1
|
Jahandar-Lashaki S, Farajnia S, Faraji-Barhagh A, Hosseini Z, Bakhtiyari N, Rahbarnia L. Phage Display as a Medium for Target Therapy Based Drug Discovery, Review and Update. Mol Biotechnol 2024:10.1007/s12033-024-01195-6. [PMID: 38822912 DOI: 10.1007/s12033-024-01195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
Phage libraries are now amongst the most prominent approaches for the identification of high-affinity antibodies/peptides from billions of displayed phages in a specific library through the biopanning process. Due to its ability to discover potential therapeutic candidates that bind specifically to targets, phage display has gained considerable attention in targeted therapy. Using this approach, peptides with high-affinity and specificity can be identified for potential therapeutic or diagnostic use. Furthermore, phage libraries can be used to rapidly screen and identify novel antibodies to develop immunotherapeutics. The Food and Drug Administration (FDA) has approved several phage display-derived peptides and antibodies for the treatment of different diseases. In the current review, we provided a comprehensive insight into the role of phage display-derived peptides and antibodies in the treatment of different diseases including cancers, infectious diseases and neurological disorders. We also explored the applications of phage display in targeted drug delivery, gene therapy, and CAR T-cell.
Collapse
Affiliation(s)
- Samaneh Jahandar-Lashaki
- Medical Biotechnology Department, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajnia
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Aref Faraji-Barhagh
- Medical Biotechnology Department, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Hosseini
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Nasim Bakhtiyari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rahbarnia
- Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Tavarozzi R, Manzato E. The Role of Bispecific Antibodies in Non-Hodgkin's Lymphoma: From Structure to Prospective Clinical Use. Antibodies (Basel) 2022; 11:16. [PMID: 35225874 PMCID: PMC8883977 DOI: 10.3390/antib11010016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Abstract
Bispecific antibodies (bsAbs) are molecules that simultaneously bind two different antigens (Ags). bsAbs represent a very active field in tumor immunotherapy with more than one hundred molecules currently being tested. More specifically, they have elicited a great interest in the setting of non-Hodgkin's lymphoma (NHLs), where they could represent a viable option for more fragile patients or those resistant to other conventional therapies. This review aims to give a brief overview of the different available bsAb formats and their mechanisms of action, pinpointing the differences between IgG-like and non-IgG-like classes and will then focus on those in advanced clinical development for NHLs.
Collapse
Affiliation(s)
- Rita Tavarozzi
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
- SCDU of Hematology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Enrica Manzato
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy;
| |
Collapse
|
3
|
Kafil V, Saei AA, Tohidkia MR, Barar J, Omidi Y. Immunotargeting and therapy of cancer by advanced multivalence antibody scaffolds. J Drug Target 2020; 28:1018-1033. [DOI: 10.1080/1061186x.2020.1772796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Vala Kafil
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ata Saei
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Kotani N, Yamaguchi A, Ohnishi T, Kuwahara R, Nakano T, Nakano Y, Ida Y, Murakoshi T, Honke K. Proximity proteomics identifies cancer cell membrane cis-molecular complex as a potential cancer target. Cancer Sci 2019; 110:2607-2619. [PMID: 31228215 PMCID: PMC6676139 DOI: 10.1111/cas.14108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer‐specific antigens expressed in the cell membrane have been used as targets for several molecular targeted strategies in the last 20 years with remarkable success. To develop more effective cancer treatments, novel targets and strategies for targeted therapies are needed. Here, we examined the cancer cell membrane‐resident “cis‐bimolecular complex” as a possible cancer target (cis‐bimolecular cancer target: BiCAT) using proximity proteomics, a technique that has attracted attention in the last 10 years. BiCAT were detected using a previously developed method termed the enzyme‐mediated activation of radical source (EMARS), to label the components proximal to a given cell membrane molecule. EMARS analysis identified some BiCAT, such as close homolog of L1 (CHL1), fibroblast growth factor 3 (FGFR3) and α2 integrin, which are commonly expressed in mouse primary lung cancer cells and human lung squamous cell carcinoma cells. Analysis of cancer specimens from 55 lung cancer patients revealed that CHL1 and α2 integrin were highly co–expressed in almost all cancer tissues compared with normal lung tissues. As an example of BiCAT application, in vitro simulation of effective drug combinations used for multiple drug treatment strategies was performed using reagents targeted to BiCAT molecules. The combination treatment based on BiCAT information moderately suppressed cancer cell proliferation compared with single administration, suggesting that the information about BiCAT in cancer cells is useful for the appropriate selection of the combination among molecular targeted reagents. Thus, BiCAT has the potential to contribute to several molecular targeted strategies in future.
Collapse
Affiliation(s)
- Norihiro Kotani
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Arisa Yamaguchi
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - Tomoko Ohnishi
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - Ryusuke Kuwahara
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Takanari Nakano
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Yuka Nakano
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Yui Ida
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | | | - Koichi Honke
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| |
Collapse
|
5
|
Vasilenko EA, Mokhonov VV, Gorshkova EN, Astrakhantseva IV. Bispecific Antibodies: Formats and Areas of Application. Mol Biol 2018. [DOI: 10.1134/s0026893318020176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
6
|
Zovko A, Novak M, Hååg P, Kovalerchick D, Holmlund T, Färnegårdh K, Ilan M, Carmeli S, Lewensohn R, Viktorsson K. Compounds from the marine sponge Cribrochalina vasculum offer a way to target IGF-1R mediated signaling in tumor cells. Oncotarget 2018; 7:50258-50276. [PMID: 27384680 PMCID: PMC5226581 DOI: 10.18632/oncotarget.10361] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/15/2016] [Indexed: 11/25/2022] Open
Abstract
In this work two acetylene alcohols, compound 1 and compound 2, which were isolated and identified from the sponge Cribrochalina vasculum, and which showed anti-tumor effects were further studied with respect to targets and action mechanisms. Gene expression analyses suggested insulin like growth factor receptor (IGF-1R) signaling to be instrumental in controlling anti-tumor efficacy of these compounds in non-small cell lung cancer (NSCLC). Indeed compounds 1 and 2 inhibited phosphorylation of IGF-1Rβ as well as reduced its target signaling molecules IRS-1 and PDK1 allowing inhibition of pro-survival signaling. In silico docking indicated that compound 1 binds to the kinase domain of IGF-1R at the same binding site as the well known tyrosine kinase inhibitor AG1024. Indeed, cellular thermal shift assay (CETSA) confirmed that C. vasculum compound 1 binds to IGF-1R but not to the membrane localized tyrosine kinase receptor EGFR. Importantly, we demonstrate that compound 1 causes IGF-1Rβ but not Insulin Receptor degradation specifically in tumor cells with no effects seen in normal diploid fibroblasts. Thus, these compounds hold potential as novel therapeutic agents targeting IGF-1R signaling for anti-tumor treatment.
