93 results on '"Mikkel W. Pedersen"'
Search Results
2. Lake Sedimentary DNA Research on Past Terrestrial and Aquatic Biodiversity: Overview and Recommendations
- Author
-
Eric Capo, Charline Giguet-Covex, Alexandra Rouillard, Kevin Nota, Peter D. Heintzman, Aurèle Vuillemin, Daniel Ariztegui, Fabien Arnaud, Simon Belle, Stefan Bertilsson, Christian Bigler, Richard Bindler, Antony G. Brown, Charlotte L. Clarke, Sarah E. Crump, Didier Debroas, Göran Englund, Gentile Francesco Ficetola, Rebecca E. Garner, Joanna Gauthier, Irene Gregory-Eaves, Liv Heinecke, Ulrike Herzschuh, Anan Ibrahim, Veljo Kisand, Kurt H. Kjær, Youri Lammers, Joanne Littlefair, Erwan Messager, Marie-Eve Monchamp, Fredrik Olajos, William Orsi, Mikkel W. Pedersen, Dilli P. Rijal, Johan Rydberg, Trisha Spanbauer, Kathleen R. Stoof-Leichsenring, Pierre Taberlet, Liisi Talas, Camille Thomas, David A. Walsh, Yucheng Wang, Eske Willerslev, Anne van Woerkom, Heike H. Zimmermann, Marco J. L. Coolen, Laura S. Epp, Isabelle Domaizon, Inger G. Alsos, and Laura Parducci
- Subjects
sedimentary ancient DNA ,sedimentary DNA ,lake sediments ,paleolimnology ,paleoecology ,paleogenetics ,Human evolution ,GN281-289 ,Stratigraphy ,QE640-699 - Abstract
The use of lake sedimentary DNA to track the long-term changes in both terrestrial and aquatic biota is a rapidly advancing field in paleoecological research. Although largely applied nowadays, knowledge gaps remain in this field and there is therefore still research to be conducted to ensure the reliability of the sedimentary DNA signal. Building on the most recent literature and seven original case studies, we synthesize the state-of-the-art analytical procedures for effective sampling, extraction, amplification, quantification and/or generation of DNA inventories from sedimentary ancient DNA (sedaDNA) via high-throughput sequencing technologies. We provide recommendations based on current knowledge and best practises.
- Published
- 2021
- Full Text
- View/download PDF
3. Food resources of the Khog Gzung site on the Tibetan Plateau revealed by sedimentary ancient DNA
- Author
-
Zhengquan Gu, Yu Gao, Yiru Wang, Jishuai Yang, Jingkun Ran, Xiaoyan Yang, Wangdue Shargan, Mikkel W. Pedersen, Guilian Sheng, Yucheng Wang, and Fahu Chen
- Subjects
General Earth and Planetary Sciences - Published
- 2023
4. Supplementary Figure Legend from Safety and Activity of the First-in-Class Sym004 Anti-EGFR Antibody Mixture in Patients with Refractory Colorectal Cancer
- Author
-
Josep Tabernero, Anthony W. Tolcher, Eric Van Cutsem, Stephan Braun, Ivan D. Horak, Michael Kragh, Mikkel W. Pedersen, Rikke Hald, Ulla H. Hansen, Niels J.Ø. Skartved, Guillem Argilés, Rachel S. van der Post, Bastiaan B.J. Tops, Susana Roselló, Marta Benavent, Andrés Cervantes, Rocio Garcia-Carbonero, Amita Patnaik, and Rodrigo Dienstmann
- Abstract
Figure legend to Supplementary Figure S1.
- Published
- 2023
5. Supplementary Figure 1 from Sym004, a Novel Anti-EGFR Antibody Mixture, Augments Radiation Response in Human Lung and Head and Neck Cancers
- Author
-
Paul M. Harari, Mikkel W. Pedersen, Michael Kragh, Eric A. Armstrong, Chunrong Li, Jarob Saker, Chimera R. Peet, and Shyhmin Huang
- Abstract
PDF - 189KB, Comparison between Sym004 and cetuximab on radiosensitivity response.
- Published
- 2023
6. Supplementary methods from Simultaneous Targeting of Two Distinct Epitopes on MET Effectively Inhibits MET- and HGF-Driven Tumor Growth by Multiple Mechanisms
- Author
-
Mikkel W. Pedersen, Morag Park, Thomas Bouquin, Johan Lantto, Michael Kragh, Ivan D. Horak, Helle J. Jacobsen, George F.Vande Woude, Dafna Kaufman, Karsten W. Eriksen, Sara Collins, Paolo Conrotto, Gunther R. Galler, Anna Dahlman, Klaus Koefoed, Thomas T. Poulsen, Serhiy Havrylov, and Michael M. Grandal
- Abstract
Supplementary methods
- Published
- 2023
7. Supplementary Table S2 from Safety and Activity of the First-in-Class Sym004 Anti-EGFR Antibody Mixture in Patients with Refractory Colorectal Cancer
- Author
-
Josep Tabernero, Anthony W. Tolcher, Eric Van Cutsem, Stephan Braun, Ivan D. Horak, Michael Kragh, Mikkel W. Pedersen, Rikke Hald, Ulla H. Hansen, Niels J.Ø. Skartved, Guillem Argilés, Rachel S. van der Post, Bastiaan B.J. Tops, Susana Roselló, Marta Benavent, Andrés Cervantes, Rocio Garcia-Carbonero, Amita Patnaik, and Rodrigo Dienstmann
- Abstract
Supplementary Table S2. Sym004 pharmacokinetic derived measures - dose-escalation and dose-expansion cohorts.
- Published
- 2023
8. Supplementary Figure S1 from Safety and Activity of the First-in-Class Sym004 Anti-EGFR Antibody Mixture in Patients with Refractory Colorectal Cancer
- Author
-
Josep Tabernero, Anthony W. Tolcher, Eric Van Cutsem, Stephan Braun, Ivan D. Horak, Michael Kragh, Mikkel W. Pedersen, Rikke Hald, Ulla H. Hansen, Niels J.Ø. Skartved, Guillem Argilés, Rachel S. van der Post, Bastiaan B.J. Tops, Susana Roselló, Marta Benavent, Andrés Cervantes, Rocio Garcia-Carbonero, Amita Patnaik, and Rodrigo Dienstmann
- Abstract
Supplementary Figure S1. Pharmacokinetic profile of Sym004.
- Published
- 2023
9. Data from Safety and Activity of the First-in-Class Sym004 Anti-EGFR Antibody Mixture in Patients with Refractory Colorectal Cancer
- Author
-
Josep Tabernero, Anthony W. Tolcher, Eric Van Cutsem, Stephan Braun, Ivan D. Horak, Michael Kragh, Mikkel W. Pedersen, Rikke Hald, Ulla H. Hansen, Niels J.Ø. Skartved, Guillem Argilés, Rachel S. van der Post, Bastiaan B.J. Tops, Susana Roselló, Marta Benavent, Andrés Cervantes, Rocio Garcia-Carbonero, Amita Patnaik, and Rodrigo Dienstmann
- Abstract
Tumor growth in the context of EGFR inhibitor resistance may remain EGFR-dependent and is mediated by mechanisms including compensatory ligand upregulation and de novo gene alterations. Sym004 is a two-antibody mixture targeting nonoverlapping EGFR epitopes. In preclinical models, Sym004 causes significant EGFR internalization and degradation, which translates into superior growth inhibition in the presence of ligands. In this phase I trial, we observed grade 3 skin toxicity and hypomagnesemia as mechanism-based dose-limiting events during dose escalation. In dose-expansion cohorts of 9 and 12 mg/kg of Sym004 weekly, patients with metastatic colorectal cancer and acquired EGFR inhibitor resistance were enrolled; 17 of 39 patients (44%) had tumor shrinkage, with 5 patients (13%) achieving partial response. Pharmacodynamic studies confirmed marked Sym004-induced EGFR downmodulation. MET gene amplification emerged in 1 patient during Sym004 treatment, and a partial response was seen in a patient with EGFRS492R mutation that is predictive of cetuximab resistance.Significance: Potent EGFR downmodulation with Sym004 in patients with metastatic colorectal cancer and acquired resistance to cetuximab/panitumumab translates into significant antitumor activity and validates the preclinical hypothesis that a proportion of tumors remains dependent on EGFR signaling. Further clinical development and expanded correlative analyses of response patterns with secondary RAS/EGFR mutations are warranted. Cancer Discov; 5(6);598–609. ©2015 AACR.See related commentary by Stintzing and Heinemann, p. 578This article is highlighted in the In This Issue feature, p. 565
- Published
- 2023
10. Supplementary Materials and Methods from Sym004, a Novel Anti-EGFR Antibody Mixture, Augments Radiation Response in Human Lung and Head and Neck Cancers
- Author
-
Paul M. Harari, Mikkel W. Pedersen, Michael Kragh, Eric A. Armstrong, Chunrong Li, Jarob Saker, Chimera R. Peet, and Shyhmin Huang
- Abstract
PDF - 7KB, Quantification of EGFR mRNA expression.
- Published
- 2023
11. Supplementary Figures 1-7 from Simultaneous Targeting of Two Distinct Epitopes on MET Effectively Inhibits MET- and HGF-Driven Tumor Growth by Multiple Mechanisms
- Author
-
Mikkel W. Pedersen, Morag Park, Thomas Bouquin, Johan Lantto, Michael Kragh, Ivan D. Horak, Helle J. Jacobsen, George F.Vande Woude, Dafna Kaufman, Karsten W. Eriksen, Sara Collins, Paolo Conrotto, Gunther R. Galler, Anna Dahlman, Klaus Koefoed, Thomas T. Poulsen, Serhiy Havrylov, and Michael M. Grandal
- Abstract
Supplementary figure 1. Sym015 inhibits viability of cell lines in a synergic manner; Supplementary figure 2. The Sym015 antibodies Hu9338 and Hu9006 bind to 2nd or 3rd blades of MET and block HGF binding; Supplementary figure 3. Sym015 induces MET internalization and degradation in MKN-45 cells; Supplementary figure 4. Sym015 induces MET internalization and degradation in EBC-1 cells; Supplementary figure 5. Sym015 induces MET degradation in MKN-45 and EBC-1 cells; Supplementary figure 6. Sym015 inhibits signaling by MET in EBC-1 cells; Supplementary figure 7. Sym015 inhibits motility of EBC-1
- Published
- 2023
12. Supplementary Figure 2 from Sym004, a Novel Anti-EGFR Antibody Mixture, Augments Radiation Response in Human Lung and Head and Neck Cancers
- Author
-
Paul M. Harari, Mikkel W. Pedersen, Michael Kragh, Eric A. Armstrong, Chunrong Li, Jarob Saker, Chimera R. Peet, and Shyhmin Huang
- Abstract
PDF - 83KB, Effect of Sym004 on nuclear translocation of EGFR.
