1
|
Etemadifar M, Nouri H, Sedaghat N, Ramezani A, Kargaran PK, Salari M, Kaveyee H. Anti-CD20 therapies for pediatric-onset multiple sclerosis: A systematic review. Mult Scler Relat Disord 2024; 91:105849. [PMID: 39243503 DOI: 10.1016/j.msard.2024.105849] [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: 02/04/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Pediatric-onset multiple sclerosis (POMS) cases, defined as multiple sclerosis (MS) with onset before the age of 18, represent between 3 and 5 % of all MS patients. Anti-CD20 drugs mainly rituximab, ocrelizumab, and ofatumumab are being widely used in adult-onset MS. Their use in POMS is also being increasingly considered by experts. OBJECTIVE to review the latest evidence on safety and efficacy of the use of anti-CD20 therapies in POMS. METHODS An extensive search was performed in PubMed, Scopus, and Web of Science databases until the end of July 1st, 2024. Two independent reviewers screened the articles, and collected data. 832 studies were screened using Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. RESULTS 12 studies on rituximab (328 patients) and 6 studies on ocrelizumab (106 patients) were synthesized. Using monoclonal antibodies in POMS patients has a noteworthy effect on reducing relapses and lesions and achieving no evidence of disease activity especially in highly active POMS patients. However, anti-CD20 therapies in MS are associated with potential adverse events (AEs). Additional data is required on the effect of anti-CD20 therapy on disability accrual. CONCLUSION Although anti-CD20 therapy is associated with some AEs, it can be provided in several circumstances, especially to patients with highly active disease, or ones resistant to platform therapies.
Collapse
Affiliation(s)
- Masoud Etemadifar
- Department of Neurosurgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hosein Nouri
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Nahad Sedaghat
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Aryana Ramezani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parisa K Kargaran
- Department of Cardiovascular Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, Rochester, MN, USA
| | - Mehri Salari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hasan Kaveyee
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Rhomberg-Kauert J, Karlsson M, Thiagarajan D, Kallas T, Karlsson F, Fredriksson S, Dahlberg J, Martinez Barrio A. Using adjusted local assortativity with Molecular Pixelation unveils colocalization of membrane proteins with immunological significance. Front Immunol 2024; 15:1309916. [PMID: 38983848 PMCID: PMC11231075 DOI: 10.3389/fimmu.2024.1309916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/09/2024] [Indexed: 07/11/2024] Open
Abstract
Advances in spatial proteomics and protein colocalization are a driving force in the understanding of cellular mechanisms and their influence on biological processes. New methods in the field of spatial proteomics call for the development of algorithms and open up new avenues of research. The newly introduced Molecular Pixelation (MPX) provides spatial information on surface proteins and their relationship with each other in single cells. This allows for in silico representation of neighborhoods of membrane proteins as graphs. In order to analyze this new data modality, we adapted local assortativity in networks of MPX single-cell graphs and created a method that is able to capture detailed information on the spatial relationships of proteins. The introduced method can evaluate the pairwise colocalization of proteins and access higher-order similarity to investigate the colocalization of multiple proteins at the same time. We evaluated the method using publicly available MPX datasets where T cells were treated with a chemokine to study uropod formation. We demonstrate that adjusted local assortativity detects the effects of the stimuli at both single- and multiple-marker levels, which enhances our understanding of the uropod formation. We also applied our method to treating cancerous B-cell lines using a therapeutic antibody. With the adjusted local assortativity, we recapitulated the effect of rituximab on the polarity of CD20. Our computational method together with MPX improves our understanding of not only the formation of cell polarity and protein colocalization under stimuli but also advancing the overall insight into immune reaction and reorganization of cell surface proteins, which in turn allows the design of novel therapies. We foresee its applicability to other types of biological spatial data when represented as undirected graphs.
Collapse
Affiliation(s)
- Jan Rhomberg-Kauert
- Pixelgen Technologies AB, Stockholm, Sweden
- Department of Geodesy and Geoinformation, TU Wien, Vienna, Austria
| | | | | | | | | | - Simon Fredriksson
- Pixelgen Technologies AB, Stockholm, Sweden
- Department of Protein Science, Royal Institute of Technology, Stockholm, Sweden
| | | | | |
Collapse
|
3
|
Yang M, Wang S, Zhang J, Yan B. Primary Sjogren syndrome - A bibliometric analysis via CiteSpace. Medicine (Baltimore) 2024; 103:e38162. [PMID: 38875384 PMCID: PMC11175928 DOI: 10.1097/md.0000000000038162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/16/2024] Open
Abstract
This study employs CiteSpace software to analyze the research status, hotspots, and trends of primary Sjogren syndrome (pSS). Relevant publications from 1999 to 2023 were searched in the Web of Science Core Collection (WoSCC) set, followed by generating a network map using CiteSpace software to identify top authors, institutions, countries, keywords, journals, references, and research trends. A total of 3564 valid articles were included in this study. The People Republic of China had the highest number of articles (n = 524), while the University of Bergen emerged as the institution with the highest publication count (n = 94). Mariette X was identified as the author with the most publications (n = 67), whereas Vitali C received recognition as the most cited author (n = 1706). Annals of Rheumatic Diseases stood out as the journal with the highest citation count (n = 2530). Notably, an article published in the Annals of Rheumatic Diseases in 2017 garnered significant attention by being cited a remarkable 304 times. The bibliometric analysis reveals that key areas of research in pSS encompass investigating pathogenesis; advancing and applying targeted biological agents; and establishing treatment and diagnostic standards.
Collapse
Affiliation(s)
- Mingrui Yang
- School of pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Shangzhi Wang
- School of pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Jin Zhang
- School of pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Bin Yan
- School of Traditional of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| |
Collapse
|
4
|
Li XX, Maitiyaer M, Tan Q, Huang WH, Liu Y, Liu ZP, Wen YQ, Zheng Y, Chen X, Chen RL, Tao Y, Yu SL. Emerging biologic frontiers for Sjogren's syndrome: Unveiling novel approaches with emphasis on extra glandular pathology. Front Pharmacol 2024; 15:1377055. [PMID: 38828450 PMCID: PMC11140030 DOI: 10.3389/fphar.2024.1377055] [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: 01/26/2024] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Primary Sjögren's Syndrome (pSS) is a complex autoimmune disorder characterized by exocrine gland dysfunction, leading to dry eyes and mouth. Despite growing interest in biologic therapies for pSS, FDA approval has proven challenging due to trial complications. This review addresses the absence of a molecular-target-based approach to biologic therapy development and highlights novel research on drug targets and clinical trials. A literature search identified potential pSS treatment targets and recent advances in molecular understanding. Overlooking extraglandular symptoms like fatigue and depression is a notable gap in trials. Emerging biologic agents targeting cytokines, signal pathways, and immune responses have proven efficacy. These novel therapies could complement existing methods for symptom alleviation. Improved grading systems accounting for extraglandular symptoms are needed. The future of pSS treatment may involve gene, stem-cell, and tissue-engineering therapies. This narrative review offers insights into advancing pSS management through innovative biologic interventions.
Collapse
Affiliation(s)
- Xiao Xiao Li
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Maierhaba Maitiyaer
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Qing Tan
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Wen Hui Huang
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yu Liu
- Department of Clinical Medicine, The First Clinical Medical School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhi Ping Liu
- Ophthalmic Center, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yue Qiang Wen
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yu Zheng
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xing Chen
- Department of Geriatrics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Rui Lin Chen
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yi Tao
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shui Lian Yu
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Ding Y, Guo N, Jiang Y, Liu S, Zhou T, Bai H, Lv Y, Han S, He L. Establishment of cluster of differentiation 20 immobilized cell membrane chromatography for the screening of active antitumor components in traditional Chinese medicine. J Chromatogr A 2024; 1721:464845. [PMID: 38552371 DOI: 10.1016/j.chroma.2024.464845] [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: 02/17/2024] [Revised: 03/19/2024] [Accepted: 03/23/2024] [Indexed: 04/13/2024]
Abstract
Non-Hodgkin lymphoma (NHL) is a heterogeneous group of malignant tumors occurring in B or T lymphocytes, and no small molecule-positive drugs to treat NHL have been marketed. Cluster of differentiation 20 (CD20) is an important molecule regulating signaling for the life and differentiation of B lymphocytes and possesses the characteristics of a drug target for treating NHL. 2-Methoxyestradiol induces apoptosis in lymphoma Raji cells and CD20 protein is highly expressed by Raji lymphoma cells. Therefore, in this study, a CD20-SNAP-tag/CMC model was developed to validate the interaction of 2-methoxyestradiol with CD20. 2-Methoxyestradiol was used as a small molecule control compound, and the system was validated for good applicability. The cell membrane chromatography model was combined with high-performance liquid chromatography ion trap time-of-flight mass spectroscopy (HPLC-IT-TOF-MS) in a two-dimensional system to successfully identify, analyze, and characterize the potential active compounds of Schisandra chinensis (Turcz.) Baill. extract and Lysionotus pauciflorus Maxim. extract, including Schisandrin A, Schizandrol A, Schizandrol B, Schisantherin B, and Nevadensin, which can act on CD20 receptors. The five potential active compounds were analyzed by non-linear chromatography. The thermodynamic and kinetic parameters of their interaction with CD20 were also analyzed, and the mode of interaction was simulated by molecular docking. Their inhibitory effects on lymphoma cell growth were assessed using a Cell Counting Kit-8 (CCK-8). Nevadensin and Schizandrin A were able to induce apoptosis in Raji cells within a certain concentration range. In conclusion, the present experiments provide some bases for improving NHL treatment and developing small molecule lead compounds targeting CD20 with low toxicity and high specificity.
Collapse
Affiliation(s)
- Yifan Ding
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Na Guo
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Yuhan Jiang
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Sihan Liu
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Tongpei Zhou
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Haoyun Bai
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Yanni Lv
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China
| | - Shengli Han
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China.
| | - Langchong He
- School of Pharmacy, Xi'an Jiaotong University, 76# Yanta West Road, Xi'an 710061, PR China; Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an 710115, PR China.
| |
Collapse
|
6
|
Hayama M, Riches JC. Taking the Next Step in Double Refractory Disease: Current and Future Treatment Strategies for Chronic Lymphocytic Leukemia. Onco Targets Ther 2024; 17:181-198. [PMID: 38476308 PMCID: PMC10929554 DOI: 10.2147/ott.s443924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a monoclonal B-cell lymphoproliferative disease with a high annual incidence in Western countries. As B-cell receptor (BCR) signaling and intrinsic apoptotic resistance play critical roles in the development and survival of CLL cells, therapeutic approaches targeting these pathways have been extensively investigated to tackle this incurable disease. Over the last decade, several Phase 3 trials have confirmed the superior efficacy of covalent Bruton tyrosine kinase inhibitors (cBTKis) and venetoclax, a selective B-cell lymphoma 2 (BCL2) inhibitor, over chemoimmunotherapy. This has been demonstrated in both the treatment-naïve and relapsed/refractory (RR) settings and includes patients with high-risk molecular features. However, these drugs are not curative, with patients continuing to relapse after treatment with both cBTKis and BCL2is, and the optimal treatment strategy for these patients has not been defined. Several novel agents with distinct mechanisms have recently been developed for CLL which have demonstrated efficacy in patients who have previously received cBTKis and BCL2i. In particular, novel BCR-signaling targeting agents have shown promising efficacy in early-phase clinical trials for RR-CLL. Furthermore, cancer immunotherapies such as bispecific antibodies and chimeric antigen receptor T-cells have also shown anti-tumor activity in patients with heavily pretreated RR-CLL. Personalised approaches with these novel agents and combination strategies based on the understanding of resistance mechanisms have the potential to overcome the clinical challenge of what to do next for a patient who has already had a cBTKi and venetoclax.
