1
|
Silva TA, Aguiar RB, Mori M, Machado GE, Hamaguchi B, Machado MFM, Moraes JZ. Potential of an anti-bevacizumab idiotype scFv DNA-based immunization to elicit VEGF-binding antibody response. Gene Ther 2023; 30:598-602. [PMID: 36482074 PMCID: PMC9734904 DOI: 10.1038/s41434-022-00376-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
Anti-idiotype antibodies have been considered for vaccination approaches against different diseases, including cancers. Based on that, we previously described an anti-bevacizumab idiotype monoclonal antibody, 10.D7, that revealed detectable antitumor effects on a vascular endothelial growth factor (VEGF)-dependent tumor model. Herein, we evaluated the possible applicability of a single-chain variable fragment (scFv) for the 10.D7 antibody in a gene immunization strategy. After checking that mammalian cells transfected to express the 10.D7 scFv are recognized by bevacizumab, it was explored the ability of our scFv construction, in a gene-based scheme, to elicit an immune response containing VEGF-binding antibodies. The results provide evidence that the designed 10.D7 scFv construct maintains the anti-bevacizumab idiotype features and has potential to activate an immune response recognizing VEGF.
Collapse
Affiliation(s)
- Tábata Almeida Silva
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rodrigo Barbosa Aguiar
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Marcelo Mori
- Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Gabriel Esquitini Machado
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Barbara Hamaguchi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Jane Zveiter Moraes
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
2
|
Orozco CT, Bersellini M, Irving LM, Howard WW, Hargreaves D, Devine PWA, Siouve E, Browne GJ, Bond NJ, Phillips JJ, Ravn P, Jackson SE. Mechanistic insights into the rational design of masked antibodies. MAbs 2022; 14:2095701. [PMID: 35799328 PMCID: PMC9272835 DOI: 10.1080/19420862.2022.2095701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Although monoclonal antibodies have greatly improved cancer therapy, they can trigger side effects due to on-target, off-tumor toxicity. Over the past decade, strategies have emerged to successfully mask the antigen-binding site of antibodies, such that they are only activated at the relevant site, for example, after proteolytic cleavage. However, the methods for designing an ideal affinity-based mask and what parameters are important are not yet well understood. Here, we undertook mechanistic studies using three masks with different properties and identified four critical factors: binding site and affinity, as well as association and dissociation rate constants, which also played an important role. HDX-MS was used to identify the location of binding sites on the antibody, which were subsequently validated by obtaining a high-resolution crystal structure for one of the mask-antibody complexes. These findings will inform future designs of optimal affinity-based masks for antibodies and other therapeutic proteins.
Collapse
Affiliation(s)
- Carolina T Orozco
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.,Biologics Engineering, R&D, AstraZeneca, Cambridge, UK.,Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | | | | | - Wesley W Howard
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Paul W A Devine
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Elise Siouve
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK.,Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | | | - Nicholas J Bond
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Peter Ravn
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK.,Department of Biotherapeutic Discovery, H. Lundbeck A/S, Copenhagen, Denmark
| | - Sophie E Jackson
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Adamson H, Nicholl A, Tiede C, Tang AA, Davidson A, Curd H, Wignall A, Ford R, Nuttall J, McPherson MJ, Johnson M, Tomlinson DC. Affimers as anti-idiotypic affinity reagents for pharmacokinetic analysis of biotherapeutics. Biotechniques 2019; 67:261-269. [DOI: 10.2144/btn-2019-0089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Therapeutic antibodies are the fastest growing class of drugs in the treatment of cancer, and autoimmune and inflammatory diseases that require the concomitant development of assays to monitor therapeutic antibody levels. Here, we demonstrate that the use of Affimer nonantibody binding proteins provides an advantage over current antibody-based detection systems. For four therapeutic antibodies, we used phage display to isolate highly specific anti-idiotypic Affimer reagents, which selectively bind to the therapeutic antibody idiotype. For each antibody target the calibration curves met US Food and Drug Administration criteria and the dynamic range compared favorably with commercially available reagents. Affimer proteins therefore represent promising anti-idiotypic reagents that are simple to select and manufacture, and that offer the sensitivity, specificity and consistency required for pharmacokinetic assays.