- Published
- 2023
13. Data from Simultaneous Targeting of Two Distinct Epitopes on MET Effectively Inhibits MET- and HGF-Driven Tumor Growth by Multiple Mechanisms
- Author
-
Mikkel W. Pedersen, Morag Park, Thomas Bouquin, Johan Lantto, Michael Kragh, Ivan D. Horak, Helle J. Jacobsen, George F.Vande Woude, Dafna Kaufman, Karsten W. Eriksen, Sara Collins, Paolo Conrotto, Gunther R. Galler, Anna Dahlman, Klaus Koefoed, Thomas T. Poulsen, Serhiy Havrylov, and Michael M. Grandal
- Abstract
Increased MET activity is linked with poor prognosis and outcome in several human cancers currently lacking targeted therapies. Here, we report on the characterization of Sym015, an antibody mixture composed of two humanized IgG1 antibodies against nonoverlapping epitopes of MET. Sym015 was selected by high-throughput screening searching for antibody mixtures with superior growth-inhibitory activity against MET-dependent cell lines. Synergistic inhibitory activity of the antibodies comprising Sym015 was observed in several cancer cell lines harboring amplified MET locus and was confirmed in vivo. Sym015 was found to exert its activity via multiple mechanisms. It disrupted interaction of MET with the HGF ligand and prompted activity-independent internalization and degradation of the receptor. In addition, Sym015 induced high levels of CDC and ADCC in vitro. The importance of these effector functions was confirmed in vivo using an Fc-effector function–attenuated version of Sym015. The enhanced effect of the two antibodies in Sym015 on both MET degradation and CDC and ADCC is predicted to render Sym015 superior to single antibodies targeting MET. Our results demonstrate strong potential for use of Sym015 as a therapeutic antibody mixture for treatment of MET-driven tumors. Sym015 is currently being tested in a phase I dose escalation clinical trial (NCT02648724). Mol Cancer Ther; 16(12); 2780–91. ©2017 AACR.
- Published
- 2023
14. Supplementary Materials and Methods from Targeting Three Distinct HER2 Domains with a Recombinant Antibody Mixture Overcomes Trastuzumab Resistance
- Author
-
Michael Kragh, Johan Lantto, Ivan D. Horak, Lars S. Nielsen, Per-Johan Meijer, Thomas T. Poulsen, Ida Kjær, Anna Dahlman, Klaus Koefoed, Helle J. Jacobsen, and Mikkel W. Pedersen
- Abstract
Short description of methods and materials used for supplemmentary figures. BioLayer Interferometry, ADCC and CDC assays.
- Published
- 2023
15. Supplementary Tables and Figures from Targeting Three Distinct HER2 Domains with a Recombinant Antibody Mixture Overcomes Trastuzumab Resistance
- Author
-
Michael Kragh, Johan Lantto, Ivan D. Horak, Lars S. Nielsen, Per-Johan Meijer, Thomas T. Poulsen, Ida Kjær, Anna Dahlman, Klaus Koefoed, Helle J. Jacobsen, and Mikkel W. Pedersen
- Abstract
Supplementary Tables and Figures. Table S1 contains affinities for antibodies binding to HER2. Table S2 contains results from epitope bin analysis of the antibodies. Figure S1 shows a comparison of Perjeta and the pertuzumab analogue used in this paper. Figure S2 shows sensograms for the anti-HER2 antibodies. Figure S3 contains dose-response curves for anti-HER2 lead mixtures in the cell lines BT474, SK-BR3, HCC202 and NCI-N87. Figure S4 shows immunoblot data on the levels of HER2, pHER2, EGFR, pEGFR, HER3 and pHER3 in the eight cell lines OE19, N87, MCF7, MDA-MB-175, BT474, HCC202 SK-BR3 and ZE-75-30. Figure S5 shows activity of anti-HER2 mAbs and mixtures in ADCC and CDC assays. Figure S6 demonstrates synergy of the tripartite mixture in OE19 cell line. Figure S7 shows quantification of HER2 and pHER2 levels in OE19 and HCC202 cell lines upon treatment with anti-HER2 mAbs and mixtures. Figure S8 shows activity of tripartite mixtures of trastuzumab, pertuzumab and lead anti-HER2 antibodies in a cell viability assay
- Published
- 2023
16. Supplementary Figure Legends from Sym004, a Novel Anti-EGFR Antibody Mixture, Augments Radiation Response in Human Lung and Head and Neck Cancers
- Author
-
Paul M. Harari, Mikkel W. Pedersen, Michael Kragh, Eric A. Armstrong, Chunrong Li, Jarob Saker, Chimera R. Peet, and Shyhmin Huang
- Abstract
PDF - 66KB, Legends for Supplementary Figures 1 and 2.
- Published
- 2023
17. Supplementary Figures S1-8 from Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity
- Author
-
Johan Lantto, Michael Kragh, Mikkel W. Pedersen, Ivan D. Horak, Christina R. Andersen, Bolette Bjerregaard, Dietmar Weilguny, Jette W. Sen, Klaus Koefoed, Ida Kjær, Anna Dahlman, Thomas T. Poulsen, and Helle J. Jacobsen
- Abstract
Supplementary Figures S1-8. Figure S1 - Analysis of nonlinear blending synergy for the pairs of antibodies that constitute Pan-HER. Figure S2 - In vitro comparison of Pan-HER and a combination of cetuximab, trastuzumab and MM-121. Figure S3 - In vivo assessment of nonlinear blending synergy for the target specificities in the Pan-HER mixture. Figure S4 - Dose titration of Pan-HER in the BxPC3 xenograft model. Figure S5 - IHC analysis of Calu-3 tumors. Figure S6 - Assessment of cell death and cell cycle arrest. Figure S7 - Assessment of ADCC in a panel of cell lines. Figure S8 - Analysis of the effect of receptor internalization on effector functions.
- Published
- 2023
18. Supplementary Methods from Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity
- Author
-
Johan Lantto, Michael Kragh, Mikkel W. Pedersen, Ivan D. Horak, Christina R. Andersen, Bolette Bjerregaard, Dietmar Weilguny, Jette W. Sen, Klaus Koefoed, Ida Kjær, Anna Dahlman, Thomas T. Poulsen, and Helle J. Jacobsen
- Abstract
Supplementary Methods. Description of methods used for assessment of synergy, immunohistochemistry, cell death, cell cycle arrest, ADCC and CDC.
- Published
- 2023
19. Supplementary Tables S1-2 from Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity
- Author
-
Johan Lantto, Michael Kragh, Mikkel W. Pedersen, Ivan D. Horak, Christina R. Andersen, Bolette Bjerregaard, Dietmar Weilguny, Jette W. Sen, Klaus Koefoed, Ida Kjær, Anna Dahlman, Thomas T. Poulsen, and Helle J. Jacobsen
- Abstract
Supplementary Tables S1-2. Table S1 - Source, origin, subtype and growth medium for each cell line. Table S2 - Characteristics of tested patient-derived xenograft models.
- Published
- 2023
20. Data from Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity
- Author
-
Johan Lantto, Michael Kragh, Mikkel W. Pedersen, Ivan D. Horak, Christina R. Andersen, Bolette Bjerregaard, Dietmar Weilguny, Jette W. Sen, Klaus Koefoed, Ida Kjær, Anna Dahlman, Thomas T. Poulsen, and Helle J. Jacobsen
- Abstract
Purpose: Accumulating evidence indicates a high degree of plasticity and compensatory signaling within the human epidermal growth factor receptor (HER) family, leading to resistance upon therapeutic intervention with HER family members.Experimental Design/Results: We have generated Pan-HER, a mixture of six antibodies targeting each of the HER family members EGFR, HER2, and HER3 with synergistic pairs of antibodies, which simultaneously remove all three targets, thereby preventing compensatory tumor promoting mechanisms within the HER family. Pan-HER induces potent growth inhibition in a range of cancer cell lines and xenograft models, including cell lines with acquired resistance to therapeutic antibodies. Pan-HER is also highly efficacious in the presence of HER family ligands, indicating that it is capable of overcoming acquired resistance due to increased ligand production. All three target specificities contribute to the enhanced efficacy, demonstrating a distinct benefit of combined HER family targeting when compared with single-receptor targeting.Conclusions: Our data show that simultaneous targeting of three receptors provides broader efficacy than targeting a single receptor or any combination of two receptors in the HER family, especially in the presence of HER family ligands. Pan-HER represents a novel strategy to deal with primary and acquired resistance due to tumor heterogeneity and plasticity in terms of HER family dependency and as such may be a viable alternative in the clinic. Clin Cancer Res; 21(18); 4110–22. ©2015 AACR.See related commentary by Yarden and Sela, p. 4030
- Published
- 2023
21. First-in-human trial exploring safety, antitumor activity, and pharmacokinetics of Sym013, a recombinant pan-HER antibody mixture, in advanced epithelial malignancies
- Author
-
Jordan Berlin, Anthony W. Tolcher, Cliff Ding, Jennifer G. Whisenant, Ivan D. Horak, Debra L. Wood, Paul I. Nadler, Ulla Holm Hansen, Johan Lantto, Niels Jørgen Ø. Skartved, Mikkel W. Pedersen, and Amita Patnaik
- Subjects
Pharmacology ,Oncology ,Pharmacology (medical) - Published
- 2022
22. Abstract CT274: Individualized APC targeting VB10.NEO cancer vaccines induce broad neoepitope-specific CD8 T cell responses in patients with advanced or metastatic solid tumors: interim results from a phase 1/2a trial
- Author
-
Jürgen Krauss, Angela Krakhardt, Stephan Eisenmann, Sebastian Ochsenreither, Kaja C.G Berg, Kushi Kushekhar, Lars-Egil Fallang, Mikkel W. Pedersen, Siri Torhaug, Karsten B. Slot, and Karoline Schjetne
- Subjects
Cancer Research ,Oncology - Abstract
Background: VB N-01 is an open label phase 1/2a basket trial to evaluate safety, feasibility, and immunogenicity of a therapeutic DNA cancer vaccine VB10.NEO in patients with locally advanced or metastatic solid cancers. Each VB10.NEO vaccine contains up to 20 neoepitopes selected by the proprietary AI platform NeoSELECT and is designed to target antigen presenting cells using Nykode’ s modular vaccine platform known as Vaccibody™. Patients and Methods: The trial enrolled patients with locally advanced or metastatic solid cancers (renal cell carcinoma, urothelial cancer, non-small cell lung cancer, squamous cell carcinoma of the head and neck, and melanoma), who did not obtain complete responses on immune checkpoint inhibitor therapy (CPI). Up to 14 VB10.NEO doses (3 mg per dose) were administered i.m. by PharmaJet Stratis® for up to 1 year in combination with standard of care (CPI and/or other anti-cancer therapies). Blood samples and tumor biopsies were collected at baseline and during treatment for evaluation of immune responses. Results: At cut-off of May 2022, 41 patients had received at least one vaccination with VB10.NEO. The vaccine was safe and well-tolerated with no new or additional toxicity reported beyond that expected for CPIs alone. All patients displayed immune responses to a minimum of 3 neoepitopes (average 53% of vaccine neoepitopes), assessed by in vitro stimulation (IVS) interferon-gamma ELISpot. T cell responses were elicited in both high and low tumor mutational burden patients (TMB range 2-69mut/Mb). Multiple vaccinations increased the breadth and magnitude of the immune responses with vaccine-induced T cell responses (de novo and/or amplified pre-existing) measured in 95% of eligible patients. IVS intracellular cytokine staining of selected patients demonstrated that the majority of neoepitopes induced polyfunctional CD8 T cells. TCR sequencing of baseline and on-treatment samples (6 patients) showed expansion of T cell clones in both blood and tumor with selected clones being expanded in both compartments. Conclusions: VB10.NEO was generally well tolerated in patients with various pre-treated and advanced cancers. Assessment of neoepitope-specific T cell reactivity demonstrated VB10.NEO-induced broad and long-lasting T cell responses, and the majority of tested neoepitopes activated polyfunctional CD8 T cells. Pre- and post-vaccination TCRseq analysis of blood and tumor samples demonstrated the presence of blood-expanded clones in the on-treatment tumor sample potentially indicating trafficking of VB10.NEO-expanded T cells to the tumor site. Citation Format: Jürgen Krauss, Angela Krakhardt, Stephan Eisenmann, Sebastian Ochsenreither, Kaja C.G Berg, Kushi Kushekhar, Lars-Egil Fallang, Mikkel W. Pedersen, Siri Torhaug, Karsten B. Slot, Karoline Schjetne. Individualized APC targeting VB10.NEO cancer vaccines induce broad neoepitope-specific CD8 T cell responses in patients with advanced or metastatic solid tumors: interim results from a phase 1/2a trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT274.
