Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Zhonghua Xue Ye Xue Za Zhi ; 44(5): 380-387, 2023 May 14.
Article in Chinese | MEDLINE | ID: mdl-37550187

ABSTRACT

Objective: To understand the current status of diagnosis and treatment of chronic lymphocytic leukemia (CLL) /small lymphocytic lymphoma (SLL) among hematologists, oncologists, and lymphoma physicians from hospitals of different levels in China. Methods: This multicenter questionnaire survey was conducted from March 2021 to July 2021 and included 1,000 eligible physicians. A combination of face-to-face interviews and online questionnaire surveys was used. A standardized questionnaire regarding the composition of patients treated for CLL/SLL, disease diagnosis and prognosis evaluation, concomitant diseases, organ function evaluation, treatment selection, and Bruton tyrosine kinase (BTK) inhibitor was used. Results: ①The interviewed physicians stated that the proportion of male patients treated for CLL/SLL is higher than that of females, and the age is mainly concentrated in 61-70 years old. ②Most of the interviewed physicians conducted tests, such as bone marrow biopsies and immunohistochemistry, for patient diagnosis, in addition to the blood test. ③Only 13.7% of the interviewed physicians fully grasped the initial treatment indications recommended by the existing guidelines. ④In terms of cognition of high-risk prognostic factors, physicians' knowledge of unmutated immunoglobulin heavy-chain variable and 11q- is far inferior to that of TP53 mutation and complex karyotype, which are two high-risk prognostic factors, and only 17.1% of the interviewed physicians fully mastered CLL International Prognostic Index scoring system. ⑤Among the first-line treatment strategy, BTK inhibitors are used for different types of patients, and physicians have formed a certain understanding that BTK inhibitors should be preferentially used in patients with high-risk factors and elderly patients, but the actual use of BTK inhibitors in different types of patients is not high (31.6%-46.0%). ⑥BTK inhibitors at a reduced dose in actual clinical treatment were used by 69.0% of the physicians, and 66.8% of the physicians had interrupted the BTK inhibitor for >12 days in actual clinical treatment. The use of BTK inhibitors is reduced or interrupted mainly because of adverse reactions, such as atrial fibrillation, severe bone marrow suppression, hemorrhage, and pulmonary infection, as well as patients' payment capacity and effective disease progression control. ⑦Some differences were found in the perceptions and behaviors of hematologists and oncologists regarding the prognostic assessment of CLL/SLL, the choice of treatment options, the clinical use of BTK inhibitors, etc. Conclusion: At present, a gap remains between the diagnosis and treatment of CLL/SLL among Chinese physicians compared with the recommendations in the guidelines regarding the diagnostic criteria, treatment indications, prognosis assessment, accompanying disease assessment, treatment strategy selection, and rational BTK inhibitor use, especially the proportion of dose reduction or BTK inhibitor discontinuation due to high adverse events.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, B-Cell , Female , Humans , Male , Aged , Middle Aged , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Prognosis , Immunohistochemistry , Immunoglobulin Heavy Chains/therapeutic use
2.
Mod Rheumatol ; 33(6): 1059-1067, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37185766

ABSTRACT

Tumour necrosis factor (TNF) inhibitors are currently the most widely used biological agents to treat rheumatoid arthritis. Ozoralizumab (OZR), a novel TNF inhibitor, is an antibody using variable heavy-chain domains of heavy-chain antibody (VHHs) and became the first VHH drug approved for the treatment of rheumatoid arthritis in September 2022. VHHs isolated from camelid heavy-chain antibodies can bind antigens with a single molecule. OZR is a trivalent VHH that consists of two anti-human TNFα VHHs and one anti-human serum albumin (anti-HSA) VHH. This review summarizes OZR's unique structural characteristics and nonclinical and clinical data. The clinical data outline the pharmacokinetics, efficacy, relationship between efficacy and pharmacokinetics, and safety of OZR, focusing on a Phase II/III confirmatory study (OHZORA trial).


Subject(s)
Arthritis, Rheumatoid , Tumor Necrosis Factor Inhibitors , Humans , Tumor Necrosis Factor Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/therapeutic use , Tumor Necrosis Factor-alpha , Arthritis, Rheumatoid/drug therapy
3.
Front Immunol ; 14: 1133967, 2023.
Article in English | MEDLINE | ID: mdl-36960053