Collapse
Affiliation(s)
- Hope Adamson
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Amanda Nicholl
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Christian Tiede
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Anna A Tang
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Alex Davidson
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Helen Curd
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Alex Wignall
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Robert Ford
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - James Nuttall
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Michael J McPherson
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Matt Johnson
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Darren C Tomlinson
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
4
|
Arab A, Behravan N, Razazn A, Barati N, Mosaffa F, Nicastro J, Slavcev R, Behravan J. The viral approach to breast cancer immunotherapy. J Cell Physiol 2018; 234:1257-1267. [PMID: 30146692 DOI: 10.1002/jcp.27150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/05/2018] [Indexed: 01/03/2023]
Abstract
Despite years of intensive research, breast cancer remains the leading cause of death in women worldwide. New technologies including oncolytic virus therapies, virus, and phage display are among the most powerful and advanced methods that have emerged in recent years with potential applications in cancer prevention and treatment. Oncolytic virus therapy is an interesting strategy for cancer treatment. Presently, a number of viruses from different virus families are under laboratory and clinical investigation as oncolytic therapeutics. Oncolytic viruses (OVs) have been shown to be able to induce and initiate a systemic antitumor immune response. The possibility of application of a multimodal therapy using a combination of the OV therapy with immune checkpoint inhibitors and cancer antigen vaccination holds a great promise in the future of cancer immunotherapy. Display of immunologic peptides on bacterial viruses (bacteriophages) is also increasingly being considered as a new and strong cancer vaccine delivery strategy. In phage display immunotherapy, a peptide or protein antigen is presented by genetic fusions to the phage coat proteins, and the phage construct formulation acts as a protective or preventive vaccine against cancer. In our laboratory, we have recently tested a few peptides (E75, AE37, and GP2) derived from HER2/neu proto-oncogene as vaccine delivery modalities for the treatment of TUBO breast cancer xenograft tumors of BALB/c mice. Here, in this paper, we discuss the latest advancements in the applications of OVs and bacterial viruses display systems as new and advanced modalities in cancer immune therapeutics.
Collapse
Affiliation(s)
- Atefeh Arab
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Atefeh Razazn
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nastaran Barati
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jessica Nicastro
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.,Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Roderick Slavcev
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.,Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, ON, Canada.,Mediphage Bioceuticals, Inc., MaRS Centre, Toronto, ON, Canada
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Mediphage Bioceuticals, Inc., MaRS Centre, Toronto, ON, Canada
| |
Collapse
|
5
|
Abo-Aziza FAM, Zaki AA, El-Shemy A, Elhalem SSA, Amer AS. Immunological and histopathological changes in sheep affected with cutaneous squamous cell carcinoma and treated immunotherapeutically. Vet World 2017; 10:1094-1103. [PMID: 29062199 PMCID: PMC5639108 DOI: 10.14202/vetworld.2017.1094-1103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/29/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND AIM Recently, it has been recorded unexpected percentage of cutaneous squamous cell carcinoma (cSCC) in sheep. Despite the improvement in surgical treatment, the outcome of animals remains limited by metastatic relapse. Although antibodies for cancer treatment have been practiced for many decades, the use of this methodology in animals is deficient. This study aimed to establish cSCC therapy by tumor cell protein antibody (Ab1) or secondary antibody (Ab2) raised by two series of immunization in the same strain of rabbits. MATERIALS AND METHODS A total of 19 Ossimi sheep were used (14 sheep suffered from cSCC and 5 were apparently healthy). Each animal from control healthy group (n=5) and control cSCC (n=4) group was treated with a course of eight injections of normal globulins. Animals in the third (n=5) and the last (n=5) groups received a course of eight injections of Ab1and Ab2, respectively. Each tumor was measured before and after treatment. The eight injections were applied at 1st, 3rd, 5th, 7th, and 9th week and the remaining three injections were at 1 week interval. Tissue specimens and blood samples were taken for histological and immunological studies. RESULTS The obtained results revealed that injection of Ab1 might prevent the bad prognostic picture of polymorph infiltration without any criteria of regression % of tumor. Treatment with Ab2 showed regression of tumor size ranged between minimum of 8.99% and maximum of 78.12%, however, the measurements in most cases reached the maximum regression after the past two injections. In additions, infiltration of lymphocytes to tumor site, normalization of leukocytes picture and also increase of antibody titer were observed. CONCLUSION This profile might confirm that Ab2 could act as an antigen and encourage us to use it as a tumor vaccine. Extensive studies are needed to isolate the idiotypic portion of Ab1 for raising Ab2 as an anti-idiotypic antibody to be used as tumor vaccine. The question of how lymphocyte traffic to the tumor site as a result of Ab2 injection needs further investigation.