- Published
- 2023
23. Sym004, a Novel EGFR Antibody Mixture, Can Overcome Acquired Resistance to Cetuximab
- Author
-
Mari Iida, Toni M Brand, Megan M Starr, Chunrong Li, Evan J Huppert, Neha Luthar, Mikkel W Pedersen, Ivan D Horak, Michael Kragh, and Deric L Wheeler
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The epidermal growth factor receptor (EGFR) is a central regulator of tumor progression in a variety of human cancers. Cetuximab is an anti-EGFR monoclonal antibody that has been approved for head and neck and colorectal cancer treatment, but many patients treated with cetuximab don't respond or eventually acquire resistance. To determine how tumor cells acquire resistance to cetuximab, we previously developed a model of acquired resistance using the non-small cell lung cancer line NCI-H226. These cetuximab-resistant (CtxR) cells exhibit increased steady-state EGFR expression secondary to alterations in EGFR trafficking and degradation and, further, retained dependence on EGFR signaling for enhanced growth potential. Here, we examined Sym004, a novel mixture of antibodies directed against distinct epitopes on the extracellular domain of EGFR, as an alternative therapy for CtxR tumor cells. Sym004 treatment of CtxR clones resulted in rapid EGFR degradation, followed by robust inhibition of cell proliferation and down-regulation of several mitogen-activated protein kinase pathways. To determine whether Sym004 could have therapeutic benefit in vivo, we established de novo CtxR NCI-H226 mouse xenografts and subsequently treated CtxR tumors with Sym004. Sym004 treatment of mice harboring CtxR tumors resulted in growth delay compared to mice continued on cetuximab. Levels of total and phospho-EGFR were robustly decreased in CtxR tumors treated with Sym004. Immunohistochemical analysis of these Sym004-treated xenograft tumors further demonstrated decreased expression of Ki67, and phospho-rpS6, as well as a modest increase in cleaved caspase-3. These results indicate that Sym004 may be an effective targeted therapy for CtxR tumors.
- Published
- 2013
- Full Text
- View/download PDF
24. Targeting of EGFR by a combination of antibodies mediates unconventional EGFR trafficking and degradation
- Author
-
Peter King, Clare E. Futter, Costin N. Antonescu, Mikkel W. Pedersen, Daniel Hochhauser, John A. Hartley, Michael Kragh, Sylwia Jones, Nicos Angelopoulos, Michael G. Sugiyama, Amandeep Bhamra, and Silvia Surinova
- Subjects
Ubiquitylation ,Endosome ,lcsh:Medicine ,Antineoplastic Agents ,Receptors, Cell Surface ,Membrane trafficking ,Endosomes ,Endocytosis ,Article ,Antibodies ,Targeted therapies ,Ubiquitin ,Cell surface receptor ,medicine ,Humans ,TSG101 ,Receptor ,lcsh:Science ,Transcription factor ,Cells, Cultured ,Multidisciplinary ,Endosomal Sorting Complexes Required for Transport ,biology ,Chemistry ,Cell Membrane ,lcsh:R ,Growth factor signalling ,Antibodies, Monoclonal ,Cancer ,Phosphoproteins ,medicine.disease ,DNA-Binding Proteins ,ErbB Receptors ,Protein Transport ,Head and Neck Neoplasms ,biology.protein ,Cancer research ,lcsh:Q ,Lysosomes ,Transcription Factors - Abstract
Antibody combinations targeting cell surface receptors are a new modality of cancer therapy. The trafficking and signalling mechanisms regulated by such therapeutics are not fully understood but could underlie differential tumour responses. We explored EGFR trafficking upon treatment with the antibody combination Sym004 which has shown promise clinically. Sym004 promoted EGFR endocytosis distinctly from EGF: it was asynchronous, not accompanied by canonical signalling events and involved EGFR clustering within detergent-insoluble plasma mebrane-associated tubules. Sym004 induced lysosomal degradation independently of EGFR ubiquitylation but dependent upon Hrs/Tsg101 that are required for the formation of intraluminal vesicles (ILVs) within late endosomes. We propose Sym004 cross-links EGFR physically triggering EGFR endocytosis and incorporation onto ILVs and so Sym004 sensitivity correlates with EGFR numbers available for binding, rather than specific signalling events. Consistently Sym004 efficacy and potentiation of cisplatin responses correlated with EGFR surface expression in head and neck cancer cells. These findings will have implications in understanding the mode of action of this new class of cancer therapeutics.
- Published
- 2020
25. Ancient Human Genomes and Environmental DNA from the Cement Attaching 2,000-Year-Old Head Lice Nits
- Author
-
Mikkel W Pedersen, Catia Antunes, Binia De Cahsan, J Víctor Moreno-Mayar, Martin Sikora, Lasse Vinner, Darren Mann, Pavel B Klimov, Stuart Black, Catalina Teresa Michieli, Henk R Braig, and M Alejandra Perotti
- Subjects
integumentary system ,Genome, Human ,parasitic diseases ,Skull ,Infant, Newborn ,Pediculus ,Genetics ,Animals ,Humans ,DNA, Environmental ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Genome-Wide Association Study - Abstract
Over the past few decades, there has been a growing demand for genome analysis of ancient human remains. Destructive sampling is increasingly difficult to obtain for ethical reasons, and standard methods of breaking the skull to access the petrous bone or sampling remaining teeth are often forbidden for curatorial reasons. However, most ancient humans carried head lice and their eggs abound in historical hair specimens. Here we show that host DNA is protected by the cement that glues head lice nits to the hair of ancient Argentinian mummies, 1,500–2,000 years old. The genetic affinities deciphered from genome-wide analyses of this DNA inform that this population migrated from north-west Amazonia to the Andes of central-west Argentina; a result confirmed using the mitochondria of the host lice. The cement preserves ancient environmental DNA of the skin, including the earliest recorded case of Merkel cell polyomavirus. We found that the percentage of human DNA obtained from nit cement equals human DNA obtained from the tooth, yield 2-fold compared with a petrous bone, and 4-fold to a bloodmeal of adult lice a millennium younger. In metric studies of sheaths, the length of the cement negatively correlates with the age of the specimens, whereas hair linear distance between nit and scalp informs about the environmental conditions at the time before death. Ectoparasitic lice sheaths can offer an alternative, nondestructive source of high-quality ancient DNA from a variety of host taxa where bones and teeth are not available and reveal complementary details of their history.
- Published
- 2021
26. First-in-human trial exploring safety, antitumor activity, and pharmacokinetics of Sym013, a recombinant pan-HER antibody mixture, in advanced epithelial malignancies
- Author
-
Jordan, Berlin, Anthony W, Tolcher, Cliff, Ding, Jennifer G, Whisenant, Ivan D, Horak, Debra L, Wood, Paul I, Nadler, Ulla Holm, Hansen, Johan, Lantto, Niels Jørgen Ø, Skartved, Mikkel W, Pedersen, and Amita, Patnaik
- Subjects
Diarrhea ,Maximum Tolerated Dose ,Neoplasms ,Antibodies, Monoclonal ,Humans ,Antineoplastic Agents ,Neoplasms, Glandular and Epithelial ,Exanthema - Abstract
Sym013 contains six humanized monoclonal antibodies that bind to non-overlapping epitopes on three human epidermal growth factor receptors (HER1-3). Preclinical studies suggested Sym013 strongly suppresses growth of multiple epithelial tumors. This is a first-in-human study exploring safety and efficacy of Sym013 in patients with advanced epithelial malignancies.Dose escalation used single-patient cohorts until the stopping rule was met, followed by 3 + 3 design. Dose levels planned were: 1, 2, 4, 6, 9, 12, 15, and 18 mg/kg. Treatment cycles were 28 days with imaging every eight weeks. Serum samples were collected at multiple time points for assessment of pharmacokinetics and development of anti-drug antibodies.Thirty-two patients were enrolled with multiple solid tumors, most common being colorectal cancer (CRC; 10/32, 31%). Due to mucositis, rash, and diarrhea at 4 mg/kg once-weekly, dosing was changed to biweekly (Q2W). Mandatory prophylaxis was added due to Grade 3 infusion-related reaction and oral mucositis at 9 mg/kg Q2W. The 15 mg/kg Q2W cohort was enrolling when the study was terminated for business reasons. Most common adverse events were skin (81%) and gastrointestinal (75%) disorders, including dermatitis/rash, stomatitis, and diarrhea. One patient with CRC achieved a partial response; 12 patients with varied malignancies had stable disease.During the conduct of the study, management of frequent infusion-related reactions, skin toxicities, and mucosal disorders, which are indicative of HER inhibition, necessitated multiple protocol amendments. The investigators, in concert with the Sponsor, agreed that achieving a tolerated regimen with acceptable target saturation was unlikely.www.gov ; NCT02906670 (September 20, 2016).
