51
|
Enya Chen YC, Burgess M, Mapp S, Mollee P, Gill D, Blumenthal A, Saunders NA. PI3K-p110δ contributes to antibody responses by macrophages in chronic lymphocytic leukemia. Leukemia 2019; 34:451-461. [PMID: 31462739 DOI: 10.1038/s41375-019-0556-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/01/2019] [Accepted: 07/15/2019] [Indexed: 11/10/2022]
Abstract
Fcγ receptor (FcγR) signalling in monocyte derived macrophages from chronic lymphocytic leukaemia (CLL) patients is poorly understood. This signalling pathway is the key determinant of the ability of the macrophages to respond to therapeutic antibodies in current clinical use for CLL. Muted FcγR signalling activity accompanies disease progression and results in resistance to therapeutic antibodies. The molecular mechanisms controlling FcγR signalling and resistance are unknown. Here, we demonstrate that the class I phosphoinositide 3-kinase (PI3K) catalytic subunit p110δ is essential for CLL-derived macrophages to respond to therapeutic antibodies. Inhibition of p110δ in the macrophages reduces FcγR-mediated antibody immune responses. Surprisingly, our studies indicated that FcγR downstream signalling is independent of SYK and BTK activity. Thus, we show that FcγR antibody responses occur via a previously unidentified p110δ-dependent pathway, which is independent of the previously described SYK/BTK activation pathway. These data provide novel insights into the effectors of antibody responses. Our data also provide mechanistic insights into therapy resistance in CLL.
Collapse
Affiliation(s)
- Yu-Chen Enya Chen
- University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia
| | - Melinda Burgess
- University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia.,Department of Haematology, Cancer Services Unit, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Sally Mapp
- Department of Haematology, Cancer Services Unit, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Peter Mollee
- Department of Haematology, Cancer Services Unit, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Devinder Gill
- Department of Haematology, Cancer Services Unit, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Antje Blumenthal
- University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia
| | - Nicholas A Saunders
- University of Queensland Diamantina Institute, Woolloongabba, QLD, Australia.
| |
Collapse
|
52
|
Houston MT, Gutierrez JB. The FRiND Model: A Mathematical Model for Representing Macrophage Plasticity in Muscular Dystrophy Pathogenesis. Bull Math Biol 2019; 81:3976-3997. [PMID: 31302876 PMCID: PMC6764940 DOI: 10.1007/s11538-019-00635-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 06/19/2019] [Indexed: 12/04/2022]
Abstract
Muscular dystrophy describes generalized progressive muscular weakness due to the wasting of muscle fibers. The progression of the disease is affected by known immunological and mechanical factors, and possibly other unknown mechanisms. This article introduces a new mathematical model, the FRiND model, to further elucidate these known immunological actions. We will perform stability and sensitivity analyses on this model. The models time course results will be verified by biological studies in the literature. This model could be the foundation for further understanding of immunological muscle repair.
Collapse
Affiliation(s)
- Matthew T Houston
- Department of Mathematics, Middle Georgia State University, Macon, GA, 31206, USA. .,University of Georgia, Athens, GA, 30602, USA.
| | | |
Collapse
|
53
|
Mastrangeli R, Palinsky W, Bierau H. Glycoengineered antibodies: towards the next-generation of immunotherapeutics. Glycobiology 2019; 29:199-210. [PMID: 30289453 DOI: 10.1093/glycob/cwy092] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/23/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
Monoclonal antibodies (mAbs) are currently the largest and fastest growing class of biopharmaceuticals, and they address unmet medical needs, e.g., in oncology and in auto-immune diseases. Their clinical efficacy and safety is significantly affected by the structure and composition of their glycosylation profile which is commonly heterogeneous, heavily dependent on the manufacturing process, and thus susceptible to variations in the cell culture conditions. Glycosylation is therefore considered a critical quality attribute for mAbs. Commonly, in currently marketed therapeutic mAbs, the glycosylation profile is suboptimal in terms of biological properties such as antibody-dependent cell-mediated cytotoxicity or may give rise to safety concerns due to the presence of non-human glycans. This article will review recent innovative developments in chemo-enzymatic glycoengineering, which allow generating mAbs carrying single, well-defined, uniform Fc glycoforms, which confers the desired biological properties for the target application. This approach offers significant benefits such as enhanced Fc effector functions, improved safety profiles, higher batch-to-batch consistency, decreased risks related to immunogenicity and manufacturing process changes, and the possibility to manufacture mAbs, in an economical manner, in non-mammalian expression systems. Overall, this approach could facilitate and reduce mAb manufacturing costs which in turn would translate into tangible benefits for both patients and manufacturers. The first glycoengineered mAbs are about to enter clinical trials and it is expected that, once glycoengineering reagents are available at affordable costs, and in-line with regulatory requirements, that targeted remodeling of antibody Fc glycosylation will become an integral part in manufacturing the next-generation of immunotherapeutics.
Collapse
Affiliation(s)
- Renato Mastrangeli
- Biotech Development Programme, CMC Science & Intelligence, Merck Serono SpA, an affiliate of Merck KgaA, Darmstadt, Germany. Via Luigi Einaudi, 11. Guidonia Montecelio (Roma), Italy
| | - Wolf Palinsky
- Biotech Development Programme, Merck Biopharma, an affiliate of Merck KgaA, Darmstadt, Germany. Zone Industrielle de l'Ouriettaz, Aubonne, Switzerland
| | - Horst Bierau
- Biotech Development Programme, CMC Science & Intelligence, Merck Serono SpA, an affiliate of Merck KgaA, Darmstadt, Germany. Via Luigi Einaudi, 11. Guidonia Montecelio (Roma), Italy
| |
Collapse
|
54
|
Pellizzari G, Hoskin C, Crescioli S, Mele S, Gotovina J, Chiaruttini G, Bianchini R, Ilieva K, Bax HJ, Papa S, Lacy KE, Jensen-Jarolim E, Tsoka S, Josephs DH, Spicer JF, Karagiannis SN. IgE re-programs alternatively-activated human macrophages towards pro-inflammatory anti-tumoural states. EBioMedicine 2019; 43:67-81. [PMID: 30956175 PMCID: PMC6562024 DOI: 10.1016/j.ebiom.2019.03.080] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Antibody Fc-driven engagement of macrophages is critical for evoking cellular activation and effector functions and influencing tumour-associated macrophage (TAM) recruitment. We previously reported that IgE class antibodies promote restriction of cancer growth in rodent models associated with significant TAM infiltration. However, the human macrophage-associated IgE-Fc Receptor (FcεR) axis remains unexplored. We investigated the effects of anti-tumour IgE stimulation on human macrophage activation. METHODS Human blood monocyte-differentiated quiescent (M0), classically-(M1) and alternatively-(M2) activated macrophages were crosslinked with IgE and polyclonal antibodies to mimic immune complex formation. We examined surface marker expression, cytokine secretion, protein kinase phosphorylation and gene expression in IgE-stimulated macrophages and IgE antibody-dependent macrophage-mediated cytotoxicity (ADCC) against tumour cells. FINDINGS A proportion (40%) of M2 and (<20%) M0 and M1 macrophages expressed the high-affinity IgE receptor FcεRI. IgE crosslinking triggered upregulation of co-stimulatory CD80, increased TNFα, IFNγ, IL-1β, IL-12, IL-10, IL-13, CXCL9, CXCL11 and RANTES secretion by M0 and M2 and additionally enhanced MCP-1 by M2 macrophages. IgE-stimulated M1 macrophages retained secretion of pro-inflammatory cytokines. IgE crosslinking enhanced the FcεRI-dependent signalling pathway, including phosphorylation of the Lyn kinase, ERK1/2 and p38 in M2 macrophages and upregulated Lyn gene expression by M1 and M2 macrophages. Anti-tumour IgE engendered ADCC of cancer cells by all macrophage subsets. INTERPRETATION IgE can engage and re-educate alternatively-activated macrophages towards pro-inflammatory phenotypes and prime all subsets to mediate anti-tumour functions. This points to IgE-mediated cascades with potential to activate immune stroma and may be significant in the clinical development of strategies targeting tumour-resident macrophages.
Collapse
Affiliation(s)
- Giulia Pellizzari
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Coran Hoskin
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London WC2B 4BG, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Jelena Gotovina
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria; Department of Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Giulia Chiaruttini
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Rodolfo Bianchini
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria; Department of Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Kristina Ilieva
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom; Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Heather J Bax
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom; School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Sophie Papa
- School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom; Guy's and St Thomas' NHS Trust, Department of Medical Oncology, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria; Department of Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London WC2B 4BG, United Kingdom
| | - Debra H Josephs
- School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom; Guy's and St Thomas' NHS Trust, Department of Medical Oncology, London, United Kingdom
| | - James F Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom.
| |
Collapse
|
55
|
Kamen L, Myneni S, Langsdorf C, Kho E, Ordonia B, Thakurta T, Zheng K, Song A, Chung S. A novel method for determining antibody-dependent cellular phagocytosis. J Immunol Methods 2019; 468:55-60. [DOI: 10.1016/j.jim.2019.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 10/27/2022]
|
56
|
Kang TH, Lee CH, Delidakis G, Jung J, Richard-Le Goff O, Lee J, Kim JE, Charab W, Bruhns P, Georgiou G. An Engineered Human Fc variant With Exquisite Selectivity for FcγRIIIa V158 Reveals That Ligation of FcγRIIIa Mediates Potent Antibody Dependent Cellular Phagocytosis With GM-CSF-Differentiated Macrophages. Front Immunol 2019; 10:562. [PMID: 30984171 PMCID: PMC6448688 DOI: 10.3389/fimmu.2019.00562] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/04/2019] [Indexed: 01/27/2023] Open
Abstract
IgG antibodies mediate the clearance of target cells via the engagement of Fc gamma receptors (FcγRs) on effector cells by eliciting antibody-dependent cellular cytotoxicity and phagocytosis (ADCC and ADCP, respectively). Because (i) the IgG Fc domain binds to multiple FcγRs with varying affinities; (ii) even low Fc:FcγRs affinity interactions can play a significant role when antibodies are engaged in high avidity immune complexes and (iii) most effector cells express multiple FcγRs, the clearance mechanisms that can be mediated by individual FcγR are not well-understood. Human FcγRIIIa (hFcγRIIIa; CD16a), which exists as two polymorphic variants at position 158, hFcγRIIIaV158 and hFcγRIIIaF158, is widely considered to only trigger ADCC, especially with natural killer (NK) cells as effectors. To evaluate the role of hFcγRIIIa ligation in myeloid-derived effector cells, and in particular on macrophages and monocytes which express multiple FcγRs, we engineered an aglycosylated engineered human Fc (hFc) variant, Fc3aV, which binds exclusively to hFcγRIIIaV158. Antibodies formatted with the Fc3aV variant bind to the hFcγRIIIaV158 allotype with a somewhat lower KD than their wild type IgG1 counterparts, but not to any other hFcγR. The exceptional selectivity for hFcγRIIIaV158 was demonstrated by SPR using increased avidity, dimerized GST-fused versions of the ectodomains of hFcγRs and from the absence of binding of large immune complex (IC) to CHO cells expressing each of the hFcγRs, including notably, the FcγRIIIaF158 variant or the highly homologous FcγRIIIb. We show that even though monocyte-derived GM-CSF differentiated macrophages express hFcγRIIIa at substantially lower levels than the other two major activating receptors, namely hFcγRI or hFcγRIIa, Fc3aV-formatted Rituximab and Herceptin perform ADCP toward CD20- and Her2-expressing cancer cells, respectively, at a level comparable to that of the respective wild-type antibodies. We further show that hFcγRIIIa activation plays a significant role on ADCC by human peripheral monocytes. Our data highlight the utility of Fc3aV and other similarly engineered exquisitely selective, aglycosylated Fc variants toward other hFcγRs as tools for the detailed molecular understanding of hFcγR biology.
