Next Issue
Volume 10, March
Previous Issue
Volume 9, September
 
 

Antibodies, Volume 9, Issue 4 (December 2020) – 22 articles

Cover Story (view full-size image): Recent years have seen great advances in our understanding of the mechanisms of action of anticancer antibodies. In particular, the importance of the antibody Fc region has become clear; leading to an array of new strategies to engineer antibodies with enhanced activity. Techniques such as mutagenesis and glycoengineering have been employed to manipulate interactions between antibody Fc and Fc gamma receptors, complements, and the neonatal Fc receptor, leading to altered function. With the rules of engagement depending on antibody target and context, having a large toolkit of engineering options allows for a modular ‘plug and play’ approach to effective drug development. These engineered antibodies are arriving in the clinic in growing numbers and are beginning to deliver patient benefits. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
10 pages, 1773 KiB  
Article
Selection and Characterization of Single-Domain Antibodies for Detection of Lassa Nucleoprotein
by George P. Anderson, Jinny L. Liu, Lisa C. Shriver-Lake and Ellen R. Goldman
Antibodies 2020, 9(4), 71; https://doi.org/10.3390/antib9040071 - 17 Dec 2020
Cited by 3 | Viewed by 3687
Abstract
Lassa virus is the etiologic agent of Lassa fever, an acute and often fatal illness endemic to West Africa. It is important to develop new reagents applicable either for the specific diagnosis or as improved therapeutics for the treatment of Lassa fever. Here, [...] Read more.
Lassa virus is the etiologic agent of Lassa fever, an acute and often fatal illness endemic to West Africa. It is important to develop new reagents applicable either for the specific diagnosis or as improved therapeutics for the treatment of Lassa fever. Here, we describe the development and initial testing of llama-derived single-domain antibodies that are specific for the Lassa virus nucleoprotein. Four sequence families based on complementarity-determining region (CDR) homology were identified by phage-based enzyme-linked immunosorbent assays, however, the highest affinity clones all belonged to the same sequence family which possess a second disulfide bond between Framework 2 and CDR3. The affinity and thermal stability were evaluated for each clone. A MagPlex-based homogeneous sandwich immunoassay for Lassa virus-like particles was also demonstrated to show their potential for further development as diagnostic reagents. Full article
(This article belongs to the Special Issue Design, Production and Characterization of Peptide Antibodies)
Show Figures

Figure 1

15 pages, 711 KiB  
Review
Fc Engineering Strategies to Advance IgA Antibodies as Therapeutic Agents
by Geert van Tetering, Mitchell Evers, Chilam Chan, Marjolein Stip and Jeanette Leusen
Antibodies 2020, 9(4), 70; https://doi.org/10.3390/antib9040070 - 15 Dec 2020
Cited by 28 | Viewed by 7692
Abstract
In the past three decades, a great interest has arisen in the use of immunoglobulins as therapeutic agents. In particular, since the approval of the first monoclonal antibody Rituximab for B cell malignancies, the progress in the antibody-related therapeutic agents has been incremental. [...] Read more.
In the past three decades, a great interest has arisen in the use of immunoglobulins as therapeutic agents. In particular, since the approval of the first monoclonal antibody Rituximab for B cell malignancies, the progress in the antibody-related therapeutic agents has been incremental. Therapeutic antibodies can be applied in a variety of diseases, ranging from cancer to autoimmunity and allergy. All current therapeutic monoclonal antibodies used in the clinic are of the IgG isotype. IgG antibodies can induce the killing of cancer cells by growth inhibition, apoptosis induction, complement activation (CDC) or antibody-dependent cellular cytotoxicity (ADCC) by NK cells, antibody-dependent cellular phagocytosis (ADCP) by monocytes/macrophages, or trogoptosis by granulocytes. To enhance these effector mechanisms of IgG, protein and glyco-engineering has been successfully applied. As an alternative to IgG, antibodies of the IgA isotype have been shown to be very effective in tumor eradication. Using the IgA-specific receptor FcαRI expressed on myeloid cells, IgA antibodies show superior tumor-killing compared to IgG when granulocytes are employed. However, reasons why IgA has not been introduced in the clinic yet can be found in the intrinsic properties of IgA posing several technical limitations: (1) IgA is challenging to produce and purify, (2) IgA shows a very heterogeneous glycosylation profile, and (3) IgA has a relatively short serum half-life. Next to the technical challenges, pre-clinical evaluation of IgA efficacy in vivo is not straightforward as mice do not naturally express the FcαR. Here, we provide a concise overview of the latest insights in these engineering strategies overcoming technical limitations of IgA as a therapeutic antibody: developability, heterogeneity, and short half-life. In addition, alternative approaches using IgA/IgG hybrid and FcαR-engagers and the impact of engineering on the clinical application of IgA will be discussed. Full article
(This article belongs to the Special Issue Antibody Engineering for Cancer Immunotherapy)
Show Figures