Collapse
Affiliation(s)
- Manabu Hayama
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - John C Riches
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
7
|
Schuster SJ, Huw LY, Bolen CR, Maximov V, Polson AG, Hatzi K, Lasater EA, Assouline SE, Bartlett NL, Budde LE, Matasar MJ, Koeppen H, Piccione EC, Wilson D, Wei MC, Yin S, Penuel E. Loss of CD20 expression as a mechanism of resistance to mosunetuzumab in relapsed/refractory B-cell lymphomas. Blood 2024; 143:822-832. [PMID: 38048694 PMCID: PMC10934296 DOI: 10.1182/blood.2023022348] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/24/2023] [Accepted: 11/15/2023] [Indexed: 12/06/2023] Open
Abstract
ABSTRACT CD20 is an established therapeutic target in B-cell malignancies. The CD20 × CD3 bispecific antibody mosunetuzumab has significant efficacy in B-cell non-Hodgkin lymphomas (NHLs). Because target antigen loss is a recognized mechanism of resistance, we evaluated CD20 expression relative to clinical response in patients with relapsed and/or refractory NHL in the phase 1/2 GO29781 trial investigating mosunetuzumab monotherapy. CD20 was studied using immunohistochemistry (IHC), RNA sequencing, and whole-exome sequencing performed centrally in biopsy specimens collected before treatment at predose, during treatment, or upon progression. Before treatment, most patients exhibited a high proportion of tumor cells expressing CD20; however, in 16 of 293 patients (5.5%) the proportion was <10%. Analyses of paired biopsy specimens from patients on treatment revealed that CD20 levels were maintained in 29 of 30 patients (97%) vs at progression, where CD20 loss was observed in 11 of 32 patients (34%). Reduced transcription or acquisition of truncating mutations explained most but not all cases of CD20 loss. In vitro modeling confirmed the effects of CD20 variants identified in clinical samples on reduction of CD20 expression and missense mutations in the extracellular domain that could block mosunetuzumab binding. This study expands the knowledge about the occurrence of target antigen loss after anti-CD20 therapeutics to include CD20-targeting bispecific antibodies and elucidates mechanisms of reduced CD20 expression at disease progression that may be generalizable to other anti-CD20 targeting agents. These results also confirm the utility of readily available IHC staining for CD20 as a tool to inform clinical decisions. This trial was registered at www.ClinicalTrials.gov as #NCT02500407.
Collapse
Affiliation(s)
- Stephen J. Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | | | | - Nancy L. Bartlett
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| | | | | | | | | | | | | | - Shen Yin
- Genentech, Inc., South San Francisco, CA
| | | |
Collapse
|
8
|
Komura K, Yanaba K, Bouaziz JD, Yoshizaki A, Hasegawa M, Varga J, Takehara K, Matsushita T. Perspective to precision medicine in scleroderma. Front Immunol 2024; 14:1298665. [PMID: 38304250 PMCID: PMC10830793 DOI: 10.3389/fimmu.2023.1298665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024] Open
Abstract
Systemic sclerosis (SSc) is a rare and heterogeneous disease with no relevant environmental trigger or significant responsible gene. It has been and will continue to be difficult to identify large enough patients to conduct classic population-based epidemiologic exposure/non-exposure studies with adequate power to ascertain environmental and genetic risk factors for these entities. The complexity of pathogenesis and heterogeneity are likely to require personalized/precision medicine for SSc. Since several potential drugs are currently available for specific patients if not whole SSc, classification of SSc seems to form the foundation for a better therapeutic strategy. To date, SSc has been classified based on the extent/severity of the affected area as well as some disease markers, including the autoantibody profile. However, such an analysis should also lead to improvements in the design of appropriately stratified clinical trials to determine the effects and prediction of targeted therapies. An approach based on drug response preclinically conducted using patients' own fibroblasts in vitro, can provide a precise disease marker/therapeutic selection for clinical practice. Because scleroderma dermal fibroblasts have a persistent hyper-productive phenotype occurring not only in person, but also in cell culture conditions. Thus, an accumulating approach based on disease markers ensures progression and de-escalation to re-establish a better life with a personally optimized drug environment after the onset of SSc.
Collapse
Affiliation(s)
- Kazuhiro Komura
- Department of Dermatology, Kanazawa Red Cross Hospital, Japanese Red Cross Society, Kanazawa, Japan
- Northwestern Scleroderma Program, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Koichi Yanaba
- Department of Dermatology, Jikei University, Tokyo, Japan
| | | | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | | | - John Varga
- Northwestern Scleroderma Program, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | | | | |
Collapse
|
9
|
Park SJ, Greer PL, Lee N. From odor to oncology: non-canonical odorant receptors in cancer. Oncogene 2024; 43:304-318. [PMID: 38087050 DOI: 10.1038/s41388-023-02908-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 01/31/2024]
Abstract
Odorant receptors, traditionally associated with olfaction as chemoreceptors, have been increasingly recognized for their presence and diverse functions in various non-nasal tissues throughout the body. Beyond their roles in sensory perception, emerging evidence suggests a compelling interplay between odorant receptors and cancer progression as well. Alongside the canonical GPCR odorant receptors, dysregulation of non-canonical odorant receptors such as trace amine-associated receptors (TAARs), formyl peptide receptors (FPRs), and membrane-spanning 4A family (MS4As) has been observed in various cancer types, suggesting their contributions to cancer progression. The roles of these non-canonical chemoreceptors in cancer are complex, with some receptors promoting tumorigenesis and others acting as tumor-suppressing factors upon activation, depending on the cancer type. These findings shed light on the potential of non-canonical odorant receptors as therapeutic targets and prognostic markers in cancer, inviting further exploration to unravel their precise mechanisms of action and implications in cancer biology. In this review, we provide a comprehensive overview of the intricate relationships between these chemoreceptors and various types of cancer, potentially paving the way for innovative odor-based therapeutics. Ultimately, this review discusses the potential development of novel therapeutic strategies targeting these non-canonical chemoreceptors.
Collapse
Affiliation(s)
- Sung Jin Park
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Paul L Greer
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Namgyu Lee
- Department of Biomedical Science and Engineering, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
10
|
Bar-Or A, Aburashed R, Chinea AR, Hendin BA, Lucassen E, Meng X, Stankiewicz J, Tullman MJ, Cross AH. Humoral immune response to COVID-19 mRNA vaccines in patients with relapsing multiple sclerosis treated with ofatumumab. Mult Scler Relat Disord 2023; 79:104967. [PMID: 37769429 DOI: 10.1016/j.msard.2023.104967] [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: 04/06/2023] [Revised: 08/03/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023]
Abstract
BACKGROUND There are limited data available regarding the impact of ofatumumab, an anti-CD20 B-cell-depleting monoclonal antibody for relapsing multiple sclerosis (RMS), on vaccination response. The study objective was to assess humoral immune response (HIR) to non-live coronavirus disease 2019 (COVID-19) messenger RNA (mRNA) vaccination in patients with RMS treated with ofatumumab. METHODS This was an open-label, single-arm, multicenter, prospective pilot study of patients with RMS aged 18-55 years who received 2 or 3 doses of a COVID-19 mRNA vaccine after ≥1 month of subcutaneous ofatumumab (20 mg/month) treatment. The primary endpoint was the proportion of patients achieving HIR, as defined by local laboratory severe acute respiratory syndrome coronavirus-2 qualitative immunoglobulin G assays. Assay No. 1 was ≥14 days after the second or third vaccine dose. Assay No. 2 was 90 days thereafter. RESULTS Of the 26 patients enrolled (median [range] age: 42 [27-54] years; median [range] ofatumumab treatment duration: 237 [50-364] days), HIR was achieved by 53.9% (14/26; 95% CI: 33.4 - 73.4%) at Assay No. 1 and 50.0% (13/26; 95% CI: 29.9 - 70.1%) at Assay No. 2. Patients who received 3 vaccine doses had higher HIR rates (Assay No. 1: 70.0% [7/10]; Assay No. 2: 77.8% [7/9]) than those who received 2 doses (Assay No. 1: 46.7% [7/15]; Assay No. 2: 42.9% [6/14]). Of patients aged <40 years without previous anti-CD20 therapy, HIR was achieved by 90.0% (9/10) at Assay No. 1 and 75.0% (6/8) at Assay No. 2. No serious adverse events were reported. CONCLUSION Patients with RMS treated with ofatumumab can mount HIRs following COVID-19 vaccination. A plain language summary, infographic and a short video summarizing the key results are provided in supplementary material. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT04847596 (https://clinicaltrials.gov/ct2/show/NCT04847596).
Collapse
Affiliation(s)
- Amit Bar-Or
- Department of Neurology, and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Rany Aburashed
- Insight Chicago Hospital and Medical Center, Chicago, IL, United States
| | | | - Barry A Hendin
- Center for Neurology and Spine, Phoenix, AZ, United States
| | | | - Xiangyi Meng
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, United States
| | - James Stankiewicz
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, United States
| | | | - Anne H Cross
- Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
11
|
Lee SM, Min SW, Kwon HS, Bae GD, Jung JH, Park HI, Lee SH, Lim CS, Ko BJ, Lee JC, Jung ST. Effective clearance of rituximab-resistant tumor cells by breaking the mirror-symmetry of immunoglobulin G and simultaneous binding to CD55 and CD20. Sci Rep 2023; 13:18275. [PMID: 37880350 PMCID: PMC10600224 DOI: 10.1038/s41598-023-45491-8] [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: 05/11/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023] Open
Abstract
Complement-dependent cytotoxicity (CDC), which eliminates aberrant target cells through the assembly and complex formation of serum complement molecules, is one of the major effector functions of anticancer therapeutic antibodies. In this study, we discovered that breaking the symmetry of natural immunoglobulin G (IgG) antibodies significantly increased the CDC activity of anti-CD20 antibodies. In addition, the expression of CD55 (a checkpoint inhibitor in the CDC cascade) was significantly increased in a rituximab-resistant cell line generated in-house, suggesting that CD55 overexpression might be a mechanism by which cancer cells acquire rituximab resistance. Based on these findings, we developed an asymmetric bispecific antibody (SBU-CD55 × CD20) that simultaneously targets both CD55 and CD20 to effectively eliminate rituximab-resistant cancer cells. In various cancer cell lines, including rituximab-resistant lymphoma cells, the SBU-CD55 × CD20 antibody showed significantly higher CDC activity than either anti-CD20 IgG antibody alone or a combination of anti-CD20 IgG antibody and anti-CD55 IgG antibody. Furthermore, the asymmetric bispecific antibody (SBU-CD55 × CD20) exhibited significantly higher CDC activity against rituximab-resistant cancer cells compared to other bispecific antibodies with symmetric features. These results demonstrate that enhancing CDC with an asymmetric CD55-binding bispecific antibody could be a new strategy for developing therapeutics to treat patients with relapsed or refractory cancers.
Collapse
Affiliation(s)
- Sang Min Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Department of Applied Chemistry, Kookmin University, 77, Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Sung-Won Min
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Hyeong Sun Kwon
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Gong-Deuk Bae
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Ji Hae Jung
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Hye In Park
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Seung Hyeon Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Chung Su Lim
- New Drug Development Center, Osong Medical Innovation Foundation 123, Cheongju, Chungcheongbuk-do, 28160, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Science, Sungshin Women's University, 55, Dobonng-Ro 76ga-gil, Gangbuk, Seoul, 01133, Republic of Korea
| | - Ji Chul Lee
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea.