- Published
- 2021
27. Deep neural networks identify signaling mechanisms of ErbB-family drug resistance from a continuous cell morphology space
- Author
-
Rune Linding, James Longden, Xavier Robin, Jesper Ferkinghoff-Borg, Mathias Engel, Mikkel W. Pedersen, Ida Kjær, and Ivan D. Horak
- Subjects
0301 basic medicine ,TOPHAT ,EGFR ,Cell ,Computational biology ,Biology ,Cell morphology ,General Biochemistry, Genetics and Molecular Biology ,Machine Learning ,03 medical and health sciences ,Deep Learning ,0302 clinical medicine ,medicine ,KINASE ,Humans ,HETEROGENEITY ,Insulin-like growth factor 1 receptor ,Cetuximab ,Cancer ,Complex cell ,medicine.disease ,CANCER ,GENE ,ErbB Receptors ,030104 developmental biology ,medicine.anatomical_structure ,TARGET ,Drug Resistance, Neoplasm ,High-content screening ,Cancer cell ,GROWTH ,Neural Networks, Computer ,KINOME ,030217 neurology & neurosurgery ,Signal Transduction ,medicine.drug - Abstract
It is well known that the development of drug resistance in cancer cells can lead to changes in cell morphology. Here, we describe the use of deep neural networks to analyze this relationship, demonstrating that complex cell morphologies can encode states of signaling networks and unravel cellular mechanisms hidden to conventional approaches. We perform high-content screening of 17 cancer cell lines, generating more than 500 billion data points from similar to 850 million cells. We analyze these data using a deep learning model, resulting in the identification of a continuous 27-dimension space describing all of the observed cell morphologies. From its morphology alone, we could thus predict whether a cell was resistant to ErbB-family drugs, with an accuracy of 74%, and predict the potential mechanism of resistance, subsequently validating the role of MET and insulin-like growth factor 1 receptor (IGF1R) as drivers of cetuximab resistance in in vitro models of lung and head/neck cancer.
- Published
- 2021
28. Colorectal cancer residual disease at maximal response to EGFR blockade displays a druggable Paneth cell–like phenotype
- Author
-
Andrea Bertotti, Barbara Lupo, Guillem Argiles, Claudio Isella, Carla Boccaccio, Marika Pinnelli, Alexandra Vatsiou, Paolo Nuciforo, Trevor A. Graham, Paolo Luraghi, Mikkel W. Pedersen, Timon Heide, Daria Manganaro, Ivan D. Horak, Enzo Medico, Francesco Sassi, Inmaculada Spiteri, Rodrigo Dienstmann, Francesco Galimi, Paolo Armando Gagliardi, Giorgia Migliardi, Elena Elez, Livio Trusolino, Michael Kragh, Elena Grassi, Luca Primo, Alberto Puliafito, Valentina Vurchio, Diego Pasini, Andrea Sottoriva, Eugenia R. Zanella, and Daniel Nichol
- Subjects
Paneth Cells ,Disease reservoir ,Neoplasm, Residual ,medicine.disease_cause ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,Epidermal growth factor receptor ,030304 developmental biology ,0303 health sciences ,Cetuximab ,biology ,General Medicine ,Intestinal epithelium ,Minimal residual disease ,3. Good health ,ErbB Receptors ,Phenotype ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Paneth cell ,Cancer research ,biology.protein ,Neoplasm Recurrence, Local ,Signal transduction ,Colorectal Neoplasms ,Carcinogenesis ,medicine.drug - Abstract
Blockade of epidermal growth factor receptor (EGFR) causes tumor regressions in selected patients with metastatic colorectal cancer (mCRC); however, residual disease reservoirs typically remain even after maximal response to therapy, leading to relapse. Using patient-derived xenografts, we observed that mCRC cells surviving EGFR inhibition exhibited gene expression patterns reminiscent of those displayed by a quiescent subpopulation of normal intestinal secretory precursors with Paneth-cell characteristics. These pseudodifferentiated remnants had reduced expression of EGFR-activating ligands, more pronounced activity of human epidermal growth factor 2 (HER2) and human epidermal growth factor receptor 3 (HER3), and persistent signaling along the phosphatidylinositol-3-kinase (PI3K) pathway compared with untreated tumors. Clinically, residual disease traits were detected in lingering tumors of responsive patients and in tumors of individuals who had experienced early recurrence. Mechanistically, residual tumor reprogramming was mediated by inactivation of Yes-associated protein (YAP) – a master regulator of post-injury intestinal epithelium recovery – following EGFR neutralization. In preclinical trials, Pan-HER antibodies minimized residual disease, blunted PI3K signaling, and induced long-term tumor control after treatment discontinuation. By showing that tolerance to EGFR inhibition is typified by the disengagement of an in-built lineage program that drives both regenerative signaling during intestinal repair and EGFR-dependent tumorigenesis, our results shed light onto CRC lineage plasticity as an adaptive escape mechanism from therapeutic insults and suggest opportunities to pre-emptively target residual disease.
- Published
- 2020
29. Sym004-induced EGFR elimination is associated with profound anti-tumor activity in EGFRvIII patient-derived glioblastoma models
- Author
-
Carlee D. Hemphill, Mikkel W. Pedersen, Michael Kragh, Michael M. Grandal, Darell D. Bigner, Vidyalakshmi Chandramohan, Ivan D. Horak, Stephen T. Keir, Maria C. Melander, Henry S. Friedman, and Annick Desjardins
- Subjects
EGFRvIII ,Male ,0301 basic medicine ,Cancer Research ,medicine.drug_class ,EGFR ,media_common.quotation_subject ,Mice, Nude ,Antineoplastic Agents ,mAbs ,Monoclonal antibody ,GBM ,Epitope ,03 medical and health sciences ,Subcutaneous Tissue ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Internalization ,media_common ,Mice, Inbred BALB C ,Sym004 ,Cetuximab ,biology ,Brain Neoplasms ,Chemistry ,Antibodies, Monoclonal ,Xenograft Model Antitumor Assays ,In vitro ,ErbB Receptors ,030104 developmental biology ,Neurology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Laboratory Investigation ,Cancer research ,biology.protein ,Female ,Neurology (clinical) ,Glioblastoma ,medicine.drug - Abstract
Background Sym004 is a mixture of two monoclonal antibodies (mAbs), futuximab and modotuximab, targeting non-overlapping epitopes on the epidermal growth factor receptor (EGFR). Previous studies have shown that Sym004 is more efficient at inducing internalization and degradation of EGFR than individual components, which translates into superior cancer cell inhibition. We investigated whether Sym004 induces removal of EGFRvIII and if this removal translates into tumor growth inhibition in hard-to-treat glioblastomas (GBMs) harboring the mutated, constitutively active EGFR variant III (EGFRvIII). Methods To address this question, we tested the effect of Sym004 versus cetuximab in eight patient-derived GBM xenograft models expressing either wild-type EGFR (EGFRwt) and/or mutant EGFRvIII. All models were tested as both subcutaneous and orthotopic intracranial xenograft models. Results In vitro studies demonstrated that Sym004 internalized and removed EGFRvIII more efficiently than mAbs, futuximab, modotuximab, and cetuximab. Removal of EGFRvIII by Sym004 translated into significant in vivo anti-tumor activity in all six EGFRvIII xenograft models. Furthermore, the anti-tumor activity of Sym004 in vivo was superior to that of its individual components, futuximab and modotuximab, suggesting a clear synergistic effect of the mAbs in the mixture. Conclusion These results demonstrate the broad activity of Sym004 in patient-derived EGFRvIII-expressing GBM xenograft models and provide a clear rationale for clinical evaluation of Sym004 in EGFRvIII-positive adult GBM patients. Electronic supplementary material The online version of this article (10.1007/s11060-018-2832-6) contains supplementary material, which is available to authorized users.
- Published
- 2018
30. Simultaneous Targeting of Two Distinct Epitopes on MET Effectively Inhibits MET- and HGF-Driven Tumor Growth by Multiple Mechanisms
- Author
-
Klaus Koefoed, Helle Jacobsen, Mikkel W. Pedersen, Thomas Tuxen Poulsen, Johan Lantto, Sara Collins, Karsten W. Eriksen, Michael M. Grandal, Morag Park, Gunther R. Galler, Michael Kragh, Ivan D. Horak, Serhiy Havrylov, Thomas Bouquin, Dafna Kaufman, Paolo Conrotto, George F. Vande Woude, and Anna Dahlman
- Subjects
0301 basic medicine ,Cancer Research ,media_common.quotation_subject ,Mice, Nude ,Epitope ,Epitopes ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Internalization ,Cell Proliferation ,media_common ,Antibody-dependent cell-mediated cytotoxicity ,biology ,Cell growth ,Proto-Oncogene Proteins c-met ,Xenograft Model Antitumor Assays ,Molecular biology ,In vitro ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody - Abstract
Increased MET activity is linked with poor prognosis and outcome in several human cancers currently lacking targeted therapies. Here, we report on the characterization of Sym015, an antibody mixture composed of two humanized IgG1 antibodies against nonoverlapping epitopes of MET. Sym015 was selected by high-throughput screening searching for antibody mixtures with superior growth-inhibitory activity against MET-dependent cell lines. Synergistic inhibitory activity of the antibodies comprising Sym015 was observed in several cancer cell lines harboring amplified MET locus and was confirmed in vivo. Sym015 was found to exert its activity via multiple mechanisms. It disrupted interaction of MET with the HGF ligand and prompted activity-independent internalization and degradation of the receptor. In addition, Sym015 induced high levels of CDC and ADCC in vitro. The importance of these effector functions was confirmed in vivo using an Fc-effector function–attenuated version of Sym015. The enhanced effect of the two antibodies in Sym015 on both MET degradation and CDC and ADCC is predicted to render Sym015 superior to single antibodies targeting MET. Our results demonstrate strong potential for use of Sym015 as a therapeutic antibody mixture for treatment of MET-driven tumors. Sym015 is currently being tested in a phase I dose escalation clinical trial (NCT02648724). Mol Cancer Ther; 16(12); 2780–91. ©2017 AACR.
- Published
- 2017
31. Sym015: A Highly Efficacious Antibody Mixture against MET-Amplified Tumors
- Author
-
Mikkel W. Pedersen, Ivan D. Horak, Anna Dahlman, Klaus Koefoed, Helle Jacobsen, Rikke Hald, Michael Kragh, Karsten W. Eriksen, Michael M. Grandal, Thomas Tuxen Poulsen, Johan Lantto, Camilla Fröhlich, Lene Alifrangis, Sofie Ellebæk Pollmann, Niels Jorgen Ostergaard Skartved, Trine Lindsted, and Thomas Bouquin
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,Cell ,Cancer ,Biology ,Pharmacology ,medicine.disease ,Epitope ,Receptor tyrosine kinase ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Monoclonal ,medicine ,biology.protein ,Antibody ,Lung cancer - Abstract
Purpose: Activation of the receptor tyrosine kinase MET is associated with poor clinical outcome in certain cancers. To target MET more effectively, we developed an antagonistic antibody mixture, Sym015, consisting of two humanized mAbs directed against nonoverlapping epitopes of MET. Experimental Design/Results: We screened a large panel of well-annotated human cancer cell lines and identified a subset with highly elevated MET expression. In particular, cell lines of lung cancer and gastric cancer origin demonstrated high MET expression and activation, and Sym015 triggered degradation of MET and significantly inhibited growth of these cell lines. Next, we tested Sym015 in patient- and cell line–derived xenograft models with high MET expression and/or MET exon 14 skipping alterations, and in models harboring MET amplification as a mechanism of resistance to EGFR-targeting agents. Sym015 effectively inhibited tumor growth in all these models and was superior to an analogue of emibetuzumab, a monoclonal IgG4 antibody against MET currently in clinical development. Sym015 also induced antibody-dependent cellular cytotoxicity (ADCC) in vitro, suggesting that secondary effector functions contribute to the efficacy of Sym015. Retrospectively, all responsive, high MET-expressing models were scored as highly MET-amplified by in situ hybridization, pointing to MET amplification as a predictive biomarker for efficacy. Preclinical toxicology studies in monkeys showed that Sym015 was well tolerated, with a pharmacokinetic profile supporting administration of Sym015 every second or third week in humans. Conclusions: The preclinical efficacy and safety data provide a clear rationale for the ongoing clinical studies of Sym015 in patients with MET-amplified tumors. Clin Cancer Res; 23(19); 5923–35. ©2017 AACR.