ABSTRACT

Introduction: B cells are acknowledged as crucial players in the pathogenesis of multiple sclerosis (MS). Several disease modifying drugs including cladribine have been shown to exert differential effects on peripheral blood B cell subsets. However, little is known regarding functional changes within the peripheral B cell populations. In this study, we obtained a detailed picture of B cell repertoire changes under cladribine treatment on a combined immunoglobulin (Ig) transcriptome and proteome level. Methods: We performed next-generation sequencing of Ig heavy chain (IGH) transcripts and Ig mass spectrometry in cladribine-treated patients with relapsing-remitting multiple sclerosis (n = 8) at baseline and after 6 and 12 months of treatment in order to generate Ig transcriptome and Ig peptide libraries. Ig peptides were overlapped with the corresponding IGH transcriptome in order to analyze B cell clones on a combined transcriptome and proteome level. Results: The analysis of peripheral blood B cell percentages pointed towards a significant decrease of memory B cells and an increase of naive B cells following cladribine therapy. While basic IGH repertoire parameters (e.g. variable heavy chain family usage and Ig subclasses) were only slightly affected by cladribine treatment, a significantly decreased number of clones and significantly lower diversity in the memory subset was noticeable at 6 months following treatment which was sustained at 12 months. When looking at B-cell clones comprising sequences from the different time-points, clones spanning between all three time-points were significantly more frequent than clones including sequences from two time-points. Furthermore, Ig proteome analyses showed that Ig transcriptome specific peptides could mostly be equally aligned to all three time-points pointing towards a proportion of B-cell clones that are maintained during treatment. Discussion: Our findings suggest that peripheral B cell related treatment effects of cladribine tablets might be exerted through a reduction of possibly disease relevant clones in the memory B cell subset without disrupting the overall clonal composition of B cells. Our results -at least partially- might explain the relatively mild side effects regarding infections and the sustained immune response after vaccinations during treatment. However, exact disease driving B cell subsets and their effects remain unknown and should be addressed in future studies.


Subject(s)
Cladribine , Multiple Sclerosis , Humans , Cladribine/therapeutic use , Cladribine/adverse effects , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Memory B Cells , Proteome , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/therapeutic use , Clone Cells
4.
Chinese Journal of Hematology ; (12): 380-387, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-984633

ABSTRACT

Objective: To understand the current status of diagnosis and treatment of chronic lymphocytic leukemia (CLL) /small lymphocytic lymphoma (SLL) among hematologists, oncologists, and lymphoma physicians from hospitals of different levels in China. Methods: This multicenter questionnaire survey was conducted from March 2021 to July 2021 and included 1,000 eligible physicians. A combination of face-to-face interviews and online questionnaire surveys was used. A standardized questionnaire regarding the composition of patients treated for CLL/SLL, disease diagnosis and prognosis evaluation, concomitant diseases, organ function evaluation, treatment selection, and Bruton tyrosine kinase (BTK) inhibitor was used. Results: ①The interviewed physicians stated that the proportion of male patients treated for CLL/SLL is higher than that of females, and the age is mainly concentrated in 61-70 years old. ②Most of the interviewed physicians conducted tests, such as bone marrow biopsies and immunohistochemistry, for patient diagnosis, in addition to the blood test. ③Only 13.7% of the interviewed physicians fully grasped the initial treatment indications recommended by the existing guidelines. ④In terms of cognition of high-risk prognostic factors, physicians' knowledge of unmutated immunoglobulin heavy-chain variable and 11q- is far inferior to that of TP53 mutation and complex karyotype, which are two high-risk prognostic factors, and only 17.1% of the interviewed physicians fully mastered CLL International Prognostic Index scoring system. ⑤Among the first-line treatment strategy, BTK inhibitors are used for different types of patients, and physicians have formed a certain understanding that BTK inhibitors should be preferentially used in patients with high-risk factors and elderly patients, but the actual use of BTK inhibitors in different types of patients is not high (31.6%-46.0%). ⑥BTK inhibitors at a reduced dose in actual clinical treatment were used by 69.0% of the physicians, and 66.8% of the physicians had interrupted the BTK inhibitor for >12 days in actual clinical treatment. The use of BTK inhibitors is reduced or interrupted mainly because of adverse reactions, such as atrial fibrillation, severe bone marrow suppression, hemorrhage, and pulmonary infection, as well as patients' payment capacity and effective disease progression control. ⑦Some differences were found in the perceptions and behaviors of hematologists and oncologists regarding the prognostic assessment of CLL/SLL, the choice of treatment options, the clinical use of BTK inhibitors, etc. Conclusion: At present, a gap remains between the diagnosis and treatment of CLL/SLL among Chinese physicians compared with the recommendations in the guidelines regarding the diagnostic criteria, treatment indications, prognosis assessment, accompanying disease assessment, treatment strategy selection, and rational BTK inhibitor use, especially the proportion of dose reduction or BTK inhibitor discontinuation due to high adverse events.


Subject(s)
Female , Humans , Male , Aged , Middle Aged , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Prognosis , Lymphoma, B-Cell , Immunohistochemistry , Immunoglobulin Heavy Chains/therapeutic use
5.
Front Immunol ; 13: 1005800, 2022.
Article in English | MEDLINE | ID: mdl-36405759

ABSTRACT

Nanobodies are well suited for constructing biologics due to their high solubility. We generated nanobodies directed against CD38, a tumor marker that is overexpressed by multiple myeloma and other hematological malignancies. We then used these CD38-specific nanobodies to construct heavy chain antibodies, bispecific killer cell engagers (BiKEs), chimeric antigen receptor (CAR)-NK cells, and nanobody-displaying AAV vectors. Here we review the utility of these nanobody-based constructs to specifically and effectively target CD38-expressing myeloma cells. The promising results of our preclinical studies warrant further clinical studies to evaluate the potential of these CD38-specific nanobody-based constructs for treatment of multiple myeloma.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Receptors, Chimeric Antigen , Single-Domain Antibodies , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use , Multiple Myeloma/drug therapy , Antibodies, Bispecific/therapeutic use , ADP-ribosyl Cyclase 1 , Immunoglobulin Heavy Chains/therapeutic use , Killer Cells, Natural
6.
Front Immunol ; 12: 728694, 2021.
Article in English | MEDLINE | ID: mdl-34646268

