1
|
Mora-Boza A, Ahmedin Z, García AJ. Controlled release of therapeutic antibody using hydrolytically degradable microgels. J Biomed Mater Res A 2024; 112:1265-1275. [PMID: 37927169 PMCID: PMC11069594 DOI: 10.1002/jbm.a.37637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Monoclonal antibodies have gained significant interest as potential therapeutics for treating various diseases. However, these therapies are not always effective due to poor treatment compliance associated with multiple administrations and drug resistance. Thus, there is a growing interest in developing advanced monoclonal antibody delivery systems that can customize pharmacokinetics to enhance therapeutic outcomes. This work aimed to engineer hydrolytic 4-arm PEG maleimide (PEG-4MAL) microgels for the controlled delivery of therapeutic antibodies, specifically anti-angiogenic bevacizumab, to overcome the limitations of current monoclonal antibody therapies. Through a PEGylation reaction with a thiol-terminated PEG linker, the antibody was covalently conjugated to the macromer backbone before microgel synthesis. The PEGylation reaction was simple, effective, and did not affect antibody bioactivity. Antibody release kinetics was tuned by changing the concentration of the hydrolytic linker (0-2 mM) and/or PEG-4MAL:protein molar ratio (1000:1, 2000:1, and 5000:1) in the macromer precursor solution during microgel fabrication. The bioactivity of the released antibody was assessed on human umbilical endothelial vascular cells (HUVEC), demonstrating that extracts from hydrolytic microgels reduced cell proliferation over time. Collectively, this study demonstrates the development of highly tunable delivery platform based on degradable PEG-4MAL microgels that can be adapted for therapeutic antibody-controlled release.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Zakir Ahmedin
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Medina Pérez VM, Baselga M, Schuhmacher AJ. Single-Domain Antibodies as Antibody-Drug Conjugates: From Promise to Practice-A Systematic Review. Cancers (Basel) 2024; 16:2681. [PMID: 39123409 PMCID: PMC11311928 DOI: 10.3390/cancers16152681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) represent potent cancer therapies that deliver highly toxic drugs to tumor cells precisely, thus allowing for targeted treatment and significantly reducing off-target effects. Despite their effectiveness, ADCs can face limitations due to acquired resistance and potential side effects. OBJECTIVES This study focuses on advances in various ADC components to improve both the efficacy and safety of these agents, and includes the analysis of several novel ADC formats. This work assesses whether the unique features of VHHs-such as their small size, enhanced tissue penetration, stability, and cost-effectiveness-make them a viable alternative to conventional antibodies for ADCs and reviews their current status in ADC development. METHODS Following PRISMA guidelines, this study focused on VHHs as components of ADCs, examining advancements and prospects from 1 January 2014 to 30 June 2024. Searches were conducted in PubMed, Cochrane Library, ScienceDirect and LILACS using specific terms related to ADCs and single-domain antibodies. Retrieved articles were rigorously evaluated, excluding duplicates and non-qualifying studies. The selected peer-reviewed articles were analyzed for quality and synthesized to highlight advancements, methods, payloads, and future directions in ADC research. RESULTS VHHs offer significant advantages for drug conjugation over conventional antibodies due to their smaller size and structure, which enhance tissue penetration and enable access to previously inaccessible epitopes. Their superior stability, solubility, and manufacturability facilitate cost-effective production and expand the range of targetable antigens. Additionally, some VHHs can naturally cross the blood-brain barrier or be easily modified to favor their penetration, making them promising for targeting brain tumors and metastases. Although no VHH-drug conjugates (nADC or nanoADC) are currently in the clinical arena, preclinical studies have explored various conjugation methods and linkers. CONCLUSIONS While ADCs are transforming cancer treatment, their unique mechanisms and associated toxicities challenge traditional views on bioavailability and vary with different tumor types. Severe toxicities, often linked to compound instability, off-target effects, and nonspecific blood cell interactions, highlight the need for better understanding. Conversely, the rapid distribution, tumor penetration, and clearance of VHHs could be advantageous, potentially reducing toxicity by minimizing prolonged exposure. These attributes make single-domain antibodies strong candidates for the next generation of ADCs, potentially enhancing both efficacy and safety.
Collapse
Affiliation(s)
- Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Marta Baselga
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
3
|
Cong Y, Devoogdt N, Lambin P, Dubois LJ, Yaromina A. Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management. Cancers (Basel) 2024; 16:371. [PMID: 38254860 PMCID: PMC10814765 DOI: 10.3390/cancers16020371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The discovery of the distinctive structure of heavy chain-only antibodies in species belonging to the Camelidae family has elicited significant interest in their variable antigen binding domain (VHH) and gained attention for various applications, such as cancer diagnosis and treatment. This article presents an overview of the characteristics, advantages, and disadvantages of VHHs as compared to conventional antibodies, and their usage in diverse applications. The singular properties of VHHs are explained, and several strategies that can augment their utility are outlined. The preclinical studies illustrating the diagnostic and therapeutic efficacy of distinct VHHs in diverse formats against solid cancers are summarized, and an overview of the clinical trials assessing VHH-based agents in oncology is provided. These investigations demonstrate the enormous potential of VHHs for medical research and healthcare.
