1
|
Zhang J, Terreni M, Liu F, Sollogoub M, Zhang Y. Ganglioside GM3-based anticancer vaccines: Reviewing the mechanism and current strategies. Biomed Pharmacother 2024; 176:116824. [PMID: 38820973 DOI: 10.1016/j.biopha.2024.116824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Ganglioside GM3 is one of the most common membrane-bound glycosphingolipids. The over-expression of GM3 on tumor cells makes it defined as a tumor-associated carbohydrate antigen (TACA). The specific expression property in cancers, especially in melanoma, make it become an important target to develop anticancer vaccines or immunotherapies. However, in the manner akin to most TACAs, GM3 is an autoantigen facing with problems of low immunogenicity and easily inducing immunotolerance, which means itself only cannot elicit a powerful enough immune response to prevent or treat cancer. With a comparative understanding of the mechanisms that how immune system responses to the carbohydrate vaccines, this review summarizes the studies on the recent efforts to development GM3-based anticancer vaccines.
Collapse
Affiliation(s)
- Jiaxu Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Marco Terreni
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Fang Liu
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France
| | - Matthieu Sollogoub
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France
| | - Yongmin Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France; College of Life Sciences, Northwest University, Xi'an 710069, China.
| |
Collapse
|
2
|
Ho WL, Hsu WM, Huang MC, Kadomatsu K, Nakagawara A. Protein glycosylation in cancers and its potential therapeutic applications in neuroblastoma. J Hematol Oncol 2016; 9:100. [PMID: 27686492 PMCID: PMC5041531 DOI: 10.1186/s13045-016-0334-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023] Open
Abstract
Glycosylation is the most complex post-translational modification of proteins. Altered glycans on the tumor- and host-cell surface and in the tumor microenvironment have been identified to mediate critical events in cancer pathogenesis and progression. Tumor-associated glycan changes comprise increased branching of N-glycans, higher density of O-glycans, generation of truncated versions of normal counterparts, and generation of unusual forms of terminal structures arising from sialylation and fucosylation. The functional role of tumor-associated glycans (Tn, sTn, T, and sLea/x) is dependent on the interaction with lectins. Lectins are expressed on the surface of immune cells and endothelial cells or exist as extracellular matrix proteins and soluble adhesion molecules. Expression of tumor-associated glycans is involved in the dysregulation of glycogenes, which mainly comprise glycosyltransferases and glycosidases. Furthermore, genetic and epigenetic mechanisms on many glycogenes are associated with malignant transformation. With better understanding of all aspects of cancer-cell glycomics, many tumor-associated glycans have been utilized for diagnostic, prognostic, and therapeutic purposes. Glycan-based therapeutics has been applied to cancers from breast, lung, gastrointestinal system, melanomas, and lymphomas but rarely to neuroblastomas (NBs). The success of anti-disialoganglioside (GD2, a glycolipid antigen) antibodies sheds light on glycan-based therapies for NB and also suggests the possibility of protein glycosylation-based therapies for NB. This review summarizes our understanding of cancer glycobiology with a focus of how protein glycosylation and associated glycosyltransferases affect cellular behaviors and treatment outcome of various cancers, especially NB. Finally, we highlight potential applications of glycosylation in drug and cancer vaccine development for NB.
Collapse
Affiliation(s)
- Wan-Ling Ho
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan.,Department of Pediatrics, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 100, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| | - Min-Chuan Huang
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan. .,Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei, 10051, Taiwan.
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
3
|
Frequent co-expression of EGFR and NeuGcGM3 ganglioside in cancer: it's potential therapeutic implications. Clin Exp Metastasis 2016; 33:717-25. [PMID: 27449755 PMCID: PMC5035325 DOI: 10.1007/s10585-016-9811-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 07/11/2016] [Indexed: 12/25/2022]
Abstract
Interaction between epidermal growth factor receptor (EGFR) signaling with GM3 ganglioside expression has been previously described. However, little is known about EGFR and NeuGcGM3 co-expression in cancer patients and their therapeutic implications. In this paper, we evaluate the co-expression of EGFR and NeuGcGM3 ganglioside in tumors from 92 patients and in two spontaneous lung metastasis models of mice (Lewis lung carcinoma (3LL-D122) in C57BL/6 and mammary carcinoma (4T1) in BALB/c). As results, co-expression of EGFR and NeuGcGM3 ganglioside was frequently observed in 63 of 92 patients (68 %), independently of histological subtype. Moreover, EGFR is co-expressed with NeuGcGM3 ganglioside in the metastasis of 3LL-D122 and 4T1 murine models. Such dual expression appears to be therapeutically relevant, since combined therapy with mAbs against these two molecules synergistically increase the survival of mice treated. Overall, our results suggest that NeuGcGM3 and EGFR may coordinately contribute to the tumor cell biology and that therapeutic combinations against these two targets might be a valid strategy to explore.