- Published
- 2017
32. Sym021, a promising anti-PD1 clinical candidate antibody derived from a new chicken antibody discovery platform
- Author
-
Maria C. Melander, Torben Gjetting, Nikolaj Dietrich, Mikkel W. Pedersen, Gunther R. Galler, Klaus Koefoed, Trine Lindsted, Michael Kragh, Monika Gad, Michael M. Grandal, Camilla Fröhlich, Vikram Kjoller Bhatia, Ivan D. Horak, Franziska Uhlenbrock, Johan Lantto, Thomas Bouquin, and Magnus Strandh
- Subjects
medicine.drug_class ,T-Lymphocytes ,Immunology ,Programmed Cell Death 1 Receptor ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Lymphocyte Activation ,Protein Engineering ,immune-oncology ,Epitope ,Immunoglobulin G ,B7-H1 Antigen ,Avian Proteins ,03 medical and health sciences ,Mice ,antibody repertoire diversity ,0302 clinical medicine ,Antibody Repertoire ,Antigen ,Report ,medicine ,Immunology and Allergy ,Animals ,Humans ,Cells, Cultured ,030304 developmental biology ,Antibody-dependent cell-mediated cytotoxicity ,0303 health sciences ,biology ,Chemistry ,Immunodominant Epitopes ,Antibodies, Monoclonal ,Programmed Cell Death 1 Ligand 2 Protein ,Molecular biology ,Mice, Inbred C57BL ,PD1 ,Macaca fascicularis ,Epitope mapping ,030220 oncology & carcinogenesis ,biology.protein ,Chicken antibodies ,Cytokines ,Antibody ,Chickens ,Epitope Mapping ,Protein Binding - Abstract
Discovery of therapeutic antibodies is a field of intense development, where immunization of rodents remains a major source of antibody candidates. However, high orthologue protein sequence homology between human and rodent species disfavors generation of antibodies against functionally conserved binding epitopes. Chickens are phylogenetically distant from mammals. Since chickens generate antibodies from a restricted set of germline genes, the possibility of adapting the Symplex antibody discovery platform to chicken immunoglobulin genes and combining it with high-throughput humanization of antibody frameworks by “mass complementarity-determining region grafting” was explored. Hence, wild type chickens were immunized with an immune checkpoint inhibitor programmed cell death 1 (PD1) antigen, and a repertoire of 144 antibodies was generated. The PD1 antibody repertoire was successfully humanized, and we found that most humanized antibodies retained affinity largely similar to that of the parental chicken antibodies. The lead antibody Sym021 blocked PD-L1 and PD-L2 ligand binding, resulting in elevated T-cell cytokine production in vitro. Detailed epitope mapping showed that the epitope recognized by Sym021 was unique compared to the clinically approved PD1 antibodies pembrolizumab and nivolumab. Moreover, Sym021 bound human PD1 with a stronger affinity (30 pM) compared to nivolumab and pembrolizumab, while also cross-reacting with cynomolgus and mouse PD1. This enabled direct testing of Sym021 in the syngeneic mouse in vivo cancer models and evaluation of preclinical toxicology in cynomolgus monkeys. Preclinical in vivo evaluation in various murine and human tumor models demonstrated a pronounced anti-tumor effect of Sym021, supporting its current evaluation in a Phase 1 clinical trial. Abbreviations: ADCC, antibody-dependent cellular cytotoxicity; CD, cluster of differentiation; CDC, complement-dependent cytotoxicity; CDR, complementarity determining region; DC, dendritic cell; ELISA, enzyme-linked immunosorbent assay; FACS, fluorescence activated cell sorting; FR, framework region; GM-CSF, granulocyte-macrophage colony-stimulating factor; HRP, horseradish peroxidase; IgG, immunoglobulin G; IL, interleukin; IFN, interferon; mAb, monoclonal antibody; MLR, mixed lymphocyte reaction; NK, natural killer; PBMC, peripheral blood mono-nuclear cell; PD1, programmed cell death 1; PDL1, programmed cell death ligand 1; RT-PCR, reverse transcription polymerase chain reaction; SEB, Staphylococcus Enterotoxin B; SPR, surface Plasmon Resonance; VL, variable part of light chain; VH, variable part of heavy chain
- Published
- 2019
33. A combination of two antibodies recognizing non‐overlapping epitopes of <scp>HER</scp> 2 induces kinase activity‐dependent internalization of <scp>HER</scp> 2
- Author
-
Mikkel W. Pedersen, Espen Stang, Michael M. Grandal, Filip Nikolaysen, Vibeke Bertelsen, Anne Marthe Fosdahl, and Monika Szymanska
- Subjects
Dynamins ,0301 basic medicine ,Receptor, ErbB-2 ,medicine.drug_class ,media_common.quotation_subject ,Endocytic cycle ,kinase activity ,Biology ,Monoclonal antibody ,Epitope ,Cell Line ,Polymerization ,Epitopes ,03 medical and health sciences ,medicine ,Humans ,Epidermal growth factor receptor ,Phosphorylation ,Kinase activity ,skin and connective tissue diseases ,Receptor ,Internalization ,neoplasms ,HER2/ErbB2 ,degradation ,media_common ,antibody combinations ,Ubiquitination ,Antibodies, Monoclonal ,Original Articles ,Cell Biology ,Molecular biology ,Actins ,Clathrin ,Endocytosis ,Cell biology ,030104 developmental biology ,Proteolysis ,biology.protein ,Pinocytosis ,Molecular Medicine ,Original Article ,monoclonal antibodies - Abstract
The human epidermal growth factor receptor 2 (HER2/ErbB2) is overexpressed in a number of human cancers. HER2 is the preferred heterodimerization partner for other epidermal growth factor receptor (EGFR) family members and is considered to be resistant to endocytic down‐regulation, properties which both contribute to the high oncogenic potential of HER2. Antibodies targeting members of the EGFR family are powerful tools in cancer treatment and can function by blocking ligand binding, preventing receptor dimerization, inhibiting receptor activation and/or inducing receptor internalization and degradation. With respect to antibody‐induced endocytosis of HER2, various results are reported, and the effect seems to depend on the HER2 expression level and whether antibodies are given as individual antibodies or as mixtures of two or more. In this study, the effect of a mixture of two monoclonal antibodies against non‐overlapping epitopes of HER2 was investigated with respect to localization and stability of HER2. Individual antibodies had limited effect, but the combination of antibodies induced internalization and degradation of HER2 by multiple endocytic pathways. In addition, HER2 was phosphorylated and ubiquitinated upon incubation with the antibody combination, and the HER2 kinase activity was found to be instrumental in antibody‐induced HER2 down‐regulation.
- Published
- 2016
34. Cetuximab Resistance in Squamous Carcinomas of the Upper Aerodigestive Tract Is Driven by Receptor Tyrosine Kinase Plasticity: Potential for mAb Mixtures
- Author
-
Michael Kragh, Trine Lindsted, Ida Kjær, Mikkel W. Pedersen, Ivan D. Horak, Camilla Fröhlich, and Jesper V. Olsen
- Subjects
0301 basic medicine ,Cancer Research ,Receptor, ErbB-3 ,Cell ,Cetuximab ,Gene Expression ,Antineoplastic Agents ,Apoptosis ,Drug resistance ,Digestive System Neoplasms ,Receptor tyrosine kinase ,Receptor, IGF Type 1 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,neoplasms ,Cell Proliferation ,Insulin-like growth factor 1 receptor ,biology ,Cell growth ,Antibodies, Monoclonal ,Receptor Protein-Tyrosine Kinases ,Cancer ,medicine.disease ,digestive system diseases ,ErbB Receptors ,body regions ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Carcinoma, Squamous Cell ,biology.protein ,Cancer research ,Female ,medicine.drug - Abstract
Squamous cell carcinomas (SCC) arising in upper parts of the aerodigestive tract are among the leading causes of death worldwide. EGFR has been found to play an essential role in driving the malignancy of SCC of the upper aerodigestive tract (SCCUAT), but, despite this, clinical results using a range of different EGFR-targeted agents have been disappointing. Cetuximab is currently the only EGFR-targeted agent approved by the FDA for treatment of SCCUAT. However, intrinsic and acquired cetuximab resistance is a major problem for effective therapy. Thus, a better understanding of the mechanisms responsible for cetuximab resistance is valuable for development of the next generation of antibody therapeutics. In order to better understand the underlying mechanisms of cetuximab resistance in SCCUAT, we established from cetuximab-sensitive models cell lines with acquired resistance to cetuximab by continuous selective pressure in vitro and in vivo. Our results show that resistant clones maintain partial dependency on EGFR and that receptor tyrosine kinase plasticity mediated by HER3 and IGF1R plays an essential role. A multitarget mAb mixture against EGFR, HER3, and IGF1R was able to overcome cetuximab resistance in vitro. To our surprise, these findings could be extended to include SCCUAT cell lines with intrinsic resistance to cetuximab, suggesting that the triad consisting of EGFR, HER3, and IGF1R plays a key role in SCCUAT. Our results thus provide a rationale for simultaneous targeting of EGFR, HER3, and IGF1R in SCCUAT. Mol Cancer Ther; 15(7); 1614–26. ©2016 AACR.
- Published
- 2016
35. Deep Neural Networks Identify Signaling Mechanisms of ErbB-Family Drug Resistance From a Continuous Cell Morphology State Space
- Author
-
Jesper Ferkinghoff Borg, Mathias Engel, Ivan D. Horak, Ida Kjær, Mikkel W. Pedersen, James Longden, Rune Linding, and Xavier Robin
- Subjects
medicine.anatomical_structure ,ERBB Family ,Cell ,Cancer cell ,medicine ,State space ,Deep neural networks ,Drug resistance ,Biology ,Cell morphology ,Cell shape ,Neuroscience - Abstract
It is well known that the development of drug resistance in cancer cells can lead to a change in cell morphology. We reasoned that machine-learning techniques could thus be used to elucidate far greater insight into the relationship between cell shape and signaling. To test this hypothesis we performed a large high content screen on drug sensitive and drug resistance cancer cells, and analysed the shape of these cells using a deep neural network. Our model identified a continuous 27-dimension space describing all of the observed cell morphologies from which we were able to predict drug resistance with an accuracy of 74%. In addition, analyzing changes in cell morphology identified signaling networks that, when perturbed, caused the death of drug resistant cells. These findings suggests that complex morphologies can decode states of signaling networks seemingly unrelated to cell shape, and that analysis of this information can unravel cellular mechanisms hidden to conventional measurements.
