1
|
Camerino M, Giacobino D, Tarone L, Dentini A, Martano M, Morello E, Ferraris EI, Manassero L, Iussich S, Maniscalco L, Cavallo F, Riccardo F, Buracco P. Clinical evaluation of HuDo-CSPG4 DNA electroporation as adjuvant treatment for canine oral malignant melanoma: comparison of two vaccination protocols. Vet Q 2025; 45:1-16. [PMID: 40059815 PMCID: PMC11894750 DOI: 10.1080/01652176.2025.2473717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/13/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Canine oral malignant melanoma (OMM) is an aggressive, spontaneously occurring tumor carrying a poor to guarded prognosis and relatively limited therapeutic strategies. In this landscape, chondroitin sulfate proteoglycan (CSPG)4 represents a promising immunotherapeutic target. The objective of this bi-center prospective study was to examine the clinical outcome of OMM-bearing dogs treated with surgery and adjuvant electroporation using a DNA vaccine (HuDo-CSPG4) encoding both human (Hu) and canine (Do) portions of CSPG4 through two different vaccination protocols. Dogs with stage I-III surgically resected CSPG4-positive OMM underwent HuDo-CSPG4 plasmid electroporation starting at the 3rd-4th post-operative week; electrovaccination was repeated after 2 weeks. In protocol 1, electrovaccination was then delivered monthly while in protocol 2, electrovaccination was performed monthly four additional times followed by semestral boosters. The survival rates of HuDo-CSPG4-vaccinated dogs were estimated and compared with a control group treated with surgery alone. Significantly longer overall survival times were observed in HuDo-CSPG4 vaccinated dogs as compared with non-vaccinated controls. Dogs receiving protocol 2 showed similar outcomes to those of dogs undergoing protocol 1, despite fewer vaccinations. The comparable humoral response against CSPG4 resulting from the administration of protocol 1 and 2 appears to have similar clinical relevance, highlighting protocol 2 as the optimal vaccination schedule.
Collapse
Affiliation(s)
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | | | - Marina Martano
- Department of Veterinary Sciences, University of Parma, Parma, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Luca Manassero
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| |
Collapse
|
2
|
Davodabadi F, Sarhadi M, Arabpour J, Sargazi S, Rahdar A, Díez-Pascual AM. Breast cancer vaccines: New insights into immunomodulatory and nano-therapeutic approaches. J Control Release 2022; 349:844-875. [PMID: 35908621 DOI: 10.1016/j.jconrel.2022.07.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Breast cancer (BC) is known to be a highly heterogeneous disease that is clinically subdivided into four primary molecular subtypes, each having distinct morphology and clinical implications. These subtypes are principally defined by hormone receptors and other proteins involved (or not involved) in BC development. BC therapeutic vaccines [including peptide-based vaccines, protein-based vaccines, nucleic acid-based vaccines (DNA/RNA vaccines), bacterial/viral-based vaccines, and different immune cell-based vaccines] have emerged as an appealing class of cancer immunotherapeutics when used alone or combined with other immunotherapies. Employing the immune system to eliminate BC cells is a novel therapeutic modality. The benefit of active immunotherapies is that they develop protection against neoplastic tissue and readjust the immune system to an anti-tumor monitoring state. Such immunovaccines have not yet shown effectiveness for BC treatment in clinical trials. In recent years, nanomedicines have opened new windows to increase the effectiveness of vaccinations to treat BC. In this context, some nanoplatforms have been designed to efficiently deliver molecular, cellular, or subcellular vaccines to BC cells, increasing the efficacy and persistence of anti-tumor immunity while minimizing undesirable side effects. Immunostimulatory nano-adjuvants, liposomal-based vaccines, polymeric vaccines, virus-like particles, lipid/calcium/phosphate nanoparticles, chitosan-derived nanostructures, porous silicon microparticles, and selenium nanoparticles are among the newly designed nanostructures that have been used to facilitate antigen internalization and presentation by antigen-presenting cells, increase antigen stability, enhance vaccine antigenicity and remedial effectivity, promote antigen escape from the endosome, improve cytotoxic T lymphocyte responses, and produce humoral immune responses in BC cells. Here, we summarized the existing subtypes of BC and shed light on immunomodulatory and nano-therapeutic strategies for BC vaccination. Finally, we reviewed ongoing clinical trials on BC vaccination and highlighted near-term opportunities for moving forward.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
3
|
Riccardo F, Tarone L, Camerino M, Giacobino D, Iussich S, Barutello G, Arigoni M, Conti L, Bolli E, Quaglino E, Merighi IF, Morello E, Dentini A, Ferrone S, Buracco P, Cavallo F. Antigen mimicry as an effective strategy to induce CSPG4-targeted immunity in dogs with oral melanoma: a veterinary trial. J Immunother Cancer 2022; 10:e004007. [PMID: 35580930 PMCID: PMC9114861 DOI: 10.1136/jitc-2021-004007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Melanoma is the most lethal form of skin cancer in humans. Conventional therapies have limited efficacy, and overall response is still unsatisfactory considering that immune checkpoint inhibitors induce lasting clinical responses only in a low percentage of patients. This has prompted us to develop a vaccination strategy employing the tumor antigen chondroitin sulfate proteoglycan (CSPG)4 as a target. METHODS To overcome the host's unresponsiveness to the self-antigen CSPG4, we have taken advantage of the conservation of CSPG4 sequence through phylogenetic evolution, so we have used a vaccine, based on a chimeric DNA molecule encompassing both human (Hu) and dog (Do) portions of CSPG4 (HuDo-CSPG4). We have tested its safety and immunogenicity (primary objectives), along with its therapeutic efficacy (secondary outcome), in a prospective, non-randomized, veterinary clinical trial enrolling 80 client-owned dogs with surgically resected, CSPG4-positive, stage II-IV oral melanoma. RESULTS Vaccinated dogs developed anti-Do-CSPG4 and Hu-CSPG4 immune response. Interestingly, the antibody titer in vaccinated dogs was significantly associated with the overall survival. Our data suggest that there may be a contribution of the HuDo-CSPG4 vaccination to the improvement of survival of vaccinated dogs as compared with controls treated with conventional therapies alone. CONCLUSIONS HuDo-CSPG4 adjuvant vaccination was safe and immunogenic in dogs with oral melanoma, with potential beneficial effects on the course of the disease. Thanks to the power of naturally occurring canine tumors as predictive models for cancer immunotherapy response, these data may represent a basis for the translation of this approach to the treatment of human patients with CSPG4-positive melanoma subtypes.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Soldano Ferrone
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
4
|
Ruiu R, Di Lorenzo A, Cavallo F, Conti L. Are Cancer Stem Cells a Suitable Target for Breast Cancer Immunotherapy? Front Oncol 2022; 12:877384. [PMID: 35530300 PMCID: PMC9069673 DOI: 10.3389/fonc.2022.877384] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
There is substantial evidence to suggest that complete tumor eradication relies on the effective elimination of cancer stem cells (CSCs). CSCs have been widely described as mediators of resistance to conventional therapies, including chemo- and radiotherapy, as well as of tumor metastasization and relapse in different tumor types, including breast cancer. However, the resistant phenotype of CSCs makes their targeting a tough task, and immunotherapy may therefore be an interesting option. Nevertheless, although immunotherapeutic approaches to cancer treatment have generated great enthusiasm due to recent success in clinics, breast cancer treatment mostly relies on standard approaches. In this context, we review the existing literature on the immunological properties of breast CSC and immunotherapeutic approaches to them. We will thus attempt to clarify whether there is room for the immunotargeting of breast CSCs in the current landscape of breast cancer therapies. Finally, we will provide our opinion on the CSC-targeting immunotherapeutic strategies that could prospectively be attempted.
