215 results on '"Alfredo, Nicosia"'
Search Results
2. 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
3. Improved memory CD8 T cell response to delayed vaccine boost is associated with a distinct molecular signature
- Author
-
Ambra Natalini, Sonia Simonetti, Gabriele Favaretto, Lorenzo Lucantonio, Giovanna Peruzzi, Miguel Muñoz-Ruiz, Gavin Kelly, Alessandra M. Contino, Roberta Sbrocchi, Simone Battella, Stefania Capone, Antonella Folgori, Alfredo Nicosia, Angela Santoni, Adrian C. Hayday, and Francesca Di Rosa
- Subjects
CD8 T cells ,memory ,prime-boost interval ,transcriptomic profile ,vaccination ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Effective secondary response to antigen is a hallmark of immunological memory. However, the extent of memory CD8 T cell response to secondary boost varies at different times after a primary response. Considering the central role of memory CD8 T cells in long-lived protection against viral infections and tumors, a better understanding of the molecular mechanisms underlying the changing responsiveness of these cells to antigenic challenge would be beneficial. We examined here primed CD8 T cell response to boost in a BALB/c mouse model of intramuscular vaccination by priming with HIV-1 gag-encoding Chimpanzee adenovector, and boosting with HIV-1 gag-encoding Modified Vaccinia virus Ankara. We found that boost was more effective at day(d)100 than at d30 post-prime, as evaluated at d45 post-boost by multi-lymphoid organ assessment of gag-specific CD8 T cell frequency, CD62L-expression (as a guide to memory status) and in vivo killing. RNA-sequencing of splenic gag-primed CD8 T cells at d100 revealed a quiescent, but highly responsive signature, that trended toward a central memory (CD62L+) phenotype. Interestingly, gag-specific CD8 T cell frequency selectively diminished in the blood at d100, relative to the spleen, lymph nodes and bone marrow. These results open the possibility to modify prime/boost intervals to achieve an improved memory CD8 T cell secondary response.
- Published
- 2023
- Full Text
- View/download PDF
4. 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
5. 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
6. Novel Human Anti-PD-L1 mAbs Inhibit Immune-Independent Tumor Cell Growth and PD-L1 Associated Intracellular Signalling
- Author
-
Margherita Passariello, Anna Morena D’Alise, Annachiara Esposito, Cinzia Vetrei, Guendalina Froechlich, Elisa Scarselli, Alfredo Nicosia, and Claudia De Lorenzo
- Subjects
Medicine ,Science - Abstract
Abstract The novel antibody-based immunotherapy in oncology exploits the activation of immune system mediated by immunomodulatory antibodies specific for immune checkpoints. Among them, the programmed death ligand-1 (PD-L1) is of particular interest as it is expressed not only on T-cells, but also on other immune cells and on a large variety of cancer cells, such as breast cancer cells, considering its high expression in both ErbB2-positive and Triple Negative Breast Cancers. We demonstrate here that PD-L1_1, a novel anti-PD-L1 T -cell stimulating antibody, inhibits PD-L1-tumor cell growth also by affecting the intracellular MAPK pathway and by activating caspase 3. Similar in vitro results were obtained for the first time here also with the clinically validated anti-PD-L1 mAb Atezolizumab and in vivo with another validated anti-mouse anti-PD-L1 mAb. Moreover, we found that two high affinity variants of PD-L1_1 inhibited tumor cell viability more efficiently than the parental PD-L1_1 by affecting the same MAPK pathways with a more potent effect. Altogether, these results shed light on the role of PD-L1 in cancer cells and suggest that PD-L1_1 and its high affinity variants could become powerful antitumor weapons to be used alone or in combination with other drugs such as the anti-ErbB2 cAb already successfully tested in in vitro combinatorial treatments.
- Published
- 2019
- Full Text
- View/download PDF
7. Adenoviral vaccine targeting multiple neoantigens as strategy to eradicate large tumors combined with checkpoint blockade
- Author
-
Anna Morena D’Alise, Guido Leoni, Gabriella Cotugno, Fulvia Troise, Francesca Langone, Imma Fichera, Maria De Lucia, Lidia Avalle, Rosa Vitale, Adriano Leuzzi, Veronica Bignone, Elena Di Matteo, Fabio Giovanni Tucci, Valeria Poli, Armin Lahm, Maria Teresa Catanese, Antonella Folgori, Stefano Colloca, Alfredo Nicosia, and Elisa Scarselli
- Subjects
Science - Abstract
Vaccination against neo-antigens has resulted in an effective antitumor response in several models. Here, the authors show that delivery of larger sets of neo-antigens using an adenovirus-based vaccination platform, results in much better tumor protection when combined with checkpoint blockade in a mouse model of advanced disease.
- Published
- 2019
- Full Text
- View/download PDF
8. Correction: A next generation vaccine against human rabies based on a single dose of a chimpanzee adenovirus vector serotype C.
- Author
-
Federico Napolitano, Rossella Merone, Adele Abbate, Virginia Ammendola, Emma Horncastle, Francesca Lanzaro, Marialuisa Esposito, Alessandra Maria Contino, Roberta Sbrocchi, Andrea Sommella, Joshua D Duncan, Joseph Hinds, Richard A Urbanowicz, Armin Lahm, Stefano Colloca, Antonella Folgori, Jonathan K Ball, Alfredo Nicosia, Benjamin Wizel, Stefania Capone, and Alessandra Vitelli
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
[This corrects the article DOI: 10.1371/journal.pntd.0008459.].
- Published
- 2021
- Full Text
- View/download PDF
9. A next generation vaccine against human rabies based on a single dose of a chimpanzee adenovirus vector serotype C.
- Author
-
Federico Napolitano, Rossella Merone, Adele Abbate, Virginia Ammendola, Emma Horncastle, Francesca Lanzaro, Marialuisa Esposito, Alessandra Maria Contino, Roberta Sbrocchi, Andrea Sommella, Joshua D Duncan, Jospeh Hinds, Richard A Urbanowicz, Armin Lahm, Stefano Colloca, Antonella Folgori, Jonathan K Ball, Alfredo Nicosia, Benjamin Wizel, Stefania Capone, and Alessandra Vitelli
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Rabies, caused by RNA viruses in the Genus Lyssavirus, is the most fatal of all infectious diseases. This neglected zoonosis remains a major public health problem in developing countries, causing the death of an estimated 25,000-159,000 people each year, with more than half of them in children. The high incidence of human rabies in spite of effective vaccines is mainly linked to the lack of compliance with the complicated administration schedule, inadequacies of the community public health system for local administration by the parenteral route and the overall costs of the vaccine. The goal of our work was the development of a simple, affordable and effective vaccine strategy to prevent human rabies virus infection. This next generation vaccine is based on a replication-defective chimpanzee adenovirus vector belonging to group C, ChAd155-RG, which encodes the rabies glycoprotein (G). We demonstrate here that a single dose of this vaccine induces protective efficacy in a murine model of rabies challenge and elicits strong and durable neutralizing antibody responses in vaccinated non-human primates. Importantly, we demonstrate that one dose of a commercial rabies vaccine effectively boosts the neutralizing antibody responses induced by ChAd155-RG in vaccinated monkeys, showing the compatibility of the novel vectored vaccine with the current post-exposure prophylaxis in the event of rabies virus exposure. Finally, we demonstrate that antibodies induced by ChAd155-RG can also neutralize European bat lyssaviruses 1 and 2 (EBLV-1 and EBLV-2) found in bat reservoirs.
- Published
- 2020
- Full Text
- View/download PDF
10. Generation of a Retargeted Oncolytic Herpes Virus Encoding Adenosine Deaminase for Tumor Adenosine Clearance
- Author
-
Chiara Gentile, Arianna Finizio, Guendalina Froechlich, Anna Morena D’Alise, Gabriella Cotugno, Sara Amiranda, Alfredo Nicosia, Elisa Scarselli, Nicola Zambrano, and Emanuele Sasso
- Subjects
oncolytic virus ,adenosine ,adenosine deaminase ,targeted therapy ,immunometabolism ,immunotherapy ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Background: Oncolytic viruses are immunotherapeutic agents that can be engineered to encode payloads of interest within the tumor microenvironment to enhance therapeutic efficacy. Their therapeutic potential could be limited by many avenues for immune evasion exerted by the tumor. One such is mediated by adenosine, which induces pleiotropic immunosuppression by inhibiting antitumor immune populations as well as activating tolerogenic stimuli. Adenosine is produced starting from the highly immunostimulatory ATP, which is progressively hydrolyzed to ADP and adenosine by CD39 and CD73. Cancer cells express high levels of CD39 and CD73 ectoenzymes, thus converting immunostimulatory purinergic signal of ATP into an immunosuppressive signal. For this reason, CD39, CD73 and adenosine receptors are currently investigated in clinical trials as targets for metabolic cancer immunotherapy. This is of particular relevance in the context of oncovirotherapy, as immunogenic cell death induced by oncolytic viruses causes the secretion of a high amount of ATP which is available to be quickly converted into adenosine. Methods: Here, we took advantage of adenosine deaminase enzyme that naturally converts adenosine into the corresponding inosine derivative, devoid of immunoregulatory function. We encoded ADA into an oncolytic targeted herpes virus redirected to human HER2. An engineered ADA with an ectopic signal peptide was also generated to improve enzyme secretion (ADA-SP). Results: Insertion of the expression cassette was not detrimental for viral yield and cancer cell cytotoxicity. The THV_ADA and THV_ADA-SP successfully mediated the secretion of functional ADA enzyme. In in vitro model of human monocytes THP1, this ability of THV_ADA and THV_ADA-SP resulted in the retrieval of eADO-exposed monocytes replication rate, suggesting the proficiency of the viruses in rescuing the immune function. Conclusions: Encoding ADA into oncolytic viruses revealed promising properties for preclinical exploitation.
- Published
- 2021
- Full Text
- View/download PDF
11. Chimpanzee adenoviral vectors as vaccines – challenges to move the technology into the fast lane
- Author
-
Alessandra Vitelli, Antonella Folgori, Elisa Scarselli, Stefano Colloca, Stefania Capone, and Alfredo Nicosia
- Subjects
vaccine ,viral vector ,chimpanzee adenovirus ,immunity ,t-cells ,mucosal immunity ,Internal medicine ,RC31-1245 - Abstract
Introduction: In recent years, replication-defective chimpanzee-derived adenoviruses have been extensively evaluated as genetic vaccines. These vectors share desirable properties with human adenoviruses like the broad tissue tropism and the ease of large-scale manufacturing. Additionally, chimpanzee adenoviruses have the advantage to overcome the negative impact of pre-existing anti-human adenovirus immunity. Areas covered: Here the authors review current pre-clinical research and clinical trials that utilize chimpanzee-derived adenoviral vectors as vaccines. A wealth of studies are ongoing to evaluate different vector backbones and administration routes with the aim of improving immune responses. The challenges associated with the identification of an optimal chimpanzee vector and immunization strategies for different immunological outcomes will be discussed. Expert commentary: The demonstration that chimpanzee adenoviruses can be safely used in humans has paved the way to the use of a whole new array of vectors of different serotypes. However, so far no predictive signature of vector immunity in humans has been identified. The high magnitude of T cell responses elicited by chimpanzee adenoviruses has allowed dissecting the qualitative aspects that may be important for protective immunity. Ultimately, only the results from the most clinically advanced products will help establish the efficacy of the vaccine vector platform in the field of disease prevention.