Collapse
|
4
|
Barranco JAJ, Millar RP, Fuentes F, Bover E, Pimentel E, Basulto R, Calzada L, Morán R, Rodríguez A, Garay H, Reyes O, Castro MD, Bringas R, Arteaga N, Toudurí H, Rabassa M, Fernández Y, Serradelo A, Hernández E, Guillén GE. Gradual reduction of testosterone using a gonadotropin-releasing hormone vaccination delays castration resistance in a prostate cancer model. Oncol Lett 2016; 12:963-970. [PMID: 27446378 DOI: 10.3892/ol.2016.4679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 12/16/2015] [Indexed: 01/21/2023] Open
Abstract
In a previous study aimed to design a novel prostate cancer vaccine, the authors of the present study demonstrated the advantage of combining the adjuvants Montanide ISA 51 with very small size proteoliposomes (VSSP) to promote a significant humoral immune response to gonadotropin-releasing hormone (GnRH) in healthy animals. The present study compared the efficacy of this vaccine formulation versus the standard treatment currently available in terms of preventing the development of tumors in DD/S mice injected with Shionogi carcinoma (SC) 115 cells. The results demonstrated that 5 non-vaccinated control mice exhibited a fast tumor growth, and succumbed to the disease within 19-31 days. Mice immunized with the GnRH/Montanide ISA 51/VSSP vaccine exhibited a moderate decline in testosterone levels that was associated with a decrease in anti-GnRH antibody titers, which lead to a sustained tumor growth inhibition. In total, 2 mice in the immunized group exhibited complete remission of the tumor for the duration of the present study. In addition, castrated mice, which were used as a control for standard hormonal therapy, exhibited an accelerated decrease in tumor size. However, tumor relapse was observed between days 50 and 54, and between days 65 and 85, following the injection of SC 155 cells. Therefore, these mice were sacrificed at day 90. The present study concludes that the slow and moderate reduction of testosterone levels observed using the GnRH-based vaccine may delay the appearance of castration resistance in a Shionogi prostate cancer model. These findings suggest that this vaccine may be used to delay castration resistance in patients with prostate cancer.
Collapse
Affiliation(s)
- Jesús A Junco Barranco
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Robert P Millar
- Mammal Research Institute and Centre for Neuroendocrinology, University of Pretoria, Pretoria, Gauteng 0028, South Africa; MRC Receptor Biology Unit, Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, Western Cape 7925, South Africa
| | - Franklin Fuentes
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Eddy Bover
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Eulogio Pimentel
- Center for Genetic Engineering and Biotechnology, Havana, Havana 10600, Cuba
| | - Roberto Basulto
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Lesvia Calzada
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Rolando Morán
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Ayni Rodríguez
- Medical University of Camagüey, Camagüey, Camagüey 70100, Cuba
| | - Hilda Garay
- Center for Genetic Engineering and Biotechnology, Havana, Havana 10600, Cuba
| | - Osvaldo Reyes
- Center for Genetic Engineering and Biotechnology, Havana, Havana 10600, Cuba
| | - Maria D Castro
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Ricardo Bringas
- Center for Genetic Engineering and Biotechnology, Havana, Havana 10600, Cuba
| | - Niurka Arteaga
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Henio Toudurí
- Medical University of Camagüey, Camagüey, Camagüey 70100, Cuba
| | - Mauricio Rabassa
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | | | - Andrés Serradelo
