101
|
Wang TT, Maamary J, Tan GS, Bournazos S, Davis CW, Krammer F, Schlesinger SJ, Palese P, Ahmed R, Ravetch JV. Anti-HA Glycoforms Drive B Cell Affinity Selection and Determine Influenza Vaccine Efficacy. Cell 2015; 162:160-9. [PMID: 26140596 DOI: 10.1016/j.cell.2015.06.026] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/13/2015] [Accepted: 05/11/2015] [Indexed: 01/05/2023]
Abstract
Protective vaccines elicit high-affinity, neutralizing antibodies by selection of somatically hypermutated B cell antigen receptors (BCR) on immune complexes (ICs). This implicates Fc-Fc receptor (FcR) interactions in affinity maturation, which, in turn, are determined by IgG subclass and Fc glycan composition within ICs. Trivalent influenza virus vaccination elicited regulation of anti-hemagglutinin (HA) IgG subclass and Fc glycans, with abundance of sialylated Fc glycans (sFc) predicting quality of vaccine response. We show that sFcs drive BCR affinity selection by binding the Type-II FcR CD23, thus upregulating the inhibitory FcγRIIB on activated B cells. This elevates the threshold requirement for BCR signaling, resulting in B cell selection for higher affinity BCR. Immunization with sFc HA ICs elicited protective, high-affinity IgGs against the conserved stalk of the HA. These results reveal a novel, endogenous pathway for affinity maturation that can be exploited for eliciting high-affinity, broadly neutralizing antibodies through immunization with sialylated immune complexes.
Collapse
Affiliation(s)
- Taia T Wang
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Jad Maamary
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Gene S Tan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Stylianos Bournazos
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Carl W Davis
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah J Schlesinger
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeffrey V Ravetch
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
102
|
Reusch D, Tejada ML. Fc glycans of therapeutic antibodies as critical quality attributes. Glycobiology 2015; 25:1325-34. [PMID: 26263923 PMCID: PMC4634315 DOI: 10.1093/glycob/cwv065] [Citation(s) in RCA: 310] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/07/2015] [Indexed: 12/12/2022] Open
Abstract
Critical quality attributes (CQA) are physical, chemical, biological or microbiological properties or characteristics that must be within an appropriate limit, range or distribution to ensure the desired product quality, safety and efficacy. For monoclonal antibody therapeutics that rely on fraction crystalizable (Fc)-mediated effector function for their clinical activity, the terminal sugars of Fc glycans have been shown to be critical for safety or efficacy. Different glycosylation variants have also been shown to influence the pharmacodynamic and pharmacokinetic behavior while other Fc glycan structural elements may be involved in adverse immune reactions. This review focuses on the role of Fc glycans as CQAs. Fc glycan information from the published literature is summarized and evaluated for impact on patient safety, immunogenicity, bioactivity and pharmacodynamics/pharmacokinetics.
Collapse
Affiliation(s)
- Dietmar Reusch
- Pharma Biotech Development Penzberg, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | - Max L Tejada
- Biological Technologies, Genentech, CA 94080, USA
| |
Collapse
|
103
|
Lee HJ, Kim JY, Park IA, Song IH, Yu JH, Ahn JH, Gong G. Prognostic Significance of Tumor-Infiltrating Lymphocytes and the Tertiary Lymphoid Structures in HER2-Positive Breast Cancer Treated With Adjuvant Trastuzumab. Am J Clin Pathol 2015; 144:278-88. [PMID: 26185313 DOI: 10.1309/ajcpixuydvz0rz3g] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Tumor-infiltrating lymphocytes (TILs) have prognostic significance in breast cancer. The tertiary lymphoid structure (TLS) is related to the influx of TILs, and expression of major histocompatibility complex (MHC) I in tumor cells is necessary for the effective action of TILs. METHODS We retrospectively evaluated the relationship of TILs and TLS and the expression of MHC I in 447 HER2-positive breast cancers treated with chemotherapy and 1 year of trastuzumab. RESULTS TILs were more abundant in hormone receptor (HR)-/HER2+ tumors than in HR+/HER2+ tumors. HR-/HER2+ breast cancers with abundant TILs showed a higher histologic grade, the absence of lymphovascular invasion, the presence of peritumoral lymphocytic infiltration, moderate to abundant TLSs in adjacent tissue, and stronger HLA-ABC and HLA-A expression. Abundant TILs and the absence of lymphovascular invasion were found to be good, independent prognostic factors for disease-free survival in patients with HR-/HER2+ breast cancer. The level of TILs was not associated with the patients' prognosis in HR+ tumors. CONCLUSIONS Abundant TILs are an independent prognostic factor in HR-/HER2+ breast cancers. Evaluation of TILs in HR-/HER2+ breast cancers may provide valuable information regarding the prognosis of patients treated using adjuvant chemotherapy and trastuzumab.
Collapse
Affiliation(s)
- Hee Jin Lee
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Joo Young Kim
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - In Ah Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - In Hye Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jong Han Yu
- Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jin-Hee Ahn
- sDepartment of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Gyungyub Gong
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
104
|
Wang W, Erbe AK, Hank JA, Morris ZS, Sondel PM. NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in Cancer Immunotherapy. Front Immunol 2015; 6:368. [PMID: 26284063 PMCID: PMC4515552 DOI: 10.3389/fimmu.2015.00368] [Citation(s) in RCA: 384] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/06/2015] [Indexed: 12/21/2022] Open
Abstract
Natural killer (NK) cells play a major role in cancer immunotherapies that involve tumor-antigen targeting by monoclonal antibodies (mAbs). NK cells express a variety of activating and inhibitory receptors that serve to regulate the function and activity of the cells. In the context of targeting cells, NK cells can be "specifically activated" through certain Fc receptors that are expressed on their cell surface. NK cells can express FcγRIIIA and/or FcγRIIC, which can bind to the Fc portion of immunoglobulins, transmitting activating signals within NK cells. Once activated through Fc receptors by antibodies bound to target cells, NK cells are able to lyse target cells without priming, and secrete cytokines like interferon gamma to recruit adaptive immune cells. This antibody-dependent cell-mediated cytotoxicity (ADCC) of tumor cells is utilized in the treatment of various cancers overexpressing unique antigens, such as neuroblastoma, breast cancer, B cell lymphoma, and others. NK cells also express a family of receptors called killer immunoglobulin-like receptors (KIRs), which regulate the function and response of NK cells toward target cells through their interaction with their cognate ligands that are expressed on tumor cells. Genetic polymorphisms in KIR and KIR-ligands, as well as FcγRs may influence NK cell responsiveness in conjunction with mAb immunotherapies. This review focuses on current therapeutic mAbs, different strategies to augment the anti-tumor efficacy of ADCC, and genotypic factors that may influence patient responses to antibody-dependent immunotherapies.
Collapse
Affiliation(s)
- Wei Wang
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacquelyn A. Hank
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
105
|
Kurogochi M, Mori M, Osumi K, Tojino M, Sugawara SI, Takashima S, Hirose Y, Tsukimura W, Mizuno M, Amano J, Matsuda A, Tomita M, Takayanagi A, Shoda SI, Shirai T. Glycoengineered Monoclonal Antibodies with Homogeneous Glycan (M3, G0, G2, and A2) Using a Chemoenzymatic Approach Have Different Affinities for FcγRIIIa and Variable Antibody-Dependent Cellular Cytotoxicity Activities. PLoS One 2015. [PMID: 26200113 PMCID: PMC4511734 DOI: 10.1371/journal.pone.0132848] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Many therapeutic antibodies have been developed, and IgG antibodies have been extensively generated in various cell expression systems. IgG antibodies contain N-glycans at the constant region of the heavy chain (Fc domain), and their N-glycosylation patterns differ during various processes or among cell expression systems. The Fc N-glycan can modulate the effector functions of IgG antibodies, such as antibody-dependent cellular cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC). To control Fc N-glycans, we performed a rearrangement of Fc N-glycans from a heterogeneous N-glycosylation pattern to homogeneous N-glycans using chemoenzymatic approaches with two types of endo-β-N-acetyl glucosaminidases (ENG'ases), one that works as a hydrolase to cleave all heterogeneous N-glycans, another that is used as a glycosynthase to generate homogeneous N-glycans. As starting materials, we used an anti-Her2 antibody produced in transgenic silkworm cocoon, which consists of non-fucosylated pauci-mannose type (Man2-3GlcNAc2), high-mannose type (Man4-9GlcNAc2), and complex type (Man3GlcNAc3-4) N-glycans. As a result of the cleavage of several ENG'ases (endoS, endoM, endoD, endoH, and endoLL), the heterogeneous glycans on antibodies were fully transformed into homogeneous-GlcNAc by a combination of endoS, endoD, and endoLL. Next, the desired N-glycans (M3; Man3GlcNAc1, G0; GlcNAc2Man3GlcNAc1, G2; Gal2GlcNAc2Man3GlcNAc1, A2; NeuAc2Gal2GlcNAc2Man3GlcNAc1) were transferred from the corresponding oxazolines to the GlcNAc residue on the intact anti-Her2 antibody with an ENG'ase mutant (endoS-D233Q), and the glycoengineered anti-Her2 antibody was obtained. The binding assay of anti-Her2 antibody with homogenous N-glycans with FcγRIIIa-V158 showed that the glycoform influenced the affinity for FcγRIIIa-V158. In addition, the ADCC assay for the glycoengineered anti-Her2 antibody (mAb-M3, mAb-G0, mAb-G2, and mAb-A2) was performed using SKBR-3 and BT-474 as target cells, and revealed that the glycoform influenced ADCC activity.