- Published
- 2017
- Full Text
- View/download PDF
12. 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
13. Mucosal Vaccination with Heterologous Viral Vectored Vaccine Targeting Subdominant SIV Accessory Antigens Strongly Inhibits Early Viral Replication
- Author
-
Huanbin Xu, Anne-Marie Andersson, Emeline Ragonnaud, Ditte Boilesen, Anders Tolver, Benjamin Anderschou Holbech Jensen, James L. Blanchard, Alfredo Nicosia, Antonella Folgori, Stefano Colloca, Riccardo Cortese, Allan Randrup Thomsen, Jan Pravsgaard Christensen, Ronald S. Veazey, and Peter Johannes Holst
- Subjects
Heterologous viral vectored prime-boost immunization ,Genetic adjuvant ,Medicine ,Medicine (General) ,R5-920 - Abstract
Conventional HIV T cell vaccine strategies have not been successful in containing acute peak viremia, nor in providing long-term control. We immunized rhesus macaques intramuscularly and rectally using a heterologous adenovirus vectored SIV vaccine regimen encoding normally weakly immunogenic tat, vif, rev and vpr antigens fused to the MHC class II associated invariant chain. Immunizations induced broad T cell responses in all vaccinees. Following up to 10 repeated low-dose intrarectal challenges, vaccinees suppressed early viral replication (P = 0.01) and prevented the peak viremia in 5/6 animals. Despite consistently undetectable viremia in 2 out of 6 vaccinees, all animals showed evidence of infection induced immune responses indicating that infection had taken place. Vaccinees, with and without detectable viremia better preserved their rectal CD4+ T cell population and had reduced immune hyperactivation as measured by naïve T cell depletion, Ki-67 and PD-1 expression on T cells. These results indicate that vaccination towards SIV accessory antigens vaccine can provide a level of acute control of SIV replication with a suggestion of beneficial immunological consequences in infected animals of unknown long-term significance. In conclusion, our studies demonstrate that a vaccine encoding subdominant antigens not normally associated with virus control can exert a significant impact on acute peak viremia.
- Published
- 2017
- Full Text
- View/download PDF
14. A Novel Vaccine Strategy Employing Serologically Different Chimpanzee Adenoviral Vectors for the Prevention of HIV-1 and HCV Coinfection
- Author
-
Felicity Hartnell, Anthony Brown, Stefania Capone, Jakub Kopycinski, Carly Bliss, Shokouh Makvandi-Nejad, Leo Swadling, Emma Ghaffari, Paola Cicconi, Mariarosaria Del Sorbo, Roberta Sbrocchi, Ilaria Esposito, Ventzislav Vassilev, Paula Marriott, Clair M. Gardiner, Ciaran Bannan, Colm Bergin, Matthias Hoffmann, Bethany Turner, Alfredo Nicosia, Antonella Folgori, Tomáš Hanke, Eleanor Barnes, and Lucy Dorrell
- Subjects
HIV-1 ,HCV (hepatitis C virus) ,vaccine ,coadministration ,clinical trial ,conserved region ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Background: Nearly 3 million people worldwide are coinfected with HIV and HCV. Affordable strategies for prevention are needed. We developed a novel vaccination regimen involving replication-defective and serologically distinct chimpanzee adenovirus (ChAd3, ChAd63) vector priming followed by modified vaccinia Ankara (MVA) boosts, for simultaneous delivery of HCV non-structural (NSmut) and HIV-1 conserved (HIVconsv) region immunogens.Methods: We conducted a phase I trial in which 33 healthy volunteers were sequentially enrolled and vaccinated via the intramuscular route as follows: 9 received ChAd3-NSmut [2.5 × 1010 vp] and MVA-NSmut [2 × 108 pfu] at weeks 0 and 8, respectively; 8 received ChAdV63.HIVconsv [5 × 1010 vp] and MVA.HIVconsv [2 × 108 pfu] at the same interval; 16 were co-primed with ChAd3-NSmut [2.5 × 1010 vp] and ChAdV63.HIVconsv [5 × 1010 vp] followed at week 8 by MVA-NSmut and MVA.HIVconsv [both 1 × 108 pfu]. Immunogenicity was assessed using peptide pools in ex vivo ELISpot and intracellular cytokine assays. Vaccine-induced whole blood transcriptome changes were assessed by microarray analysis.Results: All vaccines were well tolerated and no vaccine-related serious adverse events occurred. Co-administration of the prime-boost vaccine regimens induced high magnitude and broad T cell responses that were similar to those observed following immunization with either regimen alone. Median (interquartile range, IQR) peak responses to NSmut were 3,480 (2,728–4,464) and 3,405 (2,307–7,804) spot-forming cells (SFC)/106 PBMC for single and combined HCV vaccinations, respectively (p = 0.8). Median (IQR) peak responses to HIVconsv were 1,305 (1,095–4,967) and 1,005 (169–2,482) SFC/106 PBMC for single and combined HIV-1 vaccinations, respectively (p = 0.5). Responses were maintained above baseline to 34 weeks post-vaccination. Intracellular cytokine analysis indicated that the responding populations comprised polyfunctional CD4+ and CD8+ T cells. Canonical pathway analysis showed that in the single and combined vaccination groups, pathways associated with antiviral and innate immune responses were enriched for upregulated interferon-stimulated genes 24 h after priming and boosting vaccinations.Conclusions: Serologically distinct adenoviral vectors encoding HCV and HIV-1 immunogens can be safely co-administered without reducing the immunogenicity of either vaccine. This provides a novel strategy for targeting these viruses simultaneously and for other pathogens that affect the same populations.Clinical trial registration:https://clinicaltrials.gov, identifier: NCT02362217
- Published
- 2019
- Full Text
- View/download PDF
15. 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
16. Table S2 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
Top Nous-209-derived peptides predicted to bind to HLA-A*02:01.
- Published
- 2023
17. Generation of a Novel Mesothelin-Targeted Oncolytic Herpes Virus and Implemented Strategies for Manufacturing
- Author
-
Guendalina Froechlich, Chiara Gentile, Luigia Infante, Carmen Caiazza, Pasqualina Pagano, Sarah Scatigna, Gabriella Cotugno, Anna Morena D’Alise, Armin Lahm, Elisa Scarselli, Alfredo Nicosia, Massimo Mallardo, Emanuele Sasso, and Nicola Zambrano
- Subjects
oncolytic virus ,triple negative breast cancer ,malignant mesothelioma ,targeted therapy ,MSLN ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Background: HER2-based retargeted viruses are in advanced phases of preclinical development of breast cancer models. Mesothelin (MSLN) is a cell-surface tumor antigen expressed in different subtypes of breast and non-breast cancer. Its recent identification as a marker of some triple-negative breast tumors renders it an attractive target, presently investigated in clinical trials employing antibody drug conjugates and CAR-T cells. The availability of MSLN-retargeted oncolytic viruses may complement the current immunotherapeutic panel of biological drugs against HER2-negative breast and non-breast tumors. Methods: A fully virulent, tumor-targeted oncolytic Herpes simplex virus-1 (MSLN-THV) with a selectivity for mesothelin-expressing cancer cells was generated. Recombineering technology was used to replace an essential moiety of the viral glycoprotein D with antibody fragments derived from clinically validated MSLN monoclonal antibodies, and to allow IL12 cargo expression in infected cells. Panels of breast and female reproductive system cell lines were used to verify the oncolytic potential of the viral constructs. A platform for production of the retargeted viruses was developed in HEK 293 cells, providing stable expression of a suitable chimeric receptor. Results: We demonstrated the selectivity of viral infection and cytotoxicity by MSLN-retargeted viruses in a panel of mesothelin-positive cancer cells, originating from breast and female reproductive system tumors. We also developed a second-generation oncolytic MSLN-THV, encoding IL12, to enhance the immunotherapeutic potential of the viral backbone. A non-tumor cell line expressing a chimeric MSLN/Nectin-1 receptor, de-sensitized from antiviral responses by genetic inactivation of the Stimulator of Interferon Genes (STING)-dependent pathway was engineered, to optimize viral yields. Conclusions: Our proof-of-concept study proposes MSLN-retargeted herpesviruses as potential cancer immunotherapeutics for assessments in preclinical models of MSLN-positive tumors, complementing the available panel of oncolytic viruses to HER2-negative breast tumors.
- Published
- 2021
- Full Text
- View/download PDF
18. Safety and Immunogenicity of Malaria Vectored Vaccines Given with Routine Expanded Program on Immunization Vaccines in Gambian Infants and Neonates: A Randomized Controlled Trial
- Author
-
Victorine A. Mensah, Sophie Roetynck, Ebrima K. Kanteh, Georgina Bowyer, Amy Ndaw, Francis Oko, Carly M. Bliss, Ya Jankey Jagne, Riccardo Cortese, Alfredo Nicosia, Rachel Roberts, Flavia D’Alessio, Odile Leroy, Babacar Faye, Beate Kampmann, Badara Cisse, Kalifa Bojang, Stephen Gerry, Nicola K. Viebig, Alison M. Lawrie, Ed Clarke, Egeruan B. Imoukhuede, Katie J. Ewer, Adrian V. S. Hill, and Muhammed O. Afolabi
- Subjects
vaccines ,clinical trials ,malaria ,cellular immune response ,cytokines ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BackgroundHeterologous prime-boost vaccination with chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) encoding multiple epitope string thrombospondin-related adhesion protein (ME-TRAP) has shown acceptable safety and promising immunogenicity in African adult and pediatric populations. If licensed, this vaccine could be given to infants receiving routine childhood immunizations. We therefore evaluated responses to ChAd63 MVA ME-TRAP when co-administered with routine Expanded Program on Immunization (EPI) vaccines.MethodsWe enrolled 65 Gambian infants and neonates, aged 16, 8, or 1 week at first vaccination and randomized them to receive either ME-TRAP and EPI vaccines or EPI vaccines only. Safety was assessed by the description of vaccine-related adverse events (AEs). Immunogenicity was evaluated using IFNγ enzyme-linked immunospot, whole-blood flow cytometry, and anti-TRAP IgG ELISA. Serology was performed to confirm all infants achieved protective titers to EPI vaccines.ResultsThe vaccines were well tolerated in all age groups with no vaccine-related serious AEs. High-level TRAP-specific IgG and T cell responses were generated after boosting with MVA. CD8+ T cell responses, previously found to correlate with protection, were induced in all groups. Antibody responses to EPI vaccines were not altered significantly.ConclusionMalaria vectored prime-boost vaccines co-administered with routine childhood immunizations were well tolerated. Potent humoral and cellular immunity induced by ChAd63 MVA ME-TRAP did not reduce the immunogenicity of co-administered EPI vaccines, supporting further evaluation of this regimen in infant populations.Clinical Trial RegistrationThe clinical trial was registered on http://Clinicaltrials.gov (NCT02083887) and the Pan-African Clinical Trials Registry (PACTR201402000749217).
- Published
- 2017
- Full Text
- View/download PDF
19. 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
20. Safety, Immunogenicity and Efficacy of Prime-Boost Vaccination with ChAd63 and MVA Encoding ME-TRAP against Plasmodium falciparum Infection in Adults in Senegal.
- Author
-
Victorine A Mensah, Aly Gueye, Magatte Ndiaye, Nick J Edwards, Danny Wright, Nicholas A Anagnostou, Massamba Syll, Amy Ndaw, Annie Abiola, Carly Bliss, Jules-François Gomis, Ines Petersen, Caroline Ogwang, Tandakha Dieye, Nicola K Viebig, Alison M Lawrie, Rachel Roberts, Alfredo Nicosia, Babacar Faye, Oumar Gaye, Odile Leroy, Egeruan B Imoukhuede, Katie J Ewer, Philip Bejon, Adrian V S Hill, Badara Cisse, and MVVC group
- Subjects
Medicine ,Science - Abstract
Malaria transmission is in decline in some parts of Africa, partly due to the scaling up of control measures. If the goal of elimination is to be achieved, additional control measures including an effective and durable vaccine will be required. Studies utilising the prime-boost approach to deliver viral vectors encoding the pre-erythrocytic antigen ME-TRAP (multiple epitope thrombospondin-related adhesion protein) have shown promising safety, immunogenicity and efficacy in sporozoite challenge studies. More recently, a study in Kenyan adults, similar to that reported here, showed substantial efficacy against P. falciparum infection. One hundred and twenty healthy male volunteers, living in a malaria endemic area of Senegal were randomised to receive either the Chimpanzee adenovirus (ChAd63) ME-TRAP as prime vaccination, followed eight weeks later by modified vaccinia Ankara (MVA) also encoding ME-TRAP as booster, or two doses of anti-rabies vaccine as a comparator. Prior to follow-up, antimalarials were administered to clear parasitaemia and then participants were monitored by PCR for malaria infection for eight weeks. The primary endpoint was time-to-infection with P. falciparum malaria, determined by two consecutive positive PCR results. Secondary endpoints included adverse event reporting, measures of cellular and humoral immunogenicity and a meta-analysis of combined vaccine efficacy with the parallel study in Kenyan adults.We show that this pre-erythrocytic malaria vaccine is safe and induces significant immunogenicity, with a peak T-cell response at seven days after boosting of 932 Spot Forming Cells (SFC)/106 Peripheral Blood Mononuclear Cells(PBMC) compared to 57 SFC/ 106 PBMCs in the control group. However, a vaccine efficacy was not observed: 12 of 57 ME-TRAP vaccinees became PCR positive during the intensive monitoring period as compared to 13 of the 58 controls (P = 0.80). This trial confirms that vaccine efficacy against malaria infection in adults may be rapidly assessed using this efficient and cost-effective clinical trial design. Further efficacy evaluation of this vectored candidate vaccine approach in other malaria transmission settings and age-de-escalation into the main target age groups for a malaria vaccine is in progress.