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
- Institute of Human Genetics, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Biomedical Research Center, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| |
Collapse
|
12
|
Zhang Z, Xu Q, Huang L. B cell depletion therapies in autoimmune diseases: Monoclonal antibodies or chimeric antigen receptor-based therapy? Front Immunol 2023; 14:1126421. [PMID: 36855629 PMCID: PMC9968396 DOI: 10.3389/fimmu.2023.1126421] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/26/2023] [Indexed: 03/02/2023] Open
Abstract
Immune system detects foreign pathogens, distinguishes them from self-antigens and responds to defend human body. When this self-tolerance is disrupted, the overactive immune system attacks healthy tissues or organs and the autoimmune diseases develop. B cells and plasma cells contribute a lot to pathogenesis and persistence of autoimmune diseases in both autoantibody-dependent and autoantibody-independent ways. Accumulating data indicates that treatments aiming to eliminate antibody-secreting cells (B cells or plasma cells) are effective in a wide spectrum of autoimmune diseases. Monoclonal antibodies (mAbs) deplete B cell lineage or plasma cells by signaling disruption, complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). Engineered-T cells armed with chimeric antigen receptors (CARs) have been adopted from field of hematological malignancies as a method to eliminate B cells or plasma cells. In this review, we update our understanding of B cell depletion therapies in autoimmune diseases, review the mechanism, efficacy, safety and application of monoclonal antibodies and CAR-based immunotherapies, and discuss the strengths and weaknesses of these treatment options for patients.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China,*Correspondence: Liang Huang,
| |
Collapse
|
13
|
Modeling and affinity maturation of an anti-CD20 nanobody: a comprehensive in-silico investigation. Sci Rep 2023; 13:582. [PMID: 36631511 PMCID: PMC9834265 DOI: 10.1038/s41598-023-27926-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
B-cell Non-Hodgkin lymphomas are the malignancies of lymphocytes. CD20 is a membrane protein, which is highly expressed on the cell surface of the B-cells in NHL. Treatments using monoclonal antibodies (mAbs) have resulted in failure in some cases. Nanobodies (NBs), single-domain antibodies with low molecular weights and a high specificity in antigen recognition, could be practical alternatives for traditional mAbs with superior characteristics. To design an optimized NB as a candidate CD20 inhibitor with raised binding affinity to CD20, the structure of anti-CD20 NB was optimized to selectively target CD20. The 3D structure of the NB was constructed based on the optimal templates (6C5W and 5JQH), and the key residues were determined by applying a molecular docking study. After identifying the key residues, some mutations were introduced using a rational protocol to improve the binding affinity of the NB to CD20. The rational mutations were conducted using the experimental design (Taguchi method). Six residues (Ser27, Thr28, Phe29, Ile31, Asp99, and Asn100) were selected as the key residues, and five residues were targeted for rational mutation (Trp, Phe, His, Asp, and Tyr). Based on the mutations suggested by the experimental design, two optimized NB structures were constructed. NB2 showed a remarkable binding affinity to CD20 in docking studies with a binding energy of - 853 kcal/mol. The optimized NB was further evaluated using molecular dynamics simulation. The results revealed that CDR1 (complementarity determining regions1) and CDR3 are essential loops for recognizing the antigen. NB2 could be considered as a potential inhibitor of CD20, though experimental evaluations are needed to confirm it.
Collapse
|
14
|
89Zr-labelled Obinutuzumab: a potential immuno-PET radiopharmaceutical. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08614-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
15
|
Immanuel T, Li J, Green TN, Bogdanova A, Kalev-Zylinska ML. Deregulated calcium signaling in blood cancer: Underlying mechanisms and therapeutic potential. Front Oncol 2022; 12:1010506. [PMID: 36330491 PMCID: PMC9623116 DOI: 10.3389/fonc.2022.1010506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Intracellular calcium signaling regulates diverse physiological and pathological processes. In solid tumors, changes to calcium channels and effectors via mutations or changes in expression affect all cancer hallmarks. Such changes often disrupt transport of calcium ions (Ca2+) in the endoplasmic reticulum (ER) or mitochondria, impacting apoptosis. Evidence rapidly accumulates that this is similar in blood cancer. Principles of intracellular Ca2+ signaling are outlined in the introduction. We describe different Ca2+-toolkit components and summarize the unique relationship between extracellular Ca2+ in the endosteal niche and hematopoietic stem cells. The foundational data on Ca2+ homeostasis in red blood cells is discussed, with the demonstration of changes in red blood cell disorders. This leads to the role of Ca2+ in neoplastic erythropoiesis. Then we expand onto the neoplastic impact of deregulated plasma membrane Ca2+ channels, ER Ca2+ channels, Ca2+ pumps and exchangers, as well as Ca2+ sensor and effector proteins across all types of hematologic neoplasms. This includes an overview of genetic variants in the Ca2+-toolkit encoding genes in lymphoid and myeloid cancers as recorded in publically available cancer databases. The data we compiled demonstrate that multiple Ca2+ homeostatic mechanisms and Ca2+ responsive pathways are altered in hematologic cancers. Some of these alterations may have genetic basis but this requires further investigation. Most changes in the Ca2+-toolkit do not appear to define/associate with specific disease entities but may influence disease grade, prognosis, treatment response, and certain complications. Further elucidation of the underlying mechanisms may lead to novel treatments, with the aim to tailor drugs to different patterns of deregulation. To our knowledge this is the first review of its type in the published literature. We hope that the evidence we compiled increases awareness of the calcium signaling deregulation in hematologic neoplasms and triggers more clinical studies to help advance this field.
Collapse
Affiliation(s)
- Tracey Immanuel
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jixia Li
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan City, China
| | - Taryn N. Green
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Maggie L. Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Haematology Laboratory, Department of Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
16
|
Oostindie SC, Lazar GA, Schuurman J, Parren PWHI. Avidity in antibody effector functions and biotherapeutic drug design. Nat Rev Drug Discov 2022; 21:715-735. [PMID: 35790857 PMCID: PMC9255845 DOI: 10.1038/s41573-022-00501-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 12/16/2022]
Abstract
Antibodies are the cardinal effector molecules of the immune system and are being leveraged with enormous success as biotherapeutic drugs. A key part of the adaptive immune response is the production of an epitope-diverse, polyclonal antibody mixture that is capable of neutralizing invading pathogens or disease-causing molecules through binding interference and by mediating humoral and cellular effector functions. Avidity - the accumulated binding strength derived from the affinities of multiple individual non-covalent interactions - is fundamental to virtually all aspects of antibody biology, including antibody-antigen binding, clonal selection and effector functions. The manipulation of antibody avidity has since emerged as an important design principle for enhancing or engineering novel properties in antibody biotherapeutics. In this Review, we describe the multiple levels of avidity interactions that trigger the overall efficacy and control of functional responses in both natural antibody biology and their therapeutic applications. Within this framework, we comprehensively review therapeutic antibody mechanisms of action, with particular emphasis on engineered optimizations and platforms. Overall, we describe how affinity and avidity tuning of engineered antibody formats are enabling a new wave of differentiated antibody drugs with tailored properties and novel functions, promising improved treatment options for a wide variety of diseases.
Collapse
Affiliation(s)
- Simone C Oostindie
- Genmab, Utrecht, Netherlands.,Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Greg A Lazar
- Department of Antibody Engineering, Genentech, San Francisco, CA, USA
| | | | - Paul W H I Parren
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands. .,Sparring Bioconsult, Odijk, Netherlands. .,Lava Therapeutics, Utrecht, Netherlands.
| |
Collapse
|
17
|
Bhargava P, Hartung HP, Calabresi PA. Contribution of B cells to cortical damage in multiple sclerosis. Brain 2022; 145:3363-3373. [PMID: 35775595 DOI: 10.1093/brain/awac233] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis is associated with lesions not just in the white matter, but also involving the cortex. Cortical involvement has been linked to greater disease severity and hence understanding the factor underlying cortical pathology could help identify new therapeutic strategies for multiple sclerosis. The critical role of B cells in multiple sclerosis has been clarified by multiple pivotal trials of B cell depletion in people with multiple sclerosis. The presence of B cell rich areas of meningeal inflammation in multiple sclerosis has been identified at all stages of multiple sclerosis. Leptomeningeal inflammation is associated with greater extent of cortical demyelination and neuronal loss and with greater disease severity. Recent studies have identified several potential mechanisms by which B cells may mediate cortical injury including antibody production, extracellular vesicles containing neurotoxic substances and production of pro-inflammatory cytokines. Additionally, B cells may indirectly mediate cortical damage through effects on T cells, macrophages or microglia. Several animal models replicate the meningeal inflammation and cortical injury noted in people with multiple sclerosis. Studies in these models have identified BTK inhibition and type II anti-CD20 antibodies as potential agents that can impact meningeal inflammation. Trials of anti-CD20 monoclonal antibodies in people with multiple sclerosis have unsuccessfully attempted to eliminate B cells in the leptomeninges. New strategies to target B cells in multiple sclerosis include BTK inhibition and cell-based therapies aimed at B cells infected with Epstein Barr virus. Future studies will clarify the mechanisms by which B cells mediate cortical injury and treatment strategies that can target B cells in the leptomeninges and CNS parenchyma.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans Peter Hartung
- Department of Neurology, Heinrich-Heine University, Dusseldorf, Germany.,Brain and Mind Center, University of Sydney, Sydney, Australia.,Department of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Atzeni F, Gozza F, Cafaro G, Perricone C, Bartoloni E. Cardiovascular Involvement in Sjögren’s Syndrome. Front Immunol 2022; 13:879516. [PMID: 35634284 PMCID: PMC9134348 DOI: 10.3389/fimmu.2022.879516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/22/2022] [Indexed: 12/26/2022] Open
Abstract
Sjögren Syndrome (SS) seems to be associated with a greater “overall risk” of cardiovascular (CV) and cerebrovascular events. Although not conventionally considered a feature of the disease, CV events represent a major burden in SS patients. CV risk is the consequence of a complex combination of multiple factors, including traditional risk factors and disease-related mechanisms. A complex relationships between disease-related features, endothelial dysfunction and traditional risk factor has been suggested. Several drugs are available for treating the systemic manifestations of SS, however they have shown positive effects on different outcomes of the disease, but until today the data on the role of these drugs on CV events are scarse. Given these data, the aim of this review was to evaluate the risk of CV risk in primary SS and the effect of the drugs on this manifestation.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- *Correspondence: Fabiola Atzeni,
| | - Francesco Gozza
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giacomo Cafaro
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carlo Perricone
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
19
|
Hamlin PA, Musteata V, Park SI, Burnett C, Dabovic K, Strack T, Williams ET, Anand BS, Higgins JP, Persky DO. Safety and Efficacy of Engineered Toxin Body MT-3724 in Relapsed or Refractory B-cell Non-Hodgkin's Lymphomas and Diffuse Large B-cell Lymphoma. CANCER RESEARCH COMMUNICATIONS 2022; 2:307-315. [PMID: 36875713 PMCID: PMC9981212 DOI: 10.1158/2767-9764.crc-22-0056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022]
Abstract
MT-3724, a novel engineered toxin body comprised of an anti-CD20 single-chain variable fragment genetically fused to Shiga-like Toxin A subunit, is capable of binding to and internalizing against CD20, inducing cell killing via permanent ribosomal inactivation. This study evaluated MT-3724 in patients with relapsed/refractory B-cell non-Hodgkin lymphoma (r/rNHL). This open-label, multiple-dose phase Ia/b trial included a dose escalation in patients with r/rNHL according to a standard 3+3 design. Primary objectives were to determine the MTD and pharmacokinetics/pharmacodynamics. In a dose expansion study at MTD in serum rituximab-negative patients with diffuse large B-cell lymphoma (DLBCL), primary objectives were safety, tolerability, and pharmacokinetics/pharmacodynamics. Twenty-seven patients enrolled. MTD was 50 μg/kg/dose with 6,000 μg/dose cap. Thirteen patients experienced at least one grade ≥3 treatment-related adverse events; the most common grade ≥3 event was myalgia (11.1%). Two patients (75 μg/kg/dose) experienced grade 2 treatment-related capillary leak syndrome. Overall objective response rate was 21.7%. In serum rituximab-negative patients with DLBCL or composite DLBCL (n = 12), overall response rate was 41.7% (complete response, n = 2; partial response, n = 3). In patients with detectable baseline peripheral B cells, treatment resulted in dose-dependent B-cell depletion. The proportion of patients with anti-drug antibodies (ADA) increased during treatment and the majority appeared to be neutralizing based on an in vitro assay; nevertheless, tumor regression and responses were observed. MT-3724 demonstrated efficacy at the MTD in this population of previously treated patients with r/rDLBCL, with mild-to-moderate immunogenic safety events. Significance This work describes the safety and efficacy of a new pharmaceutical pathway that could provide a treatment option for a subset of patients with a critical unmet therapeutic need. The study drug, MT-3724, is capable of targeting B-cell lymphomas via a unique, potent cell-killing mechanism that appears to be promising.