Collapse
Affiliation(s)
- Tae Hyun Kang
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Chang-Han Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Jiwon Jung
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Odile Richard-Le Goff
- Unit of Antibodies in Therapy and Pathology, Department of Immunology, Institut Pasteur, Paris, France
| | - Jiwon Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Jin Eyun Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Wissam Charab
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Department of Immunology, Institut Pasteur, Paris, France.,INSERM, U1222, Paris, France
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States.,Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States.,Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, United States.,Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
57
|
Patel KR, Roberts JT, Barb AW. Multiple Variables at the Leukocyte Cell Surface Impact Fc γ Receptor-Dependent Mechanisms. Front Immunol 2019; 10:223. [PMID: 30837990 PMCID: PMC6382684 DOI: 10.3389/fimmu.2019.00223] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
Fc γ receptors (FcγR) expressed on the surface of human leukocytes bind clusters of immunoglobulin G (IgG) to induce a variety of responses. Many therapeutic antibodies and vaccine-elicited antibodies prevent or treat infectious diseases, cancers and autoimmune disorders by binding FcγRs, thus there is a need to fully define the variables that impact antibody-induced mechanisms to properly evaluate candidate therapies and design new intervention strategies. A multitude of factors influence the IgG-FcγR interaction; one well-described factor is the differential affinity of the six distinct FcγRs for the four human IgG subclasses. However, there are several other recently described factors that may prove more relevant for disease treatment. This review covers recent reports of several aspects found at the leukocyte membrane or outside the cell that contribute to the cell-based response to antibody-coated targets. One major focus is recent reports covering post-translational modification of the FcγRs, including asparagine-linked glycosylation. This review also covers the organization of FcγRs at the cell surface, and properties of the immune complex. Recent technical advances provide high-resolution measurements of these often-overlooked variables in leukocyte function and immune system activation.
Collapse
Affiliation(s)
- Kashyap R Patel
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Jacob T Roberts
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| |
Collapse
|
58
|
Gunn BM, Yu WH, Karim MM, Brannan JM, Herbert AS, Wec AZ, Halfmann PJ, Fusco ML, Schendel SL, Gangavarapu K, Krause T, Qiu X, He S, Das J, Suscovich TJ, Lai J, Chandran K, Zeitlin L, Crowe JE, Lauffenburger D, Kawaoka Y, Kobinger GP, Andersen KG, Dye JM, Saphire EO, Alter G. A Role for Fc Function in Therapeutic Monoclonal Antibody-Mediated Protection against Ebola Virus. Cell Host Microbe 2019; 24:221-233.e5. [PMID: 30092199 DOI: 10.1016/j.chom.2018.07.009] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/04/2018] [Accepted: 07/20/2018] [Indexed: 11/28/2022]
Abstract
The recent Ebola virus (EBOV) epidemic highlighted the need for effective vaccines and therapeutics to limit and prevent outbreaks. Host antibodies against EBOV are critical for controlling disease, and recombinant monoclonal antibodies (mAbs) can protect from infection. However, antibodies mediate an array of antiviral functions including neutralization as well as engagement of Fc-domain receptors on immune cells, resulting in phagocytosis or NK cell-mediated killing of infected cells. Thus, to understand the antibody features mediating EBOV protection, we examined specific Fc features associated with protection using a library of EBOV-specific mAbs. Neutralization was strongly associated with therapeutic protection against EBOV. However, several neutralizing mAbs failed to protect, while several non-neutralizing or weakly neutralizing mAbs could protect. Antibody-mediated effector functions, including phagocytosis and NK cell activation, were associated with protection, particularly for antibodies with moderate neutralizing activity. This framework identifies functional correlates that can inform therapeutic and vaccine design strategies against EBOV and other pathogens.
Collapse
Affiliation(s)
- Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Wen-Han Yu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Marcus M Karim
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jennifer M Brannan
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Andrew S Herbert
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Anna Z Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peter J Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin, Madison, WI 53706, USA
| | - Marnie L Fusco
- Department of Immunology and Microbiology, The Scripps Research Institute, The Skaggs Institute for Chemical Biology, La Jolla, CA 92037, USA
| | - Sharon L Schendel
- Department of Immunology and Microbiology, The Scripps Research Institute, The Skaggs Institute for Chemical Biology, La Jolla, CA 92037, USA
| | - Karthik Gangavarapu
- Department of Immunology and Microbiology, The Scripps Research Institute, The Skaggs Institute for Chemical Biology, La Jolla, CA 92037, USA
| | - Tyler Krause
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Shinhua He
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Jishnu Das
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Todd J Suscovich
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jonathan Lai
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin, Madison, WI 53706, USA
| | - Gary P Kobinger
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; Université Laval Quebec, Québec, QC G1V 0A6, Canada
| | - Kristian G Andersen
- Department of Immunology and Microbial Science, Scripps Translational Science Institute, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John M Dye
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbiology, The Scripps Research Institute, The Skaggs Institute for Chemical Biology, La Jolla, CA 92037, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
59
|
Bone Marrow Mast Cell Antibody-Targetable Cell Surface Protein Expression Profiles in Systemic Mastocytosis. Int J Mol Sci 2019; 20:ijms20030552. [PMID: 30696068 PMCID: PMC6387409 DOI: 10.3390/ijms20030552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/30/2022] Open
Abstract
Despite recent therapeutic advances, systemic mastocytosis (SM) remains an incurable disease due to limited complete remission (CR) rates even after novel therapies. To date, no study has evaluated the expression on SM bone marrow mast cells (BMMC) of large panel of cell surface suitable for antibody-targeted therapy. In this study, we analyzed the expression profile of six cell-surface proteins for which antibody-based therapies are available, on BMMC from 166 SM patients vs. 40 controls. Overall, variable patterns of expression for the markers evaluated were observed among SM BMMC. Thus, CD22, CD30, and CD123, while expressed on BMMC from patients within every subtype of SM, showed highly variable patterns with a significant fraction of negative cases among advanced SM (aggressive SM (ASM), ASM with an associated clonal non-MC lineage disease (ASM-AHN) and MC leukemia (MCL)), 36%, 46%, and 39%, respectively. In turn, CD25 and FcεRI were found to be expressed in most cases (89% and 92%) in virtually all BMMC (median: 92% and 95%) from both indolent and advanced SM, but with lower/absent levels in a significant fraction of MC leukemia (MCL) and both in MCL and well-differentiated SM (WDSM) patients, respectively. In contrast, CD33 was the only marker expressed on all BMMC from every SM patient. Thus, CD33 emerges as the best potentially targetable cell-surface membrane marker in SM, particularly in advanced SM.
Collapse
|
60
|
Prica A, Crump M. Improving CD20 antibody therapy: obinutuzumab in lymphoproliferative disorders. Leuk Lymphoma 2019; 60:573-582. [PMID: 30668192 DOI: 10.1080/10428194.2018.1498490] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Soon after the anti-CD20 monoclonal antibody rituximab began to change the management of indolent and aggressive B cell lymphomas, development of alternative antibodies - including chemoimmunoconjugates - was undertaken. Among humanized and fully human CD20 antibodies, obinutuzumab has emerged as one antibody that seems to have lived up to the promise of improved efficacy based on in vitro and preclinical experiments. The data available, thus, far establish obinutuzumab's preferred role as the anti-CD20 antibody of choice in chronic lymphocytic leukemia and untreated follicular lymphoma, as well as an important addition to the treatment of rituximab-refractory indolent lymphomas. Additional trials in aggressive lymphoma are required to define the place of this new antibody in the management of patients with curable lymphoma subtypes. There are greater toxicities associated with this treatment, including increased infusion-related reactions and cytopenias, but these are manageable with standard supportive care measures.
Collapse
Affiliation(s)
- Anca Prica
- a Division of Medical Oncology and Hematology , Princess Margaret Cancer Centre, University of Toronto , Toronto , Canada
| | - Michael Crump
- a Division of Medical Oncology and Hematology , Princess Margaret Cancer Centre, University of Toronto , Toronto , Canada
| |
Collapse
|
61
|
Abstract
In recent years, therapeutic monoclonal antibodies have made impressive progress, providing great benefit by successfully treating malignant and chronic inflammatory diseases. Monoclonal antibodies with broadly neutralizing effects against specific antigens, or that target specific immune regulators, manifest therapeutic effects via their Fab fragment specificities. Subsequently therapeutic efficacy is mediated mostly by interactions of the Fc fragments of the antibodies with their receptors (FcR) displayed on cells of the immune system. These interactions can trigger a series of immunoregulatory responses, involving both innate and adaptive immune systems and including cross-presentation of antigens, activation of CD8+ T cells and CD4+ T cells, phagocytosis, complement-mediated antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). The nature of the triggered effector functions of the antibodies is markedly affected by the glycosylation patterns of the Fc fragments. These can cause differences in the conformation of the heavy chains of antibodies, with resultant changes in antibody binding affinity and activation of the complement system. Studies of the Fc glycosylation profiles together with the associated Fc effector functions and FcR/CR interactions promoted interest and progress in engineering therapeutic antibodies. Furthermore, because antigen–antibody immune complexes (ICs) have shown similar actions, in addition to certain novel immunoregulatory mechanisms that also reshape immune responses, the properties of ICs are being explored in new approaches for prevention and therapy of diseases. In this review, both basic studies and experimental/clinical applications of ICs leading to the development of preventive and therapeutic vaccines are presented.
Collapse
|
62
|
Dominguez A, Kastritis E, Castillo JJ. Monoclonal Antibodies for Waldenström Macroglobulinemia. Hematol Oncol Clin North Am 2018; 32:841-852. [PMID: 30190022 DOI: 10.1016/j.hoc.2018.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
For the last 2 decades, anti-CD20 monoclonal antibodies have revolutionized the treatment of patients with B-cell lymphomas. These agents have shown efficacy when used as single agents and also have improved response and survival rates when added to chemotherapy. Monoclonal antibodies are safe and effective as well in patients with Waldenström macroglobulinemia (WM). The purpose of this article is to review the mechanism of action of monoclonal antibodies and to discuss current clinical data supporting their use in patients with WM. This review focuses on retrospective and prospective studies and clinical trials on anti-CD20 antibodies, anti-CD38 antibody, and anti-CXCR4 antibody.
Collapse
Affiliation(s)
- Andres Dominguez
- Department of Internal Medicine, Fundación Valle del Lili, CES University, Cali, Colombia
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University, Athens, Greece
| | - Jorge J Castillo
- Bing Center for Waldenstrom Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Mayer 221, Boston, MA 02215, USA.
| |
Collapse
|
63
|
Dalziel M, Beers SA, Cragg MS, Crispin M. Through the barricades: overcoming the barriers to effective antibody-based cancer therapeutics. Glycobiology 2018; 28:697-712. [PMID: 29800150 DOI: 10.1093/glycob/cwy043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/30/2018] [Indexed: 02/06/2023] Open
Abstract
Since the turn of the century, cancer therapy has undergone a transformation in terms of new treatment modalities and renewed optimism in achieving long-lived tumor control and even cure. This is, in large part, thanks to the widespread incorporation of monoclonal antibodies (mAbs) into standard treatment regimens. These new therapies have, across many settings, significantly contributed to improved clinical responses, patient quality of life and survival. Moreover, the flexibility of the antibody platform has led to the development of a wide range of innovative and combinatorial therapies that continue to augment the clinician's armory. Despite these successes, there is a growing awareness that in many cases mAb therapy remains suboptimal, primarily due to inherent limitations imposed by the immune system's own homeostatic controls and the immunosuppressive tumor microenvironment. Here, we discuss the principal barriers that act to constrain the tumor-killing activity of antibody-based therapeutics, particularly those involving antibody glycans, using illustrative examples from both pre-clinical and market approved mAbs. We also discuss strategies that have been, or are in development to overcome these obstacles. Finally, we outline how the growing understanding of the biological terrain in which mAbs function is shaping innovation and regulation in cancer therapeutics.
