25
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Research Progress and Applications of Multivalent, Multispecific and Modified Nanobodies for Disease Treatment

      systematic-review

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Recombinant antibodies such as nanobodies are progressively demonstrating to be a valid alternative to conventional monoclonal antibodies also for clinical applications. Furthermore, they do not solely represent a substitute for monoclonal antibodies but their unique features allow expanding the applications of biotherapeutics and changes the pattern of disease treatment. Nanobodies possess the double advantage of being small and simple to engineer. This combination has promoted extremely diversified approaches to design nanobody-based constructs suitable for particular applications. Both the format geometry possibilities and the functionalization strategies have been widely explored to provide macromolecules with better efficacy with respect to single nanobodies or their combination. Nanobody multimers and nanobody-derived reagents were developed to image and contrast several cancer diseases and have shown their effectiveness in animal models. Their capacity to block more independent signaling pathways simultaneously is considered a critical advantage to avoid tumor resistance, whereas the mass of these multimeric compounds still remains significantly smaller than that of an IgG, enabling deeper penetration in solid tumors. When applied to CAR-T cell therapy, nanobodies can effectively improve the specificity by targeting multiple epitopes and consequently reduce the side effects. This represents a great potential in treating malignant lymphomas, acute myeloid leukemia, acute lymphoblastic leukemia, multiple myeloma and solid tumors. Apart from cancer treatment, multispecific drugs and imaging reagents built with nanobody blocks have demonstrated their value also for detecting and tackling neurodegenerative, autoimmune, metabolic, and infectious diseases and as antidotes for toxins. In particular, multi-paratopic nanobody-based constructs have been developed recently as drugs for passive immunization against SARS-CoV-2 with the goal of impairing variant survival due to resistance to antibodies targeting single epitopes. Given the enormous research activity in the field, it can be expected that more and more multimeric nanobody molecules will undergo late clinical trials in the next future.

          Systematic Review Registration

          Related collections

          Most cited references208

          • Record: found
          • Abstract: found
          • Article: not found

          Bispecific antibodies: a mechanistic review of the pipeline

          The term bispecific antibody (bsAb) is used to describe a large family of molecules designed to recognize two different epitopes or antigens. BsAbs come in many formats, ranging from relatively small proteins, merely consisting of two linked antigen-binding fragments, to large immunoglobulin G (IgG)-like molecules with additional domains attached. An attractive bsAb feature is their potential for novel functionalities - that is, activities that do not exist in mixtures of the parental or reference antibodies. In these so-called obligate bsAbs, the physical linkage of the two binding specificities creates a dependency that can be temporal, with binding events occurring sequentially, or spatial, with binding events occurring simultaneously, such as in linking an effector to a target cell. To date, more than 20 different commercialized technology platforms are available for bsAb creation and development, 2 bsAbs are marketed and over 85 are in clinical development. Here, we review the current bsAb landscape from a mechanistic perspective, including a comprehensive overview of the pipeline.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial.

            We did a randomised controlled trial to assess the benefit of maintenance infliximab therapy in patients with active Crohn's disease who respond to a single infusion of infliximab. 573 patients with a score of at least 220 on the Crohn's disease activity index (CDAI) received a 5 mg/kg intravenous infusion of infliximab at week 0. After assessment of response at week 2, patients were randomly assigned repeat infusions of placebo at weeks 2 and 6 and then every 8 weeks thereafter until week 46 (group I), repeat infusions of 5 mg/kg infliximab at the same timepoints (group II), or 5 mg/kg infliximab at weeks 2 and 6 followed by 10 mg/kg (group III). The prespecified co-primary endpoints were the proportion of patients who responded at week 2 and were in remission (CDAI 54) and more than 54 weeks (21 to >54) for groups II and III, respectively, compared with 19 weeks (10-45) for group I (p=0.002 and p=0.0002, respectively). Infliximab safety was consistent with that seen in other trials of infliximab in Crohn's disease and rheumatoid arthritis. In particular, the incidence of serious infections was similar across treatment groups. Patients with Crohn's disease who respond to an initial dose of infliximab are more likely to be in remission at weeks 30 and 54, to discontinue corticosteroids, and to maintain their response for a longer period of time, if infliximab treatment is maintained every 8 weeks.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2

              The SARS-CoV-2 virus is more transmissible than previous coronaviruses and causes a more serious illness than influenza. The SARS-CoV-2 receptor binding domain (RBD) of the spike protein binds to the human angiotensin-converting enzyme 2 (ACE2) receptor as a prelude to viral entry into the cell. Using a naive llama single-domain antibody library and PCR-based maturation, we have produced two closely related nanobodies, H11-D4 and H11-H4, that bind RBD (KD of 39 and 12 nM, respectively) and block its interaction with ACE2. Single-particle cryo-EM revealed that both nanobodies bind to all three RBDs in the spike trimer. Crystal structures of each nanobody-RBD complex revealed how both nanobodies recognize the same epitope, which partly overlaps with the ACE2 binding surface, explaining the blocking of the RBD-ACE2 interaction. Nanobody-Fc fusions showed neutralizing activity against SARS-CoV-2 (4-6 nM for H11-H4, 18 nM for H11-D4) and additive neutralization with the SARS-CoV-1/2 antibody CR3022.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                18 January 2022
                2021
                : 12
                : 838082
                Affiliations
                [1] 1 Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University , Tianjin, China
                [2] 2 Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin, China
                [3] 3 Institute of Shaoxing, Tianjin University , Zhejiang, China
                [4] 4 Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University , Tianjin, China
                [5] 5 Laboratory for Environmental and Life Sciences, University of Nova Gorica , Nova Gorica, Slovenia
                Author notes

                Edited by: Yong-Sung Kim, Ajou University, South Korea

                Reviewed by: Sang Taek Jung, Korea University, South Korea; Serge Muyldermans, Vrije University Brussel, Belgium

                *Correspondence: Ario de Marco, ario.demarco@ 123456ung.si ; He Huang, huang@ 123456tju.edu.cn

                ‡These authors have contributed equally to this work

                This article was submitted to Vaccines and Molecular Therapeutics, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2021.838082
                8804282
                35116045
                f54de489-31a7-4abb-b403-54525be3cda0
                Copyright © 2022 Wang, Kang, Yuan, Cao, Huang and de Marco

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 17 December 2021
                : 30 December 2021
                Page count
                Figures: 3, Tables: 5, Equations: 0, References: 208, Pages: 19, Words: 8905
                Funding
                Funded by: National Key Research and Development Program of China , doi 10.13039/501100012166;
                Funded by: Tianjin Science and Technology Program , doi 10.13039/501100019065;
                Funded by: Major State Basic Research Development Program of China , doi 10.13039/501100012336;
                Funded by: Javna Agencija za Raziskovalno Dejavnost RS , doi 10.13039/501100004329;
                Categories
                Immunology
                Systematic Review

                Immunology
                nanobody multimers,immunomodulation,intrabodies,imaging,nanobody functionalization
                Immunology
                nanobody multimers, immunomodulation, intrabodies, imaging, nanobody functionalization

                Comments

                Comment on this article