Collapse
Affiliation(s)
| | | | - Federica Cavallo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | |
Collapse
|
5
|
Tarone L, Giacobino D, Camerino M, Ferrone S, Buracco P, Cavallo F, Riccardo F. Canine Melanoma Immunology and Immunotherapy: Relevance of Translational Research. Front Vet Sci 2022; 9:803093. [PMID: 35224082 PMCID: PMC8873926 DOI: 10.3389/fvets.2022.803093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
In veterinary oncology, canine melanoma is still a fatal disease for which innovative and long-lasting curative treatments are urgently required. Considering the similarities between canine and human melanoma and the clinical revolution that immunotherapy has instigated in the treatment of human melanoma patients, special attention must be paid to advancements in tumor immunology research in the veterinary field. Herein, we aim to discuss the most relevant knowledge on the immune landscape of canine melanoma and the most promising immunotherapeutic approaches under investigation. Particular attention will be dedicated to anti-cancer vaccination, and, especially, to the encouraging clinical results that we have obtained with DNA vaccines directed against chondroitin sulfate proteoglycan 4 (CSPG4), which is an appealing tumor-associated antigen with a key oncogenic role in both canine and human melanoma. In parallel with advances in therapeutic options, progress in the identification of easily accessible biomarkers to improve the diagnosis and the prognosis of melanoma should be sought, with circulating small extracellular vesicles emerging as strategically relevant players. Translational advances in melanoma management, whether achieved in the human or veterinary fields, may drive improvements with mutual clinical benefits for both human and canine patients; this is where the strength of comparative oncology lies.
Collapse
Affiliation(s)
- Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
6
|
Nguyen-Hoai T, Kobelt D, Hohn O, Vu MD, Schlag PM, Dörken B, Norley S, Lipp M, Walther W, Pezzutto A, Westermann J. HER2/neu DNA vaccination by intradermal gene delivery in a mouse tumor model: Gene gun is superior to jet injector in inducing CTL responses and protective immunity. Oncoimmunology 2021; 1:1537-1545. [PMID: 23264900 PMCID: PMC3525609 DOI: 10.4161/onci.22563] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
DNA vaccines are potential tools for the induction of immune responses against both infectious disease and cancer. The dermal application of DNA vaccines is of particular interest since the epidermal and dermal layers of the skin are characterized by an abundance of antigen-presenting cells (APCs). The aim of our study was to compare tumor protection as obtained by two different methods of intradermal DNA delivery (gene gun and jet injector) in a well-established HER2/neu mouse tumor model. BALB/c mice were immunized twice with a HER2/neu-coding plasmid by gene gun or jet injector. Mice were then subcutaneously challenged with HER2/neu+ syngeneic D2F2/E2 tumor cells. Protection against subsequent challenges with tumor cells as well as humoral and T-cell immune responses induced by the vaccine were monitored. Gene gun immunization was far superior to jet injector both in terms of tumor protection and induction of HER2/neu-specific immune responses. After gene gun immunization, 60% of the mice remained tumor-free until day 140 as compared with 25% after jet injector immunization. Furthermore, gene gun vaccination was able to induce both a strong TH1-polarized T-cell response with detectable cytotoxic T-lymphocyte (CTL) activity and a humoral immune response against HER2/neu, whereas the jet injector was not. Although the disadvantages that were associated with the use of the jet injector in our model may be overcome with methodological modifications and/or in larger animals, which exhibit a thicker skin and/or subcutaneous muscle tissue, we conclude that gene gun delivery constitutes the method of choice for intradermal DNA delivery in preclinical mouse models and possibly also for the clinical development of DNA-based vaccines.
Collapse
Affiliation(s)
- Tam Nguyen-Hoai
- Deptartment of Hematology, Oncology, and Tumor Immunology Charité; University Medicine Berlin; Campus Berlin-Buch, Campus Benjamin Franklin and Campus Virchow-Klinikum; Berlin, Germany ; Max Delbrück Center for Molecular Medicine; Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Quaglino E, Cavallo F, Conti L. Cancer stem cell antigens as targets for new combined anti-cancer therapies. Int J Biochem Cell Biol 2020; 129:105861. [PMID: 33031926 DOI: 10.1016/j.biocel.2020.105861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023]
Abstract
The introduction of immune checkpoint inhibitors (ICI) has ushered in a new, golden age for cancer immunotherapy. However, their clinical success remains limited in several solid cancer types because of the low intrinsic immunogenicity of tumors and the development of immune escape mechanisms. Cancer stem cells (CSC), a small population of cancer cells that are responsible for tumor onset, metastatic spread and relapse after treatment, play a pivotal role in resistance to ICIs. The development of novel therapies that can target CSCs would thus improve the outcomes of current immunotherapy regimens. In this light, vaccines that target CSCs are a promising strategy. This paper briefly describes the immunologic properties of CSCs and their antigenic profile, and reviews current preclinical and clinical approaches that combine CSC-targeting vaccines with different synergistic therapies for the development of more effective antineoplastic treatments.