- Published
- 2016
- Full Text
- View/download PDF
21. High-Throughput Monoclonal Antibody Discovery from Phage Libraries: Challenging the Current Preclinical Pipeline to Keep the Pace with the Increasing mAb Demand
- Author
-
Nicola Zambrano, Guendalina Froechlich, Dejan Lazarevic, Margherita Passariello, Alfredo Nicosia, Claudia De Lorenzo, Marco J. Morelli, Emanuele Sasso, Zambrano, Nicola, Froechlich, Guendalina, Lazarevic, Dejan, Passariello, Margherita, Nicosia, Alfredo, De Lorenzo, Claudia, Morelli, Marco J., and Sasso, Emanuele
- Subjects
Cancer Research ,Oncology - Abstract
Monoclonal antibodies are among the most powerful therapeutics in modern medicine. Since the approval of the first therapeutic antibody in 1986, monoclonal antibodies keep holding great expectations for application in a range of clinical indications, highlighting the need to provide timely and sustainable access to powerful screening options. However, their application in the past has been limited by time-consuming and expensive steps of discovery and production. The screening of antibody repertoires is a laborious step; however, the implementation of next-generation sequencing-guided screening of single-chain antibody fragments has now largely overcome this issue. This review provides a detailed overview of the current strategies for the identification of monoclonal antibodies from phage display-based libraries. We also discuss the challenges and the possible solutions to improve the limiting selection and screening steps, in order to keep pace with the increasing demand for monoclonal antibodies.
- Published
- 2022
22. Mucosal delivery of a vectored RSV vaccine is safe and elicits protective immunity in rodents and nonhuman primates
- Author
-
Angiolo Pierantoni, Maria Luisa Esposito, Virginia Ammendola, Federico Napolitano, Fabiana Grazioli, Adele Abbate, Mariarosaria del Sorbo, Loredana Siani, Anna Morena D'Alise, Alessandra Taglioni, Gemma Perretta, Antonio Siccardi, Elisa Soprana, Maddalena Panigada, Michelle Thom, Elisa Scarselli, Antonella Folgori, Stefano Colloca, Geraldine Taylor, Riccardo Cortese, Alfredo Nicosia, Stefania Capone, and Alessandra Vitelli
- Subjects
Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of severe respiratory disease in infants and the elderly. No vaccine is presently available to address this major unmet medical need. We generated a new genetic vaccine based on chimpanzee Adenovirus (PanAd3-RSV) and Modified Vaccinia Ankara RSV (MVA-RSV) encoding the F, N, and M2-1 proteins of RSV, for the induction of neutralizing antibodies and broad cellular immunity. Because RSV infection is restricted to the respiratory tract, we compared intranasal (IN) and intramuscular (M) administration for safety, immunogenicity, and efficacy in different species. A single IN or IM vaccination completely protected BALB/c mice and cotton rats against RSV replication in the lungs. However, only IN administration could prevent infection in the upper respiratory tract. IM vaccination with MVA-RSV also protected cotton rats from lower respiratory tract infection in the absence of detectable neutralizing antibodies. Heterologous prime boost with PanAd3-RSV and MVA-RSV elicited high neutralizing antibody titers and broad T-cell responses in nonhuman primates. In addition, animals primed in the nose developed mucosal IgA against the F protein. In conclusion, we have shown that our vectored RSV vaccine induces potent cellular and humoral responses in a primate model, providing strong support for clinical testing.
- Published
- 2015
- Full Text
- View/download PDF
23. 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
24. A phase Ia study to assess the safety and immunogenicity of new malaria vaccine candidates ChAd63 CS administered alone and with MVA CS.
- Author
-
Eoghan de Barra, Susanne H Hodgson, Katie J Ewer, Carly M Bliss, Kerrie Hennigan, Ann Collins, Eleanor Berrie, Alison M Lawrie, Sarah C Gilbert, Alfredo Nicosia, Samuel J McConkey, and Adrian V S Hill
- Subjects
Medicine ,Science - Abstract
Plasmodium falciparum (P. falciparum) malaria remains a significant cause of mortality and morbidity throughout the world. Development of an effective vaccine would be a key intervention to reduce the considerable social and economic impact of malaria.We conducted a Phase Ia, non-randomized, clinical trial in 24 healthy, malaria-naïve adults of the chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient viral vectored vaccines encoding the circumsporozoite protein (CS) of P. falciparum.ChAd63-MVA CS administered in a heterologous prime-boost regime was shown to be safe and immunogenic, inducing high-level T cell responses to CS. With a priming ChAd63 CS dose of 5×109 vp responses peaked at a mean of 1947 SFC/million PBMC (median 1524) measured by ELIspot 7 days after the MVA boost and showed a mixed CD4+/CD8+ phenotype. With a higher priming dose of ChAd63 CS dose 5×1010 vp T cell responses did not increase (mean 1659 SFC/million PBMC, median 1049). Serum IgG responses to CS were modest and peaked at day 14 post ChAd63 CS (median antibody concentration for all groups at day 14 of 1.3 µg/ml (range 0-11.9), but persisted throughout late follow-up (day 140 median antibody concentration groups 1B & 2B 0.9 µg/ml (range 0-4.7).ChAd63-MVA is a safe and highly immunogenic delivery platform for the CS antigen in humans which warrants efficacy testing.ClinicalTrials.gov NCT01450280.
- Published
- 2014
- Full Text
- View/download PDF
25. Enhanced vaccine-induced CD8+ T cell responses to malaria antigen ME-TRAP by fusion to MHC class ii invariant chain.
- Author
-
Alexandra J Spencer, Matthew G Cottingham, Jennifer A Jenks, Rhea J Longley, Stefania Capone, Stefano Colloca, Antonella Folgori, Riccardo Cortese, Alfredo Nicosia, Migena Bregu, and Adrian V S Hill
- Subjects
Medicine ,Science - Abstract
The orthodox role of the invariant chain (CD74; Ii) is in antigen presentation to CD4+ T cells, but enhanced CD8+ T cells responses have been reported after vaccination with vectored viral vaccines encoding a fusion of Ii to the antigen of interest. In this study we assessed whether fusion of the malarial antigen, ME-TRAP, to Ii could increase the vaccine-induced CD8+ T cell response. Following single or heterologous prime-boost vaccination of mice with a recombinant chimpanzee adenovirus vector, ChAd63, or recombinant modified vaccinia virus Ankara (MVA), higher frequencies of antigen-specific CD4+ and CD8+ T cells were observed, with the largest increases observed following a ChAd63-MVA heterologous prime-boost regimen. Studies in non-human primates confirmed the ability of Ii-fusion to augment the T cell response, where a 4-fold increase was maintained up to 11 weeks after the MVA boost. Of the numerous different approaches explored to increase vectored vaccine induced immunogenicity over the years, fusion to the invariant chain showed a consistent enhancement in CD8+ T cell responses across different animal species and may therefore find application in the development of vaccines against human malaria and other diseases where high levels of cell-mediated immunity are required.
- Published
- 2014
- Full Text
- View/download PDF
26. Randomized Trial of a Vaccine Regimen to Prevent Chronic HCV Infection
- Author
-
Kimberly E Rousseau, Elisa Scarselli, Matthew E. Winter, Kimberly Page, Karla Thornton, Ellen Stein, Andrea L. Cox, Michael R. Wierzbicki, Paula J. Lum, Soju Chang, Antonella Folgori, Alfredo Nicosia, Guido Massaccesi, Michael Forman, Katherine Wagner, Marc G. Ghany, Alice Asher, Linda C. Giudice, T. Jake Liang, Elaine Thomas, Stefania Capone, William O. Osburn, Rebecca T. Veenhuis, Lan Lin, Richard L. Gorman, Michael T. Melia, Ventzislav Vassilev, Page, K., Melia, M. T., Veenhuis, R. T., Winter, M., Rousseau, K. E., Massaccesi, G., Osburn, W. O., Forman, M., Thomas, E., Thornton, K., Wagner, K., Vassilev, V., Lin, L., Lum, P. J., Giudice, L. C., Stein, E., Asher, A., Chang, S., Gorman, R., Ghany, M. G., Liang, T. J., Wierzbicki, M. R., Scarselli, E., Nicosia, A., Folgori, A., Capone, S., and Cox, A. L.
- Subjects
Male ,Adenoviruses ,and promotion of well-being ,T-Lymphocytes ,Disease ,Substance Abuse, Intravenou ,030204 cardiovascular system & hematology ,Medical and Health Sciences ,law.invention ,Hepatitis ,0302 clinical medicine ,Immunogenicity, Vaccine ,Randomized controlled trial ,law ,Medicine ,030212 general & internal medicine ,Young adult ,Chronic ,Substance Abuse, Intravenous ,Vaccines ,Vaccines, Synthetic ,Immunogenicity ,Liver Disease ,Incidence ,Pan troglodyte ,Substance Abuse ,General Medicine ,Hepatitis C ,Middle Aged ,Infectious Diseases ,3.4 Vaccines ,5.1 Pharmaceuticals ,6.1 Pharmaceuticals ,HIV/AIDS ,Female ,Genetic Vector ,Development of treatments and therapeutic interventions ,Intravenous ,Infection ,Human ,Biotechnology ,Adult ,Viral Hepatitis Vaccines ,medicine.medical_specialty ,Adolescent ,Pan troglodytes ,Clinical Trials and Supportive Activities ,Chronic Liver Disease and Cirrhosis ,Genetic Vectors ,Virus ,Article ,Vaccine Related ,03 medical and health sciences ,Young Adult ,Hepatitis - C ,Double-Blind Method ,Clinical Research ,Internal medicine ,General & Internal Medicine ,Animals ,Humans ,Animal ,business.industry ,Prevention ,Synthetic ,Evaluation of treatments and therapeutic interventions ,Hepatitis C Antibodies ,Hepatitis C, Chronic ,medicine.disease ,Prevention of disease and conditions ,Clinical trial ,Regimen ,Emerging Infectious Diseases ,Good Health and Well Being ,T-Lymphocyte ,Adenoviruses, Simian ,Immunization ,Hepatitis C Antibodie ,business ,Simian ,Digestive Diseases ,Vaccine ,Viral Hepatitis Vaccine - Abstract
BACKGROUND: A safe and effective vaccine to prevent chronic hepatitis C virus (HCV) infection is a critical component of efforts to eliminate the disease. METHODS: In this phase 1–2 randomized, double-blind, placebo-controlled trial, we evaluated a recombinant chimpanzee adenovirus 3 vector priming vaccination followed by a recombinant modified vaccinia Ankara boost; both vaccines encode HCV nonstructural proteins. Adults who were considered to be at risk for HCV infection on the basis of a history of recent injection drug use were randomly assigned (in a 1:1 ratio) to receive vaccine or placebo on days 0 and 56. Vaccine-related serious adverse events, severe local or systemic adverse events, and laboratory adverse events were the primary safety end points. The primary efficacy end point was chronic HCV infection, defined as persistent viremia for 6 months. RESULTS: A total of 548 participants underwent randomization, with 274 assigned to each group. There was no significant difference in the incidence of chronic HCV infection between the groups. In the per-protocol population, chronic HCV infection developed in 14 participants in each group (hazard ratio [vaccine vs. placebo], 1.53; 95% confidence interval [CI], 0.66 to 3.55; vaccine efficacy, −53%; 95% CI, −255 to 34). In the modified intention-to-treat population, chronic HCV infection developed in 19 participants in the vaccine group and 17 in placebo group (hazard ratio, 1.66; 95% CI, 0.79 to 3.50; vaccine efficacy, −66%; 95% CI, −250 to 21). The geometric mean peak HCV RNA level after infection differed between the vaccine group and the placebo group (152.51×10(3) IU per milliliter and 1804.93×10(3) IU per milliliter, respectively). T-cell responses to HCV were detected in 78% of the participants in the vaccine group. The percentages of participants with serious adverse events were similar in the two groups. CONCLUSIONS: In this trial, the HCV vaccine regimen did not cause serious adverse events, produced HCV-specific T-cell responses, and lowered the peak HCV RNA level, but it did not prevent chronic HCV infection. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT01436357.)