Collapse
Affiliation(s)
- Faten A. M. Abo-Aziza
- Department of Parasitology and Animal Diseases, Veterinary Research Division, National Research Centre, Cairo, Egypt
| | - A. A. Zaki
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - A. El-Shemy
- Department of Parasitology and Animal Diseases, Veterinary Research Division, National Research Centre, Cairo, Egypt
| | - Sahar S. Abd Elhalem
- Department of Zoology, Women Faculty for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Amany S. Amer
- Department of Zoology, Women Faculty for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| |
Collapse
|
6
|
Semi-synthetic vNAR libraries screened against therapeutic antibodies primarily deliver anti-idiotypic binders. Sci Rep 2017; 7:9676. [PMID: 28852148 PMCID: PMC5575089 DOI: 10.1038/s41598-017-10513-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023] Open
Abstract
Anti-idiotypic binders which specifically recognize the variable region of monoclonal antibodies have proven to be robust tools for pharmacokinetic studies of antibody therapeutics and for the development of cancer vaccines. In the present investigation, we focused on the identification of anti-idiotypic, shark-derived IgNAR antibody variable domains (vNARs) targeting the therapeutic antibodies matuzumab and cetuximab for the purpose of developing specific capturing ligands. Using yeast surface display and semi-synthetic, CDR3-randomized libraries, we identified several highly specific binders targeting both therapeutic antibodies in their corresponding variable region, without applying any counter selections during screening. Importantly, anti-idiotypic vNAR binders were not cross-reactive towards cetuximab or matuzumab, respectively, and comprised good target recognition in the presence of human and mouse serum. When coupled to magnetic beads, anti-idiotypic vNAR variants could be used as efficient capturing tools. Moreover, a two-step procedure involving vNAR-functionalized beads was employed for the enrichment of potentially bispecific cetuximab × matuzumab antibody constructs. In conclusion, semi-synthetic and CDR3-randomized vNAR libraries in combination with yeast display enable the fast and facile identification of anti-idiotypic vNAR domains targeting monoclonal antibodies primarily in an anti-idiotypic manner.
Collapse
|
7
|
Generation of anti-idiotype scFv for pharmacokinetic measurement in lymphoma patients treated with chimera anti-CD22 antibody SM03. PLoS One 2014; 9:e96697. [PMID: 24816427 PMCID: PMC4015983 DOI: 10.1371/journal.pone.0096697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 04/10/2014] [Indexed: 11/19/2022] Open
Abstract
Pre-clinical and clinical studies of therapeutic antibodies require highly specific reagents to examine their immune responses, bio-distributions, immunogenicity, and pharmacodynamics in patients. Selective antigen-mimicking anti-idiotype antibody facilitates the assessment of therapeutic antibody in the detection, quantitation and characterization of antibody immune responses. Using mouse specific degenerate primer pairs and splenocytic RNA, we generated an idiotype antibody-immunized phage-displayed scFv library in which an anti-idiotype antibody against the therapeutic chimera anti-CD22 antibody SM03 was isolated. The anti-idiotype scFv recognized the idiotype of anti-CD22 antibody and inhibited binding of SM03 to CD22 on Raji cell surface. The anti-idiotype scFv was subsequently classified as Ab2γ type. Moreover, our results also demonstrated firstly that the anti-idiotype scFv could be used for pharmacokinetic measurement of circulating residual antibody in lymphoma patients treated with chimera anti-CD22 monoclonal antibody SM03. Of important, the present approach could be easily adopted to generate anti-idiotype antibodies for therapeutic antibodies targeting membrane proteins, saving the cost and time for producing a soluble antigen.