Collapse
Affiliation(s)
- Masaki Kurogochi
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Masako Mori
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Kenji Osumi
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Mami Tojino
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Shu-ichi Sugawara
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Shou Takashima
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Yuriko Hirose
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Wataru Tsukimura
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Junko Amano
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Akio Matsuda
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Masahiro Tomita
- Immuno-Biological Laboratories Co., Ltd., 1091-1 Naka, Fujioka-shi, Gunma, Japan
| | - Atsushi Takayanagi
- Department of Molecular Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Shin-Ichiro Shoda
- Graduate School of Engineering, Tohoku University, Aoba-ku, Sendai, Japan
| | - Takashi Shirai
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
106
|
Liu SD, Lowe JB. Implications of understanding the signaling, cellular, and cytotoxic mechanisms afforded by afucosylated antibodies. Oncoimmunology 2015; 4:e1009288. [PMID: 26155429 DOI: 10.1080/2162402x.2015.1009288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 10/23/2022] Open
Abstract
Afucosylated antibodies potentiate natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC) by enhancing signaling pathways and cellular processes, which in turn, increases cytotoxic potential. Importantly, a better understanding of these processes and properties will aid in exploiting them to help design therapeutic antibodies and strategies that may be of the greatest benefit to patients.
Collapse
Affiliation(s)
- Scot D Liu
- Department of Pathology; Genentech, Inc. ; San Francisco, CA, USA
| | - John B Lowe
- Department of Pathology; Genentech, Inc. ; San Francisco, CA, USA
| |
Collapse
|
107
|
Huang R, Sun Y, Zhang XY, Sun BW, Wang QC, Zhu J. Biological evaluation of a novel Herceptin-platinum (II) conjugate for efficient and cancer cell specific delivery. Biomed Pharmacother 2015. [DOI: 10.1016/j.biopha.2015.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
108
|
Brown EP, Normandin E, Osei-Owusu NY, Mahan AE, Chan YN, Lai JI, Vaccari M, Rao M, Franchini G, Alter G, Ackerman ME. Microscale purification of antigen-specific antibodies. J Immunol Methods 2015; 425:27-36. [PMID: 26078040 DOI: 10.1016/j.jim.2015.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 10/23/2022]
Abstract
Glycosylation of the Fc domain is an important driver of antibody effector function. While assessment of antibody glycoform compositions observed across total plasma IgG has identified differences associated with a variety of clinical conditions, in many cases it is the glycosylation state of only antibodies against a specific antigen or set of antigens that may be of interest, for example, in defining the potential effector function of antibodies produced during disease or after vaccination. Historically, glycoprofiling such antigen-specific antibodies in clinical samples has been challenging due to their low prevalence, the high sample requirement for most methods of glycan determination, and the lack of high-throughput purification methods. New methods of glycoprofiling with lower sample requirements and higher throughput have motivated the development of microscale and automatable methods for purification of antigen-specific antibodies from polyclonal sources such as clinical serum samples. In this work, we present a robot-compatible 96-well plate-based method for purification of antigen-specific antibodies, suitable for such population level glycosylation screening. We demonstrate the utility of this method across multiple antibody sources, using both purified plasma IgG and plasma, and across multiple different antigen types, with enrichment factors greater than 1000-fold observed. Using an on-column IdeS protease treatment, we further describe staged release of Fc and Fab domains, allowing for glycoprofiling of each domain.
Collapse
Affiliation(s)
- Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Erica Normandin
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Nana Yaw Osei-Owusu
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH 03755, United States
| | - Alison E Mahan
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, United States
| | - Ying N Chan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Jennifer I Lai
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Monica Vaccari
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, MD 20814, United States
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| | - Genoveffa Franchini
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, MD 20814, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, United States
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States.
| |
Collapse
|
109
|
Turini M, Chames P, Bruhns P, Baty D, Kerfelec B. A FcγRIII-engaging bispecific antibody expands the range of HER2-expressing breast tumors eligible to antibody therapy. Oncotarget 2015; 5:5304-19. [PMID: 24979648 PMCID: PMC4170649 DOI: 10.18632/oncotarget.2093] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Trastuzumab is established as treatment of HER2high metastatic breast cancers but many limitations impair its efficacy. Here, we report the design of a Fab-like bispecific antibody (HER2bsFab) that displays a moderate affinity for HER2 and a unique, specific and high affinity for FcγRIII. In vitro characterization showed that ADCC was the major mechanism of action of HER2bsFab as no significant HER2-driven effect was observed. HER2bsFab mediated ADCC at picomolar concentration against HER2high, HER2low as well as trastuzumab-refractive cell lines. In vivo HER2bsFab potently inhibited HER2high tumor growth by recruitment of mouse FcγRIII and IV-positive resident effector cells and more importantly, exhibited a net superiority over trastuzumab at inhibiting HER2low tumor growth. Moreover, FcγRIIIA-engagement by HER2bsFab was independent of V/F158 polymorphism and induced a stronger NK cells activation in response to target cell recognition. Thus, taking advantage of its epitope specificity and affinity for HER2 and FcγRIIIA, HER2bsFab exhibits potent anti-tumor activity against HER2low tumors while evading most of trastuzumab Fc-linked limitations thereby potentially enlarging the number of patients eligible for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Marc Turini
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| | - Patrick Chames
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| | - Pierre Bruhns
- Département d'Immunologie, Laboratoire Anticorps en Thérapie et Pathologie, Institut Pasteur, Paris, France. INSERM, U760, Paris, France
| | - Daniel Baty
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| | - Brigitte Kerfelec
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| |
Collapse
|
110
|
Boyerinas B, Jochems C, Fantini M, Heery CR, Gulley JL, Tsang KY, Schlom J. Antibody-Dependent Cellular Cytotoxicity Activity of a Novel Anti-PD-L1 Antibody Avelumab (MSB0010718C) on Human Tumor Cells. Cancer Immunol Res 2015; 3:1148-1157. [PMID: 26014098 DOI: 10.1158/2326-6066.cir-15-0059] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/19/2015] [Indexed: 12/18/2022]
Abstract
Several anti-PD-1/PD-L1 monoclonal antibodies (mAb) are currently providing evidence of clinical benefit in subsets of cancer patients. The mode of action of these mAbs is to inhibit PD-1 on immune cells interacting with PD-L1 on tumor cells. These mAbs are either designed or engineered to eliminate antibody-dependent cell-mediated cytotoxicity (ADCC), which, however, has been implicated as an important mechanism in several highly effective mAb-mediated cancer therapies. A fully human anti-PD-L1 mAb would potentially be able to block PD-1/PD-L1 interactions and also mediate the ADCC lysis of tumor cells. MSB0010718C (designated avelumab) is a fully human IgG1 anti-PD-L1 mAb. The studies reported here demonstrate (i) the ability of avelumab to lyse a range of human tumor cells in the presence of PBMC or NK effectors; (ii) IFNγ can enhance tumor cell PD-L1 expression and, in some cases, enhance ADCC tumor cell lysis; (iii) purified NK cells are potent effectors for avelumab; (iv) similar levels of avelumab-mediated ADCC lysis of tumor cells are seen using purified NK as effectors from either healthy donors or cancer patients; (v) very low levels of avelumab-mediated lysis are seen using whole PBMCs as targets; this finding complements results seen in analyses of PBMC subsets of patients receiving avelumab; and (vi) the addition of IL12 to NK cells greatly enhances avelumab-mediated ADCC. These studies thus provide an additional mode of action for an anti-PD-L1 mAb and support the rationale for further studies to enhance avelumab-mediated ADCC activity.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/genetics
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Biomarkers, Tumor
- Cell Line, Tumor
- Cell Membrane/metabolism
- Gene Expression
- Genotype
- Humans
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Interleukin-12/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/metabolism
- Receptors, IgG/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Benjamin Boyerinas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Massimo Fantini
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Kwong Yok Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
111
|
Physicochemical and biological characterization of a biosimilar trastuzumab. BIOMED RESEARCH INTERNATIONAL 2015; 2015:427235. [PMID: 26075238 PMCID: PMC4449878 DOI: 10.1155/2015/427235] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 03/10/2015] [Accepted: 03/24/2015] [Indexed: 12/21/2022]
Abstract
According to the World Health Organization, the incidence of malignant neoplasms and endocrine, blood, and immune disorders will increase in the upcoming decades along with the demand of affordable treatments. In response to this need, the development of biosimilar drugs is increasing worldwide. The approval of biosimilars relies on the compliance with international guidelines, starting with the demonstration of similarity in their physicochemical and functional properties against the reference product. Subsequent clinical studies are performed to demonstrate similar pharmacological behavior and to diminish the uncertainty related to their safety and efficacy. Herein we present a comparability exercise between a biosimilar trastuzumab and its reference product, by using a hierarchical strategy with an orthogonal approach, to assess the physicochemical and biological attributes with potential impact on its pharmacokinetics, pharmacodynamics, and immunogenicity. Our results showed that the high degree of similarity in the physicochemical attributes of the biosimilar trastuzumab with respect to the reference product resulted in comparable biological activity, demonstrating that a controlled process is able to provide consistently the expected product. These results also constitute the basis for the design of subsequent delimited pharmacological studies, as they diminish the uncertainty of exhibiting different profiles.