ABSTRACT

Monoclonal antibodies (mAbs) are an important class of therapeutics used to treat cancer, inflammation, and infectious diseases. Identifying highly developable mAb sequences in silico could greatly reduce the time and cost required for therapeutic mAb development. Here, we present position-specific scoring matrices (PSSMs) for antibody framework mutations developed using baseline human antibody repertoire sequences. Our analysis shows that human antibody repertoire-based PSSMs are consistent across individuals and demonstrate high correlations between related germlines. We show that mutations in existing therapeutic antibodies can be accurately predicted solely from baseline human antibody sequence data. We find that mAbs developed using humanized mice had more human-like FR mutations than mAbs originally developed by hybridoma technology. A quantitative assessment of entire framework regions of therapeutic antibodies revealed that there may be potential for improving the properties of existing therapeutic antibodies by incorporating additional mutations of high frequency in baseline human antibody repertoires. In addition, high frequency mutations in baseline human antibody repertoires were predicted in silico to reduce immunogenicity in therapeutic mAbs due to the removal of T cell epitopes. Several therapeutic mAbs were identified to have common, universally high-scoring framework mutations, and molecular dynamics simulations revealed the mechanistic basis for the evolutionary selection of these mutations. Our results suggest that baseline human antibody repertoires may be useful as predictive tools to guide mAb development in the future.


Subject(s)
Antibodies, Monoclonal/genetics , Drug Development , Epitopes, T-Lymphocyte/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Mutation , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , DNA Mutational Analysis , Databases, Genetic , Drug Approval , Drug Stability , Epitopes, T-Lymphocyte/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Variable Region/immunology , Immunoglobulin Variable Region/therapeutic use , Models, Genetic , Molecular Dynamics Simulation , Protein Stability , United States , United States Food and Drug Administration
7.
MAbs ; 13(1): 1892366, 2021.
Article in English | MEDLINE | ID: mdl-33722161

ABSTRACT

The patent literature should reflect the past 30 years of engineering efforts directed toward developing monoclonal antibody therapeutics. Such information is potentially valuable for rational antibody design. Patents, however, are designed not to convey scientific knowledge, but to provide legal protection. It is not obvious whether antibody information from patent documents, such as antibody sequences, is useful in conveying engineering know-how, rather than as a legal reference only. To assess the utility of patent data for therapeutic antibody engineering, we quantified the amount of antibody sequences in patents destined for medicinal purposes and how well they reflect the primary sequences of therapeutic antibodies in clinical use. We identified 16,526 patent families covering major jurisdictions (e.g., US Patent and Trademark Office (USPTO) and World Intellectual Property Organization) that contained antibody sequences. These families held 245,109 unique antibody chains (135,397 heavy chains and 109,712 light chains) that we compiled in our Patented Antibody Database (PAD, http://naturalantibody.com/pad). We find that antibodies make up a non-trivial proportion of all patent amino acid sequence depositions (e.g., 11% of USPTO Full Text database). Our analysis of the 16,526 families demonstrates that the volume of patent documents with antibody sequences is growing, with the majority of documents classified as containing antibodies for medicinal purposes. We further studied the 245,109 antibody chains from patent literature to reveal that they very well reflect the primary sequences of antibody therapeutics in clinical use. This suggests that the patent literature could serve as a reference for previous engineering efforts to improve rational antibody design.


Subject(s)
Antibodies, Monoclonal/chemistry , Data Mining , Databases, Protein , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Intellectual Property , Legislation, Drug , Patents as Topic , Amino Acid Sequence , Antibodies, Monoclonal/therapeutic use , Drug Design , Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Light Chains/therapeutic use
8.
Curr Oncol ; 28(1): 332-345, 2021 01 09.
Article in English | MEDLINE | ID: mdl-33435341

ABSTRACT

Background: Continuous oral targeted therapy (OTT) for chronic lymphocytic leukemia (CLL) represents an effective therapy but also a major economic burden on the healthcare system. This study aimed to estimate future direct costs, along with the prevalence, of CLL in the era of continuous OTT in Canada. Methods: The economic burden of OTT was modelled and compared to chemoimmunotherapy (CIT), for CLL treatment. The burden was assessed/projected from 2011 to 2025. For the OTT scenario, CIT was considered the standard of care before 2015, while OTT was considered standard of care for patients with either unmutated immunoglobulin heavy-chain variable (IGHV) or del(17p)/TP53 mutations starting in 2015 and, from 2020 onwards, for all first-line treatments except for patients with mutated IGHV. A Markov model was developed including four health states: watchful-waiting, first-line treatment, relapse and death. Costs of therapy, follow-up/monitoring and adverse events were included. Key clinical parameters were extracted from pivotal clinical trials. Results: As incidence rates and rate of survival are increasing, the prevalence of CLL in Canada is projected to increase 1.8-fold, from 8301 patients in 2011 to 14,654 by 2025. Correspondingly, the total annual costs of CLL management are predicted to increase 15.7-fold, from $60.8 million to $957.5 million during that same period. Conclusions: Although OTT enhances survival for patients with CLL, it is nonetheless associated with an important economic burden due to the projected vast increase in costs from 2011 to 2025. Changes in clinical strategies, such as implementation of a fixed OTT treatment duration, could help alleviate financial burden.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Administration, Oral , Cost of Illness , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Mutation
9.
FEBS J ; 288(7): 2084-2102, 2021 04.
Article in English | MEDLINE | ID: mdl-32780549