Collapse
Affiliation(s)
- Martin Dalziel
- Oxford Glycobiology Institute, Department of Biochemistry, South Parks Road, Oxford, UK
| | - Stephen A Beers
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Max Crispin
- Centre for Biological Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UK
| |
Collapse
|
64
|
Kauder SE, Kuo TC, Harrabi O, Chen A, Sangalang E, Doyle L, Rocha SS, Bollini S, Han B, Sim J, Pons J, Wan HI. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS One 2018; 13:e0201832. [PMID: 30133535 PMCID: PMC6104973 DOI: 10.1371/journal.pone.0201832] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
CD47 is a widely expressed cell surface protein that functions as an immune checkpoint in cancer. When expressed by tumor cells, CD47 can bind SIRPα on myeloid cells, leading to suppression of tumor cell phagocytosis and other innate immune functions. CD47-SIRPα signaling has also been implicated in the suppression of adaptive antitumor responses, but the relevant cellular functions have yet to be elucidated. Therapeutic blockade of the CD47 pathway may stimulate antitumor immunity and improve cancer therapy. To this end, a novel CD47-blocking molecule, ALX148, was generated by fusing a modified SIRPα D1 domain to an inactive human IgG1 Fc. ALX148 binds CD47 from multiple species with high affinity, inhibits wild type SIRPα binding, and enhances phagocytosis of tumor cells by macrophages. ALX148 has no effect on normal human blood cells in vitro or on blood cell parameters in rodent and non-human primate studies. Across several murine tumor xenograft models, ALX148 enhanced the antitumor activity of different targeted antitumor antibodies. Additionally, ALX148 enhanced the antitumor activity of multiple immunotherapeutic antibodies in syngeneic tumor models. These studies revealed that CD47 blockade with ALX148 induces multiple responses that bridge innate and adaptive immunity. ALX148 stimulates antitumor properties of innate immune cells by promoting dendritic cell activation, macrophage phagocytosis, and a shift of tumor-associated macrophages toward an inflammatory phenotype. ALX148 also stimulated the antitumor properties of adaptive immune cells, causing increased T cell effector function, pro-inflammatory cytokine production, and a reduction in the number of suppressive cells within the tumor microenvironment. Taken together, these results show that ALX148 binds and blocks CD47 with high affinity, induces a broad antitumor immune response, and has a favorable safety profile.
Collapse
Affiliation(s)
| | - Tracy C. Kuo
- ALX Oncology, Burlingame, CA, United States of America
| | - Ons Harrabi
- ALX Oncology, Burlingame, CA, United States of America
| | - Amy Chen
- ALX Oncology, Burlingame, CA, United States of America
| | | | - Laura Doyle
- ALX Oncology, Burlingame, CA, United States of America
| | - Sony S. Rocha
- ALX Oncology, Burlingame, CA, United States of America
| | | | - Bora Han
- ALX Oncology, Burlingame, CA, United States of America
| | - Janet Sim
- ALX Oncology, Burlingame, CA, United States of America
| | - Jaume Pons
- ALX Oncology, Burlingame, CA, United States of America
| | - Hong I. Wan
- ALX Oncology, Burlingame, CA, United States of America
| |
Collapse
|
65
|
Olivares N, Rodriguez Y, Zatarain-Barron ZL, Marquina B, Mata-Espinosa D, Barrios-Payán J, Parada C, Moguel B, Espitia-Pinzón C, Estrada I, Hernandez-Pando R. A significant therapeutic effect of immunoglobulins administered alone, or in combination with conventional chemotherapy, in experimental pulmonary tuberculosis caused by drug-sensitive or drug-resistant strains. Pathog Dis 2018; 75:4654846. [PMID: 29186408 DOI: 10.1093/femspd/ftx118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/22/2017] [Indexed: 11/14/2022] Open
Abstract
The recommended chemotherapy for drug-sensitive tuberculosis (TB) consists of four different antibiotics administrated for 6 months. This long treatment leads to significant compliance problems and consequently to recrudescence of the disease and to the development of multidrug-resistant (MDR) strains. Thus, new alternatives are needed to shorten or simplify the treatment of TB. Antibodies have therapeutic effects in animal models of TB, so their use as adjuvants in drug-sensitive and MDR TB is an interesting alternative. To assess the effect of antibodies, BALB/c mice with active late disease 60 days after infection with drug-sensitive TB strain H37Rv were treated with intravenous immunoglobulin (IVIg), alone or in combination with conventional chemotherapy. When compared with control non-treated animals, IVIg alone produced a significantly decreased burden of pulmonary bacilli. This decrease was even greater when IVIg was used in combination with conventional chemotherapy. The combined therapy also significantly reduced tissue damage (pneumonia) when compared to infected animals treated only with antibiotics. IVIg treatment also caused decreased bacillary burdens in mice infected with an MDR strain. In vitro experiments suggested that improving phagocytosis by efficient opsonization is perhaps the principal mechanism of this beneficial therapeutic effect.
Collapse
Affiliation(s)
- Nesty Olivares
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México.,Institute of Biomedical Research, National Autonomous University of Mexico, 04510 México, DF, México
| | - Yadira Rodriguez
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México
| | - Zyanya Lucia Zatarain-Barron
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México
| | - Brenda Marquina
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México
| | - Dulce Mata-Espinosa
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México
| | - Jorge Barrios-Payán
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México
| | - Cristina Parada
- Institute of Biomedical Research, National Autonomous University of Mexico, 04510 México, DF, México
| | - Bárbara Moguel
- Institute of Biomedical Research, National Autonomous University of Mexico, 04510 México, DF, México
| | - Clara Espitia-Pinzón
- Institute of Biomedical Research, National Autonomous University of Mexico, 04510 México, DF, México
| | - Iris Estrada
- National School of Biological Sciences, IPN, Carpio y Plan de Ayala s/n, Colonia Santo Tomas, 11340 Mexico, DF, Mexico
| | - Rogelio Hernandez-Pando
- Experimental Pathology Section. National Institute of Medical Sciences and Nutrition Salvador Zubirán. Avenida Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI, Delegación Tlalpan, CP 14080, Ciudad de México, México
| |
Collapse
|
66
|
Vermi W, Micheletti A, Finotti G, Tecchio C, Calzetti F, Costa S, Bugatti M, Calza S, Agostinelli C, Pileri S, Balzarini P, Tucci A, Rossi G, Furlani L, Todeschini G, Zamò A, Facchetti F, Lorenzi L, Lonardi S, Cassatella MA. slan + Monocytes and Macrophages Mediate CD20-Dependent B-cell Lymphoma Elimination via ADCC and ADCP. Cancer Res 2018; 78:3544-3559. [PMID: 29748373 DOI: 10.1158/0008-5472.can-17-2344] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 03/02/2018] [Accepted: 05/03/2018] [Indexed: 11/16/2022]
Abstract
Terminal tissue differentiation and function of slan+ monocytes in cancer is largely unexplored. Our recent studies demonstrated that slan+ monocytes differentiate into a distinct subset of dendritic cells (DC) in human tonsils and that slan+ cells colonize metastatic carcinoma-draining lymph nodes. Herein, we report by retrospective analysis of multi-institutional cohorts that slan+ cells infiltrate various types of non-Hodgkin lymphomas (NHL), particularly the diffuse large B-cell lymphoma (DLBCL) group, including the most aggressive, nodal and extranodal, forms. Nodal slan+ cells displayed features of either immature DC or macrophages, in the latter case ingesting tumor cells and apoptotic bodies. We also found in patients with DLBCL that peripheral blood slan+ monocytes, but not CD14+ monocytes, increased in number and displayed highly efficient rituximab-mediated antibody-dependent cellular cytotoxicity, almost equivalent to that exerted by NK cells. Notably, slan+ monocytes cultured in conditioned medium from nodal DLBCL (DCM) acquired a macrophage-like phenotype, retained CD16 expression, and became very efficient in rituximab-mediated antibody-dependent cellular phagocytosis (ADCP). Macrophages derived from DCM-treated CD14+ monocytes performed very efficient rituximab-mediated ADCP, however, using different FcγRs from those used by slan+ macrophages. Our observations shed new light on the complexity of the immune microenvironment of DLBCL and demonstrate plasticity of slan+ monocytes homing to cancer tissues. Altogether, data identify slan+ monocytes and macrophages as prominent effectors of antibody-mediated tumor cell targeting in patients with DLBCL.Significance: slan+ monocytes differentiate into macrophages that function as prominent effectors of antibody-mediated tumor cell targeting in lymphoma.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3544/F1.large.jpg Cancer Res; 78(13); 3544-59. ©2018 AACR.
Collapse
Affiliation(s)
- William Vermi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy. .,Department of Pathology and Immunology, Washington University, Saint Louis, Missouri
| | - Alessandra Micheletti
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Giulia Finotti
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Cristina Tecchio
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Federica Calzetti
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Sara Costa
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Mattia Bugatti
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefano Calza
- Unit of Biostatistics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudio Agostinelli
- Haematopathology Unit, Department of Experimental Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Stefano Pileri
- Unit of Haematopathology, European Institute of Oncology, 20141 Milan, Italy
| | - Piera Balzarini
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandra Tucci
- Division of Haematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Giuseppe Rossi
- Division of Haematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Lara Furlani
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giuseppe Todeschini
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Alberto Zamò
- Section of Pathology, Department of Public Health and Diagnostics, University of Verona, Verona, Italy
| | - Fabio Facchetti
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Lorenzi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvia Lonardi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco A Cassatella
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
67
|
Buatois V, Johnson Z, Salgado-Pires S, Papaioannou A, Hatterer E, Chauchet X, Richard F, Barba L, Daubeuf B, Cons L, Broyer L, D'Asaro M, Matthes T, LeGallou S, Fest T, Tarte K, Clarke Hinojosa RK, Genescà Ferrer E, Ribera JM, Dey A, Bailey K, Fielding AK, Eissenberg L, Ritchey J, Rettig M, DiPersio JF, Kosco-Vilbois MH, Masternak K, Fischer N, Shang L, Ferlin WG. Preclinical Development of a Bispecific Antibody that Safely and Effectively Targets CD19 and CD47 for the Treatment of B-Cell Lymphoma and Leukemia. Mol Cancer Ther 2018; 17:1739-1751. [PMID: 29743205 DOI: 10.1158/1535-7163.mct-17-1095] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/15/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022]
Abstract
CD47, an ubiquitously expressed innate immune checkpoint receptor that serves as a universal "don't eat me" signal of phagocytosis, is often upregulated by hematologic and solid cancers to evade immune surveillance. Development of CD47-targeted modalities is hindered by the ubiquitous expression of the target, often leading to rapid drug elimination and hemotoxicity including anemia. To overcome such liabilities, we have developed a fully human bispecific antibody, NI-1701, designed to coengage CD47 and CD19 selectively on B cells. NI-1701 demonstrates favorable elimination kinetics with no deleterious effects seen on hematologic parameters following single or multiple administrations to nonhuman primates. Potent in vitro and in vivo activity is induced by NI-1701 to kill cancer cells across a plethora of B-cell malignancies and control tumor growth in xenograft mouse models. The mechanism affording maximal tumor growth inhibition by NI-1701 is dependent on the coengagement of CD47/CD19 on B cells inducing potent antibody-dependent cellular phagocytosis of the targeted cells. NI-1701-induced control of tumor growth in immunodeficient NOD/SCID mice was more effective than that achieved with the anti-CD20 targeted antibody, rituximab. Interestingly, a synergistic effect was seen when tumor-implanted mice were coadministered NI-1701 and rituximab leading to significantly improved tumor growth inhibition and regression in some animals. We describe herein, a novel bispecific antibody approach aimed at sensitizing B cells to become more readily phagocytosed and eliminated thus offering an alternative or adjunct therapeutic option to patients with B-cell malignancies refractory/resistant to anti-CD20-targeted therapy. Mol Cancer Ther; 17(8); 1739-51. ©2018 AACR.
