51
|
Development of a radiolabeled caninized anti-EGFR antibody for comparative oncology trials. Oncotarget 2017; 8:83128-83141. [PMID: 29137329 PMCID: PMC5669955 DOI: 10.18632/oncotarget.20914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Due to large homology of human and canine EGFR, dogs suffering from spontaneous EGFR+ cancer can be considered as ideal translational models. Thereby, novel immunotherapeutic compounds can be developed for both human and veterinary patients. This study describes the radiolabeling of a canine anti-EGFR IgG antibody (can225IgG) with potential diagnostic and therapeutic value in comparative clinical settings. Can225IgG was functionalized with DTPA for subsequent chelation with the radionuclide 99mTc. Successful coupling of 10 DTPA molecules per antibody on average was proven by significant mass increase in MALDI-TOF spectroscopy, gel electrophoresis and immunoblots. Following functionalization and radiolabeling, 99mTc-DTPA-can225IgG fully retained its binding capacity towards human and canine EGFR in flow cytometry, immuno- and radioblots, and autoradiography. The affinity of radiolabeled can225IgG was determined to KD 0.8 ±0.0031 nM in a real-time kinetics assay on canine carcinoma cells by a competition binding technique. Stability tests of the radiolabeled compound identified TRIS buffered saline as the ideal formulation for short-term storage with 87.11 ±6.04% intact compound being still detected 60 minutes post radiolabeling. High stability, specificity and EGFR binding affinity pinpoint towards 99mTc-radiolabeled can225IgG antibody as an ideal lead compound for the first proof-of-concept diagnostic and therapeutic applications in canine cancer patients.
Collapse
|
52
|
Hadzijusufovic E, Willmann M. Comparing Human Breast Cancer with Canine Mammary Cancer. Comp Med 2017. [DOI: 10.1007/978-3-319-47007-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
53
|
A Comparative Approach of Tumor-Associated Inflammation in Mammary Cancer between Humans and Dogs. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4917387. [PMID: 28053982 PMCID: PMC5178344 DOI: 10.1155/2016/4917387] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/24/2016] [Accepted: 11/03/2016] [Indexed: 12/20/2022]
Abstract
Infiltrating cells of the immune system are widely accepted to be generic constituents of tumor microenvironment. It has been well established that the development of mammary cancer, both in humans and in dogs, is associated with alterations in numbers and functions of immune cells at the sites of tumor progression. These tumor infiltrating immune cells seem to exhibit exclusive phenotypic and functional characteristics and mammary cancer cells can take advantage of signaling molecules released by them. Cancer related inflammation has an important role in mammary carcinogenesis, contributing to the acquisition of core hallmark capabilities that allow cancer cells to survive, proliferate, and disseminate. Indeed, recent studies in human breast cancer and in canine mammary tumors have identified a growing list of signaling molecules released by inflammatory cells that serve as effectors of their tumor-promoting actions. These include the COX-2, the tumor EGF, the angiogenic VEGF, other proangiogenic factors, and a large variety of chemokines and cytokines that amplify the inflammatory state. This review describes the intertwined signaling pathways shared by T-lymphocytic/macrophage infiltrates and important tissue biomarkers in both human and dog mammary carcinogenesis.
Collapse
|
54
|
Birtoiu IA, Rizea C, Togoe D, Munteanu RM, Micsa C, Rusu MI, Tautan M, Braic L, Scoicaru LO, Parau A, Becherescu-Barbu ND, Udrea MV, Tonetto A, Notonier R, Grigorescu CEA. Diagnosing clean margins through Raman spectroscopy in human and animal mammary tumour surgery: a short review. Interface Focus 2016; 6:20160067. [PMID: 27920899 DOI: 10.1098/rsfs.2016.0067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Breast cancer frequency in human and other mammal female populations has worryingly increased lately. The acute necessity for taxonomy of the aetiological factors along with seeking for new diagnostic tools and therapy procedures aimed at reducing mortality have yielded in an intense research effort worldwide. Surgery is a regular method to counteract extensive development of breast cancer and prevent metastases provided that negative surgical margins are achieved. This highly technical challenge requires fast, extremely sensitive and selective discrimination between malignant and benign tissues even down to molecular level. The particular advantages of Raman spectroscopy, such as high chemical specificity, and the ability to measure raw samples and optical responses in the visible or near-infrared spectral range, have recently recommended it as a means with elevated potential in precise diagnostic in oncology surgery. This review spans mainly the latter 10 years of exceptional efforts of scientists implementing Raman spectroscopy as a nearly real-time diagnostic tool for clean margins assessment in mastectomy and lumpectomy. Although greatly contributing to medical discoveries for the wealth of humanity, animals as patients have benefitted less from advances in surgery diagnostic using Raman spectroscopy. This work also dedicates a few lines to applications of surface enhanced Raman spectroscopy in veterinary oncological surgery.