Collapse
|
8
|
Ladjemi MZ. Anti-idiotypic antibodies as cancer vaccines: achievements and future improvements. Front Oncol 2012; 2:158. [PMID: 23133825 PMCID: PMC3490135 DOI: 10.3389/fonc.2012.00158] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 10/18/2012] [Indexed: 12/05/2022] Open
Abstract
Since the discovery of tumor-associated antigens (TAAs), researchers have tried to develop immune-based anti-cancer therapies. Thanks to their specificity, monoclonal antibodies (mAbs) offer the major advantage to induce fewer side effects than those caused by non-specific conventional treatments (e.g., chemotherapy, radiotherapy). Passive immunotherapy by means of mAbs or cytokines has proved efficacy in oncology and validated the use of immune-based agents as part of anti-cancer treatment options. The next step was to try to induce an active immune protection aiming to boost own’s host immune defense against TAAs. Cancer vaccines are thus developed to specifically induce active immune protection targeting only tumor cells while preserving normal tissues from a non-specific toxicity. But, as most of TAAs are self antigens, an immune tolerance against them exists representing a barrier to effective vaccination against these oncoproteins. One promising approach to break this immune tolerance consists in the use of anti-idiotypic (anti-Id) mAbs, so called Ab2, as antigen surrogates. This vaccination strategy allows also immunization against non-proteic antigens (such as carbohydrates). In some clinical studies, anti-Id cancer vaccines indeed induced efficient humoral and/or cellular immune responses associated with clinical benefit. This review article will focus on recent achievements of anti-Id mAbs use as cancer vaccines in solid tumors.
Collapse
Affiliation(s)
- Maha Z Ladjemi
- Institut de Recherche Expérimentale et Clinique, Pôle de Pneumologie, ORL and Dermatologie, Université Catholique de Louvain Brussels, Belgium
| |
Collapse
|
9
|
Antibody phage display libraries: contributions to oncology. Int J Mol Sci 2012; 13:5420-5440. [PMID: 22754305 PMCID: PMC3382779 DOI: 10.3390/ijms13055420] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/05/2012] [Accepted: 04/24/2012] [Indexed: 12/16/2022] Open
Abstract
Since the advent of phage display technology, dating back to 1985, antibody libraries displayed on filamentous phage surfaces have been used to identify specific binders for many different purposes, including the recognition of tumors. Phage display represents a high-throughput technique for screening billions of random fusion antibodies against virtually any target on the surface or inside cancer cells, or even soluble markers found in patient serum. Many phage display derived binders targeting important tumor markers have been identified. Selection directed to tumoral cells’ surfaces lead to the identification of unknown tumoral markers. Also the improvement of methods that require smaller amounts of cells has opened the possibility to use this approach on patient samples. Robust techniques combining an antibody library displayed on the phage surface and protein microarray allowed the identification of auto antibodies recognized by patient sera. Many Ab molecules directly or indirectly targeting angiogenesis have been identified, and one of them, ramucirumab, has been tested in 27 phase I–III clinical trials in a broad array of cancers. Examples of such antibodies will be discussed here with emphasis on those used as probes for molecular imaging and other clinical trials.