Collapse
Affiliation(s)
- Ying Cong
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands; (Y.C.); (P.L.)
| |
Collapse
|
4
|
Kalinovsky DV, Kholodenko IV, Svirshchevskaya EV, Kibardin AV, Ryazantsev DY, Rozov FN, Larin SS, Deyev SM, Kholodenko RV. Targeting GD2-Positive Tumor Cells by Pegylated scFv Fragment-Drug Conjugates Carrying Maytansinoids DM1 and DM4. Curr Issues Mol Biol 2023; 45:8112-8125. [PMID: 37886955 PMCID: PMC10604934 DOI: 10.3390/cimb45100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Oligomerization of antibody fragments via modification with polyethylene glycol (pegylation) may alter their function and properties, leading to a multivalent interaction of the resulting constructs with the target antigen. In a recent study, we generated pegylated monomers and multimers of scFv fragments of GD2-specific antibodies using maleimide-thiol chemistry. Multimerization enhanced the antigen-binding properties and demonstrated a more efficient tumor uptake in a syngeneic GD2-positive mouse cancer model compared to monomeric antibody fragments, thereby providing a rationale for improving the therapeutic characteristics of GD2-specific antibody fragments. In this work, we obtained pegylated conjugates of scFv fragments of GD2-specific antibodies with maytansinoids DM1 or DM4 using tetravalent PEG-maleimide (PEG4). The protein products from the two-stage thiol-maleimide reaction resolved by gel electrophoresis indicated that pegylated scFv fragments constituted the predominant part of the protein bands, and most of the scFv formed pegylated monomers and dimers. The conjugates retained the ability to bind ganglioside GD2 comparable to that of the parental scFv fragment and to specifically interact with GD2-positive cells. Both induced significant inhibitory effects in the GD2-positive B78-D14 cell line, in contrast to the GD2-negative B16 cell line. The decrease in the B78-D14 cell viability when treated with scFv-PEG4-DM4 was more prominent than that for scFv-PEG4-DM1, and was characterized by a twofold lower half-maximal inhibitory concentration (IC50). Unlike the parental scFv fragment, the product of scFv and PEG4 conjugation (scFv-PEG4), consisting predominantly of pegylated scFv multimers and monomers, induced direct cell death in the GD2-positive B78-D14 cells. However, the potency of scFv-PEG4 was low in the selected concentration range, thus demonstrating that the cytotoxic effect of DM1 and DM4 within the antibody fragment-drug conjugates was primary. The suggested approach may contribute to development of novel configurations of antibody fragment-drug conjugates for cancer treatment.
Collapse
Affiliation(s)
- Daniel V. Kalinovsky
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., Moscow 119121, Russia
| | - Elena V. Svirshchevskaya
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Alexey V. Kibardin
- Laboratory of Molecular Immunology, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia; (A.V.K.); (S.S.L.)
| | - Dmitry Yu. Ryazantsev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Fedor N. Rozov
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Sergey S. Larin
- Laboratory of Molecular Immunology, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia; (A.V.K.); (S.S.L.)
| | - Sergey M. Deyev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
- Laboratory of Molecular Pharmacology, Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- “Biomarker” Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan 420008, Russia
| | - Roman V. Kholodenko
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
- Real Target LLC, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia
| |
Collapse
|
5
|
Prado NDR, Brilhante-Da-Silva N, Sousa RMO, Morais MSDS, Roberto SA, Luiz MB, Assis LCD, Marinho ACM, Araujo LFLD, Pontes RDS, Stabeli RG, Fernandes CFC, Pereira SDS. Single-domain antibodies applied as antiviral immunotherapeutics. J Virol Methods 2023; 320:114787. [PMID: 37516366 DOI: 10.1016/j.jviromet.2023.114787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Viral infections have been the cause of high mortality rates throughout different periods in history. Over the last two decades, outbreaks caused by zoonotic diseases and transmitted by arboviruses have had a significant impact on human health. The emergence of viral infections in different parts of the world encourages the search for new inputs to fight pathologies of viral origin. Antibodies represent the predominant class of new drugs developed in recent years and approved for the treatment of various human diseases, including cancer, autoimmune and infectious diseases. A promising group of antibodies are single-domain antibodies derived from camelid heavy chain immunoglobulins, or VHHs, are biomolecules with nanometric dimensions and unique pharmaceutical and biophysical properties that can be used in the diagnosis and immunotherapy of viral infections. For viral neutralization to occur, VHHs can act in different stages of the viral cycle, including the actual inhibition of infection, to hindering viral replication or assembly. This review article addresses advances involving the use of VHHs in therapeutic propositions aimed to battle different viruses that affect human health.
