1
|
Alvarez-Arzola R, Bancaro N, Lai P, Attanasio G, Pellegrini L, Troiani M, Colucci M, Mosole S, Pasquini E, Alimonti A, Mesa C. VSSP-activated macrophages mediate senescence and tumor inhibition in a preclinical model of advanced prostate cancer. Cell Commun Signal 2023; 21:76. [PMID: 37055829 PMCID: PMC10100133 DOI: 10.1186/s12964-023-01095-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/07/2023] [Indexed: 04/15/2023] Open
Abstract
Androgen deprivation therapy (ADT) is a standard therapy for prostate cancer (PCa). Though disseminated disease is initially sensitive to ADT, an important fraction of the patients progresses to castration-resistant prostate cancer (CRPC). For this reason, the identification of novel effective therapies for treating CRPC is needed. Immunotherapeutic strategies focused on macrophages as antitumor effectors, directly enhancing their tumoricidal potential at the tumor microenvironment or their adoptive transfer after ex vivo activation, have arisen as promising therapies in several cancer types. Despite several approaches centered on the activation of tumor-associated macrophages (TAMs) in PCa are under investigation, to date there is no evidence of clinical benefit in patients. In addition, the evidence of the effectiveness of macrophage adoptive transfer on PCa is poor. Here we find that VSSP, an immunomodulator of the myeloid system, decreases TAMs and inhibits prostatic tumor growth when administered to castrated Pten-deficient prostate tumor-bearing mice. In mice bearing castration-resistant Ptenpc-/-; Trp53pc-/- tumors, VSSP administration showed no effect. Nevertheless, adoptive transfer of macrophages activated ex vivo with VSSP inhibited Ptenpc-/-; Trp53pc-/- tumor growth through reduction of angiogenesis and tumor cell proliferation and induction of senescence. Taken together, our results highlight the rationale of exploiting macrophage functional programming as a promising strategy for CRPC therapy, with particular emphasis on ex vivo-activated proinflammatory macrophage adoptive transfer. Video abstract.
Collapse
Affiliation(s)
- Rydell Alvarez-Arzola
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba.
| | - Nicoló Bancaro
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Ping Lai
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Giuseppe Attanasio
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Laura Pellegrini
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Martina Troiani
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Manuel Colucci
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Simone Mosole
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Emiliano Pasquini
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
| | - Andrea Alimonti
- Department of Molecular Oncology, Institute of Oncology Research (IOR), 6500, Bellinzona, Switzerland
- Faculty of Medicine, Università della Svizzera Italiana, 1011, Lugano, Switzerland
- Department of Medicine, University of Padua, 35131, Padua, Italy
- Medical Oncology, Oncology Institute of Southern Switzerland, 6500, Bellinzona, Switzerland
| | - Circe Mesa
- Innovative Immunotherapy Alliance S.A., Mariel, Artemisa, Cuba
| |
Collapse
|
2
|
Shi T, Sun M, Lu C, Meng F. Self-assembled nanoparticles: A new platform for revolutionizing therapeutic cancer vaccines. Front Immunol 2023; 14:1125253. [PMID: 36895553 PMCID: PMC9988954 DOI: 10.3389/fimmu.2023.1125253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Cancer vaccines have had some success in the past decade. Based on in-depth analysis of tumor antigen genomics, many therapeutic vaccines have already entered clinical trials for multiple cancers, including melanoma, lung cancer, and head and neck squamous cell carcinoma, which have demonstrated impressive tumor immunogenicity and antitumor activity. Recently, vaccines based on self-assembled nanoparticles are being actively developed as cancer treatment, and their feasibility has been confirmed in both mice and humans. In this review, we summarize recent therapeutic cancer vaccines based on self-assembled nanoparticles. We describe the basic ingredients for self-assembled nanoparticles, and how they enhance vaccine immunogenicity. We also discuss the novel design method for self-assembled nanoparticles that pose as a promising delivery platform for cancer vaccines, and the potential in combination with multiple therapeutic approaches.
Collapse
Affiliation(s)
- Tianyu Shi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Mengna Sun
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Changchang Lu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
3
|
Bergado-Báez G, Gonzalez Suarez N, García LC, Pérez-Martínez D, Hernández-Fernández DR, Fundora-Barrios T, Rodríguez-Álvarez A, Díaz-Ordaz GD, Lindzen M, Yarden Y, Sánchez-Ramírez B. Polyclonal antibody-induced downregulation of HER1/EGFR and HER2 surpasses the effect of combinations of specific registered antibodies. Front Oncol 2022; 12:951267. [PMID: 36408164 PMCID: PMC9667895 DOI: 10.3389/fonc.2022.951267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Background Antitumor therapies targeting HER1/EGFR and HER2, such as monoclonal antibodies (MAbs) and tyrosine-kinase inhibitors (TKIs), have demonstrated a significant clinical benefit, but the emergence of resistance limits long-term efficacy. While secondary HER1 mutations confer tolerance to TKI, compensatory upregulation of HER2 drives resistance to anti-HER1 MAbs, which identifies MAb combinations targeting both receptors as an attractive therapeutic strategy. Nevertheless, toxicity hampers the clinical validation of this approach. Alternatively, cancer vaccines may induce antibodies directed against several antigens with less concern about induced toxicity. Methods Polyclonal antibodies (PAbs) targeting HER1 and HER2 were induced in mice or rabbits through immunization. Recognition of different epitopes on targets by PAbs was validated by phage-display technology. Receptor downregulation was evaluated by flow cytometry, immunofluorescence, and Western blot. MTT assays assessed cytotoxicity, while the antitumor effect of PAbs was assayed in nude mice. Results PAbs promoted degradation of HER1 and HER2 regarding clinical MAbs or their combinations. As a result, inhibition of cytotoxicity on tumor cell lines was improved, even in the presence of oncogenic mutations in HER1, as well as in cetuximab-insensitive cells. Accordingly, the antitumor effect of vaccination-induced PAbs was observed in lung tumor lines representative of sensitivity or resistance to HER1 targeting therapies. Conclusions Immunization against HER1 and HER2 receptors offers an alternative to passive administration of combinations of MAbs, since vaccination-induced PAbs promote the downregulation of both receptors and they have a higher impact on the survival of tumor cells.
Collapse
Affiliation(s)
- Gretchen Bergado-Báez
- Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Narjara Gonzalez Suarez
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, Université du Québec à, Montréal, QC, Canada
| | - Lisset Chao García
- Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Dayana Pérez-Martínez
- Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Talia Fundora-Barrios
- Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | - Moshit Lindzen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Belinda Sánchez-Ramírez
- Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
- *Correspondence: Belinda Sánchez-Ramírez,
| |
Collapse
|
4
|
Khan ANH, Emmons TR, Magner WJ, Alqassim E, Singel KL, Ricciuti J, Eng KH, Odunsi K, Tomasi TB, Lee K, Abrams SI, Mesa C, Segal BH. VSSP abrogates murine ovarian tumor-associated myeloid cell-driven immune suppression and induces M1 polarization in tumor-associated macrophages from ovarian cancer patients. Cancer Immunol Immunother 2022; 71:2355-2369. [PMID: 35166871 PMCID: PMC10591410 DOI: 10.1007/s00262-022-03156-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/16/2022] [Indexed: 02/07/2023]
Abstract
The ovarian tumor microenvironment (TME) is characterized by the accumulation of immunosuppressive tumor-associated macrophages (TAMs) and granulocytic cells. Very small size particles (VSSP), comprised of the ganglioside NAcGM3 and Neisseria meningitidis derived outer membrane vesicles, is being developed as a nanoparticulated modulator of innate immunity. Prior studies have shown that VSSP enhanced antigen-specific cytotoxic T cell responses and reduced the suppressive phenotype of splenic granulocytic cells in tumor-bearing mice. Here, we hypothesized that intraperitoneal VSSP would modify myeloid cell accumulation and phenotypes in the ovarian TME and abrogate suppressor function of TAMs and tumor-associated granulocytic cells. In the ID8 syngeneic model of epithelial ovarian cancer, VSSP reduced peritoneal TAMs and induced M1-like polarization in TAMs. In addition, VSSP stimulated peritoneal inflammation characterized by increased granulocytes and monocytes, including inflammatory monocytic cells. VSSP treatment resulted in peritoneal TAMs and granulocytic cells being less suppressive of ex vivo stimulated CD8+ T cell responses. VSSP alone and combined with anti-PD-1 modestly but significantly prolonged survival in tumor-bearing mice. In addition, ex vivo treatment with VSSP induced M1-like polarization in TAMs from patients with metastatic ovarian cancer and variably abrogated their suppressor phenotype. VSSP treatment also partially abrogated the induction of suppressor function in healthy donor neutrophils exposed to ascites supernatants from patients with ovarian cancer. Together, these results point to VSSP reprogramming myeloid responses resulting in abrogation of suppressive pathways and raise the potential for administration of VSSP into the TME to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Anm Nazmul H Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
| | - Tiffany R Emmons
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William J Magner
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
- Department of Microbiology & Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Emad Alqassim
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Kelly L Singel
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
- Office of Evaluation, Performance, and Reporting; Division of Program Coordination, Planning, and Strategic Initiatives; Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Jason Ricciuti
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kevin H Eng
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| | - Thomas B Tomasi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
- Department of Microbiology & Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Kelvin Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
- Department of Medicine, Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA
| | - Circe Mesa
- Center of Molecular Immunology, Havana, Cuba
- Innovative Immunotherapy Alliance, S. A. Mariel, Artemisa, Cuba
| | - Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA.
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, USA.
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
5
|
Oliver L, Alvarez R, Diaz R, Valdés A, Colligan SH, Nemeth MJ, Twum DYF, Fernández A, Fernández-Medina O, Carlson LM, Yu H, Eng KH, Hensen ML, Rábade-Chediak ML, Fernández LE, Lee KP, Perez L, Muhitch JB, Mesa C, Abrams SI. Mitigating the prevalence and function of myeloid-derived suppressor cells by redirecting myeloid differentiation using a novel immune modulator. J Immunother Cancer 2022; 10:e004710. [PMID: 36150744 PMCID: PMC9511656 DOI: 10.1136/jitc-2022-004710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Immune suppression is common in neoplasia and a major driver is tumor-induced myeloid dysfunction. Yet, overcoming such myeloid cell defects remains an untapped strategy to reverse suppression and improve host defense. Exposure of bone marrow progenitors to heightened levels of myeloid growth factors in cancer or following certain systemic treatments promote abnormal myelopoiesis characterized by the production of myeloid-derived suppressor cells (MDSCs) and a deficiency in antigen-presenting cell function. We previously showed that a novel immune modulator, termed 'very small size particle' (VSSP), attenuates MDSC function in tumor-bearing mice, which was accompanied by an increase in dendritic cells (DCs) suggesting that VSSP exhibits myeloid differentiating properties. Therefore, here, we addressed two unresolved aspects of the mechanism of action of this unique immunomodulatory agent: (1) does VSSP alter myelopoiesis in the bone marrow to redirect MDSC differentiation toward a monocyte/macrophage or DC fate? and (2) does VSSP mitigate the frequency and suppressive function of human tumor-induced MDSCs? METHODS To address the first question, we first used a murine model of granulocyte-colony stimulating factor-driven emergency myelopoiesis following chemotherapy-induced myeloablation, which skews myeloid output toward MDSCs, especially the polymorphonuclear (PMN)-MDSC subset. Following VSSP treatment, progenitors and their myeloid progeny were analyzed by immunophenotyping and MDSC function was evaluated by suppression assays. To strengthen rigor, we validated our findings in tumor-bearing mouse models. To address the second question, we conducted a clinical trial in patients with metastatic renal cell carcinoma, wherein 15 patients were treated with VSSP. Endpoints in this study included safety and impact on PMN-MDSC frequency and function. RESULTS We demonstrated that VSSP diminished PMN-MDSCs by shunting granulocyte-monocyte progenitor differentiation toward monocytes/macrophages and DCs with heightened expression of the myeloid-dependent transcription factors interferon regulatory factor-8 and PU.1. This skewing was at the expense of expansion of granulocytic progenitors and rendered the remaining MDSCs less suppressive. Importantly, these effects were also demonstrated in a clinical setting wherein VSSP monotherapy significantly reduced circulating PMN-MDSCs, and their suppressive function. CONCLUSIONS Altogether, these data revealed VSSP as a novel regulator of myeloid biology that mitigates MDSCs in cancer patients and reinstates a more normal myeloid phenotype that potentially favors immune activation over immune suppression.