Collapse
|
10
|
Ladjemi MZ, Chardes T, Corgnac S, Garambois V, Morisseau S, Robert B, Bascoul-Mollevi C, Ait Arsa I, Jacot W, Pouget JP, Pelegrin A, Navarro-Teulon I. Vaccination with human anti-trastuzumab anti-idiotype scFv reverses HER2 immunological tolerance and induces tumor immunity in MMTV.f.huHER2(Fo5) mice. Breast Cancer Res 2011; 13:R17. [PMID: 21294885 PMCID: PMC3109586 DOI: 10.1186/bcr2826] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 01/18/2011] [Accepted: 02/04/2011] [Indexed: 01/13/2023] Open
Abstract
Introduction Novel adjuvant therapies are needed to prevent metastatic relapses in HER2-expressing breast cancer. Here, we tested whether trastuzumab-selected single-chain Fv (scFv) could be used to develop an anti-idiotype-based vaccine to inhibit growth of HER2-positive tumor cells in vitro and in vivo through induction of long-lasting HER-specific immunity. Methods BALB/c mice were immunized with anti-trastuzumab anti-idiotype (anti-Id) scFv (scFv40 and scFv69), which mimic human HER2. Their sera were assessed for the presence of HER2-specific Ab1' antibodies and for their ability to reduce viability of SK-OV-3 cells, a HER2-positive cancer cell line, in nude mice. MMTV.f.huHER2(Fo5) transgenic mice were immunized with scFv40 and scFv69 and, then, growth inhibition of spontaneous HER2-positive mammary tumors, humoral response, antibody isotype as well as splenocyte secretion of IL2 and IFN-γ were evaluated. Results Adoptively-transferred sera from BALB/c mice immunized with scFv40 and scFv69 contain anti-HER2 Ab1' antibodies that can efficiently inhibit growth of SK-OV-3 cell tumors in nude mice. Similarly, prophylactic vaccination with anti-Id scFv69 fully protects virgin or primiparous FVB-MMTV.f.huHER2(Fo5) females from developing spontaneous mammary tumors. Moreover, such vaccination elicits an anti-HER2 Ab1' immune response together with a scFv69-specific Th1 response with IL2 and IFN-γ cytokine secretion. Conclusions Anti-trastuzumab anti-Id scFv69, used as a therapeutic or prophylactic vaccine, protects mice from developing HER2-positive mammary tumors by inducing both anti-HER2 Ab1' antibody production and an anti-HER2 Th2-dependent immune response. These results suggest that scFv69 could be used as an anti-Id-based vaccine for adjuvant therapy of patients with HER2-positive tumors to reverse immunological tolerance to HER2.
Collapse
Affiliation(s)
- Maha Z Ladjemi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U896, Université Montpellier1, CRLC Val d'Aurelle Paul Lamarque, 208 rue des Apothicaires, Montpellier, F-34298, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Hearty S, Conroy PJ, Ayyar BV, Byrne B, O'Kennedy R. Surface plasmon resonance for vaccine design and efficacy studies: recent applications and future trends. Expert Rev Vaccines 2010; 9:645-64. [PMID: 20518719 DOI: 10.1586/erv.10.52] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The lack of a clear correlation between design and protection continues to present a barrier to progress in vaccine research. In this article, we outline how surface plasmon resonance (SPR) biosensors are emerging as tools to help resolve some of the key biophysical determinants of protection and, thereby, facilitate more rational vaccine design campaigns. SPR technology has contributed significantly to our understanding of the complex biophysical determinants of HIV neutralization and offers a platform for preclinical evaluation of vaccine candidates. In particular, the concept of reverse-engineering HIV vaccine targets based on known broadly neutralizing antibody modalities is explored and extended to include other infectious diseases, such as malaria and influenza, and other diseases such as cancer. The analytical capacity afforded by SPR includes serum screening to monitor immune responses and highly efficient quality-control surveillance measures. These are discussed alongside key technological advances, such as developments in sample throughput, and a perspective predicting continued growth and diversification of the role of SPR in vaccine development is proposed.