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Eduardo Hernández
- Department of Vaccines, Center for Genetic Engineering and Biotechnology, Camagüey, Camagüey 70100, Cuba
| | - Gerardo E Guillén
- Center for Genetic Engineering and Biotechnology, Havana, Havana 10600, Cuba
| |
Collapse
|
5
|
Cacciavillano W, Sampor C, Venier C, Gabri MR, de Dávila MTG, Galluzzo ML, Guthmann MD, Fainboim L, Alonso DF, Chantada GL. A Phase I Study of the Anti-Idiotype Vaccine Racotumomab in Neuroblastoma and Other Pediatric Refractory Malignancies. Pediatr Blood Cancer 2015; 62:2120-4. [PMID: 26154941 DOI: 10.1002/pbc.25631] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/21/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Pediatric neuroectodermal malignancies express N-glycolylated gangliosides including N-glycolyl GM3 (NeuGcGM3) as targets for immunotherapy. PROCEDURE We evaluated the toxicity and maximum tolerated dose and immunological response of racotumomab, an anti-idiotype vaccine targeting NeuGcGM3 through a Phase I study enrolling children with relapsed or resistant tumors expressing NeuGcGM3. MATERIALS AND METHODS Drug dose was escalated to three levels (0.15-0.25-0.4 mg) of racotumomab administered intradermally. Each drug level included three patients receiving a total of three doses, every 14 days. A confirmation cohort was added to the highest dose level. Antibody response was assessed upon study entry and at 4-week intervals for at least three immunological determinations for each patient. RESULTS Fourteen patients were enrolled (10 with neuroblastoma, one with retinoblastoma, one with Wilms' tumor, and two with brainstem glioma). Three patients completed the three drug levels and three were enrolled in the confirmation cohort. One patient died of tumor progression before completing the three applications. Racotumomab was well tolerated. The only side effect observed was grade 1-2 toxicity at the injection site. Racotumomab elicited an IgM and/or IgG antibody response directed against NGcGM3 in nine patients and IgM against racotumomab in 11 of 13 evaluable patients. The maximum tolerated dose was not reached and no dose-limiting toxicity was seen. CONCLUSIONS Racotumomab vaccination has a favorable toxicity profile up to a dose of 0.4 mg, and most patients elicited an immune response. Its activity as immunotherapy for neuroectodermal malignancies will be tested in further clinical trials.
Collapse
Affiliation(s)
| | - Claudia Sampor
- Hospital de Pediatría SAMIC "Prof Dr Juan P Garrahan", Buenos Aires, Argentina
| | - Cecilia Venier
- Institute of Immunology, Genetics and Metabolism (INIGEM), University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Mariano R Gabri
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina
| | - María T G de Dávila
- Hospital de Pediatría SAMIC "Prof Dr Juan P Garrahan", Buenos Aires, Argentina
| | - Maria L Galluzzo
- Hospital de Pediatría SAMIC "Prof Dr Juan P Garrahan", Buenos Aires, Argentina
| | | | - Leonardo Fainboim
- Institute of Immunology, Genetics and Metabolism (INIGEM), University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Daniel F Alonso
- Laboratory of Molecular Oncology, National University of Quilmes, Buenos Aires, Argentina
| | | |
Collapse
|
6
|
Zheng XJ, Yang F, Zheng M, Huo CX, Zhang Y, Ye XS. Improvement of the immune efficacy of carbohydrate vaccines by chemical modification on the GM3 antigen. Org Biomol Chem 2015; 13:6399-406. [DOI: 10.1039/c5ob00405e] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
N-modified GM3 glycoconjugates improved the efficiency of the vaccination without the combination of metabolic oligosaccharide engineering technology.
