16 results on '"Irene Garzia"'
Search Results
2. Development of a Potency Assay for Nous-209, a Multivalent Neoantigens-Based Genetic Cancer Vaccine
- Author
-
Rosa Bartolomeo, Fulvia Troise, Simona Allocca, Giulia Sdruscia, Rosa Vitale, Veronica Bignone, Anna Maria Petrone, Giuseppina Romano, Anna Morena D’Alise, Valentino Ruzza, Irene Garzia, Guido Leoni, Rossella Merone, Francesca Lanzaro, Stefano Colloca, Loredana Siani, Elisa Scarselli, and Gabriella Cotugno
- Subjects
viral vectors ,quality control testing ,potency assessment ,cancer vaccine ,RT-Q-PCR potency assay ,synthetic polypeptides ,Medicine - Abstract
Quality control testing of vaccines, including potency assessment, is critical to ensure equivalence of clinical lots. We developed a potency assay to support the clinical advancement of Nous-209, a cancer vaccine based on heterologous prime/boost administration of two multivalent viral vector products: GAd-209 and MVA-209. These consist of a mix of four Adeno (Great Ape Adenovirus; GAd) and four Modified Vaccinia Ankara (MVA) vectors respectively, each containing a different transgene encoding a synthetic polypeptide composed of antigenic peptide fragments joined one after the other. The potency assay employs quantitative Reverse Transcription PCR (RT-Q-PCR) to quantitatively measure the transcripts from the four transgenes encoded by each product in in vitro infected cells, enabling simultaneous detection. Results showcase the assay’s robustness and biological relevance, as it effectively detects potency loss in one component of the mixture comparably to in vivo immunogenicity testing. This report details the assay’s setup and validation, offering valuable insights for the clinical development of similar genetic vaccines, particularly those encoding synthetic polypeptides.
- Published
- 2024
- Full Text
- View/download PDF
3. Vector Aided Microenvironment programming (VAMP): reprogramming the TME with MVA virus expressing IL-12 for effective antitumor activity
- Author
-
Fulvia Troise, Anna Morena D'Alise, Guido Leoni, Maria De Lucia, Linda Nocchi, Elisa Micarelli, Gabriella Cotugno, Irene Garzia, Elisa Scarselli, Laura Seclì, Luigia Infante, Lidia Avalle, Giulia Sdruscia, Simona Allocca, and Giuseppina Romano
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Tumor microenvironment (TME) represents a critical hurdle in cancer immunotherapy, given its ability to suppress antitumor immunity. Several efforts are made to overcome this hostile TME with the development of new therapeutic strategies modifying TME to boost antitumor immunity. Among these, cytokine-based approaches have been pursued for their known immunomodulatory effects on different cell populations within the TME. IL-12 is a potent pro-inflammatory cytokine that demonstrates striking immune activation and tumor control but causes severe adverse effects when systemically administered. Thus, local administration is considered a potential strategy to achieve high cytokine concentrations at the tumor site while sparing systemic adverse effects.Methods Modified Vaccinia Ankara (MVA) vector is a potent inducer of pro-inflammatory response. Here, we cloned IL-12 into the genome of MVA for intratumoral immunotherapy, combining the immunomodulatory properties of both the vector and the cargo. The antitumor activity of MVA-IL-12 and its effect on TME reprogramming were investigated in preclinical tumor models. RNA sequencing (RNA-Seq) analysis was performed to assess changes in the TME in treated and distal tumors and the effect on the intratumoral T-cell receptor repertoire.Results Intratumoral injection of MVA-IL-12 resulted in strong antitumor activity with the complete remission of established tumors in multiple murine models, including those resistant to checkpoint inhibitors. The therapeutic activity of MVA-IL-12 was associated with very low levels of circulating cytokine. Effective TME reprogramming was demonstrated on treatment, with the reduction of immunosuppressive M2 macrophages while increasing pro-inflammatory M1, and recruitment of dendritic cells. TME switch from immunosuppressive into immunostimulatory environment allowed for CD8 T cells priming and expansion leading to tumor attack.Conclusions Intratumoral administration of MVA-IL-12 turns immunologically ‘cold’ tumors ‘hot’ and overcomes resistance to programmed cell death protein-1 blockade.
