibility that such control might be carried out by as yet unknown receptors. Indeed, receptors for HSV gB, but not for gH/gL, have been described. In other members of the Herpesviridae family, such as Epstein-Barr virus, integrin receptors bind gH/gL and trigger conformational changes in the glycoproteins. We report that αvβ6- and αvβ8-integrins serve as receptors for HSV entry into experimental models of keratinocytes and other epithelial and neuronal cells. Evidence rests on loss of function experiments, in which integrins were blocked by antibodies or silenced, and gain of function experiments in which αvβ6-integrin was expressed in integrin-negative cells. αvβ6- and αvβ8-integrins acted independently and are thus interchangeable. Both bind gH/gL with high affinity. The interaction profoundly affects the route of HSV entry and directs the virus to acidic endosomes. In the case of αvβ8, but not αvβ6-integrin, the portal of entry is located at lipid microdomains and requires dynamin 2. Thus, a major role of αvβ6- or αvβ8-integrin in HSV infection appears to be to function as gH/gL receptors and to promote virus endocytosis. We propose that placing the gH/gL activation under the integrin trigger point enables HSV to synchronize virion endocytosis with the cascade of glycoprotein activation that culminates in execution of fusion.
- Published
- 2013
- Full Text
- View/download PDF
15. Retargeted and Multi-cytokine-Armed Herpes Virus Is a Potent Cancer Endovaccine for Local and Systemic Anti-tumor Treatment
- Author
-
Gabriella Campadelli-Fiume, Gabriella Cotugno, Irene Garzia, Alfredo Nicosia, Maria De Lucia, Francesca Langone, Emanuele Sasso, Guendalina Froechlich, Chiara Gentile, Biljana Petrovic, Anna Morena D'Alise, Nicola Zambrano, Simona Pepe, Veronica Bignone, Elisa Scarselli, Linda Nocchi, De Lucia, M., Cotugno, G., Bignone, V., Garzia, I., Nocchi, L., Langone, F., Petrovic, B., Sasso, E., Pepe, S., Froechlich, G., Gentile, C., Zambrano, N., Campadelli-Fiume, G., Nicosia, A., Scarselli, E., and D'Alise, A. M.
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,medicine.disease_cause ,lcsh:RC254-282 ,Virus ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,medicine ,cytokine ,cancer ,Pharmacology (medical) ,immune checkpoint ,retargeted Herpes virus ,oncolytic virus ,Tumor microenvironment ,business.industry ,GM-CSF ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint ,cytokines ,Oncolytic virus ,030104 developmental biology ,Herpes simplex virus ,Oncology ,oncolytic viru ,IL-12 ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Original Article ,business ,endovaccine - Abstract
Oncolytic viruses (OVs) are novel anti-tumor agents with the ability to selectively infect and kill tumor cells while sparing normal tissue. Beyond tumor cytolysis, OVs are capable of priming an anti-tumor immune response via lysis and cross-presentation of locally expressed endogenous tumor antigens, acting as an “endovaccine.” The effectiveness of OVs, similar to other immunotherapies, can be hampered by an immunosuppressive tumor microenvironment. In this study, we modified a previously generated oncolytic herpes simplex virus (oHSV) retargeted to the human HER2 (hHER2) tumor molecule and encoding murine interleukin-12 (mIL-12), by insertion of a second immunomodulatory molecule, murine granulocyte-macrophage colony-stimulating factor (mGM-CSF), to maximize therapeutic efficacy. We assessed the efficacy of this double-armed virus (R-123) compared to singly expressing GM-CSF and IL-12 oHSVs in tumor-bearing mice. While monotherapies were poorly effective, combination with α-PD1 enhanced the anti-tumor response, with the highest efficacy of 100% response rate achieved by the combination of R-123 and α-PD1. Efficacy was T cell-dependent, and the induced immunity was long lasting and able to reject a second contralateral tumor. Importantly, systemic delivery of R-123 combined with α-PD1 was effective in inhibiting the development of tumor metastasis. As such, this approach could have a significant therapeutic impact paving the way for further development of this platform in cancer immunotherapy., Graphical Abstract, Fully virulent tumor retargeted HSV oncolytic viruses (THVs) are novel immunotherapeutic agents with increased specificity, safety, and potency. De Lucia et al. propose the use of a hHER2 THV expressing IL-12 and GM-CSF as a strategy to potentiate anti-tumor efficacy in combination with anti-PD1, opening future perspectives for local and systemic treatment.
- Published
- 2020
16. Genotype of Immunologically Hot or Cold Tumors Determines the Antitumor Immune Response and Efficacy by Fully Virulent Retargeted oHSV
- Author
-
Gabriella Campadelli-Fiume, Anna Zaghini, Valerio Leoni, Tatiana Gianni, Mara Sanapo, Federico Parenti, Andrea Vannini, Daniela Bressanin, Catia Barboni, Gianni T., Leoni V., Sanapo M., Parenti F., Bressanin D., Barboni C., Zaghini A., Campadelli-Fiume G., and Vannini A.
- Subjects
Receptor, ErbB-2 ,medicine.medical_treatment ,Cell ,Herpesvirus 1, Human ,Immune checkpoint inhibitor ,Virus Replication ,immune checkpoint inhibitors ,Mice ,0302 clinical medicine ,Neoplasms ,skin and connective tissue diseases ,0303 health sciences ,Mice, Inbred BALB C ,Immunosuppression ,oncolytic herpes simplex virus ,Primary tumor ,QR1-502 ,3. Good health ,Oncolytic Viruses ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunotherapy ,Oncolytic herpes simplex viru ,Genotype ,T cell ,Mice, Transgenic ,Biology ,tumor genotype ,Microbiology ,Article ,03 medical and health sciences ,Immune system ,Virology ,HER2 ,medicine ,Animals ,Humans ,030304 developmental biology ,oncolytic virus ,retargeting ,Immunity ,Oncolytic viru ,medicine.disease ,vaccination ,Oncolytic virus ,Mice, Inbred C57BL ,Viral replication ,Cancer research - Abstract
We report on the efficacy of the non-attenuated HER2-retargeted oHSV named R-337 against the immunologically hot CT26-HER2 tumor, and an insight into the basis of the immune protection. Preliminarily, we conducted an RNA immune profiling and immune cell content characterization of CT26-HER2 tumor in comparison to the immunologically cold LLC1-HER2 tumor. CT26-HER2 tumor was implanted into HER2-transgenic BALB/c mice. Hallmarks of R-337 effects were the protection from primary tumor, long-term adaptive vaccination directed to both HER2 and CT26-wt cell neoantigens. The latter effect differentiated R-337 from OncoVEXGM-CSF. As to the basis of the immune protection, R-337 orchestrated several changes to the tumor immune profile, which cumulatively reversed the immunosuppression typical of this tumor (graphical abstract). Thus, Ido1 (inhibitor of T cell anticancer immunity) levels and T regulatory cell infiltration were decreased, Cd40 and Cd27 co-immunostimulatory markers were increased, the IFNγ cascade was activated. Of note was the dampening of IFN-I response, which we attribute to the fact that R-337 is fully equipped with genes that contrast the host innate response. The IFN-I shut-down likely favored viral replication and the expression of the mIL-12 payload, which, in turn, boosted the antitumor response. The results call for a characterization of tumor immune markers to employ oncolytic herpesviruses more precisely.
- Published
- 2021
17. Specificity, Safety, Efficacy of EGFRvIII-Retargeted Oncolytic HSV for Xenotransplanted Human Glioblastoma
- Author
-
Gabriella Campadelli-Fiume, Noemi Piga, Irene Appolloni, Francesca Piaggio, Elisa Avitabile, Paolo Malatesta, Francesco Alessandrini, Davide Ceresa, Daniela Marubbi, Laura Menotti, Appolloni I., Alessandrini F., Menotti L., Avitabile E., Marubbi D., Piga N., Ceresa D., Piaggio F., Campadelli-Fiume G., and Malatesta P.
- Subjects
HSL and HSV ,Herpesvirus 1, Human ,Mice, SCID ,medicine.disease_cause ,Virus Replication ,Mice ,0302 clinical medicine ,Chlorocebus aethiops ,Medicine ,Epidermal growth factor receptor ,Oncolytic Virotherapy ,0303 health sciences ,Mutation ,biology ,glioblastoma stem cells ,EGFRvIII ,Glioblastoma initiating cells ,Glioblastoma stem cells ,Oncolytic HSV ,Oncolytic virotherapy ,QR1-502 ,3. Good health ,ErbB Receptors ,Oncolytic Viruses ,Infectious Diseases ,030220 oncology & carcinogenesis ,Median survival ,Genetic Vectors ,Transplantation, Heterologous ,Brain tumor ,Microbiology ,Article ,03 medical and health sciences ,Virology ,Cell Line, Tumor ,glioblastoma initiating cells ,Animals ,Humans ,Glioblastoma stem cell ,Vero Cells ,030304 developmental biology ,Glioblastoma initiating cell ,business.industry ,oncolytic HSV ,medicine.disease ,Oncolytic virus ,Cancer research ,biology.protein ,business ,Glioblastoma ,Ex vivo - Abstract
Glioblastoma is a lethal primary brain tumor lacking effective therapy. The secluded onset site, combined with the infiltrative properties of this tumor, require novel targeted therapies. In this scenario, the use of oncolytic viruses retargeted to glioblastoma cells and able to spread across the tumor cells represent an intriguing treatment strategy. Here, we tested the specificity, safety and efficacy of R-613, the first oncolytic HSV fully retargeted to EGFRvIII, a variant of the epidermal growth factor receptor carrying a mutation typically found in glioblastoma. An early treatment with R-613 on orthotopically transplanted EGFRvIII-expressing human glioblastoma significantly increased the median survival time of mice. In this setting, the growth of human glioblastoma xenotransplants was monitored by a secreted luciferase reporter and showed that R-613 is able to substantially delay the development of the tumor masses. When administered as late treatment to a well-established glioblastomas, R-613 appeared to be less effective. Notably the uninfected tumor cells derived from the explanted tumor masses were still susceptible to R-613 infection ex vivo, thus suggesting that multiple treatments could enhance R-613 therapeutic efficacy, making R-613 a promising oncolytic HSV candidate for glioblastoma treatment.
- Published
- 2021
18. αvβ3-integrin regulates PD-L1 expression and is involved in cancer immune evasion
- Author
-
Catia Barboni, Anna Zaghini, Valerio Leoni, Tatiana Gianni, Gabriella Campadelli-Fiume, Andrea Vannini, Paolo Malatesta, Mara Sanapo, Vannini A., Leoni V., Barboni C., Sanapo M., Zaghini A., Malatesta P., Campadelli-Fiume G., and Gianni T.
- Subjects
medicine.medical_treatment ,Integrin ,Cell ,αvβ3-integrin ,PD-L1 expression ,Corrections ,combination therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Cancer immunotherapy ,medicine ,Splenocyte ,immune evasion ,030304 developmental biology ,0303 health sciences ,cancer immunotherapy ,Multidisciplinary ,biology ,Chemistry ,Immunotherapy ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,CD8 - Abstract
Tumors utilize a number of effective strategies, including the programmed death 1/PD ligand 1 (PD-1/PD-L1) axis, to evade immune-mediated control of their growth. PD-L1 expression is mainly induced by IFN receptor signaling or constitutively induced. Integrins are an abundantly expressed class of proteins which play multiple deleterious roles in cancer and exert proangiogenic and prosurvival activities. We asked whether αvβ3-integrin positively regulates PD-L1 expression and the anticancer immune response. We report that αvβ3-integrin regulated constitutive and IFN-induced PD-L1 expression in human and murine cancerous and noncancerous cells. αvβ3-integrin targeted STAT1 through its signaling C tail. The implantation of β3-integrin-depleted tumor cells led to a dramatic decrease in the growth of primary tumors, which exhibited reduced PD-L1 expression and became immunologically hot, with increased IFNγ content and CD8+ cell infiltration. In addition, the implantation of β3-integrin-depleted tumors elicited an abscopal immunotherapeutic effect measured as protection from the challenge tumor and durable splenocyte and serum reactivity to B16 cell antigens. These modifications to the immunosuppressive microenvironment primed cells for checkpoint (CP) blockade. When combined with anti-PD-1, β3-integrin depletion led to durable therapy and elicited an abscopal immunotherapeutic effect. We conclude that in addition to its previously known roles, αvβ3-integrin serves as a critical component of the cancer immune evasion strategy and can be an effective immunotherapy target.