Collapse
Affiliation(s)
- I A Birtoiu
- Faculty of Veterinary Medicine-University of Agronomic Sciences and Veterinary Medicine , Bucharest , Romania
| | - C Rizea
- ROXY VETERINARY S.R.L , Magurele , Romania
| | - D Togoe
- Faculty of Veterinary Medicine-University of Agronomic Sciences and Veterinary Medicine , Bucharest , Romania
| | - R M Munteanu
- Faculty of Veterinary Medicine-University of Agronomic Sciences and Veterinary Medicine , Bucharest , Romania
| | - C Micsa
- Faculty of Veterinary Medicine-University of Agronomic Sciences and Veterinary Medicine , Bucharest , Romania
| | - M I Rusu
- National Institute of Research and Development for Optoelectronics INOE 2000 , Magurele , Romania
| | - M Tautan
- National Institute of Research and Development for Optoelectronics INOE 2000 , Magurele , Romania
| | - L Braic
- National Institute of Research and Development for Optoelectronics INOE 2000 , Magurele , Romania
| | - L O Scoicaru
- National Institute of Research and Development for Optoelectronics INOE 2000 , Magurele , Romania
| | - A Parau
- National Institute of Research and Development for Optoelectronics INOE 2000 , Magurele , Romania
| | - N D Becherescu-Barbu
- APEL LASER S.R.L., Bucharest, Romania; Faculty of Physics, University of Bucharest, Bucharest, Romania
| | - M V Udrea
- APEL LASER S.R.L. , Bucharest , Romania
| | - A Tonetto
- Aix-Marseille Université , Centrale Marseille, CNRS, Fédération Sciences Chimiques Marseille (FR 1739) - PRATIM, 13000 Marseille , France
| | - R Notonier
- Aix-Marseille Université , Centrale Marseille, CNRS, Fédération Sciences Chimiques Marseille (FR 1739) - PRATIM, 13000 Marseille , France
| | - C E A Grigorescu
- National Institute of Research and Development for Optoelectronics INOE 2000 , Magurele , Romania
| |
Collapse
|
55
|
Beirão BCB, Raposo T, Jain S, Hupp T, Argyle DJ. Challenges and opportunities for monoclonal antibody therapy in veterinary oncology. Vet J 2016; 218:40-50. [PMID: 27938708 DOI: 10.1016/j.tvjl.2016.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/15/2016] [Accepted: 11/13/2016] [Indexed: 12/22/2022]
Abstract
Monoclonal antibodies (mAbs) have come to dominate the biologics market in human cancer therapy. Nevertheless, in veterinary medicine, very few clinical trials have been initiated using this form of therapy. Some of the advantages of mAb therapeutics over conventional drugs are high specificity, precise mode of action and long half-life, which favour infrequent dosing of the antibody. Further advancement in the field of biomedical sciences has led to the production of different forms of antibodies, such as single chain antibody fragment, Fab, bi-specific antibodies and drug conjugates for use in diagnostic and therapeutic purposes. This review describes the potential for mAbs in veterinary oncology in supporting both diagnosis and therapy of cancer. The technical and financial hurdles to facilitate clinical acceptance of mAbs are explored and insights into novel technologies and targets that could support more rapid clinical development are offered.