Collapse
Affiliation(s)
- Xiu-Jing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- and Center for Molecular and Translational Medicine
- Peking University
- Beijing 100191
| | - Fan Yang
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- and Center for Molecular and Translational Medicine
- Peking University
- Beijing 100191
| | - Mingwei Zheng
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- and Center for Molecular and Translational Medicine
- Peking University
- Beijing 100191
| | - Chang-Xin Huo
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- and Center for Molecular and Translational Medicine
- Peking University
- Beijing 100191
| | - Ye Zhang
- School of Basic Medical Sciences
- Peking University
- Beijing 100191
- China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- and Center for Molecular and Translational Medicine
- Peking University
- Beijing 100191
| |
Collapse
|
7
|
Joo EJ, Weyers A, Li G, Gasimli L, Li L, Choi WJ, Lee KB, Linhardt RJ. Carbohydrate-containing molecules as potential biomarkers in colon cancer. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:231-41. [PMID: 24502776 DOI: 10.1089/omi.2013.0128] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glycans play a critical role in physiological and pathological processes through interaction with a variety of ligands. Altered expression and dysregulation of these molecules can cause aberrant cellular function such as malignancy. Glycomics provide information of the structure and function of glycans, glycolipids, and glycoproteins such as proteoglycans, and may help to predict cancer development and progression as biomarkers. In this report, we compared the expression of proteoglycans, the content and structure of glycosaminoglycans and glycolipids between patient-matched normal and cancer tissues obtained from colon cancer patients. Tumor-related proteoglycans, glypican-3, and syndecan-1 showed downregulation in cancer tissues compared to normal tissues. In cancer tissue, the total amount of chondroitin sulfate (CS)/dermatan sulfate and heparan sulfate were lower and, interestingly, the level of disaccharide units of both 4S6S (CS-E) and 6S (CS-C) were higher compared to normal tissue. Also, overall lipids including glycolipids, a major glycomics target, were analyzed by hydrophilic interaction liquid chromatography mass spectrometry. Increase of lyso-phosphatidylcholine (phospholipid), sphingomyelin (sphigolipid), and four types of glycolipids (glucosylceramide, lactosylceramide, monosialic acid ganglioside, and globoside 4) in cancer tissue showed the possibility as potential biomarkers in colon cancer. While requiring the need for careful interpretation, this type of broad investigation gives us a better understanding of pathophysiological roles on glycosaminoglycans and glycolipids and might be a powerful tool for colon cancer diagnosis.
Collapse
Affiliation(s)
- Eun Ji Joo
- 1 Department of Chemical and Chemical Biology, Rensselaer Polytechnic Institute , Troy, New York
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Rojas G, Pupo A, Gómez S, Krengel U, Moreno E. Engineering the binding site of an antibody against N-glycolyl GM3: from functional mapping to novel anti-ganglioside specificities. ACS Chem Biol 2013; 8:376-86. [PMID: 23138862 DOI: 10.1021/cb3003754] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The structurally related gangliosides N-glycolyl GM3 and N-acetyl GM3 are potential targets for tumor immunotherapy. 14F7 is a monoclonal antibody able to discriminate the tumor-specific antigen N-glycolyl GM3 from the closely related N-acetyl GM3 on the basis of the presence of a single additional hydroxyl group in the former. A combinatorial phage display strategy, based on the screening of a large library followed by refined mutagenesis, allowed a thorough exploration of the binding chemistry of this unique antibody. Three essential features of the heavy chain variable region were identified: two aromatic rings (in positions 33 and 100D) contributing to the binding site architecture and an arginine residue (position 98) critical for recognition. Directed evolution of 14F7 resulted in novel variants that cross-react with the tumor-associated antigen N-acetyl GM3 and display recurrent replacements: the substitution W33Q and the appearance of additional arginine residues at several positions of CDR H1. Successful conversion of such engineered variable regions into whole cross-reactive anti-GM3 immunoglobulins validated our phage-based approach to study and modify the lead antibody 14F7. The resulting family of closely related antibodies offers new tools to study the mechanisms of cell death induced by antibodies targeting gangliosides. In vitro directed evolution was useful to overcome the technical limitations to obtain anti-ganglioside antibodies. The case of 14F7 illustrates the power of combining library screening with focused site-directed randomization for a comprehensive scanning of protein interactions.