- Published
- 2021
27. Generation of a Novel Mesothelin-Targeted Oncolytic Herpes Virus and Implemented Strategies for Manufacturing
- Author
-
Elisa Scarselli, Alfredo Nicosia, Guendalina Froechlich, Gabriella Cotugno, Anna Morena D'Alise, Nicola Zambrano, Emanuele Sasso, Pasqualina Pagano, Chiara Gentile, Armin Lahm, Carmen Caiazza, Massimo Mallardo, Sarah Scatigna, Luigia Infante, Froechlich, G., Gentile, C., Infante, L., Caiazza, C., Pagano, P., Scatigna, S., Cotugno, G., D'Alise, A. M., Lahm, A., Scarselli, E., Nicosia, A., Mallardo, M., Sasso, E., and Zambrano, N.
- Subjects
Cell Survival ,Receptor, ErbB-2 ,medicine.medical_treatment ,MSLN ,Breast Neoplasms ,Herpesvirus 1, Human ,GPI-Linked Proteins ,Article ,Catalysis ,Oncolytic herpes virus ,Targeted therapy ,lcsh:Chemistry ,Inorganic Chemistry ,Cell Line, Tumor ,medicine ,Humans ,Mesothelin ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,Triple-negative breast cancer ,oncolytic virus ,Gene Editing ,Oncolytic Virotherapy ,biology ,Organic Chemistry ,Membrane Proteins ,Cancer ,Oncolytic viru ,General Medicine ,medicine.disease ,targeted therapy ,Tumor antigen ,Computer Science Applications ,Oncolytic virus ,HEK293 Cells ,lcsh:Biology (General) ,lcsh:QD1-999 ,triple negative breast cancer ,Cancer cell ,malignant mesothelioma ,biology.protein ,Cancer research ,Female ,Immunotherapy - Abstract
Background: HER2-based retargeted viruses are in advanced phases of preclinical development of breast cancer models. Mesothelin (MSLN) is a cell-surface tumor antigen expressed in different subtypes of breast and non-breast cancer. Its recent identification as a marker of some triple-negative breast tumors renders it an attractive target, presently investigated in clinical trials employing antibody drug conjugates and CAR-T cells. The availability of MSLN-retargeted oncolytic viruses may complement the current immunotherapeutic panel of biological drugs against HER2-negative breast and non-breast tumors. Methods: A fully virulent, tumor-targeted oncolytic Herpes simplex virus-1 (MSLN-THV) with a selectivity for mesothelin-expressing cancer cells was generated. Recombineering technology was used to replace an essential moiety of the viral glycoprotein D with antibody fragments derived from clinically validated MSLN monoclonal antibodies, and to allow IL12 cargo expression in infected cells. Panels of breast and female reproductive system cell lines were used to verify the oncolytic potential of the viral constructs. A platform for production of the retargeted viruses was developed in HEK 293 cells, providing stable expression of a suitable chimeric receptor. Results: We demonstrated the selectivity of viral infection and cytotoxicity by MSLN-retargeted viruses in a panel of mesothelin-positive cancer cells, originating from breast and female reproductive system tumors. We also developed a second-generation oncolytic MSLN-THV, encoding IL12, to enhance the immunotherapeutic potential of the viral backbone. A non-tumor cell line expressing a chimeric MSLN/Nectin-1 receptor, de-sensitized from antiviral responses by genetic inactivation of the Stimulator of Interferon Genes (STING)-dependent pathway was engineered, to optimize viral yields. Conclusions: Our proof-of-concept study proposes MSLN-retargeted herpesviruses as potential cancer immunotherapeutics for assessments in preclinical models of MSLN-positive tumors, complementing the available panel of oncolytic viruses to HER2-negative breast tumors.
- Published
- 2021
- Full Text
- View/download PDF
28. A DNA/Ki67-Based Flow Cytometry Assay for Cell Cycle Analysis of Antigen-Specific CD8 T Cells in Vaccinated Mice
- Author
-
Sonia Simonetti, Angela Santoni, Antonella Folgori, Stefania Capone, Giovanna Peruzzi, Alfredo Nicosia, Francesca Di Rosa, Ambra Natalini, Simonetti, S., Natalini, A., Peruzzi, G., Nicosia, A., Folgori, A., Capone, S., Santoni, A., and Di Rosa, F.
- Subjects
Data Analysis ,Cell division ,General Chemical Engineering ,T cells ,Bone Marrow Cells ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,General Biochemistry, Genetics and Molecular Biology ,Flow cytometry ,Epitopes ,chemistry.chemical_compound ,Antigen ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Mitosis ,Mice, Inbred BALB C ,Staining and Labeling ,General Immunology and Microbiology ,biology ,medicine.diagnostic_test ,Animal ,Chemistry ,General Neuroscience ,Cell Cycle ,Vaccination ,Lymph Node ,Carboxyfluorescein succinimidyl ester ,CD8-Positive T-Lymphocyte ,DNA ,Cell cycle ,Flow Cytometry ,Cell biology ,Ki-67 Antigen ,Data Analysi ,biology.protein ,Bone Marrow Cell ,Epitope ,Female ,Lymph Nodes ,Spleen ,Human - Abstract
The cell cycle of antigen-specific T cells in vivo has been examined by using a few methods, all of which possess some limitations. Bromodeoxyuridine (BrdU) marks cells that are in or recently completed S-phase, and carboxyfluorescein succinimidyl ester (CFSE) detects daughter cells after division. However, these dyes do not allow identification of the cell cycle phase at the time of analysis. An alternative approach is to exploit Ki67, a marker that is highly expressed by cells in all phases of the cell cycle except the quiescent phase G0. Unfortunately, Ki67 does not allow further differentiation as it does not separate cells in S-phase that are committed to mitosis from those in G1 that can remain in this phase, proceed into cycling, or move into G0. Here, we describe a flow cytometric method for capturing a "snapshot" of T cells in different cell cycle phases in mouse secondary lymphoid organs. The method combines Ki67 and DNA staining with major histocompatibility complex (MHC)-peptide-multimer staining and an innovative gating strategy, allowing us to successfully differentiate between antigen-specific CD8 T cells in G0, in G1 and in S-G2/M phases of the cell cycle in the spleen and draining lymph nodes of mice after vaccination with viral vectors carrying the model antigen gag of human immunodeficiency virus (HIV)-1. Critical steps of the method were the choice of the DNA dye and the gating strategy to increase the assay sensitivity and to include highly activated/proliferating antigenspecific T cells that would have been missed by current criteria of analysis. The DNA dye, Hoechst 33342, enabled us to obtain a high-quality discrimination of the G0/ G1 and G2/M DNA peaks, while preserving membrane and intracellular staining. The method has great potential to increase knowledge about T cell response in vivo and to improve immuno-monitoring analysis.
- Published
- 2021
29. Safety and immunogenicity of heterologous prime-boost immunisation with Plasmodium falciparum malaria candidate vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in healthy Gambian and Kenyan adults.
- Author
-
Caroline Ogwang, Muhammed Afolabi, Domtila Kimani, Ya Jankey Jagne, Susanne H Sheehy, Carly M Bliss, Christopher J A Duncan, Katharine A Collins, Miguel A Garcia Knight, Eva Kimani, Nicholas A Anagnostou, Eleanor Berrie, Sarah Moyle, Sarah C Gilbert, Alexandra J Spencer, Peninah Soipei, Jenny Mueller, Joseph Okebe, Stefano Colloca, Riccardo Cortese, Nicola K Viebig, Rachel Roberts, Katherine Gantlett, Alison M Lawrie, Alfredo Nicosia, Egeruan B Imoukhuede, Philip Bejon, Britta C Urban, Katie L Flanagan, Katie J Ewer, Roma Chilengi, Adrian V S Hill, and Kalifa Bojang
- Subjects
Medicine ,Science - Abstract
Heterologous prime boost immunization with chimpanzee adenovirus 63 (ChAd63) and Modified vaccinia Virus Ankara (MVA) vectored vaccines is a strategy recently shown to be capable of inducing strong cell mediated responses against several antigens from the malaria parasite. ChAd63-MVA expressing the Plasmodium falciparum pre-erythrocytic antigen ME-TRAP (multiple epitope string with thrombospondin-related adhesion protein) is a leading malaria vaccine candidate, capable of inducing sterile protection in malaria naïve adults following controlled human malaria infection (CHMI).We conducted two Phase Ib dose escalation clinical trials assessing the safety and immunogenicity of ChAd63-MVA ME-TRAP in 46 healthy malaria exposed adults in two African countries with similar malaria transmission patterns.ChAd63-MVA ME-TRAP was shown to be safe and immunogenic, inducing high-level T cell responses (median >1300 SFU/million PBMC).ChAd63-MVA ME-TRAP is a safe and highly immunogenic vaccine regimen in adults with prior exposure to malaria. Further clinical trials to assess safety and immunogenicity in children and infants and protective efficacy in the field are now warranted.Pactr.org PACTR2010020001771828 Pactr.org PACTR201008000221638 ClinicalTrials.gov NCT01373879 NCT01373879 ClinicalTrials.gov NCT01379430 NCT01379430.
- Published
- 2013
- Full Text
- View/download PDF
30. Vaccination to conserved influenza antigens in mice using a novel Simian adenovirus vector, PanAd3, derived from the bonobo Pan paniscus.
- Author
-
Alessandra Vitelli, Mary R Quirion, Chia-Yun Lo, Julia A Misplon, Agnieszka K Grabowska, Angiolo Pierantoni, Virginia Ammendola, Graeme E Price, Mark R Soboleski, Riccardo Cortese, Stefano Colloca, Alfredo Nicosia, and Suzanne L Epstein
- Subjects
Medicine ,Science - Abstract
Among approximately 1000 adenoviruses from chimpanzees and bonobos studied recently, the Pan Adenovirus type 3 (PanAd3, isolated from a bonobo, Pan paniscus) has one of the best profiles for a vaccine vector, combining potent transgene immunogenicity with minimal pre-existing immunity in the human population. In this study, we inserted into a replication defective PanAd3 a transgene expressing a fusion protein of conserved influenza antigens nucleoprotein (NP) and matrix 1 (M1). We then studied antibody and T cell responses as well as protection from challenge infection in a mouse model. A single intranasal administration of PanAd3-NPM1 vaccine induced strong antibody and T cell responses, and protected against high dose lethal influenza virus challenge. Thus PanAd3 is a promising candidate vector for vaccines, including universal influenza vaccines.