Figure 1

12 pages, 859 KiB  
Review
IgE in the Pathogenesis of SLE: From Pathogenic Role to Therapeutic Target
by Yasmine Lamri and Nicolas Charles
Antibodies 2020, 9(4), 69; https://doi.org/10.3390/antib9040069 - 08 Dec 2020
Cited by 7 | Viewed by 5400
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial chronic autoimmune disease, marked by the presence of autoantibodies to nuclear antigens belonging to different isotype classes. For several years, IgE antibodies have been incriminated in the development of allergic diseases and parasitic infections and different [...] Read more.
Systemic lupus erythematosus (SLE) is a multifactorial chronic autoimmune disease, marked by the presence of autoantibodies to nuclear antigens belonging to different isotype classes. For several years, IgE antibodies have been incriminated in the development of allergic diseases and parasitic infections and different anti-IgE therapies have been developed to encounter the pathogenic role of IgE in these pathologies. Recently, multiple studies showed the presence of elevated total IgE levels and demonstrated a pathogenic role of autoreactive IgE in SLE. This review aims to summarize the findings incriminating IgE and autoreactive IgE in the pathophysiology of SLE, to describe their functional outcomes on their targeted cells as well as to discuss different IgE-related therapeutic modalities that emerged and that may be beneficial for SLE patient care. Full article
(This article belongs to the Special Issue IgE in Autoimmunity)
Show Figures

Figure 1

14 pages, 1330 KiB  
Review
Complement Activation in the Treatment of B-Cell Malignancies
by Clive S. Zent, Jonathan J. Pinney, Charles C. Chu and Michael R. Elliott
Antibodies 2020, 9(4), 68; https://doi.org/10.3390/antib9040068 - 01 Dec 2020
Cited by 5 | Viewed by 4841
Abstract
Unconjugated monoclonal antibodies (mAb) have revolutionized the treatment of B-cell malignancies. These targeted drugs can activate innate immune cytotoxicity for therapeutic benefit. mAb activation of the complement cascade results in complement-dependent cytotoxicity (CDC) and complement receptor-mediated antibody-dependent cellular phagocytosis (cADCP). Clinical and laboratory [...] Read more.
Unconjugated monoclonal antibodies (mAb) have revolutionized the treatment of B-cell malignancies. These targeted drugs can activate innate immune cytotoxicity for therapeutic benefit. mAb activation of the complement cascade results in complement-dependent cytotoxicity (CDC) and complement receptor-mediated antibody-dependent cellular phagocytosis (cADCP). Clinical and laboratory studies have showed that CDC is therapeutically important. In contrast, the biological role and clinical effects of cADCP are less well understood. This review summarizes the available data on the role of complement activation in the treatment of mature B-cell malignancies and proposes future research directions that could be useful in optimizing the efficacy of this important class of drugs. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Figure 1

14 pages, 2235 KiB  
Review
The Role of Complement in Angiogenesis
by Maciej M. Markiewski, Elizabeth Daugherity, Britney Reese and Magdalena Karbowniczek
Antibodies 2020, 9(4), 67; https://doi.org/10.3390/antib9040067 - 01 Dec 2020
Cited by 15 | Viewed by 5522
Abstract
The link of the complement system to angiogenesis has remained circumstantial and speculative for several years. Perhaps the most clinically relevant example of possible involvement of complement in pathological neovascularization is age-related macular degeneration. Recent studies, however, provide more direct and experimental evidence [...] Read more.
The link of the complement system to angiogenesis has remained circumstantial and speculative for several years. Perhaps the most clinically relevant example of possible involvement of complement in pathological neovascularization is age-related macular degeneration. Recent studies, however, provide more direct and experimental evidence that indeed the complement system regulates physiological and pathological angiogenesis in models of wound healing, retinal regeneration, age-related macular degeneration, and cancer. Interestingly, complement-dependent mechanisms involved in angiogenesis are very much context dependent, including anti- and proangiogenic functions. Here, we discuss these new developments that place complement among other important regulators of homeostatic and pathological angiogenesis, and we provide the perspective on how these newly discovered complement functions can be targeted for therapy. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Figure 1

24 pages, 3752 KiB  
Article
Crystal Structure and Characterization of Human Heavy-Chain Only Antibodies Reveals a Novel, Stable Dimeric Structure Similar to Monoclonal Antibodies
by Carl Mieczkowski, Soheila Bahmanjah, Yao Yu, Jeanne Baker, Gopalan Raghunathan, Daniela Tomazela, Mark Hsieh, Mark McCoy, Corey Strickland and Laurence Fayadat-Dilman
Antibodies 2020, 9(4), 66; https://doi.org/10.3390/antib9040066 - 22 Nov 2020
Cited by 2 | Viewed by 4985
Abstract
We report the novel crystal structure and characterization of symmetrical, homodimeric humanized heavy-chain-only antibodies or dimers (HC2s). HC2s were found to be significantly coexpressed and secreted along with mAbs from transient CHO HC/LC cotransfection, resulting in an unacceptable mAb developability attribute. Expression of [...] Read more.
We report the novel crystal structure and characterization of symmetrical, homodimeric humanized heavy-chain-only antibodies or dimers (HC2s). HC2s were found to be significantly coexpressed and secreted along with mAbs from transient CHO HC/LC cotransfection, resulting in an unacceptable mAb developability attribute. Expression of full-length HC2s in the absence of LC followed by purification resulted in HC2s with high purity and thermal stability similar to conventional mAbs. The VH and CH1 portion of the heavy chain (or Fd) was also efficiently expressed and yielded a stable, covalent, and reducible dimer (Fd2). Mutagenesis of all heavy chain cysteines involved in disulfide bond formation revealed that Fd2 intermolecular disulfide formation was similar to Fabs and elucidated requirements for Fd2 folding and expression. For one HC2, we solved the crystal structure of the Fd2 domain to 2.9 Å, revealing a highly symmetrical homodimer that is structurally similar to Fabs and is mediated by conserved (CH1) and variable (VH) contacts with all CDRs positioned outward for target binding. Interfacial dimer contacts revealed by the crystal structure were mutated for two HC2s and were found to dramatically affect HC2 formation while maintaining mAb bioactivity, offering a potential means to modulate novel HC2 formation through engineering. These findings indicate that human heavy-chain dimers can be secreted efficiently in the absence of light chains, may show good physicochemical properties and stability, are structurally similar to Fabs, offer insights into their mechanism of formation, and may be amenable as a novel therapeutic modality. Full article
(This article belongs to the Special Issue Design, Production and Characterization of Peptide Antibodies)
Show Figures