Collapse
|
112
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: an update for 2009-2010. MASS SPECTROMETRY REVIEWS 2015; 34:268-422. [PMID: 24863367 PMCID: PMC7168572 DOI: 10.1002/mas.21411] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 07/16/2013] [Accepted: 07/16/2013] [Indexed: 05/07/2023]
Abstract
This review is the sixth update of the original article published in 1999 on the application of MALDI mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2010. General aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, arrays and fragmentation are covered in the first part of the review and applications to various structural typed constitutes the remainder. The main groups of compound that are discussed in this section are oligo and polysaccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals. Many of these applications are presented in tabular form. Also discussed are medical and industrial applications of the technique, studies of enzyme reactions and applications to chemical synthesis.
Collapse
Affiliation(s)
- David J. Harvey
- Department of BiochemistryOxford Glycobiology InstituteUniversity of OxfordOxfordOX1 3QUUK
| |
Collapse
|
113
|
Stern HM, Gardner H, Burzykowski T, Elatre W, O'Brien C, Lackner MR, Pestano GA, Santiago A, Villalobos I, Eiermann W, Pienkowski T, Martin M, Robert N, Crown J, Nuciforo P, Bee V, Mackey J, Slamon DJ, Press MF. PTEN Loss Is Associated with Worse Outcome in HER2-Amplified Breast Cancer Patients but Is Not Associated with Trastuzumab Resistance. Clin Cancer Res 2015; 21:2065-74. [PMID: 25649019 DOI: 10.1158/1078-0432.ccr-14-2993] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/31/2014] [Indexed: 01/03/2023]
Abstract
PURPOSE To investigate the clinical relevance of PTEN in HER2-amplified and HER2-nonamplified disease. EXPERIMENTAL DESIGN We assessed PTEN status in two large adjuvant breast cancer trials (BCIRG-006 and BCIRG-005) using a PTEN immunohistochemical (IHC) assay that was previously validated in a panel of 33 breast cancer cell lines and prostate cancer tissues with known PTEN gene deletion. RESULTS In the HER2-positive patient population, absence of tumor cell PTEN staining occurred at a rate of 5.4% and was independent of ER/PR status. In contrast, 15.9% of HER2-negative patients exhibited absence of PTEN staining with the highest frequency seen in triple-negative breast cancer (TNBC) subgroup versus ER/PR-positive patients (35.1% vs. 10.9%). Complete absence of PTEN staining in tumor cells was associated with poor clinical outcome in HER2-positive disease. Those patients whose cancers demonstrated absent PTEN staining had a significant decrease in disease-free survival (DFS) and overall survival (OS) compared with patients with tumors exhibiting any PTEN staining patterns (low, moderate, or high). Trastuzumab appeared to provide clinical benefit even for patients lacking PTEN staining. In the HER2-negative population, there were no statistically significant differences in clinical outcome based on PTEN status. CONCLUSIONS This study is the largest to date examining PTEN status in breast cancer and the data suggest that the rate and significance of PTEN status differ between HER2-positive and HER2-negative disease. Furthermore, the data clearly suggest that HER2-positive patients with PTEN loss still benefit from trastuzumab.
Collapse
Affiliation(s)
- Howard M Stern
- Genentech Research and Early Development, South San Francisco, California
| | | | | | - Wafaa Elatre
- USC/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Carol O'Brien
- Genentech Research and Early Development, South San Francisco, California
| | - Mark R Lackner
- Genentech Research and Early Development, South San Francisco, California
| | | | - Angela Santiago
- USC/Norris Comprehensive Cancer Center, Los Angeles, California
| | | | | | | | | | - Nicholas Robert
- Virginia Cancer Specialists/U.S. Oncology Research Network, Fairfax, Virginia
| | - John Crown
- Irish Cooperative Oncology Research Group, St. Vincent's University Hospital, Dublin, Ireland
| | | | - Valerie Bee
- Cancer International Research Group/Translational Research in Oncology, Paris, France
| | - John Mackey
- Department of Oncology, University of Alberta, Edmonton, Canada
| | - Dennis J Slamon
- Department of Medicine, Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Michael F Press
- USC/Norris Comprehensive Cancer Center, Los Angeles, California.
| |
Collapse
|
114
|
Perez EA, Thompson EA, Ballman KV, Anderson SK, Asmann YW, Kalari KR, Eckel-Passow JE, Dueck AC, Tenner KS, Jen J, Fan JB, Geiger XJ, McCullough AE, Chen B, Jenkins RB, Sledge GW, Winer EP, Gralow JR, Reinholz MM. Genomic analysis reveals that immune function genes are strongly linked to clinical outcome in the North Central Cancer Treatment Group n9831 Adjuvant Trastuzumab Trial. J Clin Oncol 2015; 33:701-8. [PMID: 25605861 DOI: 10.1200/jco.2014.57.6298] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To develop a genomic signature that predicts benefit from trastuzumab in human epidermal growth factor receptor 2-positive breast cancer. PATIENTS AND METHODS DASL technology was used to quantify mRNA in samples from 1,282 patients enrolled onto the Combination Chemotherapy With or Without Trastuzumab in Treating Women With Breast Cancer (North Central Cancer Treatment Group N9831 [NCCTG-N9831]) adjuvant trastuzumab trial. Cox proportional hazard ratios (HRs), adjusted for significant clinicopathologic risk factors, were used to determine the association of each gene with relapse-free survival (RFS) for 433 patients who received chemotherapy alone (arm A) and 849 patients who received chemotherapy plus trastuzumab (arms B and C). Network and pathway analyses were used to identify key biologic processes linked to RFS. The signature was built by using a voting scheme. RESULTS Network and functional ontology analyses suggested that increased RFS was linked to a subset of immune function genes. A voting scheme model was used to define immune gene enrichment based on the expression of any nine or more of 14 immune function genes at or above the 0.40 quantile for the population. This model was used to identify immune gene-enriched tumors in arm A and arms B and C. Immune gene enrichment was linked to increased RFS in arms B and C (HR, 0.35; 95% CI, 0.22 to 0.55; P < .001), whereas arm B and C patients who did not exhibit immune gene enrichment did not benefit from trastuzumab (HR, 0.89; 95% CI, 0.62 to 1.28; P = .53). Enriched immune function gene expression as defined by our predictive signature was not associated with increased RFS in arm A (HR, 0.90; 95% CI, 0.60 to 1.37; P = .64). CONCLUSION Increased expression of a subset of immune function genes may provide a means of predicting benefit from adjuvant trastuzumab.