Collapse
Affiliation(s)
- Stephen Hearty
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland
| | | | | | | | | |
Collapse
|
12
|
Ladjemi MZ, Jacot W, Chardès T, Pèlegrin A, Navarro-Teulon I. Anti-HER2 vaccines: new prospects for breast cancer therapy. Cancer Immunol Immunother 2010; 59:1295-312. [PMID: 20532501 DOI: 10.1007/s00262-010-0869-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 05/11/2010] [Indexed: 12/24/2022]
Abstract
Each year, breast cancer accounts for more than 400,000 new cancer cases and more than 130,000 cancer deaths in Europe. Prognosis of nonmetastatic breast cancer patients is directly related to the extent of the disease, mainly nodal spreading and tumor size, and to the molecular profile, particularly HER2 over-expression. In patients with HER2-over-expressing tumors, different studies have shown cellular and/or humoral immune responses against HER2 associated with a lower tumor development at early stages of the disease. These findings have led to the hypothesis that the generation of an anti-HER2 immune response should protect patients from HER2-over-expressing tumor growth. Taken together with the clinical efficiency of trastuzumab-based anti-HER2 passive immunotherapy, these observations allowed to envisage various vaccine strategies against HER2. The induction of a stable and strong immunity by cancer vaccines is expected to lead to establishment of immune memory, thereby preventing tumor recurrence. However, an immunological tolerance against HER2 antigen exists representing a barrier to effective vaccination against this oncoprotein. As a consequence, the current challenge for vaccines is to find the best conditions to break this immunological tolerance. In this review, we will discuss the different anti-HER2 vaccine strategies currently developed; considering the strategies having reached the clinical phases as well as those still in preclinical development. The used antigen can be either composed of tumoral allogenic cells or autologous cells, or specific to HER2. It can be delivered by dendritic cells or in a DNA, peptidic or proteic form. Another area of research concerns the use of anti-idiotypic antibodies mimicking HER2.
Collapse
Affiliation(s)
- Maha Zohra Ladjemi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U896, Université Montpellier1, Montpellier, France
| | | | | | | | | |
Collapse
|
13
|
Immunogenicity and efficacy of a DNA vaccine encoding a human anti-idiotype single chain antibody against nasopharyngeal carcinoma. Vaccine 2010; 28:2769-74. [PMID: 20117260 DOI: 10.1016/j.vaccine.2010.01.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2009] [Revised: 01/14/2010] [Accepted: 01/16/2010] [Indexed: 11/23/2022]
Abstract
G22, an anti-idiotype single chain antibody screened from human nasopharyngeal carcinoma phage anti-idiotype antibody library, has been already identified by He et al. G22 DNA vaccine was produced by cloning G22 gene and inserting the cloned gene into pcDNA3.1. To investigate the immunogenicity of pcDNA3.1-G22, C57BL/6 mice were immunized with the vaccine, pcDNA3.1 and PBS individually and the antibody response, T cell phenocyte at the 15th, 22th, 29th, 36th day after the last immunity were detected. In the tumor protection experiment, the immunized mice were then challenged with CMT-93-G22 cells or CMT-93-mock cells. The tumor size and the survival time of the animals were compared between these groups. The results showed that DNA vaccine pcDNA3.1-G22 could raise G22-specific humoral and cellular immune responses. Furthermore, pcDNA3.1-G22 could prolong the survival time and lessen the tumor size of the CMT-93-G22-bearing mice but had no protection effect on the mice attacked by CMT-93-mock cells. These results were expected to lay foundation for further studies on the clinical application of pcDNA3.1-G22 DNA vaccine.