Collapse
Affiliation(s)
- Gertrudis Rojas
- Center of Molecular Immunology, Calle 216 esq 15, Atabey, Playa, PO Box
16040, La Habana CP 11600, Cuba
| | - Amaury Pupo
- Center of Molecular Immunology, Calle 216 esq 15, Atabey, Playa, PO Box
16040, La Habana CP 11600, Cuba
| | - Silvia Gómez
- Center of Molecular Immunology, Calle 216 esq 15, Atabey, Playa, PO Box
16040, La Habana CP 11600, Cuba
| | - Ute Krengel
- Department of Chemistry, University of Oslo, PO Box 1033 Blindern, NO-0315 Oslo,
Norway
| | - Ernesto Moreno
- Center of Molecular Immunology, Calle 216 esq 15, Atabey, Playa, PO Box
16040, La Habana CP 11600, Cuba
| |
Collapse
|
9
|
Recent advances in developing synthetic carbohydrate-based vaccines for cancer immunotherapies. Future Med Chem 2012; 4:545-84. [DOI: 10.4155/fmc.11.193] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cancer cells can often be distinguished from healthy cells by the expression of unique carbohydrate sequences decorating the cell surface as a result of aberrant glycosyltransferase activity occurring within the cell; these unusual carbohydrates can be used as valuable immunological targets in modern vaccine designs to raise carbohydrate-specific antibodies. Many tumor antigens (e.g., GM2, Ley, globo H, sialyl Tn and TF) have been identified to date in a variety of cancers. Unfortunately, carbohydrates alone evoke poor immunogenicity, owing to their lack of ability in inducing T-cell-dependent immune responses. In order to enhance their immunogenicity and promote long-lasting immune responses, carbohydrates are often chemically modified to link to an immunogenic protein or peptide fragment for eliciting T-cell-dependent responses. This review will present a summary of efforts and advancements made to date on creating carbohydrate-based anticancer vaccines, and will include novel approaches to overcoming the poor immunogenicity of carbohydrate-based vaccines.
Collapse
|
10
|
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
|
11
|
Dias BR, Rodrigues EG, Nimrichter L, Nakayasu ES, Almeida IC, Travassos LR. Identification of iGb3 and iGb4 in melanoma B16F10-Nex2 cells and the iNKT cell-mediated antitumor effect of dendritic cells primed with iGb3. Mol Cancer 2009; 8:116. [PMID: 19968878 PMCID: PMC2795753 DOI: 10.1186/1476-4598-8-116] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 12/07/2009] [Indexed: 01/23/2023] Open
Abstract
Background CD1d-restricted iNKT cells are protective against murine melanoma B16F10-Nex2 growing subcutaneously in syngeneic C57Bl/6 mice as inferred from the fast tumor development in CD1d-KO in comparison with wild type animals. CD1d glycoproteins are related to the class I MHC molecules, and are involved in the presentation, particularly by dentritic cells (DC), of lipid antigens to iNKT cells. In the present work we attempted to identify the endogenous lipid mediator expressed in melanoma cells inducing such immunesurveillance response and study the possibility of protecting animals challenged with tumor cells with lipid-primed DC. Results Crude cytosolic and membrane fractions from in vivo growing melanoma contained iNKT-stimulating substances. Lipids were then extracted from these cells and one of the fractions (i.e. F3A) was shown to prime bone marrow-derived dendritic cells (BMDC) to stimulate iNKT murine hybridoma (DN32D3) cells to produce IL-2. The active fraction was analyzed by electrospray ionization-mass spectrometry (ESI-LIT-MS) and both iGb3 and iGb4 were identified along with GM3. When iGb3 was incubated with BMDC and tested with DN32D3 cells, IL-2 was equally produced indicating iNKT cell activation. GM3 consistently inhibited this response. To assess the antitumor response-induced by iGb3, a cytotoxicity assay in vitro was used with [3H]-thymidine labeled B16F10-Nex2 cells. At target/effector (iGb3-activated iNKT) cell ratio of 100-1-100-4 tumor cell lysis was shown. The antitumor activity in vivo was tested in mice challenged i.v. with B16F10-Nex2 cells and treated with iGb3- or α-galactosylceramide-primed DCs. A 4-fold lower tumor load in the lungs was observed with either treatment. Conclusion Our results show the expression of globo and isoglobohexosylceramides in murine melanoma B16F10-Nex2. The expression of iGb3 and its precursor, iGb4, on tumor cells may prime an effective iNKT cell-dependent antitumor response, modulated negatively by GM3 which is also produced in these cells. iGb3-primed BMDC exerted a significant iNKT cell-mediated anti-tumor activity in mice challenged with melanoma cells.