Collapse
Affiliation(s)
- Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
8
|
Riccardo F, Barutello G, Petito A, Tarone L, Conti L, Arigoni M, Musiu C, Izzo S, Volante M, Longo DL, Merighi IF, Papotti M, Cavallo F, Quaglino E. Immunization against ROS1 by DNA Electroporation Impairs K-Ras-Driven Lung Adenocarcinomas . Vaccines (Basel) 2020; 8:vaccines8020166. [PMID: 32268572 PMCID: PMC7349290 DOI: 10.3390/vaccines8020166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is still the leading cause of cancer death worldwide. Despite the introduction of tyrosine kinase inhibitors and immunotherapeutic approaches, there is still an urgent need for novel strategies to improve patient survival. ROS1, a tyrosine kinase receptor endowed with oncoantigen features, is activated by chromosomal rearrangement or overexpression in NSCLC and in several tumor histotypes. In this work, we have exploited transgenic mice harboring the activated K-Ras oncogene (K-RasG12D) that spontaneously develop metastatic NSCLC as a preclinical model to test the efficacy of ROS1 immune targeting. Indeed, qPCR and immunohistochemical analyses revealed ROS1 overexpression in the autochthonous primary tumors and extrathoracic metastases developed by K-RasG12D mice and in a derived transplantable cell line. As proof of concept, we have evaluated the effects of the intramuscular electroporation (electrovaccination) of plasmids coding for mouse- and human-ROS1 on the progression of these NSCLC models. A significant increase in survival was observed in ROS1-electrovaccinated mice challenged with the transplantable cell line. It is worth noting that tumors were completely rejected, and immune memory was achieved, albeit only in a few mice. Most importantly, ROS1 electrovaccination was also found to be effective in slowing the development of autochthonous NSCLC in K-RasG12D mice.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Angela Petito
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Chiara Musiu
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Stefania Izzo
- Department of Oncology, University of Torino, 10043 Orbassano, Italy; (S.I.); (M.V.); (M.P.)
| | - Marco Volante
- Department of Oncology, University of Torino, 10043 Orbassano, Italy; (S.I.); (M.V.); (M.P.)
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), 10126 Torino, Italy;
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Mauro Papotti
- Department of Oncology, University of Torino, 10043 Orbassano, Italy; (S.I.); (M.V.); (M.P.)
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
- Correspondence: (F.C.); (E.Q.); Tel.: +39-011670-6457 (F.C. & E.Q.)
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
- Correspondence: (F.C.); (E.Q.); Tel.: +39-011670-6457 (F.C. & E.Q.)
| |
Collapse
|
9
|
Quaglino E, Conti L, Cavallo F. Breast cancer stem cell antigens as targets for immunotherapy. Semin Immunol 2020; 47:101386. [PMID: 31932198 DOI: 10.1016/j.smim.2020.101386] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/01/2020] [Indexed: 12/13/2022]
Abstract
The great success of immunotherapy is paving the way for a new era in cancer treatment and is driving major improvements in the therapy of patients suffering from a range of solid tumors. However, the choice of the appropriate tumor antigens to be targeted with cancer vaccines and T-cell therapies is still a challenge. Most antigens targeted so far have been identified on the tumor bulk and are expressed on differentiated cancer cells. The discovery of a small population of cancer stem cells (CSC), which is refractory to most current therapies and responsible for the development of metastasis and recurrence, has made it clear that the ideal targets for immunotherapies are the antigens that are expressed in CSC and play a key role in their function. Indeed, their immunotargeting would enable the eradication of CSC to be performed, thus eliminating the tumor source. We call these antigens "CSC oncoantigens". Herein, we summarize the controversial nature of breast CSC, discuss why they represent good candidates for cancer immunotherapy, and review the CSC antigens that have been used as targets for CSC immunotargeting this far. Moreover, we describe the pipeline that we have developed for the identification of fresh CSC oncoantigens, and present the pre-clinical results obtained with vaccines that target some of these antigens.
Collapse
Affiliation(s)
- Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
10
|
Riccardo F, Tarone L, Iussich S, Giacobino D, Arigoni M, Sammartano F, Morello E, Martano M, Gattino F, Maria RD, Ferrone S, Buracco P, Cavallo F. Identification of CSPG4 as a promising target for translational combinatorial approaches in osteosarcoma. Ther Adv Med Oncol 2019; 11:1758835919855491. [PMID: 31217827 PMCID: PMC6557023 DOI: 10.1177/1758835919855491] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 05/09/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Osteosarcoma (OSA) is a highly metastatic pediatric bone tumor. Adjuvant chemotherapy and surgical resection represent standard treatments; however, the prognosis is still poor. Effective strategies are urgently needed. Chondroitin sulfate proteoglycan (CSPG)4 is a transmembrane proteoglycan with a low expression in normal tissues but high expression in several solid tumors, where it plays a central tumorigenic role. Therefore, it represents a promising therapeutic target. The high homology between human and canine CSPG4 and the recognized translational power of canine tumors as preclinical models for human malignancies prompted us to evaluate CSPG4 expression and the consequences of its immune-targeting for both human and canine OSA treatment. Methods: We analyzed CSPG4 overexpression in human and canine OSA samples and its significance for the survival of OSA patients. We exploited functional in vitro experiments to assess the antitumor potential of CSPG4 immune-targeting. Results: CSPG4 is overexpressed in OSA and has possible clinical implications as suggested by an evident correlation between CSPG4 overexpression and a shorter survival for both OSA-affected humans and dogs. The potential of CSPG4 immune-targeting for OSA treatment came from the ability of anti-CSPG4 monoclonal antibodies and sera, derived from human-CSPG4-DNA vaccinated canine patients, to significantly inhibit human and canine CSPG4-positive OSA cell proliferation, migration, and osteospheres generation. Moreover, CSPG4 immune-targeting has been shown to potentiate the effect of doxorubicin. Conclusions: Overall, these results provide the rationale to investigate the CSPG4 immune-targeting as a promising weapon for the treatment of CSPG4-positive OSA canine patients, to be successfully translated to a human setting.