Collapse
Affiliation(s)
- Ana Zovko
- Department of Oncology and Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden
| | - Metka Novak
- Department of Oncology and Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden
| | - Petra Hååg
- Department of Oncology and Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden
| | - Dimitry Kovalerchick
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Teresa Holmlund
- Department of Oncology and Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Färnegårdh
- Science for Life Laboratory, Drug Discovery and Development Platform, Department of Organic Chemistry, Stockholm University, Stockholm, Sweden
| | - Micha Ilan
- Department of Zoology, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shmuel Carmeli
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rolf Lewensohn
- Department of Oncology and Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Viktorsson
- Department of Oncology and Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
8
|
Alfaleh MA, Jones ML, Howard CB, Mahler SM. Strategies for Selecting Membrane Protein-Specific Antibodies using Phage Display with Cell-Based Panning. Antibodies (Basel) 2017; 6:E10. [PMID: 31548525 PMCID: PMC6698842 DOI: 10.3390/antib6030010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 12/14/2022] Open
Abstract
Membrane proteins are attractive targets for monoclonal antibody (mAb) discovery and development. Although several approved mAbs against membrane proteins have been isolated from phage antibody libraries, the process is challenging, as it requires the presentation of a correctly folded protein to screen the antibody library. Cell-based panning could represent the optimal method for antibody discovery against membrane proteins, since it allows for presentation in their natural conformation along with the appropriate post-translational modifications. Nevertheless, screening antibodies against a desired antigen, within a selected cell line, may be difficult due to the abundance of irrelevant organic molecules, which can potentially obscure the antigen of interest. This review will provide a comprehensive overview of the different cell-based phage panning strategies, with an emphasis placed on the optimisation of four critical panning conditions: cell surface antigen presentation, non-specific binding events, incubation time, and temperature and recovery of phage binders.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
9
|
Wang Y, Wu Q, Liu Z, Guo X, Zhou L, Wang Y, Song L, Wang N, Zheng Q, Wang W, Ren G, Li D. A recombinant IgG-like bispecific antibody acting as interleukin-1β and interleukin-17A inhibitor exhibits a promising efficacy for rheumatoid arthritis. Biomed Pharmacother 2017; 89:426-437. [DOI: 10.1016/j.biopha.2017.02.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 01/04/2023] Open
|
10
|
Verzijl D, Riedl T, Parren P, Gerritsen A. A novel label-free cell-based assay technology using biolayer interferometry. Biosens Bioelectron 2017; 87:388-395. [DOI: 10.1016/j.bios.2016.08.095] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/26/2016] [Accepted: 08/27/2016] [Indexed: 01/15/2023]
|
11
|
Ströhlein MA, Heiss MM, Jauch KW. The current status of immunotherapy in peritoneal carcinomatosis. Expert Rev Anticancer Ther 2016; 16:1019-27. [PMID: 27530056 DOI: 10.1080/14737140.2016.1224666] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Peritoneal carcinomatosis (PC) is a cancer disease with an urgent need for effective treatment. Conventional chemotherapy failed to show acceptable results. Cytoreductive surgery and hyperthermic chemoperfusion (HIPEC) are only beneficial in few patients with resectable peritoneal metastasis. Immunotherapy could be attractive against PC, as all requirements for immunotherapy are available in the peritoneal cavity. AREAS COVERED This review analyzes the present literature for immunotherapy of PC. Advances from immune stimulators, radionucleotide-conjugated- and bispecific antibodies to future developments like adoptive engineered T-cells with chimeric receptors are discussed. The clinical development of catumaxomab, which was the first intraperitoneal immunotherapy to be approved for clinical treatment, is discussed. The requirements for future developments are illustrated. Expert commentary: Immunotherapy of peritoneal carcinomatosis is manageable, showing striking cancer cell killing. Improved profiles of adverse events by therapy-induced cytokine release, enhanced specific killing and optimal treatment schedules within multimodal treatment will be key factors.
Collapse
Affiliation(s)
- Michael Alfred Ströhlein
- a Department of Abdominal, Vascular and Transplant Surgery, Cologne Merheim Medical Center , Witten/Herdecke University , Cologne , Germany
| | - Markus Maria Heiss
- a Department of Abdominal, Vascular and Transplant Surgery, Cologne Merheim Medical Center , Witten/Herdecke University , Cologne , Germany
| | - Karl-Walter Jauch
- b Medical Center of the Ludwig Maximilian University Munich , Munich , Germany
| |
Collapse
|
12
|
Feiner RC, Müller KM. Recent progress in protein-protein interaction study for EGFR-targeted therapeutics. Expert Rev Proteomics 2016; 13:817-32. [PMID: 27424502 DOI: 10.1080/14789450.2016.1212665] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Epidermal growth factor receptor (EGFR) expression is upregulated in many tumors and its aberrant signaling drives progression of many cancer types. Consequently, EGFR has become a clinically validated target as extracellular tumor marker for antibodies as well as for tyrosine kinase inhibitors. Within the last years, new mechanistic insights were uncovered and, based on clinical experience as well as progress in protein engineering, novel bio-therapeutic approaches were developed and tested. AREAS COVERED The potential therapeutic targeting arsenal in the fight against cancer now encompasses bispecific or biparatopic antibodies, DARPins, Adnectins, Affibodies, peptides and combinations of these binding molecules with viral- and nano-particles. We review past and recent binding proteins from the literature and include a brief description of the various targeting approaches. Special attention is given to the binding modes with the EGFR. Expert commentary: Clinical data from the three approved anti EGFR antibodies indicate that there is room for improved therapeutic efficacy. Having choices in size, affinity, avidity and the mode of EGFR binding as well as the possibility to combine various effector functions opens the possibility to rationally design more effective therapeutics.
Collapse
Affiliation(s)
- Rebecca Christine Feiner
- a Cellular and Molecular Biotechnology group, Faculty of Technology , Bielefeld University , Bielefeld , Germany
| | - Kristian Mark Müller
- a Cellular and Molecular Biotechnology group, Faculty of Technology , Bielefeld University , Bielefeld , Germany
| |
Collapse
|
13
|
Shen Y, Zeng L, Novosyadlyy R, Forest A, Zhu A, Korytko A, Zhang H, Eastman SW, Topper M, Hindi S, Covino N, Persaud K, Kang Y, Burtrum D, Surguladze D, Prewett M, Chintharlapalli S, Wroblewski VJ, Shen J, Balderes P, Zhu Z, Snavely M, Ludwig DL. A bi-functional antibody-receptor domain fusion protein simultaneously targeting IGF-IR and VEGF for degradation. MAbs 2016; 7:931-45. [PMID: 26073904 PMCID: PMC4623440 DOI: 10.1080/19420862.2015.1055442] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bi-specific antibodies (BsAbs), which can simultaneously block 2 tumor targets, have emerged as promising therapeutic alternatives to combinations of individual monoclonal antibodies. Here, we describe the engineering and development of a novel, human bi-functional antibody-receptor domain fusion molecule with ligand capture (bi-AbCap) through the fusion of the domain 2 of human vascular endothelial growth factor receptor 1 (VEGFR1) to an antibody directed against insulin-like growth factor - type I receptor (IGF-IR). The bi-AbCap possesses excellent stability and developability, and is the result of minimal engineering. Beyond potent neutralizing activities against IGF-IR and VEGF, the bi-AbCap is capable of cross-linking VEGF to IGF-IR, leading to co-internalization and degradation of both targets by tumor cells. In multiple mouse xenograft tumor models, the bi-AbCap improves anti-tumor activity over individual monotherapies. More importantly, it exhibits superior inhibition of tumor growth, compared with the combination of anti-IGF-IR and anti-VEGF therapies, via powerful blockade of both direct tumor cell growth and tumor angiogenesis. The unique "capture-for-degradation" mechanism of the bi-AbCap is informative for the design of next-generation bi-functional anti-cancer therapies directed against independent signaling pathways. The bi-AbCap design represents an alternative approach to the creation of dual-targeting antibody fusion molecules by taking advantage of natural receptor-ligand interactions.