Collapse
Affiliation(s)
- Breno C B Beirão
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, EH25 9RG, United Kingdom
| | - Teresa Raposo
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, EH25 9RG, United Kingdom; Department of Veterinary Sciences, Universidade de Trás-os-Montes e Alto Douro, 5001-801, Portugal
| | - Saurabh Jain
- Edinburgh Cancer Research Centre, University of Edinburgh, EH4 2XR, United Kingdom
| | - Ted Hupp
- Edinburgh Cancer Research Centre, University of Edinburgh, EH4 2XR, United Kingdom
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, EH25 9RG, United Kingdom.
| |
Collapse
|
56
|
Fazekas J, Fürdös I, Singer J, Jensen-Jarolim E. Why man's best friend, the dog, could also benefit from an anti-HER-2 vaccine. Oncol Lett 2016; 12:2271-2276. [PMID: 27698788 PMCID: PMC5038860 DOI: 10.3892/ol.2016.5001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 05/12/2016] [Indexed: 01/11/2023] Open
Abstract
Human epidermal growth factor receptor-2 (HER-2) is a well-established target for anticancer anticancerprecision medicine in humans. A HER-2 homologue with 92% amino acid identity has been described in canine mammary tumors, which whichis termed here as ‘dog epidermal growth factor receptor-2 (DER-2)’, with similar biological implications as those in human breast cancer. Both antigens can principally be immunologically targeted by anti-HER-2 antibodies, such as trastuzumab; however, the in vivo application applicationof humanized antibodies to other species would lead to specific hypersensitivity reactions. Therefore, HER-2 mimotope vaccines that actively induce autologous trastuzumab-like immunoglobulins represent a novel and economic treatment option to overcome species-specific limitations. Thus, the present review proposes the implementation of clinical trials with HER-2 vaccines in canine cancer model modelpatients with spontaneous DER-2 positive mammary gland carcinomas in order to assess their safety and efficacy. This approach would not only pave the way into the veterinary oncology market, but would also similarly generate robust data for human trials and facilitate the testing of novel combinatorial treatments.
Collapse
Affiliation(s)
- Judit Fazekas
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna 1210, Austria; Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna 1090, Austria
| | - Irene Fürdös
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna 1210, Austria
| | - Josef Singer
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna 1210, Austria; Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna 1090, Austria
| | - Erika Jensen-Jarolim
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna 1210, Austria; Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna 1090, Austria
| |
Collapse
|
57
|
Gray ME, Lee S, McDowell AL, Erskine M, Loh QTM, Grice O, Argyle DJ, Bergkvist GT. Dual targeting of EGFR and ERBB2 pathways produces a synergistic effect on cancer cell proliferation and migration in vitro. Vet Comp Oncol 2016; 15:890-909. [PMID: 27229930 DOI: 10.1111/vco.12230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 12/16/2022]
Abstract
Members of the epidermal growth factor receptor (EGFR/ERBB) gene family are frequently dysregulated in a range of human cancers, and therapeutics targeting these proteins are in clinical use. We hypothesized that similar pathways are involved in feline and canine tumours and that the same drugs may be of clinical use in veterinary patients. We investigated EGFR and ERBB2 targeting using a panel of feline and canine cell lines. EGFR and ERBB2 were targeted with siRNAs or tyrosine kinase inhibitors (TKIs) and their effect on cellular proliferation, colony formation and migration was investigated in vitro. Here we report that EGFR and ERBB2 combined siRNA targeting produced synergistic effects in feline and canine cell lines similar to that reported in human cell lines. We conclude that dual EGFR and ERBB2 targeting using TKIs should be further evaluated as a potential new therapeutic strategy in feline head and neck and mammary tumours and canine mammary tumours.
Collapse
Affiliation(s)
- M E Gray
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - S Lee
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - A L McDowell
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - M Erskine
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Q T M Loh
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - O Grice
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - D J Argyle
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| | - G T Bergkvist
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, EH25 9RG, UK
| |
Collapse
|
58
|
Development of new therapy for canine mammary cancer with recombinant measles virus. MOLECULAR THERAPY-ONCOLYTICS 2016; 3:15022. [PMID: 27119113 PMCID: PMC4782952 DOI: 10.1038/mto.2015.22] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/06/2015] [Accepted: 11/06/2015] [Indexed: 12/21/2022]
Abstract
Oncolytic virotherapy is a promising treatment strategy for cancer. We previously generated a recombinant measles virus (rMV-SLAMblind) that selectively uses a poliovirus receptor-related 4 (PVRL4/Nectin4) receptor, but not signaling lymphocyte activation molecule (SLAM). We demonstrated that the virus exerts therapeutic effects against human breast cancer cells. Here, we examined the applicability of rMV-SLAMblind to treating canine mammary cancers (CMCs). We found that the susceptibilities of host cells to rMV-SLAMblind were dependent on canine Nectin-4 expression. Nectin-4 was detected in four of nine CMC cell lines. The rMV-SLAMblind efficiently infected those four Nectin-4-positive cell lines and was cytotoxic for three of them (CF33, CHMm, and CTBm). In vivo experiment showed that the administration of rMV-SLAMblind greatly suppressed the progression of tumors in mice xenografted with a CMC cell line (CF33). Immunohistochemistry revealed that canine Nectin-4 was expressed in 45% of canine mammary tumors, and the tumor cells derived from one clinical specimen were efficiently infected with rMV-SLAMblind. These results suggest that rMV-SLAMblind infects CMC cells and displays antitumor activity in vitro, in xenografts, and ex vivo. Therefore, oncolytic virotherapy with rMV-SLAMblind can be a novel method for treating CMCs.