Collapse
Affiliation(s)
- Federica Riccardo
- University of Torino, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, Via Nizza, 52, Torino, TO, 10126, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, Bioinformatics and Genomic Unit, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | | | - Emanuela Morello
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Marina Martano
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Francesca Gattino
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
11
|
Ruiu R, Tarone L, Rolih V, Barutello G, Bolli E, Riccardo F, Cavallo F, Conti L. Cancer stem cell immunology and immunotherapy: Harnessing the immune system against cancer's source. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:119-188. [PMID: 31383404 DOI: 10.1016/bs.pmbts.2019.03.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite recent advances in diagnosis and therapy having improved cancer outcome, many patients still do not respond to treatments, resulting in the progression or relapse of the disease, eventually impairing survival expectations. The limited efficacy of therapy is often attributable to its inability to affect cancer stem cells (CSCs), a small population of cells resistant to current radio- and chemo-therapies. CSCs are characterized by self-renewal and tumor-initiating capabilities, and function as a reservoir for the local and distant recurrence of the disease. Therefore, new therapeutic approaches able to effectively target and deplete CSCs are urgently needed. Immunotherapy is facing a renewed interest for its potential in cancer treatment, and the possibility of harnessing the immune system to target CSCs is being addressed by a new exciting research field. In this chapter, we discuss the cancer stem cell model and illustrate CSC biological and molecular properties, critically addressing theoretical and practical issues linked with their definition and study. We then review the existing literature regarding the immunological properties of CSCs and the complex interplay occurring between CSCs and immune cells. Finally, we present up-to-date studies on CSC immunotargeting and its potential future perspective. In conclusion, understanding the interplay between CSC biology and tumor immunology will provide a deeper understanding of the mechanisms that regulate CSC immunological properties. This will contribute to the design of new CSC-directed immunotherapeutic strategies with the potential of strongly improving cancer outcomes.
Collapse
Affiliation(s)
- Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
12
|
Ruiu R, Rolih V, Bolli E, Barutello G, Riccardo F, Quaglino E, Merighi IF, Pericle F, Donofrio G, Cavallo F, Conti L. Fighting breast cancer stem cells through the immune-targeting of the xCT cystine-glutamate antiporter. Cancer Immunol Immunother 2019; 68:131-141. [PMID: 29947961 PMCID: PMC11028170 DOI: 10.1007/s00262-018-2185-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/08/2018] [Indexed: 01/17/2023]
Abstract
Tumor relapse and metastatic spreading act as major hindrances to achieve complete cure of breast cancer. Evidence suggests that cancer stem cells (CSC) would function as a reservoir for the local and distant recurrence of the disease, due to their resistance to radio- and chemotherapy and their ability to regenerate the tumor. Therefore, the identification of appropriate molecular targets expressed by CSC may be critical in the development of more effective therapies. Our studies focused on the identification of mammary CSC antigens and on the development of CSC-targeting vaccines. We compared the transcriptional profile of CSC-enriched tumorspheres from an Her2+ breast cancer cell line with that of the more differentiated parental cells. Among the molecules strongly upregulated in tumorspheres we selected the transmembrane amino-acid antiporter xCT. In this review, we summarize the results we obtained with different xCT-targeting vaccines. We show that, despite xCT being a self-antigen, vaccination was able to induce a humoral immune response that delayed primary tumor growth and strongly impaired pulmonary metastasis formation in mice challenged with tumorsphere-derived cells. Moreover, immunotargeting of xCT was able to increase CSC chemosensitivity to doxorubicin, suggesting that it may act as an adjuvant to chemotherapy. In conclusion, our approach based on the comparison of the transcriptome of tumorspheres and parental cells allowed us to identify a novel CSC-related target and to develop preclinical therapeutic approaches able to impact on CSC biology, and therefore, hampering tumor growth and dissemination.
Collapse
Affiliation(s)
- Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | | | - Gaetano Donofrio
- Department of Medical Veterinary Science, University of Parma, Parma, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| |
Collapse
|
13
|
Palladini A, Thrane S, Janitzek CM, Pihl J, Clemmensen SB, de Jongh WA, Clausen TM, Nicoletti G, Landuzzi L, Penichet ML, Balboni T, Ianzano ML, Giusti V, Theander TG, Nielsen MA, Salanti A, Lollini PL, Nanni P, Sander AF. Virus-like particle display of HER2 induces potent anti-cancer responses. Oncoimmunology 2018; 7:e1408749. [PMID: 29399414 PMCID: PMC5790387 DOI: 10.1080/2162402x.2017.1408749] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
Overexpression of human epidermal growth factor receptor-2 (HER2) occurs in 20–30% of invasive breast cancers. Monoclonal antibody therapy is effective in treating HER2-driven mammary carcinomas, but its utility is limited by high costs, side effects and development of resistance. Active vaccination may represent a safer, more effective and cheaper alternative, although the induction of strong and durable autoantibody responses is hampered by immune-tolerogenic mechanisms. Using a novel virus-like particle (VLP) based vaccine platform we show that directional, high-density display of human HER2 on the surface of VLPs, allows induction of therapeutically potent anti-HER2 autoantibody responses. Prophylactic vaccination reduced spontaneous development of mammary carcinomas by 50%-100% in human HER2 transgenic mice and inhibited the growth of HER2-positive tumors implanted in wild-type mice. The HER2-VLP vaccine shows promise as a new cost-effective modality for prevention and treatment of HER2-positive cancer. The VLP platform may represent an effective tool for development of vaccines against other non-communicable diseases.
Collapse
Affiliation(s)
- Arianna Palladini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Susan Thrane
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christoph M Janitzek
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jessica Pihl
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stine B Clemmensen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark.,ExpreS2ion Biotechnologies, SCION-DTU Science Park, Hørsholm, Denmark
| | | | - Thomas M Clausen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Giordano Nicoletti
- Rizzoli Orthopedic Institute, Laboratory of Experimental Oncology, Bologna, Italy
| | - Lorena Landuzzi
- Rizzoli Orthopedic Institute, Laboratory of Experimental Oncology, Bologna, Italy
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery and Department of Microbiology, Immunology and Molecular Genetics, The Molecular Biology Institute, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Tania Balboni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Marianna L Ianzano
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Veronica Giusti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Thor G Theander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten A Nielsen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Patrizia Nanni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Adam F Sander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
14
|
Development of a radiolabeled caninized anti-EGFR antibody for comparative oncology trials. Oncotarget 2017; 8:83128-83141. [PMID: 29137329 PMCID: PMC5669955 DOI: 10.18632/oncotarget.20914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Due to large homology of human and canine EGFR, dogs suffering from spontaneous EGFR+ cancer can be considered as ideal translational models. Thereby, novel immunotherapeutic compounds can be developed for both human and veterinary patients. This study describes the radiolabeling of a canine anti-EGFR IgG antibody (can225IgG) with potential diagnostic and therapeutic value in comparative clinical settings. Can225IgG was functionalized with DTPA for subsequent chelation with the radionuclide 99mTc. Successful coupling of 10 DTPA molecules per antibody on average was proven by significant mass increase in MALDI-TOF spectroscopy, gel electrophoresis and immunoblots. Following functionalization and radiolabeling, 99mTc-DTPA-can225IgG fully retained its binding capacity towards human and canine EGFR in flow cytometry, immuno- and radioblots, and autoradiography. The affinity of radiolabeled can225IgG was determined to KD 0.8 ±0.0031 nM in a real-time kinetics assay on canine carcinoma cells by a competition binding technique. Stability tests of the radiolabeled compound identified TRIS buffered saline as the ideal formulation for short-term storage with 87.11 ±6.04% intact compound being still detected 60 minutes post radiolabeling. High stability, specificity and EGFR binding affinity pinpoint towards 99mTc-radiolabeled can225IgG antibody as an ideal lead compound for the first proof-of-concept diagnostic and therapeutic applications in canine cancer patients.