Collapse
Affiliation(s)
- Nidiane Dantas Reis Prado
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | - Nairo Brilhante-Da-Silva
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil
| | - Rosa Maria Oliveira Sousa
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | | | - Sibele Andrade Roberto
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Marcos Barros Luiz
- Instituto Federal de Rondônia Campus Guajará-Mirim, IFRO, Guajará-Mirim, RO, Brazil
| | - Livia Coelho de Assis
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil
| | - Anna Carolina M Marinho
- Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Luiz Felipe Lemes de Araujo
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rafael de Souza Pontes
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rodrigo Guerino Stabeli
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Carla Freire Celedonio Fernandes
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Soraya Dos Santos Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
6
|
Maali A, Gholizadeh M, Feghhi-Najafabadi S, Noei A, Seyed-Motahari SS, Mansoori S, Sharifzadeh Z. Nanobodies in cell-mediated immunotherapy: On the road to fight cancer. Front Immunol 2023; 14:1012841. [PMID: 36761751 PMCID: PMC9905824 DOI: 10.3389/fimmu.2023.1012841] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
The immune system is essential in recognizing and eliminating tumor cells. The unique characteristics of the tumor microenvironment (TME), such as heterogeneity, reduced blood flow, hypoxia, and acidity, can reduce the efficacy of cell-mediated immunity. The primary goal of cancer immunotherapy is to modify the immune cells or the TME to enable the immune system to eliminate malignancies successfully. Nanobodies, known as single-domain antibodies, are light chain-free antibody fragments produced from Camelidae antibodies. The unique properties of nanobodies, including high stability, reduced immunogenicity, enhanced infiltration into the TME of solid tumors and facile genetic engineering have led to their promising application in cell-mediated immunotherapy. They can promote the cancer therapy either directly by bridging between tumor cells and immune cells and by targeting cancer cells using immune cell-bound nanobodies or indirectly by blocking the inhibitory ligands/receptors. The T-cell activation can be engaged through anti-CD3 and anti-4-1BB nanobodies in the bispecific (bispecific T-cell engagers (BiTEs)) and trispecific (trispecific T-cell engager (TriTEs)) manners. Also, nanobodies can be used as natural killer (NK) cell engagers (BiKEs, TriKEs, and TetraKEs) to create an immune synapse between the tumor and NK cells. Nanobodies can redirect immune cells to attack tumor cells through a chimeric antigen receptor (CAR) incorporating a nanobody against the target antigen. Various cancer antigens have been targeted by nanobody-based CAR-T and CAR-NK cells for treating both hematological and solid malignancies. They can also cause the continuation of immune surveillance against tumor cells by stopping inappropriate inhibition of immune checkpoints. Other roles of nanobodies in cell-mediated cancer immunotherapy include reprogramming macrophages to reduce metastasis and angiogenesis, as well as preventing the severe side effects occurring in cell-mediated immunotherapy. Here, we highlight the critical functions of various immune cells, including T cells, NK cells, and macrophages in the TME, and discuss newly developed immunotherapy methods based on the targeted manipulation of immune cells and TME with nanobodies.
Collapse
Affiliation(s)
- Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Monireh Gholizadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyedeh Sheila Seyed-Motahari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Zahra Sharifzadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,*Correspondence: Zahra Sharifzadeh,
| |
Collapse
|
7
|
Zhang Y, Yang S, Jiang D, Li Y, Ma S, Wang L, Li G, Wang H, Zhang A, Xu G. Screening and identification of an anti-PD-1 nanobody with antitumor activity. Biosci Rep 2022; 43:BSR20221546. [PMID: 36475449 PMCID: PMC9867944 DOI: 10.1042/bsr20221546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/25/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Blocking of PD-1 or PD-L1 with corresponding antibody to enhance T cell response and mediate antitumor activity has been successfully applied in clinical practice. Several immune checkpoint inhibitors including monoclonal antibodies targeting PD-1 have been approved by the Food and Drug Administration (FDA) in cancer immunotherapy. However, the application of traditional antibodies has limited due to their drawbacks of large molecular weight and low tissue penetration. As the high specificity and strong tissue penetration of nanobodies (Nbs), efforts have been taken to develop Nbs for cancer therapy. Herein, we aim to screen a specific Nb against human PD-1 derived from a naïve camel Nb phage display library and further study its biological characteristic and anti-tumor activity. Finally, an anti-PD-1 Nb with high specificity and affinity was screened and generated, its cytotoxicity and antitumor effect was also confirmed in vitro and vivo. All of these indicate that the anti-PD-1 Nb may provide an alternative and appealing therapeutic agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Yanting Zhang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Shaoqi Yang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Dan Jiang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Yanning Li
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Shuo Ma
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Liyan Wang
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Guangqi Li
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Hongxia Wang
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Aijun Zhang
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Guangxian Xu
- Department of Laboratory Medicine, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523808, China
| |
Collapse
|
8
|
Mulero F, Oteo M, Garaulet G, Magro N, Rebollo L, Medrano G, Santiveri C, Romero E, Sellek RE, Margolles Y, Campos-Olivas R, Arroyo AG, Fernández LA, Morcillo MA, Martínez-Torrecuadrada JL. Development of anti-membrane type 1-matrix metalloproteinase nanobodies as immunoPET probes for triple negative breast cancer imaging. Front Med (Lausanne) 2022; 9:1058455. [PMID: 36507540 PMCID: PMC9729729 DOI: 10.3389/fmed.2022.1058455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by aggressiveness and high rates of metastasis. The identification of relevant biomarkers is crucial to improve outcomes for TNBC patients. Membrane type 1-matrix metalloproteinase (MT1-MMP) could be a good candidate because its expression has been reported to correlate with tumor malignancy, progression and metastasis. Moreover, single-domain variable regions (VHHs or Nanobodies) derived from camelid heavy-chain-only antibodies have demonstrated improvements in tissue penetration and blood clearance, important characteristics for cancer imaging. Here, we have developed a nanobody-based PET imaging strategy for TNBC detection that targets MT1-MMP. A llama-derived library was screened against the catalytic domain of MT1-MMP and a panel of specific nanobodies were identified. After a deep characterization, two nanobodies were selected to be labeled with gallium-68 (68Ga). ImmunoPET imaging with both ([68Ga]Ga-NOTA-3TPA14 and [68Ga]Ga-NOTA-3CMP75) in a TNBC mouse model showed precise tumor-targeting capacity in vivo with high signal-to-background ratios. (68Ga)Ga-NOTA-3CMP75 exhibited higher tumor uptake compared to (68Ga)Ga-NOTA-3TPA14. Furthermore, imaging data correlated perfectly with the immunohistochemistry staining results. In conclusion, we found a promising candidate for nanobody-based PET imaging to be further investigated as a diagnostic tool in TNBC.