ABSTRACT

A nanobody (Nb) is a registered trademark of Ablynx, referring to the single antigen-binding domain of heavy chain-only antibodies (HCAbs) that are circulating in Camelidae. Nbs are produced recombinantly in micro-organisms and employed as research tools or for diagnostic and therapeutic applications. They were - and still are - also named single-domain antibodies (sdAbs) or variable domain of the heavy chain of HCAbs (VHH). A variety of methods are currently in use for the fast and efficient generation of target-specific Nbs. Such Nbs are produced at low cost and associate with high affinity to their cognate antigen. They are robust, strictly monomeric and easy to tailor into more complex entities to meet the requirements of their application. Here, we review the various sources and different strategies that have been developed to identify rapidly, target-specific Nbs. We further discuss a variety of engineering technologies that have been explored to broaden the application range of Nbs and summarise those applications where designed Nbs might offer a marked advantage over other affinity reagents.


Subject(s)
Immunoglobulin Heavy Chains/immunology , Single-Domain Antibodies/immunology , Animals , Camelidae/immunology , Humans , Immunoglobulin Heavy Chains/therapeutic use , Single-Domain Antibodies/therapeutic use
10.
Parasite Immunol ; 42(9): e12718, 2020 09.
Article in English | MEDLINE | ID: mdl-32249437

ABSTRACT

AIM: To characterize several anti-Leishmania tropica nanobodies and to investigate their effect on Leishmania infection. METHODS: Several immunological tests were implied to characterize five different (as confirmed by sequencing) anti-L tropica nanobodies (NbLt05, NbLt06, NbLt14, NbLt24 and NbLt36) against parasite lysates or intact cells from different stages, promastigotes and amastigotes. Direct inhibitory effect of these nanobodies on parasite infection cycle on macrophages was tested in cell culture. RESULTS: All the five nanobodies (with distinguished characteristics) were more specific to L tropica than to L major, but could equally recognize the lysate and the outer surface of the intact cells from the two main stages of the parasite. Nanobodies recognized several leishmania antigens (majorly between 75 and 63 kDa), and their proteinaceous nature was confirmed. Because of its role in leishmania life cycle, gp63 was considered a potential antigen candidate for nanobodies, and bioinformatics predicted such interaction. All nanobodies have a negative effect on the infectivity of L tropica, as they decreased the number of infected macrophages and the amastigotes inside those macrophages. CONCLUSION: Such anti-leishmania nanobodies, with outstanding characteristics and important target, can be of great use in the development of promising treatment strategies against leishmaniasis.


Subject(s)
Camelus/immunology , Immunoglobulin Heavy Chains/therapeutic use , Leishmania tropica , Leishmaniasis/therapy , Single-Domain Antibodies/therapeutic use , Animals , Cells, Cultured , Humans , Immunoglobulin Heavy Chains/immunology , Leishmania tropica/drug effects , Leishmania tropica/growth & development , Leishmania tropica/immunology , Leishmaniasis/immunology , Life Cycle Stages , Macrophages/immunology , Macrophages/parasitology , Single-Domain Antibodies/immunology
11.
Theranostics ; 10(6): 2645-2658, 2020.
Article in English | MEDLINE | ID: mdl-32194826