Collapse
|
59
|
|
60
|
Matos A, Santos A. Advances in the understanding of the clinically relevant genetic pathways and molecular aspects of canine mammary tumours: Part 1. Proliferation, apoptosis and DNA repair. Vet J 2015; 205:136-43. [DOI: 10.1016/j.tvjl.2015.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 02/09/2023]
|
61
|
Marconato L, Buracco P, Aresu L. Perspectives on the design of clinical trials for targeted therapies and immunotherapy in veterinary oncology. Vet J 2015; 205:238-43. [DOI: 10.1016/j.tvjl.2015.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 02/15/2015] [Accepted: 02/25/2015] [Indexed: 12/18/2022]
|
62
|
Investigation of HER2 expression in canine mammary tumors by antibody-based, transcriptomic and mass spectrometry analysis: is the dog a suitable animal model for human breast cancer? Tumour Biol 2015; 36:9083-91. [PMID: 26088453 DOI: 10.1007/s13277-015-3661-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/09/2015] [Indexed: 11/27/2022] Open
Abstract
Canine mammary tumors (CMTs) share many features with human breast cancer (HBC), specifically concerning cancer-related pathways. Although the human epidermal growth factor receptor 2 (HER2) plays a significant role as a therapeutic and prognostic biomarker in HBC, its relevance in the pathogenesis and prognosis of CMT is still controversial. The aim of this study was to investigate HER2 expression in canine mammary hyperplasic and neoplastic tissues as well as to evaluate the specificity of the most commonly used polyclonal anti HER2 antibody by multiple molecular approaches. HER2 protein and RNA expression were determined by immunohistochemistry (IHC) and by quantitative real-time (qRT) PCR. A strong cell membrane associated with non-specific cytoplasmic staining was observed in 22% of carcinomas by IHC. Adenomas and carcinomas exhibited a significantly higher HER2 mRNA expression when compared to normal mammary glands, although no significant difference between benign and malignant tumors was noticed by qRT-PCR. The IHC results suggest a lack of specificity of the FDA-approved antibody in CMT samples as further demonstrated by Western immunoblotting (WB) and reverse phase protein arrays (RPPA). Furthemore, HER2 was not detected by mass spectrometry (MS) in a protein-expressing carcinoma at the IHC investigation. This study highlights that caution needs to be used when trying to translate from human to veterinary medicine information concerning cancer-related biomarkers and pathways. Further investigations are necessary to carefully assess the diagnostic and biological role specifically exerted by HER2 in CMTs and the use of canine mammary tumors as a model of HER2 over-expressing breast cancer.
Collapse
|
63
|
Toward immunotherapy with redirected T cells in a large animal model: ex vivo activation, expansion, and genetic modification of canine T cells. J Immunother 2015; 37:407-15. [PMID: 25198528 DOI: 10.1097/cji.0000000000000052] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Adoptive transfer of T cells expressing chimeric antigen receptors (CARs) has shown promising antitumor activity in early phase clinical studies, especially for hematological malignancies. However, most preclinical models do not reliably mimic human disease. We reasoned that developing an adoptive T-cell therapy approach for spontaneous osteosarcoma (OS) occurring in dogs would more closely reproduce the condition in human cancer. To generate CAR-expressing canine T cells, we developed expansion and transduction protocols that allow for the generation of sufficient numbers of CAR-expressing canine T cells for future clinical studies in dogs within 2 weeks of ex vivo culture. To evaluate the functionality of CAR-expressing canine T cells, we targeted HER2(+) OS. We demonstrate that canine OS is positive for HER2, and that canine T cells expressing a HER2-specific CAR with human-derived transmembrane and CD28.ζ signaling domains recognize and kill HER2(+) canine OS cell lines in an antigen-dependent manner. To reduce the potential immunogenicity of the CAR, we evaluated a CAR with canine-derived transmembrane and signaling domains, and found no functional difference between human and canine CARs. Hence, we have successfully developed a strategy to generate CAR-expressing canine T cells for future preclinical studies in dogs. Testing T-cell therapies in an immunocompetent, outbred animal model may improve our ability to predict their safety and efficacy before conducting studies in humans.