Figure 1

17 pages, 1309 KiB  
Review
Discovery Strategies to Maximize the Clinical Potential of T-Cell Engaging Antibodies for the Treatment of Solid Tumors
by Vladimir Voynov, Paul J. Adam, Andrew E. Nixon and Justin M. Scheer
Antibodies 2020, 9(4), 65; https://doi.org/10.3390/antib9040065 - 18 Nov 2020
Cited by 13 | Viewed by 6621
Abstract
T-cell Engaging bispecific antibodies (TcEs) that can re-direct cytotoxic T-cells to kill cancer cells have been validated in clinical studies. To date, the clinical success with these agents has mainly been seen in hematologic tumor indications. However, an increasing number of TcEs are [...] Read more.
T-cell Engaging bispecific antibodies (TcEs) that can re-direct cytotoxic T-cells to kill cancer cells have been validated in clinical studies. To date, the clinical success with these agents has mainly been seen in hematologic tumor indications. However, an increasing number of TcEs are currently being developed to exploit the potent mode-of-action to treat solid tumor indications, which is more challenging in terms of tumor-cell accessibility and the complexity of the tumor microenvironment (TME). Of particular interest is the potential of TcEs as an immunotherapeutic approach for the treatment of non-immunogenic (often referred to as cold) tumors that do not respond to checkpoint inhibitors such as programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) antibodies. This has led to considerable discovery efforts for, firstly, the identification of tumor selective targeting approaches that can safely re-direct cytotoxic T-cells to cancer cells, and, secondly, bispecific antibodies and their derivatives with drug-like properties that promote a potent cytolytic synapse between T-cells and tumor cells, and in the most advanced TcEs, have IgG-like pharmacokinetics for dosing convenience. Based on encouraging pre-clinical data, a growing number of TcEs against a broad range of targets, and using an array of different molecular structures have entered clinical studies for solid tumor indications, and the first clinical data is beginning to emerge. This review outlines the different approaches that have been taken to date in addressing the challenges of exploiting the TcE mode-of-action for a broad range of solid indications, as well as opportunities for future discovery potential. Full article
(This article belongs to the Special Issue Monoclonal Antibody-Directed Therapy)
Show Figures

Figure 1

34 pages, 3590 KiB  
Review
Fc-Engineering for Modulated Effector Functions—Improving Antibodies for Cancer Treatment
by Rena Liu, Robert J. Oldham, Emma Teal, Stephen A. Beers and Mark S. Cragg
Antibodies 2020, 9(4), 64; https://doi.org/10.3390/antib9040064 - 17 Nov 2020
Cited by 93 | Viewed by 21390
Abstract
The majority of monoclonal antibody (mAb) therapeutics possess the ability to engage innate immune effectors through interactions mediated by their fragment crystallizable (Fc) domain. By delivering Fc-Fc gamma receptor (FcγR) and Fc-C1q interactions, mAb are able to link exquisite specificity to powerful cellular [...] Read more.
The majority of monoclonal antibody (mAb) therapeutics possess the ability to engage innate immune effectors through interactions mediated by their fragment crystallizable (Fc) domain. By delivering Fc-Fc gamma receptor (FcγR) and Fc-C1q interactions, mAb are able to link exquisite specificity to powerful cellular and complement-mediated effector functions. Fc interactions can also facilitate enhanced target clustering to evoke potent receptor signaling. These observations have driven decades-long research to delineate the properties within the Fc that elicit these various activities, identifying key amino acid residues and elucidating the important role of glycosylation. They have also fostered a growing interest in Fc-engineering whereby this knowledge is exploited to modulate Fc effector function to suit specific mechanisms of action and therapeutic purposes. In this review, we document the insight that has been generated through the study of the Fc domain; revealing the underpinning structure-function relationships and how the Fc has been engineered to produce an increasing number of antibodies that are appearing in the clinic with augmented abilities to treat cancer. Full article
(This article belongs to the Special Issue Antibody Engineering for Cancer Immunotherapy)
Show Figures