Collapse
Affiliation(s)
- Paul A Hamlin
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vasile Musteata
- Institute of Oncology, ARENSIA EM, Chisinau, Republic of Moldova
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang X, Phan MM, Sun Y, Koerber JT, Ho H, Chen Y, Yang J. Development of an SPR-based binding assay for characterization of anti-CD20 antibodies to CD20 expressed on extracellular vesicles. Anal Biochem 2022; 646:114635. [DOI: 10.1016/j.ab.2022.114635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 11/01/2022]
|
21
|
Del Vecchio L, Allinovi M, Rocco P, Brando B. Rituximab Therapy for Adults with Nephrotic Syndromes: Standard Schedules or B Cell-Targeted Therapy? J Clin Med 2021; 10:5847. [PMID: 34945143 PMCID: PMC8709396 DOI: 10.3390/jcm10245847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Rituximab is a chimeric anti-CD20 monoclonal antibody. It acts mainly through complement-dependent cytotoxicity on B cells expressing the CD20 marker. In this review, we analyse the efficacy and possible pitfalls of rituximab to treat nephrotic syndromes by taking into account pharmacological considerations and CD19 marker testing utility. Despite the fact that the drug has been in use for years, efficacy and treatment schedules in adults with nephrotic syndrome are still a matter of debate. Clinical trials have proven the efficacy and safety of rituximab in idiopathic membranous nephropathy. Data from observational studies also showed the efficacy of rituximab in minimal change disease and focal segmental glomerulosclerosis. Rituximab use is now widely recommended by new Kidney Disease Improved Outcome (KDIGO) guidelines in membranous nephropathy and in frequent-relapsing, steroid-dependent minimal change disease or focal segmental glomerulosclerosis. However, rituximab response has a large interindividual variability. One reason could be that rituximab is lost in the urine at a higher extent in patients with nonselective nephrotic proteinuria, exposing patients to different rituximab plasma levels. Moreover, the association between CD19+ levels and clinical response or relapses is not always present, making the use of this marker in clinical practice complex. High resolution flow cytometry has increased the capability of detecting residual CD19+ B cells. Moreover, it can identify specific B-cell subsets (including IgG-switched memory B cells), which can repopulate at different rates. Its wider use could become a useful tool for better understanding reasons of rituximab failure or avoiding unnecessary retreatments.
Collapse
Affiliation(s)
- Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant’Anna Hospital, ASST Lariana, 22042 Como, Italy
| | - Marco Allinovi
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, 50134 Florence, Italy;
| | - Paolo Rocco
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via G. Colombo, 71-20133 Milan, Italy;
| | - Bruno Brando
- Haematology Laboratory and Transfusion Centre, Legnano General Hospital (Milan), 20025 Milan, Italy;
| |
Collapse
|
22
|
Durand-Panteix S, Monteil J, Sage M, Garot A, Clavel M, Saidi A, Torgue J, Cogne M, Quelven I. Preclinical study of 212Pb alpha-radioimmunotherapy targeting CD20 in non-Hodgkin lymphoma. Br J Cancer 2021; 125:1657-1665. [PMID: 34671126 DOI: 10.1038/s41416-021-01585-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Despite therapeutic advances, Non-Hodgkin lymphoma (NHL) relapses can occur. The development of radioimmunotherapy (RIT) with α-emitters is an attractive alternative. In this study, we investigated the potential of α-RIT in conjunction with 212Pb-rituximab for the treatment of NHL. METHODS EL4-hCD20-Luc cells (mouse lymphoma cell line) were used for in vitro and in vivo studies. Biodistribution and efficacy studies were performed on C57BL/6 mice injected intravenously with 25 × 103 cells. RESULTS 212Pb-rituximab (0.925-7.4 kBq/mL) inhibit proliferation of EL4-hCD20-Luc cells in vitro. Biodistribution of 203/212Pb-rituximab in mice showed a significant tumour uptake and suggested that the liver, spleen, and kidneys were the organs at risk. For efficacy studies, mice were treated at either 11 days (early stage) or 20-30 days after injection of tumour cells (late stage). Treatment with 277.5 kBq 212Pb-rituximab significantly prolonged survival. Even at an advanced tumour stage, significant tumour regression occurred, with an increase in the median survival time to 28 days, compared with 9 days in the controls. CONCLUSIONS These results show the efficacy of 212Pb-rituximab in a murine syngeneic lymphoma model, in terms of significant tumour regression and increased survival, thereby highlighting the potency of α-RIT for the treatment of NHL.
Collapse
Affiliation(s)
- Stéphanie Durand-Panteix
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | - Jacques Monteil
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France.,Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Magali Sage
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | - Armand Garot
- Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Marie Clavel
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | | | | | - Michel Cogne
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France.
| | - Isabelle Quelven
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France. .,Nuclear Medicine Department, Limoges University Hospital, Limoges, France. .,ToNIC, Toulouse NeuroImaging Center - INSERM U1214, Toulouse, France.
| |
Collapse
|
23
|
Chen YH, Wang XY, Jin X, Yang Z, Xu J. Rituximab Therapy for Primary Sjögren's Syndrome. Front Pharmacol 2021; 12:731122. [PMID: 34539411 PMCID: PMC8445329 DOI: 10.3389/fphar.2021.731122] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a systemic autoimmune diseases of the connective tissues, characteristic of the presentation of keratoconjunctivitis sicca and xerostomia. A cardinal pathogenetic feature of SS is B-cell hyperactivity, which has invited efforts on optimal B-cell targeted therapy, whereas conventional corticosteroids and disease-modifying antirheumatic drugs (DMARDs) are restricted to symptomatic relief. As per the first EULAR recommendation for pSS patients published in 2020, regimens with monoclonal antibodies targeting B cells may be initiated in patients with severe, refractory systemic disease, notably rituximab (RTX), a mouse-derived monoclonal antibody that targets CD20 antigen and contributes to B-cell depletion. Nonetheless, the data available from clinical trials with RTX are often controversial. Despite the lack of promising results from two large RCTs, several positive clinical efficacies were demonstrated. This current review addressed the efficacy and safety of clinical trials available and elucidated the potential of RTX on the immune system, especially B and T cells. Furthermore, plausible explanations for the discrepancy in clinical data were also presented.
Collapse
Affiliation(s)
| | | | | | - Zi Yang
- Key Laboratory of Oral Disease Research of Anhui Province, Stomatologic Hospital AndCollege, Anhui Medical University, Hefei, China
| | - Jianguang Xu
- Key Laboratory of Oral Disease Research of Anhui Province, Stomatologic Hospital AndCollege, Anhui Medical University, Hefei, China
| |
Collapse
|
24
|
Ghilardi N, Pappu R, Arron JR, Chan AC. 30 Years of Biotherapeutics Development-What Have We Learned? Annu Rev Immunol 2021; 38:249-287. [PMID: 32340579 DOI: 10.1146/annurev-immunol-101619-031510] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Since the birth of biotechnology, hundreds of biotherapeutics have been developed and approved by the US Food and Drug Administration (FDA) for human use. These novel medicines not only bring significant benefit to patients but also represent precision tools to interrogate human disease biology. Accordingly, much has been learned from the successes and failures of hundreds of high-quality clinical trials. In this review, we discuss general and broadly applicable themes that have emerged from this collective experience. We base our discussion on insights gained from exploring some of the most important target classes, including interleukin-1 (IL-1), tumor necrosis factor α (TNF-α), IL-6, IL-12/23, IL-17, IL-4/13, IL-5, immunoglobulin E (IgE), integrins and B cells. We also describe current challenges and speculate about how emerging technological capabilities may enable the discovery and development of the next generation of biotherapeutics.
Collapse
Affiliation(s)
- Nico Ghilardi
- Department of Immunology, Genentech, South San Francisco, California 94080, USA; , ,
| | - Rajita Pappu
- Department of Immunology, Genentech, South San Francisco, California 94080, USA; , ,
| | - Joseph R Arron
- Department of Immunology, Genentech, South San Francisco, California 94080, USA; , ,
| | - Andrew C Chan
- Research-Biology, Genentech, South San Francisco, California 94080, USA;
| |
Collapse
|
25
|
Silva-Gomes R, Mapelli SN, Boutet MA, Mattiola I, Sironi M, Grizzi F, Colombo F, Supino D, Carnevale S, Pasqualini F, Stravalaci M, Porte R, Gianatti A, Pitzalis C, Locati M, Oliveira MJ, Bottazzi B, Mantovani A. Differential expression and regulation of MS4A family members in myeloid cells in physiological and pathological conditions. J Leukoc Biol 2021; 111:817-836. [PMID: 34346525 PMCID: PMC9290968 DOI: 10.1002/jlb.2a0421-200r] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The MS4A gene family encodes 18 tetraspanin-like proteins, most of which with unknown function. MS4A1 (CD20), MS4A2 (FcεRIβ), MS4A3 (HTm4), and MS4A4A play important roles in immunity, whereas expression and function of other members of the family are unknown. The present investigation was designed to obtain an expression fingerprint of MS4A family members, using bioinformatics analysis of public databases, RT-PCR, and protein analysis when possible. MS4A3, MS4A4A, MS4A4E, MS4A6A, MS4A7, and MS4A14 were expressed by myeloid cells. MS4A6A and MS4A14 were expressed in circulating monocytes and decreased during monocyte-to-Mϕ differentiation in parallel with an increase in MS4A4A expression. Analysis of gene expression regulation revealed a strong induction of MS4A4A, MS4A6A, MS4A7, and MS4A4E by glucocorticoid hormones. Consistently with in vitro findings, MS4A4A and MS4A7 were expressed in tissue Mϕs from COVID-19 and rheumatoid arthritis patients. Interestingly, MS4A3, selectively expressed in myeloid precursors, was found to be a marker of immature circulating neutrophils, a cellular population associated to COVID-19 severe disease. The results reported here show that members of the MS4A family are differentially expressed and regulated during myelomonocytic differentiation, and call for assessment of their functional role and value as therapeutic targets.