- Published
- 2013
- Full Text
- View/download PDF
31. Highly-Immunogenic Virally-Vectored T-cell Vaccines Cannot Overcome Subversion of the T-cell Response by HCV during Chronic Infection
- Author
-
Leo Swadling, John Halliday, Christabel Kelly, Anthony Brown, Stefania Capone, M. Azim Ansari, David Bonsall, Rachel Richardson, Felicity Hartnell, Jane Collier, Virginia Ammendola, Mariarosaria Del Sorbo, Annette Von Delft, Cinzia Traboni, Adrian V. S. Hill, Stefano Colloca, Alfredo Nicosia, Riccardo Cortese, Paul Klenerman, Antonella Folgori, and Eleanor Barnes
- Subjects
therapeutic vaccination ,adenovirus ,modified vaccinia Ankara ,immunotherapy ,HCV ,T-cells ,exhaustion ,Medicine - Abstract
An effective therapeutic vaccine for the treatment of chronic hepatitis C virus (HCV) infection, as an adjunct to newly developed directly-acting antivirals (DAA), or for the prevention of reinfection, would significantly reduce the global burden of disease associated with chronic HCV infection. A recombinant chimpanzee adenoviral (ChAd3) vector and a modified vaccinia Ankara (MVA), encoding the non-structural proteins of HCV (NSmut), used in a heterologous prime/boost regimen induced multi-specific, high-magnitude, durable HCV-specific CD4+ and CD8+ T-cell responses in healthy volunteers, and was more immunogenic than a heterologous Ad regimen. We now assess the immunogenicity of this vaccine regimen in HCV infected patients (including patients with a low viral load suppressed with interferon/ribavirin therapy), determine T-cell cross-reactivity to endogenous virus, and compare immunogenicity with that observed previously in both healthy volunteers and in HCV infected patients vaccinated with the heterologous Ad regimen. Vaccination of HCV infected patients with ChAd3-NSmut/MVA-NSmut was well tolerated. Vaccine-induced HCV-specific T-cell responses were detected in 8/12 patients; however, CD4+ T-cell responses were rarely detected, and the overall magnitude of HCV-specific T-cell responses was markedly reduced when compared to vaccinated healthy volunteers. Furthermore, HCV-specific cells had a distinct partially-functional phenotype (lower expression of activation markers, granzyme B, and TNFα production, weaker in vitro proliferation, and higher Tim3 expression, with comparable Tbet and Eomes expression) compared to healthy volunteers. Robust anti-vector T-cells and antibodies were induced, showing that there is no global defect in immunity. The level of viremia at the time of vaccination did not correlate with the magnitude of the vaccine-induced T-cell response. Full-length, next-generation sequencing of the circulating virus demonstrated that T-cells were only induced by vaccination when there was a sequence mismatch between the autologous virus and the vaccine immunogen. However, these T-cells were not cross-reactive with the endogenous viral variant epitopes. Conversely, when there was complete homology between the immunogen and circulating virus at a given epitope T-cells were not induced. T-cell induction following vaccination had no significant impact on HCV viral load. In vitro T-cell culture experiments identified the presence of T-cells at baseline that could be expanded by vaccination; thus, HCV-specific T-cells may have been expanded from pre-existing low-level memory T-cell populations that had been exposed to HCV antigens during natural infection, explaining the partial T-cell dysfunction. In conclusion, vaccination with ChAd3-NSmut and MVA-NSmut prime/boost, a potent vaccine regimen previously optimized in healthy volunteers was unable to reconstitute HCV-specific T-cell immunity in HCV infected patients. This highlights the major challenge of overcoming T-cell exhaustion in the context of persistent antigen exposure.
- Published
- 2016
- Full Text
- View/download PDF
32. Integrity of the Antiviral STING-mediated DNA Sensing in Tumor Cells Is Required to Sustain the Immunotherapeutic Efficacy of Herpes Simplex Oncolytic Virus
- Author
-
Carmen Caiazza, Massimo Mallardo, Anna Morena D'Alise, Nicola Zambrano, Guendalina Froechlich, Guido Leoni, Alfredo Nicosia, Vittorio Scisciola, Maria De Lucia, Emanuele Sasso, Francesca Langone, Chiara Gentile, Gabriella Cotugno, Elisa Scarselli, Froechlich, G., Caiazza, C., Gentile, C., D'Alise, A. M., De Lucia, M., Langone, F., Leoni, G., Cotugno, G., Scisciola, V., Nicosia, A., Scarselli, E., Mallardo, M., Sasso, E., and Zambrano, N.
- Subjects
0301 basic medicine ,Herpes simplex ,Cancer Research ,RNA profiling ,lcsh:RC254-282 ,Article ,MB21D ,TMEM173 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,STING Knockout ,Interferon ,immunogenic cell death ,medicine ,oncolytic virus ,Tumor microenvironment ,business.industry ,Oncolytic viru ,Acquired immune system ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,HSV-1 ,Immune checkpoint ,eye diseases ,Oncolytic virus ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Stimulator of interferon genes ,Cancer research ,Immunogenic cell death ,business ,medicine.drug - Abstract
Simple Summary Oncolytic viruses are emerging immunotherapeutics in cancer treatments. The conflicting role of innate immunity in the antitumor activity of oncolytic viruses is still a matter of debate. The STING-dependent DNA sensing axis is considered detrimental for viral replication and cancer cell clearance. Accordingly, we observed that STING loss in tumor cells was associated with improved lytic potential by a herpes-based oncolytic virus. However, STING-knockout cancer cells infected with the oncolytic virus showed impaired immunogenicity, as immunogenic cell death was improperly triggered. In agreement with these observations, STING-knockout tumors raised in a murine syngeneic model were more resistant to a combined treatment of the oncolytic virus with PD-1 blockade. The present study demonstrates the antitumor benefit of antiviral immunity and sheds lights on the mechanisms of immune resistance to oncovirotherapy exerted by STING-loss in tumor cells. Abstract The dichotomic contribution of cancer cell lysis and tumor immunogenicity is considered essential for effective oncovirotherapy, suggesting that the innate antiviral immune response is a hurdle for efficacy of oncolytic viruses. However, emerging evidence is resizing this view. By sensing cytosolic DNA, the cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) axis can both counteract viral spread and contribute to the elicitation of adaptive immunity via type I interferon responses. In this paper, we analyzed the tumor-resident function of Sting-mediated DNA sensing in a combined approach of oncovirotherapy and PD-1 immune checkpoint blockade, in an immunocompetent murine model. While supporting increased lytic potential by oncolytic HER2-retargeted HSV-1 in vitro and in vivo, Sting-knockout tumors showed molecular signatures of an immunosuppressive tumor microenvironment. These signatures were correspondingly associated with ineffectiveness of the combination therapy in a model of established tumors. Results suggest that the impairment in antiviral response of Sting-knockout tumors, while favoring viral replication, is not able to elicit an adequate immunotherapeutic effect, due to lack of immunogenic cell death and the inability of Sting-knockout cancer cells to promote anti-tumor adaptive immune responses. Accordingly, we propose that antiviral, tumor-resident Sting provides fundamental contributions to immunotherapeutic efficacy of oncolytic viruses.
- Published
- 2020
33. Optimising T cell (re)boosting strategies for adenoviral and modified vaccinia Ankara vaccine regimens in humans
- Author
-
Antonella Folgori, Stefano Colloca, Paul Klenerman, Cinzia Traboni, Eleanor Barnes, Stefania Capone, Alfredo Nicosia, Anthony Brown, Felicity Hartnell, Leo Swadling, Ventzislav Vassilev, Riccardo Cortese, Mariarosaria Del Sorbo, Capone, S., Brown, A., Hartnell, F., Sorbo, M. D., Traboni, C., Vassilev, V., Colloca, S., Nicosia, A., Cortese, R., Folgori, A., Klenerman, P., Barnes, E., and Swadling, L.
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Modified vaccinia Ankara ,animal structures ,Live attenuated vaccines ,T cell ,Hepatitis C virus ,viruses ,Immunology ,Adaptive immunity ,medicine.disease_cause ,lcsh:RC254-282 ,complex mixtures ,Article ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Medicine ,Pharmacology (medical) ,Pharmacology ,Vaccines ,Reactogenicity ,business.industry ,hemic and immune systems ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Acquired immune system ,Virology ,3. Good health ,Vaccination ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Live attenuated vaccine ,030220 oncology & carcinogenesis ,lcsh:RC581-607 ,business - Abstract
Simian adenoviral and modified vaccinia Ankara (MVA) viral vectors used in heterologous prime-boost strategies are potent inducers of T cells against encoded antigens and are in advanced testing as vaccine carriers for a wide range of infectious agents and cancers. It is unclear if these responses can be further enhanced or sustained with reboosting strategies. Furthermore, despite the challenges involved in MVA manufacture dose de-escalation has not been performed in humans. In this study, healthy volunteers received chimpanzee-derived adenovirus-3 and MVA vaccines encoding the non-structural region of hepatitis C virus (ChAd3-NSmut/MVA-NSmut) 8 weeks apart. Volunteers were then reboosted with a second round of ChAd3-NSmut/MVA-NSmut or MVA-NSmut vaccines 8 weeks or 1-year later. We also determined the capacity of reduced doses of MVA-NSmut to boost ChAd3-NSmut primed T cells. Reboosting was safe, with no enhanced reactogenicity. Reboosting after an 8-week interval led to minimal re-expansion of transgene-specific T cells. However, after a longer interval, T cell responses expanded efficiently and memory responses were enhanced. The 8-week interval regimen induced a higher percentage of terminally differentiated and effector memory T cells. Reboosting with MVA-NSmut alone was as effective as with ChAd3-NSmut/MVA-NSmut. A ten-fold lower dose of MVA (2 × 107pfu) induced high-magnitude, sustained, broad, and functional Hepatitis C virus (HCV)-specific T cell responses, equivalent to standard doses (2 × 108 pfu). Overall, we show that following Ad/MVA prime-boost vaccination reboosting is most effective after a prolonged interval and is productive with MVA alone. Importantly, we also show that a ten-fold lower dose of MVA is as potent in humans as the standard dose.
- Published
- 2020
34. New viral vectors for infectious diseases and cancer
- Author
-
Elisa Scarselli, Emanuele Sasso, Anna Morena D'Alise, Nicola Zambrano, Alfredo Nicosia, Antonella Folgori, Sasso, E., D'Alise, A. M., Zambrano, N., Scarselli, E., Folgori, A., and Nicosia, A.
- Subjects
Herpesviru ,0301 basic medicine ,Modified vaccinia Ankara ,viruses ,Arenaviru ,Poxviru ,Immunology ,Genetic Vectors ,Biology ,Cancer Vaccines ,Virus ,Viral vector ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Paramixoviru ,Neoplasms ,Cancer vaccine ,Immunology and Allergy ,Animals ,Humans ,Arenavirus ,Viral Vaccine ,Vaccination ,Immunity ,Oncolytic viru ,Viral Vaccines ,biology.organism_classification ,Virology ,Oncolytic virus ,030104 developmental biology ,chemistry ,Vesicular stomatitis virus ,Virus Diseases ,030220 oncology & carcinogenesis ,Viruses ,Adenoviru ,Genetic vaccine ,Rhabdoviru ,Vaccinia - Abstract
Since the discovery in 1796 by Edward Jenner of vaccinia virus as a way to prevent and finally eradicate smallpox, the concept of using a virus to fight another virus has evolved into the current approaches of viral vectored genetic vaccines. In recent years, key improvements to the vaccinia virus leading to a safer version (Modified Vaccinia Ankara, MVA) and the discovery that some viruses can be used as carriers of heterologous genes encoding for pathological antigens of other infectious agents (the concept of ‘viral vectors’) has spurred a new wave of clinical research potentially providing for a solution for the long sought after vaccines against major diseases such as HIV, TB, RSV and Malaria, or emerging infectious diseases including those caused by filoviruses and coronaviruses. The unique ability of some of these viral vectors to stimulate the cellular arm of the immune response and, most importantly, T lymphocytes with cell killing activity, has also reawakened the interest toward developing therapeutic vaccines against chronic infectious diseases and cancer. To this end, existing vectors such as those based on Adenoviruses have been improved in immunogenicity and efficacy. Along the same line, new vectors that exploit viruses such as Vesicular Stomatitis Virus (VSV), Measles Virus (MV), Lymphocytic choriomeningitis virus (LCMV), cytomegalovirus (CMV), and Herpes Simplex Virus (HSV), have emerged. Furthermore, technological progress toward modifying their genome to render some of these vectors incompetent for replication has increased confidence toward their use in infant and elderly populations. Lastly, their production process being the same for every product has made viral vectored vaccines the technology of choice for rapid development of vaccines against emerging diseases and for ‘personalised’ cancer vaccines where there is an absolute need to reduce time to the patient from months to weeks or days. Here we review the recent developments in viral vector technologies, focusing on novel vectors based on primate derived Adenoviruses and Poxviruses, Rhabdoviruses, Paramixoviruses, Arenaviruses and Herpesviruses. We describe the rationale for, immunologic mechanisms involved in, and design of viral vectored gene vaccines under development and discuss the potential utility of these novel genetic vaccine approaches in eliciting protection against infectious diseases and cancer.