ABSTRACT

Rationale: CD38 is a target for the therapy of multiple myeloma (MM) with monoclonal antibodies such as daratumumab and isatuximab. Since MM patients exhibit a high rate of relapse, the development of new biologics targeting alternative CD38 epitopes is desirable. The discovery of single-domain antibodies (nanobodies) has opened the way for a new generation of antitumor therapeutics. We report the generation of nanobody-based humanized IgG1 heavy chain antibodies (hcAbs) with a high specificity and affinity that recognize three different and non-overlapping epitopes of CD38 and compare their cytotoxicity against CD38-expressing hematological cancer cells in vitro, ex vivo and in vivo. Methods: We generated three humanized hcAbs (WF211-hcAb, MU1067-hcAb, JK36-hcAb) that recognize three different non-overlapping epitopes (E1, E2, E3) of CD38 by fusion of llama-derived nanobodies to the hinge- and Fc-domains of human IgG1. WF211-hcAb shares the binding epitope E1 with daratumumab. We compared the capacity of these CD38-specific hcAbs and daratumumab to induce CDC and ADCC in CD38-expressing tumor cell lines in vitro and in patient MM cells ex vivo as well as effects on xenograft tumor growth and survival in vivo. Results: CD38-specific heavy chain antibodies (WF211-hcAb, MU1067-hcAb, JK36-hcAb) potently induced antibody-dependent cellular cytotoxicity (ADCC) in CD38-expressing tumor cell lines and in primary patient MM cells, but only little if any complement-dependent cytotoxicity (CDC). In vivo, CD38-specific heavy chain antibodies significantly reduced the growth of systemic lymphomas and prolonged survival of tumor bearing SCID mice. Conclusions: CD38-specific nanobody-based humanized IgG1 heavy chain antibodies mediate cytotoxicity against CD38-expressing hematological cancer cells in vitro, ex vivo and in vivo. These promising results of our study indicate that CD38-specific hcAbs warrant further clinical development as therapeutics for multiple myeloma and other hematological malignancies.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Hematologic Neoplasms/drug therapy , Immunoglobulin G/therapeutic use , Immunoglobulin Heavy Chains/therapeutic use , Membrane Glycoproteins/immunology , Multiple Myeloma/drug therapy , Single-Domain Antibodies/therapeutic use , Aged , Animals , Cell Line, Tumor , Epitopes/immunology , Female , Humans , Male , Mice , Mice, SCID , Middle Aged
12.
J Allergy Clin Immunol ; 144(2): 514-523.e5, 2019 08.
Article in English | MEDLINE | ID: mdl-30995457

ABSTRACT

BACKGROUND: Variable heavy chain (VH) family frameworks (FWRs) have been reported to affect antibody receptor and superantigen binding; however, such effects in IgE remain largely unknown. Given that VH family biases have been previously reported in IgE of certain allergies, there is a need to investigate this phenomenon for biotechnological and therapeutic purposes. OBJECTIVE: We sought to investigate the effects of VH families on IgE interaction with FcεRIα, anti-IgE omalizumab, antigen, and superantigen protein A (spA) by using the pertuzumab and trastuzumab IgE models. METHODS: Pertuzumab VH1-VH7 family variants of IgE with the same complementarity-determining regions were investigated with regard to their binding interactions to FcεRIα, Her2, omalizumab, and spA. Notable FcεRIα-IgE observations were cross-checked against appropriate trastuzumab IgE VH variants. Computational structural modeling and simulations were also performed for insight into the mechanism of interactions with various VH FWRs. RESULTS: The pertuzumab VH5 IgE variant, but not the trastuzumab VH5 IgE, was found to interact with FcεRIα significantly longer than the respective VH family variants within each model antibody. No significant differences in interaction were found between IgE and omalizumab for the pertuzumab VH variants. Although trastuzumab VH3 interacted with spA, none of our pertuzumab VH variants, including VH3, associated with spA. CONCLUSION: We found unexpected varying allosteric communications caused by the VH family FWRs to the FcεRIα-, Her2-, and spA-binding regions of pertuzumab IgE, with implications for use of IgE/anti-IgE therapeutics to treat allergy and IgE therapeutics in allergo-oncology.


Subject(s)
Antigens, Bacterial/chemistry , Immunoglobulin E/chemistry , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/immunology , Receptors, IgE/chemistry , Superantigens/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigens, Bacterial/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunoglobulin E/immunology , Immunoglobulin E/therapeutic use , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/therapeutic use , Immunotherapy , Omalizumab/chemistry , Omalizumab/immunology , Receptors, IgE/immunology , Superantigens/immunology , Trastuzumab/chemistry , Trastuzumab/immunology
13.
Front Immunol ; 9: 2559, 2018.
Article in English | MEDLINE | ID: mdl-30459772

ABSTRACT

The NAD+-metabolizing ectoenzyme CD38 is an established therapeutic target in multiple myeloma. The CD38-specific monoclonal antibodies daratumumab and isatuximab show promising results in the clinic. Nanobodies correspond to the single variable domains (VHH) derived from heavy chain antibodies that naturally occur in camelids. VHHs display high solubility and excellent tissue penetration in vivo. We recently generated a panel of CD38-specific nanobodies, some of which block or enhance the enzymatic activity of CD38. Fusion of such a nanobody to the hinge, CH2, and CH3 domains of human IgG1 generates a chimeric llama/human hcAb of about half the size of a conventional moAb (75 vs. 150 kDa). Similarly, a fully human CD38-specific hcAb can be generated using a CD38-specific human VH3 instead of a CD38-specific camelid nanobody. Here we discuss the advantages and disadvantages of CD38-specific hcAbs vs. conventional moAbs and provide an outlook for the potential use of CD38-specific hcAbs as novel therapeutics for multiple myeloma.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antigens, Neoplasm/immunology , Immunoglobulin G/therapeutic use , Immunoglobulin Heavy Chains/therapeutic use , Immunotherapy/methods , Multiple Myeloma/therapy , Single-Domain Antibodies/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Camelidae/immunology , Humans , Multiple Myeloma/immunology
14.
Int Rev Immunol ; 37(1): 69-76, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29182399