Collapse
Affiliation(s)
- Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Oteo
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Natalia Magro
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Lluvia Rebollo
- Protein Production Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Clara Santiveri
- Spectroscopy and Nuclear Magnetic Resonance Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Romero
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Ricela E. Sellek
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Yago Margolles
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, (CNB-CSIC), Madrid, Spain
| | - Ramón Campos-Olivas
- Spectroscopy and Nuclear Magnetic Resonance Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alicia G. Arroyo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Luis Angel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, (CNB-CSIC), Madrid, Spain
| | - Miguel Angel Morcillo
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain,*Correspondence: Miguel Angel Morcillo,
| | | |
Collapse
|
9
|
Lou H, Cao X. Antibody variable region engineering for improving cancer immunotherapy. Cancer Commun (Lond) 2022; 42:804-827. [PMID: 35822503 PMCID: PMC9456695 DOI: 10.1002/cac2.12330] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/25/2022] [Accepted: 06/22/2022] [Indexed: 04/09/2023] Open
Abstract
The efficacy and specificity of conventional monoclonal antibody (mAb) drugs in the clinic require further improvement. Currently, the development and application of novel antibody formats for improving cancer immunotherapy have attracted much attention. Variable region-retaining antibody fragments, such as antigen-binding fragment (Fab), single-chain variable fragment (scFv), bispecific antibody, and bi/trispecific cell engagers, are engineered with humanization, multivalent antibody construction, affinity optimization and antibody masking for targeting tumor cells and killer cells to improve antibody-based therapy potency, efficacy and specificity. In this review, we summarize the application of antibody variable region engineering and discuss the future direction of antibody engineering for improving cancer therapies.
Collapse
Affiliation(s)
- Hantao Lou
- Ludwig Institute of Cancer ResearchUniversity of OxfordOxfordOX3 7DRUK
- Chinese Academy for Medical Sciences Oxford InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Xuetao Cao
- Chinese Academy for Medical Sciences Oxford InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Department of ImmunologyCentre for Immunotherapy, Institute of Basic Medical SciencesChinese Academy of Medical SciencesBeijing100005P. R. China
| |
Collapse
|
10
|
Malindi Z, Barth S, Abrahamse H. The Potential of Antibody Technology and Silver Nanoparticles for Enhancing Photodynamic Therapy for Melanoma. Biomedicines 2022; 10:2158. [PMID: 36140259 PMCID: PMC9495799 DOI: 10.3390/biomedicines10092158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
Melanoma is highly aggressive and is known to be efficient at resisting drug-induced apoptotic signals. Resection is currently the gold standard for melanoma management, but it only offers local control of the early stage of the disease. Metastatic melanoma is prone to recurrence, and has a poor prognosis and treatment response. Thus, the need for advanced theranostic alternatives is evident. Photodynamic therapy has been increasingly studied for melanoma treatment; however, it relies on passive drug accumulation, leading to off-target effects. Nanoparticles enhance drug biodistribution, uptake and intra-tumoural concentration and can be functionalised with monoclonal antibodies that offer selective biorecognition. Antibody-drug conjugates reduce passive drug accumulation and off-target effects. Nonetheless, one limitation of monoclonal antibodies and antibody-drug conjugates is their lack of versatility, given cancer's heterogeneity. Monoclonal antibodies suffer several additional limitations that make recombinant antibody fragments more desirable. SNAP-tag is a modified version of the human DNA-repair enzyme, O6-alkylguanine-DNA alkyltransferase. It reacts in an autocatalytic and covalent manner with benzylguanine-modified substrates, providing a simple protein labelling system. SNAP-tag can be genetically fused with antibody fragments, creating fusion proteins that can be easily labelled with benzylguanine-modified payloads for site-directed delivery. This review aims to highlight the benefits and limitations of the abovementioned approaches and to outline how their combination could enhance photodynamic therapy for melanoma.
Collapse
Affiliation(s)
- Zaria Malindi
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, 55 Beit Street, Doornfontein, Johannesburg 2028, South Africa
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road Observatory, Cape Town 7925, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, 55 Beit Street, Doornfontein, Johannesburg 2028, South Africa
| |
Collapse
|
11
|
Li B, Qin X, Mi LZ. Nanobodies: from structure to applications in non-injectable and bispecific biotherapeutic development. NANOSCALE 2022; 14:7110-7122. [PMID: 35535618 DOI: 10.1039/d2nr00306f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The increasing demand for convenient, miniaturized and multifunctional antibodies necessitates the development of novel antigen-recognition molecules for biological and medical studies. Nanobodies, the functional variable regions of camelid heavy-chain-only antibodies, as a new tool, complement the conventional antibodies and are in the stage of rapid development. The outstanding advantages of nanobodies include a stable structure, easy production, excellent water solubility, high affinity toward antigens and low immunogenicity. With promising application potential, nanobodies are now increasingly applied to various studies, including protein structure analysis, microscopic imaging, medical diagnosis, and drug development. The approval of the first nanobody drug Caplacizumab by the FDA disclosed the therapeutic potential of nanobodies. The outbreak of COVID-19 accelerated the development of nanobody drugs in non-injectable and bispecific biotherapeutic applications. Herein, we reviewed recent studies on the nanobody structure, screening and their applications in protein structure analysis and nanobody drugs, especially on non-injectable nanobody and bispecific nanobody development.