Collapse
|
64
|
Barbieri F, Thellung S, Ratto A, Carra E, Marini V, Fucile C, Bajetto A, Pattarozzi A, Würth R, Gatti M, Campanella C, Vito G, Mattioli F, Pagano A, Daga A, Ferrari A, Florio T. In vitro and in vivo antiproliferative activity of metformin on stem-like cells isolated from spontaneous canine mammary carcinomas: translational implications for human tumors. BMC Cancer 2015; 15:228. [PMID: 25884842 PMCID: PMC4397725 DOI: 10.1186/s12885-015-1235-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/20/2015] [Indexed: 12/21/2022] Open
Abstract
Background Cancer stem cells (CSCs) are considered the cell subpopulation responsible for breast cancer (BC) initiation, growth, and relapse. CSCs are identified as self-renewing and tumor-initiating cells, conferring resistance to chemo- and radio-therapy to several neoplasias. Nowadays, th (about 10mM)e pharmacological targeting of CSCs is considered an ineludible therapeutic goal. The antidiabetic drug metformin was reported to suppress in vitro and in vivo CSC survival in different tumors and, in particular, in BC preclinical models. However, few studies are available on primary CSC cultures derived from human postsurgical BC samples, likely because of the limited amount of tissue available after surgery. In this context, comparative oncology is acquiring a relevant role in cancer research, allowing the analysis of larger samples from spontaneous pet tumors that represent optimal models for human cancer. Methods Isolation of primary canine mammary carcinoma (CMC) cells and enrichment in stem-like cell was carried out from fresh tumor specimens by culturing cells in stem-permissive conditions. Phenotypic and functional characterization of CMC-derived stem cells was performed in vitro, by assessment of self-renewal, long-lasting proliferation, marker expression, and drug sensitivity, and in vivo, by tumorigenicity experiments. Corresponding cultures of differentiated CMC cells were used as internal reference. Metformin efficacy on CMC stem cell viability was analyzed both in vitro and in vivo. Results We identified a subpopulation of CMC cells showing human breast CSC features, including expression of specific markers (i.e. CD44, CXCR4), growth as mammospheres, and tumor-initiation in mice. These cells show resistance to doxorubicin but were highly sensitive to metformin in vitro. Finally, in vivo metformin administration significantly impaired CMC growth in NOD-SCID mice, associated with a significant depletion of CSCs. Conclusions Similarly to the human counterpart, CMCs contain stem-like subpopulations representing, in a comparative oncology context, a valuable translational model for human BC, and, in particular, to predict the efficacy of antitumor drugs. Moreover, metformin represents a potential CSC-selective drug for BC, as effective (neo-)adjuvant therapy to eradicate CSC in mammary carcinomas of humans and animals. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1235-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Federica Barbieri
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy. .,Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genoa, Italy.
| | - Stefano Thellung
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy. .,Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genoa, Italy.
| | - Alessandra Ratto
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Elisa Carra
- Dipartimento di Medicina Sperimentale, University of Genova, Genoa, Italy.
| | - Valeria Marini
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Carmen Fucile
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Adriana Bajetto
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Alessandra Pattarozzi
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Roberto Würth
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Monica Gatti
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Chiara Campanella
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Guendalina Vito
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Francesca Mattioli
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Aldo Pagano
- Dipartimento di Medicina Sperimentale, University of Genova, Genoa, Italy. .,IRCCS AOU San Martino - IST, Genoa, Italy.
| | | | - Angelo Ferrari
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Tullio Florio
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy. .,Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genoa, Italy.