- Published
- 2020
35. 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
36. 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
37. Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors.
- Author
-
Susanne H Sheehy, Christopher J A Duncan, Sean C Elias, Sumi Biswas, Katharine A Collins, Geraldine A O'Hara, Fenella D Halstead, Katie J Ewer, Tabitha Mahungu, Alexandra J Spencer, Kazutoyo Miura, Ian D Poulton, Matthew D J Dicks, Nick J Edwards, Eleanor Berrie, Sarah Moyle, Stefano Colloca, Riccardo Cortese, Katherine Gantlett, Carole A Long, Alison M Lawrie, Sarah C Gilbert, Tom Doherty, Alfredo Nicosia, Adrian V S Hill, and Simon J Draper
- Subjects
Medicine ,Science - Abstract
Traditionally, vaccine development against the blood-stage of Plasmodium falciparum infection has focused on recombinant protein-adjuvant formulations in order to induce high-titer growth-inhibitory antibody responses. However, to date no such vaccine encoding a blood-stage antigen(s) alone has induced significant protective efficacy against erythrocytic-stage infection in a pre-specified primary endpoint of a Phase IIa/b clinical trial designed to assess vaccine efficacy. Cell-mediated responses, acting in conjunction with functional antibodies, may be necessary for immunity against blood-stage P. falciparum. The development of a vaccine that could induce both cell-mediated and humoral immune responses would enable important proof-of-concept efficacy studies to be undertaken to address this question.We conducted a Phase Ia, non-randomized clinical trial in 16 healthy, malaria-naïve adults of the chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient viral vectored vaccines encoding two alleles (3D7 and FVO) of the P. falciparum blood-stage malaria antigen; apical membrane antigen 1 (AMA1). ChAd63-MVA AMA1 administered in a heterologous prime-boost regime was shown to be safe and immunogenic, inducing high-level T cell responses to both alleles 3D7 (median 2036 SFU/million PBMC) and FVO (median 1539 SFU/million PBMC), with a mixed CD4(+)/CD8(+) phenotype, as well as substantial AMA1-specific serum IgG responses (medians of 49 µg/mL and 41 µg/mL for 3D7 and FVO AMA1 respectively) that demonstrated growth inhibitory activity in vitro.ChAd63-MVA is a safe and highly immunogenic delivery platform for both alleles of the AMA1 antigen in humans which warrants further efficacy testing. ChAd63-MVA is a promising heterologous prime-boost vaccine strategy that could be applied to numerous other diseases where strong cellular and humoral immune responses are required for protection.ClinicalTrials.gov NCT01095055.
- Published
- 2012
- Full Text
- View/download PDF
38. A long non-coding SINEUP RNA boosts semi-stable production of fully human monoclonal antibodies in HEK293E cells
- Author
-
Claudia De Lorenzo, Emanuele Sasso, Nicola Zambrano, Guendalina Froechlich, Alfredo Nicosia, Margherita Passariello, Mariangela Succoio, Debora Latino, Sasso, Emanuele, Latino, Debora, Froechlich, Guendalina, Succoio, Mariangela, Passariello, Margherita, De Lorenzo, Claudia, Nicosia, Alfredo, and Zambrano, Nicola
- Subjects
0301 basic medicine ,Glycosylation ,glycosylation ,medicine.drug_class ,Immunology ,Biology ,Monoclonal antibody ,scFv ,03 medical and health sciences ,chemistry.chemical_compound ,lncRNA ,SINEUP ,Peptide Library ,Cell Line, Tumor ,Claudin-1 ,medicine ,Humans ,Immunology and Allergy ,monoclonal antibodie ,antibody production ,IgG4 ,Antibodies, Monoclonal ,RNA ,Virology ,Antibody production ,HEK293 Cells ,030104 developmental biology ,Gene Expression Regulation ,chemistry ,CLDN1 ,HEK293E ,Immunoglobulin Light Chains ,RNA, Long Noncoding ,Immunoglobulin Heavy Chains - Abstract
Use of monoclonal antibodies is emerging as a highly promising and fast-developing scenario for innovative treatment of viral, autoimmune and tumour diseases. The search for diagnostic and therapeutic antibodies currently depends on in vitro screening approaches, such as phage and yeast display technologies. Antibody production still represents a critical step for preclinical and clinical evaluations. Accordingly, improving production of monoclonal antibodies represents an opportunity, to facilitate downstream target validations. SINEUP RNAs are long non-coding transcripts, possessing the ability to enhance translation of selected mRNAs. We applied SINEUP technology to semi-stable production of monoclonal antibodies in HEK293E cells, which allows for episomal propagation of the expression vectors encoding the heavy and light chains of IgGs. Co-expression of SINEUP RNA with mRNAs encoding heavy and light chains of IgG4s was able to increase the production of different anti-CLDN1 antibodies up to three-fold. Improved production of monoclonal antibodies was achieved both in transiently transfected HEK293E cells and in cellular clones with stable expression of the SINEUP. Compared to antibody preparations obtained under standard conditions, the anti-CLDN1 IgG4s produced in the presence of the SINEUP transcript showed unaltered post-translational modifications, and retained the ability to recognize their target. We thus propose SINEUP technology as a valuable tool to enhance semi-stable antibody production in human cell lines.
- Published
- 2018
39. Differential screening of phage-ab libraries by oligonucleotide microarray technology.
- Author
-
Paolo Monaci, Alessandra Luzzago, Claudia Santini, Alessandra De Pra, Mirko Arcuri, Francesca Magistri, Alessandro Bellini, Helenia Ansuini, Maria Ambrosio, Virginia Ammendola, Maria Giulia Bigotti, Agostino Cirillo, Maurizio Nuzzo, Annamaria Assunta Nasti, Philippe Neuner, Laura Orsatti, Monica Pezzanera, Andrea Sbardellati, Giuseppe Silvestre, Paolo Uva, Valentina Viti, Gaetano Barbato, Stefano Colloca, Anna Demartis, Emanuele De Rinaldis, Saverio Giampaoli, Armin Lahm, Fabio Palombo, Fabio Talamo, Alessandra Vitelli, Alfredo Nicosia, and Riccardo Cortese
- Subjects
Medicine ,Science - Abstract
A novel and efficient tagArray technology was developed that allows rapid identification of antibodies which bind to receptors with a specific expression profile, in the absence of biological information. This method is based on the cloning of a specific, short nucleotide sequence (tag) in the phagemid coding for each phage-displayed antibody fragment (phage-Ab) present in a library. In order to set up and validate the method we identified about 10,000 different phage-Abs binding to receptors expressed in their native form on the cell surface (10 k Membranome collection) and tagged each individual phage-Ab. The frequency of each phage-Ab in a given population can at this point be inferred by measuring the frequency of its associated tag sequence through standard DNA hybridization methods. Using tiny amounts of biological samples we identified phage-Abs binding to receptors preferentially expressed on primary tumor cells rather than on cells obtained from matched normal tissues. These antibodies inhibited cell proliferation in vitro and tumor development in vivo, thus representing therapeutic lead candidates.
- Published
- 2008
- Full Text
- View/download PDF
40. 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
41. Mucosal Vaccination with Heterologous Viral Vectored Vaccine Targeting Subdominant SIV Accessory Antigens Strongly Inhibits Early Viral Replication
- Author
-
Alfredo Nicosia, James Blanchard, Benjamin A. H. Jensen, Ronald S. Veazey, Stefano Colloca, Jan Pravsgaard Christensen, Emeline Ragonnaud, Allan Randrup Thomsen, Anders Tolver, Antonella Folgori, Peter Johannes Holst, Ditte Rahbæk Boilesen, Huanbin Xu, Anne-Marie C. Andersson, Riccardo Cortese, Xu, H, Andersson, Am, Ragonnaud, E, Boilesen, D, Tolver, A, Jensen, Bah, Blanchard, Jl, Nicosia, A, Folgori, A, Colloca, S, Cortese, R, Thomsen, Ar, Christensen, Jp, Veazey, R, and Holst, Pj.
- Subjects
0301 basic medicine ,Naive T cell ,T cell ,lcsh:Medicine ,Viremia ,Heterologous viral vectored prime-boost immunization ,Genetic adjuvant ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,lcsh:R5-920 ,lcsh:R ,virus diseases ,General Medicine ,medicine.disease ,Virology ,3. Good health ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Viral replication ,Immunology ,lcsh:Medicine (General) ,T-cell vaccine ,030215 immunology - Abstract
Conventional HIV T cell vaccine strategies have not been successful in containing acute peak viremia, nor in providing long-term control. We immunized rhesus macaques intramuscularly and rectally using a heterologous adenovirus vectored SIV vaccine regimen encoding normally weakly immunogenic tat, vif, rev and vpr antigens fused to the MHC class II associated invariant chain. Immunizations induced broad T cell responses in all vaccinees. Following up to 10 repeated low-dose intrarectal challenges, vaccinees suppressed early viral replication (P=0.01) and prevented the peak viremia in 5/6 animals. Despite consistently undetectable viremia in 2 out of 6 vaccinees, all animals showed evidence of infection induced immune responses indicating that infection had taken place. Vaccinees, with and without detectable viremia better preserved their rectal CD4+ T cell population and had reduced immune hyperactivation as measured by naive T cell depletion, Ki-67 and PD-1 expression on T cells. These results indicate that vaccination towards SIV accessory antigens vaccine can provide a level of acute control of SIV replication with a suggestion of beneficial immunological consequences in infected animals of unknown long-term significance. In conclusion, our studies demonstrate that a vaccine encoding subdominant antigens not normally associated with virus control can exert a significant impact on acute peak viremia.
- Published
- 2017
42. Chimpanzee Adenovirus Vector Ebola Vaccine
- Author
-
Julie E, Ledgerwood, Adam D, DeZure, Daphne A, Stanley, Emily E, Coates, Laura, Novik, Mary E, Enama, Nina M, Berkowitz, Zonghui, Hu, Gyan, Joshi, Aurélie, Ploquin, Sandra, Sitar, Ingelise J, Gordon, Sarah A, Plummer, LaSonji A, Holman, Cynthia S, Hendel, Galina, Yamshchikov, Francois, Roman, Alfredo, Nicosia, Stefano, Colloca, Riccardo, Cortese, Robert T, Bailer, Richard M, Schwartz, Mario, Roederer, John R, Mascola, Richard A, Koup, Nancy J, Sullivan, Barney S, Graham, and Sarah, Romano
- Subjects
Adult ,Male ,0301 basic medicine ,Fever ,Pan troglodytes ,T-Lymphocytes ,Genetic Vectors ,Simian ,Antibodies, Viral ,medicine.disease_cause ,03 medical and health sciences ,Animals ,Humans ,Medicine ,Vector (molecular biology) ,Ebola Vaccines ,Glycoproteins ,Ebolavirus ,Ebola virus ,Ebola vaccine ,biology ,business.industry ,Immunogenicity ,General Medicine ,Hemorrhagic Fever, Ebola ,Middle Aged ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,Vaccination ,Clinical trial ,030104 developmental biology ,Immunology ,Adenoviruses, Simian ,business - Abstract
BackgroundThe unprecedented 2014 epidemic of Ebola virus disease (EVD) prompted an international response to accelerate the availability of a preventive vaccine. A replication-defective recombinant chimpanzee adenovirus type 3–vectored ebolavirus vaccine (cAd3-EBO), encoding the glycoprotein from Zaire and Sudan species, that offers protection in the nonhuman primate model, was rapidly advanced into phase 1 clinical evaluation. MethodsWe conducted a phase 1, dose-escalation, open-label trial of cAd3-EBO. Twenty healthy adults, in sequentially enrolled groups of 10 each, received vaccination intramuscularly in doses of 2×1010 particle units or 2×1011 particle units. Primary and secondary end points related to safety and immunogenicity were assessed throughout the first 8 weeks after vaccination; in addition, longer-term vaccine durability was assessed at 48 weeks after vaccination. ResultsIn this small study, no safety concerns were identified; however, transient fever developed within 1 day after vaccinat...