- Published
- 2023
- Full Text
- View/download PDF
4. Adenovirus Encoded Adjuvant (AdEnA) anti-CTLA-4, a novel strategy to improve Adenovirus based vaccines against infectious diseases and cancer
- Author
-
Anna Morena D’Alise, Linda Nocchi, Irene Garzia, Laura Seclì, Luigia Infante, Fulvia Troise, Gabriella Cotugno, Simona Allocca, Giuseppina Romano, Armin Lahm, Guido Leoni, Emanuele Sasso, Elisa Scarselli, and Alfredo Nicosia
- Subjects
vaccine ,genetic adjuvants ,immune response ,cancer ,infectious disease ,neoantigens ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionVirus vectored genetic vaccines (Vvgv) represent a promising approach for eliciting immune protection against infectious diseases and cancer. However, at variance with classical vaccines to date, no adjuvant has been combined with clinically approved genetic vaccines, possibly due to the detrimental effect of the adjuvant-induced innate response on the expression driven by the genetic vaccine vector. We reasoned that a potential novel approach to develop adjuvants for genetic vaccines would be to “synchronize” in time and space the activity of the adjuvant with that of the vaccine.MethodsTo this aim, we generated an Adenovirus vector encoding a murine anti-CTLA-4 monoclonal antibody (Ad-9D9) as a genetic adjuvant for Adenovirus based vaccines.ResultsThe co-delivery of Ad-9D9 with an Adeno-based COVID-19 vaccine encoding the Spike protein resulted in stronger cellular and humoral immune responses. In contrast, only a modest adjuvant effect was achieved when combining the vaccine with the same anti-CTLA-4 in its proteinaceous form. Importantly, the administration of the adjuvant vector at different sites of the vaccine vector abrogates the immunostimulatory effect. We showed that the adjuvant activity of Ad-α-CTLA-4 is independent from the vaccine antigen as it improved the immune response and efficacy of an Adenovirus based polyepitope vaccine encoding tumor neoantigens.DiscussionOur study demonstrated that the combination of Adenovirus Encoded Adjuvant (AdEnA) with an Adeno-encoded antigen vaccine enhances immune responses to viral and tumor antigens, representing a potent approach to develop more effective genetic vaccines.
- Published
- 2023
- Full Text
- View/download PDF
5. 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
6. Maximizing cancer therapy via complementary mechanisms of immune activation: PD-1 blockade, neoantigen vaccination, and Tregs depletion
- Author
-
Jonathan Zalevsky, Deborah H Charych, Alfredo Nicosia, Fulvia Troise, Anna Morena D'Alise, Guido Leoni, Maria De Lucia, Francesca Langone, Linda Nocchi, Fabio Giovanni Tucci, Elisa Micarelli, Gabriella Cotugno, Irene Garzia, Rosa Vitale, Veronica Bignone, Elena Di Matteo, Rosa Bartolomeo, Armin Lahm, and Elisa Scarselli
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
7. VENUS, a Novel Selection Approach to Improve the Accuracy of Neoantigens’ Prediction
- Author
-
Guido Leoni, Anna Morena D’Alise, Fabio Giovanni Tucci, Elisa Micarelli, Irene Garzia, Maria De Lucia, Francesca Langone, Linda Nocchi, Gabriella Cotugno, Rosa Bartolomeo, Giuseppina Romano, Simona Allocca, Fulvia Troise, Alfredo Nicosia, Armin Lahm, and Elisa Scarselli
- Subjects
neoantigen ,cancer vaccine ,VENUS ,prediction ,MC38 ,Medicine - Abstract
Neoantigens are tumor-specific antigens able to induce T-cell responses, generated by mutations in protein-coding regions of expressed genes. Previous studies demonstrated that only a limited subset of mutations generates neoantigens in microsatellite stable tumors. We developed a method, called VENUS (Vaccine-Encoded Neoantigens Unrestricted Selection), to prioritize mutated peptides with high potential to be neoantigens. Our method assigns to each mutation a weighted score that combines the mutation allelic frequency, the abundance of the transcript coding for the mutation, and the likelihood to bind the patient’s class-I major histocompatibility complex alleles. By ranking mutated peptides encoded by mutations detected in nine cancer patients, VENUS was able to select in the top 60 ranked peptides, the 95% of neoantigens experimentally validated including both CD8 and CD4 T cell specificities. VENUS was evaluated in a murine model in the context of vaccination with an adeno vector encoding the top ranked mutations prioritized in the MC38 cell line. Efficacy studies demonstrated anti tumoral activity of the vaccine when used in combination with checkpoint inhibitors. The results obtained highlight the importance of a combined scoring system taking into account multiple features of each tumor mutation to improve the accuracy of neoantigen prediction.