| |
Collapse
|
65
|
Jensen-Jarolim E, Fazekas J, Singer J, Hofstetter G, Oida K, Matsuda H, Tanaka A. Crosstalk of carcinoembryonic antigen and transforming growth factor-β via their receptors: comparing human and canine cancer. Cancer Immunol Immunother 2015; 64:531-7. [PMID: 25832000 PMCID: PMC4412651 DOI: 10.1007/s00262-015-1684-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/19/2015] [Indexed: 01/27/2023]
Abstract
There is accumulating evidence that the transforming growth factor beta (TGF-β) and nuclear factor kappa-B (NFκB) pathways are tightly connected and play a key role in malignant transformation in cancer. Immune infiltration by regulatory T- and B-lymphocytes (Tregs, Bregs) has recently gained increased attention for being an important source of TGF-β. There is a plethora of studies examining the pro-tumorigenic functions of carcinoembryonic antigen (CEA), but its receptor CEAR is far less studied. So far, there is a single connecting report that TGF-β also may signal through CEAR. The crosstalk between cancer tissues is further complicated by the expression of CEAR and TGF-β receptors in stromal cells, and implications of TGF-β in epithelial–mesenchymal transition. Furthermore, tumor-infiltrating Tregs and Bregs may directly instruct cancer cells by secreting TGF-β binding to their CEAR. Therefore, both TGF-β and CEA may act synergistically in breast cancer and cause disease progression, and NFκB could be a common crossing point between their signaling. CEAR, TGF-β1–3, TGF-β-R types I–III and NFκB class I and II molecules have an outstanding human–canine sequence identity, and only a canine CEA homolog has not yet been identified. For these reasons, the dog may be a valid translational model patient for investigating the crosstalk of the interconnected CEA and TGF-β networks.
Collapse
Affiliation(s)
- Erika Jensen-Jarolim
- Department of Comparative Medicine, Comparative Immunology and Oncology, Messerli Research Institute of the University of Veterinary Medicine Vienna, c/o Institute of Pathophysiology and Allergy Research, AKH 4Q, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria,
| | | | | | | | | | | | | |
Collapse
|
66
|
Abstract
The success of antibody therapy in cancer is consistent with the ability of these molecules to activate immune responses against tumors. Experience in clinical applications, antibody design, and advancement in technology have enabled antibodies to be engineered with enhanced efficacy against cancer cells. This allows re-evaluation of current antibody approaches dominated by antibodies of the IgG class with a new light. Antibodies of the IgE class play a central role in allergic reactions and have many properties that may be advantageous for cancer therapy. IgE-based active and passive immunotherapeutic approaches have been shown to be effective in both in vitro and in vivo models of cancer, suggesting the potential use of these approaches in humans. Further studies on the anticancer efficacy and safety profile of these IgE-based approaches are warranted in preparation for translation toward clinical application.
Collapse
Affiliation(s)
- Lai Sum Leoh
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, CHS 54-140, Box 951782, Los Angeles, CA 90095-1782, USA
| | - Tracy R. Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, CHS 54-140, Box 951782, Los Angeles, CA 90095-1782, USA
| | - Manuel L. Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, CHS 54-140, Box 951782, Los Angeles, CA 90095-1782, USA. Department of Microbiology, Immunology, and Molecular Genetics, University of California, 609 Charles E. Young Dr. East, 1602 Molecular Science Building, Los Angeles, CA 90095, USA. The Jonsson Comprehensive Cancer Center, University of California, 10833 Le Conte Ave, 8-684 Factor Building, Box 951781, Los Angeles, CA 90095, USA. The Molecular Biology Institute, University of California, 611 Charles E. Young Dr., Los Angeles, CA 90095, USA
| |
Collapse
|
67
|
Researcher of the month. Wien Klin Wochenschr 2015. [DOI: 10.1007/s00508-015-0699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
68
|