Collapse
Affiliation(s)
- Liliana Oliver
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Rydell Alvarez
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Raquel Diaz
- Department of Oncology, Joaquín Albarrán Hospital, Havana, Cuba
| | - Anet Valdés
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Sean H Colligan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Michael J Nemeth
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Danielle Y F Twum
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Audry Fernández
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Olivia Fernández-Medina
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Louise M Carlson
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Han Yu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kevin H Eng
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Mary L Hensen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Maura L Rábade-Chediak
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Luis Enrique Fernández
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Leslie Perez
- Clinical Direction, Center of Molecular Immunology, Havana, Cuba
| | - Jason B Muhitch
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Circe Mesa
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
- Innovative Immunotherapy Alliance, S. A. Mariel, Artemisa, Cuba
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
6
|
Liao Z, Huang J, Lo PC, Lovell JF, Jin H, Yang K. Self-adjuvanting cancer nanovaccines. J Nanobiotechnology 2022; 20:345. [PMID: 35883176 PMCID: PMC9316869 DOI: 10.1186/s12951-022-01545-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/04/2022] [Indexed: 11/12/2022] Open
Abstract
Nanovaccines, a new generation of vaccines that use nanoparticles as carriers and/or adjuvants, have been widely used in the prevention and treatment of various diseases, including cancer. Nanovaccines have sparked considerable interest in cancer therapy due to a variety of advantages, including improved access to lymph nodes (LN), optimal packing and presentation of antigens, and induction of a persistent anti-tumor immune response. As a delivery system for cancer vaccines, various types of nanoparticles have been designed to facilitate the delivery of antigens and adjuvants to lymphoid organs and antigen-presenting cells (APCs). Particularly, some types of nanoparticles are able to confer an immune-enhancing capability and can themselves be utilized for adjuvant-like effect for vaccines, suggesting a direction for a better use of nanomaterials and the optimization of cancer vaccines. However, this role of nanoparticles in vaccines has not been well studied. To further elucidate the role of self-adjuvanting nanovaccines in cancer therapy, we review the mechanisms of antitumor vaccine adjuvants with respect to nanovaccines with self-adjuvanting properties, including enhancing cross-presentation, targeting signaling pathways, biomimicking of the natural invasion process of pathogens, and further unknown mechanisms. We surveyed self-adjuvanting cancer nanovaccines in clinical research and discussed their advantages and challenges. In this review, we classified self-adjuvanting cancer nanovaccines according to the underlying immunomodulatory mechanism, which may provide mechanistic insights into the design of nanovaccines in the future.
Collapse
Affiliation(s)
- Zhiyun Liao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
Santana-Mederos D, Perez-Nicado R, Climent Y, Rodriguez L, Ramirez BS, Perez-Rodriguez S, Rodriguez M, Labrada C, Hernandez T, Diaz M, Orosa I, Ramirez U, Oliva R, Garrido R, Cardoso F, Landys M, Martinez R, Gonzalez H, Hernandez T, Ochoa-Azze R, Perez JL, Enriquez J, Gonzalez N, Infante Y, Espinosa LA, Ramos Y, González LJ, Valenzuela C, Casadesus AV, Fernandez B, Rojas G, Pérez-Massón B, Tundidor Y, Bermudez E, Plasencia CA, Boggiano T, Ojito E, Chiodo F, Fernandez S, Paquet F, Fang C, Chen GW, Rivera DG, Valdes-Balbin Y, Garcia-Rivera D, Verez Bencomo V. A COVID-19 vaccine candidate composed of the SARS-CoV-2 RBD dimer and Neisseria meningitidis outer membrane vesicles. RSC Chem Biol 2022; 3:242-249. [PMID: 35360883 PMCID: PMC8826971 DOI: 10.1039/d1cb00200g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Soberana01 is composed of the SARS-CoV-2 dimeric RBD and Neisseria meningitidis outer membrane vesicles (OMVs) adsorbed on alum. This vaccine induces a potent neutralizing immune response and shows potential against SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
| | | | - Yanet Climent
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Laura Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | - Meybi Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Claudia Labrada
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Tays Hernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Marianniz Diaz
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ivette Orosa
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ubel Ramirez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Reynaldo Oliva
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Raine Garrido
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Felix Cardoso
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Mario Landys
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | | | | | - Jose L. Perez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Juliet Enriquez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Nibaldo Gonzalez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Yenicet Infante
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Luis A. Espinosa
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Luis Javier González
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Carmen Valenzuela
- Institute of Cybernetics, Mathematics and Physics, Havana 10400, Cuba
| | | | - Briandy Fernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Gertrudis Rojas
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | - Yaima Tundidor
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ernesto Bermudez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | - Tammy Boggiano
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Eduardo Ojito
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Fabrizio Chiodo
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands and Institute of Biomolecular Chemistry, National Research Council (CNR), Pozzuoli, Napoli, Italy
| | | | - Françoise Paquet
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, F-45071, Orléans Cedex 2, France
| | - Cheng Fang
- Shanghai Fenglin Glycodrug Promotion Center, Shanghai 200032, China
| | - Guang-Wu Chen
- Chengdu Olisynn Biotech. Co. Ltd., and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Daniel G. Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | | | | | | |
Collapse
|
8
|
Christodoulides M, Humbert MV, Heckels JE. The potential utility of liposomes for Neisseria vaccines. Expert Rev Vaccines 2021; 20:1235-1256. [PMID: 34524062 DOI: 10.1080/14760584.2021.1981865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Species of the genus Neisseria are important global pathogens. Neisseria gonorrhoeae (gonococcus) causes the sexually transmitted disease gonorrhea and Neisseria meningitidis (meningococcus) causes meningitis and sepsis. Liposomes are self-assembled spheres of phospholipid bilayers enclosing a central aqueous space, and they have attracted much interest and use as a delivery vehicle for Neisseria vaccine antigens. AREAS COVERED A brief background on Neisseria infections and the success of licensed meningococcal vaccines are provided. The absence of a gonococcal vaccine is highlighted. The use of liposomes for delivering Neisseria antigens and adjuvants, for the purposes of generating specific immune responses, is reviewed. The use of other lipid-based systems for antigen and adjuvant delivery is examined briefly. EXPERT OPINION With renewed interest in developing a gonococcal vaccine, liposomes remain an attractive option for delivering antigens. The discipline of nanotechnology provides additional nanoparticle-based options for gonococcal vaccine development. Future work would be needed to tailor the composition of liposomes and other nanoparticles to the specific vaccine antigen(s), in order to generate optimal anti-gonococcal immune responses. The potential use of liposomes and other nanoparticles to deliver anti-gonococcal compounds to treat infections also should be explored further.
Collapse
Affiliation(s)
- Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Maria Victoria Humbert
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - John E Heckels
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| |
Collapse
|
9
|
Choudhury SD. Nano-Medicines a Hope for Chagas Disease! Front Mol Biosci 2021; 8:655435. [PMID: 34141721 PMCID: PMC8204082 DOI: 10.3389/fmolb.2021.655435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Chagas disease, is a vector-mediated tropical disease whose causative agent is a parasitic protozoan named Trypanosoma cruzi. It is a very severe health issue in South America and Mexico infecting millions of people every year. Protozoan T. cruzi gets transmitted to human through Triatominae, a subfamily of the Reduviidae, and do not have any effective treatment or preventative available. The lack of economic gains from this tropical parasitic infection, has always been the reason behind its negligence by researchers and drug manufacturers for many decades. Hence there is an enormous requirement for more efficient and novel strategies to reduce the fatality associated with these diseases. Even, available diagnosis protocols are outdated and inefficient and there is an urgent need for rapid high throughput diagnostics as well as management protocol. The current advancement of nanotechnology in the field of healthcare has generated hope for better management of many tropical diseases including Chagas disease. Nanoparticulate systems for drug delivery like poloxamer coated nanosuspension of benzimidazole have shown promising results in reducing toxicity, elevating efficacy and bioavailability of the active compound against the pathogen, by prolonging release, thereby increasing the therapeutic index. Moreover, nanoparticle-based drug delivery has shown promising results in inducing the host’s immune response against the pathogen with very few side effects. Besides, advances in diagnostic assays, such as nanosensors, aided in the accurate detection of the parasite. In this review, we provide an insight into the life cycle stages of the pathogen in both vertebrate host and the insect vector, along with an overview of the current therapy for Chagas disease and its limitations; nano carrier-based delivery systems for antichagasic agents, we also address the advancement of nano vaccines and nano-diagnostic techniques, for treatment of Chagas disease, majorly focusing on the novel perspectives in combating the disease.
Collapse
|
10
|
Tamargo SB, Bui Thanh T, Pérez M, Otero O, Oliva HR, Falero G, Pérez JL, Cedré MB, Okuskhanova E, Thiruvengadam M, Shariati MA, Sierra GVG. Nanocochleates containing N-Octylglicoside extracted Vibrio cholerae antigens elicited high vibriocidal antibodies titers after intragastric immunization in a mice model. Microb Pathog 2021; 156:104902. [PMID: 33930421 DOI: 10.1016/j.micpath.2021.104902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/31/2021] [Accepted: 04/01/2021] [Indexed: 09/30/2022]
Abstract
Biological detergents are used in research laboratories, to extract or solubilize proteins from cell membranes. In order to evaluate the ability to extract antigens from the bacterial cell surface of the wild Vibrio cholerae strain C7258 and study their immunogenic potential by forming proteoliposomes and cochleate and preserving their immunogenicity, the non-ionic detergent, n-Octylglucoside (n-OG), and the Zwitterionic detergent (3-cholamidopropyl dimethylammonio 1-propanesulfonate; CHAPS) were tested in concentrations between 5 and 15%. The anionic detergent sodium deoxycholate (DOC) was used as a reference. Electrophoretic, immunochemical and electron microscopy techniques have characterized the extracts and their chromatographic fractions. With CHAPS and n-OG detergents in concentrations between 5 and 15%, a higher yield was obtained in the extraction of proteins and lipopolysaccharides (LPS) and other components from the bacterial surface compared to 10% DOC. When using 10% DOC, 15% CHAPS and n-OG between 5 and 15%, stable proteoliposomes were formed, of average size between 82 and 93 nm in diameter, with known proportions of proteins, LPS and other components. In some of the concentrations, liposomes were formed with almost pure proteins. Some cholera outer membrane proteins like the 17 kDa protein, which corresponds to the mannose-sensitive hemagglutinin (MSHA), which mediates the adhesion to the brush border of the small intestine and the outer membrane protein U (OMPU) were identified with monoclonal antibodies (mAbs) and purified. The fundamental components of liposomes, proteins and LPS, retained their molecular weights, when compared with known standards and by processing programs of electrophoretic profiles and their antigenicity, without alterations due to the extraction procedure, as could be verified by immune identification techniques with monoclonal antibodies in the case of LPS, significant antigens in this pathogen. The main purpose of the present work was to show that a new anticholera vaccine formulation based on cochleates, containing selected protein and LPS fraction extracted by detergents, is able to elicit protective high titers of bactericidal antibodies after intragastric immunization in the mice model. The objective was achieved.