Graphical abstract

15 pages, 1663 KiB  
Article
Enhancing CDC and ADCC of CD19 Antibodies by Combining Fc Protein-Engineering with Fc Glyco-Engineering
by Sophia Roßkopf, Klara Marie Eichholz, Dorothee Winterberg, Katarina Julia Diemer, Sebastian Lutz, Ira Alexandra Münnich, Katja Klausz, Thies Rösner, Thomas Valerius, Denis Martin Schewe, Andreas Humpe, Martin Gramatzki, Matthias Peipp and Christian Kellner
Antibodies 2020, 9(4), 63; https://doi.org/10.3390/antib9040063 - 17 Nov 2020
Cited by 18 | Viewed by 5776
Abstract
Background: Native cluster of differentiation (CD) 19 targeting antibodies are poorly effective in triggering antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which are crucial effector functions of therapeutic antibodies in cancer immunotherapy. Both functions can be enhanced by engineering the antibody’s Fc [...] Read more.
Background: Native cluster of differentiation (CD) 19 targeting antibodies are poorly effective in triggering antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which are crucial effector functions of therapeutic antibodies in cancer immunotherapy. Both functions can be enhanced by engineering the antibody’s Fc region by altering the amino acid sequence (Fc protein-engineering) or the Fc-linked glycan (Fc glyco-engineering). We hypothesized that combining Fc glyco-engineering with Fc protein-engineering will rescue ADCC and CDC in CD19 antibodies. Results: Four versions of a CD19 antibody based on tafasitamab’s V-regions were generated: a native IgG1, an Fc protein-engineered version with amino acid exchanges S267E/H268F/S324T/G236A/I332E (EFTAE modification) to enhance CDC, and afucosylated, Fc glyco-engineered versions of both to promote ADCC. Irrespective of fucosylation, antibodies carrying the EFTAE modification had enhanced C1q binding and were superior in inducing CDC. In contrast, afucosylated versions exerted an enhanced affinity to Fcγ receptor IIIA and had increased ADCC activity. Of note, the double-engineered antibody harboring the EFTAE modification and lacking fucose triggered both CDC and ADCC more efficiently. Conclusions: Fc glyco-engineering and protein-engineering could be combined to enhance ADCC and CDC in CD19 antibodies and may allow the generation of antibodies with higher therapeutic efficacy by promoting two key functions simultaneously. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Figure 1

22 pages, 2151 KiB  
Article
Bringing the Heavy Chain to Light: Creating a Symmetric, Bivalent IgG-Like Bispecific
by Anusuya Ramasubramanian, Rachel Tennyson, Maureen Magnay, Sagar Kathuria, Tara Travaline, Annu Jain, Dana M. Lord, Megan Salemi, Caitlin Sullivan, Tristan Magnay, Jiali Hu, Eva Bric-Furlong, Pierrick Rival, Yanfeng Zhou, Dietmar Hoffmann, William Brondyk, Katarina Radošević and Partha S. Chowdhury
Antibodies 2020, 9(4), 62; https://doi.org/10.3390/antib9040062 - 06 Nov 2020
Cited by 3 | Viewed by 6733
Abstract
Bispecific molecules are biologically significant, yet their complex structures pose important manufacturing and pharmacokinetic challenges. Nevertheless, owing to similarities with monoclonal antibodies (mAbs), IgG-like bispecifics conceptually align well with conventional expression and manufacturing platforms and often exhibit potentially favorable drug metabolism and pharmacokinetic [...] Read more.
Bispecific molecules are biologically significant, yet their complex structures pose important manufacturing and pharmacokinetic challenges. Nevertheless, owing to similarities with monoclonal antibodies (mAbs), IgG-like bispecifics conceptually align well with conventional expression and manufacturing platforms and often exhibit potentially favorable drug metabolism and pharmacokinetic (DMPK) properties. However, IgG-like bispecifics do not possess target bivalency and current designs often require tedious engineering and purification to ensure appropriate chain pairing. Here, we present a near-native IgG antibody format, the 2xVH, which can create bivalency for each target or epitope and requires no engineering for cognate chain pairing. In this modality, two different variable heavy (VH) domains with distinct binding specificities are grafted onto the first constant heavy (CH1) and constant light (CL) domains, conferring the molecule with dual specificity. To determine the versatility of this format, we characterized the expression, binding, and stability of several previously identified soluble human VH domains. By grafting these domains onto an IgG scaffold, we generated several prototype 2xVH IgG and Fab molecules that display similar properties to mAbs. These molecules avoided the post-expression purification necessary for engineered bispecifics while maintaining a capacity for simultaneous dual binding. Hence, the 2xVH format represents a bivalent, bispecific design that addresses limitations of manufacturing IgG-like bispecifics while promoting biologically-relevant dual target engagement. Full article
(This article belongs to the Special Issue Design, Production and Characterization of Peptide Antibodies)
Show Figures

Graphical abstract

16 pages, 866 KiB  
Review
Complement and Cancer—A Dysfunctional Relationship?
by Joshua M. Thurman, Jennifer Laskowski and Raphael A. Nemenoff
Antibodies 2020, 9(4), 61; https://doi.org/10.3390/antib9040061 - 05 Nov 2020
Cited by 10 | Viewed by 4860
Abstract
Although it was long believed that the complement system helps the body to identify and remove transformed cells, it is now clear that complement activation contributes to carcinogenesis and can also help tumors to escape immune-elimination. Complement is activated by several different mechanisms [...] Read more.
Although it was long believed that the complement system helps the body to identify and remove transformed cells, it is now clear that complement activation contributes to carcinogenesis and can also help tumors to escape immune-elimination. Complement is activated by several different mechanisms in various types of cancer, and complement activation fragments have multiple different downstream effects on cancer cells and throughout the tumor microenvironment. Thus, the role of complement activation in tumor biology may vary among different types of cancer and over time within a single tumor. In multiple different pre-clinical models, however, complement activation has been shown to recruit immunosuppressive myeloid cells into the tumor microenvironment. These cells, in turn, suppress anti-tumor T cell immunity, enabling the tumor to grow. Based on extensive pre-clinical work, therapeutic complement inhibitors hold great promise as a new class of immunotherapy. A greater understanding of the role of complement in tumor biology will improve our ability to identify those patients most likely to benefit from this treatment and to rationally combine complement inhibitors with other cancer therapies. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Figure 1