Collapse
|
14
|
A llama single domain anti-idiotypic antibody mimicking HER2 as a vaccine: Immunogenicity and efficacy. Vaccine 2009; 27:4826-33. [DOI: 10.1016/j.vaccine.2009.05.067] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/18/2009] [Accepted: 05/21/2009] [Indexed: 11/21/2022]
|
15
|
Ladjemi MZ, Jacot W, Pèlegrin A, Navarro-Teulon I. [Anti-HER2 vaccines: The HER2 immunotargeting future?]. ACTA ACUST UNITED AC 2009; 59:173-82. [PMID: 19481373 DOI: 10.1016/j.patbio.2009.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 04/03/2009] [Indexed: 11/17/2022]
Abstract
Breast cancer is a widely spread women's disease. In spite of progress in the field of surgery and adjuvant therapies, the risk of breast cancer metastatic relapses remains high especially in those overexpressing HER2. Different studies have shown cellular and/or humoral immune responses against HER2 in patients with HER2-overexpressing tumors. This immune response is associated with a lower tumor development at early stages of the disease. These observations, associated with the efficiency today demonstrated by a trastuzumab-based anti-HER2 immunotherapy, allowed to envisage various vaccinal strategies against HER2. These findings have so led to the hypothesis that the generation of an anti-HER2 immune response should protect patients from HER2-overexpressing tumor growth, and induction of a stable and strong immunity by cancer vaccines is expected to lead to establishment of immune memory, thereby preventing tumor recurrence. However, an immunological tolerance against HER2 antigen exists representing a barrier to effective vaccination against this oncoprotein. As a consequence, the current challenge for vaccines is to find the best conditions to break this immunological tolerance. In this review, we will discuss the different anti-HER2 vaccine strategies currently developed; considering the strategies having reached the clinical phases as well as those still in preclinical development. The used antigen can be composed of tumoral allogenic cells or autologous cells or be specific of HER2. It can be delivered by denditric cells or in a DNA, peptidic or proteic form. Another area of the research concerns the use of anti-idiotypic antibodies mimicking HER2.
Collapse
Affiliation(s)
- M Z Ladjemi
- Inserm U896, CRLC Val-d'Aurelle Paul-Lamarque, institut de recherche en cancérologie de Montpellier (IRCM), université Montpellier-1, 34298 Montpellier cedex 5, France.
| | | | | | | |
Collapse
|
16
|
Pignatari GC, Takeshita D, Parise CB, Soares FA, de Moraes JZ, Han SW. Carcinoembryonic antigen (CEA) mimicry by an anti-idiotypic scFv isolated from anti-Id 6.C4 hybridoma. J Biotechnol 2006; 127:615-25. [PMID: 16989916 DOI: 10.1016/j.jbiotec.2006.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 08/04/2006] [Accepted: 08/15/2006] [Indexed: 11/26/2022]
Abstract
Since carcinoembryonic antigen (CEA) is expressed during embryonic life, it is not immunogenic in humans. The use of anti-idiotypic (Id) antibodies as a surrogate of antigen in the immunization has been considered a promising strategy for breaking tolerance to some tumor associated antigens. We have described an anti-Id monoclonal antibody (MAb), designated 6.C4, which is able to mimic CEA functionally. The anti-Id MAb 6.C4 was shown to elicit antibodies that recognized CEA in vitro and in vivo. In the present study, we sought to verify whether a single chain (scFv) antibody obtained, the scFv 6.C4, would retain the ability to mimic CEA. Two scFv containing the variable heavy and light chain domains of 6.C4 were constructed with a 15-amino acid linker: one with and another without signal peptide. DNA immunization of mice with both forms of scFv individually elicited antibodies able to recognize CEA.
Collapse
|
17
|
Shen YC, Wang XH, Wang XM, Chen ZL, Shen XP, Zhao CC, Li J. High efficient mammalian expression and secretion of a functional humanized single-chain Fv/human interleukin-2 molecules. World J Gastroenterol 2006; 12:3859-65. [PMID: 16804971 PMCID: PMC4087934 DOI: 10.3748/wjg.v12.i24.3859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct and produce a recombinant bispecific humanized single-chain Fv (sFv) /Interleukin-2 (IL-2) fusion protein by using mammalian cells.
METHODS: The sFv/IL-2 protein was genetically engineered, and transfected to mammalian cells to determine whether the mammalian protein folding machinery can produce and secrete active sFv/IL-2 with high efficiency.
RESULTS: The fusion protein was constructed and high efficiently expressed with yields up to 102 ± 4.2 mg/L in culture supernatant of the stably transfected 293 cell line. This recombinant fusion protein consisted of humanized variable heavy (VH) and light (VL) domains of monoclonal antibody (mAb) 520C9 directed against the human HER-2/neu (c-erbB2) proto-oncogene product p185, and human IL-2 connected by polypeptide linker. The fusion protein was shown to retain the immunostimulatory activities of IL-2 as measured by IL-2-dependent cell proliferation and cytotoxicity assays. In addition to its IL-2 activities, this fusion protein also possessed antigen-binding specificity against p185, as determined by indirect ELISA using p185 positive SKOV 3ip1 cells.