- Published
- 2018
36. Efficacy of Sym004 in Patients With Metastatic Colorectal Cancer With Acquired Resistance to Anti-EGFR Therapy and Molecularly Selected by Circulating Tumor DNA Analyses
- Author
-
Clara Montagut, Guillem Argilés, Fortunato Ciardiello, Thomas T. Poulsen, Rodrigo Dienstmann, Michael Kragh, Scott Kopetz, Trine Lindsted, Cliff Ding, Joana Vidal, Jenifer Clausell-Tormos, Giulia Siravegna, Francisco J. Sánchez-Martín, Klaus Koefoed, Mikkel W. Pedersen, Michael M. Grandal, Mikhail Dvorkin, Lucjan Wyrwicz, Ana Rovira, Antonio Cubillo, Ramon Salazar, Françoise Desseigne, Cristin
- Published
- 2018
- Full Text
- View/download PDF
37. A phase I study of Sym021, an anti-PD-1 antibody (Ab), alone and in combination with Sym022 (anti-LAG-3) or Sym023 (anti-TIM-3)
- Author
-
Anna Spreafico, Marc Oliva, Filip Janku, Jordi Rodon, Johan Lantto, S. Musalli, Thorarinn Blondal, Ivan D. Horak, Mikkel W. Pedersen, Nehal Lakhani, L. Knauss, Paul Nadler, Lene Alifrangis, Camilla Fröhlich, S.R. Chandana, L.L. Siu, Maria C. Melander, Michael Kragh, Anthony W. Tolcher, and D. Wood
- Subjects
Eccrine ductal carcinoma ,biology ,business.industry ,Immunogenicity ,Anti pd 1 ,Complete remission ,Hematology ,Pharmacology ,medicine.disease_cause ,Autoimmunity ,Phase i study ,Oncology ,Aldesleukin ,medicine ,biology.protein ,Antibody ,business - Published
- 2019
38. Targeting Three Distinct HER2 Domains with a Recombinant Antibody Mixture Overcomes Trastuzumab Resistance
- Author
-
Lars S. Nielsen, Mikkel W. Pedersen, Ida Kjær, Thomas Tuxen Poulsen, Johan Lantto, Ivan D. Horak, Anna Dahlman, Per-Johan Meijer, Michael Kragh, Klaus Koefoed, and Helle Jacobsen
- Subjects
Cancer Research ,Receptor, ErbB-2 ,media_common.quotation_subject ,Mice, Nude ,Breast Neoplasms ,Biology ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Epitope ,Trastuzumab ,In vivo ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Internalization ,neoplasms ,Cell Proliferation ,media_common ,Mice, Inbred BALB C ,Antibodies, Monoclonal ,Drug Synergism ,medicine.disease ,Xenograft Model Antitumor Assays ,Metastatic breast cancer ,ErbB Receptors ,Mice, Inbred C57BL ,Oncology ,Drug Resistance, Neoplasm ,Monoclonal ,biology.protein ,Female ,Pertuzumab ,Antibody ,Signal Transduction ,medicine.drug - Abstract
HER2 plays an important role in the development and maintenance of the malignant phenotype of several human cancers. As such, it is a frequently pursued therapeutic target and two antibodies targeting HER2 have been clinically approved, trastuzumab and pertuzumab. It has been suggested that optimal inhibition of HER2 is achieved when utilizing two or more antibodies targeting nonoverlapping epitopes. Superior clinical activity of the trastuzumab plus pertuzumab combination in metastatic breast cancer supports this hypothesis. Because trastuzumab and pertuzumab were not codeveloped, there may be potential for further optimizing HER2 targeting. The study herein evaluated functional activity of anti-HER2 antibody combinations identifying optimal epitope combinations that provide efficacious HER2 inhibition. High-affinity antibodies to all four extracellular domains on HER2 were identified and tested for ability to inhibit growth of different HER2-dependent tumor cell lines. An antibody mixture targeting three HER2 subdomains proved to be superior to trastuzumab, pertuzumab, or a combination in vitro and to trastuzumab in two in vivo models. Specifically, the tripartite antibody mixture induced efficient HER2 internalization and degradation demonstrating increased sensitivity in cell lines with HER2 amplification and high EGFR levels. When compared with individual and clinically approved mAbs, the synergistic tripartite antibody targeting HER2 subdomains I, II, and IV demonstrates superior anticancer activity. Mol Cancer Ther; 14(3); 669–80. ©2015 AACR.
- Published
- 2015
39. Targeting the ERBB family in cancer: couples therapy
- Author
-
Mikkel W. Pedersen, Terrance Grant Johns, and Niall C. Tebbutt
- Subjects
biology ,Receptor, ErbB-2 ,ERBB Family ,business.industry ,Applied Mathematics ,General Mathematics ,Antibodies, Monoclonal ,Cancer ,Pharmacology ,medicine.disease ,Receptor tyrosine kinase ,ErbB Receptors ,Clinical trial ,Drug development ,Drug Resistance, Neoplasm ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,biology.protein ,medicine ,Cancer research ,Humans ,business ,Protein Kinase Inhibitors - Abstract
The ERBB family of receptor tyrosine kinases has a central role in the tumorigenesis of many types of solid tumour. Various therapeutics targeting these receptors have been approved for the treatment of several cancers. Considerable preclinical data have shown that the administration of two inhibitors against an individual ERBB family member--particularly epidermal growth factor receptor (EGFR) or ERBB2--leads to markedly higher antitumour activity than the administration of single agents. This Opinion article describes the preclinical and clinical performance of these dual-targeting approaches, discusses the key mechanisms that mediate their increased efficacy and highlights areas for ongoing investigation.
- Published
- 2013
40. Rational identification of an optimal antibody mixture for targeting the epidermal growth factor receptor
- Author
-
Josefine Nielsen Søderberg, Lucilla Steinaa, Mikkel W. Pedersen, John S. Haurum, Peter S. Andersen, Allan Jensen, Ida Kjær, Michael Kragh, Per-Johan Meijer, Klaus Koefoed, and Helle Jacobsen
- Subjects
Immunology ,Antineoplastic Agents ,Pharmacology ,Epitope ,Epitopes ,Mice ,In vivo ,Report ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Panitumumab ,Epidermal growth factor receptor ,Mice, Inbred BALB C ,Cetuximab ,biology ,business.industry ,Antibodies, Monoclonal ,Surface Plasmon Resonance ,Xenograft Model Antitumor Assays ,ErbB Receptors ,Treatment Outcome ,Epitope mapping ,Cancer cell ,Monoclonal ,biology.protein ,Female ,Immunization ,business ,Epitope Mapping ,medicine.drug - Abstract
The epidermal growth factor receptor (EGFR) is frequently dysregulated in human malignancies and a validated target for cancer therapy. Two monoclonal anti-EGFR antibodies (cetuximab and panitumumab) are approved for clinical use. However, the percentage of patients responding to treatment is low and many patients experiencing an initial response eventually relapse. Thus, the need for more efficacious treatments remains. Previous studies have reported that mixtures of antibodies targeting multiple distinct epitopes are more effective than single mAbs at inhibiting growth of human cancer cells in vitro and in vivo. The current work describes the rational approach that led to discovery and selection of a novel anti-EGFR antibody mixture Sym004, which is currently in Phase 2 clinical testing. Twenty-four selected anti-EGFR antibodies were systematically tested in dual and triple mixtures for their ability to inhibit cancer cells in vitro and tumor growth in vivo. The results show that targeting EGFR dependent cancer cells with mixtures of antibodies is superior at inhibiting their growth both in vitro and in vivo. In particular, antibody mixtures targeting non-overlapping epitopes on domain III are efficient and indeed Sym004 is composed of two monoclonal antibodies targeting this domain. The superior growth inhibitory activity of mixtures correlated with their ability to induce efficient EGFR degradation.
- Published
- 2011
41. Preclinical Pharmacokinetics and Safety of Sym004: A Synergistic Antibody Mixture Directed against Epidermal Growth Factor Receptor
- Author
-
Jette Wagtberg Sen, Mikkel W. Pedersen, Helle Jacobsen, Niels Jorgen Ostergaard Skartved, Thomas Kjærsgaard Jørgensen, Adam S. Hey, Pernille Foged Jensen, and Michael Kragh
- Subjects
Male ,Cancer Research ,medicine.drug_class ,Guinea Pigs ,Mice, Nude ,Antineoplastic Agents ,Cross Reactions ,Pharmacology ,Monoclonal antibody ,Mice ,Pharmacokinetics ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Epidermal growth factor receptor ,Adverse effect ,Mice, Inbred BALB C ,biology ,Cetuximab ,business.industry ,Antibodies, Monoclonal ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Antibodies, Anti-Idiotypic ,Rats ,ErbB Receptors ,Macaca fascicularis ,Oncology ,Toxicity ,biology.protein ,Female ,Rabbits ,Antibody ,business ,medicine.drug - Abstract
Purpose: Sym004 is a novel therapeutic antibody mixture product comprising two unmarketed monoclonal antibodies (mAb) targeting the epidermal growth factor receptor (EGFR). In previous preclinical proof-of-concept studies, Sym004 was shown to elicit superior cancer cell growth inhibition activities compared with marketed anti-EGFR mAbs. This article describes the design and results of the preclinical safety program conducted to support early clinical development of Sym004. Experimental Design: Tissue cryosections from various species were stained with Sym004 to evaluate tissue cross reactivity. The pharmacokinetics of Sym004 were evaluated in a mouse xenograft model and in Cynomolgus monkeys. Monkeys received once weekly intravenous infusions of Sym004 in the range 2 to 24 mg/kg for 6 to 8 weeks. Cetuximab (a marketed anti-EGFR mAb) and the individual antibodies comprising Sym004 were included in the repeat-dose toxicity studies at single-dose level. Results: Sym004 had a staining pattern similar to cetuximab in tissue panels from both human and non-human primates. Once weekly dosing of Sym004 to Cynomolgus monkeys did not cause accumulation, whereas administration of the individual antibodies resulted in prolonged half-life and accumulation. In direct comparisons with cetuximab, Sym004 did not induce any distinct or novel adverse findings in the animals. However, an early onset of pronounced, reversible, and anticipated anti-EGFR–mediated pharmacologic effects, such as skin rash, dehydration, and liquid feces, was observed. Only minor adverse effects were recorded in animals treated with the individual antibodies comprising Sym004. Conclusion: Sym004 was well tolerated and did not induce any unexpected toxicities. The preclinical safety data enabled initiation of the ongoing clinical development. Clin Cancer Res; 17(18); 5962–72. ©2011 AACR.