- Published
- 2021
- Full Text
- View/download PDF
8. Data from A Genetic Vaccine Encoding Shared Cancer Neoantigens to Treat Tumors with Microsatellite Instability
- Author
-
Elisa Scarselli, Maria Teresa Catanese, Armin Lahm, Alfredo Nicosia, Pierre van der Bruggen, Stefano Colloca, Antonella Folgori, Antonio Siccardi, Elisa Soprana, Maddalena Panigada, Cristophe Vanhaver, Monica Gordon-Alonso, Mahesh Yadav, Maria Grazia Diodoro, Valentino Ruzza, Rossella Merone, Adele Abbate, Fulvia Troise, Elena Di Matteo, Adriano Leuzzi, Federica Mori, Fabio Giovanni Tucci, Veronica Bignone, Rosa Vitale, Imma Fichera, Maria De Lucia, Irene Garzia, Francesca Langone, Gabriella Cotugno, Anna Morena D'Alise, and Guido Leoni
- Abstract
Tumors with microsatellite instability (MSI) are caused by a defective DNA mismatch repair system that leads to the accumulation of mutations within microsatellite regions. Indels in microsatellites of coding genes can result in the synthesis of frameshift peptides (FSP). FSPs are tumor-specific neoantigens shared across patients with MSI. In this study, we developed a neoantigen-based vaccine for the treatment of MSI tumors. Genetic sequences from 320 MSI tumor biopsies and matched healthy tissues in The Cancer Genome Atlas database were analyzed to select shared FSPs. Two hundred nine FSPs were selected and cloned into nonhuman Great Ape Adenoviral and Modified Vaccinia Ankara vectors to generate a viral-vectored vaccine, referred to as Nous-209. Sequencing tumor biopsies of 20 independent patients with MSI colorectal cancer revealed that a median number of 31 FSPs out of the 209 encoded by the vaccine was detected both in DNA and mRNA extracted from each tumor biopsy. A relevant number of peptides encoded by the vaccine were predicted to bind patient HLA haplotypes. Vaccine immunogenicity was demonstrated in mice with potent and broad induction of FSP-specific CD8 and CD4 T-cell responses. Moreover, a vaccine-encoded FSP was processed in vitro by human antigen-presenting cells and was subsequently able to activate human CD8 T cells. Nous-209 is an “off-the-shelf” cancer vaccine encoding many neoantigens shared across sporadic and hereditary MSI tumors. These results indicate that Nous-209 can induce the optimal breadth of immune responses that might achieve clinical benefit to treat and prevent MSI tumors.Significance:These findings demonstrate the feasibility of an “off-the-shelf” vaccine for treatment and prevention of tumors harboring frameshift mutations and neoantigenic peptides as a result of microsatellite instability.
- Published
- 2023
- Full Text
- View/download PDF
9. Table S1 from A Genetic Vaccine Encoding Shared Cancer Neoantigens to Treat Tumors with Microsatellite Instability
- Author
-
Elisa Scarselli, Maria Teresa Catanese, Armin Lahm, Alfredo Nicosia, Pierre van der Bruggen, Stefano Colloca, Antonella Folgori, Antonio Siccardi, Elisa Soprana, Maddalena Panigada, Cristophe Vanhaver, Monica Gordon-Alonso, Mahesh Yadav, Maria Grazia Diodoro, Valentino Ruzza, Rossella Merone, Adele Abbate, Fulvia Troise, Elena Di Matteo, Adriano Leuzzi, Federica Mori, Fabio Giovanni Tucci, Veronica Bignone, Rosa Vitale, Imma Fichera, Maria De Lucia, Irene Garzia, Francesca Langone, Gabriella Cotugno, Anna Morena D'Alise, and Guido Leoni
- Abstract
List of 1087 FSPs suitable for the inclusion in NOUS-209 vaccine.