- Published
- 2019
19. Targeted Delivery of IL-12 Adjuvants Immunotherapy by Oncolytic Viruses
- Author
-
Andrea, Vannini, Valerio, Leoni, and Gabriella, Campadelli-Fiume
- Subjects
Oncolytic Virotherapy ,Mice ,Oncolytic Viruses ,Tumor Microenvironment ,Animals ,Humans ,Immunotherapy ,Interleukin-12 - Abstract
The great hopes raised by the discovery of the immunoregulatory cytokine interleukin 12 (IL-12) as an anticancer agent were marred during early clinical experimentation because of severe adverse effects, which prompted a search for alternative formulations and routes of administration. Onco-immunotherapeutic viruses (OIVs) are wild-type or genetically engineered viruses that exert antitumor activity by causing death of the tumor cells they infect and by overcoming a variety of immunosuppressive mechanisms put in place by the tumors. OIVs have renewed the interest in IL-12, as they offer the opportunity to encode the cytokine transgenically from the viral genome and to produce it at high concentrations in the tumor bed. A large body of evidence indicates that IL-12 serves as a potent adjuvant for the immunotherapeutic response elicited by OIVs in murine tumor models. The list of OIVs includes onco-immunotherapeutic herpes simplex, adeno, measles, Newcastle disease, and Maraba viruses, among others. The large increase in IL-12-mediated adjuvanticity was invariably observed for all the OIVs analyzed. Indirect evidence suggests that locally delivered IL-12 may also increase tumor antigenicity. Importantly, the OIV/IL-12 treatment was not accompanied by adverse effects and elicited a long-lasting immune response capable of halting the growth of distant tumors. Thus, OIVs provide an avenue for reducing the clinical toxicity associated with systemic IL-12 therapy, by concentrating the cytokine at the site of disease. The changes to the tumor microenvironment induced by the IL-12-armed OIVs primed the tumors to an improved response to the checkpoint blockade therapy, suggesting that the triple combination is worth pursuing in the future. The highly encouraging results in preclinical models have prompted translation to the clinic. How well the IL-12-OIV-checkpoint inhibitors' combination will perform in humans remains to be fully investigated.
- Published
- 2021
20. Immunotherapeutic Efficacy of Retargeted oHSVs Designed for Propagation in an Ad Hoc Cell Line
- Author
-
Catia Barboni, Anna Zaghini, Tatiana Gianni, Valentina Gatta, Andrea Vannini, Giorgia Giordani, Mara Sanapo, Gabriella Campadelli-Fiume, Valerio Leoni, Vannini A., Leoni V., Sanapo M., Gianni T., Giordani G., Gatta V., Barboni C., Zaghini A., and Campadelli-fiume G.
- Subjects
0301 basic medicine ,Cancer Research ,tumor ,medicine.medical_treatment ,antigen-agnostic vaccination ,Review ,Biology ,medicine.disease_cause ,Retargeted viru ,lcsh:RC254-282 ,tropism retargeting ,Antigen‐agnostic vaccination ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,HER2 ,medicine ,Herpes simplex viru ,oncolytic virus ,Tumor microenvironment ,retargeted virus ,Oncolytic viru ,Immunotherapy ,vaccination ,herpes simplex virus ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Oncolytic virus ,checkpoint inhibitor ,030104 developmental biology ,Herpes simplex virus ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Retargeting ,Cancer research ,Vero cell ,immunotherapy - Abstract
Simple Summary The onco-immunotherapeutic viruses, among which stand the onco-immunotherapeutic herpes simplex viruses, have gained renewed interest due to their ability to unlock the potential of checkpoint inhibitors in preclinical and clinical settings. In prior decades, safety concerns led to the generation of overall safe, partially or highly attenuated oncolytic viruses. Current focus is on more efficacious onco-immunotherapeutic viruses with limited ability to cause off-tumor and off-target infections and the capability to subvert the tumor microenvironment immunosuppression—hence to potentiate checkpoint inhibitors. These viruses might serve as potential partners of T-cell therapies. Abstract Our laboratory has pursued the generation of cancer-specific oncolytic herpes simplex viruses (oHSVs) which ensure high efficacy while maintaining a high safety profile. Their blueprint included retargeting to a Tumor-Associated Antigen, e.g., HER2, coupled to detargeting from natural receptors to avoid off-target and off-tumor infections and preservation of the full complement of unmodified viral genes. These oHSVs are “fully virulent in their target cancer cells”. The 3rd generation retargeted oHSVs carry two distinct retargeting moieties, which enable infection of a producer cell line and of the target cancer cells, respectively. They can be propagated in an ad hoc Vero cell derivative at about tenfold higher yields than 1st generation recombinants, and more effectively replicate in human cancer cell lines. The R-335 and R-337 prototypes were armed with murine IL-12. Intratumorally-administered R-337 conferred almost complete protection from LLC-1-HER2 primary tumors, unleashed the tumor microenvironment immunosuppression, synergized with the checkpoint blockade and conferred long-term vaccination against distant challenge tumors. In summary, the problem intrinsic to the propagation of retargeted oHSVs—which strictly require cells positive for targeted receptors—was solved in 3rd generation viruses. They are effective as immunotherapeutic agents against primary tumors and as antigen-agnostic vaccines.
- Published
- 2021
21. Rescue, Purification, and Characterization of a Recombinant HSV Expressing a Transgenic Protein
- Author
-
Tatiana Gianni, Laura Menotti, Valentina Gatta, Simona Pepe, Biljana Petrovic, Gabriella Campadelli-Fiume, Andrea Vannini, Valerio Leoni, Diefenbach R., Fraefel C. (eds), and Andrea Vannini, Biljana Petrovic, Valentina Gatta, Valerio Leoni, Simona Pepe, Laura Menotti, Gabriella Campadelli-Fiume, Tatiana Gianni
- Subjects
0301 basic medicine ,Virus quantification ,Transgene ,Biology ,Recombinant virus ,Genome ,Recombineering ,law.invention ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,mIL12 transgene expression ,In vivo ,law ,Plaque purification ,030220 oncology & carcinogenesis ,Recombinant DNA ,Interleukin 12 ,HSV rescue ,Virion purification ,Plaque assay - Abstract
In the previous chapter, we describe the engineering of a HSV-BAC genome by galK recombineering. Here we describe the procedures to reconstitute, or regenerate, the replicating recombinant virus, and the methods to purify it and characterize it for the correct expression of the transgene. We present the example of R-115, a recombinant expressing murine interleukin 12 (mIL12) from the US1-US2 intergenic region. A specific method for the production of highly purified virions by iodixanol gradient, suitable for in vivo applications, is also detailed.
- Published
- 2019
22. oHSV Genome Editing by Means of galK Recombineering
- Author
-
Andrea Vannini, Tatiana Gianni, Laura Menotti, Valerio Leoni, Biljana Petrovic, Simona Pepe, Gabriella Campadelli-Fiume, Valentina Gatta, Russell J. Diefenbach, Cornel Fraefel, and Laura Menotti, Valerio Leoni, Valentina Gatta, Biljana Petrovic, Andrea Vannini, Simona Pepe, Tatiana Gianni, Gabriella Campadelli-Fiume
- Subjects
0301 basic medicine ,Bacterial artificial chromosome ,Virus arming ,viruses ,Virus engineering ,Computational biology ,Amplicon ,Biology ,Recombinant virus ,medicine.disease_cause ,Genome ,Recombineering ,galK recombineering ,Interleukin 12 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Herpes simplex virus ,Genome editing ,030220 oncology & carcinogenesis ,medicine ,Herpes simplex viru ,Transgene expression ,Oncolytic virotherapy ,Gene - Abstract
Since the cloning of the herpes simplex virus (HSV) genome as BAC (bacterial artificial chromosome), the genetic engineering of the viral genome has become readily feasible. The advantage is that the modification of the animal virus genome is carried out in bacteria, with no replication or production of viral progeny, and is separated from the reconstitution or regeneration of the recombinant virus in mammalian cells. This allows an easy engineering of essential genes, as well. Many technologies have been developed for herpesvirus BAC engineering. In our hands the most powerful is galK recombineering that exploits a single marker (galK) for positive and negative selection and PCR amplicons for seamless modification in the desired genome locus. Here we describe the engineering of the HSV recombinant BAC 115 by the insertion of a heterologous cassette for the expression of murine interleukin 12 (mIL12) in the intergenic sequence between US1 and US2 ORFs.
- Published
- 2019
23. Replicative conditioning of Herpes simplex type 1 virus by Survivin promoter, combined to ERBB2 retargeting, improves tumour cell-restricted oncolysis
- Author
-
Gabriella Campadelli-Fiume, Gabriella Cotugno, Alfredo Nicosia, Nicola Zambrano, Emanuele Sasso, Chiara Gentile, Elisa Scarselli, Anna Morena D'Alise, Guendalina Froechlich, Veronica Bignone, Maria De Lucia, Biljana Petrovic, Sasso, E., Froechlich, G., Cotugno, G., D'Alise, A. M., Gentile, C., Bignone, V., De Lucia, M., Petrovic, B., Campadelli-Fiume, G., Scarselli, E., Nicosia, A., and Zambrano, N.
- Subjects
Xenograft Model Antitumor Assay ,Receptor, ErbB-2 ,viruses ,Survivin ,Cell ,Virulence ,lcsh:Medicine ,Cancer immunotherapy ,Herpesvirus 1, Human ,Biology ,Virus Replication ,Virus ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Targeted therapies ,medicine ,Tumor Cells, Cultured ,Cytotoxic T cell ,Herpes virus ,Animals ,Humans ,lcsh:Science ,Promoter Regions, Genetic ,Tropism ,030304 developmental biology ,Oncolytic Virotherapy ,Ovarian Neoplasms ,0303 health sciences ,Multidisciplinary ,Animal ,Ovarian Neoplasm ,lcsh:R ,Xenograft Model Antitumor Assays ,Immune checkpoint ,3. Good health ,Oncolytic virus ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Viral replication ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:Q ,Female ,Human - Abstract
Oncolytic virotherapy is emerging as a promising therapeutic option for solid tumours. Several oncolytic vectors in clinical testing are based on attenuated viruses; thus, efforts are being taken to develop a new repertoire of oncolytic viruses, based on virulent viral genomes. This possibility, however, raises concerns dealing with the safety features of the virulent phenotypes. We generated a double regulated Herpes simplex type-1 virus (HSV-1), in which tumour cell restricted replicative potential was combined to selective entry via ERBB2 receptor retargeting. The transcriptional control of the viral alpha4 gene encoding for the infected cell protein-4 (ICP4) by the cellular Survivin/BIRC5 promoter conferred a tumour cell-restricted replicative potential to a virulent HSV-1 genome. The combination of the additional ERBB2 retargeting further improved the selectivity for tumour cells, conferring to the double regulated virus a very limited ability to infect and propagate in non-cancerous cells. Accordingly, a suitable replicative and cytotoxic potential was maintained in tumour cell lines, allowing the double regulated virus to synergize in vivo with immune checkpoint (anti-PD-1) blockade in immunocompetent mice. Thus, restricting the replicative spectrum and tropism of virulent HSV-1 genomes by combination of conditional replication and retargeting provides an improved safety, does not alter the oncolytic strength, and is exploitable for its therapeutic potential with immune checkpoint blockade in cancer.