Collapse
Affiliation(s)
- Rita Silva-Gomes
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde and Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | | | - Marie-Astrid Boutet
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Regenerative Medicine and Skeleton, RMeS, Inserm UMR 1229, Oniris, CHU Nantes, Université de Nantes, Nantes, France
| | - Irene Mattiola
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Berlin, Germany
| | - Marina Sironi
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Fabio Grizzi
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Domenico Supino
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Silvia Carnevale
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Fabio Pasqualini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Rémi Porte
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Andrea Gianatti
- Unit of Pathology, Azienda Ospedaliera Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Constantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Massimo Locati
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Maria José Oliveira
- ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde and Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
26
|
Cencioni MT, Mattoscio M, Magliozzi R, Bar-Or A, Muraro PA. B cells in multiple sclerosis - from targeted depletion to immune reconstitution therapies. Nat Rev Neurol 2021; 17:399-414. [PMID: 34075251 DOI: 10.1038/s41582-021-00498-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Increasing evidence indicates the involvement of B cells in the pathogenesis of multiple sclerosis (MS), but their precise roles are unclear. In this Review, we provide an overview of the development and physiological functions of B cells and the main mechanisms through which B cells are thought to contribute to CNS autoimmunity. In MS, abnormalities of B cell function include pro-inflammatory cytokine production, defective B cell regulatory function and the formation of tertiary lymphoid-like structures in the CNS, which are the likely source of abnormal immunoglobulin production detectable in the cerebrospinal fluid. We also consider the hypothesis that Epstein-Barr virus (EBV) is involved in the B cell overactivation that leads to inflammatory injury to the CNS in MS. We also review the immunological effects - with a focus on the effects on B cell subsets - of several successful therapeutic approaches in MS, including agents that selectively deplete B cells (rituximab, ocrelizumab and ofatumumab), agents that less specifically deplete lymphocytes (alemtuzumab and cladribine) and autologous haematopoietic stem cell transplantation, in which the immune system is unselectively ablated and reconstituted. We consider the insights that these effects on B cell populations provide and their potential to further our understanding and targeting of B cells in MS.
Collapse
Affiliation(s)
- Maria T Cencioni
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Miriam Mattoscio
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Roberta Magliozzi
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.,Department of Neurology, University of Verona, Verona, Italy
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paolo A Muraro
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
27
|
Vlachonikola E, Stamatopoulos K, Chatzidimitriou A. T Cell Defects and Immunotherapy in Chronic Lymphocytic Leukemia. Cancers (Basel) 2021; 13:3255. [PMID: 34209724 PMCID: PMC8268526 DOI: 10.3390/cancers13133255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 12/31/2022] Open
Abstract
In the past few years, independent studies have highlighted the relevance of the tumor microenvironment (TME) in cancer, revealing a great variety of TME-related predictive markers, as well as identifying novel therapeutic targets in the TME. Cancer immunotherapy targets different components of the immune system and the TME at large in order to reinforce effector mechanisms or relieve inhibitory and suppressive signaling. Currently, it constitutes a clinically validated treatment for many cancers, including chronic lymphocytic leukemia (CLL), an incurable malignancy of mature B lymphocytes with great dependency on microenvironmental signals. Although immunotherapy represents a promising therapeutic option with encouraging results in CLL, the dysfunctional T cell compartment remains a major obstacle in such approaches. In the scope of this review, we outline the current immunotherapeutic treatment options in CLL in the light of recent immunogenetic and functional evidence of T cell impairment. We also highlight possible approaches for overcoming T cell defects and invigorating potent anti-tumor immune responses that would enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Elisavet Vlachonikola
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, 57001 Thessaloniki, Greece; (E.V.); (K.S.)
- Department of Genetics and Molecular Biology, Faculty of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Kostas Stamatopoulos
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, 57001 Thessaloniki, Greece; (E.V.); (K.S.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Anastasia Chatzidimitriou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, 57001 Thessaloniki, Greece; (E.V.); (K.S.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
28
|
Single Domain Antibodies as Carriers for Intracellular Drug Delivery: A Proof of Principle Study. Biomolecules 2021; 11:biom11070927. [PMID: 34206656 PMCID: PMC8301836 DOI: 10.3390/biom11070927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are currently used for the targeted delivery of drugs to diseased cells, but intracellular drug delivery and therefore efficacy may be suboptimal because of the large size, slow internalization and ineffective intracellular trafficking of the antibody. Using a phage display method selecting internalizing phages only, we developed internalizing single domain antibodies (sdAbs) with high binding affinity to rat PDGFRβ, a receptor involved in different types of diseases. We demonstrate that these constructs have different characteristics with respect to internalization rates but all traffic to lysosomes. To compare their efficacy in targeted drug delivery, we conjugated the sdAbs to a cytotoxic drug. The conjugates showed improved cytotoxicity correlating to their internalization speed. The efficacy of the conjugates was inhibited in the presence of vacuolin-1, an inhibitor of lysosomal maturation, suggesting lysosomal trafficking is needed for efficient drug release. In conclusion, sdAb constructs with different internalization rates can be designed against the same target, and sdAbs with a high internalization rate induce more cell killing than sdAbs with a lower internalization rate in vitro. Even though the overall efficacy should also be tested in vivo, sdAbs are particularly interesting formats to be explored to obtain different internalization rates.
Collapse
|
29
|
Han JW, Heo W, Lee D, Kang C, Kim HY, Jun I, So I, Hur H, Lee MG, Jung M, Kim JY. Plasma Membrane Localized GCaMP-MS4A12 by Orai1 Co-Expression Shows Thapsigargin- and Ca 2+-Dependent Fluorescence Increases. Mol Cells 2021; 44:223-232. [PMID: 33935043 PMCID: PMC8112172 DOI: 10.14348/molcells.2021.2031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 11/27/2022] Open
Abstract
Uniquely expressed in the colon, MS4A12 exhibits store-operated Ca2+ entry (SOCE) activity. However, compared to MS4A1 (CD20), a Ca2+ channel and ideal target for successful leukaemia immunotherapy, MS4A12 has rarely been studied. In this study, we investigated the involvement of MS4A12 in Ca2+ influx and expression changes in MS4A12 in human colonic malignancy. Fluorescence of GCaMP-fused MS4A12 (GCaMP-M12) was evaluated to analyse MS4A12 activity in Ca2+ influx. Plasma membrane expression of GCaMP-M12 was achieved by homo- or hetero-complex formation with no-tagged MS4A12 (nt-M12) or Orai1, respectively. GCaMP-M12 fluorescence in plasma membrane increased only after thapsigargin-induced depletion of endoplasmic reticulum Ca2+ stores, and this fluorescence was inhibited by typical SOCE inhibitors and siRNA for Orai1. Furthermore, GCaMP-MS4A12 and Orai1 co-transfection elicited greater plasma membrane fluorescence than GCaMP-M12 co-transfected with nt-M12. Interestingly, the fluorescence of GCaMP-M12 was decreased by STIM1 over-expression, while increased by siRNA for STIM1 in the presence of thapsigargin and extracellular Ca2+. Moreover, immunoprecipitation assay revealed that Orai1 co-expression decreased protein interactions between MS4A12 and STIM1. In human colon tissue, MS4A12 was expressed in the apical region of the colonic epithelium, although its expression was dramatically decreased in colon cancer tissues. In conclusion, we propose that MS4A12 contributes to SOCE through complex formation with Orai1, but does not cooperate with STIM1. Additionally, we discovered that MS4A12 is expressed in the apical membrane of the colonic epithelium and that its expression is decreased with cancer progression.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Woon Heo
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Donghyuk Lee
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Choeun Kang
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Hye-Yeon Kim
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Ikhyun Jun
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyuk Hur
- Department of Surgery, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
30
|
Hatterer E, Chauchet X, Richard F, Barba L, Moine V, Chatel L, Broyer L, Pontini G, Bautzova T, Juan F, Calloud S, Bosson N, Charreton M, Masternak K, Buatois V, Shang L. Targeting a membrane-proximal epitope on mesothelin increases the tumoricidal activity of a bispecific antibody blocking CD47 on mesothelin-positive tumors. MAbs 2021; 12:1739408. [PMID: 32191151 PMCID: PMC7153835 DOI: 10.1080/19420862.2020.1739408] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mesothelin (MSLN) is a cell surface glycoprotein overexpressed in several solid malignancies, including gastric, lung, mesothelioma, pancreatic and ovarian cancers. While several MSLN-targeting therapeutic approaches are in development, only limited efficacy has been achieved in patients. A potential shortcoming of several described antibody-based approaches is that they target the membrane distal region of MSLN and, additionally, are known to be handicapped by the high levels of circulating soluble MSLN in patients. We show here, using monoclonal antibodies (mAbs) targeting different MSLN-spanning epitopes, that the membrane-proximal region resulted in more efficient killing of MSLN-positive tumor cells in antibody-dependent cell-mediated cytotoxicity (ADCC) assays. Surprisingly, no augmented killing was observed in antibody-dependent cellular phagocytosis (ADCP) by mAbs targeting this membrane-proximal region. To further increase the ADCP potential, we, therefore, generated bispecific antibodies (bsAbs) coupling a high-affinity MSLN binding arm to a blocking CD47 arm. Here, targeting the membrane-proximal domain of MSLN demonstrated enhanced ADCP activity compared to membrane-distal domains when the bsAbs were used in in vitro phagocytosis killing assays. Importantly, the superior anti-tumor activity was also translated in xenograft tumor models. Furthermore, we show that the bsAb approach targeting the membrane-proximal epitope of MSLN optimized ADCC activity by augmenting FcγR-IIIA activation and enhanced ADCP via a more efficient blockade of the CD47/SIRPα axis.
Collapse
Affiliation(s)
- Eric Hatterer
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Xavier Chauchet
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Françoise Richard
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Leticia Barba
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Valéry Moine
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Laurence Chatel
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Lucile Broyer
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | - Tereza Bautzova
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Flora Juan
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Sebastien Calloud
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Nicolas Bosson
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Maud Charreton
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | - Vanessa Buatois
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Limin Shang
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| |
Collapse
|
31
|
Luo C, Wu G, Huang X, Ma Y, Zhang Y, Song Q, Xie M, Sun Y, Huang Y, Huang Z, Hou Y, Xu S, Chen J, Li X. Efficacy and safety of new anti-CD20 monoclonal antibodies versus rituximab for induction therapy of CD20 + B-cell non-Hodgkin lymphomas: a systematic review and meta-analysis. Sci Rep 2021; 11:3255. [PMID: 33547368 PMCID: PMC7864901 DOI: 10.1038/s41598-021-82841-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 01/20/2021] [Indexed: 12/25/2022] Open
Abstract
Rituximab combined with chemotherapy is the first-line induction therapy of CD20 positive B-cell non-Hodgkin lymphomas (CD20+ B-NHL). Recently new anti-CD20 monoclonal antibodies (mAbs) have been developed, but their efficacy and safety compared with rituximab are still controversial. We searched MEDLINE, Embase, and Cochrane Library for eligible randomized controlled trials (RCTs) that compared new anti-CD20 mAbs with rituximab in induction therapy of B-NHL. The primary outcomes are progression-free survival (PFS) and overall survival (OS), additional outcomes include event-free survival (EFS), disease-free survival (DFS), overall response rate (ORR), complete response rate (CRR) and incidences of adverse events (AEs). Time-to-event data were pooled as hazard ratios (HRs) using the generic inverse-variance method and dichotomous outcomes were pooled as odds ratios (ORs) using the Mantel-Haenszel method with their respective 95% confidence interval (CI). Eleven RCTs comprising 5261 patients with CD20+ B-NHL were included. Compared with rituximab, obinutuzumab significantly prolonged PFS (HR 0.84, 95% CI 0.73-0.96, P = 0.01), had no improvement on OS, ORR, and CRR, but increased the incidences of serious AEs (OR 1.29, 95% CI 1.13-1.48, P < 0.001). Ofatumumab was inferior to rituximab in consideration of ORR (OR 0.73, 95% CI 0.55-0.96, P = 0.02), and had no significant differences with rituximab in regard to PFS, OS and CRR. 131I-tositumomab yielded similar PFS, OS, ORR and CRR with rituximab. 90Y-ibritumomab tiuxetan increased ORR (OR 3.07, 95% CI 1.47-6.43, P = 0.003), but did not improve PFS, DFS, OS and CRR compared with rituximab. In conclusion, compared with rituximab in induction therapy of CD20+ B-NHL, obinutuzumab significantly improves PFS but with higher incidence of AEs, ofatumumab decreases ORR, 90Y-ibritumomab tiuxetan increases ORR.