Collapse
Affiliation(s)
- Edith A Perez
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA.
| | - E Aubrey Thompson
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Karla V Ballman
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - S Keith Anderson
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Yan W Asmann
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Krishna R Kalari
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Jeanette E Eckel-Passow
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Amylou C Dueck
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Kathleen S Tenner
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Jin Jen
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Jian-Bing Fan
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Xochiquetzal J Geiger
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Ann E McCullough
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Beiyun Chen
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Robert B Jenkins
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - George W Sledge
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Eric P Winer
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Julie R Gralow
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| | - Monica M Reinholz
- Edith A. Perez, E. Aubrey Thompson, Yan W. Asmann, and Xochiquetzal J. Geiger, Mayo Clinic, Jacksonville, FL; Karla V. Ballman, S. Keith Anderson, Krishna R. Kalari, Jeanette E. Eckel-Passow, Kathleen S. Tenner, Jin Jen, Beiyun Chen, Robert B. Jenkins, and Monica M. Reinholz, Mayo Clinic, Rochester, MN; Amylou C. Dueck, and Ann E. McCullough, Mayo Clinic, Scottsdale, AZ; Jian-Bing Fan, Illumina, San Diego, CA; George W. Sledge, Stanford School of Medicine, Palo Alto, CA; Eric P. Winer, Harvard Medical School, Boston, MA; and Julie R. Gralow, Seattle Cancer Care Alliance, Seattle, WA
| |
Collapse
|
115
|
Moorthy BS, Xie B, Moussa EM, Iyer LK, Chandrasekhar S, Panchal JP, Topp EM. Effect of Hydrolytic Degradation on the In Vivo Properties of Monoclonal Antibodies. BIOBETTERS 2015. [DOI: 10.1007/978-1-4939-2543-8_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
116
|
HER2-family signalling mechanisms, clinical implications and targeting in breast cancer. Breast Cancer Res Treat 2014; 149:5-15. [PMID: 25542271 DOI: 10.1007/s10549-014-3250-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/17/2014] [Indexed: 12/18/2022]
Abstract
Approximately 20 % of human breast cancers (BC) overexpress HER2 protein, and HER2-positivity is associated with a worse prognosis. Although HER2-targeted therapies have significantly improved outcomes for HER2-positive BC patients, resistance to trastuzumab-based therapy remains a clinical problem. In order to better understand resistance to HER2-targeted therapies in HER2-positive BC, it is necessary to examine HER family signalling as a whole. An extensive literature search was carried out to critically assess the current knowledge of HER family signalling in HER2-positive BC and response to HER2-targeted therapy. Known mechanisms of trastuzumab resistance include reduced receptor-antibody binding (MUC4, p95HER2), increased signalling through alternative HER family receptor tyrosine kinases (RTK), altered intracellular signalling involving loss of PTEN, reduced p27kip1, or increased PI3K/AKT activity and altered signalling via non-HER family RTKs such as IGF1R. Emerging strategies to circumvent resistance to HER2-targeted therapies in HER2-positive BC include co-targeting HER2/PI3K, pan-HER family inhibition, and novel therapies such as T-DM1. There is evidence that immunity plays a key role in the efficacy of HER-targeted therapy, and efforts are being made to exploit the immune system in order to improve the efficacy of current anti-HER therapies. With our rapidly expanding understanding of HER2 signalling mechanisms along with the repertoire of HER family and other targeted therapies, it is likely that the near future holds further dramatic improvements to the prognosis of women with HER2-positive BC.
Collapse
|
117
|
Tomasevic N, Luehrsen K, Baer M, Palath V, Martinez D, Williams J, Yi C, Sujatha-Bhaskar S, Lanke R, Leung J, Ching W, Lee A, Bai L, Yarranton G, Bebbington C. A high affinity recombinant antibody to the human EphA3 receptor with enhanced ADCC activity. Growth Factors 2014; 32:223-35. [PMID: 25413948 DOI: 10.3109/08977194.2014.984808] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
EphA3 is expressed in solid tumors and leukemias and is an attractive target for the therapy. We have generated a panel of Humaneered® antibodies to the ligand-binding domain using a Fab epitope-focused library that has the same specificity as monoclonal antibody mIIIA4. A high-affinity antibody was selected that competes with the mIIIA4 antibody for binding to EphA3 and has an improved affinity of ∼1 nM. In order to generate an antibody with potent cell-killing activity the variable regions were assembled with human IgG1k constant regions and expressed in a Chinese hamster ovary (CHO) cell line deficient in fucosyl transferase. Non-fucosylated antibodies have been reported to have enhanced binding affinity for the IgG receptor CD16a (FcγRIIIa). The affinity of the antibody for recombinant CD16a was enhanced approximately 10-fold. This resulted in enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) activity against EphA3-expressing leukemic cells, providing a potent antibody for the evaluation as a therapeutic agent.
Collapse
|
118
|
Shefet-Carasso L, Benhar I. Antibody-targeted drugs and drug resistance--challenges and solutions. Drug Resist Updat 2014; 18:36-46. [PMID: 25476546 DOI: 10.1016/j.drup.2014.11.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/16/2014] [Accepted: 11/17/2014] [Indexed: 11/24/2022]
Abstract
Antibody-based therapy of various human malignancies has shown efficacy in the past 30 years and is now one of the most successful and leading strategies for targeted treatment of patients harboring hematological malignancies and solid tumors. Antibody-drug conjugates (ADCs) aim to take advantage of the affinity and specificity of monoclonal antibodies (mAbs) to selectively deliver potent cytotoxic drugs to antigen-expressing tumor cells. Key parameters for ADC include choosing the optimal components of the ADC (the antibody, the linker and the cytotoxic drug) and selecting the suitable cell-surface target antigen. Building on the success of recent FDA approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla), ADCs are currently a class of drugs with a robust pipeline with clinical applications that are rapidly expanding. The more ADCs are being evaluated in preclinical models and clinical trials, the clearer are becoming the parameters and the challenges required for their therapeutic success. This rapidly growing knowledge and clinical experience are revealing novel modalities and mechanisms of resistance to ADCs, hence offering plausible solutions to such challenges. Here, we review the key parameters for designing a powerful ADC, focusing on how ADCs are addressing the challenge of multiple drug resistance (MDR) and its rational overcoming.
Collapse
Affiliation(s)
- LeeRon Shefet-Carasso
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
119
|
Liu SD, Chalouni C, Young JC, Junttila TT, Sliwkowski MX, Lowe JB. Afucosylated antibodies increase activation of FcγRIIIa-dependent signaling components to intensify processes promoting ADCC. Cancer Immunol Res 2014; 3:173-83. [PMID: 25387893 DOI: 10.1158/2326-6066.cir-14-0125] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is a key mechanism by which therapeutic antibodies mediate their antitumor effects. The absence of fucose on the heavy chain of the antibody increases the affinity between the antibody and FcγRIIIa, which results in increased in vitro and in vivo ADCC compared with the fucosylated form. However, the cellular and molecular mechanisms responsible for increased ADCC are unknown. Through a series of biochemical and cellular studies, we find that human natural killer (NK) cells stimulated with afucosylated antibody exhibit enhanced activation of proximal FcγRIIIa signaling and downstream pathways, as well as enhanced cytoskeletal rearrangement and degranulation, relative to stimulation with fucosylated antibody. Furthermore, analysis of the interaction between human NK cells and targets using a high-throughput microscope-based antibody-dependent cytotoxicity assay shows that afucosylated antibodies increase the number of NK cells capable of killing multiple targets and the rate with which targets are killed. We conclude that the increase in affinity between afucosylated antibodies and FcγRIIIa enhances activation of signaling molecules, promoting cytoskeletal rearrangement and degranulation, which, in turn, potentiates the cytotoxic characteristics of NK cells to increase efficiency of ADCC.