ABSTRACT

Camelid heavy-chain variable domains (VHHs) are the smallest, intact, antigen-binding units to occur in nature. VHHs possess high degrees of solubility and robustness enabling generation of multivalent constructs with increased avidity - characteristics that mark their superiority to other antibody fragments and monoclonal antibodies. Capable of effectively binding to molecular targets inaccessible to classical immunotherapeutic agents and easily produced in microbial culture, VHHs are considered promising tools for pharmaceutical biotechnology. With the aim to demonstrate the perspective and potential of VHHs for the development of prophylactic and therapeutic drugs to target diseases caused by bacterial and viral infections, this review article will initially describe the structural features that underlie the unique properties of VHHs and explain the methods currently used for the selection and recombinant production of pathogen-specific VHHs, and then thoroughly summarize the experimental findings of five distinct studies that employed VHHs as inhibitors of host-pathogen interactions or neutralizers of infectious agents. Past and recent studies suggest the potential of camelid heavy-chain variable domains as a novel modality of immunotherapeutic drugs and a promising alternative to monoclonal antibodies. VHHs demonstrate the ability to interfere with bacterial pathogenesis by preventing adhesion to host tissue and sequestering disease-causing bacterial toxins. To protect from viral infections, VHHs may be employed as inhibitors of viral entry by binding to viral coat proteins or blocking interactions with cell-surface receptors. The implementation of VHHs as immunotherapeutic agents for infectious diseases is of considerable potential and set to contribute to public health in the near future.


Subject(s)
Bacterial Infections/prevention & control , Camelidae/immunology , Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Variable Region/therapeutic use , Virus Diseases/prevention & control , Animals , Bacterial Infections/immunology , Bacterial Infections/therapy , Camelidae/genetics , Dental Caries/microbiology , Dental Caries/therapy , Diarrhea/microbiology , Diarrhea/prevention & control , Escherichia coli Infections/prevention & control , Humans , Immunization, Passive , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Poliomyelitis/prevention & control , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Respiratory Syncytial Virus Infections/therapy , Streptococcus mutans/immunology , Virus Diseases/immunology , Virus Diseases/therapy
15.
PLoS One ; 11(9): e0162351, 2016.
Article in English | MEDLINE | ID: mdl-27603013

ABSTRACT

Species A Rotaviruses (RVA) remain a leading cause of mortality in children under 5 years of age. Current treatment options are limited. We assessed the efficacy of two VP6-specific llama-derived heavy chain antibody fragments (VHH) -2KD1 and 3B2- as an oral prophylactic and therapeutic treatment against RVA-induced diarrhea in a neonatal mouse model inoculated with virulent murine RVA (ECw, G16P[16]I7). Joint therapeutic administration of 2KD1+3B2 (200 µg/dose) successfully reduced diarrhea duration, RVA infection severity and virus shedding in feces. While the same dose of 2KD1 or 3B2 (200 µg) significantly reduced duration of RVA-induced diarrhea, 2KD1 was more effective in diminishing the severity of intestinal infection and RVA shedding in feces, perhaps because 2KD1 presented higher binding affinity for RVA particles than 3B2. Neither prophylactic nor therapeutic administration of the VHH interfered with the host's humoral immune response against RVA. When 2KD1 (200 µg) was administered after diarrhea development, it also significantly reduced RVA intestinal infection and fecal shedding. Host antibody responses against the oral VHH treatment were not detected, nor did viral escape mutants. Our findings show that oral administration of anti-VP6 VHH constitute, not only an effective prophylactic treatment against RVA-associated diarrhea, but also a safe therapeutic tool against RVA infection, even once diarrhea is present. Anti-VP6 VHH could be used complementary to ongoing vaccination, especially in populations that have shown lower immunization efficacy. These VHH could also be scaled-up to develop pediatric medication or functional food like infant milk formulas that might help treat RVA diarrhea.


Subject(s)
Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Variable Region/therapeutic use , Rotavirus Infections/drug therapy , Rotavirus Infections/virology , Rotavirus/physiology , Animals , Animals, Newborn , Camelids, New World , Diarrhea/drug therapy , Diarrhea/virology , Feces/virology , Hydrogen-Ion Concentration , Immunity, Humoral/immunology , Immunoglobulin Heavy Chains/administration & dosage , Immunoglobulin Variable Region/administration & dosage , Intestines/pathology , Intestines/virology , Mice, Inbred BALB C , Mutation/genetics , Phylogeny , Proteolysis , Rotavirus Infections/immunology , Virion/metabolism , Virus Shedding
16.
Mol Pharm ; 12(6): 2151-7, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25955255