- Published
- 2020
24. Towards a Precision Medicine Approach and In Situ Vaccination against Prostate Cancer by PSMA-Retargeted oHSV
- Author
-
Tatiana Gianni, Gabriella Campadelli-Fiume, Catia Barboni, Federico Parenti, Daniela Bressanin, Anna Zaghini, Andrea Vannini, Vannini A., Parenti F., Bressanin D., Barboni C., Zaghini A., Campadelli-Fiume G., and Gianni T.
- Subjects
Male ,medicine.medical_treatment ,Immune checkpoint inhibitor ,urologic and male genital diseases ,immune checkpoint inhibitors ,Prostate cancer ,0302 clinical medicine ,Prostate ,Tumor Microenvironment ,Precision Medicine ,In Situ Hybridization ,Oncolytic Virotherapy ,0303 health sciences ,oncolytic herpes simplex virus ,prostate cancer ,Primary tumor ,QR1-502 ,3. Good health ,Oncolytic Viruses ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Kallikreins ,immunotherapy ,Oncolytic herpes simplex viru ,Antineoplastic Agents ,Microbiology ,Article ,03 medical and health sciences ,Immune system ,Antigen ,Cell Line, Tumor ,Virology ,LNCaP ,PSMA ,medicine ,Humans ,oncolytic virus ,retargeting ,030304 developmental biology ,business.industry ,Oncolytic viru ,Prostatic Neoplasms ,Cancer ,Immunotherapy ,Prostate-Specific Antigen ,vaccination ,medicine.disease ,in situ vaccine ,Cancer research ,business ,Biomarkers - Abstract
Prostate specific membrane antigen (PSMA) is a specific high frequency cell surface marker of prostate cancers. Theranostic approaches targeting PSMA show no major adverse effects and rule out off-tumor toxicity. A PSMA-retargeted oHSV (R-405) was generated which both infected and was cytotoxic exclusively for PSMA-positive cells, including human prostate cancer LNCaP and 22Rv1 cells, and spared PSMA-negative cells. R-405 in vivo efficacy against LLC1-PSMA and Renca-PSMA tumors consisted of inhibiting primary tumor growth, establishing long-term T immune response, immune heating of the microenvironment, de-repression of the anti-tumor immune phenotype, and sensitization to checkpoint blockade. The in situ vaccination protected from distant challenge tumors, both PSMA-positive and PSMA-negative, implying that it was addressed also to LLC1 tumor antigens. PSMA-retargeted oHSVs are a precision medicine tool worth being additionally investigated in the immunotherapeutic and in situ vaccination landscape against prostate cancers.
- Published
- 2021
25. oHSV Genome Editing by Means of galK Recombineering
- Author
-
Laura, Menotti, Valerio, Leoni, Valentina, Gatta, Biljana, Petrovic, Andrea, Vannini, Simona, Pepe, Tatiana, Gianni, and Gabriella, Campadelli-Fiume
- Subjects
Gene Editing ,Chromosomes, Artificial, Bacterial ,Galactokinase ,Mice ,Viral Proteins ,Escherichia coli Proteins ,Escherichia coli ,Animals ,Herpesvirus 1, Human - Abstract
Since the cloning of the herpes simplex virus (HSV) genome as BAC (bacterial artificial chromosome), the genetic engineering of the viral genome has become readily feasible. The advantage is that the modification of the animal virus genome is carried out in bacteria, with no replication or production of viral progeny, and is separated from the reconstitution or regeneration of the recombinant virus in mammalian cells. This allows an easy engineering of essential genes, as well. Many technologies have been developed for herpesvirus BAC engineering. In our hands the most powerful is galK recombineering that exploits a single marker (galK) for positive and negative selection and PCR amplicons for seamless modification in the desired genome locus. Here we describe the engineering of the HSV recombinant BAC 115 by the insertion of a heterologous cassette for the expression of murine interleukin 12 (mIL12) in the intergenic sequence between US1 and US2 ORFs.
- Published
- 2019
26. Rescue, Purification, and Characterization of a Recombinant HSV Expressing a Transgenic Protein
- Author
-
Andrea, Vannini, Biljana, Petrovic, Valentina, Gatta, Valerio, Leoni, Simona, Pepe, Laura, Menotti, Gabriella, Campadelli-Fiume, and Tatiana, Gianni
- Subjects
Recombination, Genetic ,Chromosomes, Artificial, Bacterial ,Mice ,Cell Line, Tumor ,Animals ,Gene Expression ,Herpesvirus 1, Human ,Interleukin-12 ,Recombinant Proteins - Abstract
In the previous chapter, we describe the engineering of a HSV-BAC genome by galK recombineering. Here we describe the procedures to reconstitute, or regenerate, the replicating recombinant virus, and the methods to purify it and characterize it for the correct expression of the transgene. We present the example of R-115, a recombinant expressing murine interleukin 12 (mIL12) from the US1-US2 intergenic region. A specific method for the production of highly purified virions by iodixanol gradient, suitable for in vivo applications, is also detailed.
- Published
- 2019
27. Correction for Vannini et al., αvβ3-integrin regulates PD-L1 expression and is involved in cancer immune evasion
- Author
-
Paolo Malatesta, Valerio Leoni, Andrea Vannini, Gabriella Campadelli-Fiume, Mara Sanapo, Catia Barboni, Tatiana Gianni, and Anna Zaghini
- Subjects
αvβ3 integrin ,Multidisciplinary ,cancer immunotherapy ,business.industry ,Cancer ,αvβ3-integrin ,Biological Sciences ,PD-L1 expression ,Evasion (ethics) ,medicine.disease ,Microbiology ,combination therapy ,Immune system ,PNAS Plus ,Cancer research ,Medicine ,Pd l1 expression ,business ,immune evasion - Abstract
Significance The PD-1/PD-L1 axis is a master player in the tumor immune evasion strategy. Checkpoint inhibitors, including anti–PD-1/PD-L1, are revolutionizing cancer immunotherapy. There is intense interest in dissecting their regulation and improving their application, mainly by combination therapies. The significance of the current findings is 2-fold. Our results suggest αvβ3-integrin as a critical regulator of PD-L1 expression and a key component of the tumor immune evasion machinery. Indeed, αvβ3-integrin depletion impairs tumor growth and elicits immunotherapeutic protection. Second, αvβ3-integrin blockade primes tumors for anti–PD-1 therapy and induces durable anticancer immune protection when combined with anti–PD-1 therapy. αvβ3-integrin is a readily druggable target that adds to the list of molecules suitable for combinatorial cancer immunotherapy., Tumors utilize a number of effective strategies, including the programmed death 1/PD ligand 1 (PD-1/PD-L1) axis, to evade immune-mediated control of their growth. PD-L1 expression is mainly induced by IFN receptor signaling or constitutively induced. Integrins are an abundantly expressed class of proteins which play multiple deleterious roles in cancer and exert proangiogenic and prosurvival activities. We asked whether αvβ3-integrin positively regulates PD-L1 expression and the anticancer immune response. We report that αvβ3-integrin regulated constitutive and IFN-induced PD-L1 expression in human and murine cancerous and noncancerous cells. αvβ3-integrin targeted STAT1 through its signaling C tail. The implantation of β3-integrin–depleted tumor cells led to a dramatic decrease in the growth of primary tumors, which exhibited reduced PD-L1 expression and became immunologically hot, with increased IFNγ content and CD8+ cell infiltration. In addition, the implantation of β3-integrin–depleted tumors elicited an abscopal immunotherapeutic effect measured as protection from the challenge tumor and durable splenocyte and serum reactivity to B16 cell antigens. These modifications to the immunosuppressive microenvironment primed cells for checkpoint (CP) blockade. When combined with anti–PD-1, β3-integrin depletion led to durable therapy and elicited an abscopal immunotherapeutic effect. We conclude that in addition to its previously known roles, αvβ3-integrin serves as a critical component of the cancer immune evasion strategy and can be an effective immunotherapy target.
- Published
- 2019
28. Eradication of glioblastoma by immuno-virotherapy with a retargeted oncolytic HSV in a preclinical model
- Author
-
Davide Ceresa, Gabriella Campadelli-Fiume, Elisa Avitabile, Laura Menotti, Paolo Malatesta, Irene Appolloni, Francesco Alessandrini, Daniela Marubbi, Alessandrini, Francesco, Menotti, Laura, Avitabile, Elisa, Appolloni, Irene, Ceresa, Davide, Marubbi, Daniela, Campadelli-Fiume, Gabriella, and Malatesta, Paolo
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Receptor, ErbB-2 ,medicine.medical_treatment ,Virulence ,Kaplan-Meier Estimate ,Biology ,herpes simplex, retargeting, glioblastoma, IL12, immuno-virotherapy ,medicine.disease_cause ,Interleukin-12 Subunit p35 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Simplexvirus ,Virotherapy ,Molecular Biology ,Oncolytic Virotherapy ,Mice, Inbred BALB C ,Brain Neoplasms ,Proto-Oncogene Proteins c-sis ,3. Good health ,Oncolytic virus ,Clinical trial ,Disease Models, Animal ,Oncolytic Viruses ,030104 developmental biology ,Herpes simplex virus ,Treatment Outcome ,Cell culture ,030220 oncology & carcinogenesis ,Mutation ,Interleukin 12 ,Cancer research ,Female ,Immunotherapy ,Neoplasm Recurrence, Local ,Glioblastoma - Abstract
Oncolytic herpes simplex viruses are proving to be effective in clinical trials against a number of cancers. Here, R-115, an oncolytic herpes simplex virus retargeted to human erbB-2, fully virulent in its target cells, and armed with murine interleukin-12 was evaluated in a murine model of glioblastoma. We show that a single R-115 injection in established tumors resulted, in about 30% of animals, in the complete eradication of the tumor, otherwise invariably lethal. The treatment also induced a significant improvement in the overall median survival time of mice and a resistance to recurrence from the same neoplasia. Such a high degree of protection was unprecedented; it was not observed before following treatments with the commonly used, mutated/attenuated oncolytic viruses. This is the first study providing the evidence of benefits offered by a fully virulent, retargeted, and armed herpes simplex virus in the treatment of glioblastoma and paves the way for clinical translation.