Collapse
Affiliation(s)
- S B Tamargo
- Latin American School of Medicine, Havana, Cuba; Jiangxi Institute of Biological Products, Ji'An, China
| | - T Bui Thanh
- Pharmacy and Foods Institute, University of Havana, Cuba
| | - M Pérez
- Pharmacy and Foods Institute, University of Havana, Cuba
| | - O Otero
- Finlay Institute for Vaccines, Havana City, Cuba
| | - H R Oliva
- Finlay Institute for Vaccines, Havana City, Cuba
| | - G Falero
- Finlay Institute for Vaccines, Havana City, Cuba
| | - J L Pérez
- Finlay Institute for Vaccines, Havana City, Cuba
| | - M B Cedré
- Finlay Institute for Vaccines, Havana City, Cuba
| | - E Okuskhanova
- Shakarim State University of Semey, Semey, 071412, Kazakhstan
| | - M Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - M A Shariati
- Laboratory of Biological Control and Antimicrobial Resistance, Orel State University, Orel City, 302026, Russia; K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation; Kazakh Research Institute of Processing and Food Industry (Semey Branch), Kazakhstan
| | - G V G Sierra
- Group of Biotechnology and Pharmaceutical Industries of Cuba (BioCubaFarma), Havana, Cuba; Jiangxi Institute of Biological Products, Ji'An, China.
| |
Collapse
|
11
|
Lampe AT, Farris EJ, Brown DM, Pannier AK. High- and low-molecular-weight chitosan act as adjuvants during single-dose influenza A virus protein vaccination through distinct mechanisms. Biotechnol Bioeng 2020; 118:1224-1243. [PMID: 33289090 PMCID: PMC7897297 DOI: 10.1002/bit.27647] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/20/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022]
Abstract
The investigation of new adjuvants is essential for the development of efficacious vaccines. Chitosan (CS), a derivative of chitin, has been shown to act as an adjuvant, improving vaccine-induced immune responses. However, the effect of CS molecular weight (MW) on this adjuvanticity has not been investigated, despite MW having been shown to impact CS biological properties. Here, two MW variants of CS were investigated for their ability to enhance vaccine-elicited immune responses in vitro and in vivo, using a single-dose influenza A virus (IAV) protein vaccine model. Both low-molecular-weight (LMW) and high-molecular-weight (HMW) CS-induced interferon regulatory factor pathway signaling, antigen-presenting cell activation, and cytokine messenger RNA (mRNA) production, with LMW inducing higher mRNA levels at 24 h and HMW elevating mRNA responses at 48 h. LMW and HMW CS also induced adaptive immune responses after vaccination, indicated by enhanced immunoglobulin G production in mice receiving LMW CS and increased CD4 interleukin 4 (IL-4) and IL-2 production in mice receiving HMW CS. Importantly, both LMW and HMW CS adjuvantation reduced morbidity following homologous IAV challenge. Taken together, these results support that LMW and HMW CS can act as adjuvants, although this protection may be mediated through distinct mechanisms based on CS MW.
Collapse
Affiliation(s)
- Anna T Lampe
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.,Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Eric J Farris
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.,Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.,Trudeau Institute, Saranac Lake, NY, USA
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
12
|
Quijia Quezada C, Azevedo CS, Charneau S, Santana JM, Chorilli M, Carneiro MB, Bastos IMD. Advances in nanocarriers as drug delivery systems in Chagas disease. Int J Nanomedicine 2019; 14:6407-6424. [PMID: 31496694 PMCID: PMC6691952 DOI: 10.2147/ijn.s206109] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Chagas disease is one of the most important public health problems in Latin America due to its high mortality and morbidity levels. There is no effective treatment for this disease since drugs are usually toxic with low bioavailability. Serious efforts to achieve disease control and eventual eradication have been unsuccessful to date, emphasizing the need for rapid diagnosis, drug development, and a reliable vaccine. Novel systems for drug and vaccine administration based on nanocarriers represent a promising avenue for Chagas disease treatment. Nanoparticulate systems can reduce toxicity, and increase the efficacy and bioavailability of active compounds by prolonging release, and therefore improve the therapeutic index. Moreover, nanoparticles are able to interact with the host's immune system, modulating the immune response to favour the elimination of pathogenic microorganisms. In addition, new advances in diagnostic assays, such as nanobiosensors, are beneficial in that they enable precise identification of the pathogen. In this review, we provide an overview of the strategies and nanocarrier-based delivery systems for antichagasic agents, such as liposomes, micelles, nanoemulsions, polymeric and non-polymeric nanoparticles. We address recent progress, with a particular focus on the advances of nanovaccines and nanodiagnostics, exploring new perspectives on Chagas disease treatment.
Collapse
Affiliation(s)
- Christian Quijia Quezada
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
- Department of Drugs and Medicines, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Clênia S Azevedo
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Sébastien Charneau
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Jaime M Santana
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marcella B Carneiro
- Electron Microscopy Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Izabela Marques Dourado Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| |
Collapse
|
13
|
Junco JA, Rodríguez R, Fuentes F, Baladrón I, Castro MD, Calzada L, Valenzuela C, Bover E, Pimentel E, Basulto R, Arteaga N, Cid-Arregui A, Sariol F, González L, Porres-Fong L, Medina M, Rodríguez A, Garay AH, Reyes O, López M, de Quesada L, Alvarez A, Martínez C, Marrero M, Molero G, Guerra A, Rosales P, Capote C, Acosta S, Vela I, Arzuaga L, Campal A, Ruiz E, Rubio E, Cedeño P, Sánchez MC, Cardoso P, Morán R, Fernández Y, Campos M, Touduri H, Bacardi D, Feria I, Ramirez A, Cosme K, Saura PL, Quintana M, Muzio V, Bringas R, Ayala M, Mendoza M, Fernández LE, Carr A, Herrera L, Guillén G. Safety and Therapeutic Profile of a GnRH-Based Vaccine Candidate Directed to Prostate Cancer. A 10-Year Follow-Up of Patients Vaccinated With Heberprovac. Front Oncol 2019; 9:49. [PMID: 30859088 PMCID: PMC6397853 DOI: 10.3389/fonc.2019.00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 01/17/2019] [Indexed: 12/25/2022] Open
Abstract
Heberprovac is a GnRH based vaccine candidate containing 2.4 mg of the GnRHm1-TT peptide as the main active principle; 245 μg of the very small size proteoliposomes adjuvant (VSSP); and 350 μL of Montanide ISA 51 VG oil adjuvant. The aim of this study was to assess the safety and tolerance of the Heberprovac in advanced prostate cancer patients as well as its capacity to induce anti-GnRH antibodies, the subsequent effects on serum levels of testosterone and PSA and the patient overall survival. The study included eight patients with histologically-proven advanced prostate cancer with indication for hormonal therapy, who received seven intramuscular immunizations with Heberprovac within 18 weeks. Anti-GnRH antibody titers, testosterone and PSA levels, as well as clinical parameters were recorded and evaluated. The vaccine was well tolerated. Significant reductions in serum levels of testosterone and PSA were seen after four immunizations. Castrate levels of testosterone were observed in all patients at the end of the immunization schedule, which remained at the lowest level for at least 20 months. In a 10-year follow-up three out of six patients who completed the entire trial survived. In contrast only one out eight patients survived in the same period in a matched randomly selected group receiving standard anti-hormonal treatment. Heberprovac vaccination showed a good security profile, as well as immunological, biochemical and, most importantly, clinical benefit. The vaccinated group displayed survival advantage compared with the reference group that received standard treatment. These results warrant further clinical trials with Heberprovac involving a larger cohort.
Collapse
Affiliation(s)
- Jesús A. Junco
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Ranfis Rodríguez
- Uro-oncology Department of National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Franklin Fuentes
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Idania Baladrón
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Maria D. Castro
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Lesvia Calzada
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | | | - Eddy Bover
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | | | - Roberto Basulto
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Niurka Arteaga
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | | | | | | | | | - María Medina
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Ayni Rodríguez
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - A. Hilda Garay
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Osvaldo Reyes
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Matilde López
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | | | | | | | | | - Alfredo Guerra
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Pedro Rosales
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Carlos Capote
- Amalia Simoni Clinical-Surgical Hospital, Camagüey, Cuba
| | - Sahily Acosta
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Idania Vela
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Lina Arzuaga
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Ana Campal
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Erlán Ruiz
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Elier Rubio
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Pável Cedeño
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - María Carmen Sánchez
- Clinical Laboratory of the Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Pedro Cardoso
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Rolando Morán
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Yairis Fernández
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Magalys Campos
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Henio Touduri
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Dania Bacardi
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Indalecio Feria
- Clinical Trials Department of Oncologic Hospital Marie Curie of Camaguey, Marie Curie, Camagüey, Cuba
| | - Amilcar Ramirez
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Karelia Cosme
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | | | - Verena Muzio
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Ricardo Bringas
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Marta Ayala
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mario Mendoza
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | | | | | - Luis Herrera
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
- BioCubafarma, Havana, Cuba
| | - Gerardo Guillén
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
14
|
Bergado Báez G, Hernández Fernández DR, Mazorra Herrera Z, Sánchez Ramírez B. HER1-based vaccine: Simultaneous activation of humoral and cellular immune response. Semin Oncol 2018; 45:75-83. [PMID: 30318087 DOI: 10.1053/j.seminoncol.2018.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023]
Abstract
The human epidermal growth factor receptor 1 (HER1) is a tumor-associated antigen that has been validated as a clinical target for several passive, non-immune therapies currently approved for the treatment of epithelial tumors. HER1 is an oncogene that not only promotes tumor progression and survival, but also immune escape. Its overexpression in some epithelial malignancies has been correlated with a poor prognosis. We developed an approach to target HER1 by specific active immunotherapy, recognizing the extracellular domain of the receptor, using a combination of VSSP and Montanide ISA 51 as adjuvants. We summarize the results obtained with this vaccine in both the preclinical and clinical settings, emphasizing the importance of the induction of both humoral and cellular responses for the success of cancer vaccines as safe therapeutic alternatives for the treatment of cancer.
Collapse
|
15
|
Alvarez R, Oliver L, Valdes A, Mesa C. Cancer-induced systemic myeloid dysfunction: Implications for treatment and a novel nanoparticle approach for its correction. Semin Oncol 2018; 45:84-94. [PMID: 30318088 DOI: 10.1053/j.seminoncol.2018.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 05/17/2018] [Indexed: 02/08/2023]
Abstract
Unlike other regulatory circuits, cancer-induced myeloid dysfunction involves more than an accumulation of impaired dendritic cells, protumoral macrophages, and myeloid derived suppressor cells in the tumor microenvironment. It is also characterized by "aberrant" myelopoiesis that results in the accumulation and expansion of immature myeloid precursors with a suppressive phenotype in the systemic circulation. The first part of this review briefly describes the evidence for and consequences of this systemic dysfunctional myelopoiesis and the possible reinforcement of this phenomenon by conventional treatments used in patients with cancer, in particular chemotherapy and granulocyte-colony stimulating factor. The second half of this review describes very small size particles, a novel immune-modulatory nanoparticle, and the evidence indicating a possible role of this agent in correcting or re-programming the dysfunctional myelopoiesis in different scenarios.