Collapse
Affiliation(s)
| | - Zoë Johnson
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | | | | | | | | | | | - Laura Cons
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | - Thomas Matthes
- Medical Faculty, University of Geneva, Genève, Switzerland
| | | | - Thierry Fest
- Rennes 1 University, Inserm U1236, Rennes, France
| | - Karin Tarte
- Rennes 1 University, Inserm U1236, Rennes, France
| | - Robert K Clarke Hinojosa
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - Eulàlia Genescà Ferrer
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - José María Ribera
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - Aditi Dey
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Katharine Bailey
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Adele K Fielding
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Linda Eissenberg
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Julie Ritchey
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | - Limin Shang
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | |
Collapse
|
68
|
Bacac M, Colombetti S, Herter S, Sam J, Perro M, Chen S, Bianchi R, Richard M, Schoenle A, Nicolini V, Diggelmann S, Limani F, Schlenker R, Hüsser T, Richter W, Bray-French K, Hinton H, Giusti AM, Freimoser-Grundschober A, Lariviere L, Neumann C, Klein C, Umaña P. CD20-TCB with Obinutuzumab Pretreatment as Next-Generation Treatment of Hematologic Malignancies. Clin Cancer Res 2018; 24:4785-4797. [PMID: 29716920 DOI: 10.1158/1078-0432.ccr-18-0455] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/24/2018] [Accepted: 04/25/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Despite promising clinical activity, T-cell-engaging therapies including T-cell bispecific antibodies (TCB) are associated with severe side effects requiring the use of step-up-dosing (SUD) regimens to mitigate safety. Here, we present a next-generation CD20-targeting TCB (CD20-TCB) with significantly higher potency and a novel approach enabling safer administration of such potent drug.Experimental Design: We developed CD20-TCB based on the 2:1 TCB molecular format and characterized its activity preclinically. We also applied a single administration of obinutuzumab (Gazyva pretreatment, Gpt; Genentech/Roche) prior to the first infusion of CD20-TCB as a way to safely administer such a potent drug.Results: CD20-TCB is associated with a long half-life and high potency enabled by high-avidity bivalent binding to CD20 and head-to-tail orientation of B- and T-cell-binding domains in a 2:1 molecular format. CD20-TCB displays considerably higher potency than other CD20-TCB antibodies in clinical development and is efficacious on tumor cells expressing low levels of CD20. CD20-TCB also displays potent activity in primary tumor samples with low effector:target ratios. In vivo, CD20-TCB regresses established tumors of aggressive lymphoma models. Gpt enables profound B-cell depletion in peripheral blood and secondary lymphoid organs and reduces T-cell activation and cytokine release in the peripheral blood, thus increasing the safety of CD20-TCB administration. Gpt is more efficacious and safer than SUD.Conclusions: CD20-TCB and Gpt represent a potent and safer approach for treatment of lymphoma patients and are currently being evaluated in phase I, multicenter study in patients with relapsed/refractory non-Hodgkin lymphoma (NCT03075696). Clin Cancer Res; 24(19); 4785-97. ©2018 AACR See related commentary by Prakash and Diefenbach, p. 4631.
Collapse
Affiliation(s)
- Marina Bacac
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland.
| | - Sara Colombetti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Sylvia Herter
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Johannes Sam
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Mario Perro
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Stanford Chen
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Roberta Bianchi
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Marine Richard
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Anne Schoenle
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Valeria Nicolini
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Sarah Diggelmann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Florian Limani
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Ramona Schlenker
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Tamara Hüsser
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Wolfgang Richter
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, pRED, Basel, Switzerland
| | - Katharine Bray-French
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, pRED, Basel, Switzerland
| | - Heather Hinton
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Anna Maria Giusti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Anne Freimoser-Grundschober
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Laurent Lariviere
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, pRED, Munich, Germany
| | - Christiane Neumann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, pRED, Zurich, Switzerland.
| |
Collapse
|
69
|
Hassenrück F, Knödgen E, Göckeritz E, Midda SH, Vondey V, Neumann L, Herter S, Klein C, Hallek M, Krause G. Sensitive Detection of the Natural Killer Cell-Mediated Cytotoxicity of Anti-CD20 Antibodies and Its Impairment by B-Cell Receptor Pathway Inhibitors. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1023490. [PMID: 29750146 PMCID: PMC5884282 DOI: 10.1155/2018/1023490] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 11/17/2022]
Abstract
The antibody-dependent cell-mediated cytotoxicity (ADCC) of the anti-CD20 monoclonal antibodies (mAbs) rituximab and obinutuzumab against the cell line Raji and isolated CLL cells and its potential impairment by kinase inhibitors (KI) was determined via lactate dehydrogenase release or calcein retention, respectively, using genetically modified NK92 cells expressing CD16-176V as effector cells. Compared to peripheral blood mononuclear cells, recombinant effector cell lines showed substantial alloreactivity-related cytotoxicity without addition of mAbs but afforded determination of ADCC with reduced interassay variability. The cytotoxicity owing to alloreactivity was less susceptible to interference by KI than the ADCC of anti-CD20 mAbs, which was markedly diminished by ibrutinib, but not by idelalisib. Compared to rituximab, the ADCC of obinutuzumab against primary CLL cells showed approximately 30% higher efficacy and less interference with KI. Irreversible BTK inhibitors at a clinically relevant concentration of 1 μM only weakly impaired the ADCC of anti-CD20 mAbs, with less influence in combinations with obinutuzumab than with rituximab and by acalabrutinib than by ibrutinib or tirabrutinib. In summary, NK cell line-based assays permitted the sensitive detection of ADCC of therapeutic anti-CD20 mAbs against CLL cells and of the interference of KI with this important killing mechanism.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antigens, CD20/metabolism
- B-Lymphocytes/drug effects
- Benzamides/pharmacology
- Cell Line, Tumor
- Cytotoxins/pharmacology
- Humans
- Killer Cells, Natural/drug effects
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Piperidines
- Protein Kinase Inhibitors/pharmacology
- Purines/pharmacology
- Pyrazines/pharmacology
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Quinazolinones/pharmacology
- Receptors, Antigen, B-Cell/metabolism
- Rituximab/pharmacology
Collapse
Affiliation(s)
- Floyd Hassenrück
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Eva Knödgen
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Elisa Göckeritz
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Safi Hasan Midda
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Verena Vondey
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Lars Neumann
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Sylvia Herter
- Roche Innovation Center Zurich, Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Günter Krause
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| |
Collapse
|
70
|
Elias S, Kahlon S, Kotzur R, Kaynan N, Mandelboim O. Obinutuzumab activates FcγRI more potently than other anti-CD20 antibodies in chronic lymphocytic leukemia (CLL). Oncoimmunology 2018; 7:e1428158. [PMID: 29872553 PMCID: PMC5980409 DOI: 10.1080/2162402x.2018.1428158] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/07/2018] [Accepted: 01/09/2018] [Indexed: 11/03/2022] Open
Abstract
Treatment with monoclonal antibodies has revolutionized clinical medicine, especially in the fields of cancer and immunology. One of the oldest antibodies, which is widely used for the treatment of lymphomas and autoimmune diseases, is the anti-CD20 antibody rituximab. In recent years, new antibodies against CD20 have been developed including ofatumumab and obinutuzumab. An important mechanism of action of therapeutic monoclonal antibodies is activation of immune cells via Fc receptors (FcγRs). However, surprisingly, little is known about triggering of FcγRs by different therapeutic antibodies in general and anti-CD20 antibodies in particular. Here we establish a reporter assay to assess whether a particular antibody activates a certain Fc receptor. Using this assay we corroborated previous reports demonstrating obinutuzumab's ability to highly activate FcγRIIIa (CD16a). Importantly, we discovered that obinutuzumab also activates FcγRI (CD64) significantly more than rituximab and ofatumumab in response to chronic lymphocytic leukemia (CLL) cells obtained from patients. Mechanistically we show that this is due to the lack of FcγRIIb-mediated internalization of obinutuzumab following binding to CD20. Moreover, we show that obinutuzumab induces increased phagocytosis by primary macrophages in an FcγRI-dependent manner. Beyond the discovery of a new mechanism of obinutuzumab activity, the reporter assay can be applied to other therapeutic antibodies and may assist in developing antibodies with improved immunological properties.
Collapse
Affiliation(s)
- Shlomo Elias
- Department of Hematology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.,The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shira Kahlon
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Rebecca Kotzur
- Department of Transfusion Medicine, Institute of Immunogenetics, Hannover Medical School, Hannover, Germany
| | - Noah Kaynan
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
71
|
Boesch AW, Kappel JH, Mahan AE, Chu TH, Crowley AR, Osei-Owusu NY, Alter G, Ackerman ME. Enrichment of high affinity subclasses and glycoforms from serum-derived IgG using FcγRs as affinity ligands. Biotechnol Bioeng 2018; 115:1265-1278. [PMID: 29315477 DOI: 10.1002/bit.26545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/18/2017] [Accepted: 01/03/2018] [Indexed: 12/26/2022]
Abstract
As antibodies continue to gain predominance in drug discovery and development pipelines, efforts to control and optimize their activity in vivo have matured to incorporate sophisticated abilities to manipulate engagement of specific Fc binding partners. Such efforts to promote diverse functional outcomes include modulating IgG-Fc affinity for FcγRs to alternatively potentiate or reduce effector functions, such as antibody-dependent cellular cytotoxicity and phagocytosis. While a number of natural and engineered Fc features capable of eliciting variable effector functions have been demonstrated in vitro and in vivo, elucidation of these important functional relationships has taken significant effort through use of diverse genetic, cellular and enzymatic techniques. As an orthogonal approach, we demonstrate use of FcγR as chromatographic affinity ligands to enrich and therefore simultaneously identify favored binding species from a complex mixture of serum-derived pooled polycloncal human IgG, a load material that contains the natural repertoire of Fc variants and post-translational modifications. The FcγR-enriched IgG was characterized for subclass and glycoform composition and the impact of this bioseparation step on antibody activity was measured in cell-based effector function assays including Natural Killer cell activation and monocyte phagocytosis. This work demonstrates a tractable means to rapidly distinguish complex functional relationships between two or more interacting biological agents by leveraging affinity chromatography followed by secondary analysis with high-resolution biophysical and functional assays and emphasizes a platform capable of surveying diverse natural post-translational modifications that may not be easily produced with high purity or easily accessible with recombinant expression techniques.
Collapse
Affiliation(s)
- Austin W Boesch
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
- Zepteon, Inc., Boston, Massachusetts
| | - James H Kappel
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Alison E Mahan
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, Massachusetts
| | - Thach H Chu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Nana Y Osei-Owusu
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, Massachusetts
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| |
Collapse
|
72
|
Abstract
The persistence of West Nile virus (WNV) infections throughout the USA since its inception in 1999 and its continuous spread throughout the globe calls for an urgent need of effective treatments and prevention measures. Although the licensing of several WNV vaccines for veterinary use provides a proof of concept, similar efforts on the development of an effective vaccine for humans remain still unsuccessful. Increased understanding of biology and pathogenesis of WNV together with recent technological advancements have raised hope that an effective WNV vaccine may be available in the near future. In addition, rapid progress in the structural and functional characterization of WNV and other flaviviral proteins have provided a solid base for the design and development of several classes of inhibitors as potential WNV therapeutics. Moreover, the therapeutic monoclonal antibodies demonstrate an excellent efficacy against WNV in animal models and represent a promising class of WNV therapeutics. However, there are some challenges as to the design and development of a safe and efficient WNV vaccine or therapeutic. In this chapter, we discuss the current approaches, progress, and challenges toward the development of WNV vaccines, therapeutic antibodies, and antiviral drugs.
Collapse
|
73
|
Bougherara H, Némati F, Nicolas A, Massonnet G, Pugnière M, Ngô C, Le Frère-Belda MA, Leary A, Alexandre J, Meseure D, Barret JM, Navarro-Teulon I, Pèlegrin A, Roman-Roman S, Prost JF, Donnadieu E, Decaudin D. The humanized anti-human AMHRII mAb 3C23K exerts an anti-tumor activity against human ovarian cancer through tumor-associated macrophages. Oncotarget 2017; 8:99950-99965. [PMID: 29245952 PMCID: PMC5725143 DOI: 10.18632/oncotarget.21556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 05/31/2017] [Indexed: 02/05/2023] Open
Abstract
Müllerian inhibiting substance, also called anti-Müllerian hormone (AMH), inhibits proliferation and induces apoptosis of AMH type II receptor-positive tumor cells, such as human ovarian cancers (OCs). On this basis, a humanized glyco-engineered monoclonal antibody (3C23K) has been developed. The aim of this study was therefore to experimentally confirm the therapeutic potential of 3C23K in human OCs. We first determined by immunofluorescence, immunohistochemistry and cytofluorometry analyses the expression of AMHRII in patient’s tumors and found that a majority (60 to 80% depending on the detection technique) of OCs were positive for this marker. We then provided evidence that the tumor stroma of OC is enriched in tumor-associated macrophages and that these cells are responsible for 3C23K-induced killing of tumor cells through ADCP and ADCC mechanisms. In addition, we showed that 3C23K reduced macrophages induced-T cells immunosuppression. Finally, we evaluated the therapeutic efficacy of 3C23K alone and in combination with a carboplatin-paclitaxel chemotherapy in a panel of OC Patient-Derived Xenografts. In those experiments, we showed that 3C23K significantly increased the proportion and the quality of chemotherapy-based in vivo responses. Altogether, our data support the potential interest of AMHRII targeting in human ovarian cancers and the evaluation of 3C23K in further clinical trials.