Collapse
Affiliation(s)
- Scot D Liu
- Department of Pathology, Genentech, Inc., South San Francisco, California.
| | - Cecile Chalouni
- Department of Pathology, Genentech, Inc., South San Francisco, California
| | - Judy C Young
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California
| | - Teemu T Junttila
- Department of Cancer Immunotherapy and Hematology, Genentech, Inc., South San Francisco, California
| | - Mark X Sliwkowski
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - John B Lowe
- Department of Pathology, Genentech, Inc., South San Francisco, California
| |
Collapse
|
120
|
Zhang Z, Zhang Y, Sun Q, Feng F, Huhe M, Mi L, Chen Z. Preclinical Pharmacokinetics, Tolerability, and Pharmacodynamics of Metuzumab, a Novel CD147 Human–Mouse Chimeric and Glycoengineered Antibody. Mol Cancer Ther 2014; 14:162-73. [DOI: 10.1158/1535-7163.mct-14-0104] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
121
|
|
122
|
Kapur R, Della Valle L, Sonneveld M, Hipgrave Ederveen A, Visser R, Ligthart P, de Haas M, Wuhrer M, van der Schoot CE, Vidarsson G. Low anti-RhD IgG-Fc-fucosylation in pregnancy: a new variable predicting severity in haemolytic disease of the fetus and newborn. Br J Haematol 2014; 166:936-45. [PMID: 24909983 PMCID: PMC4282073 DOI: 10.1111/bjh.12965] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/23/2014] [Indexed: 12/19/2022]
Abstract
Haemolytic disease of the fetus and newborn (HDFN) may occur when maternal IgG antibodies against red blood cells (RBCs), often anti-RhD (anti-D) antibodies, cross the placenta and mediate the destruction of RBCs via phagocytic IgG-Fc-receptors (FcγR). Clinical severity is not strictly related to titre and is more accurately predicted by the diagnostically-applied monocyte-based antibody-dependent cellular cytotoxicity (ADCC), a sensitive test with relatively low specificity. This suggests that other factors are involved in the pathogenesis of HDFN. Binding of IgG to FcγR requires the N-linked glycan at position 297 in the IgG-Fc-region, consisting of several different glycoforms. We therefore systematically analysed IgG-derived glycopeptides by mass spectrometry from 70 anti-D IgG1 antibodies purified from the plasma of alloimmunized pregnant women. This revealed a variable decrease in Fc-fucosylation in the majority of anti-D IgG1 (even down to 12%), whereas the total IgG of these patients remained highly fucosylated, like in healthy individuals (>90%). The degree of anti-D fucosylation correlated significantly with CD16 (FcγRIIIa)-mediated ADCC, in agreement with increased affinity of defucosylated IgG to human FcγRIIIa. Additionally, low anti-D fucosylation correlated significantly with low fetal-neonatal haemoglobin levels, thus with increased haemolysis, suggesting IgG-fucosylation to be an important pathological feature in HDFN with diagnostic potential.
Collapse
Affiliation(s)
- Rick Kapur
- Department of Experimental Immunohaematology, Sanquin Research, Amsterdam and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Gillis C, Gouel-Chéron A, Jönsson F, Bruhns P. Contribution of Human FcγRs to Disease with Evidence from Human Polymorphisms and Transgenic Animal Studies. Front Immunol 2014; 5:254. [PMID: 24910634 PMCID: PMC4038777 DOI: 10.3389/fimmu.2014.00254] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/14/2014] [Indexed: 11/13/2022] Open
Abstract
The biological activities of human IgG antibodies predominantly rely on a family of receptors for the Fc portion of IgG, FcγRs: FcγRI, FcγRIIA, FcγRIIB, FcγRIIC, FcγRIIIA, FcγRIIIB, FcRL5, FcRn, and TRIM21. All FcγRs bind IgG at the cell surface, except FcRn and TRIM21 that bind IgG once internalized. The affinity of FcγRs for IgG is determined by polymorphisms of human FcγRs and ranges from 2 × 104 to 8 × 107 M−1. The biological functions of FcγRs extend from cellular activation or inhibition, IgG-internalization/endocytosis/phagocytosis to IgG transport and recycling. This review focuses on human FcγRs and intends to present an overview of the current understanding of how these receptors may contribute to various pathologies. It will define FcγRs and their polymorphic variants, their affinity for human IgG subclasses, and review the associations found between FcγR polymorphisms and human pathologies. It will also describe the human FcγR-transgenic mice that have been used to study the role of these receptors in autoimmune, inflammatory, and allergic disease models.
Collapse
Affiliation(s)
- Caitlin Gillis
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France
| | - Aurélie Gouel-Chéron
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France ; Department of Anesthesia and Intensive Care, Hospital of Bichat-Claude Bernard, Public Assistance-Hospitals of Paris , Paris , France
| | - Friederike Jönsson
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France
| | - Pierre Bruhns
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France
| |
Collapse
|
124
|
Fabi A, Mottolese M, Segatto O. Therapeutic targeting of ERBB2 in breast cancer: understanding resistance in the laboratory and combating it in the clinic. J Mol Med (Berl) 2014; 92:681-95. [PMID: 24861025 DOI: 10.1007/s00109-014-1169-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/12/2014] [Accepted: 05/14/2014] [Indexed: 01/21/2023]
Abstract
ERBB2 gene amplification occurs in about one quarter of breast carcinomas (BCs) and identifies a distinct clinical subset of BC. The introduction in the clinic of Trastuzumab, a humanized monoclonal antibody (mAb) directed to the ERBB2 extracellular domain, has had a great impact on the therapeutic management of ERBB2+ BC. Yet, not all patients respond to Trastuzumab and resistance develops also among patients that initially benefit from Trastuzumab-based regimens. Pre-clinical studies have discovered several mechanisms through which tumor cells may escape from Trastuzumab-mediated ERBB2 inhibition. These include rewiring of the ErbB signaling network, loss of ERBB2 expression, expression of ERBB2 isoforms refractory to Trastuzumab inhibition, vicarious signaling by non-ErbB tyrosine kinases and constitutive activation of downstream signaling routes, such as the PI3K pathway. While the relative contribution of each of these mechanisms to establishing Trastuzumab resistance in the clinical setting is not fully understood, much attention has been focused on abating resistance by achieving complete blockade of ERBB2-containing dimers. This approach, propelled by the development of novel anti-ERBB2 therapeutics, has led to the recent approval of Lapatinib, Pertuzumab and T-DM1 as additional anti-ERBB2 therapeutics in BC. However, full success is far from being achieved and resistance to ERBB2 targeting remains a relevant problem in the clinical management of BC. Herein, we provide an overview of biological and molecular bases underpinning resistance to ERBB2 therapeutics in BC, discuss outstanding issues in the field of ERBB2 therapeutic targeting and elaborate on future directions of translational research on ERBB2+ breast cancer.
Collapse
Affiliation(s)
- Alessandra Fabi
- Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy
| | | | | |
Collapse
|
125
|
Ackerman ME, Alter G. Opportunities to exploit non-neutralizing HIV-specific antibody activity. Curr HIV Res 2014; 11:365-77. [PMID: 24191934 DOI: 10.2174/1570162x113116660058] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/16/2013] [Accepted: 08/03/2013] [Indexed: 12/22/2022]
Abstract
Antibodies act as a nexus between innate and adaptive immunity: they provide a means to engage a spectrum of innate immune effector cells in order to clear viral particles and infected cells and prime antigen presentation. This functional landscape is remarkably complex, and depends on antibody isotype, subclass, and glycosylation; the expression levels and patterns of a suite of Fc receptors with both complementary and opposing activities; and a host of innate immune cells capable of differential responses to opsonized particles and present at different sites. In vivo, even neutralizing antibodies rely on their ability to act as molecular beacons and recruit innate immune effector cells in order to provide protection, and results from both human and macaque studies have implicated these effector functions in vaccinemediated protection. Thus, while enhancing effector function is a tractable handle for potentiating antibody-mediated protection from HIV infection, success will depend critically on leveraging understanding of the means by which antibodies with specific functional profiles could be elicited, which effector functions could provide optimal protection, and perhaps most critically, how to efficiently recruit the innate effector cells present at sites of infection.