- Published
- 2017
43. Novel Human Anti-PD-L1 mAbs Inhibit Immune-Independent Tumor Cell Growth and PD-L1 Associated Intracellular Signalling
- Author
-
Alfredo Nicosia, Margherita Passariello, Elisa Scarselli, Guendalina Froechlich, Cinzia Vetrei, Anna Morena D'Alise, Annachiara Esposito, Claudia De Lorenzo, Passariello, M, D'Alise, Am, Esposito, A, Vetrei, C, Froechlich, Guendalina, Scarselli, E, Nicosia, A, and De Lorenzo, C.
- Subjects
0301 basic medicine ,Receptor, ErbB-2 ,medicine.drug_class ,Science ,medicine.medical_treatment ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Article ,B7-H1 Antigen ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Atezolizumab ,Target identification ,PD-L1 ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cancer ,Cell Proliferation ,Mice, Inbred BALB C ,Multidisciplinary ,Molecular medicine ,biology ,Chemistry ,Cell growth ,Immunotherapy ,Trastuzumab ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Medicine ,Female ,Antibody - Abstract
The novel antibody-based immunotherapy in oncology exploits the activation of immune system mediated by immunomodulatory antibodies specific for immune checkpoints. Among them, the programmed death ligand-1 (PD-L1) is of particular interest as it is expressed not only on T-cells, but also on other immune cells and on a large variety of cancer cells, such as breast cancer cells, considering its high expression in both ErbB2-positive and Triple Negative Breast Cancers. We demonstrate here that PD-L1_1, a novel anti-PD-L1 T -cell stimulating antibody, inhibits PD-L1-tumor cell growth also by affecting the intracellular MAPK pathway and by activating caspase 3. Similar in vitro results were obtained for the first time here also with the clinically validated anti-PD-L1 mAb Atezolizumab and in vivo with another validated anti-mouse anti-PD-L1 mAb. Moreover, we found that two high affinity variants of PD-L1_1 inhibited tumor cell viability more efficiently than the parental PD-L1_1 by affecting the same MAPK pathways with a more potent effect. Altogether, these results shed light on the role of PD-L1 in cancer cells and suggest that PD-L1_1 and its high affinity variants could become powerful antitumor weapons to be used alone or in combination with other drugs such as the anti-ErbB2 cAb already successfully tested in in vitro combinatorial treatments.
- Published
- 2019
44. Rapid Affinity Maturation of Novel Anti-PD-L1 Antibodies by a Fast Drop of the Antigen Concentration and FACS Selection of Yeast Libraries
- Author
-
Claudia De Lorenzo, Valentino Ruzza, Nicola Zambrano, Riccardo Cortese, Margherita Passariello, Fulvia Troise, Emanuele Sasso, Elisa Scarselli, Luigi Del Vecchio, Feliciano Visconte, Maria Luisa Esposito, Anna Morena D'Alise, Alfredo Nicosia, Valeria Cafaro, Maddalena Raia, Biancamaria Cembrola, Cembrola, B, Ruzza, V, Troise, F, Esposito, Ml, Sasso, E, Cafaro, V, Passariello, M, Visconte, F, Raia, M, Del Vecchio, L, D'Alise, Am, Cortese, R, Scarselli, E, Zambrano, N, De Lorenzo, C, and Nicosia, A
- Subjects
Phage display ,Article Subject ,medicine.drug_class ,Antibody Affinity ,lcsh:Medicine ,Complementarity determining region ,Lymphocyte proliferation ,Saccharomyces cerevisiae ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,B7-H1 Antigen ,Cell Line ,Affinity maturation ,Antigen ,Peptide Library ,medicine ,Humans ,Lymphocytes ,Peptide library ,Cell Proliferation ,General Immunology and Microbiology ,Base Sequence ,Chemistry ,lcsh:R ,Antibodies, Monoclonal ,High-Throughput Nucleotide Sequencing ,General Medicine ,Surface Plasmon Resonance ,Flow Cytometry ,Complementarity Determining Regions ,Biochemistry ,Mutagenesis ,Immunoglobulin G ,Single-Chain Antibodies ,Research Article - Abstract
The affinity engineering is a key step to increase the efficacy of therapeutic monoclonal antibodies and yeast surface display is the most widely used and powerful affinity maturation approach, achieving picomolar binding affinities. In this study, we provide an optimization of the yeast surface display methodology, applied to the generation of potentially therapeutic high affinity antibodies targeting the immune checkpoint PD-L1. In this approach, we coupled a 10-cycle error-prone mutagenesis of heavy chain complementarity determining region 3 of an anti‐PD-L1 scFv, previously identified by phage display, with high-throughput sequencing, to generate scFv-yeast libraries with high mutant frequency and diversity. In addition, we set up a novel, faster and effective selection scheme by fluorescence-activated cell sorting, based on a fast drop of the antigen concentration between the first and the last selection cycles, unlike the gradual decrease typical of current selection protocols. In this way we isolated 6 enriched mutated scFv-yeast clones overall, showing an affinity improvement for soluble PD-L1 protein compared to the parental scFv. As a proof of the potency of the novel approach, we confirmed that the antibodies converted from all the mutated scFvs retained the affinity improvement. Remarkably, the best PD-L1 binder among them also bound with a higher affinity to PD-L1 expressed in its native conformation on human-activated lymphocytes, and it was able to stimulate lymphocyte proliferation in vitro more efficiently than its parental antibody. This optimized technology, besides the identification of a new potential checkpoint inhibitor, provides a tool for the quick isolation of high affinity binders.
- Published
- 2019
45. Novel genetically-modified chimpanzee adenovirus and MVA-vectored respiratory syncytial virus vaccine safely boosts humoral and cellular immunity in healthy older adults
- Author
-
L Silva-Reyes, Elisa Scarselli, Alfredo Nicosia, Stefania Capone, Paul Klenerman, Tamsin Cargill, C de Lara, Andrew J. Pollard, Charles J. Sande, Amber J. Thompson, Kathryn Mary Taylor, Claire Hutchings, Brian Angus, Christopher A Green, Alessandra Vitelli, K Haworth, Green, Christopher A., Sande, Charles J., Scarselli, Elisa, Capone, Stefania, Vitelli, Alessandra, Nicosia, Alfredo, Silva-Reyes, Laura, Thompson, Amber J., de Lara, Catherine M., Taylor, Kathryn S., Haworth, Kathryn, Hutchings, Claire L., Cargill, Tamsin, Angus, Brian, Klenerman, Paul, and Pollard, Andrew J.
- Subjects
Viral vectors ,Male ,0301 basic medicine ,Cellular immunity ,T-Lymphocytes ,viruses ,Respiratory syncytial virus ,Antibodies, Viral ,Elderly ,0302 clinical medicine ,Medicine ,030212 general & internal medicine ,Older adult ,B-Lymphocytes ,Drug Carriers ,Immunity, Cellular ,Vaccines, Synthetic ,Immunogenicity ,ELISPOT ,virus diseases ,Respiratory infection ,respiratory system ,Middle Aged ,Healthy Volunteers ,Vaccination ,Infectious Diseases ,Older adults ,Respiratory syncytial viru ,Female ,Adult ,Microbiology (medical) ,Adolescent ,Drug-Related Side Effects and Adverse Reactions ,030106 microbiology ,Vaccinia virus ,Respiratory Syncytial Virus Infections ,Injections, Intramuscular ,Article ,Mastadenovirus ,Young Adult ,03 medical and health sciences ,Viral vector ,Immune system ,Immunity ,Respiratory Syncytial Virus Vaccines ,Humans ,Antibody-Producing Cells ,Administration, Intranasal ,Immunization Schedule ,Aged ,business.industry ,Vaccine trial ,Antibodies, Neutralizing ,Immunity, Humoral ,Respiratory Syncytial Virus, Human ,Immunology ,business ,Vaccine - Abstract
Highlights • There is no licensed vaccine to prevent severe disease caused by respiratory syncytial virus (RSV) infection. • RSV is a major cause of hospitalisation and death in the elderly. • The novel viral-vectored vaccines PanAd3-RSV and MVA-RSV appeared safe and boosted both humoral and cellular RSV-specific immune responses in healthy older adults. • The magnitude of immune responses to vaccination appeared similar to what was observed in younger adults., Objectives Respiratory syncytial virus (RSV) causes respiratory infection across the world, with infants and the elderly at particular risk of developing severe disease and death. The replication-defective chimpanzee adenovirus (PanAd3-RSV) and modified vaccinia virus Ankara (MVA-RSV) vaccines were shown to be safe and immunogenic in young healthy adults. Here we report an extension to this first-in-man vaccine trial to include healthy older adults aged 60–75 years. Methods We evaluated the safety and immunogenicity of a single dose of MVA-RSV given by intra-muscular (IM) injection (n = 6), two doses of IM PanAd3-RSV given 4-weeks apart (n = 6), IM PanAd3-RSV prime and IM MVA-RSV boost 8-weeks later (n = 6), intra-nasal (IN) spray of PanAd3-RSV prime and IM MVA-RSV boost 8-weeks later (n = 6), or no vaccine (n = 6). Safety measures included all adverse events within one week of vaccination and blood monitoring. Immunogenicity measures included serum antibody responses (RSV- and PanAd3-neutralising antibody titres measured by plaque-reduction neutralisation and SEAP assays, respectively), peripheral B-cell immune responses (frequencies of F-specific IgG and IgA antibody secreting cells and memory B-cells by ex vivo and cultured dual-colour ELISpot assays respectively), and peripheral RSV-specific T-cell immune responses (frequencies of IFNγ-producing T-cells by ex vivo ELISpot and CD4+/CD8+/Tfh-like cell frequencies by ICS/FACS assay). Results The vaccines were safe and well tolerated. Compared with each individual baseline immunity the mean fold-changes in serum RSV-neutralising antibody, appearance and magnitude of F-specific IgG and IgA ASCs and expansion of CD4+/CD8+ IFNγ-producing T-cells in peripheral circulation were comparable to the results seen from younger healthy adults who received the same vaccine combination and dose. There were little/no IgA memory B-cell responses in younger and older adults. Expansion of IFNγ-producing T-cells was most marked in older adults following IM prime, with balanced CD4+ and CD8+ T cell responses. The RSV-specific immune responses to vaccination did not appear to be attenuated in the presence of PanAd3 (vector) neutralising antibody. Conclusions PanAd3-RSV and MVA-RSV was safe and immunogenic in older adults and the parallel induction of RSV-specific humoral and cellular immunity merits further assessment in providing protection from severe disease.
- Published
- 2019
46. Antigen‐specific CD8 T cells in cell cycle circulate in the blood after vaccination
- Author
-
Alfredo Nicosia, Ambra Natalini, Francesca Di Rosa, Stefania Capone, Antonella Folgori, Sonia Simonetti, Angela Santoni, Simonetti, Sonia, Natalini, Ambra, Folgori, Antonella, Capone, Stefania, Nicosia, Alfredo, Santoni, Angela, and Di Rosa, Francesca
- Subjects
0301 basic medicine ,spleen and lymph nodes ,Cell ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,spleen and lymph node ,Mice ,0302 clinical medicine ,HEK293 Cell ,Cytotoxic T cell ,Mice, Inbred BALB C ,medicine.diagnostic_test ,viral vector ,antigen-specific response ,Cell Cycle ,Vaccination ,Lymph Node ,General Medicine ,Cell cycle ,Acquired immune system ,Flow Cytometry ,medicine.anatomical_structure ,flow cytometry analysis ,Antigen ,Blood Circulation ,Viruses ,Female ,Lymph ,Genetic Vector ,Human ,viral vectors ,Cell Survival ,Immunology ,Genetic Vectors ,T cells ,Spleen ,clonal expansion ,Biology ,Flow cytometry ,Viral vector ,03 medical and health sciences ,blood ,medicine ,Animals ,Humans ,antigen‐specific response ,Antigens ,Cell Proliferation ,Experimental Immunology ,Animal ,Viruse ,vaccination ,T cell ,flow cytometry analysi ,CD8-Positive T-Lymphocyte ,DNA ,Molecular biology ,030104 developmental biology ,HEK293 Cells ,Ki-67 Antigen ,Lymph Nodes ,030215 immunology - Abstract
Although clonal expansion is a hallmark of adaptive immunity, the location(s) where antigen‐responding T cells enter cell cycle and complete it have been poorly explored. This lack of knowledge stems partially from the limited experimental approaches available. By using Ki67 plus DNA staining and a novel strategy for flow cytometry analysis, we distinguished antigen‐specific CD8 T cells in G0, in G1 and in S‐G2/M phases of cell cycle after intramuscular vaccination of BALB/c mice with antigen‐expressing viral vectors. Antigen‐specific cells in S‐G2/M were present at early times after vaccination in lymph nodes (LNs), spleen and, surprisingly, also in the blood, which is an unexpected site for cycling of normal non‐leukaemic cells. Most proliferating cells had high scatter profile and were undetected by current criteria of analysis, which under‐estimated up to 6 times antigen‐specific cell frequency in LNs. Our discovery of cycling antigen‐specific CD8 T cells in the blood opens promising translational perspectives.