19 pages, 774 KiB  
Review
Intravenous Immune Globulin Uses in the Fetus and Neonate: A Review
by Mahdi Alsaleem
Antibodies 2020, 9(4), 60; https://doi.org/10.3390/antib9040060 - 04 Nov 2020
Cited by 18 | Viewed by 10109
Abstract
Intravenous immune globulin (IVIG) is made after processing plasma from healthy donors. It is composed mainly of pooled immunoglobulin and has clinical evidence-based applications in adult and pediatric populations. Recently, several clinical applications have been proposed for managing conditions in the neonatal population, [...] Read more.
Intravenous immune globulin (IVIG) is made after processing plasma from healthy donors. It is composed mainly of pooled immunoglobulin and has clinical evidence-based applications in adult and pediatric populations. Recently, several clinical applications have been proposed for managing conditions in the neonatal population, such as hemolytic disease of the newborn, treatment, and prophylaxis for sepsis in high-risk neonates, enterovirus parvovirus and COVID-19 related neonatal infections, fetal and neonatal immune-induced thrombocytopenia, neonatal hemochromatosis, neonatal Kawasaki disease, and some types of immunodeficiency. The dosing, mechanism of action, effectiveness, side effects, and adverse reactions of IVIG have been relatively well studied in adults but are not well described in the neonatal population. This review aims to provide the most recent evidence and consensus guidelines about the use of IVIG in the fetus and neonate. Full article
Show Figures

Graphical abstract

14 pages, 8532 KiB  
Review
CD46 and Oncologic Interactions: Friendly Fire against Cancer
by Michelle Elvington, M. Kathryn Liszewski and John P. Atkinson
Antibodies 2020, 9(4), 59; https://doi.org/10.3390/antib9040059 - 02 Nov 2020
Cited by 23 | Viewed by 5904
Abstract
One of the most challenging aspects of cancer therapeutics is target selection. Recently, CD46 (membrane cofactor protein; MCP) has emerged as a key player in both malignant transformation as well as in cancer treatments. Normally a regulator of complement activation, CD46 is co-expressed [...] Read more.
One of the most challenging aspects of cancer therapeutics is target selection. Recently, CD46 (membrane cofactor protein; MCP) has emerged as a key player in both malignant transformation as well as in cancer treatments. Normally a regulator of complement activation, CD46 is co-expressed as four predominant isoforms on almost all cell types. CD46 is highly overexpressed on a variety of human tumor cells. Clinical and experimental data support an association between increased CD46 expression and malignant transformation and metastasizing potential. Further, CD46 is a newly discovered driver of metabolic processes and plays a role in the intracellular complement system (complosome). CD46 is also known as a pathogen magnet due to its role as a receptor for numerous microbes, including several species of measles virus and adenoviruses. Strains of these two viruses have been exploited as vectors for the therapeutic development of oncolytic agents targeting CD46. In addition, monoclonal antibody-drug conjugates against CD46 also are being clinically evaluated. As a result, there are multiple early-phase clinical trials targeting CD46 to treat a variety of cancers. Here, we review CD46 relative to these oncologic connections. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Graphical abstract

23 pages, 1168 KiB  
Review
The Role of Complement in the Mechanism of Action of Therapeutic Anti-Cancer mAbs
by Josée Golay and Ronald P. Taylor
Antibodies 2020, 9(4), 58; https://doi.org/10.3390/antib9040058 - 28 Oct 2020
Cited by 39 | Viewed by 8516
Abstract
Unconjugated anti-cancer IgG1 monoclonal antibodies (mAbs) activate antibody-dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells and antibody-dependent cellular phagocytosis (ADCP) by macrophages, and these activities are thought to be important mechanisms of action for many of these mAbs in vivo. Several mAbs [...] Read more.
Unconjugated anti-cancer IgG1 monoclonal antibodies (mAbs) activate antibody-dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells and antibody-dependent cellular phagocytosis (ADCP) by macrophages, and these activities are thought to be important mechanisms of action for many of these mAbs in vivo. Several mAbs also activate the classical complement pathway and promote complement-dependent cytotoxicity (CDC), although with very different levels of efficacy, depending on the mAb, the target antigen, and the tumor type. Recent studies have unraveled the various structural factors that define why some IgG1 mAbs are strong mediators of CDC, whereas others are not. The role of complement activation and membrane inhibitors expressed by tumor cells, most notably CD55 and CD59, has also been quite extensively studied, but how much these affect the resistance of tumors in vivo to IgG1 therapeutic mAbs still remains incompletely understood. Recent studies have demonstrated that complement activation has multiple effects beyond target cell lysis, affecting both innate and adaptive immunity mediated by soluble complement fragments, such as C3a and C5a, and by stimulating complement receptors expressed by immune cells, including NK cells, neutrophils, macrophages, T cells, and dendritic cells. Complement activation can enhance ADCC and ADCP and may contribute to the vaccine effect of mAbs. These different aspects of complement are also briefly reviewed in the specific context of FDA-approved therapeutic anti-cancer IgG1 mAbs. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Graphical abstract