ABSTRACT

Glypican-3 (GPC3) represents an attractive target for hepatocellular carcinoma (HCC) therapy because it is highly expressed in HCC but not in adult normal tissue. Recently, high affinity anti-GPC3 antibodies have been developed; however, full antibodies may not penetrate evenly into tumor parenchyma, reducing their effectiveness. In this study, we compared a whole IgG antibody, anti-GPC3 YP7, with an anti-GPC3 human heavy chain antibody, HN3, with regard to their relative therapeutic effects. Both YP7 and HN3 bound to GPC3-positive A431/G1 cells and were internalized by the cells by in vitro evaluation with (125)I- and (111)In-radiolabeling antibodies. In vivo biodistribution and tumor accumulation was performed with (111)In-labeled antibodies, and intratumoral microdistribution was evaluated using fluorescently labeled antibodies (IR700). HN3 showed similar high tumor accumulation but superior homogeneity within the tumor compared with YP7. Using the same IR700 conjugated antibodies photoimmunotherapy (PIT) was performed in vitro and in a tumor-bearing mouse model in vivo. PIT with IR700-HN3 and IR700-YP7 demonstrated that comparable results could be achieved despite of low reaccumulation 24 h after the first NIR light exposure. These results indicated that a heavy-chain antibody, HN3, showed more favorable characteristics than YP7, a conventional IgG, as a therapeutic antibody platform for designing molecularly targeted agents against HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Glypicans/immunology , Immunoglobulin Heavy Chains/therapeutic use , Liver Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Drug Carriers/chemistry , Female , Humans , Immunoglobulin Heavy Chains/immunology , Immunotherapy , Liver Neoplasms/immunology , Mice
17.
Biochimie ; 111: 82-106, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25656912

ABSTRACT

The deposition of misfolded peptides and proteins in the form of amyloid fibrils is the hallmark of nearly fifty medical disorders, including Alzheimer's disease, Parkinson's disease, prion diseases and type II diabetes. These disorders, referred to as amyloidoses, generally become apparent late in life. Their psycho-sociological and economic incidence in western societies will be therefore considerable in the coming decades due to the ageing of the population. Neither preventing nor curative treatments are available yet. These disorders constitute therefore a medical challenge of great importance. Thus, an extensive research is being carried out to understand, at the molecular level, (i) how amyloidogenic proteins misfold and convert from their soluble form into amyloid fibrils, and (ii) how these aggregates or some of their oligomeric precursor species are toxic. The formation of amyloid fibrils proceeds through a complex nucleation/polymerisation mechanism with the formation of various species, including small oligomers. In this review, we focus on how VHHs or nanobodies, the antigen-binding domains of camelid heavy-chain antibodies, are being increasingly used to characterise each of the species formed on the pathway of fibril formation in terms of structure, stability, kinetics of formation and toxicity. We first introduce the characteristic features of nanobodies compared to those of conventional antibody fragments. Thereafter, we discuss how nanobodies, due to their unique properties, are used as probes to dissect the molecular mechanisms of misfolding and aggregation of six proteins associated with diseases, i.e. human lysozyme, ß2-microglobulin, α-synuclein, prion, polyadenylate binding protein nuclear 1 and amyloid ß-peptide. A brief general presentation of each disease and the associated peptide/protein is also provided. In addition, we discuss how nanobodies could be used as early diagnostic tools and as novel strategies to treat diseases associated with protein misfolding and aggregation.


Subject(s)
Camelids, New World/immunology , Immunoglobulin Heavy Chains/therapeutic use , Protein Aggregation, Pathological/drug therapy , Proteostasis Deficiencies/drug therapy , Single-Domain Antibodies/therapeutic use , Animals , Camelids, New World/genetics , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Protein Aggregation, Pathological/immunology , Proteostasis Deficiencies/immunology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology
18.
J Control Release ; 203: 40-50, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25668771

ABSTRACT

Treatment of neurodegenerative disorders such as Alzheimer's disease is hampered by the blood-brain barrier (BBB). This tight cerebral vascular endothelium regulates selective diffusion and active transport of endogenous molecules and xenobiotics into and out of the brain parenchyma. In this study, glutathione targeted PEGylated (GSH-PEG) liposomes were designed to deliver amyloid-targeting antibody fragments across the BBB into the brain. Two different formulations of GSH-PEG liposomes based on 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) and egg-yolk phosphatidylcholine (EYPC) were produced. Both formulations encapsulate 15kDa amyloid beta binding llama single domain antibody fragments (VHH-pa2H). To follow the biodistribution of VHH-pa2H rather than the liposome, the antibody fragment was labeled with the radioisotope indium-111. To prolong the shelf life of the construct beyond the limit of radioactive decay, an active-loading method was developed to efficiently radiolabel the antibody fragments after encapsulation into the liposomes, with radiolabeling efficiencies of up to 68% after purification. The radiolabeled liposomes were administered via a single intravenous bolus injection to APPswe/PS1dE9 double transgenic mice, a mouse model of Alzheimer's disease, and their wildtype littermates. Both GSH-PEG DMPC and GSH-PEG EYPC liposomes significantly increased the standard uptake values (SUV) of VHH-pa2H in the blood of the animals compared to free VHH-pa2H. Encapsulation in GSH-PEG EYPC liposomes resulted in the highest increase in SUV in the brains of transgenic animals. Overall, these data provide evidence that GSH-PEG liposomes may be suitable for specific delivery of single domain antibody fragments over the BBB into the brain.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/antagonists & inhibitors , Brain/metabolism , Glutathione/metabolism , Liposomes/metabolism , Single-Chain Antibodies/administration & dosage , Alzheimer Disease/metabolism , Animals , Blood-Brain Barrier/metabolism , Camelids, New World , Disease Models, Animal , Drug Delivery Systems , Humans , Immunoglobulin Heavy Chains/administration & dosage , Immunoglobulin Heavy Chains/therapeutic use , Mice , Mice, Transgenic , Polyethylene Glycols/metabolism , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/therapeutic use , Tissue Distribution
19.
Nucl Med Biol ; 41 Suppl: e44-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24530366