- Published
- 2019
47. Longitudinal transcriptomic and genetic landscape of radiotherapy response in canine melanoma
- Author
-
Piero Fariselli, Diana Giannuzzi, Mauro Dacasto, Ramy Elgendy, Mery Giantin, Laura Marconato, Vito F. Leone, Alfredo Nicosia, P Laganga, Sara Pegolo, Guido Leoni, Elisa Scarselli, Serena Ferraresso, Fulvia Troise, Luca Aresu, Giannuzzi D., Marconato L., Elgendy R., Ferraresso S., Scarselli E., Fariselli P., Nicosia A., Pegolo S., Leoni G., Laganga P., Leone V.F., Giantin M., Troise F., Dacasto M., Aresu L., Giannuzzi, Diana, Marconato, Laura, Ramy, Elgendy, Ferraresso, Serena, Scarselli, Elisa, Fariselli, Piero, Nicosia, Alfredo, Pegolo, Sara, Leoni, Guido, Laganga, Paola, Ferdinando, Leone Vito, Giantin, Mery, Troise, Fulvia, Dacasto, Mauro, and Aresu, Luca
- Subjects
Male ,040301 veterinary sciences ,medicine.medical_treatment ,Biology ,radiation therapy ,0403 veterinary science ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Dogs ,medicine ,Canine Melanoma ,melanoma ,Animals ,Dog Diseases ,Exome ,Exome sequencing ,PI3K/AKT/mTOR pathway ,Chromosome Aberrations ,General Veterinary ,Base Sequence ,Melanoma ,Gene Expression Profiling ,Cancer ,04 agricultural and veterinary sciences ,medicine.disease ,Radiation therapy ,Gene Expression Regulation, Neoplastic ,Dog ,RNA-seq ,030220 oncology & carcinogenesis ,Mutation ,dog ,Cancer research ,Female ,exome sequencing - Abstract
Canine malignant melanoma is a highly aggressive tumor with a low survival rate and represents an ideal spontaneous model for the human counterpart. Considerable progress has been recently obtained, but the therapeutic success for canine melanoma is still challenging. Little is known about the mechanisms beyond pathogenesis and melanoma development, and the molecular response to radiotherapy has never been explored before. A faster and deeper understanding of cancer mutational processes and developing mechanisms are now possible through next generation sequencing technologies. In this study, we matched whole exome and transcriptome sequencing in four dogs affected by malignant melanoma at diagnosis and at disease progression to identify possible genetic mechanisms associated with therapy failure. According to previous studies, a genetic similarity between canine malignant melanoma and its human counterpart was observed. Several somatic mutations were functionally related to MAPK, PI3K/AKT and p53 signaling pathways, but located in genes other than BRAF, RAS and KIT. At disease progression, several mutations were related to therapy effects. Natural killer cell-mediated cytotoxicity and several immune-system-related pathways resulted activated opening a new scenario on the microenvironment in this tumor. In conclusion, this study suggests a potential role of the immune system associated to radiotherapy in canine melanoma, but a larger sample size associated with functional studies are needed. This article is protected by copyright. All rights reserved.
- Published
- 2019
48. Persistent hepatitis C viral replication despite priming of functional CD8+ T cells by combined therapy with a vaccine and a direct‐acting antiviral
- Author
-
Benoit Callendret, Riccardo Cortese, William C. Satterfield, Antonella Folgori, Stefania Capone, Alfredo Nicosia, Christopher M. Walker, Heather B. Eccleston, B., Callendret, Hb., Eccleston, W., Satterfield, S., Capone, A., Folgori, Nicosia, Alfredo, and Cm., Walker
- Subjects
Viral Hepatitis Vaccines ,0301 basic medicine ,Pan troglodytes ,Hepacivirus ,CD8-Positive T-Lymphocytes ,Viral Nonstructural Proteins ,Biology ,Virus Replication ,Antiviral Agents ,Epitope ,Virus ,03 medical and health sciences ,Immune system ,Interferon ,medicine ,Animals ,Cytotoxic T cell ,Hepatology ,Vaccination ,virus diseases ,Hepatitis C, Chronic ,Virology ,030104 developmental biology ,Viral replication ,Immunology ,CD8 ,medicine.drug - Abstract
Exhaustion of antiviral CD8(+) T cells contributes to persistence of hepatitis C viral (HCV) infection. This immune response has proved difficult to restore by therapeutic vaccination, even when HCV replication is suppressed using antiviral regimens containing type I interferon. Because immunomodulatory effects of type I interferon may be a factor in poor T-cell priming, we undertook therapeutic vaccination in two chronically infected chimpanzees during treatment with a direct-acting antiviral (DAA) targeting the HCV NS5b polymerase protein. Immunization with genetic vaccines encoding the HCV NS3-NS5b nonstructural proteins during DAA treatment resulted in a multifunctional CD8(+) T-cell response. However, these antiviral CD8(+) T cells did not prevent persistent replication of DAA-resistant HCV variants that emerged during treatment. Most vaccine-induced CD8(+) T cells targeted class I epitopes that were not conserved in the circulating virus. Exhausted intrahepatic CD8(+) T-cell targeting-conserved epitopes did not expand after vaccination, with a notable exception. A sustained, multifunctional CD8(+) T-cell response against at least one intact class I epitope was detected in blood after vaccination. Persistence of HCV was not due to mutational escape of this epitope. Instead, failure to control HCV replication was likely caused by localized exhaustion in the liver, where CD8(+) T-cell expression of the inhibitory receptor programmed cell death 1 increased 25-fold compared with those in circulation. CONCLUSION: Treatment with a DAA during therapeutic vaccination provided transient control of HCV replication and a multifunctional T-cell response, primarily against nonconserved class I epitopes; exhaustion of liver-infiltrating CD8(+) T cells that target conserved epitopes may not be averted when DAA therapy fails prematurely due to emergence of resistant HCV variants.
- Published
- 2015
49. Isolation of Two Novel Human Anti-CTLA-4 mAbs with Intriguing Biological Properties on Tumor and NK Cells
- Author
-
Margherita Passariello, Claudia De Lorenzo, Cinzia Vetrei, Anna Morena D'Alise, Alfredo Nicosia, Elisa Scarselli, Guendalina Froechlich, Emanuele Sasso, Chiara Gentile, Nicola Zambrano, Passariello, M, Vetrei, C, Sasso, E, Froechlich, G, Gentile, C, D'Alise, Am, Zambrano, N, Scarselli, E, Nicosia, A, and De Lorenzo, C.
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Natural killer cell ,chemical and pharmacologic phenomena ,Ipilimumab ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,Cytotoxic T cell ,immune checkpoint ,Immunomodulatory mAb ,Antibody-dependent cell-mediated cytotoxicity ,cancer immunotherapy ,natural killer cells ,biology ,Chemistry ,CD28 ,hemic and immune systems ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,030104 developmental biology ,Oncology ,CTLA-4 ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,immunomodulatory mAbs ,Antibody ,medicine.drug - Abstract
The cytotoxic T lymphocyte-antigen 4 (CTLA-4) has been considered an IC exclusively expressed on T cells, where it counteracts the co-stimulatory CD28 receptor, by competing for its binding to CD-80 and CD-86. We recently found that it is expressed also on tumor and NK cells, suggesting other possible unknown roles of CTLA-4. To shed light on these novel aspects of CTLA-4, we used Ipilimumab, the first FDA approved human antibody targeting CTLA-4, in parallel studies with two novel human mAbs we isolated by using an efficient phage display selection strategy on live activated lymphocytes and purified mouse and human CTLA-4. The selection for cross-reactive mAbs was guaranteed by a high throughput sequencing to identify the sequences commonly enriched by two parallel pannings on human and mouse CTLA-4. Two isolated antibodies were found to bind with high affinity to both human and mouse CTLA-4 and lymphocytes, showing nanomolar or sub-nanomolar Kd values. They were able to kill Treg cells by ADCC, and to activate both human and mouse PBMCs, by strongly increasing cytokines secretion. Interestingly, they activated NK cells, exhibited cytotoxicity against cancer cells by inducing ADCC and inhibited tumor cell growth by affecting CTLA-4 downstream pathways in a similar fashion to CD-80 and CD-86 ligands and differently from Ipilimumab. Moreover, the novel mAbs showed a reduced ability to interfere in the binding of CD-80 ligands to CTLA-4 on T cells with respect to Ipilimumab, suggesting that they could allow for anti-tumor effects without the irAEs associated with the potent antagonistic activity of Ipilimumab.
- Published
- 2020
50. Adenoviral vaccine targeting multiple neoantigens as strategy to eradicate large tumors combined with checkpoint blockade
- Author
-
Adriano Leuzzi, Anna Morena D'Alise, Maria De Lucia, Antonella Folgori, Maria Teresa Catanese, Stefano Colloca, Veronica Bignone, Francesca Langone, Valeria Poli, Lidia Avalle, Elena Di Matteo, Fulvia Troise, Gabriella Cotugno, Fabio Giovanni Tucci, Imma Fichera, Elisa Scarselli, Alfredo Nicosia, Rosa Maria Vitale, Guido Leoni, and Armin Lahm
- Subjects
Cancer therapy ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,General Physics and Astronomy ,Cancer immunotherapy ,Lymphocyte Activation ,Mice ,Antineoplastic Agents, Immunological ,Neoplasms ,Gene expression ,Medicine ,Lymphocytes ,lcsh:Science ,Vaccines ,Vaccines, Synthetic ,Multidisciplinary ,Tumor ,Combined Modality Therapy ,Tumor Burden ,Vaccination ,medicine.anatomical_structure ,Immunological ,Treatment Outcome ,Female ,Immunotherapy ,T cell ,Science ,Antineoplastic Agents ,Cancer Vaccines ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Adenoviridae ,Lymphocytes, Tumor-Infiltrating ,Antigen ,Immunity ,Antigens, Neoplasm ,Cell Line, Tumor ,Animals ,Humans ,Tumor-Infiltrating ,Antigens ,business.industry ,Disease Models, Animal ,Viral Vaccines ,Animal ,Synthetic ,Cancer ,General Chemistry ,medicine.disease ,Blockade ,Cell culture ,Tumour vaccines ,Disease Models ,Cancer research ,Neoplasm ,lcsh:Q ,business - Abstract
Neoantigens (nAgs) are promising tumor antigens for cancer vaccination with the potential of inducing robust and selective T cell responses. Genetic vaccines based on Adenoviruses derived from non-human Great Apes (GAd) elicit strong and effective T cell-mediated immunity in humans. Here, we investigate for the first time the potency and efficacy of a novel GAd encoding multiple neoantigens. Prophylactic or early therapeutic vaccination with GAd efficiently control tumor growth in mice. In contrast, combination of the vaccine with checkpoint inhibitors is required to eradicate large tumors. Gene expression profile of tumors in regression shows abundance of activated tumor infiltrating T cells with a more diversified TCR repertoire in animals treated with GAd and anti-PD1 compared to anti-PD1. Data suggest that effectiveness of vaccination in the presence of high tumor burden correlates with the breadth of nAgs-specific T cells and requires concomitant reversal of tumor suppression by checkpoint blockade., Vaccination against neo-antigens has resulted in an effective antitumor response in several models. Here, the authors show that delivery of larger sets of neo-antigens using an adenovirus-based vaccination platform, results in much better tumor protection when combined with checkpoint blockade in a mouse model of advanced disease.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.