21 pages, 2984 KiB  
Review
Complement System: Promoter or Suppressor of Cancer Progression?
by Margot Revel, Marie V. Daugan, Catherine Sautés-Fridman, Wolf H. Fridman and Lubka T. Roumenina
Antibodies 2020, 9(4), 57; https://doi.org/10.3390/antib9040057 - 25 Oct 2020
Cited by 51 | Viewed by 7757
Abstract
Constituent of innate immunity, complement is present in the tumor microenvironment. The functions of complement include clearance of pathogens and maintenance of homeostasis, and as such could contribute to an anti-tumoral role in the context of certain cancers. However, multiple lines of evidence [...] Read more.
Constituent of innate immunity, complement is present in the tumor microenvironment. The functions of complement include clearance of pathogens and maintenance of homeostasis, and as such could contribute to an anti-tumoral role in the context of certain cancers. However, multiple lines of evidence show that in many cancers, complement has pro-tumoral actions. The large number of complement molecules (over 30), the diversity of their functions (related or not to the complement cascade), and the variety of cancer types make the complement-cancer topic a very complex matter that has just started to be unraveled. With this review we highlight the context-dependent role of complement in cancer. Recent studies revealed that depending of the cancer type, complement can be pro or anti-tumoral and, even for the same type of cancer, different models presented opposite effects. We aim to clarify the current knowledge of the role of complement in human cancers and the insights from mouse models. Using our classification of human cancers based on the prognostic impact of the overexpression of complement genes, we emphasize the strong potential for therapeutic targeting the complement system in selected subgroups of cancer patients. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Graphical abstract

8 pages, 232 KiB  
Communication
High Seroprevalence of Antibodies against Arboviruses among Pregnant Women in Rural Caribbean Colombia in the Context of the Zika Virus Epidemic
by Elena Marbán-Castro, Germán J. Arrieta, Miguel J. Martínez, Raquel González, Azucena Bardají, Clara Menéndez and Salim Mattar
Antibodies 2020, 9(4), 56; https://doi.org/10.3390/antib9040056 - 21 Oct 2020
Cited by 6 | Viewed by 3511
Abstract
Mosquito-borne viruses such as dengue (DENV), chikungunya (CHIKV), and Zika (ZIKV) have spread in recent decades. We aimed to assess seroprevalence of arboviral infections in pregnant women living in Cereté, Caribbean Colombia. In 2016 a cross-sectional facility-based sero-survey study was performed among pregnant [...] Read more.
Mosquito-borne viruses such as dengue (DENV), chikungunya (CHIKV), and Zika (ZIKV) have spread in recent decades. We aimed to assess seroprevalence of arboviral infections in pregnant women living in Cereté, Caribbean Colombia. In 2016 a cross-sectional facility-based sero-survey study was performed among pregnant women (N = 90). Most of them (66%) reported at least one symptom or sign compatible with arboviral infection over the previous 15 days. All screened women had a positive IgG for DENV, 89% for ZIKV, and 82% for CHIKV. One woman tested positive for ZIKV IgM. This study shows the high exposure among pregnant women to arboviruses in endemic areas, shown by the high seroprevalence of past arboviral infections. Given the evidence on the potential risks of these arboviral infections on pregnancy and infant outcomes, these results highlight the need for continuous epidemiological surveillance of arboviral diseases, particularly among those most of risk of their harmful consequences. Full article
27 pages, 2734 KiB  
Review
IgE Antibodies against Cancer: Efficacy and Safety
by Jitesh Chauhan, Alex J. McCraw, Mano Nakamura, Gabriel Osborn, Heng Sheng Sow, Vivienne F. Cox, Chara Stavraka, Debra H. Josephs, James F. Spicer, Sophia N. Karagiannis and Heather J. Bax
Antibodies 2020, 9(4), 55; https://doi.org/10.3390/antib9040055 - 16 Oct 2020
Cited by 18 | Viewed by 9100
Abstract
Immunoglobulin E (IgE) antibodies are well known for their role in allergic diseases and for contributions to antiparasitic immune responses. Properties of this antibody class that mediate powerful effector functions may be redirected for the treatment of solid tumours. This has led to [...] Read more.
Immunoglobulin E (IgE) antibodies are well known for their role in allergic diseases and for contributions to antiparasitic immune responses. Properties of this antibody class that mediate powerful effector functions may be redirected for the treatment of solid tumours. This has led to the rise of a new class of therapeutic antibodies to complement the armamentarium of approved tumour targeting antibodies, which to date are all IgG class. The perceived risk of type I hypersensitivity reactions following administration of IgE has necessitated particular consideration in the development of these therapeutic agents. Here, we bring together the properties of IgE antibodies pivotal to the hypothesis for superior antitumour activity compared to IgG, observations of in vitro and in vivo efficacy and mechanisms of action, and a focus on the safety considerations for this novel class of therapeutic agent. These include in vitro studies of potential hypersensitivity, selection of and observations from appropriate in vivo animal models and possible implications of the high degree of glycosylation of IgE. We also discuss the use of ex vivo predictive and monitoring clinical tools, as well as the risk mitigation steps employed in, and the preliminary outcomes from, the first-in-human clinical trial of a candidate anticancer IgE therapeutic. Full article
(This article belongs to the Special Issue Antibody Engineering for Cancer Immunotherapy)
Show Figures