- Published
- 2023
- Full Text
- View/download PDF
10. 706 NOUS-PEV, a novel personalized viral-based prime/boost cancer immunotherapy targeting patient-specific neoantigens: interim results from the first subjects in the phase 1b study
- Author
-
Oliver Bechter, Juan Martin-Liberal, Anna D’Alise, Guido Leoni, Gabriella Cotugno, Loredana Siani, Rosa Vitale, Valentino Ruzza, Elisa Micarelli, Irene Garzia, Thea Faivre, Sven Gogov, Patricia Delaite, Stefano Colloca, Maria Ambrosio, Rossella Merone, Elisa Scarselli, and Stefan Symeonides
- Published
- 2022
- Full Text
- View/download PDF
11. Abstract LB196: NOUS-PEV, a personalized cancer immunotherapy targeting neoantigens, induces long lasting, tumor infiltrating memory T cells
- Author
-
Oliver Bechter, Anna Morena D'Alise, Guido Leoni, Gabriella Cotugno, Loredana Siani, Rosa Vitale, Valentino Ruzza, Irene Garzia, Laura Antonucci, Elisa Micarelli, Sven Gogov, Alessia Capone, Juan Martin-Liberal, Emiliano Calvo, Victor Moreno, Stefan Symeonides, and Elisa Scarselli
- Subjects
Cancer Research ,Oncology - Abstract
Personalized vaccines hold great promise to exert meaningful clinical efficacy, with durable tumor control maintained by a vaccine–induced memory response. NOUS–PEV is a personalized viral prime–boost cancer vaccine that expresses 60 patient–specific neoantigens identified by next generation sequencing (NGS) and selected with a proprietary algorithm VENUS (Leoni & D’Alise et al, Vaccines, 9, 2021). Administration is intramuscular, with a priming Great Ape Adenovirus (GAd) vaccination, followed by Modified Vaccinia Ankara (MVA) “boosts”, administered in combination with the PD–1 blocking antibody pembrolizumab in patients with metastatic malignant melanoma and non–small cell lung cancer. Data from the Part 1 dose–confirmation cohort of 3 patients demonstrated the combination of NOUS–PEV and pembrolizumab to be safe and well–tolerated, with early indications of efficacy and immunogenicity (Bechter, et al SITC 2022 Poster number: 706). Now in Part 2 extension–expansion cohorts, we present extended safety, immunogenicity and clinical data at 11 months median follow–up for 6 vaccinated melanoma patients. Tolerability remains good with no grade 3 or 4 vaccine related adverse events and activity encouraging, with 4 PRs, 1 SD and only 1 PD as best response. Immune responses were evaluated by ex–vivo interferon–gamma ELISpot on PBMC collected at baseline, post pembrolizumab, and post vaccination. Vaccine immunogenicity was demonstrated in all evaluable patients receiving the prime/boost regimen (n=4), with a mean of T cell response of ~ 650 IFN–γ spot forming cells (SFC) per million of PBMC (range 380–1,250 SFC/106) and with observed induction of both CD4 and CD8 T cell responses which lasted for at least 6 months. By analyzing the intratumoral TCR repertoire, we found increase of T cells by ~3 fold on average post treatment with NOUS–PEV in all evaluable patients (n=3), with expansion and diversification of intratumoral T cell clones. Vaccine–induced TCR clonotypes were found in on–treatment tumor biopsies of 2 vaccinated patients, providing the proof–of–concept for neoantigen induced T cells homing and infiltrating into the tumor. Overall, these data show that NOUS–PEV continues to be safe, and elicits a robust long lasting immune response and clinical activity. Citation Format: Oliver Bechter, Anna Morena D'Alise, Guido Leoni, Gabriella Cotugno, Loredana Siani, Rosa Vitale, Valentino Ruzza, Irene Garzia, Laura Antonucci, Elisa Micarelli, Sven Gogov, Alessia Capone, Juan Martin-Liberal, Emiliano Calvo, Victor Moreno, Stefan Symeonides, Elisa Scarselli. NOUS-PEV, a personalized cancer immunotherapy targeting neoantigens, induces long lasting, tumor infiltrating memory T cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr LB196.