Collapse
Affiliation(s)
- Houcine Bougherara
- Inserm, U1016, Institut Cochin, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fariba Némati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, Paris, France
| | - André Nicolas
- Department of Tumor Biology, Institut Curie, Paris, France
| | - Gérald Massonnet
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Martine Pugnière
- INSERM U896, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Charlotte Ngô
- Department of Gynaecological and Oncological Surgery, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marie-Aude Le Frère-Belda
- Department of Pathology, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Jérôme Alexandre
- Inserm, U1016, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Medical Oncology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Didier Meseure
- Department of Tumor Biology, Institut Curie, Paris, France
| | | | | | - André Pèlegrin
- INSERM U896, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Sergio Roman-Roman
- Department of Translational Research, Institut Curie, PSL University, Paris, France
| | | | - Emmanuel Donnadieu
- Inserm, U1016, Institut Cochin, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL University, Paris, France.,Department of Medical Oncology, Institut Curie, Paris, France
| |
Collapse
|
74
|
Reddy V, Klein C, Isenberg DA, Glennie MJ, Cambridge G, Cragg MS, Leandro MJ. Obinutuzumab induces superior B-cell cytotoxicity to rituximab in rheumatoid arthritis and systemic lupus erythematosus patient samples. Rheumatology (Oxford) 2017; 56:1227-1237. [PMID: 28407142 PMCID: PMC5808665 DOI: 10.1093/rheumatology/kex067] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Indexed: 01/21/2023] Open
Abstract
Objective A proportion of RA and SLE patients treated with standard doses of rituximab (RTX) display inefficient B cell deletion and poor clinical responses that can be augmented by delivering higher doses, indicating that standard-dose RTX is a sub-optimal therapy in these patients. This study aimed to investigate whether better responses could be achieved with mechanistically different anti-CD20 mAbs. Methods We compared RTX with obinutuzumab (OBZ), a new-generation, glycoengineered type II anti-CD20 mAb, in a series of in vitro assays measuring B cell cytotoxicity in RA and SLE patient samples. Results We found that OBZ was at least 2-fold more efficient than RTX at inducing B-cell cytotoxicity in in vitro whole blood assays. Dissecting this difference, we found that RTX elicited more potent complement-dependent cellular cytotoxicity than OBZ. In contrast, OBZ was more effective at evoking Fc gamma receptor-mediated effector mechanisms, including activation of NK cells and neutrophils, probably due to stronger interaction with Fc gamma receptors and the ability of OBZ to remain at the cell surface following CD20 engagement, whereas RTX became internalized. OBZ was also more efficient at inducing direct cell death. This was true for all CD19 + B cells as a whole and in naïve (IgD + CD27 - ) and switched (IgD - CD27 + ) memory B cells specifically, a higher frequency of which is associated with poor clinical response after RTX. Conclusion Taken together, these data provide a mechanistic basis for resistance to rituximab-induced B-cell depletion, and for considering obinutuzumab as an alternative B-cell depleting agent in RA and SLE.
Collapse
Affiliation(s)
- Venkat Reddy
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| | - Christian Klein
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| | - David A Isenberg
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| | - Martin J Glennie
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| | - Geraldine Cambridge
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| | - Mark S Cragg
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| | - Maria J Leandro
- Centre for Rheumatology, University College London, London, UK2Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland3Academic Unit of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
75
|
Li D, He W, Liu X, Zheng S, Qi Y, Li H, Mao F, Liu J, Sun Y, Pan L, Du K, Ye K, Li W, Sui J. A potent human neutralizing antibody Fc-dependently reduces established HBV infections. eLife 2017; 6. [PMID: 28949917 PMCID: PMC5614562 DOI: 10.7554/elife.26738] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major global health problem. Currently-available therapies are ineffective in curing chronic HBV infection. HBV and its satellite hepatitis D virus (HDV) infect hepatocytes via binding of the preS1 domain of its large envelope protein to sodium taurocholate cotransporting polypeptide (NTCP). Here, we developed novel human monoclonal antibodies that block the engagement of preS1 with NTCP and neutralize HBV and HDV with high potency. One antibody, 2H5-A14, functions at picomolar level and exhibited neutralization-activity-mediated prophylactic effects. It also acts therapeutically by eliciting antibody-Fc-dependent immunological effector functions that impose durable suppression of viral infection in HBV-infected mice, resulting in reductions in the levels of the small envelope antigen and viral DNA, with no emergence of escape mutants. Our results illustrate a novel antibody-Fc-dependent approach for HBV treatment and suggest 2H5-A14 as a novel clinical candidate for HBV prevention and treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Dan Li
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Wenhui He
- National Institute of Biological Sciences, Beijing, China
| | - Ximing Liu
- National Institute of Biological Sciences, Beijing, China.,PTN Joint Graduate Program, College of Life Sciences, Peking University, Beijing, China
| | - Sanduo Zheng
- National Institute of Biological Sciences, Beijing, China
| | - Yonghe Qi
- National Institute of Biological Sciences, Beijing, China
| | - Huiyu Li
- National Institute of Biological Sciences, Beijing, China
| | - Fengfeng Mao
- National Institute of Biological Sciences, Beijing, China.,Graduate Program in College of Life Sciences, Beijing Normal University, Beijing, China
| | - Juan Liu
- National Institute of Biological Sciences, Beijing, China
| | - Yinyan Sun
- National Institute of Biological Sciences, Beijing, China
| | - Lijing Pan
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Kaixin Du
- National Institute of Biological Sciences, Beijing, China.,Graduate Program in College of Life Sciences, Beijing Normal University, Beijing, China
| | - Keqiong Ye
- National Institute of Biological Sciences, Beijing, China
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, China
| | - Jianhua Sui
- National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
76
|
Klein C, Bacac M, Umana P, Fingerle-Rowson G. Combination therapy with the type II anti-CD20 antibody obinutuzumab. Expert Opin Investig Drugs 2017; 26:1145-1162. [DOI: 10.1080/13543784.2017.1373087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Christian Klein
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Pablo Umana
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | | |
Collapse
|
77
|
Skopelja-Gardner S, Jones JD, Hamilton BJ, Danilov AV, Rigby WFC. Role for ZAP-70 Signaling in the Differential Effector Functions of Rituximab and Obinutuzumab (GA101) in Chronic Lymphocytic Leukemia B Cells. THE JOURNAL OF IMMUNOLOGY 2017; 199:1275-1282. [DOI: 10.4049/jimmunol.1602105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/16/2017] [Indexed: 11/19/2022]
|
78
|
Hart F, Danielczyk A, Goletz S. Human Cell Line-Derived Monoclonal IgA Antibodies for Cancer Immunotherapy. Bioengineering (Basel) 2017; 4:bioengineering4020042. [PMID: 28952521 PMCID: PMC5590476 DOI: 10.3390/bioengineering4020042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/30/2017] [Accepted: 05/02/2017] [Indexed: 12/17/2022] Open
Abstract
IgA antibodies have great potential to improve the functional diversity of current IgG antibody-based cancer immunotherapy options. However, IgA production and purification is not well established, which can at least in part be attributed to the more complex glycosylation as compared to IgG antibodies. IgA antibodies possess up to five N-glycosylation sites within their constant region of the heavy chain as compared to one site for IgG antibodies. The human GlycoExpress expression system was developed to produce biotherapeutics with optimized glycosylation and used here to generate a panel of IgA isotype antibodies directed against targets for solid (TA-mucin 1, Her2, EGFR, Thomsen–Friedenreich) and hematological (CD20) cancer indications. The feasibility of good manufacturing practice was shown by the production of 11 g IgA within 35 days in a one liter perfusion bioreactor, and IgA antibodies in high purity were obtained after purification. The monoclonal IgA antibodies possessed a high sialylation degree, and no non-human glycan structures were detected. Kinetic analysis revealed increased avidity antigen binding for IgA dimers as compared to monomeric antibodies. The IgA antibodies exhibited potent Fab- and Fc-mediated functionalities against cancer cell lines, whereby especially granulocytes are recruited. Therefore, for patients who do not sufficiently benefit from therapeutic IgG antibodies, IgA antibodies may complement current regiment options and represent a promising strategy for cancer immunotherapy. In conclusion, a panel of novel biofunctional IgA antibodies with human glycosylation was successfully generated.
Collapse
Affiliation(s)
- Felix Hart
- Glycotope GmbH, Robert-Roessle-Street 10, 13125 Berlin, Germany.
| | - Antje Danielczyk
- Glycotope GmbH, Robert-Roessle-Street 10, 13125 Berlin, Germany.
| | - Steffen Goletz
- Glycotope GmbH, Robert-Roessle-Street 10, 13125 Berlin, Germany.
| |
Collapse
|
79
|
Yu X, Marshall MJE, Cragg MS, Crispin M. Improving Antibody-Based Cancer Therapeutics Through Glycan Engineering. BioDrugs 2017; 31:151-166. [DOI: 10.1007/s40259-017-0223-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
80
|
Largy E, Cantais F, Van Vyncht G, Beck A, Delobel A. Orthogonal liquid chromatography-mass spectrometry methods for the comprehensive characterization of therapeutic glycoproteins, from released glycans to intact protein level. J Chromatogr A 2017; 1498:128-146. [PMID: 28372839 DOI: 10.1016/j.chroma.2017.02.072] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/04/2017] [Accepted: 02/28/2017] [Indexed: 01/16/2023]
Abstract
Proteins are increasingly used as therapeutics. Their characterization is challenging due to their size and inherent heterogeneity notably caused by post-translational modifications, among which glycosylation is probably the most prominent. The glycosylation profile of therapeutic proteins must therefore be thoroughly analyzed. Here, we illustrate how the use of a combination of various cutting-edge LC or LC/MS(/MS) methods, and operating at different levels of analysis allows the comprehensive characterization of both the N- and O-glycosylations of therapeutic proteins without the need for other approaches (capillary electrophoresis, MALDI-TOF). This workflow does not call for the use of highly specialized/custom hardware and software nor an extensive knowledge of glycan analysis. Most notably, we present the point of view of a contract research organization, with the constraints associated to the work in a regulated environment (GxP). Two salient points of this work are i) the use of mixed-mode chromatography as a fast and straightforward mean of profiling N-glycans sialylation as well as an orthogonal method to separate N-glycans co-eluting in the HILIC mode; and ii) the use of widepore HILIC/MS to analyze challenging N/O-glycosylation profiles at both the peptide and subunit levels. A particular attention was given to the sample preparations in terms of duration, specificity, versatility, and robustness, as well as the ease of data processing.
Collapse
Affiliation(s)
- Eric Largy
- Quality Assistance sa, Technoparc de Thudinie 2, 6536, Donstiennes, Belgium
| | - Fabrice Cantais
- Quality Assistance sa, Technoparc de Thudinie 2, 6536, Donstiennes, Belgium
| | - Géry Van Vyncht
- Quality Assistance sa, Technoparc de Thudinie 2, 6536, Donstiennes, Belgium
| | - Alain Beck
- Centre d'Immunologie Pierre Fabre (CIPF), 5 Av. Napoléon III, BP 60497, 74164, Saint-Julien-en-Genevois, France
| | - Arnaud Delobel
- Quality Assistance sa, Technoparc de Thudinie 2, 6536, Donstiennes, Belgium.
| |
Collapse
|
81
|
Chen YCE, Mapp S, Blumenthal A, Burgess ML, Mazzieri R, Mattarollo SR, Mollee P, Gill D, Saunders NA. The duality of macrophage function in chronic lymphocytic leukaemia. Biochim Biophys Acta Rev Cancer 2017; 1868:176-182. [PMID: 28347751 DOI: 10.1016/j.bbcan.2017.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/09/2017] [Accepted: 03/21/2017] [Indexed: 12/23/2022]
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common adult leukaemia and, in some patients, is accompanied by resistance to both chemotherapeutics and immunotherapeutics. In this review we will discuss the role of tumour associated macrophages (TAMs) in promoting CLL cell survival and resistance to immunotherapeutics. In addition, we will discuss mechanisms by which TAMs suppress T-cell mediated antitumour responses. Thus, targeting macrophages could be used to i) reduce the leukaemic burden via the induction of T-cell-mediated antitumour responses, ii) to reduce pro-survival signalling and enhance response to conventional chemotherapeutics or iii) enhance the response to therapeutic antibodies in current clinical use.