- Published
- 2011
42. Abstract 3822: Characterization of the first chicken-derived anti-PD-1 clinical stage antibody with a unique epitope and promising anticancer activity
- Author
-
Maria C. Melander, Monika Gad, Gunther R. Galler, Johan Lantto, Mikkel W. Pedersen, Klaus Koefoed, Thomas Bouquin, Camilla Fröhlich, Torben Gjetting, Ivan D. Horak, and Michael Kragh
- Subjects
Cancer Research ,Epitope mapping ,Oncology ,biology ,Immunization ,Antigen ,Antibody Repertoire ,biology.protein ,Syngenic ,Pembrolizumab ,Antibody ,Molecular biology ,Epitope - Abstract
Inhibition of immunologic checkpoints like Programmed Cell Death 1 (PD-1) has shown clinical efficacy in a broad range of cancers by improving or restoring T-cell activity. Anti-PD-1 antibodies show great promise in treating cancer malignances when administered alone or in combination with other immune activating approaches. However, high protein sequence identity between human and mammalian species used for antibody generation often disfavor generation of antibodies against functionally conserved epitopes, or prevents isolating antibodies cross reacting with ortholog species used for evaluating potential toxicity. Chickens are phylogenetically distant from mammals and are better at generating antibodies against epitopes that are conserved in mammals. Because chickens generate antibodies from a very restricted set of V-gene germline genes that are diversified by “gene conversion”, we envisioned that high throughput humanization of antibody frameworks was achievable by “mass CDR grafting” after recovering antibodies by immunization and B-cell cloning. Wild type chickens were immunized with PD-1 antigen, and a repertoire of 120 antibodies was generated with Symplex™ technology, by combining single B-cell FACS sorting and high throughput RT-PCR cloning of cognate VH and VL chains. The isolated PD-1 repertoire was cloned with an inert Fc backbone and humanized by a combination of in silico CDR grafting and gene synthesis. Humanized antibodies were expressed and screened for retained binding affinity and functionality in T-cell based assays. We successfully generated a humanized PD-1 antibody repertoire and found that most antibodies retained affinity and functionality similar to that of parental chicken antibodies. Furthermore, the antibody repertoire displayed broad binding epitope coverage on PD-1, often with strong pM affinity, and showed biophysical properties acceptable for drug development. Our lead antibody Sym021 blocked PD-L1 and PD-L2 ligand binding and downstream PD-1 signaling, resulting in elevated T-cell cytokine production in vitro. Moreover, Sym021 bound human PD-1 with much stronger affinity of 30 pM compared to clinical PD-1 mAbs nivolumab and pembrolizumab, while also cross reacting to cynomolgous and mouse PD-1. This enabled direct testing of Sym021 in syngenic mouse in vivo models and evaluation of preclinical toxicology in cynomolgus monkeys. Detailed epitope mapping showed that the epitope recognized by Sym021 was unique compared to clinical antibodies pembrolizumab and nivolumab. These results supported entry of PD-1 targeting Sym021 into clinical trials. Citation Format: Torben Gjetting, Monika Gad, Camilla Fröhlich, Maria C. Melander, Gunther Galler, Johan Lantto, Thomas Bouquin, Ivan D. Horak, Michael Kragh, Mikkel W. Pedersen, Klaus Koefoed. Characterization of the first chicken-derived anti-PD-1 clinical stage antibody with a unique epitope and promising anticancer activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3822.
- Published
- 2018
43. Abstract 5629: Preclinical characterization of Sym023 a human anti-TIM3 antibody with a novel mechanism of action
- Author
-
Torben Gjetting, Michael Kragh, Mikkel W. Pedersen, Klaus Koefoed, Johan Lantto, Camilla Frölich, Monika Gad, Ivan D. Horak, Vikram Kjoller Bhatia, Michael V. Grandal, and Trine Lindsted
- Subjects
0301 basic medicine ,Cancer Research ,biology ,medicine.medical_treatment ,T cell ,Immunotherapy ,03 medical and health sciences ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Immune system ,Oncology ,Mechanism of action ,medicine ,biology.protein ,Cancer research ,Cytotoxic T cell ,medicine.symptom ,Antibody ,Galectin - Abstract
Immunotherapy has become a major focus of research in oncology and blockade of immune checkpoints such as cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4) and programmed cell death protein 1 (PD1) has been some of the most successful immunotherapies. The next wave of immunomodulatory targets that are being explored for cancer therapy include T cell immunoglobulin and mucin domain protein 3 (TIM3). TIM3 is constitutively expressed on cells of myeloid origin whereas the TIM3 expression is induced on T-cells upon activation. The exact function of TIM3 on the different immune cells is not clear and may be context dependent suggesting that TIM3 is not a classical immune check-point. Sym023 is a human anti-TIM3 antibody, which binds human TIM3 and cross-reacts with cynomolgus monkey TIM3. Sym023 blocks binding of phosphatidyl serine but not galectin 9 and stimulates T-cell proliferation in mixed lymphocyte reactions and tumor growth inhibition in vivo. Here, we present data demonstrating that ligation of TIM3 by Sym023 increase cytokine production and T cell proliferation in vitro through a novel mechanism of action. Citation Format: Trine Lindsted, Monika Gad, Michael V. Grandal, Camilla Frölich, Vikram K. Bhatia, Torben Gjetting, Johan Lantto, Ivan D. Horak, Michael Kragh, Klaus Koefoed, Mikkel W. Pedersen. Preclinical characterization of Sym023 a human anti-TIM3 antibody with a novel mechanism of action [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5629.
- Published
- 2018
44. Abstract 5626: Preclinical characterization of Sym022, a novel anti-LAG3 antibody
- Author
-
Mikkel W. Pedersen, Ivan D. Horak, Vikram Kjoller Bhatia, Torben Gjetting, Maria C. Melander, Michael M. Grandal, Klaus Kofoed, Trine Lindsted, Camilla Fröhlich, Michael Kragh, and Johan Lantto
- Subjects
0301 basic medicine ,Cancer Research ,LAG3 ,biology ,Chemistry ,medicine.drug_class ,Tumor-infiltrating lymphocytes ,medicine.medical_treatment ,Immunotherapy ,Monoclonal antibody ,03 medical and health sciences ,030104 developmental biology ,Cytokine ,Oncology ,Cancer research ,biology.protein ,medicine ,Cytotoxic T cell ,Antibody ,Antigen-presenting cell - Abstract
Immunotherapy has become a major focus of research in oncology and blockade of immune checkpoints such as cytotoxic T-Lymphocyte associated protein 4 (CTLA4) and programmed cell death protein 1 (PD1) has been some of the most successful immunotherapies. Lymphocyte-activation gene 3 (LAG3) belongs to the second-generation immune modulatory targets. LAG3 is expressed by activated T-cells and tumor infiltrating lymphocytes (TILs) and negatively regulates T-cell activity upon ligand engagement. LAG3 binds major histocompatibility complex class II (MHCII) molecules found on the surface of antigen presenting cells and tumor cells. Sym022 is a Fc-inert human monoclonal antibody targeting LAG3. Sym022 binds to human and cynomolgus monkey LAG3 with high affinity and blocks the interaction between LAG3 and MHCII molecules. Functionally, Sym022 increases cytokine production by T-cells in vitro and tumor growth inhibition in vivo. Mechanistically, Sym022 not only blocks ligand binding, but also decreases total LAG3 surface levels through internalization and/or shedding. Citation Format: Michael M. Grandal, Maria C. Melander, Vikram K. Bhatia, Torben Gjetting, Trine Lindsted, Camilla Fröhlich, Johan Lantto, Ivan D. Horak, Michael Kragh, Klaus Kofoed, Mikkel W. Pedersen. Preclinical characterization of Sym022, a novel anti-LAG3 antibody [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5626.
- Published
- 2018
45. Circulating tumor DNA profiling to identify patients with metastatic colorectal cancer with improved overall survival following Sym004 treatment
- Author
-
Thorarinn Blondal, Ivan D. Horak, Mikkel W. Pedersen, Josep Tabernero, Scott Kopetz, Rodrigo Dienstmann, C. Ding, Clara Montagut, Michael Kragh, Thomas Tuxen Poulsen, and Trine Lindsted
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,Colorectal cancer ,business.industry ,medicine.disease ,digestive system diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,biology.protein ,Overall survival ,In patient ,Antibody ,business - Abstract
e15577Background: Sym004, an antibody mixture targeting EGFR, has demonstrated promising activity in patients (pts) with anti-EGFR refractory metastatic colorectal cancer (mCRC). A Phase 2b trial e...