ABSTRACT

INTRODUCTION: Small domain antibodies (sdAbs) present high potential for both molecular in vivo imaging and therapy. Owing to the low molecular weight they are rapidly cleared from blood circulation, and new strategies to extend their half-lifes are needed for therapeutic applications. We have selected a bacterial albumin-binding domain (ABD) from protein Zag to be fused to an anti-tumor necrosis factor (TNF) single variable-domain heavy-chain region antibody (VHH) to delay blood clearance, and evaluated the biodistribution profile of the fusion protein. METHODS: The anti-TNF VHH and the fusion protein VHH-Zag were conjugated to S-2-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA). The anti-TNF and albumin-binding properties of the conjugates NOTA-VHH and NOTA-VHH-Zag were assessed by enzyme-linked immunosorbent assay (ELISA). The radioconjugates (67)Ga-NOTA-VHH and (67)Ga-NOTA-VHH-Zag were obtained by reaction of (67)GaCl3 with the corresponding conjugates at room temperature. Biodistribution studies were performed in healthy female CD-1 mice. RESULTS: The immunoreactivity of the VHH-based proteins is preserved upon conjugation to NOTA as well as after radiometallation. The radiochemical purity of the radioconjugates was higher than 95% as determined by ITLC-SG after purification by gel filtration. The biodistribution studies showed that the Zag domain affected the pharmacokinetic properties of VHH, with impressive differences in blood clearance (0.028 ± 0.004 vs 1.7 ± 0.8 % I.A./g) and total excretion (97.8 ± 0.6 vs 25.5 ± 2.1 % I.A.) for (67)Ga-NOTA-VHH and (67)Ga-NOTA-VHH-Zag, respectively, at 24h p.i. CONCLUSION: The Zag domain prolonged the circulation time of VHH by reducing the blood clearance of the labeled fusion protein (67)Ga-NOTA-VHH-Zag. In this way, the anti-TNF VHH in fusion with the Zag ABD presents a higher therapeutic potential than the unmodified VHH.


Subject(s)
Bacterial Proteins/genetics , Immunoglobulin Heavy Chains/therapeutic use , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Single-Domain Antibodies/therapeutic use , Tumor Necrosis Factor-alpha/immunology , Animals , Female , Gallium Radioisotopes/therapeutic use , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Tissue Distribution
20.
PLoS One ; 9(12): e115893, 2014.
Article in English | MEDLINE | ID: mdl-25551637

ABSTRACT

Trypanosomes are protozoan parasites that cause diseases in humans and livestock for which no vaccines are available. Disease eradication requires sensitive diagnostic tools and efficient treatment strategies. Immunodiagnostics based on antigen detection are preferable to antibody detection because the latter cannot differentiate between active infection and cure. Classical monoclonal antibodies are inaccessible to cryptic epitopes (based on their size-150 kDa), costly to produce and require cold chain maintenance, a condition that is difficult to achieve in trypanosomiasis endemic regions, which are mostly rural. Nanobodies are recombinant, heat-stable, small-sized (15 kDa), antigen-specific, single-domain, variable fragments derived from heavy chain-only antibodies in camelids. Because of numerous advantages over classical antibodies, we investigated the use of nanobodies for the targeting of trypanosome-specific antigens and diagnostic potential. An alpaca was immunized using lysates of Trypanosoma evansi. Using phage display and bio-panning techniques, a cross-reactive nanobody (Nb392) targeting all trypanosome species and isolates tested was selected. Imunoblotting, immunofluorescence microscopy, immunoprecipitation and mass spectrometry assays were combined to identify the target recognized. Nb392 targets paraflagellar rod protein (PFR1) of T. evansi, T. brucei, T. congolense and T. vivax. Two different RNAi mutants with defective PFR assembly (PFR2RNAi and KIF9BRNAi) were used to confirm its specificity. In conclusion, using a complex protein mixture for alpaca immunization, we generated a highly specific nanobody (Nb392) that targets a conserved trypanosome protein, i.e., PFR1 in the flagella of trypanosomes. Nb392 is an excellent marker for the PFR and can be useful in the diagnosis of trypanosomiasis. In addition, as demonstrated, Nb392 can be a useful research or PFR protein isolation tool.


Subject(s)
Antigens, Protozoan/immunology , Protozoan Proteins/immunology , Single-Chain Antibodies/therapeutic use , Trypanocidal Agents/therapeutic use , Trypanosomiasis/prevention & control , Amino Acid Sequence , Animals , Camelids, New World , Flagella/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/therapeutic use , Molecular Sequence Data , RNA Interference , RNA, Small Interfering , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Sequence Alignment , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Trypanocidal Agents/immunology , Trypanosoma/immunology , Trypanosomiasis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...