Collapse
Affiliation(s)
- Rydell Alvarez
- Immunobiology Division, Institute of Molecular Immunology, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Liliana Oliver
- Immunobiology Division, Institute of Molecular Immunology, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Anet Valdes
- Immunobiology Division, Institute of Molecular Immunology, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Circe Mesa
- Immunobiology Division, Institute of Molecular Immunology, Center of Molecular Immunology (CIM), Havana, Cuba.
| |
Collapse
|
16
|
Sánchez Ramírez J, Morera Díaz Y, Bequet-Romero M, Hernández-Bernal F, Selman-Housein Bernal KH, de la Torre Santos A, Santiesteban Álvarez ER, Martín Bauta Y, Bermúdez Badell CH, de la Torre Pupo J, Gavilondo JV, Ayala Avila M. Characteristics of the specific humoral response in patients with advanced solid tumors after active immunotherapy with a VEGF vaccine, at different antigen doses and using two distinct adjuvants. BMC Immunol 2017; 18:39. [PMID: 28747172 PMCID: PMC5530503 DOI: 10.1186/s12865-017-0222-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 07/20/2017] [Indexed: 12/19/2022] Open
Abstract
Background CIGB-247, a VSSP-adjuvanted VEGF-based vaccine, was evaluated in a phase I clinical trial in patients with advanced solid tumors (CENTAURO). Vaccination with the maximum dose of antigen showed an excellent safety profile, exhibited the highest immunogenicity and was the only one showing a reduction on platelet VEGF bioavailability. However, this antigen dose level did not achieve a complete seroconversion rate in vaccinated patients. These clinical results led us to the question whether a “reserve” of untapped immune response potential against VEGF could exist in cancer patients. To address this matter, CENTAURO-2 clinical trial was conducted where antigen and VSSP dose scale up were studied, and also incorporated the exploration of aluminum phosphate as adjuvant. These changes were made with the aim to increase immune response against VEGF. Results The present study reports the characterization of the humoral response elicited by CIGB-247 from the combining of different antigen doses and adjuvants. Cancer patients were immunologically monitored for approximately 1 year. Vaccination with different CIGB-247 formulations exhibited a very positive safety profile. Cancer patients developed IgM, IgG or IgA antibodies specific to VEGF. Elicited polyclonal antibodies had the ability to block the interaction between VEGF and its receptors, VEGFR1 and VEGFR2. The highest humoral response was detected in patients immunized with 800 μg of antigen + 200 μg of VSSP. Off-protocol long-term vaccination did not produce negative changes in humoral response. Conclusions Vaccination with a human VEGF variant molecule as antigen in combination with VSSP or aluminum phosphate is immunogenic. The results of this study could contribute to the investigation of this vaccine therapy in an adequately powered efficacy trial. Trial registration Trial registration number: RPCEC00000155. Cuban Public Clinical Trial Registry. Date of registration: June 06, 2013. Available from: http://registroclinico.sld.cu/. Electronic supplementary material The online version of this article (doi:10.1186/s12865-017-0222-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Javier Sánchez Ramírez
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Playa Cubanacán, Havana, 10600, Cuba.
| | - Yanelys Morera Díaz
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Playa Cubanacán, Havana, 10600, Cuba
| | - Mónica Bequet-Romero
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Playa Cubanacán, Havana, 10600, Cuba
| | | | | | | | | | - Yenima Martín Bauta
- Department of Clinical Research, CIGB, P.O. Box 6162, Playa Cubanacán, Havana, 10600, Cuba
| | | | | | - Jorge V Gavilondo
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Playa Cubanacán, Havana, 10600, Cuba
| | | | - Marta Ayala Avila
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, Playa Cubanacán, Havana, 10600, Cuba
| |
Collapse
|
17
|
Chesney JA, Mitchell RA, Yaddanapudi K. Myeloid-derived suppressor cells-a new therapeutic target to overcome resistance to cancer immunotherapy. J Leukoc Biol 2017; 102:727-740. [PMID: 28546500 DOI: 10.1189/jlb.5vmr1116-458rrr] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that accumulate during pathologic conditions, such as cancer. Patients diagnosed with advanced metastatic cancers have an average survival of 12-24 mo, a survival time that hasn't changed significantly in the past 30 yr. Despite some encouraging improvements in response rates and overall survival in patients receiving immunotherapies, such as immune checkpoint inhibitors, most patients will ultimately progress. MDSCs contribute to immunotherapeutic resistance by actively inhibiting antitumor T cell proliferation and cytotoxic activity as well as by promoting expansion of protumorigenic T regulatory cells, thereby, dampening the host immune responses against the tumor. In addition, MDSCs promote angiogenesis, tumor invasion, and metastasis. Thus, MDSCs are potential therapeutic targets in cases of multiple cancers. This review focuses on the phenotypic and functional characteristics of MDSCs and provides an overview of the mono- and combinatorial-therapeutic strategies that target MDSCs with an objective of enhancing the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Jason A Chesney
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and
| | - Robert A Mitchell
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Kavitha Yaddanapudi
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
18
|
Caballero I, Aira LE, Lavastida A, Popa X, Rivero J, González J, Mesa M, González N, Coba K, Lorenzo-Luaces P, Wilkinson B, Santiesteban Y, Santiesteban Y, Troche M, Suarez E, Crombet T, Sánchez B, Casacó A, Macías A, Mazorra Z. Safety and Immunogenicity of a Human Epidermal Growth Factor Receptor 1 (HER1)-Based Vaccine in Prostate Castration-Resistant Carcinoma Patients: A Dose-Escalation Phase I Study Trial. Front Pharmacol 2017; 8:263. [PMID: 28539888 PMCID: PMC5423955 DOI: 10.3389/fphar.2017.00263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (CRPC) remains incurable due to the lack of effective therapies. Activation of the human epidermal growth factor receptor 1 (HER1) in prostate cancer contributes to metastatic progression as well as to disease relapse. Here, we determined the toxicity and immunogenicity of a HER1-based cancer vaccine in CRPC patients included in a phase I clinical trial. CRPC patients (n = 24) were intramuscularly vaccinated with HER1 vaccine consisting of the extracellular domain of HER1 molecule (ECD) and very small size proteoliposome from Neisseria meningitidis (VSSP) and Montanide ISA-51 VG as adjuvants. Patients were included in five groups according to the vaccine dose (100, 200, 400, 600, and 800 μg). The primary endpoints were safety and immunogenicity. The anti-HER1 antibodies were measured by an ELISA, the recognition of an HER1 positive tumor cell line and the inhibition of HER1 phosphorylation by sera were determined by flow cytometry and western blot analysis, respectively. The HER1-specific T cell response was assessed by determination of IFN-γ-producing T cells using ELISpot assay. The vaccine was well tolerated. No grade III or IV adverse events were reported. High titers of anti-HER1 antibodies were observed in most of the evaluated patients. There were no significant differences regarding the geometric means of the anti-HER1 titers among the dose groups except the group of 100 μg in which antibody titers were significantly lower. A Th1-type IgG subclasses pattern was predominant in most patients. Only patients receiving the higher doses of vaccine showed significant tumor cell recognition and HER1 phosphorylation inhibition by hyperimmune sera. Forty two percent of the patients showed a specific T cell response against HER1 peptides pool in post-treatment samples. There was a trend toward survival benefit in those patients showing high anti-HER1 specific antibody titers and a significant association between cellular immune response and clinical outcome.
Collapse
Affiliation(s)
- Iraida Caballero
- Department of Oncology, Hermanos Ameijeiras HospitalHavana, Cuba
| | - Lazaro E Aira
- Department of Clinical Immunology, Center of Molecular ImmunologyHavana, Cuba
| | - Anabel Lavastida
- Department of Clinical Immunology, Center of Molecular ImmunologyHavana, Cuba
| | - Xitlally Popa
- Department of Clinical Immunology, Center of Molecular ImmunologyHavana, Cuba
| | | | - Joaquín González
- Department of Oncology, Hermanos Ameijeiras HospitalHavana, Cuba
| | - Mónica Mesa
- Tumor Immunology Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Narjara González
- Tumor Immunology Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Kelly Coba
- Faculty of Medicine "Victoria de Girón"Havana, Cuba
| | | | - Barbara Wilkinson
- Clinical Trials Direction, Center of Molecular ImmunologyHavana, Cuba
| | | | | | - Mayelin Troche
- Clinical Trials Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Eduardo Suarez
- Department of Innovation, Center of Molecular ImmunologyHavana, Cuba
| | - Tania Crombet
- Clinical Trials Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Belinda Sánchez
- Tumor Immunology Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Angel Casacó
- Clinical Trials Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Amparo Macías
- Clinical Trials Direction, Center of Molecular ImmunologyHavana, Cuba
| | - Zaima Mazorra
- Department of Clinical Immunology, Center of Molecular ImmunologyHavana, Cuba
| |
Collapse
|
19
|
Anani W, Shurin MR. Targeting Myeloid-Derived Suppressor Cells in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:105-128. [PMID: 29275468 DOI: 10.1007/978-3-319-67577-0_8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Myeloid derived suppressor cells (MDSC) represent only a minor fraction of circulating blood cells but play an important role in tumor formation and progression. They are a heterogeneous group of cells that influence the tumor microenvironment by depletion of amino acids, oxidative stress, decreased trafficking of antitumor effector cells, and increased regulatory T and regulatory dendritic cell responses. Investigational treatment strategies targeting MDSCs have attempted to inhibit MDSC development and expansion (stem cell factor blockade, modulate of cell signaling, and target MDSC migration and recruitment), inhibit MDSC function (nitric oxide inhibition and reactive oxygen and nitrogen species inhibition), differentiate MDSCs into more mature cells (Vitamins A and D, all-trans retinoic acid, interleukin-2, toll-like receptor 9 inhibitors, taxanes, beta-glucan particles, tumor-derived exosome inhibition, and very small size proteoliposomes), and destroy MDSCs (cytotoxic agents, ephrin A2 degradation, anti-interleukin 13, and histamine blockers). To date, there are no Food and Drug Administration approved therapies selectively targeting MDSCs, but such therapies are likely to be implemented in the future, due to the key role of MDSCs in antitumor immunity.
Collapse
Affiliation(s)
- Waseem Anani
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Pérez Sánchez L, Morera Díaz Y, Bequet-Romero M, Ramses Hernández G, Rodríguez Y, Castro Velazco J, Puente Pérez P, Ayala Avila M, Gavilondo JV. Experimental studies of a vaccine formulation of recombinant human VEGF antigen with aluminum phosphate. Hum Vaccin Immunother 2016; 11:2030-7. [PMID: 25891359 DOI: 10.1080/21645515.2015.1029213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
CIGB-247 is a cancer vaccine that is a formulation of a recombinant protein antigen representative of the human vascular endothelial growth factor (VEGF) with a bacterially-derived adjuvant (VSSP). The vaccine has shown an excellent safety profile in mice, rats, rabbits, not-human primates and in recent clinical trials in cancer patients. Response to the vaccine is characterized by specific antibody titers that neutralize VEGF/VEGFR2 binding and a cytotoxic tumor-specific response. To expand our present anti-VEGF active immunotherapy strategies, we have now studied in mice and non-human primates the effects of vaccination with a formulation of our recombinant VEGF antigen and aluminum phosphate adjuvant (hereafter denominated CIGB-247-A). Administered bi-weekly, CIGB-247-A produces high titers of anti-VEGF IgG blocking antibodies in 2 mice strains. Particularly in BALB/c, the treatment impaired subcutaneous F3II mammary tumor growth and reduced the number of spontaneous lung macro metastases, increasing animals' survival. Spleen cells from specifically immunized mice directly killed F3II tumor cells in vitro. CIGB-247-A also showed to be immunogenic in non-human primates, which developed anti-VEGF blocking antibodies and the ability for specific direct cell cytotoxic responses, all without impairing the healing of deep skin wounds or other side effect. Our results support consideration of aluminum phosphate as a suitable adjuvant for the development of new vaccine formulations using VEGF as antigen.