Collapse
Affiliation(s)
- Chengxin Luo
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Guixian Wu
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiangtao Huang
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanni Ma
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yali Zhang
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qiuyue Song
- Department of Health Statistics, Third Military Medical University, Chongqing, China
| | - Mingling Xie
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanni Sun
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yarui Huang
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhen Huang
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yu Hou
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shuangnian Xu
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China.
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Jieping Chen
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China.
- Key Laboratory of Cancer Immunotherapy of Chongqing, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Xi Li
- Institute of Infectious Disease, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
32
|
Abstract
Patients with chronic lymphocytic leukemia can be divided into three categories: those who are minimally affected by the problem, often never requiring therapy; those that initially follow an indolent course but subsequently progress and require therapy; and those that from the point of diagnosis exhibit an aggressive disease necessitating treatment. Likewise, such patients pass through three phases: development of the disease, diagnosis, and need for therapy. Finally, the leukemic clones of all patients appear to require continuous input from the exterior, most often through membrane receptors, to allow them to survive and grow. This review is presented according to the temporal course that the disease follows, focusing on those external influences from the tissue microenvironment (TME) that support the time lines as well as those internal influences that are inherited or develop as genetic and epigenetic changes occurring over the time line. Regarding the former, special emphasis is placed on the input provided via the B-cell receptor for antigen and the C-X-C-motif chemokine receptor-4 and the therapeutic agents that block these inputs. Regarding the latter, prominence is laid upon inherited susceptibility genes and the genetic and epigenetic abnormalities that lead to the developmental and progression of the disease.
Collapse
MESH Headings
- Disease Progression
- Humans
- Immunotherapy
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Mutation
- PAX5 Transcription Factor/metabolism
- Receptors, Antigen, B-Cell
- Signal Transduction
- Tumor Microenvironment
Collapse
Affiliation(s)
- Nicholas Chiorazzi
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Shih-Shih Chen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Kanti R Rai
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York 11549, USA
| |
Collapse
|
33
|
Graf J, Mares J, Barnett M, Aktas O, Albrecht P, Zamvil SS, Hartung HP. Targeting B Cells to Modify MS, NMOSD, and MOGAD: Part 1. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e918. [PMID: 33406479 PMCID: PMC8063619 DOI: 10.1212/nxi.0000000000000918] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/11/2020] [Indexed: 01/16/2023]
Abstract
Ocrelizumab, rituximab, ofatumumab, ublituximab, inebilizumab, and evobrutinib are immunotherapies that target various B cell-related proteins. Most of these treatments have proven efficacy in relapsing and progressive forms of MS and neuromyelitis optica spectrum disease (NMOSD), or are in advanced stages of clinical development. Currently, ocrelizumab, ofatumumab, and inebilizumab are licensed for treatment of MS and NMOSD, respectively. This review focuses on the current state of knowledge about the role of B lymphocytes in immune-mediated pathophysiology and its implications for the mode of action. To understand the significance of this breakthrough in the context of the current MS therapeutic armamentarium, this review more closely examines the clinical development of CD20 depletion and the pioneering contribution of rituximab. Phase 3 and the recently published postmarketing studies will be highlighted to better understand the relevant efficacy data and safety aspects of long-term B-cell depletion.
Collapse
Affiliation(s)
- Jonas Graf
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco
| | - Jan Mares
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco
| | - Michael Barnett
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco
| | - Orhan Aktas
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco
| | - Philipp Albrecht
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco
| | - Scott S Zamvil
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco
| | - Hans-Peter Hartung
- From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology (M.B., H.-P.H.), Brain and Mind Centre, Department of Neurology, University of Sydney, New South Wales, Australia; and UCSF Weill Institute of Neurosciences (S.S.Z.), Department of Neurology, University of California at San Francisco.
| |
Collapse
|
34
|
Mudd TW, Lu C, Klement JD, Liu K. MS4A1 expression and function in T cells in the colorectal cancer tumor microenvironment. Cell Immunol 2020; 360:104260. [PMID: 33352466 DOI: 10.1016/j.cellimm.2020.104260] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 01/26/2023]
Abstract
The majority of human colorectal cancer remains resistant to immune checkpoint inhibitor (ICI) immunotherapy, but the underlying mechanism is incompletely understood. We report here that MS4A1, the gene encoding B cell surface marker CD20, is significantly downregulated in human colorectal carcinoma. Furthermore, MS4A1 expression level in colorectal carcinoma is positively correlated with patient survival. Analysis of scRNA-Seq dataset from public database revealed that MS4A1 is also expressed in subsets of T cells. A CD8+CD20+ subset of T cells exists in the neighboring non-neoplastic colon but disappears in tumor in human colorectal carcinoma. Furthermore, analysis of a published nivolumab treatment dataset indicated that nivolumab-bound T cells from human patients during anti-PD-1 immunotherapy exhibit significantly higher MS4A1 expression. Our findings indicate that CD8+CD20+ T subset functions in host cancer immunosurveillance and tumor microenvironment suppresses this T subset through a PD-L1-dependent mechanism.
Collapse
Affiliation(s)
- T William Mudd
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | - Chunwan Lu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - John D Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| |
Collapse
|
35
|
Liu R, Oldham RJ, Teal E, Beers SA, Cragg MS. Fc-Engineering for Modulated Effector Functions-Improving Antibodies for Cancer Treatment. Antibodies (Basel) 2020; 9:E64. [PMID: 33212886 PMCID: PMC7709126 DOI: 10.3390/antib9040064] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022] Open
Abstract
The majority of monoclonal antibody (mAb) therapeutics possess the ability to engage innate immune effectors through interactions mediated by their fragment crystallizable (Fc) domain. By delivering Fc-Fc gamma receptor (FcγR) and Fc-C1q interactions, mAb are able to link exquisite specificity to powerful cellular and complement-mediated effector functions. Fc interactions can also facilitate enhanced target clustering to evoke potent receptor signaling. These observations have driven decades-long research to delineate the properties within the Fc that elicit these various activities, identifying key amino acid residues and elucidating the important role of glycosylation. They have also fostered a growing interest in Fc-engineering whereby this knowledge is exploited to modulate Fc effector function to suit specific mechanisms of action and therapeutic purposes. In this review, we document the insight that has been generated through the study of the Fc domain; revealing the underpinning structure-function relationships and how the Fc has been engineered to produce an increasing number of antibodies that are appearing in the clinic with augmented abilities to treat cancer.
Collapse
Affiliation(s)
- Rena Liu
- GlaxoSmithKline Research and Development, Stevenage SG1 2NY, UK;
| | - Robert J. Oldham
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Emma Teal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| |
Collapse
|
36
|
Tang JJJ, Sung AP, Guglielmo MJ, Navarrete-Galvan L, Redelman D, Smith-Gagen J, Hudig D. Natural Killer (NK) Cell Expression of CD2 as a Predictor of Serial Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC). Antibodies (Basel) 2020; 9:antib9040054. [PMID: 33081115 PMCID: PMC7709134 DOI: 10.3390/antib9040054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
NK cell ADCC supports monoclonal antibody anti-tumor therapies. We investigated serial ADCC and whether it could be predicted by NK phenotypes, including expression of CD16A, CD2 and perforin. CD16A, the NK receptor for antibodies, has AA158 valine or phenylalanine variants with different affinities for IgG. CD2, a costimulatory protein, associates with CD16A and can augment CD16A-signaling. Pore-forming perforin is essential for rapid NK-mediated killing. NK cells were monitored for their ADCC serial killing frequency (KF). KF is the average number of target cells killed per cell by a cytotoxic cell population. KF comparisons were made at 1:4 CD16pos NK effector:target ratios. ADCC was toward Daudi cells labeled with 51Cr and obinutuzumab anti-CD20 antibody. CD16A genotypes were determined by DNA sequencing. CD2, CD16A, and perforin expression was monitored by flow cytometry. Serial killing KFs varied two-fold among 24 donors and were independent of CD16A genotypes and perforin levels. However, high percentages of CD2pos of the CD16Apos NK cells and high levels of CD16A were associated with high KFs. ROC analysis indicated that the %CD2pos of CD16Apos NK cells can predict KFs. In conclusion, the extent of serial ADCC varies significantly among donors and appears predictable by the CD2posCD16Apos NK phenotype.
Collapse
Affiliation(s)
- Jennifer J.-J. Tang
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Alexander P. Sung
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Michael J. Guglielmo
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Lydia Navarrete-Galvan
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Doug Redelman
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, Reno, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Dorothy Hudig
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
- Correspondence: ; Tel.: +1-775-784-4430
| |
Collapse
|
37
|
Gordan S, Albert H, Danzer H, Lux A, Biburger M, Nimmerjahn F. The Immunological Organ Environment Dictates the Molecular and Cellular Pathways of Cytotoxic Antibody Activity. Cell Rep 2020; 29:3033-3046.e4. [PMID: 31801071 DOI: 10.1016/j.celrep.2019.10.111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/30/2019] [Accepted: 10/25/2019] [Indexed: 02/07/2023] Open
Abstract
Cytotoxic immunoglobulin G antibodies are an essential component of therapeutic approaches aimed at depleting self-reactive or malignant cells. More recent evidence suggests that the tissue in which the target cell resides influences the underlying molecular and cellular pathways responsible for cytotoxic antibody activity. By studying cytotoxic IgG activity directed against natural killer cells in primary and secondary immunological organs, we show that distinct organ-specific effector pathways are responsible for target cell depletion. While in the bone marrow, the classical complement pathway and the high-affinity Fcγ-receptor I expressed on organ-resident macrophages were both involved in removing opsonized target cells; in the spleen and blood, all activating FcγRs but not the classical complement pathway were critical for target cell killing. Our study suggests that future strategies aimed at optimizing overall cytotoxic antibody activity may need to consider organ-specific pathways to achieve a maximal therapeutic effect.