- Published
- 2023
- Full Text
- View/download PDF
12. Vector Aided Microenvironment programming (VAMP): reprogramming the TME with MVA virus expressing IL-12 for effective antitumor activity
- Author
-
Laura Seclì, Luigia Infante, Linda Nocchi, Maria De Lucia, Gabriella Cotugno, Guido Leoni, Elisa Micarelli, Irene Garzia, Lidia Avalle, Giulia Sdruscia, Fulvia Troise, Simona Allocca, Giuseppina Romano, Elisa Scarselli, and Anna Morena D'Alise
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Immunology ,Molecular Medicine ,Immunology and Allergy - Abstract
BackgroundTumor microenvironment (TME) represents a critical hurdle in cancer immunotherapy, given its ability to suppress antitumor immunity. Several efforts are made to overcome this hostile TME with the development of new therapeutic strategies modifying TME to boost antitumor immunity. Among these, cytokine-based approaches have been pursued for their known immunomodulatory effects on different cell populations within the TME. IL-12 is a potent pro-inflammatory cytokine that demonstrates striking immune activation and tumor control but causes severe adverse effects when systemically administered. Thus, local administration is considered a potential strategy to achieve high cytokine concentrations at the tumor site while sparing systemic adverse effects.MethodsModified Vaccinia Ankara (MVA) vector is a potent inducer of pro-inflammatory response. Here, we cloned IL-12 into the genome of MVA for intratumoral immunotherapy, combining the immunomodulatory properties of both the vector and the cargo. The antitumor activity of MVA-IL-12 and its effect on TME reprogramming were investigated in preclinical tumor models. RNA sequencing (RNA-Seq) analysis was performed to assess changes in the TME in treated and distal tumors and the effect on the intratumoral T-cell receptor repertoire.ResultsIntratumoral injection of MVA-IL-12 resulted in strong antitumor activity with the complete remission of established tumors in multiple murine models, including those resistant to checkpoint inhibitors. The therapeutic activity of MVA-IL-12 was associated with very low levels of circulating cytokine. Effective TME reprogramming was demonstrated on treatment, with the reduction of immunosuppressive M2 macrophages while increasing pro-inflammatory M1, and recruitment of dendritic cells. TME switch from immunosuppressive into immunostimulatory environment allowed for CD8 T cells priming and expansion leading to tumor attack.ConclusionsIntratumoral administration of MVA-IL-12 turns immunologically ‘cold’ tumors ‘hot’ and overcomes resistance to programmed cell death protein-1 blockade.
- Published
- 2023
- Full Text
- View/download PDF
13. 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
14. VENUS, a Novel Selection Approach to Improve the Accuracy of Neoantigens’ Prediction
- Author
-
Simona Allocca, Linda Nocchi, Gabriella Cotugno, Guido Leoni, Rosa Bartolomeo, Irene Garzia, Elisa Scarselli, Fulvia Troise, Maria De Lucia, Elisa Micarelli, Anna Morena D'Alise, Armin Lahm, Alfredo Nicosia, Giuseppina Romano, Fabio Giovanni Tucci, Francesca Langone, Leoni, G., D'Alise, A. M., Tucci, F. G., Micarelli, E., Garzia, I., De Lucia, M., Langone, F., Nocchi, L., Cotugno, G., Bartolomeo, R., Romano, G., Allocca, S., Troise, F., Nicosia, A., Lahm, A., and Scarselli, E.
- Subjects
Immunology ,Context (language use) ,Computational biology ,Major histocompatibility complex ,Article ,Antigen ,Drug Discovery ,Pharmacology (medical) ,Allele ,Allele frequency ,Gene ,Pharmacology ,biology ,integumentary system ,prediction ,neoantigen ,Infectious Diseases ,cancer vaccine ,VENUS ,MC38 ,Mutation (genetic algorithm) ,biology.protein ,Medicine ,Cancer vaccine - Abstract
Neoantigens are tumor-specific antigens able to induce T-cell responses, generated by mutations in protein-coding regions of expressed genes. Previous studies demonstrated that only a limited subset of mutations generates neoantigens in microsatellite stable tumors. We developed a method, called VENUS (Vaccine-Encoded Neoantigens Unrestricted Selection), to prioritize mutated peptides with high potential to be neoantigens. Our method assigns to each mutation a weighted score that combines the mutation allelic frequency, the abundance of the transcript coding for the mutation, and the likelihood to bind the patient’s class-I major histocompatibility complex alleles. By ranking mutated peptides encoded by mutations detected in nine cancer patients, VENUS was able to select in the top 60 ranked peptides, the 95% of neoantigens experimentally validated including both CD8 and CD4 T cell specificities. VENUS was evaluated in a murine model in the context of vaccination with an adeno vector encoding the top ranked mutations prioritized in the MC38 cell line. Efficacy studies demonstrated anti tumoral activity of the vaccine when used in combination with checkpoint inhibitors. The results obtained highlight the importance of a combined scoring system taking into account multiple features of each tumor mutation to improve the accuracy of neoantigen prediction.