Collapse
Affiliation(s)
- Bianca R Dias
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
12
|
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
|
13
|
Gabri MR, Mazorra Z, Ripoll GV, Mesa C, Fernandez LE, Gomez DE, Alonso DF. Complete antitumor protection by perioperative immunization with GM3/VSSP vaccine in a preclinical mouse melanoma model. Clin Cancer Res 2007; 12:7092-8. [PMID: 17145833 DOI: 10.1158/1078-0432.ccr-06-1075] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE The GM3/VSSP vaccine is composed of very small sized proteoliposomes resulting from the hydrophobic conjugation of GM3 ganglioside with membrane proteins from Neisseria meningitidis. Previously, we showed that preventive vaccination with GM3/VSSP induces a specific antitumor response and elicits the rejection of syngeneic GM3-positive melanoma cells in immunized mice. Our aim was to explore the antitumor properties of perioperative GM3/VSSP vaccination in a preclinical mouse model. EXPERIMENTAL DESIGN The highly metastatic B16F10 mouse melanoma was used to investigate perioperative vaccination with GM3/VSSP. The vaccine was administered i.m. in doses of 120 microg emulsified with the adjuvant Montanide ISA 51 at weekly or biweekly intervals, and s.c. tumors were excised 25 to 31 days after tumor cell implantation. The persistence of antitumor protection and dose dependency was also examined in preimmunized animals. To evaluate the immune performance of tumor-bearing and tumor-operated mice, ovoalbumin-specific delayed-type hypersensitivity, cytokine secretion, and cell proliferation responses were studied. RESULTS Surgical excision of B16F10 tumors improved survival, and perioperative immunization with four biweekly GM3/VSSP doses yielded survival for all animals (P = 0.04; log-rank test). Mice showed neither local recurrence nor lung metastasis at the end of the experiment. An impairment of CD4(+) T-cell responses was observed in tumor-bearing animals measured as neoantigen-specific delayed-type hypersensitivity, with a significant recovery after surgery. A strong interleukin-4 secretion was induced in B16F10-operated mice, whereas IFN-gamma remained unaffected. CONCLUSION Preclinical evidence suggests that GM3/VSSP vaccine might have therapeutic potential to induce antitumor immunity in patients with minimal residual disease after surgery, thereby preventing or prolonging the time to recurrence.
Collapse
Affiliation(s)
- Mariano R Gabri
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
14
|
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
|
15
|
Mesa C, de León J, Rigley K, Fernández LE. Very small size proteoliposomes derived from Neisseria meningitidis: an effective adjuvant for dendritic cell activation. Vaccine 2006; 24 Suppl 2:S2-42-3. [PMID: 16823920 DOI: 10.1016/j.vaccine.2005.01.114] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Recent findings in the interactions between pathogens and the innate immune system, particularly with dendritic cells (DC), have opened new opportunities for adjuvants designs. We have elaborated a new approach, in which gangliosides are incorporated into the outer membrane complex of Neisseria meningitidis to form very small size proteoliposomes (VSSP). VSSP demonstrated a unique ability to render highly tolerated gangliosides immunogenic. These results drove our attention to the immunopotentiating properties of VSSP. Here we examined the VSSP adjuvant effect on dendritic cell activation. Also the role of LPS on this effect was dissected. This study reveals that VSSP is a potent adjuvant for DC activation.
Collapse
Affiliation(s)
- Circe Mesa
- Vaccine's Department, Centre of Molecular Immunology, 216 esq 15, Atabey, Playa, CP 16040, C. Habana, Cuba.
| | | | | | | |
Collapse
|
16
|
Ségui B, Andrieu-Abadie N, Jaffrézou JP, Benoist H, Levade T. Sphingolipids as modulators of cancer cell death: potential therapeutic targets. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1758:2104-20. [PMID: 16925980 DOI: 10.1016/j.bbamem.2006.05.024] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 05/04/2006] [Accepted: 05/06/2006] [Indexed: 02/07/2023]
Abstract
Through modifications in the fine membrane structure, cell-cell or cell-matrix interactions, and/or modulation of intracellular signaling pathways, sphingolipids can affect the tumorigenic potential of numerous cell types. Whereas ceramide and its metabolites have been described as regulators of cell growth and apoptosis, these lipids as well as other sphingolipid molecules can modulate the ability of malignant cells to grow and resist anticancer treatments, and their susceptibility to non-apoptotic cell deaths. This review summarizes our current knowledge on the properties of sphingolipids in the regulation of cancer cell death and tumor development. It also provides an update on the potential perspectives of manipulating sphingolipid metabolism and using sphingolipid analogues in anticancer therapy.