CONCLUSION: The large-scale preparation of the recombinant humanized sFv antibody/IL-2 fusion protein is performed with 293 cells. The recombinant humanized sFv antibody/IL-2 fusion protein may provide an effective means of targeting therapeutic doses of IL-2 to p185 positive tumors without increasing systemic toxicity or immunogenicity.
Collapse
Affiliation(s)
- Yue-Chun Shen
- Division of Cardiology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, 48109, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Schwegler C, Dorn-Beineke A, Nittka S, Stocking C, Neumaier M. Monoclonal Anti-idiotype Antibody 6G6.C4 Fused to GM-CSF Is Capable of Breaking Tolerance to Carcinoembryonic Antigen (CEA) in CEA–Transgenic Mice. Cancer Res 2005; 65:1925-33. [PMID: 15753392 DOI: 10.1158/0008-5472.can-04-3591] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Internal image anti-idiotypic antibodies are capable of mimicking tumor-associated antigens and thus may serve as surrogate for vaccination strategies in cancer patients. The monoclonal antibody (mAb) 6G6.C4 mimics an epitope specific for the human carcinoembryonic antigen (CEA) and generates a CEA-specific response (Ab3) in various experimental animals. In humans, however, 6G6.C4 only yields a very limited humoral anti-CEA reaction presumably due to tolerance against the CEA autoantigen. In this study, we investigated the CEA-specific Ab3 response in mice transgenic for the human CEA and tested whether the antigen tolerance could be overcome by fusing a recombinant single-chain variable fragment of 6G6.C4 (scFv6G6.C4) to the murine granulocyte macrophage colony-stimulating factor (GM-CSF). Like mAb 6G6.C4, the fusion protein (scFv6G6.C4/GM-CSF) retained binding to the CEA-specific idiotype mAb T84.66. Also, scFv6G6.C4/GM-CSF was biologically active as measured by proliferation of the GM-CSF-dependent murine FDC-P1 cells in vitro. After immunization with the scFv6G6.C4/GM-CSF fusion protein, CEA-transgenic animals showed significantly enhanced Ab3 antibody responses to scFv6G6.C4 (P=0.005) and to CEA (P=0.012) compared with the scFV6G6.C4 alone. Sera from mice immunized with the fusion protein specifically recognized CEA in Western blot analyses with no cross-reaction to CEA-related antigens. Finally, the Ab3 antisera detected single CEA-expressing tumor cells in suspension as shown by flow cytometry. Taken together, these data show in a model antigenically related to the human system that vaccination with scFv6G6.C4/GM-CSF improves vaccination against an endogenous tumor-associated antigen resulting in a highly specific humoral Ab3 response in vivo that is capable of bind single circulating CEA-positive tumor cells.
Collapse
Affiliation(s)
- Christian Schwegler
- Department of Internal Medicine and Clinical Immunology Bad Bramstedt, University of Lübeck, Lübeck, Germany
| | | | | | | | | |
Collapse
|
19
|
Abstract
The year 2004 represents a milestone for the biosensor research community: in this year, over 1000 articles were published describing experiments performed using commercially available systems. The 1038 papers we found represent an approximately 10% increase over the past year and demonstrate that the implementation of biosensors continues to expand at a healthy pace. We evaluated the data presented in each paper and compiled a 'top 10' list. These 10 articles, which we recommend every biosensor user reads, describe well-performed kinetic, equilibrium and qualitative/screening studies, provide comparisons between binding parameters obtained from different biosensor users, as well as from biosensor- and solution-based interaction analyses, and summarize the cutting-edge applications of the technology. We also re-iterate some of the experimental pitfalls that lead to sub-optimal data and over-interpreted results. We are hopeful that the biosensor community, by applying the hints we outline, will obtain data on a par with that presented in the 10 spotlighted articles. This will ensure that the scientific community at large can be confident in the data we report from optical biosensors.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|