Collapse
Affiliation(s)
- Sina Gordan
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Heike Albert
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Heike Danzer
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Markus Biburger
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erwin-Rommelstr. 3, 91058 Erlangen, Germany.
| |
Collapse
|
38
|
Caldito NG, Shirani A, Salter A, Stuve O. Adverse event profile differences between rituximab and ocrelizumab: Findings from the FDA Adverse Event Reporting Database. Mult Scler 2020; 27:1066-1076. [PMID: 32820687 DOI: 10.1177/1352458520949986] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Rituximab and ocrelizumab are anti-CD20 monoclonal antibodies that have shown a marked reduction in multiple sclerosis (MS) inflammatory activity. However, their real-world safety profile has not been adequately compared. OBJECTIVE To investigate the adverse event (AE) profile of rituximab and ocrelizumab reported to the Food and Drug Administration Adverse Event Reporting System (FAERS) database. METHODS The FAERS database was filtered by indication (MS) and drug (rituximab or ocrelizumab). Disproportionality analyses including but not limited to reporting odds ratio (ROR) were conducted to identify drug-AE associations. A signal was detected if the lower limit of the 95% confidence interval of ROR (ROR025) exceeded 1. RESULTS There were 623 and 7948 reports for rituximab and ocrelizumab, respectively. The most frequent AEs with rituximab and ocrelizumab were infusion-related reaction (4.82%) and urinary tract infection (10.52%), respectively. The strongest drug-AE association for rituximab and ocrelizumab were ear pruritus (ROR025: 47.53) and oral herpes (ROR025: 38.99), respectively. Ocrelizumab was associated with an almost two times higher frequency of infections than rituximab (21.93% vs 11.05%, respectively). CONCLUSION This study revealed differences in reporting AEs between rituximab and ocrelizumab. Infections were reported more frequently with ocrelizumab. Although speculative, a potentially different or more extensive B-cell depletion by ocrelizumab might explain these findings. Additional pharmacovigilance studies need to be performed to better characterize differences in the AE profile in B-cell-depleting therapies.
Collapse
Affiliation(s)
- Natalia Gonzalez Caldito
- Department of Neurology & Neurotherapeutics, Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Afsaneh Shirani
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amber Salter
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Olaf Stuve
- Department of Neurology & Neurotherapeutics, Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA/Neurology Section, VA North Texas Health Care System, Dallas, TX, USA
| |
Collapse
|
39
|
Abstract
Ocrelizumab ist ein monoklonaler Antikörper, der sich gegen das Differenzierungsantigen CD20 richtet und zu einer effektiven längerfristigen Depletion von Lymphozyten, insbesondere von B‑Zellen, führt. Unlängst publizierte Phase-3-Studien belegen, dass Ocrelizumab sowohl bei der Behandlung der schubförmigen als auch der primär progressiven Multiplen Sklerose (MS) wirksam ist. Darauf basierend wurde Ocrelizumab als erstes Medikament zur Behandlung der primär chronisch-progredienten MS zugelassen. Um diesen Durchbruch besser in den Kontext des heutigen MS-Therapiekanons einordnen zu können, lohnt sowohl ein Blick zurück auf die Entwicklung der antikörpervermittelten CD20-Depletion als auch auf die der Zulassung zugrunde liegenden Studien sowie deren Extensionsphasen. Diese Übersichtsarbeit diskutiert die verfügbaren Daten zur Wirksamkeit und Sicherheit der langfristigen B‑Zell-Depletion bei MS-Patienten und erörtert den aktuellen Kenntnisstand zur Rolle von B‑Lymphozyten in der Immunpathogenese der MS.
Collapse
|
40
|
Wang L, Qin W, Huo YJ, Li X, Shi Q, Rasko JEJ, Janin A, Zhao WL. Advances in targeted therapy for malignant lymphoma. Signal Transduct Target Ther 2020; 5:15. [PMID: 32296035 PMCID: PMC7058622 DOI: 10.1038/s41392-020-0113-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022] Open
Abstract
The incidence of lymphoma has gradually increased over previous decades, and it ranks among the ten most prevalent cancers worldwide. With the development of targeted therapeutic strategies, though a subset of lymphoma patients has become curable, the treatment of refractory and relapsed diseases remains challenging. Many efforts have been made to explore new targets and to develop corresponding therapies. In addition to novel antibodies targeting surface antigens and small molecular inhibitors targeting oncogenic signaling pathways and tumor suppressors, immune checkpoint inhibitors and chimeric antigen receptor T-cells have been rapidly developed to target the tumor microenvironment. Although these targeted agents have shown great success in treating lymphoma patients, adverse events should be noted. The selection of the most suitable candidates, optimal dosage, and effective combinations warrant further investigation. In this review, we systematically outlined the advances in targeted therapy for malignant lymphoma, providing a clinical rationale for mechanism-based lymphoma treatment in the era of precision medicine.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Wei Qin
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - Yu-Jia Huo
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - Xiao Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - Qing Shi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China
| | - John E J Rasko
- Gene & Stem Cell Therapy Program Centenary Institute, Sydney Medical School, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Anne Janin
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
- U1165 Inserm/Université Paris 7, Hôpital Saint Louis, Paris, France
| | - Wei-Li Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, China.
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
41
|
Sellebjerg F, Blinkenberg M, Sorensen PS. Anti-CD20 Monoclonal Antibodies for Relapsing and Progressive Multiple Sclerosis. CNS Drugs 2020; 34:269-280. [PMID: 31994023 DOI: 10.1007/s40263-020-00704-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) was previously thought to be a T-cell-mediated, demyelinating disease of the central nervous system. Disease-modifying therapies targeting T cells have, indeed, shown remarkable efficacy in patients with relapsing-remitting MS. However, these therapies do also target B cells, and a B-cell-depleting monoclonal antibody (ocrelizumab) has recently been approved for MS therapy and is efficacious not only in relapsing forms of MS but also in some patients with primary progressive MS. This suggests that B cells may play a more important role in the pathogenesis of MS than previously appreciated. We review the potential roles of B cells, which are the precursors of antibody-secreting plasma cells in the pathogenesis of MS. Furthermore, we provide an overview of the characteristics and clinical data for the four monoclonal antibodies (ocrelizumab, ofatumumab, rituximab, and ublituximab) that have been approved, are currently been used off-label or are being investigated as treatments for MS. These antibodies all target the cluster of differentiation (CD)-20 molecule and bind to distinct or overlapping epitopes on B cells and a subset of T cells that express CD20. This leads to B-cell depletion and, possibly, to depletion of CD20-positive T cells. The net result is strong suppression of clinical and radiological disease activity as well as slowing of the development of persisting neurological impairment.
Collapse
Affiliation(s)
- Finn Sellebjerg
- Danish Multiple Sclerosis Clinic, Department of Neurology 2082, University of Copenhagen, Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen, Denmark
| | - Morten Blinkenberg
- Danish Multiple Sclerosis Clinic, Department of Neurology 2082, University of Copenhagen, Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen, Denmark
| | - Per Soelberg Sorensen
- Danish Multiple Sclerosis Clinic, Department of Neurology 2082, University of Copenhagen, Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen, Denmark.
| |
Collapse
|
42
|
Heo W, Jin N, Park MS, Kim HY, Yoon SM, Lee J, Kim JY. STIM1 knock-down decreases the affinity of obinutuzumab for CD20 by altering CD20 localization to Triton-soluble membrane. Clin Exp Immunol 2020; 200:260-271. [PMID: 32056202 DOI: 10.1111/cei.13427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2020] [Indexed: 01/16/2023] Open
Abstract
Obinutuzumab is thought to exert its effects through its high antibody-dependent cellular cytotoxicity (ADCC) via glyco-engineering of the Fc region. In addition, obinutuzumab causes direct binding-induced cell death (DCD) only by specifically binding to its target CD20, a Ca2+ channel. However, the specific features of CD20 related to obinutuzumab binding-induction of cell death are not clearly understood. In this study, we evaluated the relationship between the Ca2+ channel features of CD20 as a store-operated Ca2+ channel (SOC) and obinutuzumab binding-induced cell death. Ca2+ channel function and biochemical analysis revealed that CD20 is an Orai1- and stromal interaction molecule (STIM1)-dependent Ca2+ pore. However, binding of obinutuzumab on CD20 did not have any effect on Ca2+ influx activity of CD20; the direct cell death rate mediated by obinutuzumab binding was almost equivalent with or without the extracellular Ca2+ condition. Given the apparent interaction between STIM1 and CD20, we observed Triton-X solubilized obinutuzumab-bound CD20 accompanied by STIM1. Subsequently, obinutuzumab binding and cell death were decreased by STIM1 knock-down in Ramos B cells. Thus, STIM1 directly contributes to cell death by increasing the affinity of cells for obinutuzumab by transferring CD20 to the Triton-soluble membrane region.
Collapse
Affiliation(s)
- W Heo
- Department of Pharmacology and Brain, Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - N Jin
- Department of Pharmacology and Brain, Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - M S Park
- Department of Pharmacology and Brain, Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - H-Y Kim
- Department of Pharmacology and Brain, Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - S M Yoon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - J Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - J Y Kim
- Department of Pharmacology and Brain, Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Song L, Chen Y, Ding J, Wu H, Zhang W, Ma M, Zang F, Wang Z, Gu N, Zhang Y. Rituximab conjugated iron oxide nanoparticles for targeted imaging and enhanced treatment against CD20-positive lymphoma. J Mater Chem B 2020; 8:895-907. [DOI: 10.1039/c9tb02521a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Fe3O4-PEG-nAb multivalent nanoprobes provide a possible avenue to improve the cancer therapy of rituximab towards clinical application.
Collapse
|
44
|
Goulet DR, Atkins WM. Considerations for the Design of Antibody-Based Therapeutics. J Pharm Sci 2020; 109:74-103. [PMID: 31173761 PMCID: PMC6891151 DOI: 10.1016/j.xphs.2019.05.031] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/02/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Antibody-based proteins have become an important class of biologic therapeutics, due in large part to the stability, specificity, and adaptability of the antibody framework. Indeed, antibodies not only have the inherent ability to bind both antigens and endogenous immune receptors but also have proven extremely amenable to protein engineering. Thus, several derivatives of the monoclonal antibody format, including bispecific antibodies, antibody-drug conjugates, and antibody fragments, have demonstrated efficacy for treating human disease, particularly in the fields of immunology and oncology. Reviewed here are considerations for the design of antibody-based therapeutics, including immunological context, therapeutic mechanisms, and engineering strategies. First, characteristics of antibodies are introduced, with emphasis on structural domains, functionally important receptors, isotypic and allotypic differences, and modifications such as glycosylation. Then, aspects of therapeutic antibody design are discussed, including identification of antigen-specific variable regions, choice of expression system, use of multispecific formats, and design of antibody derivatives based on fragmentation, oligomerization, or conjugation to other functional moieties. Finally, strategies to enhance antibody function through protein engineering are reviewed while highlighting the impact of fundamental biophysical properties on protein developability.
Collapse
Affiliation(s)
- Dennis R Goulet
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195.
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
45
|
Urlaub D, Zhao S, Blank N, Bergner R, Claus M, Tretter T, Lorenz HM, Watzl C, Merkt W. Activation of natural killer cells by rituximab in granulomatosis with polyangiitis. Arthritis Res Ther 2019; 21:277. [PMID: 31829278 PMCID: PMC6907269 DOI: 10.1186/s13075-019-2054-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/08/2019] [Indexed: 11/28/2022] Open
Abstract
Objective In the last few years, anti-CD20 antibody rituximab profoundly changed the therapeutic landscape of granulomatosis with polyangiitis (GPA). Here, we investigated whether natural killer (NK) cells may play a role in rituximab’s mechanism of action in GPA. Methods B cell depletion, NK cell degranulation, and the expression of CD69 and CD16 on NK cells were measured in a series of in vitro experiments using peripheral blood mononuclear cells (PBMCs). In vivo activation of NK cells was investigated in patients receiving rituximab infusions. Cells were analyzed by seven-color flow cytometry. Results NK cells from GPA patients were activated by immobilized rituximab. Also soluble rituximab activated NK cells, provided that B cells were present. NK cells degranulated and expressed the activation marker CD69 while CD16 expression was decreased. This activation of NK cells by soluble rituximab was accompanied by a reduction of B cells. The next-generation anti-CD20 antibody obinutuzumab showed stronger effects compared to rituximab on both the reduction of B cells and the activation of NK cells. Finally, we found that rituximab led to the activation of NK cells in vivo, provided that B cells were not depleted due to prior rituximab infusions. Conclusion B cell-bound rituximab activates NK cells in GPA. While NK cells therefore participate in rituximab’s mechanism of action in humans, their potential may be more efficiently exploited, e.g., by Fc engineering of therapeutic antibodies.