Collapse
Key Words
- ANOVA, Analysis of Variance
- Aluminum phosphate
- CFSE, Carboxyfluorescein succinimidyl ester
- CTL, Cytotoxic T lymphocyte
- ELISA, Enzyme-linked immune-sorbent assay
- FACS, Fluorescence-activated cell sorting
- GST, Glutathione S-transferase
- HPLC, High-performance liquid chromatography
- KDR, kinase domain receptor
- Ni-NTA, nickel-nitrilotriacetic acid
- PBMC, Peripheral blood mononuclear cells
- VEGF
- VEGF, vascular endothelial growth factor
- VEGFR2, vascular endothelial growth factor receptor 2
- VSSP, very small sized proteoliposomes
- adjuvant
- antibodies
- cancer therapeutic vaccine
Collapse
Affiliation(s)
- Lincidio Pérez Sánchez
- a Cancer Immunotherapy Laboratory; Department of Pharmaceuticals; Center for Genetic Engineering and Biotechnology (CIGB) ; Playa Cubanacan , Havana , Cuba
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Fernández A, Oliver L, Alvarez R, Fernández LE, Mesa C. GM3-containing nanoparticles in immunosuppressed hosts: Effect on myeloid-derived suppressor cells. World J Immunol 2014; 4:98-106. [DOI: 10.5411/wji.v4.i2.98] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/27/2014] [Accepted: 06/27/2014] [Indexed: 02/05/2023] Open
Abstract
Cancer vaccines to date have not broadly achieved a significant impact on the overall survival of patients. The negative effect on the immune system of the tumor itself and conventional anti-tumor treatments such as chemotherapy is, undoubtedly, a key reason for these disappointing results. Myeloid-derived suppressor cells (MDSCs) are considered a central node of the immunosuppressive network associated with tumors. These cells inhibit the effector function of natural killer and CD8+ T cells, expand regulatory T cells and can differentiate into tumor-associated macrophages within the tumor microenvironment. Thus, overcoming the suppressive effects of MDSCs is likely to be critical for cancer immunotherapy to generate effective anti-tumor immune responses. However, the capacity of cancer vaccines and particularly their adjuvants to overcome this inhibitory population has not been well characterized. Very small size proteoliposomes (VSSP) is a nanoparticulated adjuvant specifically designed to be formulated with vaccines used in the treatment of immunocompromised patients. This adjuvant contains immunostimulatory bacterial signals together with GM3 ganglioside. VSSP promotes dendritic cell maturation, antigen cross-presentation to CD8+ T cells, Th1 polarization, and enhances CD8+ T cell response in tumor-free mice. Currently, four cancer vaccines using VSSP as the adjuvant are in Phase I and II clinical trials. In this review, we summarize our work characterizing the unique ability of VSSP to stimulate antigen-specific CD8+ T cell responses in two immunocompromised scenarios; in tumor-bearing mice and during chemotherapy-induced leukopenia. Particular emphasis has been placed on the interaction of these nanoparticles with MDSCs, as well as comparison with other cancer vaccine adjuvants currently in preclinical or clinical studies.
Collapse
|
22
|
Fernández A, Oliver L, Alvarez R, Hernández A, Raymond J, Fernández LE, Mesa C. Very small size proteoliposomes abrogate cross-presentation of tumor antigens by myeloid-derived suppressor cells and induce their differentiation to dendritic cells. J Immunother Cancer 2014; 2:5. [PMID: 24829762 PMCID: PMC4019907 DOI: 10.1186/2051-1426-2-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 03/04/2014] [Indexed: 12/22/2022] Open
Abstract
Background Myeloid-derived suppressor cells (MDSCs) are among the major obstacles that adjuvants for cancer vaccines have to overcome. These cells cross-present tumor-associated antigens (TAA) to naive T lymphocytes with a tolerogenic outcome. Very Small Size Proteoliposomes (VSSP) is used as adjuvant by four therapeutic cancer vaccines currently in Phase I and II clinical trials. We previously found that VSSP reduces the suppressive function of MDSCs, then activating antigen-specific CTL responses in tumor-bearing (TB) mice, with the consequent reduction of tumor growth. However the mechanistic explanation for the immunomodulatory effect of this adjuvant in TB hosts has not been addressed before. Methods TB mice were inoculated with VSSP and MDSCs isolated and characterized by their expression of Arg1 and Nos2 genes by RT-PCR. The effect of VSSP on antigen cross-presentation by MDSCs, regulatory T cells (Tregs) expansion and MDSCs differentiation towards dendritic cells (DCs) was analyzed by FACS. Student’s t test or ANOVA and Tukey’s tests were used for statistical analyses. Results After inoculating VSSP into TB mice, a significant reduction of Arg1 and Nos2 gene expression was observed in recovered MDSCs. Concurrently the ability of these cells to induce down-regulation of CD3ζ chain on T cells was lost. Likewise in mice inoculated with the adjuvant lower percentages of Tregs were detected. In vitro, VSSP treatment was enough to differentiate MDSCs into phenotypically mature DCs, eliminating the former suppressive effect. Noteworthy, in vivo administration of VSSP to OVA-expressing (EG.7) TB mice abrogated this model antigen cross-presentation by splenic MDSCs. Similar results were obtained even when OVA antigen was administered into these TB mice formulated in VSSP. On the contrary, immunization with the same protein in polyI:C did not change the percentage of MDSCs expressing SIINFEKL/H-2Kb complexes, whereas a concomitant injection of VSSP aborted the limitations of polyI:C in this setting. Conclusions Altogether, these results indicate that VSSP has the peculiar capacity of inhibiting TAA cross-presentation and certain suppressive mechanisms on MDSCs which in turn, combined with the ability to induce differentiation of these cells into antigen-presenting cells (APCs), sustains this adjuvant as an ideal immunomodulator for cancer immunotherapy.
Collapse
Affiliation(s)
- Audry Fernández
- Immunobiology Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| | - Liliana Oliver
- Immunobiology Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| | - Rydell Alvarez
- Immunobiology Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| | - Arletty Hernández
- Immunobiology Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| | - Judith Raymond
- Systems Biology Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| | - Luis E Fernández
- Innovation Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| | - Circe Mesa
- Immunobiology Division, Center of Molecular Immunology, 216 St and 15th Ave., Atabey, Playa, P.O. Box 16040, Havana 11600, Cuba
| |
Collapse
|
23
|
Specific active immunotherapy with a VEGF vaccine in patients with advanced solid tumors. results of the CENTAURO antigen dose escalation phase I clinical trial. Vaccine 2014; 32:2241-50. [PMID: 24530151 DOI: 10.1016/j.vaccine.2013.11.102] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/28/2013] [Accepted: 11/27/2013] [Indexed: 12/30/2022]
Abstract
UNLABELLED CIGB-247 is a novel cancer therapeutic vaccine that uses a human VEGF variant molecule as antigen, in combination with a bacterial adjuvant. In mice, CIGB-247 has anti-tumor and anti-metastatic effects. The vaccine induces anti-VEGF blocking antibodies and a cellular response targeting tumor cells producing VEGF, and has proven to be safe in mice, rats, rabbits and non-human primates. Herein we report the results of a Phase I clinical trial (code name CENTAURO) where safety, tolerance, and immunogenicity of CIGB-247 were studied in 30 patients with advanced solid tumors, at three antigen dose levels. Individuals were subcutaneously immunized for 8 consecutive weeks with 50, 100 or 400 μg of antigen, and re-immunized on week twelve. On week sixteen, evaluations of safety, tolerance, clinical status, and immunogenicity (seroconversion for anti-VEGF IgG, serum VEGF/KDR-Fc blocking ability, and gamma-IFN ELISPOT with blood cells stimulated in vitro with mutated VEGF) were done. Surviving patients were eligible for off-trial additional 4-week re-immunizations with 400 μg of antigen. Immunogenicity and clinical status were again studied on weeks 25 and 49. Vaccination was shown to be safe at the three dose levels, with only grade 1-2 adverse events. CIGB-247 was immunogenic and higher numbers of individuals positive to the three immune response tests were seen with increasing antigen dose. Off-protocol long-term vaccination produced no additional adverse events or negative changes in immunogenicity. Eleven patients are still alive, with overall survivals ranging from 20 to 24 months. Twelve of the thirty patients exhibited objective clinical benefits, and two individuals have complete responses. Most patients with higher survivals are positive in the three immune response tests. In summary, this is the first clinical testing report of a cancer therapeutic vaccine based on a human VEGF related molecule as antigen. The CIGB-247 vaccine is safe, immunogenic, and merits further clinical development. REGISTRATION NUMBER AND NAME OF TRIAL REGISTRY RPCEC00000102. Cuban Public Clinical Trial Registry (WHO accepted Primary Registry). Available from: http://registroclinico.sld.cu/.
Collapse
|
24
|
Jones HE, Copland A, Hamstra HJ, Cohen J, Brown J, Klein N, van der Ley P, Dixon G. LOS oligosaccharide modification enhances dendritic cell responses to meningococcal native outer membrane vesicles expressing a non-toxic lipid A. Cell Microbiol 2013; 16:519-34. [PMID: 24152255 PMCID: PMC4204155 DOI: 10.1111/cmi.12231] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/16/2013] [Indexed: 01/01/2023]
Abstract
Outer membrane vesicles (OMV) are released by many bacteria, and contain immunogenic antigens in addition to harmful inflammatory factors, like lipopolysaccharides. Chemically detoxified OMV have been used in vaccines against Neisseria meningitidis (Nm); however, little is known about their interaction with antigen presenting cells. In this study, we investigated the interaction of Nm OMV with human dendritic cells (DC) to gain further understanding of their biological activity. We engineered a novel serogroup B Nm that is unencapsulated (siaD), expresses pentacylated lipid A (lpxL1), hence conferring reduced toxicity, and expresses an lgtB oligosaccharide structure designed to target OMV to DC via DC-SIGN. We show that the lgtB moiety is critical for internalization of NOMV by DC. Furthermore, the lgtB moiety significantly enhances DC maturation, IL-10 and IL-23 production in the presence of a pentacylated lipid A. While different DC phenotypes were observed for each NOMV, this had little effect on Th1 and Th2 cell differentiation; however, lgtBsignificantly increased Th17 cell expansion in the presence of pentacylated lipid A. We believe that lpxL1/lgtB NOMV should be considered further as a vaccine vector, particularly considering the importance of lgtB in antigen uptake and further human studies on antigen-specific responses should be considered.
Collapse
Affiliation(s)
- Hannah E Jones
- Infectious Diseases Microbiology Unit, Institute of Child Health, UCL, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Non-clinical immuno-toxicological evaluation of HER1 cancer vaccine in non-human primates: a 12-month study. Vaccine 2012; 31:89-95. [DOI: 10.1016/j.vaccine.2012.10.098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 10/25/2012] [Accepted: 10/28/2012] [Indexed: 11/22/2022]
|
26
|
Mancebo A, Casacó A, Sánchez B, González B, Gómez D, León A, Bada A, Arteaga M, González Y, González C, Pupo M, Fuentes D. Repeated dose (14days) rat intramuscular toxicology study of Her1 vaccine. Regul Toxicol Pharmacol 2012; 64:425-34. [DOI: 10.1016/j.yrtph.2012.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 10/01/2012] [Accepted: 10/04/2012] [Indexed: 01/27/2023]
|
27
|
Oliver L, Fernández A, Raymond J, López-Requena A, Fernández LE, Mesa C. Very small size proteoliposomes derived from Neisseria meningitidis: an effective adjuvant for antigen-specific cytotoxic T lymphocyte response stimulation under leukopenic conditions. Vaccine 2012; 30:2963-72. [PMID: 22391399 DOI: 10.1016/j.vaccine.2012.02.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 02/16/2012] [Accepted: 02/19/2012] [Indexed: 10/28/2022]
Abstract
Leukopenia is a severe condition resulting from both pathological processes and some treatments, like chemotherapy in cancer patients. However, the activation of the patient immune system is required for the success of immunotherapeutic strategies, as cancer vaccines. In this regard, leukopenia constitutes a major hurdle to overcome, mainly due to the impairment of cytotoxic T lymphocyte (CTL) responses. Adjuvants are basic components of vaccine formulations, which might be useful to stimulate immunity under this immunosuppressed condition. To this aim, we tested the capacity of a novel nanoparticulated complex, very small size proteoliposomes (VSSP), to promote CTL even in a leukopenic scenario. Noteworthy, we observed that a VSSP-based OVA vaccine induced a normal antigen-specific CTL response in mice rendered leukopenia by the administration of high doses of the chemotherapeutic agent cyclophosphamide (CY), while under the same conditions the OVA antigen formulated in the TLR-3 agonist polyinosinic-polycytidylic acid (P(I:C)) was ineffective. Moreover, an appropriate combination of VSSP with the P(I:C) vaccine was able to restore the CD8(+) T cell effector function in leukopenic mice. VSSP induced not only a faster repopulation of immune cells in CY-receiving animals, but also enhanced the recovery of memory T lymphocytes and myeloid dendritic cells (DCs) while simultaneously abrogated the immunosuppressive capacity of myeloid-derived suppressor cells (MDSCs). Our results suggest that VSSP could be a particularly suitable immunomodulator to be used in CTL-promoting active immunotherapy strategies operating in severe immune compromised scenarios.