Collapse
Affiliation(s)
- Margaret E Ackerman
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, USA.
| | | |
Collapse
|
126
|
Veomett N, Dao T, Liu H, Xiang J, Pankov D, Dubrovsky L, Whitten JA, Park SM, Korontsvit T, Zakhaleva V, Casey E, Curcio M, Kharas MG, O'Reilly RJ, Liu C, Scheinberg DA. Therapeutic efficacy of an Fc-enhanced TCR-like antibody to the intracellular WT1 oncoprotein. Clin Cancer Res 2014; 20:4036-46. [PMID: 24850840 DOI: 10.1158/1078-0432.ccr-13-2756] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE RMFPNAPYL (RMF), a Wilms' tumor gene 1 (WT1)-derived CD8 T-cell epitope presented by HLA-A*02:01, is a validated target for T-cell-based immunotherapy. We previously reported ESK1, a high avidity (Kd < 0.2 nmol/L), fully-human monoclonal antibody (mAb) specific for the WT1 RMF peptide/HLA-A*02:01 complex, which selectively bound and killed WT1(+) and HLA-A*02:01(+) leukemia and solid tumor cell lines. EXPERIMENTAL DESIGN We engineered a second-generation mAb, ESKM, to have enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) function due to altered Fc glycosylation. ESKM was compared with native ESK1 in binding assays, in vitro ADCC assays, and mesothelioma and leukemia therapeutic models and pharmacokinetic studies in mice. ESKM toxicity was assessed in HLA-A*02:01(+) transgenic mice. RESULTS ESK antibodies mediated ADCC against hematopoietic and solid tumor cells at concentrations below 1 μg/mL, but ESKM was about 5- to 10-fold more potent in vitro against multiple cancer cell lines. ESKM was more potent in vivo against JMN mesothelioma, and effective against SET2 AML and fresh ALL xenografts. ESKM had a shortened half-life (4.9 days vs. 6.5 days), but an identical biodistribution pattern in C57BL/6J mice. At therapeutic doses of ESKM, there was no difference in half-life or biodistribution in HLA-A*02:01(+) transgenic mice compared with the parent strain. Importantly, therapeutic doses of ESKM in these mice caused no depletion of total WBCs or hematopoetic stem cells, or pathologic tissue damage. CONCLUSIONS The data provide proof of concept that an Fc-enhanced mAb can improve efficacy against a low-density, tumor-specific, peptide/MHC target, and support further development of this mAb against an important intracellular oncogenic protein.
Collapse
Affiliation(s)
- Nicholas Veomett
- Sloan Kettering Institute; Weill Cornell Medical College, New York, New York
| | | | - Hong Liu
- Eureka Therapeutics Inc., Emeryville, California; and
| | - Jingyi Xiang
- Eureka Therapeutics Inc., Emeryville, California; and
| | | | | | | | | | | | | | | | | | | | | | - Cheng Liu
- Eureka Therapeutics Inc., Emeryville, California; and
| | - David A Scheinberg
- Sloan Kettering Institute; Weill Cornell Medical College, New York, New York;
| |
Collapse
|
127
|
Fc glycan-modulated immunoglobulin G effector functions. J Clin Immunol 2014; 34 Suppl 1:S51-5. [PMID: 24760108 DOI: 10.1007/s10875-014-0018-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 03/19/2014] [Indexed: 10/25/2022]
Abstract
Immunoglobulin G (IgG) molecules are glycoproteins and residues in the sugar moiety attached to the IgG constant fragment (Fc) are essential for IgG functionality such as binding to cellular Fc receptors and complement activation. The core of this sugar moiety consists of a bi-antennary heptameric structure of mannose and N-acetylglucosamine (GlcNAc), further decorated with terminal and branching residues including galactose, sialic acid, fucose, and GlcNAc. Presence or absence of distinct residues such as fucose and sialic acid can dramatically alter pro- and anti-inflammatory IgG activities which could be harnessed for immunotherapeutic purposes. Here we review recent advances in understanding the role of the IgG-Fc glycan during immune responses and for immunotherapy with a focus on sialic acid and intravenous immunoglobulin (IVIG) treatment.
Collapse
|
128
|
Abstract
Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that is effective and generally well tolerated when administered as a single agent to treat advanced breast cancer. Efficacy has now been demonstrated in randomized trials as first line, second line, and later than the second line treatment of advanced breast cancer. T-DM1 is currently being evaluated as adjuvant treatment for early breast cancer. It has several mechanisms of action consisting of the anti-tumor effects of trastuzumab and those of DM1, a cytotoxic anti-microtubule agent released within the target cells upon degradation of the human epidermal growth factor receptor-2 (HER2)-T-DM1 complex in lysosomes. The cytotoxic effect of T-DM1 likely varies depending on the intracellular concentration of DM1 accumulated in cancer cells, high intracellular levels resulting in rapid apoptosis, somewhat lower levels in impaired cellular trafficking and mitotic catastrophe, while the lowest levels lead to poor response to T-DM1. Primary resistance of HER2-positive metastatic breast cancer to T-DM1 appears to be relatively infrequent, but most patients treated with T-DM1 develop acquired drug resistance. The mechanisms of resistance are incompletely understood, but mechanisms limiting the binding of trastuzumab to cancer cells may be involved. The cytotoxic effect of T-DM1 may be impaired by inefficient internalization or enhanced recycling of the HER2-T-DM1 complex in cancer cells, or impaired lysosomal degradation of trastuzumab or intracellular trafficking of HER2. The effect of T-DM1 may also be compromised by multidrug resistance proteins that pump DM1 out of cancer cells. In this review we discuss the mechanism of action of T-DM1 and the key clinical results obtained with it, the combinations of T-DM1 with other cytotoxic agents and anti-HER drugs, and the potential resistance mechanisms and the strategies to overcome resistance to T-DM1.
Collapse
|
129
|
Abstract
PURPOSE OF REVIEW Although a large number of novel broadly neutralizing antibodies has been recently described, the induction of such antibodies via vaccination has proven difficult. By contrast, nonneutralizing antibodies arise early during infection and have been repeatedly associated with both protection from infection and disease progression. RECENT FINDINGS We are beginning to gain new insights into the broader landscape of antiviral mechanisms that nonneutralizing antibodies may harness to fight HIV, providing an unprecedented breadth of approaches by which HIV can be blocked and contained. SUMMARY In this review, we summarize the characteristics of nonneutralizing antibodies, their role in HIV infection, and new paradigm-shifting functions that may be exploited by next-generation vaccine approaches aimed at blocking HIV infection.
Collapse
|
130
|
IgG-effector functions: "the good, the bad and the ugly". Immunol Lett 2014; 160:139-44. [PMID: 24495619 DOI: 10.1016/j.imlet.2014.01.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/15/2014] [Accepted: 01/24/2014] [Indexed: 01/06/2023]
Abstract
IgG-antibodies are potent and versatile mediators of host protection. They elicit their biological effects through specific interaction of the Fc-part with complement, specific cellular receptors, or both. Several factors should be taken into consideration when analyzing the nature and intensity of the immunological response elicited via IgG-effector functions, especially for the family of IgG-Fc receptors (FcγRs) exclusively expressed on immune cells. These include the various classes of leukocyte FcγR, expressed variably on different immune cells, each with distinct affinity for every IgG subclass, as well as genetic FcγR-polymorphisms affecting expression and affinity for IgG. Furthermore, various aspects of the IgG itself are also crucial for the outcome of the biological response. These include endogenously encoded IgG-polymorphisms, such as IgG3 polymorphisms, and post-transcriptional IgG-modifications, in particular IgG-Fc-glycosylation, affecting IgG effector functions through modified binding affinity to FcγR. These latter aspects concerning the variability in IgG3 on its half-life and placental transport and the clinical consequences of altered IgG-quality through glycosylation, will be the focus of this review.
Collapse
|
131
|
Abstract
Immunoglobulin G (IgG) formed during pregnancy against human platelet antigens (HPAs) of the fetus mediates fetal or neonatal alloimmune thrombocytopenia (FNAIT). Because antibody titer or isotype does not strictly correlate with disease severity, we investigated by mass spectrometry variations in the glycosylation at Asn297 in the IgG Fc because the composition of this glycan can be highly variable, affecting binding to phagocyte IgG-Fc receptors (FcγR). We found markedly decreased levels of core fucosylation of anti-HPA-1a-specific IgG1 from FNAIT patients (n = 48), but not in total serum IgG1. Antibodies with a low amount of fucose displayed higher binding affinity to FcγRIIIa and FcγRIIIb, but not to FcγRIIa, compared with antibodies with a high amount of Fc fucose. Consequently, these antibodies with a low amount of Fc fucose showed enhanced phagocytosis of platelets using FcγRIIIb(+) polymorphonuclear cells or FcγRIIIa(+) monocytes as effector cells, but not with FcγRIIIa(-) monocytes. In addition, the degree of anti-HPA-1a fucosylation correlated positively with the neonatal platelet counts in FNAIT, and negatively to the clinical disease severity. In contrast to the FNAIT patients, no changes in core fucosylation were observed for anti-HLA antibodies in refractory thrombocytopenia (post platelet transfusion), indicating that the level of fucosylation may be antigen dependent and/or related to the immune milieu defined by pregnancy.