Collapse
Affiliation(s)
- Y C E Chen
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - S Mapp
- Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - A Blumenthal
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - M L Burgess
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - R Mazzieri
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - S R Mattarollo
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - P Mollee
- Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - D Gill
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - N A Saunders
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia.
| |
Collapse
|
82
|
Human neutrophils mediate trogocytosis rather than phagocytosis of CLL B cells opsonized with anti-CD20 antibodies. Blood 2017; 129:2636-2644. [PMID: 28288980 DOI: 10.1182/blood-2016-08-735605] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/03/2017] [Indexed: 02/05/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) have previously been reported to mediate phagocytosis of anti-CD20-opsonized B cells from patients with chronic lymphocytic leukemia (CLL). However, recent data have suggested that PMNs, like macrophages, can also mediate trogocytosis. We have performed experiments to more precisely investigate this point and to discriminate between trogocytosis and phagocytosis. In live-cell time-lapse microscopy experiments, we could not detect any significant phagocytosis by purified PMNs of anti-CD20-opsonized CLL B cells, but could detect only the repeated close contact between effectors and targets, which suggested trogocytosis. Similarly, in flow cytometry assays using CLL B-cell targets labeled with the membrane dye PKH67 and opsonized with rituximab or obinutuzumab, we observed that a mean of 50% and 75% of PMNs had taken a fraction of the dye from CLL B cells at 3 and 20 hours, respectively, with no significant decrease in absolute live or total CLL B-cell numbers, confirming that trogocytosis occurs, rather than phagocytosis. Trogocytosis was accompanied by loss of membrane CD20 from CLL B cells, which was evident with rituximab but not obinutuzumab. We conclude that PMNs mediate mostly trogocytosis rather than phagocytosis of anti-CD20-opsonized CLL B cells, and we discuss the implications of this finding in patients with CLL treated with rituximab or obinutuzumab in vivo.
Collapse
|
83
|
Hierarchical and Redundant Roles of Activating FcγRs in Protection against Influenza Disease by M2e-Specific IgG1 and IgG2a Antibodies. J Virol 2017; 91:JVI.02500-16. [PMID: 28077656 DOI: 10.1128/jvi.02500-16] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 02/01/2023] Open
Abstract
The ectodomain of matrix protein 2 is a universal influenza A virus vaccine candidate that provides protection through antibody-dependent effector mechanisms. Here we compared the functional engagement of Fcγ receptor (FcγR) family members by two M2e-specific monoclonal antibodies (MAbs), MAb 37 (IgG1) and MAb 65 (IgG2a), which recognize a similar epitope in M2e with similar affinities. The binding of MAb 65 to influenza A virus-infected cells triggered all three activating mouse Fcγ receptors in vitro, whereas MAb 37 activated only FcγRIII. The passive transfer of MAb 37 or MAb 65 in wild-type, Fcer1g-/-, Fcgr3-/-, and Fcgr1-/-Fcgr3-/- BALB/c mice revealed the importance of these receptors for protection against influenza A virus challenge, with a clear requirement of FcγRIII for IgG1 MAb 37 being found. We also report that FcγRIV contributes to protection by M2e-specific IgG2a antibodies.IMPORTANCE There is increased awareness that protection by antibodies directed against viral antigens is also mediated by the Fc domain of these antibodies. These Fc-mediated effector functions are often missed in clinical assays, which are used, for example, to define correlates of protection induced by vaccines. The use of antibodies to prevent and treat infectious diseases is on the rise and has proven to be a promising approach in our battle against newly emerging viral infections. It is now also realized that Fcγ receptors significantly enhance the in vivo protective effect of broadly neutralizing antibodies directed against the conserved parts of the influenza virus hemagglutinin. We show here that two M2e-specific monoclonal antibodies with close to identical antigen-binding specificities and affinities have a very different in vivo protective potential that is controlled by their capacity to interact with activating Fcγ receptors.
Collapse
|
84
|
Quast I, Peschke B, Lünemann JD. Regulation of antibody effector functions through IgG Fc N-glycosylation. Cell Mol Life Sci 2017; 74:837-847. [PMID: 27639381 PMCID: PMC11107549 DOI: 10.1007/s00018-016-2366-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/13/2016] [Indexed: 02/03/2023]
Abstract
Immunoglobulin gamma (IgG) antibodies are key effector proteins of the immune system. They recognize antigens with high specificity and are indispensable for immunological memory following pathogen exposure or vaccination. The constant, crystallizable fragment (Fc) of IgG molecules mediates antibody effector functions such as complement-dependent cytotoxicity, antibody-mediated cellular cytotoxicity, and antibody-dependent cell-mediated phagocytosis. These functions are regulated by a single N-linked, biantennary glycan of the heavy chain, which resides just below the hinge region, and the presence of specific sugar moieties on the glycan has profound implications on IgG effector functions. Emerging knowledge of how Fc glycans contribute to IgG structure and functions has opened new avenues for the therapeutic exploitation of defined antibody glycoforms in the treatment of cancer and autoimmune diseases. Here, we review recent advances in understanding proinflammatory IgG effector functions and their regulation by Fc glycans.
Collapse
Affiliation(s)
- Isaak Quast
- Laboratory of Neuroinflammation, Department of Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Benjamin Peschke
- Laboratory of Neuroinflammation, Department of Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Jan D Lünemann
- Laboratory of Neuroinflammation, Department of Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
85
|
Capuano C, Pighi C, Molfetta R, Paolini R, Battella S, Palmieri G, Giannini G, Belardinilli F, Santoni A, Galandrini R. Obinutuzumab-mediated high-affinity ligation of FcγRIIIA/CD16 primes NK cells for IFNγ production. Oncoimmunology 2017; 6:e1290037. [PMID: 28405525 PMCID: PMC5384385 DOI: 10.1080/2162402x.2017.1290037] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Accepted: 01/26/2017] [Indexed: 01/30/2023] Open
Abstract
Natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC), based on the recognition of IgG-opsonized targets by the low-affinity receptor for IgG FcγRIIIA/CD16, represents one of the main mechanisms by which therapeutic antibodies (mAbs) mediate their antitumor effects. Besides ADCC, CD16 ligation also results in cytokine production, in particular, NK-derived IFNγ is endowed with a well-recognized role in the shaping of adaptive immune responses. Obinutuzumab is a glycoengineered anti-CD20 mAb with a modified crystallizable fragment (Fc) domain designed to increase the affinity for CD16 and consequently the killing of mAb-opsonized targets. However, the impact of CD16 ligation in optimized affinity conditions on NK functional program is not completely understood. Herein, we demonstrate that the interaction of NK cells with obinutuzumab-opsonized cells results in enhanced IFNγ production as compared with parental non-glycoengineered mAb or the reference molecule rituximab. We observed that affinity ligation conditions strictly correlate with the ability to induce CD16 down-modulation and lysosomal targeting of receptor-associated signaling elements. Indeed, a preferential degradation of FcεRIγ chain and Syk kinase was observed upon obinutuzumab stimulation independently from CD16-V158F polymorphism. Although the downregulation of FcεRIγ/Syk module leads to the impairment of cytotoxic function induced by NKp46 and NKp30 receptors, obinutuzumab-experienced cells exhibit an increased ability to produce IFNγ in response to different stimuli. These data highlight a relationship between CD16 aggregation conditions and the ability to promote a degradative pathway of CD16-coupled signaling elements associated to the shift of NK functional program.
Collapse
Affiliation(s)
- Cristina Capuano
- Department of Experimental Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Chiara Pighi
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Simone Battella
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Gabriella Palmieri
- Department of Experimental Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Francesca Belardinilli
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Ricciarda Galandrini
- Department of Experimental Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| |
Collapse
|
86
|
Lagassé HAD, Alexaki A, Simhadri VL, Katagiri NH, Jankowski W, Sauna ZE, Kimchi-Sarfaty C. Recent advances in (therapeutic protein) drug development. F1000Res 2017; 6:113. [PMID: 28232867 PMCID: PMC5302153 DOI: 10.12688/f1000research.9970.1] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2017] [Indexed: 01/11/2023] Open
Abstract
Therapeutic protein drugs are an important class of medicines serving patients most in need of novel therapies. Recently approved recombinant protein therapeutics have been developed to treat a wide variety of clinical indications, including cancers, autoimmunity/inflammation, exposure to infectious agents, and genetic disorders. The latest advances in protein-engineering technologies have allowed drug developers and manufacturers to fine-tune and exploit desirable functional characteristics of proteins of interest while maintaining (and in some cases enhancing) product safety or efficacy or both. In this review, we highlight the emerging trends and approaches in protein drug development by using examples of therapeutic proteins approved by the U.S. Food and Drug Administration over the previous five years (2011–2016, namely January 1, 2011, through August 31, 2016).
Collapse
Affiliation(s)
- H A Daniel Lagassé
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Aikaterini Alexaki
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Vijaya L Simhadri
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Nobuko H Katagiri
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Wojciech Jankowski
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Zuben E Sauna
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Chava Kimchi-Sarfaty
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
87
|
Tobinai K, Klein C, Oya N, Fingerle-Rowson G. A Review of Obinutuzumab (GA101), a Novel Type II Anti-CD20 Monoclonal Antibody, for the Treatment of Patients with B-Cell Malignancies. Adv Ther 2017; 34:324-356. [PMID: 28004361 PMCID: PMC5331088 DOI: 10.1007/s12325-016-0451-1] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Indexed: 12/26/2022]
Abstract
Obinutuzumab (GA101) is a novel, type II, glycoengineered, humanized anti-CD20 monoclonal antibody that has been developed to address the need for new therapeutics with improved efficacy in patients with lymphocytic leukemia and lymphoma of B-cell origin. Obinutuzumab has a distinct mode of action relative to type I anti-CD20 antibodies, such as rituximab, working primarily by inducing direct cell death and antibody-dependent cell-mediated cytotoxicity. Obinutuzumab is under investigation in a wide-ranging program of clinical trials in patients with B-cell malignancies. Efficacy as monotherapy has been reported in patients with relapsed/refractory indolent and aggressive non-Hodgkin lymphoma (NHL) and in chronic lymphocytic leukemia (CLL) of B-cell origin. Improved outcomes have also been noted when obinutuzumab is added to chemotherapy in patients with B-cell NHL, and superiority over rituximab has been reported with combination therapy in patients with CLL. Ongoing research is focusing on developing options for chemotherapy-free treatment and on new combinations of obinutuzumab with novel targeted agents.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD20/immunology
- Antineoplastic Agents/pharmacology
- B-Lymphocytes/immunology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/immunology
Collapse
Affiliation(s)
- Kensei Tobinai
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan.
| | - Christian Klein
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Naoko Oya
- Oncology Lifecycle Management Department, Chugai Pharmaceutical Co. Ltd, Tokyo, Japan
| | | |
Collapse
|
88
|
Unexpected cross-reactivity of anti-cathepsin B antibodies leads to uncertainties regarding the mechanism of action of anti-CD20 monoclonal antibody GA101. Leuk Res 2017; 55:41-48. [PMID: 28122282 DOI: 10.1016/j.leukres.2017.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/17/2016] [Accepted: 01/04/2017] [Indexed: 12/21/2022]
Abstract
GA101, also known as obinutuzumab or Gazyva (Gazyvaro), is a glycoengineered type II humanized antibody that targets the CD20 antigen expressed at the surface of B-cells. This novel anti-CD20 antibody is currently assessed in clinical trials with promising results as a single agent or as part of therapeutic combinations for the treatment of B-cell malignancies. Detailed understanding of the mechanisms of GA101-induced cell death is needed to get insight into possible resistance mechanisms occurring in patients. Although multiple in vitro and in vivo mechanisms have been suggested to describe the effects of GA101 on B-cells, currently available data are ambiguous. The aim of our study was to clarify the cellular mechanisms involved in GA101-induced cell death in vitro, and more particularly the respective roles played by lysosomal and mitochondrial membrane permeabilization. Our results confirm previous reports suggesting that GA101 triggers homotypic adhesion and caspase-independent cell death, two processes that are dependent on actin remodeling and involve the production of reactive oxygen species. With respect to lysosomal membrane permeabilization (LMP), our data suggest that lack of specificity of available antibodies directed against cathepsin B may have confounded previously published results, possibly challenging current LMP-driven model of GA101 action mode.