- Published
- 2021
15. A genetic vaccine encoding shared cancer neoantigens to treat tumors with microsatellite instability
- Author
-
Valentino Ruzza, Adele Abbate, Adriano Leuzzi, Veronica Bignone, Christophe Vanhaver, Fulvia Troise, Mónica Gordón-Alonso, Anna Morena D'Alise, Irene Garzia, Pierre van der Bruggen, Maddalena Panigada, Francesca Langone, Alfredo Nicosia, Guido Leoni, Elisa Scarselli, Federica Mori, Rosa Maria Vitale, Mahesh Yadav, Maria Grazia Diodoro, Imma Fichera, Rossella Merone, Maria Teresa Catanese, Stefano Colloca, Armin Lahm, Maria De Lucia, Fabio Giovanni Tucci, Elena Di Matteo, Elisa Soprana, Gabriella Cotugno, Antonella Folgori, Antonio G. Siccardi, Leoni, G., D'Alise, A. M., Cotugno, G., Langone, F., Garzia, I., de Lucia, M., Fichera, I., Vitale, R., Bignone, V., Tucci, F. G., Mori, F., Leuzzi, A., Di Matteo, E., Troise, F., Abbate, A., Merone, R., Ruzza, V., Diodoro, M. G., Yadav, M., Gordon-Alonso, M., Vanhaver, C., Panigada, M., Soprana, E., Siccardi, A., Folgori, A., Colloca, S., van der Bruggen, P., Nicosia, A., Lahm, A., Catanese, M. T., Scarselli, E., and UCL - SSS/DDUV/GECE - Génétique cellulaire
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Cancer Research ,Modified vaccinia Ankara ,Antigen-Presenting Cells ,Colorectal Neoplasm ,Biology ,CD8-Positive T-Lymphocytes ,Cancer Vaccines ,Frameshift mutation ,Neoplasm Protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunogenicity, Vaccine ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Frameshift Mutation ,Gene ,Antigen-Presenting Cell ,Animal ,Microsatellite instability ,CD8-Positive T-Lymphocyte ,medicine.disease ,Neoplasm Proteins ,030104 developmental biology ,Oncology ,CD4-Positive T-Lymphocyte ,030220 oncology & carcinogenesis ,Cancer research ,Microsatellite ,DNA mismatch repair ,Female ,Microsatellite Instability ,Cancer vaccine ,Colorectal Neoplasms ,Cancer Vaccine ,Human - Abstract
Tumors with microsatellite instability (MSI) are caused by a defective DNA mismatch repair system that leads to the accumulation of mutations within microsatellite regions. Indels in microsatellites of coding genes can result in the synthesis of frameshift peptides (FSP). FSPs are tumor-specific neoantigens shared across patients with MSI. In this study, we developed a neoantigen-based vaccine for the treatment of MSI tumors. Genetic sequences from 320 MSI tumor biopsies and matched healthy tissues in The Cancer Genome Atlas database were analyzed to select shared FSPs. Two hundred nine FSPs were selected and cloned into nonhuman Great Ape Adenoviral and Modified Vaccinia Ankara vectors to generate a viral-vectored vaccine, referred to as Nous-209. Sequencing tumor biopsies of 20 independent patients with MSI colorectal cancer revealed that a median number of 31 FSPs out of the 209 encoded by the vaccine was detected both in DNA and mRNA extracted from each tumor biopsy. A relevant number of peptides encoded by the vaccine were predicted to bind patient HLA haplotypes. Vaccine immunogenicity was demonstrated in mice with potent and broad induction of FSP-specific CD8 and CD4 T-cell responses. Moreover, a vaccine-encoded FSP was processed in vitro by human antigen-presenting cells and was subsequently able to activate human CD8 T cells. Nous-209 is an “off-the-shelf” cancer vaccine encoding many neoantigens shared across sporadic and hereditary MSI tumors. These results indicate that Nous-209 can induce the optimal breadth of immune responses that might achieve clinical benefit to treat and prevent MSI tumors. Significance: These findings demonstrate the feasibility of an “off-the-shelf” vaccine for treatment and prevention of tumors harboring frameshift mutations and neoantigenic peptides as a result of microsatellite instability.