Collapse
Affiliation(s)
- Liliana Oliver
- Center of Molecular Immunology, Atabey, Playa, Havana 11600, Cuba
| | | | | | | | | | | |
Collapse
|
28
|
CIGB-247: a VEGF-based therapeutic vaccine that reduces experimental and spontaneous lung metastasis of C57Bl/6 and BALB/c mouse tumors. Vaccine 2012; 30:1790-9. [PMID: 22240345 DOI: 10.1016/j.vaccine.2012.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 12/29/2011] [Accepted: 01/02/2012] [Indexed: 12/22/2022]
Abstract
CIGB-247 is a novel cancer therapeutic vaccine that uses a mutated form of human VEGF as antigen. Being metastatic disease the most dramatic factor of tumor biology affecting patient survival and cure, preclinical evaluation of the impact of CIGB-247 vaccination on experimental metastasis mouse models is highly relevant, and constitutes the focus of this work. CIGB-247 was administered in a weekly schedule known to effectively reduce primary tumor growth. The vaccine was tested in experimental and spontaneous metastasis models of colon (CT26), lung (3LL-D122) and breast (F3II) carcinomas growing in C57Bl/6 or BALB/c mice. Primary tumor growth parameters, metastatic counts, and/or animal survival were recorded. Histology and specific humoral and cellular responses to the vaccine were evaluated. As compared to control groups, CIGB-247 vaccination significantly reduced the number and size of metastatic tumor foci in lungs after intravenous inoculation of CT26 and 3LL-D122 tumor cells. Spontaneous lung dissemination from 3LL-D122 and F3II breast tumor cells implanted in the footpad, or subcutaneously, was also reduced by immunization with CIGB-247. The vaccine elicited in both mouse strains antibodies specific for human and murine VEGF that effectively blocked the interaction of VEGF with VEGF receptor 2. Differing from other experimental reports that describe the use of VEGF for active tumor immunotherapy, CIGB-247 elicited a specific cellular response, measured both by a DTH increment and the induction of spleen cells cytotoxic to syngeneic tumor cells producing murine VEGF. In summary our results reinforce the potential of CIGB-247 vaccination to reduce both tumor growth and the number and size of tumor metastasis in lungs, the latter both after direct inoculations of cells in the blood stream, or as part of primary tumor progression in immunocompetent mice.
Collapse
|
29
|
Rodriguez L, Tirado Y, Reyes F, Puig A, Kadir R, Borrero R, Fernandez S, Reyes G, Alvarez N, Garcia MA, Sarmiento ME, Norazmi MN, Perez Quinoy JL, Acosta A. Proteoliposomes from Mycobacterium smegmatis induce immune cross-reactivity against Mycobacterium tuberculosis antigens in mice. Vaccine 2011; 29:6236-41. [PMID: 21736914 DOI: 10.1016/j.vaccine.2011.06.077] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/18/2011] [Accepted: 06/21/2011] [Indexed: 02/06/2023]
Abstract
Proteoliposomes (PL) obtained from Mycobacterium smegmatis (Ms) were evaluated for their capacity to elicit cross-reactive responses against Mycobacterium tuberculosis (Mtb) antigens in BALB/c mice. Animals immunized with PL adjuvanted with alum (PL-AL) or Freund's Incomplete Adjuvant (PL-IFA) showed significant IgG responses against the PL as well as total Ms lipids. Both groups of animals also showed significant IgG responses against BCG, but only animals immunized with PL-AL produced significant IgG responses against soluble cell wall proteins (SCWP) or whole cell lysate (WCL) of Mtb. Significant DTH responses against WCL were observed in both groups of animals after 24 h, but only PL-AL-immunized mice showed significant DTH responses after 48 h and 72 h. PL-Ms are capable of eliciting cross-reactive humoral and cellular responses against Mtb antigens and thus may be a potential vaccine strategy against tuberculosis.
Collapse
|
30
|
Solares AM, Baladron I, Ramos T, Valenzuela C, Borbon Z, Fanjull S, Gonzalez L, Castillo D, Esmir J, Granadillo M, Batte A, Cintado A, Ale M, Fernandez de Cossio ME, Ferrer A, Torrens I, Lopez-Saura P. Safety and Immunogenicity of a Human Papillomavirus Peptide Vaccine (CIGB-228) in Women with High-Grade Cervical Intraepithelial Neoplasia: First-in-Human, Proof-of-Concept Trial. ISRN OBSTETRICS AND GYNECOLOGY 2011; 2011:292951. [PMID: 21748025 PMCID: PMC3118643 DOI: 10.5402/2011/292951] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 01/26/2011] [Indexed: 12/22/2022]
Abstract
Objective. CIGB-228 is a novel therapeutic vaccine consisting of HLA-restricted HPV16 E7 epitope adjuvated with VSSP. This trial was designed to evaluate the toxicity, safety, immunogenicity, HPV clearance, and lesion regression. Methods. Seven women were entered. All were HLA-A2 positive, had biopsy-proven high-grade CIN, histologically positive for HPV16, and beared persistent postbiopsy lesions visible by digital colposcopy. HLA-A2 women with biopsy-proven high-grade CIN, HPV16-positive, and beared persistent postbiopsy lesions visible by digital colposcopy were vaccinated. One weekly injections of CIGB-228 vaccine was given for four weeks. Then, loop electrosurgical excision procedure (LEEP) of the transformation zone was performed. Study subjects were followed for 1 year after LEEP. Results. No toxicity beyond grade 1 was observed during and after the four vaccinations. Five of seven women had complete and partial regression. Cellular immune response was seen in all patients. HPV was cleared in three of the patients with complete response.
Conclusion. CIGB-228 vaccination was well tolerated and capable to induce IFNγ-associated T-cell response in women with high-grade CIN. In several patients, lesion regression and HPV clearance were observed.
Collapse
Affiliation(s)
- Ana M Solares
- Gyneco-obstetric Hospital Ramon Gonzalez Coro, Havana 10400, Cuba
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fernández A, Mesa C, Marigo I, Dolcetti L, Clavell M, Oliver L, Fernández LE, Bronte V. Inhibition of tumor-induced myeloid-derived suppressor cell function by a nanoparticulated adjuvant. THE JOURNAL OF IMMUNOLOGY 2010; 186:264-74. [PMID: 21135171 DOI: 10.4049/jimmunol.1001465] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The interaction between cancer vaccine adjuvants and myeloid-derived suppressor cells (MDSCs) is currently poorly understood. Very small size proteoliposomes (VSSP) are a nanoparticulated adjuvant under investigation in clinical trials in patients with renal carcinoma, breast cancer, prostate cancer, and cervical intraepithelial neoplasia grade III. We found that VSSP adjuvant induced a significant splenomegaly due to accumulation of CD11b(+)Gr-1(+) cells. However, VSSP-derived MDSCs showed a reduced capacity to suppress both allogeneic and Ag-specific CTL response compared with that of tumor-induced MDSCs. Moreover, splenic MDSCs isolated from tumor-bearing mice treated with VSSP were phenotypically more similar to those isolated from VSSP-treated tumor-free mice and much less suppressive than tumor-induced MDSCs, both in vitro and in vivo. Furthermore, different from dendritic cell vaccination, inoculation of VSSP-based vaccine in EG.7-OVA tumor-bearing mice was sufficient to avoid tumor-induced tolerance and stimulate an immune response against OVA Ag, similar to that observed in tumor-free mice. This effect correlated with an accelerated differentiation of MDSCs into mature APCs that was promoted by VSSP. VSSP used as a cancer vaccine adjuvant might thus improve antitumor efficacy not only by stimulating a potent immune response against tumor Ags but also by reducing tumor-induced immunosuppression.
Collapse
|
32
|
Morera Y, Bequet-Romero M, Ayala M, Velazco JC, Pérez PP, Alba JS, Ancizar J, Rodríguez M, Cosme K, Gavilondo JV. Immunogenicity and some safety features of a VEGF-based cancer therapeutic vaccine in rats, rabbits and non-human primates. Vaccine 2010; 28:3453-61. [PMID: 20197134 DOI: 10.1016/j.vaccine.2010.02.069] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 02/12/2010] [Accepted: 02/15/2010] [Indexed: 11/16/2022]
Abstract
We have developed a cancer vaccine candidate (hereafter denominated CIGB-247), based on recombinant modified human vascular endothelial growth factor (VEGF) as antigen, and the adjuvant VSSP (very small sized proteoliposomes of Neisseria meningitidis outer membrane). In mice, previous work of our group had shown that vaccination with CIGB-247 extended tumor-take time, slowed tumor growth, and increased animal survival. Immunization elicited anti-human and murine VEGF-neutralizing antibodies, and spleen cells of vaccinated mice are cytotoxic in vitro to tumor cells that produce VEGF. We have now tested the immunogenicity of CIGB-247 in Wistar rats, New Zealand White rabbits and the non-human primate Chlorocebus aethiops sabaeus. Using weekly, biweekly and biweekly plus montanide immunization schemes, all three species develop antigen-specific IgG antibodies that can block the interaction of VEGF and VEGF receptor 2 in an ELISA assay. Antibody titers decline after vaccination stops, but can be boosted with new immunizations. In monkeys, DTH and direct cell cytotoxicity experiments suggest that specific T-cell responses are elicited by vaccination. Immunization with CIGB-247 had no effect on normal behavior, hematology, blood biochemistry and histology of critical organs, in the tested animals. Skin deep wound healing was not affected in vaccinated rats and monkeys.
Collapse
Affiliation(s)
- Yanelys Morera
- Recombinant Antibodies Laboratory, Dept. of Pharmaceuticals, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Cubanacán, Playa Havana 10600, Cuba.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yang D, Zhao Y, Guo H, Li Y, Tewary P, Xing G, Hou W, Oppenheim JJ, Zhang N. [Gd@C(82)(OH)(22)](n) nanoparticles induce dendritic cell maturation and activate Th1 immune responses. ACS NANO 2010; 4:1178-86. [PMID: 20121217 PMCID: PMC2835518 DOI: 10.1021/nn901478z] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Dendritic cells play a pivotal role in host immune defense, such as elimination of foreign pathogen and inhibition of tumorigenesis. In this paper, we report that [Gd@C(82)(OH)(22)](n) could induce phenotypic maturation of dendritic cells by stimulating DC production of cytokines including IL-12p70, upregulating DC co-stimulatory (CD80, CD83, and CD86) and MHC (HLA-A,B,C and HLA-DR) molecules, and switching DCs from a CCL5-responsive to a CCL19-responsive phenotype. We found that [Gd@C(82)(OH)(22)](n) can induce dendritic cells to become functionally mature as illustrated by their capacity to activate allogeneic T cells. Mice immunized with ovalbumin in the presence of [Gd@C(82)(OH)(22)](n) exhibit enhanced ovalbumin-specific Th1-polarized immune response as evidenced by the predominantly increased production of IFNgamma, IL-1beta, and IL-2. The [Gd@C(82)(OH)(22)](n) nanoparticle is a potent activator of dendritic cells and Th1 immune responses. These new findings also provide a rational understanding of the potent anticancer activities of [Gd@C(82)(OH)(22)](n) nanoparticles reported previously.