Collapse
|
15
|
Riccardo F, Réal A, Voena C, Chiarle R, Cavallo F, Barutello G. Maternal Immunization: New Perspectives on Its Application Against Non-Infectious Related Diseases in Newborns. Vaccines (Basel) 2017; 5:E20. [PMID: 28763018 PMCID: PMC5620551 DOI: 10.3390/vaccines5030020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
The continuous evolution in preventive medicine has anointed vaccination a versatile, human-health improving tool, which has led to a steady decline in deaths in the developing world. Maternal immunization represents an incisive step forward for the field of vaccination as it provides protection against various life-threatening diseases in pregnant women and their children. A number of studies to improve prevention rates and expand protection against the largest possible number of infections are still in progress. The complex unicity of the mother-infant interaction, both during and after pregnancy and which involves immune system cells and molecules, is an able partner in the success of maternal immunization, as intended thus far. Interestingly, new studies have shed light on the versatility of maternal immunization in protecting infants from non-infectious related diseases, such as allergy, asthma and congenital metabolic disorders. However, barely any attempt at applying maternal immunization to the prevention of childhood cancer has been made. The most promising study reported in this new field is a recent proof of concept on the efficacy of maternal immunization in protecting cancer-prone offspring against mammary tumor progression. New investigations into the possibility of exploiting maternal immunization to prevent the onset and/or progression of neuroblastoma, one of the most common childhood malignancies, are therefore justified. Maternal immunization is presented in a new guise in this review. Attention will be focused on its versatility and potential applications in preventing tumor progression in neuroblastoma-prone offspring.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Aline Réal
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| |
Collapse
|
16
|
Rolih V, Barutello G, Iussich S, De Maria R, Quaglino E, Buracco P, Cavallo F, Riccardo F. CSPG4: a prototype oncoantigen for translational immunotherapy studies. J Transl Med 2017; 15:151. [PMID: 28668095 PMCID: PMC5494135 DOI: 10.1186/s12967-017-1250-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/21/2017] [Indexed: 12/21/2022] Open
Abstract
Thanks to striking progress in both the understanding of anti-tumor immune response and the characterization of several tumor associated antigens (TAA), a more rational design and more sophisticated strategies for anti-tumor vaccination have been possible. However, the effectiveness of cancer vaccines in clinical trial is still partial, indicating that additional studies are needed to optimize their design and their pre-clinical testing. Indeed, anti-tumor vaccination success relies on the choice of the best TAA to be targeted and on the translational power of the pre-clinical model used to assess its efficacy. The chondroitin sulfate proteoglycan-4 (CSPG4) is a cell surface proteoglycan overexpressed in a huge range of human and canine neoplastic lesions by tumor cells, tumor microenvironment and cancer initiating cells. CSPG4 plays a central role in the oncogenic pathways required for malignant progression and metastatization. Thanks to these features and to its poor expression in adult healthy tissues, CSPG4 represents an ideal oncoantigen and thus an attractive target for anti-tumor immunotherapy. In this review we explore the potential of CSPG4 immune-targeting. Moreover, since it has been clearly demonstrated that spontaneous canine tumors mimic the progression of human malignancies better than any other pre-clinical model available so far, we reported also our results indicating that CSPG4 DNA vaccination is safe and effective in significantly increasing the survival of canine melanoma patients. Therefore, anti-CSPG4 vaccination strategy could have a substantial impact for the treatment of the wider population of spontaneous CSPG4-positive tumor affected dogs with a priceless translational value and a revolutionary implication for human oncological patients.
Collapse
Affiliation(s)
- Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| |
Collapse
|
17
|
Bandini S, Macagno M, Hysi A, Lanzardo S, Conti L, Bello A, Riccardo F, Ruiu R, Merighi IF, Forni G, Iezzi M, Quaglino E, Cavallo F. The non-inflammatory role of C1q during Her2/neu-driven mammary carcinogenesis. Oncoimmunology 2016; 5:e1253653. [PMID: 28123895 DOI: 10.1080/2162402x.2016.1253653] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/12/2016] [Accepted: 10/23/2016] [Indexed: 12/13/2022] Open
Abstract
There is an ever increasing amount of evidence to support the hypothesis that complement C1q, the first component of the classical complement pathway, is involved in the regulation of cancer growth, in addition to its role in fighting infections. It has been demonstrated that C1q is expressed in the microenvironment of various types of human tumors, including breast adenocarcinomas. This study compares carcinogenesis progression in C1q deficient (neuT-C1KO) and C1q competent neuT mice in order to investigate the role of C1q in mammary carcinogenesis. Significantly accelerated autochthonous neu+ carcinoma progression was paralleled by accelerated spontaneous lung metastases occurrence in C1q deficient mice. Surprisingly, this effect was not caused by differences in the tumor-infiltrating cells or in the activation of the complement classical pathway, since neuT-C1KO mice did not display a reduction in C3 fragment deposition at the tumor site. By contrast, a significant higher number of intratumor blood vessels and a decrease in the activation of the tumor suppressor WW domain containing oxidoreductase (WWOX) were observed in tumors from neuT-C1KO as compare with neuT mice. In parallel, an increase in Her2/neu expression was observed on the membrane of tumor cells. Taken together, our findings suggest that C1q plays a direct role both on halting tumor angiogenesis and on inducing apoptosis in mammary cancer cells by coordinating the signal transduction pathways linked to WWOX and, furthermore, highlight the role of C1q in mammary tumor immune surveillance regardless of complement system activation.