- Published
- 2020
16. Maximizing cancer therapy via complementary mechanisms of immune activation: PD-1 blockade, neoantigen vaccination, and Tregs depletion
- Author
-
Deborah H. Charych, Elena Di Matteo, Anna Morena D'Alise, Gabriella Cotugno, Maria De Lucia, Veronica Bignone, Fabio Giovanni Tucci, Guido Leoni, Rosa Bartolomeo, Elisa Scarselli, Elisa Micarelli, Jonathan Zalevsky, Alfredo Nicosia, Linda Nocchi, Francesca Langone, Rosa Maria Vitale, Fulvia Troise, Irene Garzia, Armin Lahm, D'Alise, A. M., Leoni, G., de Lucia, M., Langone, F., Nocchi, L., Tucci, F. G., Micarelli, E., Cotugno, G., Troise, F., Garzia, I., Vitale, R., Bignone, V., Matteo, E. D., Bartolomeo, R., Charych, D. H., Lahm, A., Zalevsky, J., Nicosia, A., and Scarselli, E.
- Subjects
Cancer Research ,combined modality therapy ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Immunology ,Gene Expression ,chemical and pharmacologic phenomena ,Cancer Vaccines ,T-Lymphocytes, Regulatory ,Mice ,Immune system ,medicine ,Animals ,Humans ,tumor microenvironment ,Immunology and Allergy ,RC254-282 ,Pharmacology ,Tumor microenvironment ,Animal ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,Basic Tumor Immunology ,adaptive immunity ,Immunotherapy ,vaccination ,medicine.disease ,Acquired immune system ,Vaccination ,medicine.anatomical_structure ,Oncology ,Cancer cell ,Cancer research ,Molecular Medicine ,Female ,business ,Cancer Vaccine ,Human - Abstract
BackgroundA number of different immune pathways are involved in the effective killing of cancer cells, collectively named as the ‘Cancer Immunity Cycle’. Anti-PD-1 checkpoint blockade (CPB) therapy is active on one of these pathways and reinvigorates anticancer T cell immunity, leading to long-term responses in a limited fraction of patients with cancer. We have previously shown that neoantigens-based adenovirus vectored vaccine in combination with anti-PD-1 further expands pre-existing anticancer immunity and elicits novel neoantigen-specific T cells thereby increasing efficacy to 50% of tumor clearance in mice. Here we added a third component to the CPB plus vaccine combination, which is able to modify the suppressive tumor microenvironment by reducing the number of tumor-infiltrating regulatory T cells (Tregs), as strategy for improving the therapeutic efficacy and overcoming resistance.MethodsThe antitumor efficacy of anti-PD-1, neoantigen vaccine and Treg modulating agents, either Bempegaldesleukin (BEMPEG: NKTR-214) or an anti-CTLA-4 mAb with Treg-depleting activity, was investigated in murine tumor models. We evaluated tumor growth in treated animals, neoantigen-specific T cells in tumors, tumor-infiltrating lymphocytes (TILs) and intratumoral Tregs.ResultsThe addition of BEMPEG or anti-CTLA-4 to the combination of vaccine and anti-PD-1 led to complete eradication of large tumors in nearby 100% of treated animals, in association with expansion and activation of cancer neoantigen-specific T cells and reduction of tumor-infiltrating Tregs.ConclusionThese data support the notion that the integrated regulation of three steps of the cancer immunity cycle, including expansion of neoantigen-specific T cells, reversal of the exhausted T cell phenotype together with the reduction of intratumoral Tregs may represent a novel rationally designed drug combination approach to achieve higher cure rates.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.