- Published
- 2019
29. HSV as A Platform for the Generation of Retargeted, Armed, and Reporter-Expressing Oncolytic Viruses
- Author
-
Biljana Petrovic, Valentina Gatta, Gabriella Campadelli-Fiume, Elisa Avitabile, Paolo Malatesta, Laura Menotti, Menotti, Laura, Avitabile, Elisa, Gatta, Valentina, Malatesta, Paolo, Petrovic, Biljana, and Campadelli-Fiume, Gabriella
- Subjects
0301 basic medicine ,Glutamate Carboxypeptidase II ,lcsh:QR1-502 ,retargeting, oncolytic herpesvirus, PSMA, EGFR, EGFRvIII, mIL12, Gaussia Luciferase, HER2 ,Gene Expression ,oncolytic herpesvirus ,Virus Replication ,lcsh:Microbiology ,law.invention ,Mice ,0302 clinical medicine ,law ,Genes, Reporter ,Chlorocebus aethiops ,Gene Order ,Simplexvirus ,Epidermal growth factor receptor ,Transgenes ,Interleukin-12 ,3. Good health ,ErbB Receptors ,Oncolytic Viruses ,Infectious Diseases ,030220 oncology & carcinogenesis ,Antigens, Surface ,Gene Targeting ,Recombinant DNA ,Genetic Engineering ,EGFRvIII ,EGFR ,Genetic Vectors ,Biology ,Article ,03 medical and health sciences ,Gaussia ,Gaussia Luciferase ,Virology ,Cell Line, Tumor ,HER2 ,PSMA ,Animals ,Humans ,mIL12 ,Luciferase ,oncolytic herpesviru ,Tropism ,retargeting ,biology.organism_classification ,Oncolytic virus ,Viral Tropism ,030104 developmental biology ,Viral replication ,Mutation ,Tissue tropism ,biology.protein - Abstract
Previously, we engineered oncolytic herpes simplex viruses (o-HSVs) retargeted to the HER2 (epidermal growth factor receptor 2) tumor cell specific receptor by the insertion of a single chain antibody (scFv) to HER2 in gD, gH, or gB. Here, the insertion of scFvs to three additional cancer targets&mdash, EGFR (epidermal growth factor receptor), EGFRvIII, and PSMA (prostate specific membrane antigen)&mdash, in gD &Delta, 6&ndash, 38 enabled the generation of specifically retargeted o-HSVs. Viable recombinants resulted from the insertion of an scFv in place of aa 6&ndash, 38, but not in place of aa 61&ndash, 218. Hence, only the gD N-terminus accepted all tested scFv inserts. Additionally, the insertion of mIL12 in the US1-US2 intergenic region of the HER2- or EGFRvIII-retargeted o-HSVs, and the further insertion of Gaussia Luciferase, gave rise to viable recombinants capable of secreting the cytokine and the reporter. Lastly, we engineered two known mutations in gB, they increased the ability of an HER2-retargeted recombinant to spread among murine cells. Altogether, current data show that the o-HSV carrying the aa 6&ndash, 38 deletion in gD serves as a platform for the specific retargeting of o-HSV tropism to a number of human cancer targets, and the retargeted o-HSVs serve as simultaneous vectors for two molecules.
- Published
- 2018
30. Spotlight on… Gabriella Campadelli-Fiume
- Author
-
Gabriella Campadelli-Fiume
- Subjects
0301 basic medicine ,Professional Development ,History ,MEDLINE ,Neoplasms therapy ,Historical Article ,Microbiology ,03 medical and health sciences ,030104 developmental biology ,Electrical current ,Portrait ,Genetics ,Profile Spotlight ,Molecular Biology ,Classics - Published
- 2018
31. Dual Ligand Insertion in gB and gD of Oncolytic Herpes Simplex Viruses for Retargeting to a Producer Vero Cell Line and to Cancer Cells
- Author
-
Gabriella Campadelli-Fiume, Andrea Vannini, Valentina Gatta, Biljana Petrovic, Anna Zaghini, Valerio Leoni, Petrovic, Biljana, Leoni, Valerio, Gatta, Valentina, Zaghini, Anna, Vannini, Andrea, and Campadelli-Fiume, Gabriella
- Subjects
0301 basic medicine ,gB ,gD ,viruses ,Immunology ,Mice, Transgenic ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,Mice ,Gene Delivery ,03 medical and health sciences ,HER2, HSV, retargeting, gD, gB, Vero, oncolytic virus ,Viral Envelope Proteins ,Cell Line, Tumor ,HER2 ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Vero Cells ,Tropism ,retargeting ,oncolytic virus ,Oncolytic Virotherapy ,HSV ,Cancer ,Neoplasms, Experimental ,medicine.disease ,Xenograft Model Antitumor Assays ,Vero ,3. Good health ,Oncolytic virus ,Virus Cultivation ,Oncolytic Viruses ,030104 developmental biology ,Herpes simplex virus ,Insect Science ,Cancer cell ,Vero cell ,Single-Chain Antibodies - Abstract
Oncolytic viruses gain cancer specificity in several ways. Like the majority of viruses, they grow better in cancer cells that are defective in mounting the host response to viruses. Often, they are attenuated by deletion or mutation of virulence genes that counteract the host response or are naturally occurring oncolytic mutants. In contrast, retargeted viruses are not attenuated or deleted; their cancer specificity rests on a modified, specific tropism for cancer receptors. For herpes simplex virus (HSV)-based oncolytics, the detargeting-retargeting strategies employed so far were based on genetic modifications of gD. Recently, we showed that even gH or gB can serve as retargeting tools. To enable the growth of retargeted HSVs in cells that can be used for clinical-grade virus production, a double-retargeting strategy has been developed. Here we show that several sites in the N terminus of gB are suitable to harbor the 20-amino-acid (aa)-long GCN4 peptide, which readdresses HSV tropism to Vero cells expressing the artificial GCN4 receptor and thus enables virus cultivation in the producer noncancer Vero-GCN4R cell line. The gB modifications can be combined with a minimal detargeting modification in gD, consisting in the deletion of two residues, aa 30 and 38, and replacement of aa 38 with the scFv to human epidermal growth factor receptor 2 (HER2), for retargeting to the cancer receptor. The panel of recombinants was analyzed comparatively in terms of virus growth, cell-to-cell spread, cytotoxicity, and in vivo antitumor efficacy to define the best double-retargeting strategy. IMPORTANCE There is increasing interest in oncolytic viruses, following FDA and the European Medicines Agency (EMA) approval of HSV Oncovex GM-CSF , and, mainly, because they greatly boost the immune response to the tumor and can be combined with immunotherapeutic agents, particularly checkpoint inhibitors. A strategy to gain cancer specificity and avoid virus attenuation is to retarget the virus tropism to cancer-specific receptors of choice. Cultivation of fully retargeted viruses is challenging, since they require cells that express the cancer receptor. We devised a strategy for their cultivation in producer noncancer Vero cell derivatives. Here, we developed a double-retargeting strategy, based on insertion of one ligand in gB for retargeting to a Vero cell derivative and of anti-HER2 ligand in gD for cancer retargeting. These modifications were combined with a minimally destructive detargeting strategy. This study and its companion paper explain the clinical-grade cultivation of retargeted oncolytic HSVs and promote their translation to the clinic.
- Published
- 2018
32. Simultaneous Insertion of Two Ligands in gD for Cultivation of Oncolytic Herpes Simplex Viruses in Noncancer Cells and Retargeting to Cancer Receptors
- Author
-
Valentina Gatta, Valerio Leoni, Tatiana Gianni, Gabriella Campadelli-Fiume, and Biljana Petrovic
- Subjects
0301 basic medicine ,gD ,Immunology ,Herpesvirus 1, Human ,Biology ,Microbiology ,Virus ,Gene Delivery ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Viral Envelope Proteins ,HER2 ,Virology ,Chlorocebus aethiops ,Animals ,Humans ,Receptor ,Vero Cells ,retargeting ,oncolytic virus ,HSV ,Vero ,3. Good health ,Oncolytic virus ,Oncolytic Viruses ,Viral Tropism ,030104 developmental biology ,030220 oncology & carcinogenesis ,Insect Science ,Cancer cell ,Retargeting ,Vero cell ,biology.protein ,Antibody ,Peptides - Abstract
Insertion of a single-chain variable-fragment antibody (scFv) to HER2 (human epidermal growth factor receptor 2) in gD, gH, or gB gives rise to herpes simplex viruses (HSVs) specifically retargeted to HER2-positive cancer cells, hence to highly specific nonattenuated oncolytic agents. Clinical-grade virus production cannot rely on cancer cells. Recently, we developed a double-retargeting strategy whereby gH carries the GCN4 peptide for retargeting to the noncancer producer Vero-GCN4R cell line and gD carries the scFv to HER2 for cancer retargeting. Here, we engineered double-retargeted recombinants, which carry both the GCN4 peptide and the scFv to HER2 in gD. Novel, more-advantageous detargeting strategies were devised so as to optimize the cultivation of the double-retargeted recombinants. Nectin1 detargeting was achieved by deletion of amino acids (aa) 35 to 39, 214 to 223, or 219 to 223 and replacement of the deleted sequences with one of the two ligands. The last two deletions were not attempted before. All recombinants exhibited the double retargeting to HER2 and to the Vero-GCN4R cells, as well as detargeting from the natural receptors HVEM and nectin1. Of note, some recombinants grew to higher yields than others. The best-performing recombinants carried a gD deletion as small as 5 amino acids and grew to titers similar to those exhibited by the singly retargeted R-LM113 and by the nonretargeted R-LM5. This study shows that double retargeting through insertion of two ligands in gD is feasible and, when combined with appropriate detargeting modifications, can result in recombinants highly effective in vitro and in vivo . IMPORTANCE There is increasing interest in oncolytic viruses following the FDA and European Medicines Agency (EMA) approval of the oncolytic HSV Oncovex GM-CSF and, mainly, because they greatly boost the immune response to the tumor and can be combined with immunotherapeutic agents, particularly immune checkpoint inhibitors. A strategy to gain high cancer specificity and avoid virus attenuation is to retarget the virus tropism to cancer-specific receptors of choice. However, cultivation of retargeted oncolytics in cells expressing the cancer receptor may not be approvable by regulatory agencies. We devised a strategy for their cultivation in noncancer cells. Here, we describe a double-retargeting strategy, based on the simultaneous insertion of two ligands in gD, one for retargeting to a producer, universal Vero cell derivative and one for retargeting to the HER2 cancer receptor. These insertions were combined with novel, minimally disadvantageous detargeting modifications. The current and accompanying studies indicate how to best achieve the clinical-grade cultivation of retargeted oncolytics.
- Published
- 2018
33. Systemic delivery of HER2-retargeted oncolytic-HSV by mesenchymal stromal cells protects from lung and brain metastases
- Author
-
Valentina Gatta, Pier Luigi Lollini, Dario Ranieri, Francesco Alviano, Patrizia Nanni, Martina Rossi, Arianna Palladini, Massimiliano Dall'Ora, Valentina Grosso, Giordano Nicoletti, Valerio Leoni, Gabriella Campadelli-Fiume, Laura Bonsi, Leoni, Valerio, Gatta, Valentina, Palladini, Arianna, Nicoletti, Giordano, Ranieri, Dario, Dall'Ora, Massimiliano, Grosso, Valentina, Rossi, Martina, Alviano, Francesco, Bonsi, Laura, Nanni, Patrizia, Lollini, Pier-Luigi, and Campadelli-Fiume, Gabriella
- Subjects
Simplexvirus ,Lung Neoplasms ,food.ingredient ,Receptor, ErbB-2 ,viruses ,Mice, Nude ,Breast Neoplasms ,Biology ,Mesenchymal Stem Cell Transplantation ,medicine.disease_cause ,Virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,food ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,metastases ,mesenchymal stem cell ,Tropism ,030304 developmental biology ,Oncolytic Virotherapy ,Ovarian Neoplasms ,viral retargeting ,mesenchymal stem cells ,0303 health sciences ,Brain Neoplasms ,Mesenchymal stem cell ,oncolytic herpes simplex viru ,oncolytic herpes simplex virus ,Flow Cytometry ,medicine.disease ,Xenograft Model Antitumor Assays ,Virology ,systemic delivery ,3. Good health ,Oncolytic virus ,Oncolytic Viruses ,Herpes simplex virus ,Oncology ,metastase ,030220 oncology & carcinogenesis ,Cancer cell ,Female ,Ovarian cancer ,Research Paper - Abstract
// Valerio Leoni 1,* , Valentina Gatta 1,* , Arianna Palladini 1 , Giordano Nicoletti 1 , Dario Ranieri 1 , Massimiliano Dall’Ora 1 , Valentina Grosso 1 , Martina Rossi 1 , Francesco Alviano 1 , Laura Bonsi 1 , Patrizia Nanni 1 , Pier-Luigi Lollini 1 and Gabriella Campadelli-Fiume 1 1 Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy * These authors have contributed equally to this work Correspondence to: Gabriella Campadelli-Fiume, email: // Pier-Luigi Lollini, email: // Keywords : oncolytic herpes simplex virus, viral retargeting, mesenchymal stem cells, systemic delivery, metastases Received : July 02, 2015 Accepted : September 04, 2015 Published : September 27, 2015 Abstract Fully retargeted oncolytic herpes simplex viruses (o-HSVs) gain cancer-specificity from redirection of tropism to cancer-specific receptors, and are non-attenuated. To overcome the hurdles of systemic delivery, and enable oncolytic viruses (o-viruses) to reach metastatic sites, carrier cells are being exploited. Mesenchymal stromal cells (MSCs) were never tested as carriers of retargeted o-viruses, given their scarse-null expression of the cancer-specific receptors. We report that MSCs from different sources can be forcedly infected with a HER2-retargeted oncolytic HSV. Progeny virus spread from MSCs to cancer cells in vitro and in vivo . We evaluated the organ distribution and therapeutic efficacy in two murine models of metastatic cancers, following a single i.v. injection of infected MSCs. As expected, the highest concentration of carrier-cells and of viral genomes was in the lungs. Viral genomes persisted throughout the body for at least two days. The growth of ovarian cancer lung metastases in nude mice was strongly inhibited, and the majority of treated mice appeared metastasis-free. The treatment significantly inhibited also breast cancer metastases to the brain in NSG mice, and reduced by more than one-half the metastatic burden in the brain.