Collapse
Affiliation(s)
- Yang Shen
- a Antibody Technology; Eli Lilly and Company ; New York , NY USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Schanzer JM, Wartha K, Moessner E, Hosse RJ, Moser S, Croasdale R, Trochanowska H, Shao C, Wang P, Shi L, Weinzierl T, Rieder N, Bacac M, Ries CH, Kettenberger H, Schlothauer T, Friess T, Umana P, Klein C. XGFR*, a novel affinity-matured bispecific antibody targeting IGF-1R and EGFR with combined signaling inhibition and enhanced immune activation for the treatment of pancreatic cancer. MAbs 2016; 8:811-27. [PMID: 26984378 PMCID: PMC4966845 DOI: 10.1080/19420862.2016.1160989] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) and the insulin-like growth factor-1 receptor (IGF-1R) play critical roles in tumor growth, providing a strong rationale for the combined inhibition of IGF-1R and EGFR signaling in cancer therapy. We describe the design, affinity maturation, in vitro and in vivo characterization of the bispecific anti-IGF-1R/EGFR antibody XGFR*. XGFR* is based on the bispecific IgG antibody XGFR, which enabled heterodimerization of an IGF-1R binding scFab heavy chain with an EGFR-binding light and heavy chain by the "knobs-into-holes" technology. XGFR* is optimized for monovalent binding of human EGFR and IGF-1R with increased binding affinity for IGF-1R due to affinity maturation and highly improved protein stability to oxidative and thermal stress. It bears an afucosylated Fc-portion for optimal induction of antibody-dependent cell-mediated cytotoxicity (ADCC). Stable Chinese hamster ovary cell clones with production yields of 2-3 g/L were generated, allowing for large scale production of the bispecific antibody. XGFR* potently inhibits EGFR- and IGF-1R-dependent receptor phosphorylation, reduces tumor cell proliferation in cells with heterogeneous levels of IGF-1R and EGFR receptor expression and induces strong ADCC in vitro. A comparison of pancreatic and colorectal cancer lines demonstrated superior responsiveness to XGFR*-mediated signaling and tumor growth inhibition in pancreatic cancers that frequently show a high degree of IGF-1R/EGFR co-expression. XGFR* showed potent anti-tumoral efficacy in the orthotopic MiaPaCa-2 pancreatic xenograft model, resulting in nearly complete tumor growth inhibition with significant number of tumor remissions. In summary, the bispecific anti-IGF-1R/EGFR antibody XGFR* combines potent signaling and tumor growth inhibition with enhanced ADCC induction and represents a clinical development candidate for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Juergen M Schanzer
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Katharina Wartha
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Ekkehard Moessner
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Ralf J Hosse
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Samuel Moser
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Rebecca Croasdale
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Halina Trochanowska
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Cuiying Shao
- c Pharma Research and Early Development, Roche Innovation Center Shanghai , Cai Lun Road, Shanghai , China
| | - Peng Wang
- c Pharma Research and Early Development, Roche Innovation Center Shanghai , Cai Lun Road, Shanghai , China
| | - Lei Shi
- c Pharma Research and Early Development, Roche Innovation Center Shanghai , Cai Lun Road, Shanghai , China
| | - Tina Weinzierl
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Natascha Rieder
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Marina Bacac
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Carola H Ries
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Hubert Kettenberger
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Tilman Schlothauer
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Thomas Friess
- a Roche Pharma Research and Early Development, Roche Innovation Center Munich , Nonnenwald, Penzberg , Germany
| | - Pablo Umana
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| | - Christian Klein
- b Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Wagistrasse, Schlieren , Switzerland
| |
Collapse
|
15
|
Huang WC, Burnouf PA, Su YC, Chen BM, Chuang KH, Lee CW, Wei PK, Cheng TL, Roffler SR. Engineering Chimeric Receptors To Investigate the Size- and Rigidity-Dependent Interaction of PEGylated Nanoparticles with Cells. ACS NANO 2016; 10:648-662. [PMID: 26741147 DOI: 10.1021/acsnano.5b05661] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Attachment of ligands to the surface of nanoparticles (NPs) is an attractive approach to target specific cells and increase intracellular delivery of nanocargos. To expedite investigation of targeted NPs, we engineered human cancer cells to express chimeric receptors that bind polyethylene glycol (PEG) and internalize stealth NPs in a fashion similar to ligand-targeted liposomes against epidermal growth factor receptor 1 or 2 (HER1 or HER2), which are validated targets for cancer therapy. Measurement of the rate of endocytosis and lysosomal accumulation of small (80-94 nm) or large (180-220 nm) flexible liposomes or more rigid lipid-coated mesoporous silica particles in human HT29 colon cancer and SKBR3 breast cancer cells that express chimeric receptors revealed that larger and more rigid NPs were internalized more slowly than smaller and more flexible NPs. An exception is when both the small and large liposomes underwent endocytosis via HER2. HER1 mediated faster and greater uptake of NPs into cells but retained NPs less well as compared to HER2. Lysosomal accumulation of NPs internalized via HER1 was unaffected by NP rigidity but was inversely related to NP size, whereas large rigid NPs internalized by HER2 displayed increased lysosomal accumulation. Our results provide insight into the effects of NP properties on receptor-mediated endocytosis and suggest that anti-PEG chimeric receptors may help accelerate investigation of targeted stealth NPs.
Collapse
Affiliation(s)
- Wei-Chiao Huang
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| | - Pierre-Alain Burnouf
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica , Taipei, Taiwan
| | - Yu-Cheng Su
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University , Taipei 110, Taiwan
| | - Chia-Wei Lee
- Research Center for Applied Sciences, Academia Sinica , Taipei 11529, Taiwan
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica , Taipei 11529, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical and Environmental Biology, Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University , Kaohsiung 80708, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Science, Academia Sinica , Taipei 11529, Taiwan
| |
Collapse
|
16
|
Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol 2015; 67:95-106. [DOI: 10.1016/j.molimm.2015.01.003] [Citation(s) in RCA: 417] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 12/21/2022]
|
17
|
Greillier L, Tomasini P, Barlesi F. Necitumumab for non-small cell lung cancer. Expert Opin Biol Ther 2015; 15:1231-9. [DOI: 10.1517/14712598.2015.1055243] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
18
|
Sacco PC, Maione P, Rossi A, Sgambato A, Casaluce F, Palazzolo G, Gridelli C. Necitumumab for the treatment of stage IV metastatic squamous non-small-cell lung cancer. Expert Rev Respir Med 2015; 9:245-54. [PMID: 25797462 DOI: 10.1586/17476348.2015.1027688] [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] [Indexed: 11/08/2022]
Abstract
Over the past two decades, progress in the treatment of patients with metastatic squamous non-small-cell lung cancer has been limited. The EGFR is involved in tumor progression and invasion and therefore it has become the target of several studies in lung cancer. Strategies to block this pathway are focused on the development of small molecule (tyrosine kinase inhibitor) and monoclonal antibodies (mAbs). Some mAbs have been studied in patients with advanced non-small-cell lung cancer. For the first time, a fully human immunoglobulin G (IMC-11F8), subclass 1 (IgG1) mAb targeting the EGFR, in combination with standard chemotherapy (cisplatin + gemcitabine), has been shown to increase overall survival in chemo-naïve patients with metastatic confirmed squamous cell histology.