- Published
- 2018
46. Efficacy of Sym004 in Patients With Metastatic Colorectal Cancer With Acquired Resistance to Anti-EGFR Therapy and Molecularly Selected by Circulating Tumor DNA Analyses
- Author
-
Françoise Desseigne, Klaus Koefoed, Mikkel W. Pedersen, Rodrigo Dienstmann, C. Ding, Thomas Tuxen Poulsen, Trine Lindsted, Joana Vidal, Jenifer Clausell-Tormos, Michael M. Grandal, Salvatore Siena, Ana Rovira, Giulia Siravegna, Francisco J. Sánchez-Martín, Giuseppe Rospo, Guillem Argiles, Antonio Cubillo, Alberto Bardelli, Cristina Nadal, Michael Kragh, Clara Montagut, Ivan D. Horak, Lucjan Wyrwicz, Mikhail Dvorkin, Joan Albanell, Scott Kopetz, Guglielmo Fumi, Paul Nadler, Fortunato Ciardiello, Ramon Salazar, Josep Tabernero, Vittorina Zagonel, Montagut, Clara, Argilés, Guillem, Ciardiello, Fortunato, Poulsen, Thomas T, Dienstmann, Rodrigo, Kragh, Michael, Kopetz, Scott, Lindsted, Trine, Ding, Cliff, Vidal, Joana, Clausell-Tormos, Jenifer, Siravegna, Giulia, Sánchez-Martín, Francisco J, Koefoed, Klau, Pedersen, Mikkel W, Grandal, Michael M, Dvorkin, Mikhail, Wyrwicz, Lucjan, Rovira, Ana, Cubillo, Antonio, Salazar, Ramon, Desseigne, Françoise, Nadal, Cristina, Albanell, Joan, Zagonel, Vittorina, Siena, Salvatore, Fumi, Guglielmo, Rospo, Giuseppe, Nadler, Paul, Horak, Ivan D, Bardelli, Alberto, and Tabernero, Josep
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Population ,Loading dose ,Circulating Tumor DNA ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Neoplasm Metastasis ,education ,Protein Kinase Inhibitors ,Survival analysis ,Aged ,Aged, 80 and over ,education.field_of_study ,business.industry ,Patient Selection ,Hazard ratio ,Antibodies, Monoclonal ,Correction ,Middle Aged ,medicine.disease ,Survival Analysis ,3. Good health ,ErbB Receptors ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,Colorectal Neoplasms ,business - Abstract
Importance Acquired resistance to anti-EGFR therapy (epidermal growth factor receptor) is frequently due toRASandEGFRextracellular domain (ECD) mutations in metastatic colorectal cancer (mCRC). Some anti-EGFR–refractory patients retain tumor EGFR dependency potentially targetable by agents such as Sym004, which is a mixture of 2 nonoverlapping monoclonal antibodies targeting EGFR. Objective To determine if continuous blockade of EGFR by Sym004 has survival benefit. Design, Setting, and Participants Multicenter, phase 2, randomized, clinical trial comparing 2 regimens of Sym004 with investigator’s choice from March 6, 2014, through October 15, 2015. Circulating tumor DNA (ctDNA) was analyzed for biomarker and tracking clonal dynamics during treatment. Participants had wild-typeKRASexon 2 mCRC refractory to standard chemotherapy and acquired resistance to anti-EGFR monoclonal antibodies. Interventions Participants were randomly assigned in a 1:1:1 ratio to Sym004, 12 mg/kg/wk (arm A), Sym004, 9 mg/kg loading dose followed by 6 mg/kg/wk (arm B), or investigator’s choice of treatment (arm C). Main Outcomes and Measures Overall survival (OS). Secondary end points included preplanned exploratory biomarker analysis in ctDNA. Results A total of 254 patients were randomized (intent-to-treat [ITT] population) (median age, 63 [range, 34-91] years; 63% male; n = 160). Median OS in the ITT population was 7.9 months (95% CI, 6.5-9.9 months), 10.3 months (95% CI, 9.0-12.9 months), and 9.6 months (95% CI, 8.3-12.2 months) for arms A, B, and C, respectively (hazard ratio [HR], 1.31; 95% CI, 0.92-1.87 for A vs C; and HR, 0.97; 95% CI, 0.68-1.40 for B vs C). The ctDNA revealed high intrapatient genomic heterogeneity following anti-EGFR therapy. Sym004 effectively targetedEGFRECD-mutated cancer cells, and a decrease inEGFRECD ctDNA occurred in Sym004-treated patients. However, this did not translate into clinical benefit in patients withEGFRECD mutations, likely owing to co-occurring resistance mechanisms. A subgroup of patients was defined by ctDNA (RAS/BRAF/EGFRECD-mutation negative) associated with improved OS in Sym004-treated patients in arm B compared with arm C (median OS, 12.8 and 7.3 months, respectively). Conclusions and Relevance Sym004 did not improve OS in an unselected population of patients with mCRC and acquired anti-EGFR resistance. A prospective clinical validation of Sym004 efficacy in a ctDNA molecularly defined subgroup of patients with refractory mCRC is warranted. Trial Registration clinicaltrialsregister.eu Identifier:2013-003829-29
- Published
- 2018
47. Sym004: A Novel Synergistic Anti–Epidermal Growth Factor Receptor Antibody Mixture with Superior Anticancer Efficacy
- Author
-
Mikkel W. Pedersen, Klaus Koefoed, Helle Jacobsen, Charles Pyke, Michael Kragh, John S. Haurum, and Adam S. Hey
- Subjects
Cancer Research ,medicine.drug_class ,media_common.quotation_subject ,Down-Regulation ,Mice, Nude ,Cell Growth Processes ,Pharmacology ,Monoclonal antibody ,Epitope ,Epitopes ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Phosphorylation ,Internalization ,Receptor ,media_common ,Mice, Inbred BALB C ,biology ,Antibodies, Monoclonal ,Drug Synergism ,Xenograft Model Antitumor Assays ,Protein Structure, Tertiary ,ErbB Receptors ,Oncology ,chemistry ,Cancer cell ,biology.protein ,Growth inhibition - Abstract
Epidermal growth factor receptor (EGFR) is a validated therapeutic target in cancer and EGFR antagonists with greater effectiveness than existing clinical agents remain of interest. Here, we report a novel approach based on Sym004, a mixture of two anti-EGFR monoclonal antibodies directed against distinct nonoverlapping epitopes in EGFR extracellular domain III. Like anti-EGFR monoclonal antibodies in current clinical use, Sym004 inhibits cancer cell growth and survival by blocking ligand-binding receptor activation and phosphorylation and downstream receptor signaling. However, unlike the other antibodies, Sym004 induces rapid and efficient removal of the receptor from the cancer cell surface by triggering EGFR internalization and degradation. Compared with reference anti-EGFR monoclonal antibodies, Sym004 exhibited more pronounced growth inhibition in vitro and superior efficacy in vivo. Together, these findings illustrate a strategy to target EGFR more effectively than existing clinical antibodies. Cancer Res; 70(2); 588–97
- Published
- 2010
48. EGFR induces expression of IRF-1viaSTAT1 and STAT3 activation leading to growth arrest of human cancer cells
- Author
-
Anders Woetmann, Mikkel W. Pedersen, Marie Thérèse Stockhausen, Niels Ødum, Mette Villingshøj, Peter Andersen, and Hans Skovgaard Poulsen
- Subjects
Cancer Research ,biology ,Cell growth ,Kinase ,medicine.medical_treatment ,Cytokine ,Oncology ,Cancer cell ,Cancer research ,medicine ,biology.protein ,Epidermal growth factor receptor ,Signal transduction ,STAT3 ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Recently, we reported that epidermal growth factor receptor (EGFR) induce expression of a module of genes known to be inducible by interferons and particularly interferon-gamma. Here we show that the module is tightly regulated by EGFR in the 2 human cancer cell lines that overexpress EGFR, A431 and HN5. The module of genes included the tumor suppressor IRF-1, which was used as a prototypical member to further investigate the regulation and function of the module. Ligand-activated EGFR induce expression of IRF-1 via phosphorylation of STAT1 and STAT3. In contrast, cells expressing the constitutively active cancer specific receptor EGFRvIII are unable to mediate phosphorylation of these STATs and thereby incapable of inducing IRF-1. We also demonstrate that IRF-1 is expressed in an EGF dose-dependent manner, which correlates with inhibition of cell proliferation, and that the regulation of IRF-1 is partially dependent on intracellular Src family kinase activity. Treatment with the dual specific Abl/c-Src kinase inhibitor AZD0530 significantly reduces the growth inhibitory effect of high EGF concentrations, signifying that EGFR induced IRF-1 is responsible for the observed growth inhibition. In addition, we show that media from these EGF treated cancer cells upregulate the activation marker CD69 on both B-cells and T-cells in peripheral blood. Taken together, these results suggest that cells acquiring sustained high activity of oncogenes such as EGFR are able to activate genes, whose products mediate growth arrest and activate immune effector cells, and which potentially could be involved in alerting the immune system in vivo leading to elimination of the transformed cells.
- Published
- 2007
49. EGFRvIII escapes down-regulation due to impaired internalization and sorting to lysosomes
- Author
-
Mikkel W. Pedersen, Berthe M. Willumsen, Hans Skovgaard Poulsen, Bo van Deurs, Michael Vibo Grandal, and Roza Zandi
- Subjects
Cancer Research ,media_common.quotation_subject ,Down-Regulation ,Cell Line, Tumor ,Humans ,Proto-Oncogene Proteins c-cbl ,Epidermal growth factor receptor ,Phosphorylation ,Internalization ,Receptor ,Ubiquitins ,GRB2 Adaptor Protein ,media_common ,Epidermal Growth Factor ,biology ,Cell Membrane ,General Medicine ,Growth Factor Receptor-Bound Protein 2 ,Endocytosis ,Ubiquitin ligase ,Cell biology ,ErbB Receptors ,Protein Transport ,Biochemistry ,biology.protein ,GRB2 ,Signal transduction ,Lysosomes ,Signal Transduction - Abstract
EGFRvIII is a mutant variant of the epidermal growth factor receptor (EGFR) found exclusively in various cancer types. EGFRvIII lacks a large part of the extracellular domain and is unable to bind ligands; however, the receptor is constitutively phosphorylated and able to activate downstream signaling pathways. Failure to attenuate signaling by receptor down-regulation could be one of the major mechanisms by which EGFRvIII becomes oncogenic. Using a cell system expressing either EGFR or EGFRvIII with no expression of other EGFR family members and with endogenous levels of key degradation proteins, we have investigated the down-regulation of EGFRvIII and compared it to that of EGFR. We show that, in contrast to EGFR, EGFRvIII is inefficiently degraded. EGFRvIII is internalized, but the internalization rate of the mutated receptor is significantly less than that of unstimulated EGFR. Moreover, internalized EGFRvIII is recycled rather than delivered to lysosomes. EGFRvIII binds the ubiquitin ligase c-Cbl via Grb2, whereas binding via phosphorylated tyrosine residue 1045 seems to be limited. Despite c-Cbl binding, the receptor fails to become effectively ubiquitinylated. Thus, our results suggest that the long lifetime of EGFRvIII is caused by inefficient internalization and impaired sorting to lysosomes due to lack of effective ubiquitinylation.
- Published
- 2007
50. Activation of the EGFR Gene Target EphA2 Inhibits Epidermal Growth Factor–Induced Cancer Cell Motility
- Author
-
Alice Bjerregaard Larsen, Mikkel W. Pedersen, Michael Vibo Grandal, Marie-Thérése Stockhausen, Bo van Deurs, and Hans Skovgaard Poulsen
- Subjects
Transcriptional Activation ,Cancer Research ,Biology ,Metastasis ,Cell Movement ,Epidermal growth factor ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,ERBB3 ,Epidermal growth factor receptor ,RNA, Small Interfering ,Promoter Regions, Genetic ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Epidermal Growth Factor ,Receptor, EphA2 ,Cell Membrane ,Cancer ,MAP Kinase Kinase Kinases ,medicine.disease ,Cell biology ,ErbB Receptors ,Oncology ,Protein Biosynthesis ,Cancer cell ,Cancer research ,biology.protein ,A431 cells - Abstract
EphA2 overexpression has been reported in many cancers and is believed to play an important role in tumor metastasis and angiogenesis. We show that the activated epidermal growth factor receptor (EGFR) and the cancer-specific constitutively active EGFR type III deletion mutant (EGFRvIII) induce the expression of EphA2 in mammalian cell lines, including the human cancer cell lines A431 and HN5. The regulation is partially dependent on downstream activation of mitogen-activated protein kinase/extracellular signal–regulated kinase kinase and is a direct effect on the EphA2 promoter. Furthermore, EGFR and EphA2 both localize to the plasma membrane and EphA2 coimmunoprecipitates with activated EGFR and EGFRvIII. Ligand activation of EphA2 and EphA2 knockdown by small interfering RNA inhibit EGF-induced cell motility of EGFR-overexpressing human cancer cells, indicating a functional role of EphA2 in EGFR-expressing cancer cells. (Mol Cancer Res 2007;5(3):283–93)
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.