Singer J, Fazekas J, Wang W, Weichselbaumer M, Matz M, Mader A, Steinfellner W, Meitz S, Mechtcheriakova D, Sobanov Y, Willmann M, Stockner T, Spillner E, Kunert R, Jensen-Jarolim E. Generation of a canine anti-EGFR (ErbB-1) antibody for passive immunotherapy in dog cancer patients. Mol Cancer Ther 2014; 13:1777-1790. [PMID: 24755200 DOI: 10.1158/1535-7163.mct-13-0288] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Passive immunotherapy with monoclonal antibodies represents a cornerstone of human anticancer therapies, but has not been established in veterinary medicine yet. As the tumor-associated antigen EGFR (ErbB-1) is highly conserved between humans and dogs, and considering the effectiveness of the anti-EGFR antibody cetuximab in human clinical oncology, we present here a "caninized" version of this antibody, can225IgG, for comparative oncology studies. Variable region genes of 225, the murine precursor of cetuximab, were fused with canine constant heavy gamma and kappa chain genes, respectively, and transfected into Chinese hamster ovary (CHO) DUKX-B11 cells. Of note, 480 clones were screened and the best clones were selected according to productivity and highest specificity in EGFR-coated ELISA. Upon purification with Protein G, the recombinant cetuximab-like canine IgG was tested for integrity, correct assembly, and functionality. Specific binding to the surface of EGFR-overexpressing cells was assessed by flow cytometry and immunofluorescence; moreover, binding to canine mammary tissue was demonstrated by immunohistochemistry. In cell viability and proliferation assays, incubation with can225IgG led to significant tumor cell growth inhibition. Moreover, this antibody mediated significant tumor cell killing via phagocytosis in vitro. We thus present here, for the first time, the generation of a canine IgG antibody and its hypothetical structure. On the basis of its cetuximab-like binding site, on the one hand, and the expression of a 91% homologous EGFR molecule in canine cancer, on the other hand, this antibody may be a promising research compound to establish passive immunotherapy in dog patients with cancer.
Collapse
Affiliation(s)
- Josef Singer
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna
| | - Judit Fazekas
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna
- Department for Applied Life Sciences, University of Applied Sciences, FH Campus Wien
| | - Wei Wang
- Department of Immunology, Capital Medical University, Beijing, PR China
| | - Marlene Weichselbaumer
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna
| | - Miroslawa Matz
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
| | - Alexander Mader
- Department of Biotechnology, VIBT-BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Willibald Steinfellner
- Department of Biotechnology, VIBT-BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Sarah Meitz
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna
| | - Diana Mechtcheriakova
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
| | - Yuri Sobanov
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
| | - Michael Willmann
- Department for Companion Animals and Horses, University of Veterinary Medicine Vienna
| | - Thomas Stockner
- Centre for Physiology and Pharmacology, Medical University of Vienna
| | - Edzard Spillner
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Renate Kunert
- Department of Biotechnology, VIBT-BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Erika Jensen-Jarolim
- Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna
| |
Collapse
|
69
|
Singer J, Jensen‐Jarolim E. IgE-based immunotherapy of cancer: challenges and chances. Allergy 2014; 69:137-49. [PMID: 24117861 PMCID: PMC4022995 DOI: 10.1111/all.12276] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2013] [Indexed: 12/16/2022]
Abstract
Passive immunotherapy with monoclonal antibodies is an indispensable cornerstone of clinical oncology. Notably, all FDA-approved antibodies comprise the IgG class, although numerous research articles proposed monoclonal antibodies of the IgM, IgG, IgA and IgE classes directed specifically against tumor-associated antigens. In particular, for the IgE isotype class, several recent studies could demonstrate high tumoricidic efficacy. Therefore, this review specifically highlights the latest developments toward IgE-based immunotherapy of cancer. Possible mechanisms and safety aspects of IgE-mediated tumor cell death are discussed with special focus on the attracted immune cells. An outlook is given on how especially comparative oncology could contribute to further developments. Humans and dogs have a highly comparable IgE biology, suggesting that translational AllergoOncology studies in patients with canine cancer could have predictive value for the potential of IgE-based anticancer immunotherapy in human clinical oncology.