Collapse
|
19
|
Deyev SM, Lebedenko EN, Petrovskaya LE, Dolgikh DA, Gabibov AG, Kirpichnikov MP. Man-made antibodies and immunoconjugates with desired properties: function optimization using structural engineering. RUSSIAN CHEMICAL REVIEWS 2015. [DOI: 10.1070/rcr4459] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Schanzer JM, Wartha K, Croasdale R, Moser S, Künkele KP, Ries C, Scheuer W, Duerr H, Pompiati S, Pollman J, Stracke J, Lau W, Ries S, Brinkmann U, Klein C, Umana P. A novel glycoengineered bispecific antibody format for targeted inhibition of epidermal growth factor receptor (EGFR) and insulin-like growth factor receptor type I (IGF-1R) demonstrating unique molecular properties. J Biol Chem 2014; 289:18693-706. [PMID: 24841203 DOI: 10.1074/jbc.m113.528109] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the present study, we have developed a novel one-arm single chain Fab heterodimeric bispecific IgG (OAscFab-IgG) antibody format targeting the insulin-like growth factor receptor type I (IGF-1R) and the epidermal growth factor receptor (EGFR) with one binding site for each target antigen. The bispecific antibody XGFR is based on the "knob-into-hole" technology for heavy chain heterodimerization with one heavy chain consisting of a single chain Fab to prevent wrong pairing of light chains. XGFR was produced with high expression yields and showed simultaneous binding to IGF-1R and EGFR with high affinity. Due to monovalent binding of XGFR to IGF-1R, IGF-1R internalization was strongly reduced compared with the bivalent parental antibody, leading to enhanced Fc-mediated cellular cytotoxicity. To further increase immune effector functions triggered by XGFR, the Fc portion of the bispecific antibody was glycoengineered, which resulted in strong antibody-dependent cell-mediated cytotoxicity activity. XGFR-mediated inhibition of IGF-1R and EGFR phosphorylation as well as A549 tumor cell proliferation was highly effective and was comparable with a combined treatment with EGFR (GA201) and IGF-1R (R1507) antibodies. XGFR also demonstrated potent anti-tumor efficacy in multiple mouse xenograft tumor models with a complete growth inhibition of AsPC1 human pancreatic tumors and improved survival of SCID beige mice carrying A549 human lung tumors compared with treatment with antibodies targeting either IGF-1R or EGFR. In summary, we have applied rational antibody engineering technology to develop a heterodimeric OAscFab-IgG bispecific antibody, which combines potent signaling inhibition with antibody-dependent cell-mediated cytotoxicity induction and results in superior molecular properties over two established tetravalent bispecific formats.
Collapse
Affiliation(s)
| | | | | | - Samuel Moser
- Roche Glycart AG, CH-8952 Schlieren, Switzerland, and
| | | | | | | | - Harald Duerr
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Sandra Pompiati
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Jan Pollman
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Jan Stracke
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Wilma Lau
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Stefan Ries
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Ulrich Brinkmann
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | | | - Pablo Umana
- Roche Glycart AG, CH-8952 Schlieren, Switzerland, and
| |
Collapse
|
21
|
Abstract
ABSTRACT
Advanced molecular biology techniques developed during the past few decades have allowed the industry to exploit and commercialize the natural defense mechanisms that antibodies provide. This review discusses the latest advances in antibody-engineering technologies to enhance clinical efficacy and outcomes. For the constant regions, the choice of the antibody class and isotype has to be made carefully to suit the therapeutic applications. Engineering of the Fc region, either by direct targeted mutagenesis or by modifying the nature of its
N
-glycan, has played an important role in recent years in increasing half-life or controlling effector functions. The variable regions of the antibody are responsible for binding affinity and exquisite specificity to the target molecule, which together with the Fc determine the drug's efficacy and influence the drug dose required to obtain the desired effectiveness. A key requirement during antibody development is therefore to affinity mature the variable regions when necessary, so that they bind the therapeutic target with sufficiently high affinity to guarantee effective occupancy over prolonged periods. If the antibody was obtained from a non-human source, such as rodents, a humanization process has to be applied to minimize immunogenicity while maintaining the desired binding affinity and selectivity. Finally, we discuss the next next-generation antibodies, such as antibody-drug conjugates, bispecific antibodies, and immunocytokines, which are being developed to meet future challenges.
Collapse
|
22
|
Gu J, Ghayur T. Rationale and development of multispecific antibody drugs. Expert Rev Clin Pharmacol 2014; 3:491-508. [DOI: 10.1586/ecp.10.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Wen PY, Kesari S, Drappatz J. Malignant gliomas: strategies to increase the effectiveness of targeted molecular treatment. Expert Rev Anticancer Ther 2014; 6:733-54. [PMID: 16759164 DOI: 10.1586/14737140.6.5.733] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, there has been increasing interest in the use of targeted molecular agents for the treatment of malignant gliomas. These agents are generally well tolerated but have demonstrated only modest activity. In this article, the current status of targeted molecular agents for malignant gliomas will be reviewed and strategies to improve their effectiveness will be discussed.
Collapse
Affiliation(s)
- Patrick Y Wen
- Harvard Medical School, Dana-Farber/Brigham and Women's Cancer Center, SW430D, Boston, MA 02115, USA.
| | | | | |
Collapse
|
24
|
A tale of two specificities: bispecific antibodies for therapeutic and diagnostic applications. Trends Biotechnol 2013; 31:621-32. [PMID: 24094861 PMCID: PMC7114091 DOI: 10.1016/j.tibtech.2013.08.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/01/2013] [Accepted: 08/27/2013] [Indexed: 12/20/2022]
Abstract
Recombinant DNA technologies are leading the rapid expansion of bispecific antibody formats. The therapeutic potential of bispecific antibodies is being realized through creative design. Bispecific antibodies are potentially underutilized reagents for diagnostics.
Artificial manipulation of antibody genes has facilitated the production of several unique recombinant antibody formats, which have highly important therapeutic and biotechnological applications. Although bispecific antibodies (bsAbs) are not new, they are coming to the forefront as our knowledge of the potential efficacy of antibody-based therapeutics expands. The next generation of bsAbs is developing due to significant improvements in recombinant antibody technologies. This review focuses on recent advances with a particular focus on improvements in format and design that are contributing to the resurgence of bsAbs, and in particular, on innovative structures applicable to next generation point-of-care (POC) devices with applicability to low resource environments.