Collapse
Affiliation(s)
- Silvio Bandini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Marco Macagno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Albana Hysi
- Department of Medicine Science, Center of Excellence on Aging and Translational Medicine (CeSI-Met), G. d'Annunzio University of Chieti Pescara , Italy
| | - Stefania Lanzardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Amanda Bello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Guido Forni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Manuela Iezzi
- Department of Medicine Science, Center of Excellence on Aging and Translational Medicine (CeSI-Met), G. d'Annunzio University of Chieti Pescara , Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino , Torino, Italy
| |
Collapse
|
18
|
CCL4 as an adjuvant for DNA vaccination in a Her2/neu mouse tumor model. Cancer Gene Ther 2016; 23:162-7. [PMID: 27056671 DOI: 10.1038/cgt.2016.9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/28/2022]
Abstract
Chemokines are key regulators of both innate and adaptive immune responses. CCL4 (macrophage inflammatory protein-1β, MIP-1β) is a CC chemokine that has a broad spectrum of target cells including immature dendritic cells, which express the cognate receptor CCR5. We asked whether a plasmid encoding CCL4 is able to improve tumor protection and immune responses in a Her2/neu+ mouse tumor model. Balb/c mice were immunized twice intramuscularly with plasmid DNA on days 1 and 15. On day 25, a tumor challenge was performed with 2 × 10(5) syngeneic Her2/neu+ D2F2/E2 tumor cells. Different groups of mice were vaccinated with pDNA(Her2/neu) plus pDNA(CCL4), pDNA(Her2/neu), pDNA(CCL4) or mock vector alone. Our results show that CCL4 is able to (i) improve tumor protection and (ii) augment a TH1-polarized immune response against Her2/neu. Although Her2/neu-specific humoral and T-cell immune responses were comparable with that induced in previous studies using CCL19 or CCL21 as adjuvants, tumor protection conferred by CCL4 was inferior. Whether this is due to a different spectrum of (innate) immune cells, remains to be clarified. However, combination of CCL19/21 with CCL4 might be a reasonable approach in the future, particularly for DNA vaccination in Her2/neu+ breast cancer in the situation of minimal residual disease.
Collapse
|
19
|
Matić S, Quaglino E, Arata L, Riccardo F, Pegoraro M, Vallino M, Cavallo F, Noris E. The rat ErbB2 tyrosine kinase receptor produced in plants is immunogenic in mice and confers protective immunity against ErbB2(+) mammary cancer. PLANT BIOTECHNOLOGY JOURNAL 2016; 14:153-9. [PMID: 25865255 PMCID: PMC11388968 DOI: 10.1111/pbi.12367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 06/04/2023]
Abstract
The rat ErbB2 (rErbB2) protein is a 185-kDa glycoprotein belonging to the epidermal growth factor-related proteins (ErbB) of receptor tyrosine kinases. Overexpression and mutations of ErbB proteins lead to several malignancies including breast, lung, pancreatic, bladder and ovary carcinomas. ErbB2 is immunogenic and is an ideal candidate for cancer immunotherapy. We investigated the possibility of expressing the extracellular (EC) domain of rErbB2 (653 amino acids, aa) in Nicotiana benthamiana plants, testing the influence of the 23 aa transmembrane (TM) sequence on protein accumulation. Synthetic variants of the rErbB2 gene portion encoding the EC domain, optimized with a human codon usage and either linked to the full TM domain (rErbB2_TM, 676 aa), to a portion of it (rErbB2-pTM, 662 aa), or deprived of it (rErbB2_noTM, 653 aa) were cloned in the pEAQ-HT expression vector as 6X His tag fusions. All rErbB2 variants (72-74.5 kDa) were transiently expressed, but the TM was detrimental for rErbB2 EC accumulation. rERbB2_noTM was the most expressed protein; it was solubilized and purified with Nickel affinity resin. When crude soluble extracts expressing rErbB2_noTM were administered to BALB/c mice, specific rErbB2 immune responses were triggered. A potent antitumour activity was induced when vaccinated mice were challenged with syngeneic transplantable ErbB2(+) mammary carcinoma cells. To our knowledge, this is the first report of expression of rErbB2 in plants and of its efficacy in inducing a protective antitumour immune response, opening interesting perspectives for further immunological testing.
Collapse
Affiliation(s)
- Slavica Matić
- Institute for Sustainable Plant Protection, National Research Council, Torino, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lucia Arata
- Institute for Sustainable Plant Protection, National Research Council, Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Mattia Pegoraro
- Institute for Sustainable Plant Protection, National Research Council, Torino, Italy
| | - Marta Vallino
- Institute for Sustainable Plant Protection, National Research Council, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emanuela Noris
- Institute for Sustainable Plant Protection, National Research Council, Torino, Italy
| |
Collapse
|
20
|
Marconato L, Buracco P, Aresu L. Perspectives on the design of clinical trials for targeted therapies and immunotherapy in veterinary oncology. Vet J 2015; 205:238-43. [DOI: 10.1016/j.tvjl.2015.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 02/15/2015] [Accepted: 02/25/2015] [Indexed: 12/18/2022]
|
21
|
Zavala VA, Kalergis AM. New clinical advances in immunotherapy for the treatment of solid tumours. Immunology 2015; 145:182-201. [PMID: 25826229 PMCID: PMC4427384 DOI: 10.1111/imm.12459] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/08/2015] [Accepted: 02/24/2015] [Indexed: 12/16/2022] Open
Abstract
Advances in understanding the mechanisms of cancer cells for evading the immune system surveillance, including how the immune system modulates the phenotype of tumours, have allowed the development of new therapies that benefit from this complex cellular network to specifically target and destroy cancer cells. Immunotherapy researchers have mainly focused on the discovery of tumour antigens that could confer specificity to immune cells to detect and destroy cancer cells, as well as on the mechanisms leading to an improved activation of effector immune cells. The Food and Drug Administration approval in 2010 of ipilumumab for melanoma treatment and of pembrolizumab in 2014, monoclonal antibodies against T-lymphocyte-associated antigen 4 and programmed cell death 1, respectively, are encouraging examples of how research in this area can successfully translate into clinical use with promising results. Currently, several ongoing clinical trials are in progress testing new anti-cancer therapies based on the enhancement of immune cell activity against tumour antigens. Here we discuss the general concepts related to immunotherapy and the recent application to the treatment of cancer with positive results that support their consideration of clinical application to patients.