Graphical abstract

18 pages, 2886 KiB  
Article
Natural Killer (NK) Cell Expression of CD2 as a Predictor of Serial Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC)
by Jennifer J.-J. Tang, Alexander P. Sung, Michael J. Guglielmo, Lydia Navarrete-Galvan, Doug Redelman, Julie Smith-Gagen and Dorothy Hudig
Antibodies 2020, 9(4), 54; https://doi.org/10.3390/antib9040054 - 16 Oct 2020
Cited by 2 | Viewed by 4796
Abstract
NK cell ADCC supports monoclonal antibody anti-tumor therapies. We investigated serial ADCC and whether it could be predicted by NK phenotypes, including expression of CD16A, CD2 and perforin. CD16A, the NK receptor for antibodies, has AA158 valine or phenylalanine variants with different affinities [...] Read more.
NK cell ADCC supports monoclonal antibody anti-tumor therapies. We investigated serial ADCC and whether it could be predicted by NK phenotypes, including expression of CD16A, CD2 and perforin. CD16A, the NK receptor for antibodies, has AA158 valine or phenylalanine variants with different affinities for IgG. CD2, a costimulatory protein, associates with CD16A and can augment CD16A-signaling. Pore-forming perforin is essential for rapid NK-mediated killing. NK cells were monitored for their ADCC serial killing frequency (KF). KF is the average number of target cells killed per cell by a cytotoxic cell population. KF comparisons were made at 1:4 CD16pos NK effector:target ratios. ADCC was toward Daudi cells labeled with 51Cr and obinutuzumab anti-CD20 antibody. CD16A genotypes were determined by DNA sequencing. CD2, CD16A, and perforin expression was monitored by flow cytometry. Serial killing KFs varied two-fold among 24 donors and were independent of CD16A genotypes and perforin levels. However, high percentages of CD2pos of the CD16Apos NK cells and high levels of CD16A were associated with high KFs. ROC analysis indicated that the %CD2pos of CD16Apos NK cells can predict KFs. In conclusion, the extent of serial ADCC varies significantly among donors and appears predictable by the CD2posCD16Apos NK phenotype. Full article
Show Figures

Graphical abstract

35 pages, 5945 KiB  
Review
Structure, Function, and Therapeutic Use of IgM Antibodies
by Bruce A. Keyt, Ramesh Baliga, Angus M. Sinclair, Stephen F. Carroll and Marvin S. Peterson
Antibodies 2020, 9(4), 53; https://doi.org/10.3390/antib9040053 - 13 Oct 2020
Cited by 100 | Viewed by 20368
Abstract
Natural immunoglobulin M (IgM) antibodies are pentameric or hexameric macro-immunoglobulins and have been highly conserved during evolution. IgMs are initially expressed during B cell ontogeny and are the first antibodies secreted following exposure to foreign antigens. The IgM multimer has either 10 (pentamer) [...] Read more.
Natural immunoglobulin M (IgM) antibodies are pentameric or hexameric macro-immunoglobulins and have been highly conserved during evolution. IgMs are initially expressed during B cell ontogeny and are the first antibodies secreted following exposure to foreign antigens. The IgM multimer has either 10 (pentamer) or 12 (hexamer) antigen binding domains consisting of paired µ heavy chains with four constant domains, each with a single variable domain, paired with a corresponding light chain. Although the antigen binding affinities of natural IgM antibodies are typically lower than IgG, their polyvalency allows for high avidity binding and efficient engagement of complement to induce complement-dependent cell lysis. The high avidity of IgM antibodies renders them particularly efficient at binding antigens present at low levels, and non-protein antigens, for example, carbohydrates or lipids present on microbial surfaces. Pentameric IgM antibodies also contain a joining (J) chain that stabilizes the pentameric structure and enables binding to several receptors. One such receptor, the polymeric immunoglobulin receptor (pIgR), is responsible for transcytosis from the vasculature to the mucosal surfaces of the lung and gastrointestinal tract. Several naturally occurring IgM antibodies have been explored as therapeutics in clinical trials, and a new class of molecules, engineered IgM antibodies with enhanced binding and/or additional functional properties are being evaluated in humans. Here, we review the considerable progress that has been made regarding the understanding of biology, structure, function, manufacturing, and therapeutic potential of IgM antibodies since their discovery more than 80 years ago. Full article
(This article belongs to the Special Issue Structure and Function of Antibodies)
Show Figures