Collapse
|
25
|
Castoldi R, Ecker V, Wiehle L, Majety M, Busl-Schuller R, Asmussen M, Nopora A, Jucknischke U, Osl F, Kobold S, Scheuer W, Venturi M, Klein C, Niederfellner G, Sustmann C. A novel bispecific EGFR/Met antibody blocks tumor-promoting phenotypic effects induced by resistance to EGFR inhibition and has potent antitumor activity. Oncogene 2013; 32:5593-601. [PMID: 23812422 PMCID: PMC3898114 DOI: 10.1038/onc.2013.245] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 02/08/2023]
Abstract
Simultaneous targeting of epidermal growth factor receptor (EGFR) and Met in cancer therapy is under pre-clinical and clinical evaluation. Here, we report the finding that treatment with EGFR inhibitors of various tumor cells, when stimulated with hepatocyte growth factor (HGF) and EGF, results in transient upregulation of phosphorylated AKT. Furthermore, EGFR inhibition in this setting stimulates a pro-invasive phenotype as assessed in Matrigel-based assays. Simultaneous treatment with AKT and EGFR inhibitors abrogates this invasive growth, hence functionally linking signaling and phenotype. This observation implies that during treatment of tumors a balanced ratio of EGFR and Met inhibition is required. To address this, we designed a bispecific antibody targeting EGFR and Met, which has the advantage of a fixed 2:1 stoichiometry. This bispecific antibody inhibits proliferation in tumor cell cultures and co-cultures with fibroblasts in an additive manner compared with treatment with both single agents. In addition, cell migration assays reveal a higher potency of the bispecific antibody in comparison with the antibodies' combination at low doses. We demonstrate that the bispecific antibody inhibits invasive growth, which is specifically observed with cetuximab. Finally, the bispecific antibody potently inhibits tumor growth in a non-small cell lung cancer xenograft model bearing a strong autocrine HGF-loop. Together, our findings strongly support a combination treatment of EGFR and Met inhibitors and further evaluation of resistance mechanisms to EGFR inhibition in the context of active Met signaling.
Collapse
Affiliation(s)
- R Castoldi
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - V Ecker
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - L Wiehle
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - M Majety
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - R Busl-Schuller
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - M Asmussen
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - A Nopora
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - U Jucknischke
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - F Osl
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - S Kobold
- Division of Clinical Pharmacology, Department of Internal Medicine IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - W Scheuer
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - M Venturi
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - C Klein
- pRED, Roche Glycart AG, Schlieren, Switzerland
| | - G Niederfellner
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - C Sustmann
- Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
26
|
Ferguson R, Gallagher E, Cohen D, Tobin-Hess A, Alikhani N, Novosyadlyy R, Haddad N, Yakar S, LeRoith D. Hyperinsulinemia promotes metastasis to the lung in a mouse model of Her2-mediated breast cancer. Endocr Relat Cancer 2013; 20:391-401. [PMID: 23572162 PMCID: PMC4093836 DOI: 10.1530/erc-12-0333] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Her2 oncogene is expressed in ∼25% of human breast cancers and is associated with metastatic progression and poor outcome. Epidemiological studies report that breast cancer incidence and mortality rates are higher in women with type 2 diabetes. Here, we use a mouse model of Her2-mediated breast cancer on a background of hyperinsulinemia to determine how elevated circulating insulin levels affect Her2-mediated primary tumor growth and lung metastasis. Hyperinsulinemic (MKR(+/+)) mice were crossed with doxycycline-inducible Neu-NT (MTB/TAN) mice to produce the MTB/TAN/MKR(+/+) mouse model. Both MTB/TAN and MTB/TAN/MKR(+/+) mice were administered doxycycline in drinking water to induce Neu-NT mammary tumor formation. In tumor tissues removed at 2, 4, and 6 weeks of Neu-NT overexpression, we observed increased tumor mass and higher phosphorylation of the insulin receptor/IGF1 receptor, suggesting that activation of these receptors in conditions of hyperinsulinemia could contribute to the increased growth of mammary tumors. After 12 weeks on doxycycline, although no further increase in tumor weight was observed in MTB/TAN/MKR(+/+) compared with MTB/TAN mice, the number of lung metastases was significantly higher in MTB/TAN/MKR(+/+) mice compared with controls (MTB/TAN/MKR(+/+) 16.41±4.18 vs MTB/TAN 5.36±2.72). In tumors at the 6-week time point, we observed an increase in vimentin, a cytoskeletal protein and marker of mesenchymal cells, associated with epithelial-to-mesenchymal transition and cancer-associated fibroblasts. We conclude that hyperinsulinemia in MTB/TAN/MKR(+/+) mice resulted in larger primary tumors, with more mesenchymal cells and therefore more aggressive tumors with more numerous pulmonary metastases.
Collapse
|
27
|
The Development of Bispecific Hexavalent Antibodies as a Novel Class of DOCK-AND-LOCKTM (DNLTM) Complexes. Antibodies (Basel) 2013. [DOI: 10.3390/antib2020353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
28
|
Way J, Super M. The potential for cancer combination therapy with multi-targeted, single-protein pharmaceuticals. Expert Opin Drug Discov 2013; 3:147-52. [PMID: 23480218 DOI: 10.1517/17460441.3.2.147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Many diseases and disorders are best treated by a combination of drugs. Unlike small molecules, engineered proteins can be designed to incorporate multiple independent drug activities. Compared with a combination of two or more proteins, the potential benefits of a single biologic that inhibits multiple targets may include lower cost, simplified clinical testing and greater patient convenience. The evolution of HIV combination therapy is a useful point of comparison, as it occurred in an unusual regulatory environment that allowed the testing of combinations when the clinical benefit of component drugs was unproven. The epidermal growth factor receptor family of cancer targets illustrates how a particular single-molecule combination therapy might be used for cancer therapy, so some attempts to construct single-protein agents with multiple activities that include anti-EGFR moieties are reviewed. Protein engineers have created an armory of multiply-targeted antibody derivatives, but such engineered molecules often have a shorter serum half-life than IgG antibodies. New protein engineering approaches may be needed to address this problem. Nonetheless, multiply-targeted single-protein agents may be an economical solution to the problem of antibody combination therapy for cancer.
Collapse
Affiliation(s)
- Jeffrey Way
- EMD Serono Lexigen Research Center, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | | |
Collapse
|
29
|
Abstract
Monoclonal antibodies (mAbs) have become a major class of therapeutic agents providing effective alternatives to treating various human diseases. To date, 15 mAbs have been approved by regulatory agencies in the world for clinical use in oncology indications. The selectivity and specificity, the unique pharmacokinetics, and the ability to engage and activate the host immune system differentiate these biologics from traditional small molecule anticancer drugs. mAb-based regimens have brought clinical benefits, including improvements in overall survival, to patients with a variety of cancers. Many challenges still remain, however, to fully realize the potential of these new medicines. With our further understanding of cancer biology, mechanism of antibody action, and advancement of antibody engineering technologies, many novel antibody formats or antibody-derived molecules are emerging as promising new generation therapeutics. Carefully designed and engineered, they retain the advantage of specificity and selectivity of original antibodies, but in the meantime acquire additional special features such as improved pharmacokinetics, increased selectivity, and enhanced anticancer efficacy. Promising clinical results are being generated with these newly improved antibody-based therapeutics.