Collapse
|
89
|
An FDA perspective on the assessment of proposed biosimilar therapeutic proteins in rheumatology. Nat Rev Rheumatol 2017; 13:123-128. [DOI: 10.1038/nrrheum.2016.204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
90
|
Johannssen T, Lepenies B. Glycan-Based Cell Targeting To Modulate Immune Responses. Trends Biotechnol 2016; 35:334-346. [PMID: 28277249 DOI: 10.1016/j.tibtech.2016.10.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023]
Abstract
Glycosylation is an integral post-translational modification present in more than half of all eukaryotic proteins. It affects key protein functions, including folding, stability, and immunogenicity. Glycoengineering approaches, such as the use of bacterial N-glycosylation systems, or expression systems, including yeasts, insect cells, and mammalian cells, have enabled access to defined and homogenous glycoproteins. Given that glycan structures on proteins can be recognized by host lectin receptors, they may facilitate cell-specific targeting and immune modulation. Myeloid C-type lectin receptors (CLRs) expressed by antigen-presenting cells are attractive targets to shape immune responses. Multivalent glycan display on nanoparticles, liposomes, or dendrimers has successfully enabled CLR targeting. In this review, we discuss novel strategies to access defined glycan structures and highlight CLR targeting approaches for immune modulation.
Collapse
Affiliation(s)
- Timo Johannssen
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Mühlenberg 1, 14476 Potsdam, Germany; Freie Universität Berlin, Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Arnimallee 22, 14195 Berlin, Germany; University of Veterinary Medicine Hannover, Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany
| | - Bernd Lepenies
- University of Veterinary Medicine Hannover, Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
91
|
Grugan KD, Dorn K, Jarantow SW, Bushey BS, Pardinas JR, Laquerre S, Moores SL, Chiu ML. Fc-mediated activity of EGFR x c-Met bispecific antibody JNJ-61186372 enhanced killing of lung cancer cells. MAbs 2016; 9:114-126. [PMID: 27786612 PMCID: PMC5240640 DOI: 10.1080/19420862.2016.1249079] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) mutant non-small cell lung cancers acquire resistance to EGFR tyrosine kinase inhibitors through multiple mechanisms including c-Met receptor pathway activation. We generated a bispecific antibody targeting EGFR and c-Met (JNJ-61186372) demonstrating anti-tumor activity in wild-type and mutant EGFR settings with c-Met pathway activation. JNJ-61186372 was engineered with low fucosylation (<10 %), resulting in enhanced antibody-dependent cell-mediated cytotoxicity and FcγRIIIa binding. In vitro and in vivo studies with the single-arm EGFR or c-Met versions of JNJ-61186372 identified that the Fc-activity of JNJ-61186372 is mediated by binding of the anti-EGFR arm and required for inhibition of EGFR-driven tumor cells. In a tumor model driven by both EGFR and c-Met, treatment with Fc-silent JNJ-61186372 or with c-Met single-arm antibody reduced tumor growth inhibition compared to treatment with JNJ-61186372, suggesting that the Fc function of JNJ-61186372 is essential for maximal tumor inhibition. Moreover in this same model, downregulation of both EGFR and c-Met receptors was observed upon treatment with Fc-competent JNJ-61186372, suggesting that the Fc interactions are necessary for down-modulation of the receptors in vivo and for efficacy. These Fc-mediated activities, in combination with inhibition of both the EGFR and c-Met signaling pathways, highlight the multiple mechanisms by which JNJ-61186372 combats therapeutic resistance in EGFR mutant patients.
Collapse
Affiliation(s)
- Katharine D Grugan
- a Biologics Research, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Keri Dorn
- a Biologics Research, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Stephen W Jarantow
- a Biologics Research, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Barbara S Bushey
- b Oncology, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Jose R Pardinas
- a Biologics Research, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Sylvie Laquerre
- b Oncology, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Sheri L Moores
- b Oncology, Janssen Research and Development, LLC , Spring House , PA , USA
| | - Mark L Chiu
- a Biologics Research, Janssen Research and Development, LLC , Spring House , PA , USA
| |
Collapse
|
92
|
Increased FcγRIIB dominance contributes to the emergence of resistance to therapeutic antibodies in chronic lymphocytic leukaemia patients. Oncogene 2016; 36:2366-2376. [PMID: 27748757 DOI: 10.1038/onc.2016.387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/23/2016] [Accepted: 09/06/2016] [Indexed: 12/19/2022]
Abstract
Resistance to therapeutic antibodies in chronic lymphocytic leukaemia (CLL) is common. In this study, we show that therapeutic antibodies against CD62L (CD62L-Ab) or CD20 (obinutuzumab) were able to induce antibody-dependent cell-mediated cytotoxicity (ADCC) and phagocytosis (ADP) in primary cultures of CLL cells. CLL cells derived from patients with active disease requiring treatment displayed resistance to these antibodies, whereas patients with stable disease were sensitive. Using enrichment strategies and transcriptomic analyses, we show that antibody-dependent tumour cell killing was FcγR-dependent and mediated by macrophages. Moreover, we show that resistance cannot be attributed to total numbers or established subtypes of monocytes/macrophages, or the efficiency with which they bind an immune complex. Rather, ADCC/ADP resistance was due to reduced signalling activity through the activating FcγRs resulting in the transfer of dominance to the inhibitory FcγRIIb within macrophages. Most significantly, we show that resistance is an actionable event that could be reversed using inhibitors of FcγRIIb signalling in primary cultures of CLL cells that were previously insensitive to obinutuzumab or CD62L-Ab.
Collapse
|
93
|
Lu LL, Chung AW, Rosebrock T, Ghebremichael M, Yu WH, Grace PS, Schoen MK, Tafesse F, Martin C, Leung V, Mahan AE, Sips M, Kumar M, Tedesco J, Robinson H, Tkachenko E, Draghi M, Freedberg KJ, Streeck H, Suscovich TJ, Lauffenburger D, Restrepo BI, Day C, Fortune SM, Alter G. A Functional Role for Antibodies in Tuberculosis. Cell 2016; 167:433-443.e14. [PMID: 27667685 PMCID: PMC5526202 DOI: 10.1016/j.cell.2016.08.072] [Citation(s) in RCA: 413] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/04/2016] [Accepted: 08/26/2016] [Indexed: 11/18/2022]
Abstract
While a third of the world carries the burden of tuberculosis, disease control has been hindered by a lack of tools, including a rapid, point-of-care diagnostic and a protective vaccine. In many infectious diseases, antibodies (Abs) are powerful biomarkers and important immune mediators. However, in Mycobacterium tuberculosis (Mtb) infection, a discriminatory or protective role for humoral immunity remains unclear. Using an unbiased antibody profiling approach, we show that individuals with latent tuberculosis infection (Ltb) and active tuberculosis disease (Atb) have distinct Mtb-specific humoral responses, such that Ltb infection is associated with unique Ab Fc functional profiles, selective binding to FcγRIII, and distinct Ab glycosylation patterns. Moreover, compared to Abs from Atb, Abs from Ltb drove enhanced phagolysosomal maturation, inflammasome activation, and, most importantly, macrophage killing of intracellular Mtb. Combined, these data point to a potential role for Fc-mediated Ab effector functions, tuned via differential glycosylation, in Mtb control.
Collapse
Affiliation(s)
- Lenette L. Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Amy W. Chung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Microbiology and Immunology, University of Melbourne, Doherty Institute for Infection and Immunity, Melbourne, 3000, Australia
| | - Tracy Rosebrock
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | | | - Wen Han Yu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | | | - Fikadu Tafesse
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Constance Martin
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Vivian Leung
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Alison E. Mahan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Magdalena Sips
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biomedical Molecular Biology, Ghent University, Ghent, 9000, Belgium
| | - Manu Kumar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | - Hannah Robinson
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | - Monia Draghi
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | | | | | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Blanca I. Restrepo
- School of Public Health, University of Texas Health Houston, Brownsville, TX, 78520, USA
| | - Cheryl Day
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30332, USA
- South African Tuberculosis Vaccine Initiative (SATVI) and School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory 7925 South Africa
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| |
Collapse
|
94
|
Grandjean CL, Montalvao F, Celli S, Michonneau D, Breart B, Garcia Z, Perro M, Freytag O, Gerdes CA, Bousso P. Intravital imaging reveals improved Kupffer cell-mediated phagocytosis as a mode of action of glycoengineered anti-CD20 antibodies. Sci Rep 2016; 6:34382. [PMID: 27698437 PMCID: PMC5048169 DOI: 10.1038/srep34382] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
Anti-CD20 monoclonal antibodies (mAbs) represent an effective treatment for a number of B cell malignancies and autoimmune disorders. Glycoengineering of anti-CD20mAb may contribute to increased anti-tumor efficacy through enhanced antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADP) as reported by in vitro studies. However, where and how glycoengineered Ab may potentiate therapeutic responses in vivo is yet to be elucidated. Here, we have performed mouse liver transplants to demonstrate that the liver is sufficient to mediate systemic B cells depletion after anti-CD20 treatment. Relying on intravital two-photon imaging of human CD20-expressing mice, we provide evidence that ADP by Kupffer cells (KC) is a major mechanism for rituximab-mediated B cell depletion. Notably, a glycoengineered anti-mouse CD20 Ab but not its wild-type counterpart triggered potent KC-mediated B cell depletion at low doses. Finally, distinct thresholds for KC phagocytosis were also observed for GA101 (obinutuzumab), a humanized glycoengineered type II anti-CD20 Ab and rituximab. Thus, we propose that enhanced phagocytosis of circulating B cells by KC represents an important in vivo mechanism underlying the improved activity of glycoengineered anti-CD20 mAbs.
Collapse
Affiliation(s)
- Capucine L Grandjean
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Fabricio Montalvao
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Susanna Celli
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - David Michonneau
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Beatrice Breart
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| | - Mario Perro
- Roche Innovation Center Zurich, Roche Pharma Research &Early Development, Wagistrasse 18,8952 Schlieren, Switzerland
| | - Olivier Freytag
- Roche Innovation Center Zurich, Roche Pharma Research &Early Development, Wagistrasse 18,8952 Schlieren, Switzerland
| | - Christian A Gerdes
- Roche Innovation Center Zurich, Roche Pharma Research &Early Development, Wagistrasse 18,8952 Schlieren, Switzerland
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labéllisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.,INSERM U1223, 75015 Paris, France
| |
Collapse
|
95
|
Chiang AW, Li S, Spahn PN, Richelle A, Kuo CC, Samoudi M, Lewis NE. Modulating carbohydrate-protein interactions through glycoengineering of monoclonal antibodies to impact cancer physiology. Curr Opin Struct Biol 2016; 40:104-111. [PMID: 27639240 DOI: 10.1016/j.sbi.2016.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/08/2016] [Accepted: 08/29/2016] [Indexed: 01/05/2023]
Abstract
Diverse glycans on proteins impact cell and organism physiology, along with drug activity. Since many protein-based biotherapeutics are glycosylated and these glycans have biological activity, there is a desire to engineer glycosylation for recombinant protein-based biotherapeutics. Engineered glycosylation can impact the recombinant protein efficacy and also influence many cell pathways by first changing glycan-protein interactions and consequently modulating disease physiologies. However, its complexity is enormous. Recent advances in glycoengineering now make it easier to modulate protein-glycan interactions. Here, we discuss how engineered glycans contribute to therapeutic monoclonal antibodies (mAbs) in the treatment of cancers, how these glycoengineered therapeutic mAbs affect the transformed phenotypes and downstream cell pathways. Furthermore, we suggest how systems biology can help in the next generation mAb glycoengineering process by aiding in data analysis and guiding engineering efforts to tailor mAb glycan and ultimately drug efficacy, safety and affordability.