Collapse
|
132
|
Seliger B, Kiessling R. The two sides of HER2/neu: immune escape versus surveillance. Trends Mol Med 2013; 19:677-84. [PMID: 24035606 DOI: 10.1016/j.molmed.2013.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 08/14/2013] [Accepted: 08/15/2013] [Indexed: 11/28/2022]
Abstract
The oncogene HER2 is one of the prototypes for targeted immunotherapy of cancer using both monoclonal antibodies as well as T cell based immunotherapies. Effective humoral and cellular immune responses against HER2 can be induced, but these responses can be influenced by the effects of this oncogene on the target tumor cells. The processes involved in HER2-mediated adaptive and innate immunity and the molecular mechanisms underlying the escape of HER2-expressing tumor cells from immune surveillance, particularly from cytotoxic T cells, are discussed. Implementing this knowledge in clinical trials to revert immune evasion may help optimize immunotherapies directed against HER2-expressing tumors.
Collapse
Affiliation(s)
- Barbara Seliger
- University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, 06112 Halle (Saale), Germany.
| | | |
Collapse
|
133
|
Lin J, Spidel JL, Maddage CJ, Rybinski KA, Kennedy RP, Krauthauser CLM, Park YC, Albone EF, Jacob S, Goserud MT, Martinez BP, Chao Q, Zhou Y, Nicolaides NC, Kline JB, Grasso L. The antitumor activity of the human FOLR1-specific monoclonal antibody, farletuzumab, in an ovarian cancer mouse model is mediated by antibody-dependent cellular cytotoxicity. Cancer Biol Ther 2013; 14:1032-8. [PMID: 24025360 DOI: 10.4161/cbt.26106] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Because of its high mortality rate, ovarian cancer is a leading cause of death among women and a highly unmet medical need. New therapeutic agents that are effective and well tolerated are needed and cancer antigen-specific monoclonal antibodies that have direct pharmacologic effects or can stimulate immunological responses represent a promising class of agents for the treatment of this disease. The human folate receptor α (FOLR1), which is overexpressed in ovarian cancer but largely absent in normal tissues, appears to play a role in the transformed phenotype in ovarian cancer, cisplatin sensitivity, and growth in depleted folate conditions and therefore has potential as a target for passive immunotherapy. The anti-FOLR1 monoclonal antibody MORAb-003 (farletuzumab) was previously shown to elicit antibody dependent cellular cytotoxicity (ADCC) and inhibit tumor growth of human tumor xenografts in nude mice. Because of its promising preclinical profile, farletuzumab has been evaluated in clinical trials as a potential therapeutic agent for ovarian cancer. In this report, we demonstrated that farletuzumab's antitumor effect against an experimental model of ovarian cancer is mediated by its ADCC activity.
Collapse
|
134
|
Bulliard Y, Jolicoeur R, Windman M, Rue SM, Ettenberg S, Knee DA, Wilson NS, Dranoff G, Brogdon JL. Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. ACTA ACUST UNITED AC 2013; 210:1685-93. [PMID: 23897982 PMCID: PMC3754864 DOI: 10.1084/jem.20130573] [Citation(s) in RCA: 369] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Antibodies that coengage activating FcγRs expressed by tumor-associated leukocytes facilitate the selective elimination of intratumoral T cells. Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.
Collapse
Affiliation(s)
- Yannick Bulliard
- Department of Oncology and 2 Laboratory Animal Services, Novartis Institute for Biomedical Research, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Kellner C, Derer S, Valerius T, Peipp M. Boosting ADCC and CDC activity by Fc engineering and evaluation of antibody effector functions. Methods 2013; 65:105-13. [PMID: 23851282 DOI: 10.1016/j.ymeth.2013.06.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/26/2013] [Accepted: 06/27/2013] [Indexed: 01/18/2023] Open
Abstract
In recent years, therapy with monoclonal antibodies has become standard of care in various clinical applications. Despite obvious clinical activity, not all patients respond and benefit from this generally well tolerated treatment option. Therefore, rational optimization of antibody therapy represents a major area of interest in translational research. Animal models and clinical data suggested important roles of Fc-mediated effector mechanisms such as antibody dependent cell-mediated cytotoxicity (ADCC) or complement dependent cytotoxicity (CDC) in antibody therapy. These novel insights into the mechanisms of action mediated by monoclonal antibodies inspired the development of different engineering approaches to enhance/optimize antibodies' effector functions. Fc-engineering approaches by altering the Fc-bound glycosylation profile or by exchanging amino acids in the protein backbone have been intensively studied. Here, advanced and emerging technologies in Fc-engineering resulting in altered ADCC and CDC activity are summarized and experimental strategies to evaluate antibodies' effector functions are discussed.
Collapse
Affiliation(s)
- Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Christian-Albrechts-University Kiel, Germany.
| |
Collapse
|
136
|
|
137
|
Grainger RK, James DC. CHO cell line specific prediction and control of recombinant monoclonal antibodyN-glycosylation. Biotechnol Bioeng 2013; 110:2970-83. [DOI: 10.1002/bit.24959] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/30/2013] [Accepted: 05/06/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Rhian K. Grainger
- ChELSI Institute, Department of Chemical and Biological Engineering; University of Sheffield; Mappin Street Sheffield S1 3JD UK
| | - David C. James
- ChELSI Institute, Department of Chemical and Biological Engineering; University of Sheffield; Mappin Street Sheffield S1 3JD UK
| |
Collapse
|
138
|
Zhao L, Liu M, Gao Y, Huang Y, Lu G, Gao Y, Guo X, She B. Glycosylation of sera thyroglobulin antibody in patients with thyroid diseases. Eur J Endocrinol 2013; 168:585-92. [PMID: 23360821 DOI: 10.1530/eje-12-0964] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Thyroglobulin antibody (TgAb) is an important autoantibody in thyroid diseases, which is a glycoprotein, predominantly of IgG class. Glycosylation of the IgG-Fc contributes to many effector functions exhibited by antibodies. The aim of our study was to investigate the glycosylation of sera TgAb in patients with different thyroid diseases. DESIGN AND METHODS Sera from 146 patients were collected and divided into four groups: Hashimoto's thyroiditis (HT, n=90), Graves' disease (GD, n=20), papillary thyroid carcinoma (PTC, n=17), and PTC with histological lymphocytic thyroiditis (PTC-T, n=19). HT patients were further divided into euthyroidism and subclinical and overt hypothyroidism groups. Lectin-ELISAs were performed to detect the relative amount of core fucose, terminal galactose, and sialic acid on each TgAb respectively. RESULTS Among HT, GD, and PTC groups, HT patients had significantly lower core fucose content on TgAb than the other two groups; an increasing trend of sialylation was found in PTC sera (P=0.076) compared with HT groups. PTC-T patients had significantly higher sialylated TgAb than HT and GD patients, and no significant difference was found between PTC and PTC-T. There was no significant difference in the three carbohydrate residue contents on sera TgAb among HT subgroups. In all the patients, negative correlation was found between sialic acid content and TgAb IgG levels (r=-0.736, P<0.001). CONCLUSIONS Our study showed that glycosylation of sera TgAb varied in different thyroid diseases and it might be involved in pathogenesis of thyroid disorders.