Collapse
Affiliation(s)
- Zhenping Zhu
- Kadmon Pharmaceuticals, New York, New York 10016, USA.
| | | |
Collapse
|
30
|
May C, Sapra P, Gerber HP. Advances in bispecific biotherapeutics for the treatment of cancer. Biochem Pharmacol 2012; 84:1105-12. [PMID: 22858161 DOI: 10.1016/j.bcp.2012.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 12/16/2022]
Abstract
Conventional monoclonal antibody (mAb) therapeutics interfering with cellular signaling of their respective target antigens are frequently limited in their ability to induce significant anti-tumor activities when administered as single agents in patients with solid tumors. To overcome these limitations, several new technologies are being developed to empower biotherapeutics and to improve their anti-tumor activities, while maintaining their high tumor selectivity and superior safety profiles. The various efficacy enhancement technologies developed for mAbs can be divided broadly into two categories: First, technologies that improve the intrinsic anti-tumor activities of conventional immunoglobulin mAb formats, including the enhancement of effector cell functions and modulations of target binding properties, including interference with multiple signaling pathways. The second category of empowered biologics combines complementary anti-tumor modalities independent of the IgG format, including antibody drug conjugates (ADCs). In addition, bispecific compounds designed to recruit different subsets of inflammatory cells to the tumor environment, also belong to the mechanistic complementation strategy. This approach termed redirected immune cell killing, belongs to one the most promising new biotherapeutic platforms developed in oncology. Over 20 bispecific compounds are currently being developed pre-clinically, and several compounds are undergoing early stage clinical trials. In this report, we review the progress made in the development of bispecific biotherapeutics in the context of ADCs, redirected T- and B-cell killing and targeting of multiple signaling pathways. We also discuss the status of the clinical development of this class of compounds in oncology and the promises and challenges this field is currently facing.
Collapse
Affiliation(s)
- Chad May
- Bioconjugate Discovery and Development, Oncology Research Unit, Pfizer Worldwide Research and Development, 401 North Middletown Road, Pearl River, NY 10965, USA
| | | | | |
Collapse
|
31
|
Croasdale R, Wartha K, Schanzer JM, Kuenkele KP, Ries C, Mayer K, Gassner C, Wagner M, Dimoudis N, Herter S, Jaeger C, Ferrara C, Hoffmann E, Kling L, Lau W, Staack RF, Heinrich J, Scheuer W, Stracke J, Gerdes C, Brinkmann U, Umana P, Klein C. Development of tetravalent IgG1 dual targeting IGF-1R-EGFR antibodies with potent tumor inhibition. Arch Biochem Biophys 2012; 526:206-18. [PMID: 22464987 DOI: 10.1016/j.abb.2012.03.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 03/13/2012] [Accepted: 03/14/2012] [Indexed: 11/30/2022]
Abstract
In this study we present novel bispecific antibodies that simultaneously target the insulin-like growth factor receptor type I (IGF-1R) and epidermal growth factor receptor (EGFR). For this purpose disulfide stabilized scFv domains of the EGFR/ADCC antibody GA201 were fused via serine-glycine connectors to the C-terminus of the heavy (XGFR2) or light chain (XGFR4), or the N-termini of the light (XGFR5) or heavy chain (XGFR3) of the IGF-1R antibody R1507 as parental IgG1 antibody. The resulting bispecific IGF-1R-EGFR antibodies XGFR2, XGFR3 and XGFR4 were successfully generated with yields and stability comparable to conventional IgG1 antibodies. They effectively inhibited IGF-1R and EGFR phosphorylation and 3D proliferation of H322M and H460M2 tumor cells, induced strong down-modulation of IGF-1R as well as enhanced EGFR down-modulation compared to the parental EGFR antibody GA201 and were ADCC competent. The bispecific XGFR derivatives showed a strong format dependent influence of N- or C-terminal heavy and light chain scFv attachment on ADCC activity and an increase in receptor downregulation over the parental combination in vitro. XGFR2 and XGFR4 were selected for in vivo evaluation and showed potent anti-tumoral efficacy comparable to the combination of monospecific IGF-1R and EGFR antibodies in subcutaneous BxPC3 and H322M xenograft models. In summary, we have managed to overcome issues of stability and productivity of bispecific antibodies, discovered important antibody fusion protein design related differences on ADCC activity and receptor downmodulation and show that IGF-1R-EGFR antibodies represent an attractive therapeutic strategy to simultaneously target two key components de-regulated in multiple cancer types, with the ultimate goal to avoid the formation of resistance to therapy.
Collapse
Affiliation(s)
- Rebecca Croasdale
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, D-82372 Penzberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Monoclonal antibodies are widely used for the treatment of cancer, inflammatory and infectious diseases and other disorders. Most of the marketed antibodies are monospecific and therefore capable of interacting and interfering with a single target. However, complex diseases are often multifactorial in nature, and involve redundant or synergistic action of disease mediators or upregulation of different receptors, including crosstalk between their signaling networks. Consequently, blockade of multiple, different pathological factors and pathways may result in improved therapeutic efficacy. This result can be achieved by combining different drugs, or use of the dual targeting strategies applying bispecific antibodies that have emerged as an alternative to combination therapy. This review discusses the various dual targeting strategies for which bispecific antibodies have been developed and provides an overview of the established bispecific antibody formats.
Collapse
Affiliation(s)
- Roland E Kontermann
- Institut für Zellbiologie und Immunologie; Universität Stuttgart; Stuttgart, Germany
| |
Collapse
|
33
|
Grote M, Haas AK, Klein C, Schaefer W, Brinkmann U. Bispecific antibody derivatives based on full-length IgG formats. Methods Mol Biol 2012; 901:247-63. [PMID: 22723106 DOI: 10.1007/978-1-61779-931-0_16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Monoclonal antibodies have emerged as an effective therapeutic modality, and numerous antibodies have been approved for the treatment of several severe diseases or are currently in clinical development. To improve their therapeutic potential, monoclonal antibodies are constantly evolved by protein engineering. Particularly, the generation of bispecific antibodies raised special interest because of their ability to bind two different antigens at the same time, and the efficiency of these formats has been demonstrated in several clinical and preclinical studies. Up to now, the major drawbacks in using bispecific antibodies as a therapeutic agent have been difficult design and low-yield expression of homogeneous antibody populations. However, major technological improvements were made in protein engineering during the last years. This allows the design of several new IgG-based bispecific antibody formats that can be prepared in high yields and high homogeneity using conventional expression and purification techniques. Especially, recent development of IgG-fusions with disulfide-stabilized Fv fragments and of CrossMab-technologies facilitates the generation of bispecific antibodies with IgG-like architectures. Here we describe design principles and methods to express and purify different bispecific antibody formats derived from full-length IgGs.
Collapse
Affiliation(s)
- Michael Grote
- Large Molecule Research, Roche Pharma Research and Early Development, Penzberg, Germany
| | | | | | | | | |
Collapse
|
34
|
Muda M, Gross AW, Dawson JP, He C, Kurosawa E, Schweickhardt R, Dugas M, Soloviev M, Bernhardt A, Fischer D, Wesolowski JS, Kelton C, Neuteboom B, Hock B. Therapeutic assessment of SEED: a new engineered antibody platform designed to generate mono- and bispecific antibodies. Protein Eng Des Sel 2011; 24:447-54. [PMID: 21498564 DOI: 10.1093/protein/gzq123] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The strand-exchange engineered domain (SEED) platform was designed to generate asymmetric and bispecific antibody-like molecules, a capability that expands therapeutic applications of natural antibodies. This new protein engineered platform is based on exchanging structurally related sequences of immunoglobulin within the conserved CH3 domains. Alternating sequences from human IgA and IgG in the SEED CH3 domains generate two asymmetric but complementary domains, designated AG and GA. The SEED design allows efficient generation of AG/GA heterodimers, while disfavoring homodimerization of AG and GA SEED CH3 domains. Using a clinically validated antibody (C225), we tested whether Fab derivatives constructed on the SEED platform retain desirable therapeutic antibody features such as in vitro and in vivo stability, favorable pharmacokinetics, ligand binding and effector functions including antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. In addition, we tested SEED with combinations of binder domains (scFv, VHH, Fab). Mono- and bivalent Fab-SEED fusions retain full binding affinity, have excellent biochemical and biophysical stability, and retain desirable antibody-like characteristics conferred by Fc domains. Furthermore, SEED is compatible with different combinations of Fab, scFv and VHH domains. Our assessment shows that the new SEED platform expands therapeutic applications of natural antibodies by generating heterodimeric Fc-analog proteins.