Collapse
Affiliation(s)
- Bingxuan Li
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Xiaohong Qin
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Li-Zhi Mi
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| |
Collapse
|
12
|
Awad RM, Meeus F, Ceuppens H, Ertveldt T, Hanssens H, Lecocq Q, Mateusiak L, Zeven K, Valenta H, De Groof TWM, De Vlaeminck Y, Krasniqi A, De Veirman K, Goyvaerts C, D'Huyvetter M, Hernot S, Devoogdt N, Breckpot K. Emerging applications of nanobodies in cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:143-199. [PMID: 35777863 DOI: 10.1016/bs.ircmb.2022.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cancer is a heterogeneous disease, requiring treatment tailored to the unique phenotype of the patient's tumor. Monoclonal antibodies (mAbs) and variants thereof have enabled targeted therapies to selectively target cancer cells. Cancer cell-specific mAbs have been used for image-guided surgery and targeted delivery of radionuclides or toxic agents, improving classical treatment strategies. Cancer cell-specific mAbs can further inhibit tumor cell growth or can stimulate immune-mediated destruction of cancer cells, a feature that has also been achieved through mAb-mediated manipulation of immune cells and pathways. Drawbacks of mAbs and their variants, together with the discovery of camelid heavy chain-only antibodies and the many advantageous features of their variable domains, referred to as VHHs, single domain antibodies or nanobodies (Nbs), resulted in the exploration of Nbs as an alternative targeting moiety. We therefore review the state-of-the-art as well as novel exploitation strategies of Nbs for targeted cancer therapy.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Heleen Hanssens
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lukasz Mateusiak
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katty Zeven
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hana Valenta
- Lab for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Timo W M De Groof
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
13
|
Hagemans IM, Wierstra PJ, Steuten K, Molkenboer-Kuenen JDM, van Dalen D, Ter Beest M, van der Schoot JMS, Ilina O, Gotthardt M, Figdor CG, Scheeren FA, Heskamp S, Verdoes M. Multiscale imaging of therapeutic anti-PD-L1 antibody localization using molecularly defined imaging agents. J Nanobiotechnology 2022; 20:64. [PMID: 35109860 PMCID: PMC8811974 DOI: 10.1186/s12951-022-01272-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND While immune checkpoint inhibitors such as anti-PD-L1 antibodies have revolutionized cancer treatment, only subgroups of patients show durable responses. Insight in the relation between clinical response, PD-L1 expression and intratumoral localization of PD-L1 therapeutics could improve patient stratification. Therefore, we present the modular synthesis of multimodal antibody-based imaging tools for multiscale imaging of PD-L1 to study intratumoral distribution of PD-L1 therapeutics. RESULTS To introduce imaging modalities, a peptide containing a near-infrared dye (sulfo-Cy5), a chelator (DTPA), an azide, and a sortase-recognition motif was synthesized. This peptide and a non-fluorescent intermediate were used for site-specific functionalization of c-terminally sortaggable mouse IgG1 (mIgG1) and Fab anti-PD-L1. To increase the half-life of the Fab fragment, a 20 kDa PEG chain was attached via strain-promoted azide-alkyne cycloaddition (SPAAC). Biodistribution and imaging studies were performed with 111In-labeled constructs in 4T1 tumor-bearing mice. Comparing our site-specific antibody-conjugates with randomly conjugated antibodies, we found that antibody clone, isotype and method of DTPA conjugation did not change tumor uptake. Furthermore, addition of sulfo-Cy5 did not affect the biodistribution. PEGylated Fab fragment displayed a significantly longer half-life compared to unPEGylated Fab and demonstrated the highest overall tumor uptake of all constructs. PD-L1 in tumors was clearly visualized by SPECT/CT, as well as whole body fluorescence imaging. Immunohistochemistry staining of tumor sections demonstrated that PD-L1 co-localized with the fluorescent and autoradiographic signal. Intratumoral localization of the imaging agent could be determined with cellular resolution using fluorescent microscopy. CONCLUSIONS A set of molecularly defined multimodal antibody-based PD-L1 imaging agents were synthesized and validated for multiscale monitoring of PD-L1 expression and localization. Our modular approach for site-specific functionalization could easily be adapted to other targets.
Collapse
Affiliation(s)
- Iris M Hagemans
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Peter J Wierstra
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kas Steuten
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Janneke D M Molkenboer-Kuenen
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Duco van Dalen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan M S van der Schoot
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Olga Ilina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
- Institute for Chemical Immunology, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Zadeh Mehrizi T, Mousavi Hosseini K. An overview on the investigation of nanomaterials' effect on plasma components: immunoglobulins and coagulation factor VIII, 2010-2020 review. NANOSCALE ADVANCES 2021; 3:3730-3745. [PMID: 36133015 PMCID: PMC9419877 DOI: 10.1039/d1na00119a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/16/2021] [Indexed: 05/04/2023]
Abstract
FVIII and immunoglobulins (Igs) are the most prominent plasma proteins, which play a vital role in plasma hemostasis. These proteins have been implemented frequently in protein therapy. Therefore, their maintenance, durability, and stability are highly essential. Herein, various approaches to improve protein functions have been investigated, such as using recombinant protein replacement. In comparison, advances in nanotechnology have provided adequate context to boost biomaterial utilization. In this regard, the applications of various nanoparticles such as polymeric nanomaterials (PEG and PLGA), metal nanoparticles, dendrimers, and lipid based nanomaterials (liposomes and lipid nanoparticles) in stability and the functional improvement of antibodies and coagulation factor VIII (FVIII) have been reviewed from 2010 to 2020. Reviewing related articles has shown that not only can nanomaterials adequately protect the structure of proteins, but have also improved proteins' functions in some cases. For example, the high rate of FVIII instability has been successfully enhanced by bio-PEGylation. Also, utilizing PEGylated liposomes, using the PEG-lip technique for coating nanostructures, leads to FIIIV half-life prolongation. Hence, PEGylation had most impact on the stability of FVIII. Likewise, PEG-coated liposome nano-carriers also presented such a good effect on stability improvements for FVIII due to their ability to tune the immune system by reducing FVIII immunogenicity. Similarly, Ig PEGylation and conjugation to magnetic nanoparticles resulted in increased half-life and better purification of Igs, respectively, without any loss in structural or functional features. Consequently, metal-organic frameworks and recent hybrid systems have been introduced as promising nanomaterials in biomedical applications. As far as we know, this is the first study in this field, which considers the applications of nanoparticles for improving the storage and stability of antibodies and coagulation FVIII.