- Published
- 2015
34. A Strategy for Cultivation of Retargeted Oncolytic Herpes Simplex Viruses in Non-cancer Cells
- Author
-
Biljana Petrovic, Alfredo Nicosia, Valentina Gatta, Costanza Casiraghi, Gabriella Campadelli-Fiume, Valerio Leoni, Leoni, Valerio, Gatta, Valentina, Casiraghi, Costanza, Nicosia, Alfredo, Petrovic, Biljana, Campadelli-Fiume, Gabriella, Leoni, V, Gatta, V, Casiraghi, C, Nicosia, A, Petrovic, B, and Campadelli-Fiume, G
- Subjects
0301 basic medicine ,Virus Cultivation ,Receptor, ErbB-2 ,viruses ,Immunology ,Herpesvirus 1, Human ,medicine.disease_cause ,Microbiology ,Cell Line ,03 medical and health sciences ,Gene Delivery ,Virology ,HER2 ,Oncolytic viruse ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Simplexvirus ,Vero Cells ,Tropism ,retargeting ,Oncolytic Virotherapy ,biology ,HSV ,3. Good health ,Oncolytic virus ,Vero ,Oncolytic Viruses ,Viral Tropism ,030104 developmental biology ,Herpes simplex virus ,Cell culture ,Insect Science ,Cancer cell ,Tissue tropism ,biology.protein ,Vero cell ,Antibody ,Genetic Engineering - Abstract
The oncolytic herpes simplex virus (HSV) that has been approved for clinical practice and those HSVs in clinical trials are attenuated viruses, often with the neurovirulence gene γ 1 34.5 and additional genes deleted. One strategy to engineer nonattenuated oncolytic HSVs consists of retargeting the viral tropism to a cancer-specific receptor of choice, exemplified by HER2 (human epidermal growth factor receptor 2), which is present in breast, ovary, and other cancers, and in detargeting from the natural receptors. Because the HER2-retargeted HSVs strictly depend on this receptor for infection, the viruses employed in preclinical studies were cultivated in HER2-positive cancer cells. The production of clinical-grade viruses destined for humans should avoid the use of cancer cells. Here, we engineered the R-213 recombinant, by insertion of a 20-amino-acid (aa) short peptide (named GCN4) in the gH of R-LM113; this recombinant was retargeted to HER2 through insertion in gD of a single-chain antibody (scFv) to HER2. Next, we generated a Vero cell line expressing an artificial receptor (GCN4R) whose N terminus consists of an scFv to GCN4 and therefore is capable of interacting with GCN4 present in gH of R-213. R-213 replicated as well as R-LM113 in SK-OV-3 cells, implying that addition of the GCN4 peptide was not detrimental to gH. R-213 grew to relatively high titers in Vero-GCN4R cells, efficiently spread from cell to cell, and killed both Vero-GCN4R and SK-OV-3 cells, as expected for an oncolytic virus. Altogether, Vero-GCN4R cells represent an efficient system for cultivation of retargeted oncolytic HSVs in non-cancer cells. IMPORTANCE There is growing interest in viruses as oncolytic agents, which can be administered in combination with immunotherapeutic compounds, including immune checkpoint inhibitors. The oncolytic HSV approved for clinical practice and those in clinical trials are attenuated viruses. An alternative to attenuation is a cancer specificity achieved by tropism retargeting to selected cancer receptors. However, the retargeted oncolytic HSVs strictly depend on cancer receptors for infection. Here, we devised a strategy for in vitro cultivation of retargeted HSVs in non-cancer cells. The strategy envisions a double-retargeting approach: one retargeting is via gD to the cancer receptor, and the second retargeting is via gH to an artificial receptor expressed in Vero cells. The double-retargeted HSV uses alternatively the two receptors to infect cancer cells or producer cells. A universal non-cancer cell line for growth of clinical-grade retargeted HSVs represents a step forward in the translational phase.
- Published
- 2017
35. Type I Interferon and NF-κB Activation Elicited by Herpes Simplex Virus gH/gL via αvβ3 Integrin in Epithelial and Neuronal Cell Lines
- Author
-
Tatiana Gianni, Gabriella Campadelli-Fiume, Valerio Leoni, T. Gianni, V. Leoni, and G. Campadelli-Fiume
- Subjects
viruses ,Immunology ,Cellular Response to Infection ,Herpesvirus 1, Human ,Integrin alpha5 ,Biology ,medicine.disease_cause ,DENDRITIC CELLS ,Microbiology ,Cell Line ,Viral Proteins ,Viral Envelope Proteins ,Interferon ,Viral entry ,Virology ,medicine ,TLR2 ,Humans ,Receptor ,Neurons ,Membrane Glycoproteins ,αvβ3 integrin in epithelial and neuronal cell line ,TOLL-LIKE RECEPTOR-2 ,Contraindications ,Integrin beta3 ,NF-kappa B ,Epithelial Cells ,Herpes Simplex ,Herpesvirus glycoprotein B ,Molecular biology ,infection ,POLIOVIRUS RECEPTOR ,LIPID RAFTS ,Herpes simplex virus ,Cell culture ,Insect Science ,ENTRY ,REPLICATION ,Interferon Type I ,recognition ,protein ,Interferon type I ,Molecular Chaperones ,medicine.drug - Abstract
αvβ3 integrin represents a novel sensing system which detects herpes simplex virus (HSV) and bacterial constituents. In cooperation with Toll-like receptor 2 (TLR2), it elicits an innate response that leads to activation of type I interferon (IFN), NF-κB, and a specific set of cytokines. We report that this defensive branch is functional in cells which represent experimental models of epithelial, including keratinocytic, and neuronal cells. These are the major targets of HSV in vivo . HSV entered the three cell lines via distinct routes. Hence, the defensive response was independent of the route of virus entry. Soluble gH/gL sufficed to elicit type I IFN and NF-κB activation and represents the viral pathogen-associated molecular pattern (PAMP) of this defense system.
- Published
- 2013
36. Integrins as Herpesvirus Receptors and Mediators of the Host Signalosome
- Author
-
Donna Collins-McMillen, Tatiana Gianni, Andrew D. Yurochko, Gabriella Campadelli-Fiume, and DIPARTIMENTO DI MEDICINA SPECIALISTICA, DIAGNOSTICA E SPERIMENTALE
- Subjects
0301 basic medicine ,Herpesvirus 4, Human ,Integrins ,viruses ,Integrin ,Cytomegalovirus ,Virus Attachment ,KSHV ,medicine.disease_cause ,Herpesviridae ,Receptor tyrosine kinase ,03 medical and health sciences ,Species Specificity ,Viral entry ,EBV ,Virology ,medicine ,Humans ,Simplexvirus ,HCMV ,HSV ,Signal transduction ,Receptor ,biology ,Virus Internalization ,Herpesvirus glycoprotein B ,Endocytosis ,3. Good health ,Cell biology ,030104 developmental biology ,Herpes simplex virus ,Herpesvirus 8, Human ,biology.protein ,Receptors, Virus ,Signal Transduction - Abstract
none 4 si NIH National Institute of General Medical Sciences (NIGMS), NIH National Institute of Allergy & Infectious Diseases (NIAID), NIH National Institute of General Medical Sciences (NIGMS), NIH National Institute of Allergy & Infectious Diseases (NIAID) The repertoire of herpesvirus receptors consists of nonintegrin and integrin molecules. Integrins interact with the conserved glycoproteins gH/gL or gB. This interaction is a conserved biology across the Herpesviridae family, likely directed to promote virus entry and endocytosis. Herpesviruses exploit this interaction to execute a range of critical functions that include (a) relocation of nonintegrin receptors (e.g., herpes simplex virus nectin1 and Kaposi's sarcoma-associated herpesvirus EphA2), or association with nonintegrin receptors (i.e., human cytomegalovirus EGFR), to dictate species-specific entry pathways; (b) activation of multiple signaling pathways (e.g., Ca2+ release, c-Src, FAK, MAPK, and PI3K); and (c) association with Rho GTPases, tyrosine kinase receptors, Toll-like receptors, which result in cytoskeletal remodeling, differential cell type targeting, and innate responses. In turn, integrins can be modulated by viral proteins (e.g., Epstein-Barr virus LMPs) to favor spread of transformed cells. We propose that herpesviruses evolved a multipartite entry system to allow interaction with multiple receptors, including integrins, required for their sophisticated life cycle. mixed Campadelli, MARIA GABRIELLA; Collins Mcmillen, Donna; Gianni, Tatiana; Yurochko, Andrew D. Campadelli, MARIA GABRIELLA; Collins Mcmillen, Donna; Gianni, Tatiana; Yurochko, Andrew D.
- Published
- 2016
37. Retargeting Strategies for Oncolytic Herpes Simplex Viruses
- Author
-
Tatiana Gianni, Elisa Avitabile, Costanza Casiraghi, Gabriella Campadelli-Fiume, Valerio Leoni, Biljana Petrovic, Valentina Gatta, Campadelli, MARIA GABRIELLA, Petrovic, Biljana, Leoni, Valerio, Gianni, Tatiana, Avitabile, Elisa, Casiraghi, Costanza, and Gatta, Valentina
- Subjects
0301 basic medicine ,Receptor, ErbB-2 ,Infectious Disease ,Review ,Biology ,Virus ,03 medical and health sciences ,Virology ,Simplexvirus ,Receptor ,Tropism ,retargeting ,Viral Structural Proteins ,oncolytic HSV ,Virus Internalization ,Recombinant Proteins ,3. Good health ,Oncolytic virus ,Oncolytic Viruses ,Viral Tropism ,030104 developmental biology ,Infectious Diseases ,Apoptosis ,HER-2 ,Retargeting ,Cancer cell ,Cancer research ,Tissue tropism ,Receptors, Virus ,Single-Chain Antibodies - Abstract
Most of the oncolytic herpes simplex viruses (HSVs) exhibit a high safety profile achieved through attenuation. They carry defects in virulence proteins that antagonize host cell response to the virus, including innate response, apoptosis, authophagy, and depend on tumor cell proliferation. They grow robustly in cancer cells, provided that these are deficient in host cell responses, which is often the case. To overcome the attenuation limits, a strategy is to render the virus highly cancer-specific, e.g., by retargeting their tropism to cancer-specific receptors, and detargeting from natural receptors. The target we selected is HER-2, overexpressed in breast, ovarian and other cancers. Entry of wt-HSV requires the essential glycoproteins gD, gH/gL and gB. Here, we reviewed that oncolytic HSV retargeting was achieved through modifications in gD: the addition of a single-chain antibody (scFv) to HER-2 coupled with appropriate deletions to remove part of the natural receptors' binding sites. Recently, we showed that also gH/gL can be a retargeting tool. The insertion of an scFv to HER-2 at the gH N-terminus, coupled with deletions in gD, led to a recombinant capable to use HER-2 as the sole receptor. The retargeted oncolytic HSVs can be administered systemically by means of carrier cells-forcedly-infected mesenchymal stem cells. Altogether, the retargeted oncolytic HSVs are highly cancer-specific and their replication is not dependent on intrinsic defects of the tumor cells. They might be further modified to express immunomodulatory molecules.