Collapse
Affiliation(s)
- De Yang
- Tianjin Medical University, Research Center of Basic Medic Sciences, Cancer Institute and Hospital, Key Laboratory of Breast Cancer Research (Ministry of Education), Tianjin 300060, China
- Basic Research Program, SAIC-Frederick, Inc.; Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China, and National Center for Nanoscience and Technology of China
| | - Hua Guo
- Tianjin Medical University, Research Center of Basic Medic Sciences, Cancer Institute and Hospital, Key Laboratory of Breast Cancer Research (Ministry of Education), Tianjin 300060, China
| | - Yana Li
- Basic Research Program, SAIC-Frederick, Inc.; Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Poonam Tewary
- Basic Research Program, SAIC-Frederick, Inc.; Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China, and National Center for Nanoscience and Technology of China
- Correspondence should be addressed to: Ning Zhang, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China. (Tel: 086-13502179648; ). Gengmei Xing, Laboratory for Bio-Environmental Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, The Chinese Academy of Sciences, Beijing 100049, China, and National Center for Nanoscience and Technology
| | - Wei Hou
- Tianjin Medical University, Research Center of Basic Medic Sciences, Cancer Institute and Hospital, Key Laboratory of Breast Cancer Research (Ministry of Education), Tianjin 300060, China
| | - Joost J. Oppenheim
- Basic Research Program, SAIC-Frederick, Inc.; Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research; National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Ning Zhang
- Tianjin Medical University, Research Center of Basic Medic Sciences, Cancer Institute and Hospital, Key Laboratory of Breast Cancer Research (Ministry of Education), Tianjin 300060, China
- Correspondence should be addressed to: Ning Zhang, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China. (Tel: 086-13502179648; ). Gengmei Xing, Laboratory for Bio-Environmental Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, The Chinese Academy of Sciences, Beijing 100049, China, and National Center for Nanoscience and Technology
| |
Collapse
|
34
|
Virus-Like Particles as vaccine antigens and adjuvants: application to chronic disease, cancer immunotherapy and infectious disease preventive strategies. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.provac.2010.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
35
|
Mazorra Z, Mesa C, Fernández A, Fernández LE. Immunization with a GM3 ganglioside nanoparticulated vaccine confers an effector CD8(+) T cells-mediated protection against melanoma B16 challenge. Cancer Immunol Immunother 2008; 57:1771-80. [PMID: 18351335 PMCID: PMC11029898 DOI: 10.1007/s00262-008-0503-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Accepted: 03/03/2008] [Indexed: 01/11/2023]
Abstract
Preventive immunotherapy is an attractive strategy for patients at a high risk of having cancer. The success of prophylactic cancer vaccines would depend on the selection of target antigens that are essential for tumour growth and progression. The overexpression of GM3 ganglioside in murine and human melanomas and its important role in tumour progression makes this self antigen a potential target for preventive immunotherapy of this neoplasm. We have previously shown that preventive administration of a GM3-based vaccine to C57BL/6 mice elicited the rejection of the GM3 positive-B16 melanoma cells in most of the animals. Despite the crucial role of cellular immune response in tumour protection, the involvement of T cells in anti-tumour immunity of ganglioside vaccines is not described. Here, we examined the mechanisms by which this immunogen confers tumour protection. We have found that induction of anti-GM3 IgG antibodies correlated with tumour protection. Surprisingly, CD8(+) T cells, but not NK1.1(+) cells, are required in the effector phase of the antitumour immune response. The depletion of CD4(+) T cells during immunization phase did not affect the anti-tumour activity. In addition, T cells from surviving-immunized animals secreted IFNgamma when were co-cultured with IFNalpha-treated B16 melanoma cells or DCs pulsed with melanoma extract. Paradoxically, in spite of the glycolipidic nature of this antigen, these findings demonstrate the direct involvement of the cellular immune response in the anti-tumour protection induced by a ganglioside-based vaccine.
Collapse
Affiliation(s)
- Zaima Mazorra
- Department of Vaccines, Center of Molecular Immunology, Havana, Cuba.
| | | | | | | |
Collapse
|
36
|
Morera Y, Bequet-Romero M, Ayala M, Lamdán H, Agger EM, Andersen P, Gavilondo JV. Anti-tumoral effect of active immunotherapy in C57BL/6 mice using a recombinant human VEGF protein as antigen and three chemically unrelated adjuvants. Angiogenesis 2008; 11:381-93. [PMID: 19034678 DOI: 10.1007/s10456-008-9121-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 10/01/2008] [Indexed: 11/30/2022]
Abstract
Following the clinical success of Bevacizumab, a humanized monoclonal antibody that affects the interaction between vascular endothelial growth factor (VEGF) and its receptors, blocking tumor-induced angiogenesis has become one of the most important targets for the development of new cancer therapeutic drugs and procedures. Among the latter, therapeutic vaccination using VEGF as antigen presents itself as very attractive, with the potential of generating not only a growth factor blocking antibody response but also a cellular response against tumor cells and stromal elements, which appear to be a major source of tumor VEGF. In this paper, we report the development of a protein vaccine candidate, based on a human modified VEGF antigen that is expressed at high levels in E. coli. With respect to controls, immunization experiments in C57BL/6 mice using weekly doses of this antigen and three adjuvants of different chemical natures show that time for tumor development after subcutaneous injection of Melanoma B16-F10 cells increases, tumors that develop grow slower, and overall animal survival is higher. Immunization also prevents tumor development in some mice, making them resistant to second tumor challenges. Vaccination of mice with the human modified VEGF recombinant antigen produces antibodies against the human antigen and the homologous mouse VEGF molecule. We also show that sera from immunized mice block human VEGF-induced HUVEC proliferation. Finally, a possible contribution of T cell cytotoxicity to the overall anti-tumor effect is suggested from the results of vaccination experiments where CD8+ lymphocytes were impaired using neutralizing rat antibodies.
Collapse
Affiliation(s)
- Yanelys Morera
- Recombinant Antibody Group, Cancer Research Department, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Ave 31 and 190 Cubanacán, 10600, Playa, Havana, Cuba
| | | | | | | | | | | | | |
Collapse
|
37
|
Ramírez BS, Alpízar YA, Fernández DRH, Hidalgo GG, Capote AR, Rodríguez RP, Fernández LE. Anti-EGFR activation, anti-proliferative and pro-apoptotic effects of polyclonal antibodies induced by EGFR-based cancer vaccine. Vaccine 2008; 26:4918-26. [DOI: 10.1016/j.vaccine.2008.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 07/03/2008] [Accepted: 07/10/2008] [Indexed: 11/30/2022]
|
38
|
|
39
|
Use of proteoliposome as a vaccine against Trypanosoma cruzi in mice. Chem Phys Lipids 2008; 152:86-94. [PMID: 18262496 DOI: 10.1016/j.chemphyslip.2007.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 12/20/2007] [Accepted: 12/27/2007] [Indexed: 11/23/2022]
Abstract
We have generated proteoliposomes carrying proteins of Trypanosoma cruzi for use as immunogens in BALB/c mice. T. cruzi trypomastigote and amastigote forms were sonicated and mixed with SDS, with 94% recovery of soluble proteins. To prepare proteoliposomes, we have used a protocol in which dipalmitoylphosphatidylcholine, dipalmitoyl-phosphatidylserine and cholesterol were incubated with the parasite proteins. BALB/c mice immunized with 20microg were able to generate antibodies which, in Western blotting, reacted with the proteins of T. cruzi. We further investigated the ability of peritoneal cells from immunized mice to arrest the intracellular replication of trypomastigotes, in vitro. After 72h of culture, the number of intracellular parasites in immunized macrophages decreased significantly, as compared to controls. Despite the fact that exposure of mice to T. cruzi proteins incorporated into proteoliposomes generate antibodies and activate macrophages, the immunized mice were not protected against T. cruzi intraperitoneal challenge.
Collapse
|
40
|
Pajón R, Niebla O, Yero D, Pérez O, Cabrera O, Findlow J, Balmer P, Borrow R. On the neisserial vaccine quest: Neisseria Vaccines 2007. Expert Rev Anti Infect Ther 2007; 5:545-50. [PMID: 17678419 DOI: 10.1586/14787210.5.4.545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Rolando Pajón
- Meningococcal Research Department, Center for Genetic Engineering and Biotechnology, Cubanacán Havana, Cuba.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Venier C, Guthmann MD, Fernández LE, Fainboim L. Innate-immunity cytokines induced by very small size proteoliposomes, a Neisseria-derived immunological adjuvant. Clin Exp Immunol 2007; 147:379-88. [PMID: 17223981 PMCID: PMC1810462 DOI: 10.1111/j.1365-2249.2006.03297.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neisserial outer membrane proteins have been combined with monosialoganglioside GM3 to form very small size proteoliposomes (VSSP), a nanoparticulated formulation used as a cancer vaccine for the treatment of cancer patients with GM3-positive tumours. VSSP were shown to elicit anti-GM3 and anti-tumour immune responses. VSSP have also been shown to be an efficient adjuvant for tumour-cell and peptide-antigen vaccines in mice. In vitro studies showed that VSSP promote maturation of both murine and human dendritic cells, suggesting that VSSP could be used as efficient adjuvants. In order to study further the capacity of VSSP to elicit innate immune responses, human peripheral blood mononuclear cells and monocytes derived thereof were assessed for in vitro secretion of interleukin (IL)-10, IL-6, IL-12 and interferon (IFN)-gamma. VSSP most prominently induced the secretion of IL-6. IL-10 was secreted at a lower level. IL-12 p40 (but no p70) was also detected. IFN-gamma response was observed in 56% of the tested samples. Cytokine secretion was not related to lipopolysaccharide (LPS) content and involved Toll-like receptor 2 (TLR2)-mediated signal transduction. VSSP also induced DC maturation and a cytokine secretion pattern (high IL-6/low IL-10) which differs from that induced by LPS. The observed proinflammatory cytokine secretion pattern and the capacity of VSSP to drive DC maturation are examined in the light of the properties of other bacterial derivatives currently being user for immunotherapy purposes. Our results suggest that VSSP could be tested in clinical settings where T helper 1-type immune responses would be beneficial.
Collapse
Affiliation(s)
- C Venier
- Hospital de Clínicas José de San Martín, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
42
|
Abstract
Immunotherapy was formerly the treatment of allergic disease by repeated exposure to allergen. Other approaches to immune modulation have emerged using new knowledge of T cell function. The so-called hygiene hypothesis argues that alterations in our environment have resulted in a failure of the immune system to develop normally, such that excessive Th2 type responses to antigen exposures occur. These observations have prompted therapies designed to promote a shift from Th-2 to Th-1 responses. These therapies include bacterial vaccines and stimulation of the immune system with Toll like receptor ligands or bacterial nucleotide immunostimulatory sequences (CpG motifs). Traditional desensitization immunotherapy is being re-examined and anti-IgE therapy is also enjoying a measure of success.