Collapse
Affiliation(s)
- Bruno Ségui
- INSERM U.466, Laboratoire de Biochimie, Institut Louis Bugnard, Centre Hospitalier Universitaire de Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | | | | | | | | |
Collapse
|
17
|
Abstract
In the past decades, a gradual increase in the resistance to antibiotics has been observed, leading to a serious thread for successful treatment of bacterial infections. This feature in addition to difficulties in developing adequate drugs against (tropical) diseases caused by parasites has stimulated the interest in vaccines to prevent infections. In principle, various types of cell surface epitopes, characteristic for the invading organism or related to aberrant growth of cells, can be applied to develop vaccines. The progress in establishing the structure of carbohydrate immuno-determinants in conjunction with improvements in carbohydrate synthesis has rendered it feasible to develop new generations of carbohydrate-based vaccines.
Collapse
Affiliation(s)
- Johannes F G Vliegenthart
- Bijvoet Center, Division Bioorganic Chemistry, Utrecht University, Padualaan 8, NL-3584 CH Utrecht, The Netherlands.
| |
Collapse
|
18
|
Guthmann MD, Bitton RJ, Carnero AJL, Gabri MR, Cinat G, Koliren L, Lewi D, Fernandez LE, Alonso DF, Gómez DE, Fainboim L. Active specific immunotherapy of melanoma with a GM3 ganglioside-based vaccine: a report on safety and immunogenicity. J Immunother 2005; 27:442-51. [PMID: 15534488 DOI: 10.1097/00002371-200411000-00004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A novel cancer vaccine was obtained by combining GM3 ganglioside with Neisseria meningitidis outer membrane protein complex to obtain very-small-size proteoliposomes (GM3/VSSP). The authors report the results of a phase 1 study of intramuscular administration of GM3/VSSP/Montanide ISA 51 to patients with metastatic melanoma. Twenty-six patients were included in three dose-level cohorts of 120, 240, and 360 mug. The first five doses (induction phase) were given at 2-week intervals, and the remaining four doses were given monthly. Patients were evaluated for dose-related toxicities and antitumor effects. In addition, serum and peripheral blood mononuclear cells were obtained at baseline and throughout treatment to evaluate humoral and cellular immune responses. One episode of severe hypotension and fever was observed in a patient included at the highest dose level. Other toxicities consisted of local reactions at the site of injection and mild fever and chills. Five doses of GM3/VSSP induced an anti-GM3 IgM response in 44% of patients. Serum reactivity was also observed against melanoma cell lines and tumor biopsies. GM3/VSSP was shown to induce very strong in vitro IFNgamma secretion in all evaluated melanoma patients. Furthermore, in one patient IFNgamma secretion was shown to be GM3-specific. A 62% reduction of a mediastinal mass was documented in one patient (partial response), while a second patient benefited from initial disease stabilization followed by tumor reduction in nonmeasurable soft tissue lesions accompanied by vitiligo.
Collapse
Affiliation(s)
- Marcelo D Guthmann
- Immunogenetics Division, Hospital de Clínicas, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mesa C, De León J, Rigley K, Fernández LE. Very small size proteoliposomes derived from Neisseria meningitidis: an effective adjuvant for Th1 induction and dendritic cell activation. Vaccine 2004; 22:3045-52. [PMID: 15297054 DOI: 10.1016/j.vaccine.2004.02.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2003] [Accepted: 02/05/2004] [Indexed: 10/26/2022]
Abstract
Recent findings about pathogens and innate immune system interactions have opened new opportunities for adjuvants designs. We have elaborated a new approach, in which gangliosides are incorporated into the outer membrane complex of Neisseria meningitidis (Nm) to form very small size proteoliposomes (VSSP). VSSP, used as monotherapy, demonstrated a unique ability to render immunogenic highly tolerated gangliosides. These results drove our attention to the immunopotentiating properties of VSSP. Here, we examined the VSSP adjuvant effect on the humoral and cellular responses, dendritic cell (DC) activation, and differentiation of Th cells. Also, the role of LPS in VSSP effect was dissected. This study reveals that VSSP is a potent adjuvant for dendritic cells activation and Th1 differentiation.