- Published
- 2016
38. αvβ3-integrin is a major sensor and activator of innate immunity to herpes simplex virus-1
- Author
-
Liudmila S. Chesnokova, Tatiana Gianni, Gabriella Campadelli-Fiume, Lindsey M. Hutt-Fletcher, Valerio Leoni, Gianni T., Leoni V., Chesnokova L.S., Hutt-Fletcher L.M., and Campadelli-Fiume G.
- Subjects
Gene Expression ,Syk ,Enzyme-Linked Immunosorbent Assay ,Herpesvirus 1, Human ,Biology ,Virus Replication ,medicine.disease_cause ,Polymerase Chain Reaction ,Cell Line ,Innate/physiology ,medicine ,Gene Silencing ,Receptor ,Human/immunology ,Multidisciplinary ,Innate immune system ,Herpesvirus 1 ,NF-kappa B ,Cytokines/metabolism ,Immunity ,Pattern recognition receptor ,HUMANS ,Biological Sciences ,Integrin alphaVbeta3/physiology ,Integrin alphaVbeta3 ,Virology ,Immunity, Innate ,Cell biology ,TLR2 ,Herpes simplex virus ,NF-kappa B/metabolism ,Human/physiology ,Cytokines ,Proto-oncogene tyrosine-protein kinase Src ,Interferon regulatory factors - Abstract
Pathogens are sensed by Toll-like receptors (TLRs) and a growing number of non-TLR receptors. Integrins constitute a family of signaling receptors exploited by viruses and bacteria to access cells. By gain- and loss-of-function approaches we found that αvβ3-integrin is a sensor of and plays a crucial role in the innate defense against herpes simplex virus (HSV). αvβ3-integrin signaled through two pathways. One concurred with TLR2, affected activation/induction of interferons type 1 (IFNs-1), NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), and a polarized set of cytokines and receptors. The virion glycoproteins gH/gL sufficed to induce IFN1 and NF-κB via this pathway. The other pathway was TLR2-independent, involved sarcoma (SRC)-spleen tyrosine kinase (SYK)-Caspase recruitment domain-containing protein 9 (CARD9)-TRIF (TIR-domain-containing adapter-inducing interferon-β), and affected interferon regulatory factor 3 and 7 (IRF3-IRF7). The importance of αvβ3-integrin-mediated defense is reflected in the observation that HSV evolved the immediate-early infected cellular protein 0 (ICP0) protein to counteract it. We propose that αvβ3-integrin is considered a class of non-TLR pattern recognition receptors, a role likely exerted toward viruses and bacteria that interact with integrins and mount an innate response.
- Published
- 2012
39. αvβ3 Integrin Boosts the Innate Immune Response Elicited in Epithelial Cells through Plasma Membrane and Endosomal Toll-Like Receptors
- Author
-
Tatiana Gianni, Costanza Casiraghi, Gabriella Campadelli-Fiume, Casiraghi, Costanza, Gianni, Tatiana, and Campadelli-Fiume, Gabriella
- Subjects
0301 basic medicine ,Integrin ,Immunology ,Cellular Response to Infection ,Endosomes ,Microbiology ,Collagen receptor ,03 medical and health sciences ,HEK293 Cell ,Virology ,Endosome ,Humans ,Simplexvirus ,Gene Silencing ,Simplexviru ,Integrin alphaVbeta3 ,Epithelial Cell ,Innate immune system ,030102 biochemistry & molecular biology ,biology ,Cell Membrane ,NF-kappa B ,Epithelial Cells ,Immunity, Innate ,Cell biology ,Toll-Like Receptor 3 ,030104 developmental biology ,HEK293 Cells ,Integrin alpha M ,Insect Science ,TLR3 ,biology.protein ,Integrin, beta 6 ,Signal transduction ,Human ,Signal Transduction - Abstract
We report that αvβ3 integrin strongly affects the innate immune response in epithelial cells. αvβ3 integrin greatly increased the response elicited via plasma membrane Toll-like receptors (TLRs) by herpes simplex virus or bacterial ligands. The endosomal TLR3, not the cytosolic sensor interferon gamma-inducible protein 16 (IFI16), was also boosted by αvβ3 integrin. The boosting was exerted specifically by αvβ3 integrin but not by αvβ6 or αvβ8 integrin. Current and previous work indicates that integrin-TLR cooperation occurs in epithelial and monocytic cells. The TLR response should be considered an integrin-TLR response.
- Published
- 2015
40. Rethinking herpes simplex virus: the way to oncolytic agents
- Author
-
Patrizia Nanni, CARLA DE GIOVANNI, VALENTINA GATTA, Gabriella Campadelli-Fiume, LAURA MENOTTI, Valentina GATTA, and Pier Luigi Lollini
- Subjects
education.field_of_study ,biology ,viruses ,Population ,medicine.disease_cause ,Protein kinase R ,Virology ,Oncolytic virus ,Infectious Diseases ,Herpes simplex virus ,Immune system ,Tissue tropism ,medicine ,biology.protein ,Antibody ,education ,Tropism - Abstract
Oncolytic viruses infect, replicate in and kill cancer cells. HSV has emerged as a most promising candidate because it exerts a generally moderate pathogenicity in humans; it is amenable to attenuation and tropism retargeting; the ample genome provides space for heterologous genes; specific antiviral therapy is available in a worst case scenario. The first strategy to convert HSV into an oncolytic agent consisted in deletion of the γ(1) 34.5 gene which counteracts the protein kinase R (PKR) response, and of the UL39 gene which encodes the large ribonucleotide reductase subunit. Tumor specificity resided in low PKR activity, and high deoxyribonucleotides content of cancer cells. These highly attenuated viruses have been and presently are in clinical trials with encouraging results. The preferred route of administration has been intratumor or in tissues adjacent to resected tumors. Although the general population has a high seroprevalence of antibodies to HSV, studies in animals and humans demonstrate that prior immunity is not an obstacle to systemic routes of administration, and that oncolytic HSV (o-HSVs) do populate tumors. As the attenuated viruses undergo clinical experimentation, the research pipeline is developing novel, more potent and highly tumor-specific o-HSVs. These include viruses which overcome tumor heterogeneity in PKR level by insertion of anti-PKR genes, viruses which reinforce the host tumor clearance capacity by encoding immune cytokines (IL-12 or granulocyte-macrophage colony-stimulating factor), and non-attenuated viruses fully retargeted to tumor specific receptors. A strategy to generate o-HSVs fully retargeted to human epidermal growth factor receptor-2 (HER-2) or other cancer-specific surface receptors is detailed.
- Published
- 2011
41. A fully-virulent retargeted oncolytic HSV armed with IL-12 elicits local immunity and vaccine therapy towards distant tumors
- Author
-
Catia Barboni, Mara Sanapo, Gabriella Campadelli-Fiume, Pier Luigi Lollini, Valentina Gatta, Anna Zaghini, Valerio Leoni, Julie Rambaldi, Costanza Casiraghi, Patrizia Nanni, Andrea Vannini, Leoni, Valerio, Vannini, Andrea, Gatta, Valentina, Rambaldi, Julie, Sanapo, Mara, Barboni, Catia, Zaghini, Anna, Nanni, Patrizia, Lollini, Pier-Luigi, Casiraghi, Costanza, and Campadelli-Fiume, Gabriella
- Subjects
0301 basic medicine ,Physiology ,medicine.medical_treatment ,Cancer Treatment ,Carcinoma, Lewis Lung ,Mice ,Cancer immunotherapy ,Immune Physiology ,Medicine and Health Sciences ,Simplexvirus ,Immune Response ,lcsh:QH301-705.5 ,Oncolytic Virotherapy ,Mammalian Genomics ,HSV ,Animal Models ,Genomics ,Interleukin-12 ,3. Good health ,Oncolytic Viruses ,Oncology ,Experimental Organism Systems ,oncolytic ,Interleukin 12 ,Research Article ,lcsh:Immunologic diseases. Allergy ,Immunology ,Mouse Models ,Antineoplastic Agents ,Research and Analysis Methods ,Cancer Vaccines ,Microbiology ,03 medical and health sciences ,Model Organisms ,Immune system ,Virology ,Genetics ,medicine ,Animals ,Molecular Biology Techniques ,Molecular Biology ,Tumor microenvironment ,business.industry ,Biology and Life Sciences ,Immunotherapy, Active ,Immunotherapy ,Viral Replication ,Vaccine therapy ,Oncolytic virus ,030104 developmental biology ,lcsh:Biology (General) ,Animal Genomics ,HER-2 ,virus retargeting ,Cancer cell ,Cancer research ,interleukin 12 ,Parasitology ,lcsh:RC581-607 ,business ,Spleen ,Cloning - Abstract
Oncolytic herpes simplex viruses (oHSVs) showed efficacy in clinical trials and practice. Most of them gain cancer-specificity from deletions/mutations in genes that counteract the host response, and grow selectively in cancer cells defective in anti-viral response. Because of the deletions/mutations, they are frequently attenuated or over-attenuated. We developed next-generation oHSVs, which carry no deletion/mutation, gain cancer-specificity from specific retargeting to tumor cell receptors—e.g. HER2 (human epidermal growth factor receptor 2)—hence are fully-virulent in the targeted cancer cells. The type of immunotherapy they elicit was not predictable, since non-attenuated HSVs induce and then dampen the innate response, whereas deleted/attenuated viruses fail to contrast it, and since the retargeted oHSVs replicate efficiently in tumor cells, but spare other cells in the tumor. We report on the first efficacy study of HER2-retargeted, fully-virulent oHSVs in immunocompetent mice. Their safety profile was very high. Both the unarmed R-LM113 and the IL-12-armed R-115 inhibited the growth of the primary HER2-Lewis lung carcinoma-1 (HER2-LLC1) tumor, R-115 being constantly more efficacious. All the mice that did not die because of the primary treated tumors, were protected from the growth of contralateral untreated tumors. The long-term survivors were protected from a second contralateral tumor, providing additional evidence for an abscopal immunotherapeutic effect. Analysis of the local response highlighted that particularly R-115 unleashed the immunosuppressive tumor microenvironment, i.e. induced immunomodulatory cytokines, including IFNγ, T-bet which promoted Th1 polarization. Some of the tumor infiltrating cells, e.g. CD4+, CD335+ cells were increased in the tumors of all responders mice, irrespective of which virus was employed, whereas CD8+, Foxp3+, CD141+ were increased and CD11b+ cells were decreased preferentially in R-115-treated mice. The durable response included a breakage of tolerance towards both HER2 and the wt tumor cells, and underscored a systemic immunotherapeutic vaccine response., Author summary There is increasing interest in oncolytic viruses (OVs), following the approval of OncovexGM-CSF, and the success of a number of them in clinical trials. Most OVs, particularly the oHSVs, are attenuated to varying degree. In contrast, the tropism-retargeted oHSVs are fully-virulent, highly effective oncolytic agents, and appear to be highly safe in mice. Up to now, it was unknown how efficacious the retargeted oHSVs are as immunotherapeutic agents. Here, the demonstration that they elicit local immune response and systemic therapy vaccine effects opens the possibility that the fully-virulent retargeted oHSVs may be highly efficacious oncolytic-immunotherapeutic agents.