Figure 1

17 pages, 2661 KiB  
Review
Structural Features and PF4 Functions that Occur in Heparin-Induced Thrombocytopenia (HIT) Complicated by COVID-19
by Zheng Cai, Mark I. Greene, Zhiqiang Zhu and Hongtao Zhang
Antibodies 2020, 9(4), 52; https://doi.org/10.3390/antib9040052 - 10 Oct 2020
Cited by 26 | Viewed by 7068
Abstract
Platelet factor 4 (PF4, CXCL4) is a small chemokine protein released by activated platelets. Although a major physiological function of PF4 is to promote blood coagulation, this cytokine is involved in innate and adaptive immunity in events when platelets are activated in response [...] Read more.
Platelet factor 4 (PF4, CXCL4) is a small chemokine protein released by activated platelets. Although a major physiological function of PF4 is to promote blood coagulation, this cytokine is involved in innate and adaptive immunity in events when platelets are activated in response to infections. Coronavirus disease 2019 (COVID-19) patients have abnormal coagulation activities, and severe patients develop higher D-dimer levels. D-dimers are small protein products present in the blood after blood clots are degraded by fibrinolysis. To prevent clotting, heparin is often clinically used in COVID-19 patients. Some clinical procedures for the management of COVID-19 patients may include extracorporeal membrane oxygenation (ECMO) and renal replacement therapy (CRRT), which also require the use of heparin. Anti-PF4 antibodies are frequently detected in severe patients and heparin-induced thrombocytopenia (HIT) can also be observed. PF4 and its role in HIT as well as in pathologies seen in COVID-19 patients define a potential therapeutic option of using blocking antibodies in the treatment of COVID-19. Full article
Show Figures

Figure 1

12 pages, 6146 KiB  
Article
Anti gC1qR/p32/HABP1 Antibody Therapy Decreases Tumor Growth in an Orthotopic Murine Xenotransplant Model of Triple Negative Breast Cancer
by Ellinor I. Peerschke, Elisa de Stanchina, Qing Chang, Katia Manova-Todorova, Afsar Barlas, Anne G. Savitt, Brian V. Geisbrecht and Berhane Ghebrehiwet
Antibodies 2020, 9(4), 51; https://doi.org/10.3390/antib9040051 - 06 Oct 2020
Cited by 6 | Viewed by 4029
Abstract
gC1qR is highly expressed in breast cancer and plays a role in cancer cell proliferation. This study explored therapy with gC1qR monoclonal antibody 60.11, directed against the C1q binding domain of gC1qR, in a murine orthotopic xenotransplant model of triple negative breast cancer. [...] Read more.
gC1qR is highly expressed in breast cancer and plays a role in cancer cell proliferation. This study explored therapy with gC1qR monoclonal antibody 60.11, directed against the C1q binding domain of gC1qR, in a murine orthotopic xenotransplant model of triple negative breast cancer. MDA231 breast cancer cells were injected into the mammary fat pad of athymic nu/nu female mice. Mice were segregated into three groups (n = 5, each) and treated with the vehicle (group 1) or gC1qR antibody 60.11 (100 mg/kg) twice weekly, starting at day 3 post-implantation (group 2) or when the tumor volume reached 100 mm3 (group 3). At study termination (d = 35), the average tumor volume in the control group measured 895 ± 143 mm3, compared to 401 ± 48 mm3 and 701 ± 100 mm3 in groups 2 and 3, respectively (p < 0.05). Immunohistochemical staining of excised tumors revealed increased apoptosis (caspase 3 and TUNEL staining) in 60.11-treated mice compared to controls, and decreased angiogenesis (CD31 staining). Slightly decreased white blood cell counts were noted in 60.11-treated mice. Otherwise, no overt toxicities were observed. These data are the first to demonstrate an in vivo anti-tumor effect of 60.11 therapy in a mouse model of triple negative breast cancer. Full article
(This article belongs to the Special Issue The Role of Complement in Cancer Immunotherapy)
Show Figures

Graphical abstract

14 pages, 3472 KiB  
Article
Taking the Hinge off: An Approach to Effector-Less Monoclonal Antibodies
by Jamie Valeich, Dan Boyd, Manu Kanwar, Daniel Stenzel, Deblina De Ghosh, Arpa Ebrahimi, James Woo, Jenny Wang and Alexandre Ambrogelly
Antibodies 2020, 9(4), 50; https://doi.org/10.3390/antib9040050 - 23 Sep 2020
Cited by 4 | Viewed by 6024
Abstract
A variety of Fc domain engineering approaches for abrogating the effector functions of mAbs exists. To address some of the limitations of the current Fc domain silencing approaches, we are exploring a less commonly considered option which relies on the deletion of the [...] Read more.
A variety of Fc domain engineering approaches for abrogating the effector functions of mAbs exists. To address some of the limitations of the current Fc domain silencing approaches, we are exploring a less commonly considered option which relies on the deletion of the hinge. Removal of the hinge domain in humanized IgG1 and IgG4 mAbs obliterates their ability to bind to activating human Fc gamma receptors I and IIIA, while leaving their ability to engage their target antigen intact. Deletion of the hinge also reduces binding to the Fc neonatal receptor, although Fc engineering allows partial recovery of affinity. Engineering of the CH3 domain, stabilizes hinge deleted IgG4s and prevents Fab arm exchange. The faster clearing properties together with the pacified Fc make modality of the hinge deleted mAb an appealing solution for therapeutic and diagnostic applications. Full article
(This article belongs to the Special Issue New, Old, and Shared Antibody Specificities in Autoimmune Diseases)
Show Figures

Graphical abstract

Previous Issue
Next Issue
Back to TopTop