Collapse
Affiliation(s)
- Circe Mesa
- Vaccine's Department, Centre of Molecular Immunology, 216 esq 15, Atabey, Playa, CP 16040 Havana, Cuba.
| | | | | | | |
Collapse
|
20
|
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
|
21
|
Ravindranath MH, Wood TF, Soh D, Gonzales A, Muthugounder S, Perez C, Morton DL, Bilchik AJ. Cryosurgical ablation of liver tumors in colon cancer patients increases the serum total ganglioside level and then selectively augments antiganglioside IgM. Cryobiology 2002; 45:10-21. [PMID: 12445546 DOI: 10.1016/s0011-2240(02)00102-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cryosurgical ablation (CSA) of tumors induces disruptive necrosis. Necrosis may release tumor gangliosides into circulation and they may augment serum antiganglioside antibodies depending on the nature of gangliosides released. The hypothesis is tested by determining the level of serum total gangliosides (STG) and their antibody titers in the sera of colon cancer patients with cryoablated liver tumors. As controls, we examined the sera of patients who underwent radiofrequency ablation (RFA) and regular surgery (RS), none of which cause disruptive necrosis. The STG level (expressed as lipid-bound sialic acids, LBSA) is higher (p(2)<0.001) in 35 patients (stage IV) than in 38 healthy case-controls (median 23.48 mg/dL, Q-range 7.1 vs 16.04 mg/dL, Q-range 4.5). The mean STG level increased significantly to 31.2+/-6.0mg/dL (p(2)<0.03) after CSA. Concomitantly, the IgM titer against colon cancer-associated gangliosides (GM(2), GD(1a), GT(1b)), increased significantly, but no increase was observed against normal tissue gangliosides (GM(3) or GM(1)). Also after RFA and RS, no such increase was observed either in the level of STG or in IgM titer against tumor gangliosides. The results suggest that CSA-induced necrosis might have acted as an adjuvant, because purified gangliosides without exogenous adjuvants even after repeated immunization failed to elicit antibody response. The post-CSA decline in the STG level correlated with the increase in the antibodies, suggesting a homeostatic role of the antibodies.
Collapse
Affiliation(s)
- Mepur H Ravindranath
- Laboratory of GlycoImmunotherapy, John Wayne Cancer Institute, 2200 Santa Monica Boulevard, Santa Monica, CA 90404-2302, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Bada A, Casacó Parada A, Arteaga M, Martínez J, León A, Santana E, Hernández O, Orphee R, González A, Mesa C, González C, Montero E, Fernández LE. Toxicity of a GM3 cancer vaccine in Macaca fascicularis monkey: a 12-month study. Hum Exp Toxicol 2002; 21:263-7. [PMID: 12141397 DOI: 10.1191/0960327102ht248oa] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
GM3 is a ganglioside that has been biochemically identified as dominating the cell surface of several human tumours, but is also found on human normal cells at much lower density. Since GM3 is widely distributed in essentially all types of animal cells, there is a conflict with the concepts of tumour-associated antigen, immunogen, and toxicity. We have designed a GM3-based cancer vaccine for the treatment of human breast and melanoma tumours. Prior to the Phase I clinical trial, we carried out a 12-month dose repeated toxicity study in five male Macaca fascicularis monkeys. Four male monkeys were treated with placebo in a similar way. During the study, no differences were observed between control and treated monkeys related to daily clinical observations (other than local damage) including rectal temperature, blood pressure, respiratory and cardiac rates, weight gain, biochemical and hematological parameters (with the exception of transitory pathological changes), and anti-DNA and anti-nuclear antibodies, although treated monkeys consistently developed both IgM- and IgG-specific anti-GM3 antibodies. Sixty per cent of treated monkeys developed moderate local reactions at the injection site, which disappeared without sequels. We concluded that this GM3 cancer vaccine overcame in monkeys the natural tolerance to GM3 ganglioside evidenced by a strong immune response, while the local reactions elicited-were transitory without apparent important systemic toxicity effects.
Collapse
Affiliation(s)
- A Bada
- Centro Nacional para la Producción de Animales de Laboratorio, Habana, Cuba
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|