Collapse
Affiliation(s)
- James G Martin
- Meakins Christie Laboratories, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
43
|
Ramírez BS, Pestana ES, Hidalgo GG, García TH, Rodríguez RP, Ullrich A, Férnandez LE. Active antimetastatic immunotherapy in Lewis lung carcinoma with self EGFR extracellular domain protein in VSSP adjuvant. Int J Cancer 2006; 119:2190-9. [PMID: 16841332 DOI: 10.1002/ijc.22085] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The epidermal growth factor receptor (EGFR) plays a central role in regulating neoplastic processes. The EGFR overexpression in many human epithelial tumors has been correlated with disease progression and bad prognosis. Passive EGFR-directed immunotherapy, but not active specific approaches, has already been introduced in medical oncology practice. Then we wonder if mice immunization with the extracellular domain of murine EGFR (mEGFR-ECD) in adjuvants can circumvent tolerance to self EGFR, by inducing an immune response with consequent antitumor effect. The present study demonstrated that despite mEGFR expression in thymus, strong DTH response was induced by inoculation of mice with the mEGFR-ECD. This self-immunization, using both CFA and very small sized proteoliposomes from Neisseria meningitidis (VSSP), promoted highly specific IgG titers, predominantly IgG2a and IgG2b. Sera from mice immunized with mEGFR-ECD/VSSP not only recognized EGFR+ tumor cell lines by FACS, but also inhibited their in vitro growth, even in the absence of complement. Noteworthy, vaccination of mice with mEGFR-ECD/VSSP stimulated a potent antimetastatic effect in the EGFR+ Lewis lung carcinoma model, while reproduction-associated side effects were absent. Curiously, mice immunized with the human EGFR-ECD (Her1-ECD) in VSSP though induced highly specific IgG antibodies with strong in vitro cytotoxic effect over EGFR+ human cell lines, showed low cross-reactivity with the mEGFR-ECD. These results further encouraged the development of the Her1-ECD/VSSP vaccine project for patients with EGFR+ tumors.
Collapse
|
44
|
Uli L, Castellanos-Serra L, Betancourt L, Domínguez F, Barberá R, Sotolongo F, Guillén G, Pajón Feyt R. Outer membrane vesicles of the VA-MENGOC-BC vaccine against serogroup B of Neisseria meningitidis: Analysis of protein components by two-dimensional gel electrophoresis and mass spectrometry. Proteomics 2006; 6:3389-99. [PMID: 16673438 DOI: 10.1002/pmic.200500502] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Neisseria meningitidis is a Gram-negative bacterium responsible for significant mortality worldwide. While effective polysaccharides-based vaccines exist against serogroups A, C, W135, and Y, no similar vaccine is suitable for children under 4 years against disease caused by serogroup B strains. Therefore, major vaccine efforts against this serogroup are based on outer membrane vesicles (OMVs), containing major outer membrane proteins. The OMV-based vaccine produced by the Finlay Institute in Cuba (VA-MENGOC-BC) contributed to the rapid decline of the epidemic in this Caribbean island. While the content of major proteins in this vaccine has been discussed, no detailed work of an outer membrane proteomic map of this, or any other, commercially available OMV-derived product has been published so far. Since OMVs exhibit a large bias toward a few major proteins and usually contain a high content of lipids, establishing the adequate conditions for high resolution, 2-DE of this kind of preparation was definitely a technical challenge. In this work, 2-DE and MS have been used to generate a proteomic map of this product, detailing the presence of 31 different proteins, and it allows the identification of new putative protective protein components it contains.
Collapse
Affiliation(s)
- Liliam Uli
- Finlay Institute, Serum and Vaccines Production Center, Habana, Cuba
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Mulet A, Garrido G, Alvarez A, Menéndez T, Böhmer FD, Pérez R, Fernández LE. The enlargement of the hormone immune deprivation concept to the blocking of TGFalpha-autocrine loop: EGFR signaling inhibition. Cancer Immunol Immunother 2006; 55:628-38. [PMID: 16328386 PMCID: PMC11030461 DOI: 10.1007/s00262-005-0030-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2004] [Accepted: 05/12/2005] [Indexed: 11/30/2022]
Abstract
Transforming growth factor alpha (TGFalpha) is a potent ligand of the epidermal growth factor receptor (EGFR). EGFR is frequently over-expressed in epithelial tumors and endogenous ligands, mostly TGFalpha, are frequently co-expressed with EGFR, potentially resulting in autocrine stimulation of tumor cell growth. Therefore, different therapeutic approaches aim for the inactivation of TGFalpha/EGF/EGFR signaling system, but no approach is based on TGFalpha as a target. The principal goal of this work was to assess the potential of an active specific immunotherapy approach to block the TGFalpha/EGFR autocrine loop. For the proof of the concept, a fusion protein between human TGFalpha (hTGFalpha) and P64k protein from Neisseria meningitidis was generated, and its immunogenicity characterized in a mouse model using different adjuvants. All immunogens were effective for the generation of specific humoral responses against hTGFalpha. The inmunodominant epitope of hTGFalpha when immunizing mice with the fusion protein involved the C-loop/C-terminal region. This region includes key residues for hTGFalpha binding to EGFR. The anti-hTGFalpha immune mice sera recognized the natural hTGFalpha precursor in A431 cells and hTGFalpha-transfected 3T3 fibroblasts as revealed by flow cytometry analysis and immunoblotting. They inhibited the binding of (125)I-TGFalpha to the EGFR, EGFR-autophosphorylation, and downstream activation of MAP kinases as well as proliferation of two EGFR-expressing human carcinoma cell lines. These data suggest that EGFR signaling activation by the hTGFalpha autocrine loop may be inhibited in vivo by induction of specifically blocking antibodies. The fusion protein reported in this paper could be a potential immunogen for the development of a new cancer vaccine.
Collapse
Affiliation(s)
- Aillette Mulet
- Center of Molecular Immunology, CIM, P.O.Box 16040, Havana 11600, Cuba.
| | | | | | | | | | | | | |
Collapse
|
46
|
Sardiñas G, Reddin K, Pajon R, Gorringe A. Outer membrane vesicles of Neisseria lactamica as a potential mucosal adjuvant. Vaccine 2006; 24:206-14. [PMID: 16115701 DOI: 10.1016/j.vaccine.2005.07.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Accepted: 07/25/2005] [Indexed: 11/24/2022]
Abstract
The muscosal delivery of vaccines has many advantages including ease of administration and the induction of a mucosal immune response at the natural site of infection for many pathogens. Mice were immunised with outer membrane vesicles (OMV) prepared from Neisseria lactamica or Neisseria meningitidis by subcutaneous (SC) or intranasal (IN) routes, or live cells of N. lactamica given IN or by SC injection. A systemic IgG and mucosal IgA response was demonstrated and N. lactamica OMV induced antibodies cross-reactive with N. meningitidis; however, a cross-reactive response following IN administration was only evident after three doses of vaccine. OMV from both organisms were also an effective intranasal adjuvant for a co-administered model antigen, hepatitis B surface antigen (HBsAg), inducing systemic IgG against HBsAg and IgA in lung and vaginal washes. IN administration of N. meningitidis OMV elicited serum antibodies that were bactericidal for meningococci and provided passive protection in an infant rat model of meningococcal bacteraemia. The antibody response to N. lactamica OMV given IN was only weakly bactericidal but still afforded passive protection. Thus, OMV from N. lactamica given IN elicit immune responses cross-reactive with N. meningitidis and act as an effective mucosal adjuvant.
Collapse
Affiliation(s)
- Gretel Sardiñas
- Center For Genetic Engineering and Biotechnology, Ave. 31e/158 y 190, Cubanacán, P.O. Box 6162, 10600 Habana, Cuba
| | | | | | | |
Collapse
|
47
|
Oliva JP, Valdés Z, Casacó A, Pimentel G, González J, Alvarez I, Osorio M, Velazco M, Figueroa M, Ortiz R, Escobar X, Orozco M, Cruz J, Franco S, Díaz M, Roque L, Carr A, Vázquez AM, Mateos C, Rubio MC, Pérez R, Fernández LE. Clinical evidences of GM3 (NeuGc) ganglioside expression in human breast cancer using the 14F7 monoclonal antibody labelled with (99m)Tc. Breast Cancer Res Treat 2005; 96:115-21. [PMID: 16322892 DOI: 10.1007/s10549-005-9064-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Accepted: 08/23/2005] [Indexed: 11/24/2022]
Abstract
The relevance of certain gangliosides in tumour growth and metastatic dissemination has been well documented, reasons for considering these molecules as potential targets for cancer immunotherapy and diagnosis. GM3(NeuGc) ganglioside is particularly interesting due to its restrictive expression in normal human tissues according to immunohistochemical studies, using either polyclonal or monoclonal antibodies. But both immunohistochemical and biochemical methods have strongly suggested its over-expression in human breast tumours. Nevertheless, the lack of a direct evidence of this antigenic display in human breast cancer has kept the subject controversial. For the first time, we described herein the "in vivo" detection of GM3(NeuGc) ganglioside in human breast primary tumours using a radioimmunoscintigraphic technique with 14F7, a highly specific anti-GM3(NeuGc) ganglioside monoclonal antibody, labelled with (99m)Tc. In an open, prospective Phase I/II clinical trial, including women diagnosed in stage II breast cancer, the 14F7 monoclonal antibody accumulation in tumours at doses of 0.3 (n=5), 1 (n=5) and 3 mg (n=4) was evaluated. Noteworthy, the immunoscintigraphic study showed antibody accumulation in 100% of patients' tumours for the 1 mg dose group. In turn, the radioimmunoconjugate injected at doses of 0.3 mg or 3 mg of the antibody, was uptaken by 60 and 33.3% of breast tumours, respectively. "In vivo" immune recognition of GM3(NeuGc) in breast tumours reinforces the value of this peculiar target for cancer immunotherapy.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/administration & dosage
- Breast Neoplasms/diagnostic imaging
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/diagnostic imaging
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Dose-Response Relationship, Drug
- Female
- G(M3) Ganglioside/metabolism
- Humans
- Infusions, Intravenous
- Middle Aged
- Neoplasm Staging
- Prospective Studies
- Radioimmunodetection
- Technetium/administration & dosage
Collapse
Affiliation(s)
- Juan P Oliva
- Department of Nuclear Medicine, National Institute of Oncology and Radiobiology, Plaza, Havana, Cuba
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
An adjuvant is defined as a product that increases or modulates the immune response against an antigen (Ag). Based on this general definition many authors have postulated that the ideal adjuvant should increase the potency of the immune response, while being non-toxic and safe. Although dozens of different adjuvants have been shown to be effective in preclinical and clinical studies, only aluminium-based salts (Alum) and squalene-oil-water emulsion (MF59) have been approved for human use. However, for the development of therapeutic vaccines to treat cancer patients, the prerequisites for an ideal cancer adjuvant differ from conventional adjuvants for many reasons. First, the patients that will receive the vaccines are immuno-compromised because of, for example, impaired mechanisms of antigen presentation, non-responsiveness of activated T cells and enhanced inhibition of self-reactivity by regulatory T cells. Second, the tumour Ag are usually self-derived and are, therefore, poorly immunogenic. Third, tumours develop escape mechanisms to avoid the immune system, such as tumour editing, low or non-expression of MHC class I molecules or secretion of suppressive cytokines. Thus, adjuvants for cancer vaccines need to be more potent than for prophylactic vaccines and consequently may be more toxic and may even induce autoimmune reactions. In summary, the ideal cancer adjuvant should rescue and increase the immune response against tumours in immuno-compromised patients, with acceptable profiles of toxicity and safety. The present review discusses the role of cancer adjuvants at the different phases of the generation of antitumour immunity following vaccination.
Collapse
Affiliation(s)
- Circe Mesa
- Vaccine's Department, Centre of Molecular Immunology, Havana, Cuba.
| | | |
Collapse
|