Collapse
Affiliation(s)
- Valentina A Zavala
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de ChileSantiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
- Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de ChileSantiago, Chile
- INSERM U1064Nantes, France
| |
Collapse
|
22
|
Barutello G, Curcio C, Spadaro M, Arigoni M, Trovato R, Bolli E, Zheng Y, Ria F, Quaglino E, Iezzi M, Riccardo F, Holmgren L, Forni G, Cavallo F. Antitumor immunization of mothers delays tumor development in cancer-prone offspring. Oncoimmunology 2015; 4:e1005500. [PMID: 26155401 PMCID: PMC4485839 DOI: 10.1080/2162402x.2015.1005500] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 10/25/2022] Open
Abstract
Maternal immunization is successfully applied against some life-threatening infectious diseases as it can protect the mother and her offspring through the passive transfer of maternal antibodies. Here, we sought to evaluate whether the concept of maternal immunization could also be applied to cancer immune-prevention. We have previously shown that antibodies induced by DNA vaccination against rat Her2 (neu) protect heterozygous neu-transgenic female (BALB-neuT) mice from autochthonous mammary tumor development. We, herein, seek to evaluate whether a similar maternal immunization can confer antitumor protection to BALB-neuT offspring. Significantly extended tumor-free survival was observed in BALB-neuT offspring born and fed by mothers vaccinated against neu, as compared to controls. Maternally derived anti-neu immunoglobulin G (IgG) was successfully transferred from mothers to newborns and was responsible for the protective effect. Vaccinated mothers and offspring also developed active immunity against neu as revealed by the presence of T-cell-mediated cytotoxicity against the neu immunodominant peptide. This active response was due to the milk transfer of immune complexes that were formed between the neu extracellular domain, shed from vaccine-transfected muscle cells, and the anti-neu IgG induced by the vaccine. These findings show that maternal immunization has the potential to hamper mammary cancer in genetically predestinated offspring and to develop into applications against lethal neonatal cancer diseases for which therapeutic options are currently unavailable.
Collapse
Affiliation(s)
- Giuseppina Barutello
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Claudia Curcio
- Aging Research Centre; "Gabriele d'Annunzio" University ; Chieti, Italy
| | - Michela Spadaro
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Maddalena Arigoni
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Rosalinda Trovato
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Elisabetta Bolli
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Yujuan Zheng
- Department of Oncology and Pathology; Cancer Centre Karolinska; Karolinska Institutet ; Stockholm, Sweden
| | - Francesco Ria
- Institute of General Pathology; Catholic University Sacro Cuore ; Roma, Italy
| | - Elena Quaglino
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Manuela Iezzi
- Aging Research Centre; "Gabriele d'Annunzio" University ; Chieti, Italy
| | - Federica Riccardo
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Lars Holmgren
- Department of Oncology and Pathology; Cancer Centre Karolinska; Karolinska Institutet ; Stockholm, Sweden
| | - Guido Forni
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino, Italy
| |
Collapse
|
23
|
Cavallo F, Aurisicchio L, Mancini R, Ciliberto G. Xenogene vaccination in the therapy of cancer. Expert Opin Biol Ther 2014; 14:1427-42. [DOI: 10.1517/14712598.2014.927433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Microenvironment, oncoantigens, and antitumor vaccination: lessons learned from BALB-neuT mice. BIOMED RESEARCH INTERNATIONAL 2014; 2014:534969. [PMID: 25136593 PMCID: PMC4065702 DOI: 10.1155/2014/534969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/12/2014] [Indexed: 12/20/2022]
Abstract
The tyrosine kinase human epidermal growth factor receptor 2 (HER2) gene is amplified in approximately 20% of human breast cancers and is associated with an aggressive clinical course and the early development of metastasis. Its crucial role in tumor growth and progression makes HER2 a prototypic oncoantigen, the targeting of which may be critical for the development of effective anticancer therapies. The setup of anti-HER2 targeting strategies has revolutionized the clinical outcome of HER2+ breast cancer. However, their initial success has been overshadowed by the onset of pharmacological resistance that renders them ineffective. Since the tumor microenvironment (TME) plays a crucial role in drug resistance, the design of more effective anticancer therapies should depend on the targeting of both cancer cells and their TME as a whole. In this review, starting from the successful know-how obtained with a HER2+ mouse model of mammary carcinogenesis, the BALB-neuT mice, we discuss the role of TME in mammary tumor development. Indeed, a deeper knowledge of antigens critical for cancer outbreak and progression and of the mechanisms that regulate the interplay between cancer and stromal cell populations could advise promising ways for the development of the best anticancer strategy.
Collapse
|
25
|
Riccardo F, Iussich S, Maniscalco L, Lorda Mayayo S, La Rosa G, Arigoni M, De Maria R, Gattino F, Lanzardo S, Lardone E, Martano M, Morello E, Prestigio S, Fiore A, Quaglino E, Zabarino S, Ferrone S, Buracco P, Cavallo F. CSPG4-specific immunity and survival prolongation in dogs with oral malignant melanoma immunized with human CSPG4 DNA. Clin Cancer Res 2014; 20:3753-62. [PMID: 24874834 DOI: 10.1158/1078-0432.ccr-13-3042] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Due to the many similarities with its human counterpart, canine malignant melanoma (cMM) is a valuable model in which to assess the efficacy of novel therapeutic strategies. The model is herein used to evaluate the immunogenicity, safety, and therapeutic efficacy of a human chondroitin sulfate proteoglycan-4 (hCSPG4) DNA-based vaccine. The fact that homology between hCSPG4 and cCSPG4 amino-acidic sequences stands at more than 80% provides the rationale for using an hCSPG4 DNA vaccine in the cMM model. EXPERIMENTAL DESIGN Dogs with stage II-III surgically resected CSPG4-positive oral MM were subjected to monthly intramuscular plasmid administration, which was followed immediately by electroporation (electrovaccination) for at least 6, and up to 20, months. The immunogenicity, safety, and therapeutic efficacy of the vaccine have been evaluated. RESULTS hCSPG4 electrovaccination caused no clinically relevant local or systemic side effects and resulted in significantly longer overall and disease-free survival times in 14 vaccinated dogs as compared with 13 nonvaccinated controls. All vaccinated dogs developed antibodies against both hCSPG4 and cCSPG4. Seven vaccinated dogs were also tested for a cCSPG4-specific T-cell response and only two gave a detectable interferon (IFN)γ response. CONCLUSION Xenogeneic electrovaccination against CSPG4 is able to overcome host unresponsiveness to the "self" antigen and seems to be effective in treating cMM, laying the foundation for its translation to a human clinical setting.