Collapse
Affiliation(s)
- Austin Wt Chiang
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Shangzhong Li
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, CA, USA
| | - Philipp N Spahn
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Anne Richelle
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Chih-Chung Kuo
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, CA, USA
| | - Mojtaba Samoudi
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, CA, USA; The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, CA, USA.
| |
Collapse
|
96
|
Fiedler W, DeDosso S, Cresta S, Weidmann J, Tessari A, Salzberg M, Dietrich B, Baumeister H, Goletz S, Gianni L, Sessa C. A phase I study of PankoMab-GEX, a humanised glyco-optimised monoclonal antibody to a novel tumour-specific MUC1 glycopeptide epitope in patients with advanced carcinomas. Eur J Cancer 2016; 63:55-63. [PMID: 27285281 DOI: 10.1016/j.ejca.2016.05.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND A phase I open-label dose-escalation study was conducted to define the safety, tolerability, and pharmacokinetics (PK) of PankoMab-GEX, a glyco-optimised humanised IgG1, with high affinity to a novel tumour-specific glycopeptide epitope of MUC1 (TA-MUC1) with excellent preclinical anti-tumour activity. PATIENTS AND METHODS Seventy-four patients with advanced TA-MUC1-positive carcinomas received PankoMab-GEX intravenously every 3 (Q3W), 2 (Q2W), or 1 (QW) week in doses of 1-2200 mg in a three-plus-three dose-escalation design until disease progression (NCT01222624). RESULTS No maximum tolerated dose was reached. Adverse events were mainly mild-to-moderate infusion-related reactions (IRRs) by the first infusion in 45% of patients. Only one dose-limiting toxicity, a grade III IRR, was observed. PankoMab-GEX exhibited linear PK over all doses. Mean terminal half-life was 189 ± 66 h (Q3W), without dose dependency. A target trough level ≥50 μg/mL was reached after one infusion with doses ≥1700 mg Q3W in 80% of patients. Clinical benefit in 60 evaluable patients included one complete response in a patient with ovarian cancer treated 483 d and confirmed disease stabilisation in 19 patients lasting a median (range) of 23 (10-109) weeks. All but two of the patients with clinical benefit had received a compounded total dose ≥700 mg over a 3-week period, including 8 of 12 (67%) patients with ovarian cancer. CONCLUSION PankoMab-GEX is safe, well tolerated, and showed promising anti-tumour activity in advanced disease. A phase IIb study is ongoing evaluating the efficacy of PankoMab-GEX as a maintenance therapy in advanced ovarian cancer.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/therapeutic use
- Carcinoma/drug therapy
- Carcinoma/immunology
- Dose-Response Relationship, Drug
- Epitopes
- Female
- Humans
- Male
- Middle Aged
- Mucin-1/immunology
Collapse
Affiliation(s)
- W Fiedler
- Hubertus-Wald University Cancer Center, Dept. of Medicine II, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - S DeDosso
- Oncology Institute of Southern Switzerland, Ospedale Regionale Bellinzona e Valli, 6500 Bellinzona, Switzerland.
| | - S Cresta
- Fondazione IRCCS, Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milano, Italy.
| | - J Weidmann
- Hubertus-Wald University Cancer Center, Dept. of Medicine II, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - A Tessari
- Fondazione IRCCS, Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milano, Italy
| | - M Salzberg
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany.
| | - B Dietrich
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany.
| | - H Baumeister
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany.
| | - S Goletz
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany.
| | - L Gianni
- Department of Medical Oncology, Ospedale San Raffaele, Via Olgettina 60, 20132 Milano, Italy.
| | - C Sessa
- Oncology Institute of Southern Switzerland, Ospedale Regionale Bellinzona e Valli, 6500 Bellinzona, Switzerland.
| |
Collapse
|
97
|
Velmurugan R, Challa DK, Ram S, Ober RJ, Ward ES. Macrophage-Mediated Trogocytosis Leads to Death of Antibody-Opsonized Tumor Cells. Mol Cancer Ther 2016; 15:1879-89. [PMID: 27226489 DOI: 10.1158/1535-7163.mct-15-0335] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/17/2016] [Indexed: 12/16/2022]
Abstract
Understanding the complex behavior of effector cells such as monocytes or macrophages in regulating cancerous growth is of central importance for cancer immunotherapy. Earlier studies using CD20-specific antibodies have demonstrated that the Fcγ receptor (FcγR)-mediated transfer of the targeted receptors from tumor cells to these effector cells through trogocytosis can enable escape from antibody therapy, leading to the viewpoint that this process is protumorigenic. In the current study, we demonstrate that persistent trogocytic attack results in the killing of HER2-overexpressing breast cancer cells. Further, antibody engineering to increase FcγR interactions enhances this tumoricidal activity. These studies extend the complex repertoire of activities of macrophages to trogocytic-mediated cell death of HER2-overexpressing target cells and have implications for the development of effective antibody-based therapies. Mol Cancer Ther; 15(8); 1879-89. ©2016 AACR.
Collapse
Affiliation(s)
- Ramraj Velmurugan
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas. Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - Dilip K Challa
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas. Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - Sripad Ram
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Biomedical Engineering, Texas A&M University, College Station, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas.
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas. Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas. Department of Immunology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
98
|
Xu H, Guo H, Cheung IY, Cheung NKV. Antitumor Efficacy of Anti-GD2 IgG1 Is Enhanced by Fc Glyco-Engineering. Cancer Immunol Res 2016; 4:631-8. [PMID: 27197064 DOI: 10.1158/2326-6066.cir-15-0221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
The affinity of therapeutic antibodies for Fcγ receptors (FcγRs) strongly influences their antitumor potency. To generate antibodies with optimal binding and immunologic efficacy, we compared the affinities of different versions of an IgG1 Fc region that had an altered peptide backbone, altered glycans, or both. To produce IgG1 with glycans that lacked α1,6-fucose, we used CHO cells that were deficient in the enzyme UDP-N-acetylglucosamine: α-3-d-mannoside-β-1,2-N-acetylglucosaminyltransferase I (GnT1), encoded by the MGAT1 gene. Mature N-linked glycans require this enzyme, and without it, CHO cells synthesize antibodies carrying only Man5-GlcNAc2, which were more effective in antibody-dependent cell-mediated cytotoxicity (ADCC). Our engineered IgG1, hu3F8-IgG1, is specific for GD2, a neuroendocrine tumor ganglioside. Its peptide mutant is IgG1-DEL (S239D/I332E/A330L), both produced in wild-type CHO cells. When produced in GnT1-deficient CHO cells, we refer to them as IgG1n and IgG1n-DEL, respectively. Affinities for human FcγRs were measured using Biacore T-100 (on CD16 and CD32 polymorphic alleles), their immunologic properties compared for ADCC and complement-mediated cytotoxicity (CMC) in vitro, and pharmacokinetics and antitumor effects were compared in vivo in humanized mice. IgG1n and IgG1n-DEL contained only mannose and acetylglucosamine and had preferential affinity for activating CD16s, over inhibitory CD32B, receptors. In vivo, the antitumor effects of IgG1, IgG1-DEL, and IgG1n-DEL were similar but modest, whereas IgG1n was significantly more effective (P < 0.05). Thus, IgG1n antibodies produced in GnT1-deficient CHO cells may have potential as improved anticancer therapeutics. Cancer Immunol Res; 4(7); 631-8. ©2016 AACR.
Collapse
Affiliation(s)
- Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hongfen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
99
|
Cheeseman HM, Carias AM, Evans AB, Olejniczak NJ, Ziprin P, King DFL, Hope TJ, Shattock RJ. Expression Profile of Human Fc Receptors in Mucosal Tissue: Implications for Antibody-Dependent Cellular Effector Functions Targeting HIV-1 Transmission. PLoS One 2016; 11:e0154656. [PMID: 27164006 PMCID: PMC4862624 DOI: 10.1371/journal.pone.0154656] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/15/2016] [Indexed: 12/31/2022] Open
Abstract
The majority of new Human Immunodeficiency Virus (HIV)-1 infections are acquired via sexual transmission at mucosal surfaces. Partial efficacy (31.2%) of the Thai RV144 HIV-1 vaccine trial has been correlated with Antibody-dependent Cellular Cytotoxicity (ADCC) mediated by non-neutralizing antibodies targeting the V1V2 region of the HIV-1 envelope. This has led to speculation that ADCC and other antibody-dependent cellular effector functions might provide an important defense against mucosal acquisition of HIV-1 infection. However, the ability of antibody-dependent cellular effector mechanisms to impact on early mucosal transmission events will depend on a variety of parameters including effector cell type, frequency, the class of Fc-Receptor (FcR) expressed, the number of FcR per cell and the glycoslyation pattern of the induced antibodies. In this study, we characterize and compare the frequency and phenotype of IgG (CD16 [FcγRIII], CD32 [FcγRII] and CD64 [FcγRI]) and IgA (CD89 [FcαR]) receptor expression on effector cells within male and female genital mucosal tissue, colorectal tissue and red blood cell-lysed whole blood. The frequency of FcR expression on CD14+ monocytic cells, myeloid dendritic cells and natural killer cells were similar across the three mucosal tissue compartments, but significantly lower when compared to the FcR expression profile of effector cells isolated from whole blood, with many cells negative for all FcRs. Of the three tissues tested, penile tissue had the highest percentage of FcR positive effector cells. Immunofluorescent staining was used to determine the location of CD14+, CD11c+ and CD56+ cells within the three mucosal tissues. We show that the majority of effector cells across the different mucosal locations reside within the subepithelial lamina propria. The potential implication of the observed FcR expression patterns on the effectiveness of FcR-dependent cellular effector functions to impact on the initial events in mucosal transmission and dissemination warrants further mechanistic studies.
Collapse
Affiliation(s)
- Hannah M Cheeseman
- Imperial College London, Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, London, United Kingdom
| | - Ann M Carias
- Northwestern University, Feinberg School of Medicine, Cell and Molecular Biology Department, Chicago, Illinois, United States of America
| | - Abbey B Evans
- Imperial College London, Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, London, United Kingdom
| | - Natalia J Olejniczak
- Imperial College London, Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, London, United Kingdom
| | - Paul Ziprin
- Imperial College London, Department of Surgery, St. Mary's Hospital, London, United Kingdom
| | - Deborah F L King
- Imperial College London, Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, London, United Kingdom
| | - Thomas J Hope
- Northwestern University, Feinberg School of Medicine, Cell and Molecular Biology Department, Chicago, Illinois, United States of America
| | - Robin J Shattock
- Imperial College London, Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, London, United Kingdom
| |
Collapse
|
100
|
Herting F, Herter S, Friess T, Muth G, Bacac M, Sulcova J, Umana P, Dangl M, Klein C. Antitumour activity of the glycoengineered type
II
anti‐
CD
20 antibody obinutuzumab (
GA
101) in combination with the
MDM
2‐selective antagonist idasanutlin (
RG
7388). Eur J Haematol 2016; 97:461-470. [DOI: 10.1111/ejh.12756] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2016] [Indexed: 01/16/2023]
Affiliation(s)
- Frank Herting
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Munich Munich Germany
| | - Sylvia Herter
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Zurich Schlieren Switzerland
| | - Thomas Friess
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Munich Munich Germany
| | - Gunther Muth
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Munich Munich Germany
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Zurich Schlieren Switzerland
| | - Jitka Sulcova
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Zurich Schlieren Switzerland
| | - Pablo Umana
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Zurich Schlieren Switzerland
| | - Markus Dangl
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Munich Munich Germany
| | - Christian Klein
- Roche Pharmaceutical Research and Early Development Roche Innovation Center Zurich Schlieren Switzerland
| |
Collapse
|