Collapse
Affiliation(s)
- J. Singer
- Comparative Immunology and Oncology Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - E. Jensen‐Jarolim
- Comparative Immunology and Oncology Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
- Comparative Medicine Messerli Research Institute of the University of Veterinary Medicine Vienna Medical University Vienna and University Vienna Vienna Austria
| |
Collapse
|
70
|
EGFR, HER-2 and KRAS in canine gastric epithelial tumors: a potential human model? PLoS One 2014; 9:e85388. [PMID: 24454858 PMCID: PMC3893207 DOI: 10.1371/journal.pone.0085388] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/26/2013] [Indexed: 12/26/2022] Open
Abstract
Epidermal growth factor receptor (EGFR or HER-1) and its analog c-erbB-2 (HER-2) are protein tyrosine kinases correlated with prognosis and response to therapy in a variety of human cancers. KRAS mediates the transduction of signals between EGFR and the nucleus, and its mutation has been identified as a predictor of resistance to anti-EGFR drugs. In human oncology, the importance of the EGFR/HER-2/KRAS signalling pathway in gastric cancer is well established, and HER-2 testing is required before initiating therapy. Conversely, this pathway has never been investigated in canine gastric tumours. A total of 19 canine gastric epithelial neoplasms (5 adenomas and 14 carcinomas) were retrospectively evaluated for EGFR/HER-2 immunohistochemical expression and KRAS mutational status. Five (35.7%) carcinomas were classified as intestinal-type and 9 (64.3%) as diffuse-type. EGFR was overexpressed (≥1+) in 8 (42.1%) cases and HER-2 (3+) in 11 (57.9%) cases, regardless of tumour location or biological behaviour. The percentage of EGFR-positive tumours was significantly higher in the intestinal-type (80%) than in the diffuse-type (11.1%, p = 0.023). KRAS gene was wild type in 18 cases, whereas one mucinous carcinoma harboured a point mutation at codon 12 (G12R). EGFR and HER-2 may be promising prognostic and therapeutic targets in canine gastric epithelial neoplasms. The potential presence of KRAS mutation should be taken into account as a possible mechanism of drug resistance. Further studies are necessary to evaluate the role of dog as a model for human gastric cancer.
Collapse
|
71
|
Common Concepts of Immune Defense. Comp Med 2014. [DOI: 10.1007/978-3-7091-1559-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
72
|
Abstract
Antibody-based immunotherapies are important therapy options in human oncology. Although human humoral specific immunity is constituted of five different immunoglobulin classes, currently only IgG-based immunotherapies have proceeded to clinical application. This review, however, discusses the benefits and difficulties of IgE-based immunotherapy of cancer, with special emphasis on how to translate promising preclinical results into clinical studies. Pursuing the “Comparative Oncology” approach, novel drug candidates are investigated in clinical trials with veterinary cancer patients, most often dogs. By this strategy drug development could be speeded up, animal experiments could be reduced and novel therapy options could be introduced benefitting humans as well as man’s best friend.
Collapse
Affiliation(s)
- Josef Singer
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, and University Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, and University Vienna, Vienna, Austria ; Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
73
|
Tyrosine kinase inhibitors (TKIs) in human and pet tumours with special reference to breast cancer: a comparative review. Crit Rev Oncol Hematol 2013; 88:293-308. [PMID: 23768779 DOI: 10.1016/j.critrevonc.2013.05.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 04/27/2013] [Accepted: 05/17/2013] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase receptors (TKRs) play a key role in tumour cell proliferation and survival since they are involved in endothelial cell activation leading to tumour neoangiogenesis. In particular, vascular endothelial growth factor receptors (VEGFRs), platelet-derived growth factor receptor (PDGFR), stem cell factor receptor (c-KitR), and colony-stimulating factor 1 (CSF-1) are overexpressed or constitutively activated in human and pet malignancies. A variety of small molecule inhibitors targeting specific tyrosine kinases (known as tyrosine kinase inhibitors or TKIs) have recently been approved, or are under investigation, for the treatment of human cancer. TKI application in animal cancer is however relatively recent. This review aims to illustrate the major aspects of tyrosine kinase dysfunctions, with special regard to human and animal cancer of the mammary gland, providing an update on the background of the anti-angiogenic and anti-neoplastic properties of TKIs in human and veterinary cancer.
Collapse
|
74
|
Lecoindre P, Bystricka M, Chevallier M, Peyron C. Gastric carcinoma associated with Menetrier's-like disease in a West Highland white terrier. J Small Anim Pract 2012; 53:714-8. [DOI: 10.1111/j.1748-5827.2012.01291.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- P. Lecoindre
- Clinique Vétérinaire des Cerisioz; 5 Route de Saint Symphorien d'Ozon; 69 800; Saint Priest; France
| | | | | | | |
Collapse
|
75
|
New therapeutic targets in veterinary oncology: man and dog definitely are best friends. Vet J 2012; 195:6-7. [PMID: 22795959 DOI: 10.1016/j.tvjl.2012.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/07/2012] [Indexed: 01/12/2023]
|