Collapse
Affiliation(s)
- Federica Riccardo
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Selina Iussich
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Lorella Maniscalco
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Saray Lorda Mayayo
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | | | - Maddalena Arigoni
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Raffaella De Maria
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Francesca Gattino
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Stefania Lanzardo
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Elena Lardone
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Marina Martano
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Emanuela Morello
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Simone Prestigio
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Alessandra Fiore
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Elena Quaglino
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Sara Zabarino
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paolo Buracco
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy;
| | - Federica Cavallo
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino;
| |
Collapse
|
26
|
Lollini PL, Cavallo F, De Giovanni C, Nanni P. Preclinical vaccines against mammary carcinoma. Expert Rev Vaccines 2014; 12:1449-63. [DOI: 10.1586/14760584.2013.845530] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Riccardo F, Bolli E, Macagno M, Arigoni M, Cavallo F, Quaglino E. Chimeric DNA Vaccines: An Effective Way to Overcome Immune Tolerance. Curr Top Microbiol Immunol 2014; 405:99-122. [PMID: 25294003 DOI: 10.1007/82_2014_426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The fact that cancer immunotherapy is considered to be a safe and successful weapon for use in combination with surgery, radiation, and chemotherapy treatments means that it has recently been chosen as Breakthrough of the Year 2013 by Science editors. Anticancer vaccines have been extensively tested, in this field, both in preclinical cancer models and in the clinic. However, tumor-associated antigens (TAAs) are often self-tolerated molecules and cancer patients suffer from strong immunosuppressive effects, meaning that the triggering of an effective anti-tumor immune response is difficult. One possible means to overcome immunological tolerance to self-TAAs is of course the use of vaccines that code for xenogeneic proteins. However, a low-affinity antibody response against the self-homologous protein expressed by cancer cells is generally induced by xenovaccination. This issue becomes extremely limiting when working with tumors in which the contribution of the humoral rather than the cellular immune response is required if tumor growth is to be hampered. A possible way to avoid this problem is to use hybrid vaccines which code for chimeric proteins that include both homologous and xenogeneic moieties. In fact, a superior protective anti-tumor immune response against ErbB2+ transplantable and autochthonous mammary tumors was observed over plasmids that coded for the fully rat or fully human proteins when hybrid plasmids that coded for chimeric rat/human ErbB2 protein were tested in ErbB2 transgenic mice. In principle, these findings may become the basis for a new rational means of designing effective vaccines against TAAs.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Marco Macagno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
28
|
Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Lenalidomide-based immunochemotherapy. Oncoimmunology 2013; 2:e26494. [PMID: 24482747 PMCID: PMC3897503 DOI: 10.4161/onci.26494] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 09/14/2013] [Indexed: 12/19/2022] Open
Abstract
Lenalidomide is a synthetic derivative of thalidomide currently approved by the US Food and Drug Administration for use in patients affected by multiple myeloma (in combination with dexamethasone) and low or intermediate-1 risk myelodysplastic syndromes that harbor 5q cytogenetic abnormalities. For illustrative purposes, the mechanism of action of lenalidomide can be subdivided into a cancer cell-intrinsic, a stromal, and an immunological component. Indeed, lenalidomide not only exerts direct cell cycle-arresting and pro-apoptotic effects on malignant cells, but also interferes with their physical and functional interaction with the tumor microenvironment and mediates a robust, pleiotropic immunostimulatory activity. In particular, lenalidomide has been shown to stimulate the cytotoxic functions of T lymphocytes and natural killer cells, to limit the immunosuppressive impact of regulatory T cells, and to modulate the secretion of a wide range of cytokines, including tumor necrosis factor α, interferon γ as well as interleukin (IL)-6, IL-10, and IL-12. Throughout the last decade, the antineoplastic and immunostimulatory potential of lenalidomide has been investigated in patients affected by a wide variety of hematological and solid malignancies. Here, we discuss the results of these studies and review the status of clinical trials currently assessing the safety and efficacy of this potent immunomodulatory drug in oncological indications.
Collapse
Affiliation(s)
- Michaela Semeraro
- Gustave Roussy; Villejuif, France ; INSERM, U1015, CICBT507; Villejuif, France
| | - Erika Vacchelli
- Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | - Jerome Galon
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France ; INSERM, U872; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015, CICBT507; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France
| |
Collapse
|
29
|
Lan KH, Liu YC, Shih YS, Tsaid CL, Yen SH, Lan KL. A DNA vaccine against cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) prevents tumor growth. Biochem Biophys Res Commun 2013; 440:222-8. [DOI: 10.1016/j.bbrc.2013.09.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 09/05/2013] [Indexed: 12/20/2022]
|
30
|
Wang YM, Zhou JJ, Wang Y, Watson D, Zhang GY, Hu M, Wu H, Zheng G, Wang Y, Durkan AM, Harris DCH, Alexander SI. Daedalic DNA vaccination against self antigens as a treatment for chronic kidney disease. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:326-333. [PMID: 23412421 PMCID: PMC3563188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/17/2013] [Indexed: 06/01/2023]
Abstract
Chronic kidney disease (CKD) is a major cause of death and morbidity in Australia and worldwide. DNA vaccination has been used for targeting foreign antigens to induce immune responses and prevent autoimmune disease, viral infection and cancer. However, the use of DNA vaccination has been restricted by a limited ability to induce strong immune responses, especially against self-antigens which are limited by mechanisms of self-tolerance. Furthermore, there have been few studies on the potential of DNA vaccination in chronic inflammatory diseases, including CKD. We have established strategies of DNA vaccination targeting specific self-antigens in the immune system including co-stimulatory pathways, T cell receptors and chemokine molecules, which have been effective in protecting against the development of CKD in a variety of animal models. In particular, we find that the efficacy of DNA vaccination is improved by dendritic cell (DC) targeting and can protect against animal models of autoimmune nephritis mimicking human membranous nephropathy. In this review, we summarize several approaches that have been tested to improve the efficacy of DNA vaccination in CKD models, including enhanced DNA vaccine delivery methods, DNA vaccine modifications and new molecular targets for DNA vaccination. Finally, we discuss the specific application of DNA vaccination for preventing and treating CKD.
Collapse
Affiliation(s)
- Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead Sydney, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|