- Published
- 2018
42. Updates and achievements in virology
- Author
-
Gabriella Campadelli-Fiume, Giorgio Palù, Carlo De Giuli Morghen, and Franco M. Buonaguro
- Subjects
Pharmacology ,Medal ,Biomedical Research ,Bacteria ,business.industry ,Archaea ,Eukaryota ,Humans ,Italy ,Virology ,Virus Diseases ,Immunology ,Drug Discovery3003 Pharmaceutical Science ,Molecular Medicine ,Virus diseases ,Drug Discovery ,Medicine ,business ,Clinical virology - Abstract
The 4th European Congress of Virology, hosted by the Italian Society for Virology, attracted approximately 1300 scientists from 46 countries worldwide. It also represented the first conference of the European Society for Virology, which was established in Campidoglio, Rome, Italy in 2009. The main goal of the meeting was to share research activities and results achieved in European virology units/institutes and to strengthen collaboration with colleagues from both western and developing countries. The worldwide representation of participants is a testament to the strength and attraction of European virology. The 5-day conference brought together the best of current virology; topics covered all three living domains (bacteria, archaea and eucarya), with special sessions on plant and veterinary virology as well as human virology, including two oral presentations on mimiviruses. The conference included five plenary sessions, 31 workshops, one hepatitis C virus roundtable, ten special workshops and three poster sessions, as well as 45 keynote lectures, 191 oral presentations and 845 abstracts. Furthermore, the Gesellschaft fur Virologie Loeffler-Frosch medal award was given to Peter Vogt for his long-standing career and achievements; the Gardner Lecture of the European Society for Clinical Virology was presented by Yoshihiro Kawaoka, and the Pioneer in Virology Lecture of the Italian Society for Virology was presented by Ulrich Koszinowski.
- Published
- 2010
43. Glycoprotein D of Bovine Herpesvirus 5 (BoHV-5) Confers an Extended Host Range to BoHV-1 but Does Not Contribute to Invasion of the Brain
- Author
-
Claudia Senn, Kurt Tobler, Marco Franchini, Carlos Abril, Gabriella Campadelli-Fiume, Cornel Fraefel, Mathias Ackermann, Monika Hilbe, Evgeni Gabev, University of Zurich, Ackermann, M, Gabev E., Tobler K., Abril C., Hilbe M., Senn C., Franchini M., Campadelli-Fiume G., Fraefel C., and Ackermann M.
- Subjects
1109 Insect Science ,Nectins ,Immunology ,10184 Institute of Veterinary Pathology ,Virulence ,Microbiology ,Virulence factor ,Virus ,Mice ,Viral Envelope Proteins ,In vivo ,Virology ,Animals ,Herpesvirus 1, Bovine ,2403 Immunology ,Herpesvirus 5, Bovine ,biology ,2404 Microbiology ,Brain ,biology.organism_classification ,Phenotype ,Bovine herpesvirus 1 ,Bovine herpesvirus 5 ,Viral replication ,Insect Science ,Host-Pathogen Interactions ,2406 Virology ,570 Life sciences ,Pathogenesis and Immunity ,Cattle ,Nervous System Diseases ,Cell Adhesion Molecules ,10244 Institute of Virology ,Protein Binding - Abstract
Bovine herpesvirus 1 (BoHV-1) and BoHV-5 are closely related pathogens of cattle, but only BoHV-5 is considered a neuropathogen. We engineered intertypic gD exchange mutants with BoHV-1 and BoHV-5 backbones in order to address their in vitro and in vivo host ranges, with particular interest in invasion of the brain. The new viruses replicated in cell culture with similar dynamics and to titers comparable to those of their wild-type parents. However, gD of BoHV-5 (gD5) was able to interact with a surprisingly broad range of nectins. In vivo , gD5 provided a virulent phenotype to BoHV-1 in AR129 mice, featuring a high incidence of neurological symptoms and early onset of disease. However, only virus with the BoHV-5 backbone, independent of the gD type, was detected in the brain by immunohistology. Thus, gD of BoHV-5 confers an extended cellular host range to BoHV-1 and may be considered a virulence factor but does not contribute to the invasion of the brain.
- Published
- 2010
44. Construction of a Fully Retargeted Herpes Simplex Virus 1 Recombinant Capable of Entering Cells Solely via Human Epidermal Growth Factor Receptor 2
- Author
-
Gabriella Campadelli-Fiume, Laura Menotti, Arianna Cerretani, Hartmut Hengel, Menotti L., Cerretani A., Hengel H., and Campadelli-Fiume G.
- Subjects
Receptor, ErbB-2 ,Nectins ,Immunology ,Antineoplastic Agents ,Herpesvirus 1, Human ,Antibodies, Monoclonal, Humanized ,Protein Engineering ,medicine.disease_cause ,THERAPY ,Tropism ,Microbiology ,Virus ,Cell Line ,law.invention ,Gene Delivery ,law ,Neoplasms ,Virology ,medicine ,VIRUS GENETIC ENGINEERING ,Animals ,Humans ,Point Mutation ,Epidermal growth factor receptor ,Receptor ,HER2 - ERBB2 RECEPTOR ,biology ,Antibodies, Monoclonal ,Herpes Simplex ,Trastuzumab ,CANCER ,Oncolytic virus ,Herpes simplex virus ,Cell culture ,Insect Science ,Recombinant DNA ,Cancer research ,biology.protein ,Antibody ,Cell Adhesion Molecules ,Receptors, Tumor Necrosis Factor, Member 14 ,HERPES SIMPLEX VIRUS - Abstract
A novel frontier in the treatment of tumors that are difficult to treat is oncolytic virotherapy, in which a replication-competent virus selectively infects and destroys tumor cells. Herpes simplex virus (HSV) represents a particularly attractive system. Effective retargeting to tumor-specific receptors has been achieved by insertion in gD of heterologous ligands. Previously, our laboratory generated an HSV retargeted to human epidermal growth factor receptor 2 (HER2), a receptor overexpressed in about one-third of mammary tumors and in some ovarian tumors. HER2 overexpression correlates with increased metastaticity and poor prognosis. Because HER2 has no natural ligand, the inserted ligand was a single-chain antibody to HER2. The objective of this work was to genetically engineer an HSV that selectively targets the HER2-expressing tumor cells and that has lost the ability to enter cells through the natural gD receptors, HVEM and nectin1. Detargeting from nectin1 was attempted by two different strategies, point mutations and insertion of the single-chain antibody at a site in gD different from previously described sites of insertion. We report that point mutations at gD amino acids 34, 215, 222, and 223 failed to generate a nectin1-detargeted HSV. An HSV simultaneously detargeted from nectin1 and HVEM and retargeted to HER2 was successfully engineered by moving the site of single-chain antibody insertion at residue 39, i.e., in front of the nectin1-interacting surface and not lateral to it, and by deleting amino acid residues 6 to 38. The resulting recombinant, R-LM113, entered cells and spread from cell to cell solely via HER2.
- Published
- 2008
45. Intracellular Trafficking and Maturation of Herpes Simplex Virus Type 1 gB and Virus Egress Require Functional Biogenesis of Multivesicular Bodies
- Author
-
Arianna Calistri, Cristina Forghieri, Heinrich G. Göttlinger, Cristiano Salata, Paola Sette, Cristina Parolin, Enrico Cancellotti, Giorgio Palù, Alessandra Comin, and Gabriella Campadelli-Fiume
- Subjects
viruses ,Immunology ,Vesicular Transport Proteins ,Herpesvirus 1, Human ,macromolecular substances ,GLYCOPROTEIN-B ,VIRION ENVELOPE ,HSV-1 ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,Viral Envelope Proteins ,Ubiquitin ,Virology ,medicine ,Animals ,Humans ,chemistry.chemical_classification ,Endosomal Sorting Complexes Required for Transport ,biology ,Virus Assembly ,Virion ,Virus-Cell Interactions ,Cell biology ,Transport protein ,Protein Transport ,Herpes simplex virus ,chemistry ,Cytoplasm ,Insect Science ,biology.protein ,Glycoprotein ,Intracellular ,Biogenesis - Abstract
The biogenesis of multivesicular bodies (MVBs) is topologically equivalent to virion budding. Hence, a number of viruses exploit the MVB pathway to build their envelope and exit from the cell. By expression of dominant negative forms of Vps4 and Vps24, two components of the MVB pathway, we observed an impairment in infectious herpes simplex virus (HSV) assembly/egress, in agreement with a recent report showing the involvement in HSV envelopment of Vps4, the MVB-specific ATPase (C. M. Crump, C. Yates, and T. Minson, J. Virol. 81:7380-7387). Furthermore, HSV infection resulted in morphological changes to MVBs. Glycoprotein B (gB), one of the most highly conserved glycoproteins across theHerpesviridaefamily, was sorted to MVB membranes. In cells expressing the dominant negative form of Vps4, the site of intracellular gB accumulation was altered; part of gB accumulated as an endoglycosidase H-sensitive immature form at a calreticulin-positive compartment, indicating that gB traffic was dependent on a functional MVB pathway. gB was ubiquitinated in both infected and transfected cells. Ubiquitination was in part dependent on ubiquitin lysine 63, a signal for cargo sorting to MVBs. Partial deletion of the gB cytoplasmic tail resulted in a dramatic reduction of ubiquitination, as well as of progeny virus assembly and release to the extracellular compartment. Thus, HSV envelopment/egress and gB intracellular trafficking are dependent on functional MVB biogenesis. Our data support the view that the sorting of gB to MVB membranes may represent a critical step in HSV envelopment and egress and that modified MVB membranes constitute a platform for HSV cytoplasmic envelopment or that MVB components are recruited to the site(s) of envelopment.