Collapse
Affiliation(s)
- Tahereh Zadeh Mehrizi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine Tehran Iran +989338606292
| | - Kamran Mousavi Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine Tehran Iran +989338606292
| |
Collapse
|
15
|
Wu T, Liu M, Huang H, Sheng Y, Xiao H, Liu Y. Clustered nanobody-drug conjugates for targeted cancer therapy. Chem Commun (Camb) 2021; 56:9344-9347. [PMID: 32672289 DOI: 10.1039/d0cc03396k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A clustered Nb-drug conjugate (cNDC@PEG) was designed using anti-EGFR Nb to specifically deliver Pt(iv) prodrugs to tumors. cNDC@PEG efficiently targets EGFR positive tumor cells, and the clustered cNDC@PEG is more efficient in inhibiting tumor growth in vivo than the monomeric NDC. This work provides a novel strategy for the construction of a multi-valent NDC using dendrimers.
Collapse
Affiliation(s)
- Tiantian Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Manman Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Hai Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Yaping Sheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yangzhong Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
16
|
Hong H, Li C, Gong L, Wang J, Li D, Shi J, Zhou Z, Huang Z, Wu Z. Universal endogenous antibody recruiting nanobodies capable of triggering immune effectors for targeted cancer immunotherapy. Chem Sci 2021; 12:4623-4630. [PMID: 34163726 PMCID: PMC8179521 DOI: 10.1039/d0sc05332e] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/07/2021] [Indexed: 01/21/2023] Open
Abstract
Developing monoclonal antibodies (mAbs) for cancer immunotherapy is expensive and complicated. Nanobodies are small antibodies possessing favorable pharmacological properties compared with mAbs, but have limited anticancer efficacy due to the lack of an Fc region and poor pharmacokinetics. In this context, engineered universal endogenous antibody-recruiting nanobodies (UEAR Nbs), as a general and cost-effective approach, were developed to generate functional antibody-like nanobodies that could recapitulate the Fc biological functions for cancer immunotherapy. The UEAR Nbs, composed of the IgG binding domain and nanobody, were recombinantly expressed in E. coli and could recruit endogenous IgGs onto the cancer cell surface and trigger potent immune responses to kill cancer cells in vitro. Moreover, it was proved that UEAR Nbs displayed significantly improved half-lives in vivo. The in vivo antitumor efficacy of UEAR Nbs was demonstrated in a murine model using EGFR positive triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Chen Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Liang Gong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Jinfeng Wang
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Dan Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Jie Shi
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Zhifang Zhou
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University Wuxi 214062 China
- Laboratory of Cancer Epigenetics, School of Medicine, Jiangnan University Wuxi 214122 China
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University Wuxi 214122 China
| |
Collapse
|
17
|
Bao G, Tang M, Zhao J, Zhu X. Nanobody: a promising toolkit for molecular imaging and disease therapy. EJNMMI Res 2021; 11:6. [PMID: 33464410 PMCID: PMC7815856 DOI: 10.1186/s13550-021-00750-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Nanobodies are the recombinant variable domains of heavy-chain-only antibodies, with many unique properties such as small size, excellent solubility, superior stability, quick clearance from blood, and deep tissue penetration. As a result, nanobodies have become a promising tool for the diagnosis and therapy of diseases. As imaging tracers, nanobodies allow an early acquisition of high-quality images, provide a comprehensive evaluation of the disease, and subsequently enable a personalized precision therapy. As therapeutic agents, nanobodies enable a targeted therapy by lesion-specific delivery of drugs and effector domains, thereby improving the specificity and efficacy of the therapy. Up to date, a wide variety of nanobodies have been developed for a broad range of molecular targets and have played a significant role in patients with a broad spectrum of diseases. In this review, we aim to outline the current state-of-the-art research on the nanobodies for medical applications and then discuss the challenges and strategies for their further clinical translation.
Collapse
Affiliation(s)
- Guangfa Bao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Ming Tang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Jun Zhao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
18
|
DNA-based delivery of anti-DR5 Nanobodies improves exposure and anti-tumor efficacy over protein-based administration. Cancer Gene Ther 2020; 28:828-838. [PMID: 32733055 DOI: 10.1038/s41417-020-0204-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 11/09/2022]
Abstract
Nanobodies present an appealing class of potential cancer therapeutics. The current study explores the in vivo expression of these molecules through DNA-encoded delivery. We hypothesized that this approach could address the rapid clearance of Nanobodies and, through half-life modulation, increase the produced levels in circulation. We therefore evaluated pharmacokinetics and efficacy of variants of an anti-death receptor 5 Nanobody (NbDR5), either monovalent or multivalent with half-life extension properties, after DNA-based administration. Intramuscular electrotransfer of a monovalent NbDR5-encoding plasmid (pNbDR5) did not result in detectable plasma levels in BALB/c mice. A tetravalent NbDR5-encoding plasmid (pNbDR54) provided peak concentrations of 54 ng/mL, which remained above 24 ng/mL during a 12-week follow-up. DNA-based delivery of these Nanobody formats fused to a Nanobody binding to serum albumin (NbSA), pNbDR5-NbSA and pNbDR54-NbSA, resulted in significantly higher plasma levels, with peak titers of 5.2 and 7.7 µg/mL, respectively. In an athymic-nude mice COLO 205 colon-cancer model, a quadrupled intramuscular DNA dose led to peak plasma levels of 270 ng/mL for pNbDR54 and 38 µg/mL for pNbDR54-NbSA. Potent anti-tumor responses were only observed for pNbDR54, following either intramuscular or intratumoral delivery. Despite comparable in vitro activity and superior plasma exposure, NbDR54-NbSA was less effective than NbDR54 in vivo, regardless of whether delivered as DNA or protein. Overall, DNA-based Nanobody delivery resulted in more potent and durable anti-tumor responses than protein-based Nanobody delivery. In conclusion, this study demonstrates pre-clinical proof of concept for DNA-based Nanobodies in oncology and highlights the improved outcome over conventional protein administration.