Collapse
Affiliation(s)
- Lanlan Zhao
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Evaluation of treatment response for breast cancer: are we entering the era of “biological complete remission”? Chin J Cancer Res 2013. [DOI: 10.1007/s11670-012-0281-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
140
|
|
141
|
Mimoto F, Igawa T, Kuramochi T, Katada H, Kadono S, Kamikawa T, Shida-Kawazoe M, Hattori K. Novel asymmetrically engineered antibody Fc variant with superior FcγR binding affinity and specificity compared with afucosylated Fc variant. MAbs 2013; 5:229-36. [PMID: 23406628 PMCID: PMC3893233 DOI: 10.4161/mabs.23452] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Fc engineering is a promising approach to enhance the antitumor efficacy of monoclonal antibodies (mAbs) through antibody-dependent cell-mediated cytotoxicity (ADCC). Glyco- and protein-Fc engineering have been employed to enhance FcγR binding and ADCC activity of mAbs; the drawbacks of previous approaches lie in their binding affinity to both FcγRIIIa allotypes, the ratio of activating FcγR binding to inhibitory FcγR binding (A/I ratio) or the melting temperature (TM) of the CH2 domain. To date, no engineered Fc variant has been reported that satisfies all these points. Herein, we present a novel Fc engineering approach that introduces different substitutions in each Fc domain asymmetrically, conferring optimal binding affinity to FcγR and specificity to the activating FcγR without impairing the stability. We successfully designed an asymmetric Fc variant with the highest binding affinity for both FcγRIIIa allotypes and the highest A/I ratio compared with previously reported symmetrically engineered Fc variants, and superior or at least comparable in vitro ADCC activity compared with afucosylated Fc variants. In addition, the asymmetric Fc engineering approach offered higher stability by minimizing the use of substitutions that reduce the TM of the CH2 domain compared with the symmetric approach. These results demonstrate that the asymmetric Fc engineering platform provides best-in-class effector function for therapeutic antibodies against tumor antigens.
Collapse
Affiliation(s)
- Futa Mimoto
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Bian L, Wang T, Liu Y, Zhang HQ, Song JJ, Zhang SH, Wu SK, Song ST, Jiang ZF. Evaluation of treatment response for breast cancer: are we entering the era of "biological complete remission"? Chin J Cancer Res 2013; 24:403-7. [PMID: 23359646 DOI: 10.3978/j.issn.1000-9604.2012.11.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/01/2012] [Indexed: 01/04/2023] Open
Abstract
Breast cancer is one of the most common malignancies in women. The post-operative recurrence and metastasis are the leading causes of breast cancer-related mortality. In this study, we tried to explore the role of circulating tumor cell (CTC) detection combination PET/CT technology evaluating the prognosis and treatment response of patients with breast cancer; meanwhile, we attempted to assess the concept of "biological complete remission" (bCR) in this regard. A 56-year-old patient with breast cancer (T(2)N(1)M(1), stage IV left breast cancer, with metastasis to axillary lymph nodes and lungs) received 6 cycles of salvage treatment with albumin-bound paclitaxel plus capecitabine and trastuzumab. Then, she underwent CTC detection and PET/CT for efficacy evaluation. CTC detection combination PET/CT is useful for the evaluation of the biological efficacy of therapies for breast cancer. The bCR of the patient appeared earlier than the conventional clinical imaging complete remission and promised the histological (pathological) complete remission. The integrated application of the concepts including bCR, imageological CR, and histological CR can achieve the early and accurate assessment of biological therapeutic reponse and prognosis of breast cancer.
Collapse
Affiliation(s)
- Li Bian
- Department of Breast Cancer, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Champion T, Beck A. Capture of the human IgG1 antibodies by protein A for the kinetic study of h-IgG/FcγR interaction using SPR-based biosensor technology. Methods Mol Biol 2013; 988:331-43. [PMID: 23475730 DOI: 10.1007/978-1-62703-327-5_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Surface plasmon resonance (SPR) is a key technology to evaluate IgG effector functions in vitro involving binding to Fcgamma receptors (FcγR). We describe here the chemical coupling of protein A to a sensor chip. IgGs prepared either from purified antibodies or from crude cell media supernatants are captured by the protein A and are used as the ligand. Soluble forms of FcγRs are injected at different concentrations and are used as the analyte. This setup allows the definition of the kinetic rates of binding of the FcγRI (high affinity) or the FcγRIIIa (low affinity) on the Fc domain of IgGs.
Collapse
Affiliation(s)
- Thierry Champion
- Physico-Chemistry Department, Centre d'Immunologie Pierre-Fabre, Saint Julien-en-Genevois, France
| | | |
Collapse
|
144
|
Marcucci F, Bellone M, Rumio C, Corti A. Approaches to improve tumor accumulation and interactions between monoclonal antibodies and immune cells. MAbs 2012; 5:34-46. [PMID: 23211740 DOI: 10.4161/mabs.22775] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Monoclonal antibodies (mAb) have become a mainstay in tumor therapy. Clinical responses to mAb therapy, however, are far from optimal, with many patients presenting native or acquired resistance or suboptimal responses to a mAb therapy. MAbs exert antitumor activity through different mechanisms of action and we propose here a classification of these mechanisms. In many cases mAbs need to interact with immune cells to exert antitumor activity. We summarize evidence showing that interactions between mAbs and immune cells may be inadequate for optimal antitumor activity. This may be due to insufficient tumor accumulation of mAbs or immune cells, or to low-affinity interactions between these components. The possibilities to improve tumor accumulation of mAbs and immune cells, and to improve the affinity of the interactions between these components are reviewed. We also discuss future directions of research that might further improve the therapeutic efficacy of antitumor mAbs.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Centro Nazionale di Epidemiologia, Sorveglianza e Promozione della Salute, Istituto Superiore di Sanita', Roma, Italy.
| | | | | | | |
Collapse
|
145
|
Gerdes CA, Nicolini VG, Herter S, van Puijenbroek E, Lang S, Roemmele M, Moessner E, Freytag O, Friess T, Ries CH, Bossenmaier B, Mueller HJ, Umaña P. GA201 (RG7160): A Novel, Humanized, Glycoengineered Anti-EGFR Antibody with Enhanced ADCC and Superior In Vivo Efficacy Compared with Cetuximab. Clin Cancer Res 2012. [DOI: 10.1158/1078-0432.ccr-12-0989] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
146
|
Loos A, Steinkellner H. IgG-Fc glycoengineering in non-mammalian expression hosts. Arch Biochem Biophys 2012; 526:167-73. [PMID: 22634260 PMCID: PMC3442181 DOI: 10.1016/j.abb.2012.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/15/2012] [Accepted: 05/16/2012] [Indexed: 12/28/2022]
Abstract
The remarkable success of therapeutic applications of immunoglobulin G (IgG) in form of monoclonal antibodies and pooled immunoglobulin G preparations has directed attention to this class of glycoproteins. It is commonly appreciated that oligosaccharides attached to the Fc-region play a critical role in the biological activity of IgGs. Thus, glycosylation has been a focus of interest for many scientists and the biopharmaceutical industry and expression hosts have been engineered in order to optimize antibody products. In this review we focus on efforts towards a targeted manipulation of IgG-Fc N-glycans using non-mammalian expression hosts, i.e. yeast, insect cells and plants. Current achievements in generating human-like N-glycan structures will be presented and recent data on the molecular mechanisms that might explain how these potent drugs mediate in vivo activities will be discussed.
Collapse
Affiliation(s)
| | - Herta Steinkellner
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| |
Collapse
|
147
|
Marcucci F, Corti A. Improving drug penetration to curb tumor drug resistance. Drug Discov Today 2012; 17:1139-46. [DOI: 10.1016/j.drudis.2012.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/24/2012] [Accepted: 06/08/2012] [Indexed: 12/21/2022]
|
148
|
Golay J, Introna M. Mechanism of action of therapeutic monoclonal antibodies: Promises and pitfalls of in vitro and in vivo assays. Arch Biochem Biophys 2012; 526:146-53. [DOI: 10.1016/j.abb.2012.02.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/14/2012] [Accepted: 02/16/2012] [Indexed: 02/08/2023]
|
149
|
Fuksa L, Micuda S, Grim J, Ryska A, Hornychova H. Predictive Biomarkers in Breast Cancer: Their Value in Neoadjuvant Chemotherapy. Cancer Invest 2012; 30:663-78. [DOI: 10.3109/07357907.2012.725441] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
150
|
Abstract
Amplification of the ERBB2 gene, which encodes human epidermal growth factor receptor 2 (HER2), causes the overexpression of a major proliferative driver for a subset of breast and gastric cancers. Treatments for patients with HER2-positive cancer include the monoclonal antibody trastuzumab and, in the case of metastatic breast cancer, the tyrosine kinase inhibitor lapatinib. Despite significant improvement in patient outcome as a result of these therapies, challenges remain. This Review focuses on proposed mechanisms of action and resistance in the context of potential new therapeutic options. Therapeutic approaches currently in development likely will yield additional clinically meaningful improvements for patients with HER2-positive cancer.
Collapse
Affiliation(s)
- Howard M Stern
- Department of Research Pathology, Genentech Inc., South San Francisco, CA 94080-4990, USA.
| |
Collapse
|