Collapse
Affiliation(s)
- Marco Muda
- EMD Serono Research Institute, Inc, One Technology Place, Rockland, MA 02370, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bispecific digoxigenin-binding antibodies for targeted payload delivery. Proc Natl Acad Sci U S A 2011; 108:8194-9. [PMID: 21536919 DOI: 10.1073/pnas.1018565108] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bispecific antibodies that bind cell-surface targets as well as digoxigenin (Dig) were generated for targeted payload delivery. Targeting moieties are IgGs that bind the tumor antigens Her2, IGF1R, CD22, or LeY. A Dig-binding single-chain Fv was attached in disulfide-stabilized form to C termini of CH3 domains of targeting antibodies. Bispecific molecules were expressed in mammalian cells and purified in the same manner as unmodified IgGs. They are stable without aggregation propensity and retain binding specificity/affinity to cell-surface antigens and Dig. Digoxigeninylated payloads were generated that retain full functionality and can be complexed to bispecific antibodies in a defined 21 ratio. Payloads include small compounds (Dig-Cy5, Dig-Doxorubicin) and proteins (Dig-GFP). Complexed payloads are targeted by the bispecifics to cancer cells and because these complexes are stable in serum, they can be applied for targeted delivery. Because Dig bispecifics also effectively capture digoxigeninylated compounds under physiological conditions, separate administration of uncharged Dig bispecifics followed by application of Dig payload is sufficient to achieve antibody-mediated targeting in vitro and in vivo.
Collapse
|
36
|
Tu JH, Yu YH. Role of epidermal growth factor receptor inhibitors in the treatment of gastric carcinoma. Shijie Huaren Xiaohua Zazhi 2011; 19:1150-1155. [DOI: 10.11569/wcjd.v19.i11.1150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most common digestive system malignancies. Traditional chemoradiotherapy has modest efficacy in the treatment of gastric cancer. The epidermal growth factor receptor (EGFR) signal transduction pathway plays an important role in tumor proliferation, angiogenesis, invasion, and migration. EGFR inhibitors have been and are being developed to treat gastric carcinoma. In this paper, we review the role of EGFR inhibitors in the treatment of gastric carcinoma.
Collapse
|
37
|
Emanuel SL, Engle LJ, Chao G, Zhu RR, Cao C, Lin Z, Yamniuk AP, Hosbach J, Brown J, Fitzpatrick E, Gokemeijer J, Morin P, Morse BA, Carvajal IM, Fabrizio D, Wright MC, Das Gupta R, Gosselin M, Cataldo D, Ryseck RP, Doyle ML, Wong TW, Camphausen RT, Cload ST, Marsh HN, Gottardis MM, Furfine ES. A fibronectin scaffold approach to bispecific inhibitors of epidermal growth factor receptor and insulin-like growth factor-I receptor. MAbs 2011; 3:38-48. [PMID: 21099371 PMCID: PMC3038010 DOI: 10.4161/mabs.3.1.14168] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 11/09/2010] [Indexed: 11/19/2022] Open
Abstract
Engineered domains of human fibronectin (Adnectins™) were used to generate a bispecific Adnectin targeting epidermal growth factor receptor (EGFR) and insulin-like growth factor-I receptor (IGF-IR), two transmembrane receptors that mediate proliferative and survival cell signaling in cancer. Single-domain Adnectins that specifically bind EGFR or IGF-IR were generated using mRNA display with a library containing as many as 10 ( 13) Adnectin variants. mRNA display was also used to optimize lead Adnectin affinities, resulting in clones that inhibited EGFR phosphorylation at 7 to 38 nM compared to 2.6 μM for the parental clone. Individual, optimized, Adnectins specific for blocking either EGFR or IGF-IR signaling were engineered into a single protein (EI-Tandem Adnectin). The EI-Tandems inhibited phosphorylation of EGFR and IGF-IR, induced receptor degradation, and inhibited down-stream cell signaling and proliferation of human cancer cell lines (A431, H292, BxPC3 and RH41) with IC 50 values ranging from 0.1 to 113 nM. Although Adnectins bound to EGFR at a site distinct from those of anti-EGFR antibodies cetuximab, panitumumab and nimotuzumab, like the antibodies, the anti-EGFR Adnectins blocked the binding of EGF to EGFR. PEGylated EI-Tandem inhibited the growth of both EGFR and IGF-IR driven human tumor xenografts, induced degradation of EGFR, and reduced EGFR phosphorylation in tumors. These results demonstrate efficient engineering of bispecific Adnectins with high potency and desired specificity. The bispecificity may improve biological activity compared to monospecific biologics as tumor growth is driven by multiple growth factors. Our results illustrate a technological advancement for constructing multi-specific biologics in cancer therapy.
Collapse
Affiliation(s)
- Stuart L Emanuel
- Oncology Drug Discovery, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Bispecific antibodies, in contrast to conventional monoclonal antibodies, can bind simultaneously two different antigens. Taking advantage of this virtue, they are mostly designed for immune effector cell redirection to tumors and for radionuclide pretargeting to tumors. Bispecific antibodies of the first generation were produced by chemical cross-linking or cell-fusion technologies. More recently, the application of genetic engineering technologies gave rise to numerous formats of bispecific antibody fragments and whole IgG molecules. Because bispecific antibodies enable therapeutic strategies that are not possible with conventional monoclonal antibodies, they attract strong interest. Several bispecific antibody formats have already shown clinical efficacy in cancer patients, catalyzing efforts to translate the imaginative bispecific antibody concepts into effective therapies.
Collapse
Affiliation(s)
- Nurit Hollander
- Department of Human Microbiology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
39
|
Isolation of a human anti-epidermal growth factor receptor Fab antibody, EG-19-11, with subnanomolar affinity from naïve immunoglobulin repertoires using a hierarchical antibody library system. Immunol Lett 2010; 134:55-61. [DOI: 10.1016/j.imlet.2010.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 08/07/2010] [Accepted: 08/17/2010] [Indexed: 11/20/2022]
|
40
|
Li J, Zhu Z. Research and development of next generation of antibody-based therapeutics. Acta Pharmacol Sin 2010; 31:1198-207. [PMID: 20694021 DOI: 10.1038/aps.2010.120] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Monoclonal antibodies (mAb) are emerging as one of the major class of therapeutic agents in the treatment of many human diseases, in particular in cancer and immunological disorders. To date, 28 mAb have been approved by the United States Food and Drug Administration for clinical applications. In addition, several hundreds of mAb are being developed clinically by many biotech and pharmaceutical companies for various disease indications. Many challenges still remain, however, and the full potential of therapeutic antibodies has yet to be realized. With the advancement of antibody engineering technologies and our further understanding of disease biology as well as antibody mechanism of action, many classes of novel antibody formats or antibody derived molecules are emerging as promising new generation therapeutics. These new antibody formats or molecules are carefully designed and engineered to acquire special features, such as improved pharmacokinetics, increased selectivity, and enhanced efficacy. These new agents may have the potential to revolutionize both our thinking and practice in the efforts to research and develop next generation antibody-based therapeutics.