Collapse
|
19
|
Yang EY, Shah K. Nanobodies: Next Generation of Cancer Diagnostics and Therapeutics. Front Oncol 2020; 10:1182. [PMID: 32793488 PMCID: PMC7390931 DOI: 10.3389/fonc.2020.01182] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The development of targeted medicine has greatly expanded treatment options and spurred new research avenues in cancer therapeutics, with monoclonal antibodies (mAbs) emerging as a prevalent treatment in recent years. With mixed clinical success, mAbs still hold significant shortcomings, as they possess limited tumor penetration, high manufacturing costs, and the potential to develop therapeutic resistance. However, the recent discovery of “nanobodies,” the smallest-known functional antibody fragment, has demonstrated significant translational potential in preclinical and clinical studies. This review highlights their various applications in cancer and analyzes their trajectory toward their translation into the clinic.
Collapse
Affiliation(s)
- Emily Y Yang
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Departments of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Departments of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
| |
Collapse
|
20
|
The old CEACAMs find their new role in tumor immunotherapy. Invest New Drugs 2020; 38:1888-1898. [PMID: 32488569 DOI: 10.1007/s10637-020-00955-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/21/2020] [Indexed: 12/16/2022]
Abstract
Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) contain 12 family members(CEACAM1、CEACAM3、CEACAM4、CEACAM5、CEACAM6、CEACAM7、CEACAM8、CEACAM16、CEACAM18、CEACAM19、CEACAM20 and CEACAM21)and are expressed diversely in different normal and tumor tissues. CEA (CEACAM5) has been used as a tumor biomarker since 1965. Here we review the latest research and development of the structures, expression, and function of CEACAMs in normal and tumor tissues, and their application in the tumor diagnosis, prognosis, and treatment. We focus on recent clinical studies of CEA targeted cancer immunotherapies, including bispecific antibody (BsAb) for radio-immuno-therapy and imaging, bispecific T cell engager (BiTE) and chimeric antigen receptor T cells (CAR-T). We summarize the promising clinical relevance and challenges of these approaches and give perspective view for future research. This review has important implications in understanding the diversified biology of CEACAMs in normal and tumor tissues, and their new role in tumor immunotherapy.
Collapse
|
21
|
Lejeune M, Köse MC, Duray E, Einsele H, Beguin Y, Caers J. Bispecific, T-Cell-Recruiting Antibodies in B-Cell Malignancies. Front Immunol 2020; 11:762. [PMID: 32457743 PMCID: PMC7221185 DOI: 10.3389/fimmu.2020.00762] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (BsAbs) are designed to recognize and bind to two different antigens or epitopes. In the last few decades, BsAbs have been developed within the context of cancer therapies and in particular for the treatment of hematologic B-cell malignancies. To date, more than one hundred different BsAb formats exist, including bispecific T-cell engagers (BiTEs), and new constructs are constantly emerging. Advances in protein engineering have enabled the creation of BsAbs with specific mechanisms of action and clinical applications. Moreover, a better understanding of resistance and evasion mechanisms, as well as advances in the protein engineering and in immunology, will help generating a greater variety of BsAbs to treat various cancer types. This review focuses on T-cell-engaging BsAbs and more precisely on the various BsAb formats currently being studied in the context of B-cell malignancies, on ongoing clinical trials and on the clinical concerns to be taken into account in the development of new BsAbs.
Collapse
Affiliation(s)
- Margaux Lejeune
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Murat Cem Köse
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Elodie Duray
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Hermann Einsele
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Yves Beguin
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| |
Collapse
|
22
|
V Kholodenko I, V Kalinovsky D, V Svirshchevskaya E, I Doronin I, V Konovalova M, V Kibardin A, V Shamanskaya T, S Larin S, M Deyev S, V Kholodenko R. Multimerization through Pegylation Improves Pharmacokinetic Properties of scFv Fragments of GD2-Specific Antibodies. Molecules 2019; 24:molecules24213835. [PMID: 31653037 PMCID: PMC6864547 DOI: 10.3390/molecules24213835] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Antigen-binding fragments of antibodies specific to the tumor-associated ganglioside GD2 are well poised to play a substantial role in modern GD2-targeted cancer therapies, however, rapid elimination from the body and reduced affinity compared to full-length antibodies limit their therapeutic potential. In this study, scFv fragments of GD2-specific antibodies 14.18 were produced in a mammalian expression system that specifically bind to ganglioside GD2, followed by site-directed pegylation to generate mono-, di-, and tetra-scFv fragments. Fractionated pegylated dimers and tetramers of scFv fragments showed significant increase of the binding to GD2 which was not accompanied by cross-reactivity with other gangliosides. Pegylated multimeric di-scFvs and tetra-scFvs exhibited cytotoxic effects in GD2-positive tumor cells, while their circulation time in blood significantly increased compared with monomeric antibody fragments. We also demonstrated a more efficient tumor uptake of the multimers in a syngeneic GD2-positive mouse cancer model. The findings of this study provide the rationale for improving therapeutic characteristics of GD2-specific antibody fragments by multimerization and propose a strategy to generate such molecules. On the basis of multimeric antibody fragments, bispecific antibodies and conjugates with cytotoxic drugs or radioactive isotopes may be developed that will possess improved pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
- Irina V Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., Moscow 119121, Russia.