- Published
- 2007
46. αvβ6- and αvβ8-integrins serve as interchangeable receptors for HSV gH/gL to promote endocytosis and activation of membrane fusion
- Author
-
Gabriella Campadelli-Fiume, Tatiana Gianni, Liudmila S. Chesnokova, Lindsey M. Hutt-Fletcher, Stefano Salvioli, Tatiana Gianni, Stefano Salvioli, Liudmila S. Chesnokova, Lindsey M. Hutt-Fletcher, and Gabriella Campadelli-Fiume
- Subjects
lcsh:Immunologic diseases. Allergy ,Integrins ,Endosome ,viruses ,membrane fusion ,Immunology ,Integrin ,Spodoptera ,Endocytosis ,medicine.disease_cause ,Microbiology ,Viral Envelope Proteins ,Antigens, Neoplasm ,Virology ,Genetics ,medicine ,Sf9 Cells ,Animals ,Humans ,Simplexvirus ,Receptor ,lcsh:QH301-705.5 ,Molecular Biology ,Cells, Cultured ,Dynamin ,biology ,Lipid bilayer fusion ,Virus Internalization ,Molecular biology ,Herpesvirus glycoprotein B ,Herpes simplex virus ,HEK293 Cells ,lcsh:Biology (General) ,biology.protein ,Receptors, Virus ,αvβ6- and αvβ8-integrin ,Parasitology ,lcsh:RC581-607 ,K562 Cells ,receptors for HSV gH/gL ,HERPES SIMPLEX VIRUS ,HeLa Cells ,Research Article - Abstract
Herpes simplex virus (HSV) - and herpesviruses in general - encode for a multipartite entry/fusion apparatus. In HSV it consists of the HSV-specific glycoprotein D (gD), and three additional glycoproteins, gH/gL and gB, conserved across the Herpesviridae family and responsible for the execution of fusion. According to the current model, upon receptor binding, gD propagates the activation to gH/gL and to gB in a cascade fashion. Questions remain about how the cascade of activation is controlled and how it is synchronized with virion endocytosis, to avoid premature activation and exhaustion of the glycoproteins. We considered the possibility that such control might be carried out by as yet unknown receptors. Indeed, receptors for HSV gB, but not for gH/gL, have been described. In other members of the Herpesviridae family, such as Epstein-Barr virus, integrin receptors bind gH/gL and trigger conformational changes in the glycoproteins. We report that αvβ6- and αvβ8-integrins serve as receptors for HSV entry into experimental models of keratinocytes and other epithelial and neuronal cells. Evidence rests on loss of function experiments, in which integrins were blocked by antibodies or silenced, and gain of function experiments in which αvβ6-integrin was expressed in integrin-negative cells. αvβ6- and αvβ8-integrins acted independently and are thus interchangeable. Both bind gH/gL with high affinity. The interaction profoundly affects the route of HSV entry and directs the virus to acidic endosomes. In the case of αvβ8, but not αvβ6-integrin, the portal of entry is located at lipid microdomains and requires dynamin 2. Thus, a major role of αvβ6- or αvβ8-integrin in HSV infection appears to be to function as gH/gL receptors and to promote virus endocytosis. We propose that placing the gH/gL activation under the integrin trigger point enables HSV to synchronize virion endocytosis with the cascade of glycoprotein activation that culminates in execution of fusion., Author Summary In order to infect their hosts and cause disease, viruses must enter their host cells. The human pathogen herpes simplex virus (HSV) - and herpesviruses in general - are equipped with a complex, multipartite entry apparatus, made of four glycoproteins – gD, gH/gL, gB. These glycoproteins must be activated in a timely, coordinated manner. According to the current model, the flux of activation goes from receptor-bound gD, to gH/gL and gB. The premature activation, and hence exhaustion of the glycoproteins must also be prevented. We report on a checkpoint at the gH/gL level. Specifically, αvβ6- and αvβ8-integrins serve as receptors for HSV entry into keratinocytes and other epithelial and neuronal cells. Both bind gH/gL with high affinity. The interaction profoundly affects the pathway of HSV entry, promoting HSV endocytosis into acidic endosomes. For αvβ8-integrin, the portal of entry is at lipid microdomains and requires dynamin 2. We propose that, by placing the activation of gH/gL under control of an integrin trigger point, HSV can synchronize virion endocytosis with the cascade of activation that culminates in the execution of fusion between the virion envelope and cellular membranes.
- Published
- 2013
47. Dissociation of HSV gL from gH by αvβ6- or αvβ8-integrin promotes gH activation and virus entry
- Author
-
Tatiana Gianni, Gabriella Campadelli-Fiume, Raffaele Massaro, Gianni, T., Massaro, R., and Campadelli-Fiume, G.
- Subjects
glycoprotein ,Integrins ,viruses ,Amino Acid Motifs ,Nectins ,Integrin ,Herpesvirus 1, Human ,gL ,virus entry ,Biology ,medicine.disease_cause ,Models, Biological ,Herpesviridae ,gH ,Viral Envelope Proteins ,Antigens, Neoplasm ,Viral entry ,Cell Line, Tumor ,medicine ,Humans ,Receptor ,chemistry.chemical_classification ,Brefeldin A ,Multidisciplinary ,Cell adhesion molecule ,Cell Membrane ,Virion ,Antibodies, Monoclonal ,Epithelial Cells ,Herpes Simplex ,Virus Internalization ,herpes simplex viru ,Antibodies, Neutralizing ,Virology ,Molecular biology ,Herpesvirus glycoprotein B ,Endocytosis ,Protein Structure, Tertiary ,Herpes simplex virus ,Microscopy, Fluorescence ,Solubility ,PNAS Plus ,chemistry ,Mutation ,Proteolysis ,biology.protein ,Glycoprotein ,Cell Adhesion Molecules - Abstract
Significance Entry of enveloped viruses into the cell requires the activation of viral glycoproteins, often mediated by cellular receptors. Herpesviruses infect cells via a multipartite system, which includes species-specific glycoproteins plus conserved apparatus gH/gL and gB. HSV makes use of αvβ6- or αvβ8-integrins as gH/gL receptors. The interaction of HSV gH/gL with integrins resulted in the dissociation of gL. The dissociation took place if all the actors of the entry apparatus were present, i.e., under conditions that lead to glycoprotein activation and virus entry. We propose that ( i ) gL is a regulator of gH and prevents its activation until integrins promote gL dissociation from gH/gL. ( ii ) Dissociation from an inhibitory regulator represents a previously unidentified mechanism of activation of viral fusion glycoproteins.
- Published
- 2015
48. The herpesvirus glycoproteins B and H·L are sequentially recruited to the receptor-bound gD to effect membrane fusion at virus entry
- Author
-
Gabriella Campadelli-Fiume, Cristina Forghieri, and Tatiana Gianni
- Subjects
Multidisciplinary ,Viral protein ,viruses ,Lipid bilayer fusion ,Transfection ,Biological Sciences ,Biology ,medicine.disease_cause ,Virology ,Herpesvirus glycoprotein B ,Virus ,Cell biology ,Herpes simplex virus ,Viral envelope ,Viral entry ,medicine - Abstract
Four glycoproteins (gD, gB, gH, and gL) are required for herpes simplex virus entry into the cell or for cell–cell fusion in transfected cells. gD serves as the receptor-binding glycoprotein and as the trigger of fusion; the other three execute fusion between the viral envelope and the plasma and endocytic membranes or the membranes of adjacent cells and are highly conserved among members of the herpesvirus family. Details of the interaction of gD with gB, gH, and gL were not known. Here, we report that the four glycoproteins assemble into a complex initiated by the interaction of gD with its cellular receptor. gB is recruited to the gD–receptor complex next, even in the absence of gH·gL. gH·gL is recruited next, but only to the receptor–gD–gB ensemble. A complex with the composition receptor–gD–gB–gH·gL is assembled transiently with a life span of 15–30 min in cells exposed to virus but can also be found in infected cells and in cells committed to form polykaryocytes after transfection of the glycoprotein quartet. The results indicate that the complex assembly is a critical step in the process of virus entry and fusion, and that no viral protein other than those that participate in the complex itself is required for complex assembly. These findings imply critical protein–protein interactions among the quartet as herpes simplex virions enter the cells and at cell–cell fusion, define a specific order of recruitment, and place gH·gL as the last link in the process of glycoprotein recruitment to the complex.
- Published
- 2006
49. A Heptad Repeat in Herpes Simplex Virus 1 gH, Located Downstream of the α-Helix with Attributes of a Fusion Peptide, Is Critical for Virus Entry and Fusion
- Author
-
Laura Menotti, Tatiana Gianni, and Gabriella Campadelli-Fiume
- Subjects
Author's Correction ,Repetitive Sequences, Amino Acid ,viruses ,Molecular Sequence Data ,Immunology ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Membrane Fusion ,Microbiology ,Protein Structure, Secondary ,Cell Line ,Viral Envelope Proteins ,Viral entry ,Cricetinae ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Amino Acid Sequence ,Peptide sequence ,Coiled coil ,Base Sequence ,Virulence ,Genetic Complementation Test ,Lipid bilayer fusion ,Herpesvirus glycoprotein B ,Virus-Cell Interactions ,Protein Structure, Tertiary ,Cell biology ,Heptad repeat ,Herpes simplex virus ,Ectodomain ,Insect Science ,COS Cells ,DNA, Viral - Abstract
Entry of herpes simplex virus 1 (HSV-1) into cells occurs by fusion with cell membranes; it requires gD as the receptor binding glycoprotein and the trigger of fusion, and the trio of the conserved glycoproteins gB, gH, and gL to execute fusion. Recently, we reported that the ectodomain of HSV-1 gH carries a hydrophobic α-helix (residues 377 to 397) with attributes of an internal fusion peptide (T. Gianni, P. L. Martelli, R. Casadio, and G. Campadelli-Fiume, J. Virol. 79: 2931-2940, 2005). Downstream of this α-helix, a heptad repeat (HR) with a high propensity to form a coiled coil was predicted between residues 443 and 471 and was designated HR-1. The simultaneous substitution of two amino acids in HR-1 (E450G and L453A), predicted to abolish the coiled coil, abolished the ability of gH to complement the infectivity of a gH-null HSV mutant. When coexpressed with gB, gD, and gL, the mutant gH was unable to promote cell-cell fusion. These defects were not attributed to a defect in heterodimer formation with gL, the gH chaperone, or in trafficking to the plasma membrane. A 25-amino-acid synthetic peptide with the sequence of HR-1 (pep-gH wt25 ) inhibited HSV replication if present at the time of virus entry into the cell. A scrambled peptide had no effect. The effect was specific, as pep-gH wt25 did not reduce HSV-2 and pseudorabies virus infection. The presence of a functional HR in the HSV-1 gH ectodomain strengthens the view that gH has attributes typical of a viral fusion glycoprotein.
- Published
- 2005
50. Entry of Herpes Simplex Virus Mediated by Chimeric Forms of Nectin1 Retargeted to Endosomes or to Lipid Rafts Occurs through Acidic Endosomes
- Author
-
Gabriella Campadelli-Fiume, Laura Menotti, Tatiana Gianni, GIANNI T., CAMPADELLI-FIUME G., and MENOTTI L.
- Subjects
Herpesvirus entry mediator ,Endosome ,Nectins ,Immunology ,Endocytic cycle ,VIRAL ENTRY ,Biology ,Endocytosis ,Membrane Fusion ,Microbiology ,Ammonium Chloride ,Membrane Microdomains ,NECTIN1 RECEPTOR RETARGETING ,Viral entry ,Cricetinae ,Virology ,Animals ,Simplexvirus ,Lipid raft ,Tyrphostins ,ENDOSOMES ,Virus-Cell Interactions ,Cell biology ,Androstadienes ,ErbB Receptors ,LIPID RAFTS ,Ectodomain ,Biochemistry ,Insect Science ,Quinazolines ,Receptors, Virus ,Macrolides ,Signal transduction ,Wortmannin ,Cell Adhesion Molecules ,HERPES SIMPLEX VIRUS - Abstract
Herpes simplex virus (HSV) enters cells by fusion with target membranes, commonly the plasma membrane. In some cells, including CHO cells expressing the nectin1 or herpesvirus entry mediator receptors, entry occurs through an endocytic route. We report the following results. (i) When expressed in J cells, nectin1 and HVEM mediated a pathway of entry insensitive to endosome acidification inhibitors. (ii) A chimeric nectin1 receptor competent for endosomal uptake by fusion of the nectin1 ectodomain with the transmembrane sequence and cytoplasmic tail of the epidermal growth factor receptor (EGFR1) (nectin1-EGFR1) and chimeric nectin1 sorted to lipid rafts by a glycosylphosphatidylinositol anchor mediated endocytic entry blocked by the early endosome inhibitor wortmannin and by the endosome acidification inhibitors bafilomycin and NH 4 Cl. (iii) Entry mediated by nectin1-EGFR1 was selectively inhibited by AG1478, a tyrosine phosphorylation inhibitor that targets the EGFR1 cytoplasmic tail and blocks the signaling pathway that culminates in clathrin-dependent uptake of the receptor into endosomes. We draw the following conclusions. (i) The same receptor may initiate different routes of infection, depending on the cell in which it is expressed. Hence, the cell is a determinant that controls whether a given receptor initiates a plasma membrane or an endocytic route of entry. (ii) Receptors whose physiology involves uptake into endosomes or sorting to lipid rafts are suitable to serve as HSV receptors. (iii) Structural features of the receptors are additional determinants that control whether HSV entry occurs at the plasma membrane or at endosomes. These findings are relevant to studies of HSV retargeting to specific receptors.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.