Collapse
|
41
|
Li R, Pourpak A, Morris SW. Inhibition of the insulin-like growth factor-1 receptor (IGF1R) tyrosine kinase as a novel cancer therapy approach. J Med Chem 2010; 52:4981-5004. [PMID: 19610618 DOI: 10.1021/jm9002395] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rongshi Li
- Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Oncologic Sciences, University of South Florida, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
42
|
Abstract
The development of therapeutic antibodies has evolved over the past decade into a mainstay of therapeutic options for patients with autoimmune and inflammatory diseases. Substantial advances in understanding the biology of human diseases have been made and tremendous benefit to patients has been gained with the first generation of therapeutic antibodies. The lessons learnt from these antibodies have provided the foundation for the discovery and development of future therapeutic antibodies. Here we review how key insights obtained from the development of therapeutic antibodies complemented by newer antibody engineering technologies are delivering a second generation of therapeutic antibodies with promise for greater clinical efficacy and safety.
Collapse
|
43
|
|
44
|
Deev SM, Labedenko EN. [Antibody engineering: barnase-barstar module as a molecular constructor]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2010; 35:761-78. [PMID: 20208576 DOI: 10.1134/s1068162009060041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Today, antibody engineering for clinical applications is a rapidly progressing field of science and a big business. The basic functions of an antibody can be spatially differentiated and attributed to various structural domains of a molecule. Therefore, each of them may be an object for engineering with the aim of using a definite antibody function. In this sense, the potential of antibodies is unique. In this article, recent achievements and current problems of antibody engineering are briefly reviewed. The main attention is focused on a molecular constructor that allows for obtaining, with the help of a versatile barnase-barstar module, mono- and multiva-lent miniantibodies and their derivatives with outlined properties.
Collapse
|
45
|
Mabry R, Lewis KE, Moore M, McKernan PA, Bukowski TR, Bontadelli K, Brender T, Okada S, Lum K, West J, Kuijper JL, Ardourel D, Franke S, Lockwood L, Vu T, Frank A, Appleby MW, Wolf A, Reardon B, Hamacher NB, Stevens B, Lewis P, Lewis KB, Gilbertson DG, Lantry M, Julien SH, Ostrander C, Chan C, Byrnes-Blake K, Brody J, Presnell S, Meengs B, Levin SD, Snavely M. Engineering of stable bispecific antibodies targeting IL-17A and IL-23. Protein Eng Des Sel 2009; 23:115-27. [DOI: 10.1093/protein/gzp073] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
46
|
Huang L, De Baetselier P, Beyaert R. Targeting the EGF receptor ectodomain in the context of cancer. Expert Opin Ther Targets 2009; 13:1347-61. [DOI: 10.1517/14728220903277264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
47
|
Vardatsikos G, Sahu A, Srivastava AK. The insulin-like growth factor family: molecular mechanisms, redox regulation, and clinical implications. Antioxid Redox Signal 2009; 11:1165-90. [PMID: 19014342 DOI: 10.1089/ars.2008.2161] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulin-like growth factor (IGF)-induced signaling networks are vital in modulating multiple fundamental cellular processes, such as cell growth, survival, proliferation, and differentiation. Aberrations in the generation or action of IGF have been suggested to play an important role in several pathological conditions, including metabolic disorders, neurodegenerative diseases, and multiple types of cancer. Yet the exact mechanism involved in the pathogenesis of these diseases by IGFs remains obscure. Redox pathways involving reactive oxygen species (ROS) and reactive nitrogen species (RNS) contribute to the pathogenetic mechanism of various diseases by modifying key signaling pathways involved in cell growth, proliferation, survival, and apoptosis. Furthermore, ROS and RNS have been demonstrated to alter IGF production and/or action, and vice versa, and thereby have the ability to modulate cellular functions, leading to clinical manifestations of diseases. In this review, we provide an overview on the IGF system and discuss the potential role of IGF-1/IGF-1 receptor and redox pathways in the pathophysiology of several diseases.
Collapse
Affiliation(s)
- George Vardatsikos
- Laboratory of Cell Signaling, Montreal Diabetes Research Center, Centre Hospitalier de l'Université de Montréal, Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
48
|
Abstract
In past years, both laboratory and early clinical studies have demonstrated that biospecific antibodies (BsAbs) may have significant potential application in cancer therapy either by targeting tumor cells with cytotoxic agents including effector cells, radionuclides, drugs, and toxins, or by simultaneously blocking two relevant tumor targets, that is, growth factor receptors, thus neutralizing multiple receptor activation and downstream signal transduction pathways. A major obstacle in the development of BsAb has been the difficulty of producing the materials in sufficient quality and quantity by traditional technologies such as the hybrid hybridoma and chemical conjugation methods. The development of IgG-like BsAbs as therapeutic agents will depend heavily on the advances made in the design of recombinant BsAb constructs (or formats) and production efficiency. Here we describe a recombinant method for the construction and production of a novel IgG-like BsAb molecule, using the variable domains of two fully human antibodies as the building blocks.
Collapse
|
49
|
Schmitz KR, Ferguson KM. Interaction of antibodies with ErbB receptor extracellular regions. Exp Cell Res 2008; 315:659-70. [PMID: 18992239 DOI: 10.1016/j.yexcr.2008.10.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 10/07/2008] [Accepted: 10/07/2008] [Indexed: 12/11/2022]
Abstract
Antibodies to the extracellular region of the ErbB receptors have played key roles in the development of a mechanistic understanding of this family of receptor tyrosine kinases. An extensively studied class of such antibodies inhibits activation of ErbB receptors, and these antibodies have been the focus of intense development as anti-cancer agents. In this review we consider the properties of ErbB receptors antibodies in light of the current structure-based model for ErbB receptor homo- and hetero-dimerization and activation. Crystal structures of the Fab fragments from five different inhibitory antibodies in complex with the extracellular regions of EGFR and ErbB2 have been determined. These structures highlight several different modes of binding and mechanisms of receptor inhibition. Information about antibody interactions with the structurally well-characterized soluble extracellular regions of ErbB receptors can be combined with the rich knowledge of the effects of these antibodies in cultured cells, and in vivo, to provide insights into the conformation and activation of ErbB receptors at the cell surface.
Collapse
Affiliation(s)
- Karl R Schmitz
- Department of Physiology, University of Pennsylvania School of Medicine, B400 Richards Building, 3700 Hamilton Walk, Philadelphia, PA 19104-6085, USA
| | | |
Collapse
|
50
|
Diabody-based Recombinant Formats of Humanized IgG-like Bispecific Antibody With Effective Retargeting of Lymphocytes to Tumor Cells. J Immunother 2008; 31:752-61. [DOI: 10.1097/cji.0b013e3181849071] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|