| | - Daniel V Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
| | - Elena V Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
| | - Igor I Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
- Real Target LLC, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia.
| | - Maria V Konovalova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
| | - Alexey V Kibardin
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia.
| | - Tatyana V Shamanskaya
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia.
| | - Sergey S Larin
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia.
| | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia.
| | - Roman V Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia.
- Real Target LLC, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia.
| |
Collapse
|
23
|
Lecocq Q, De Vlaeminck Y, Hanssens H, D'Huyvetter M, Raes G, Goyvaerts C, Keyaerts M, Devoogdt N, Breckpot K. Theranostics in immuno-oncology using nanobody derivatives. Am J Cancer Res 2019; 9:7772-7791. [PMID: 31695800 PMCID: PMC6831473 DOI: 10.7150/thno.34941] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 07/11/2019] [Indexed: 12/25/2022] Open
Abstract
Targeted therapy and immunotherapy have become mainstream in cancer treatment. However, only patient subsets benefit from these expensive therapies, and often responses are short‐lived or coincide with side effects. A growing modality in precision oncology is the development of theranostics, as this enables patient selection, treatment and monitoring. In this approach, labeled compounds and an imaging technology are used to diagnose patients and select the best treatment option, whereas for therapy, related compounds are used to target cancer cells or the tumor stroma. In this context, nanobodies and nanobody-directed therapeutics have gained interest. This interest stems from their high antigen specificity, small size, ease of labeling and engineering, allowing specific imaging and design of therapies targeting antigens on tumor cells, immune cells as well as proteins in the tumor environment. This review provides a comprehensive overview on the state-of-the-art regarding the use of nanobodies as theranostics, and their importance in the emerging field of personalized medicine.
Collapse
|
24
|
Strategies for the production of long-acting therapeutics and efficient drug delivery for cancer treatment. Biomed Pharmacother 2019; 113:108750. [DOI: 10.1016/j.biopha.2019.108750] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 11/21/2022] Open
|
25
|
Liu J, Wu X, Lin L, Pan H, Wang Y, Li Y, Zhao Y, Wang Z. Bp-Bs, a Novel T-cell Engaging Bispecific Antibody with Biparatopic Her2 Binding, Has Potent Anti-tumor Activities. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:66-73. [PMID: 31020038 PMCID: PMC6475711 DOI: 10.1016/j.omto.2019.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/26/2019] [Indexed: 12/13/2022]
Abstract
Patients with Human epidermal growth factor receptor type 2 (Her2) overexpression are associated with aggressive tumor growth and poor clinical outcomes. Bispecific antibodies targeting Her2 have recently exhibited potent effects on Her2 signal inhibition. In this study, a novel biparatopic anti-Her2 bispecific antibody (Bp-Bs) was constructed by linking a single anti-CD3 Fab with two different anti-Her2 single-domain antibodies targeting non-overlapping epitopes of Her2. The Bp-Bs demonstrated strong binding on Her2-positive cells and potent cytotoxicity on Her2-positive tumor cells, even Her2-low expression cells, suggesting that biparatopic bispecific antibodies may have improved therapeutic benefits on broad Her2 patient populations.
Collapse
Affiliation(s)
- Jiayu Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaoqiong Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Limin Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Haitao Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yanlan Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yumei Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yining Zhao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhong Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
26
|
Chanier T, Chames P. Nanobody Engineering: Toward Next Generation Immunotherapies and Immunoimaging of Cancer. Antibodies (Basel) 2019; 8:E13. [PMID: 31544819 PMCID: PMC6640690 DOI: 10.3390/antib8010013] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
In the last decade, cancer immunotherapies have produced impressive therapeutic results. However, the potency of immunotherapy is tightly linked to immune cell infiltration within the tumor and varies from patient to patient. Thus, it is becoming increasingly important to monitor and modulate the tumor immune infiltrate for an efficient diagnosis and therapy. Various bispecific approaches are being developed to favor immune cell infiltration through specific tumor targeting. The discovery of antibodies devoid of light chains in camelids has spurred the development of single domain antibodies (also called VHH or nanobody), allowing for an increased diversity of multispecific and/or multivalent formats of relatively small sizes endowed with high tissue penetration. The small size of nanobodies is also an asset leading to high contrasts for non-invasive imaging. The approval of the first therapeutic nanobody directed against the von Willebrand factor for the treatment of acquired thrombotic thrombocypenic purpura (Caplacizumab, Ablynx), is expected to bolster the rise of these innovative molecules. In this review, we discuss the latest advances in the development of nanobodies and nanobody-derived molecules for use in cancer immunotherapy and immunoimaging.
Collapse
Affiliation(s)
- Timothée Chanier
- Aix Marseille University, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, 13009 Marseille, France.
| | - Patrick Chames
- Aix Marseille University, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, 13009 Marseille, France.
| |
Collapse
|
27
|
|