Collapse
Affiliation(s)
- Doris Urlaub
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Shuyang Zhao
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Norbert Blank
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Raoul Bergner
- Department of Rheumatology, Nephrology, Oncology, Klinikum Ludwigshafen, Ludwigshafen, Germany
| | - Maren Claus
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Theresa Tretter
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Carsten Watzl
- Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Wolfgang Merkt
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
46
|
Luan C, Chen B. Clinical application of obinutuzumab for treating chronic lymphocytic leukemia. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2899-2909. [PMID: 31692500 PMCID: PMC6707935 DOI: 10.2147/dddt.s212500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/14/2019] [Indexed: 12/02/2022]
Abstract
Alkylators and nucleoside analogs were the main drugs for treatingchronic lymphoblastic leukemia (CLL), which have been replaced by monoclonal antibodies, such as rituximab in the past 10 years for refractory or relapsed CLL. The first-line immunochemotherapy regimen, rituximab combined with nucleoside analogs, significantly increased CLL patients’ first-reaction rate and improved progression-free survival. Despite the long-lasting remissions by the use of chemoimmunotherapy, most CLL patients will relapse eventually. The obinutuzumab (GA101), an updated CD20 antibody, that is thought to achieve a more durable response with unique molecular and functional characteristics. Obinutuzumab is a humanized, monoclonal type II CD20 antibody modified by glycoengineering. The glycoengineered Fc portion enhances the binding affinity to the FcγRIII receptor on immune effector cells, resulting in increased antibody-dependent cellular cytotoxicity and phagocytosis. In addition, the type II antibody binding characteristics of obinutuzumab to CD20 lead to an efficient induction of direct non-apoptotic cell death. This review summarizes the results of clinical studies using obinutuzumab and looks forward to its further application in treating CLL clinically.
Collapse
Affiliation(s)
- Chunyan Luan
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| |
Collapse
|
47
|
Milcent B, Josseaume N, Riller Q, Giglioli I, Rabia E, Deligne C, Latouche JB, Hamieh M, Couture A, Toutirais O, Lone YC, Jeger-Madiot R, Graff-Dubois S, Amorim S, Loiseau P, Toubert A, Brice P, Thieblemont C, Teillaud JL, Sibéril S. Presence of T cells directed against CD20-derived peptides in healthy individuals and lymphoma patients. Cancer Immunol Immunother 2019; 68:1561-1572. [PMID: 31494742 PMCID: PMC6805815 DOI: 10.1007/s00262-019-02389-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
Preclinical and clinical studies have suggested that cancer treatment with antitumor antibodies induces a specific adaptive T cell response. A central role in this process has been attributed to CD4+ T cells, but the relevant T cell epitopes, mostly derived from non-mutated self-antigens, are largely unknown. In this study, we have characterized human CD20-derived epitopes restricted by HLA-DR1, HLA-DR3, HLA-DR4, and HLA-DR7, and investigated whether T cell responses directed against CD20-derived peptides can be elicited in human HLA-DR-transgenic mice and human samples. Based on in vitro binding assays to recombinant human MHC II molecules and on in vivo immunization assays in H-2 KO/HLA-A2+-DR1+ transgenic mice, we have identified 21 MHC II-restricted long peptides derived from intracellular, membrane, or extracellular domains of the human non-mutated CD20 protein that trigger in vitro IFN-γ production by PBMCs and splenocytes from healthy individuals and by PBMCs from follicular lymphoma patients. These CD20-derived MHC II-restricted peptides could serve as a therapeutic tool for improving and/or monitoring anti-CD20 T cell activity in patients treated with rituximab or other anti-CD20 antibodies.
Collapse
Affiliation(s)
- Benoit Milcent
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Nathalie Josseaume
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Quentin Riller
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Ilenia Giglioli
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Emilia Rabia
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Claire Deligne
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Jean-Baptiste Latouche
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Mohamad Hamieh
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Alexandre Couture
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Olivier Toutirais
- Unicaen, Inserm 1237, Physiopathology and Imaging of Neurological Disorders, Normandie University, Caen, France.,French Blood Service (Etablissement Français du Sang, EFS), Caen, France
| | - Yu-Chun Lone
- Inserm U1014, Hôpital Paul Brousse, Villejuif, France
| | - Raphaël Jeger-Madiot
- Inserm U1135, CNRS ERL8255, Center for Immunology and Microbial Infection, Paris, France
| | - Stéphanie Graff-Dubois
- Inserm U1135, CNRS ERL8255, Center for Immunology and Microbial Infection, Paris, France
| | - Sandy Amorim
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France
| | - Pascale Loiseau
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot-Paris 7, Paris, France
| | - Antoine Toubert
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot-Paris 7, Paris, France
| | - Pauline Brice
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France
| | - Catherine Thieblemont
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France.,EA7324 Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Luc Teillaud
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France.,Laboratory "Immune Microenvironment and Immunotherapy", Sorbonne Université UMRS 1135, INSERM U.1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Sophie Sibéril
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France. .,Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, 15 rue de l'Ecole de Médecine, 75006, Paris, France.
| |
Collapse
|
48
|
Ljungars A, Svensson C, Carlsson A, Birgersson E, Tornberg UC, Frendéus B, Ohlin M, Mattsson M. Deep Mining of Complex Antibody Phage Pools Generated by Cell Panning Enables Discovery of Rare Antibodies Binding New Targets and Epitopes. Front Pharmacol 2019; 10:847. [PMID: 31417405 PMCID: PMC6683657 DOI: 10.3389/fphar.2019.00847] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/02/2019] [Indexed: 01/11/2023] Open
Abstract
Phage display technology is a common approach for discovery of therapeutic antibodies. Drug candidates are typically isolated in two steps: First, a pool of antibodies is enriched through consecutive rounds of selection on a target antigen, and then individual clones are characterized in a screening procedure. When whole cells are used as targets, as in phenotypic discovery, the output phage pool typically contains thousands of antibodies, binding, in theory, hundreds of different cell surface receptors. Clonal expansion throughout the phage display enrichment process is affected by multiple factors resulting in extremely complex output phage pools where a few antibodies are highly abundant and the majority is very rare. This is a huge challenge in the screening where only a fraction of the antibodies can be tested using a conventional binding analysis, identifying mainly the most abundant clones typically binding only one or a few targets. As the expected number of antibodies and specificities in the pool is much higher, complementing methods, to reach deeper into the pool, are required, called deep mining methods. In this study, four deep mining methods were evaluated: 1) isolation of rare sub-pools of specific antibodies through selection on recombinant proteins predicted to be expressed on the target cells, 2) isolation of a sub-pool enriched for antibodies of unknown specificities through depletion of the primary phage pool on recombinant proteins corresponding to receptors known to generate many binders, 3) isolation of a sub-pool enriched for antibodies through selection on cells blocked with antibodies dominating the primary phage pool, and 4) next-generation sequencing-based analysis of isolated antibody pools followed by antibody gene synthesis and production of rare but enriched clones. We demonstrate that antibodies binding new targets and epitopes, not discovered through screening alone, can be discovered using described deep mining methods. Overall, we demonstrate the complexity of phage pools generated through selection on cells and show that a combination of conventional screening and deep mining methods are needed to fully utilize such pools. Deep mining will be important in future phenotypic antibody drug discovery efforts to increase the diversity of identified antibodies and targets.
Collapse
Affiliation(s)
- Anne Ljungars
- BioInvent International AB, Lund, Sweden
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | | | | | | | - Mats Ohlin
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | |
Collapse
|
49
|
de Taeye SW, Rispens T, Vidarsson G. The Ligands for Human IgG and Their Effector Functions. Antibodies (Basel) 2019; 8:E30. [PMID: 31544836 PMCID: PMC6640714 DOI: 10.3390/antib8020030] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 01/17/2023] Open
Abstract
Activation of the humoral immune system is initiated when antibodies recognize an antigen and trigger effector functions through the interaction with Fc engaging molecules. The most abundant immunoglobulin isotype in serum is Immunoglobulin G (IgG), which is involved in many humoral immune responses, strongly interacting with effector molecules. The IgG subclass, allotype, and glycosylation pattern, among other factors, determine the interaction strength of the IgG-Fc domain with these Fc engaging molecules, and thereby the potential strength of their effector potential. The molecules responsible for the effector phase include the classical IgG-Fc receptors (FcγR), the neonatal Fc-receptor (FcRn), the Tripartite motif-containing protein 21 (TRIM21), the first component of the classical complement cascade (C1), and possibly, the Fc-receptor-like receptors (FcRL4/5). Here we provide an overview of the interactions of IgG with effector molecules and discuss how natural variation on the antibody and effector molecule side shapes the biological activities of antibodies. The increasing knowledge on the Fc-mediated effector functions of antibodies drives the development of better therapeutic antibodies for cancer immunotherapy or treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Steven W de Taeye
- Sanquin Research, Dept Immunopathology and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands.
- Sanquin Research, Dept Experimental Immunohematology and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands.
| | - Theo Rispens
- Sanquin Research, Dept Immunopathology and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands.
| | - Gestur Vidarsson
- Sanquin Research, Dept Experimental Immunohematology and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
50
|
Shi L, Liu T, Gross ML, Huang Y. Recognition of Human IgG1 by Fcγ Receptors: Structural Insights from Hydrogen-Deuterium Exchange and Fast Photochemical Oxidation of Proteins Coupled with Mass Spectrometry. Biochemistry 2019; 58:1074-1080. [PMID: 30666863 DOI: 10.1021/acs.biochem.8b01048] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) is an effector function of immunoglobulins (IgGs) involved in the killing of target cells by a cytotoxic effector cell. Recognition of IgG by Fc receptors expressed on natural killer cells, mostly FcγIII receptors (FcγRIII), underpins the ADCC mechanism, thus motivating investigations of these interactions. In this paper, we describe the combination of hydrogen-deuterium exchange and fast photochemical oxidation of proteins (FPOP) coupled with mass spectrometry to study the interactions of the human IgG1/FcγRIII complex. Using these orthogonal approaches, we identified critical peptide regions and residues involved in the recognition of IgG1 by FcγRIII. The footprinting results are consistent with the previously published crystal structure of the IgG1 Fc/FcγRIII complex. Additionally, our FPOP results reveal the conformational changes in the Fab domain upon binding of the Fc domain to FcγRIII. These data demonstrate the value of footprinting as part of a comprehensive toolbox for identifying the changes in the higher-order structure of therapeutic antibodies in solution.
Collapse
Affiliation(s)
- Liuqing Shi
- Department of Chemistry , Washington University in St. Louis , Campus Box 1134, One Brookings Drive , St. Louis , Missouri 63130 , United States
| | - Tun Liu
- Bioproduct Research and Development, Lilly Research Laboratories , Eli Lilly and Company , Indianapolis , Indiana 46285 , United States
| | - Michael L Gross
- Department of Chemistry , Washington University in St. Louis , Campus Box 1134, One Brookings Drive , St. Louis , Missouri 63130 , United States
| | - Yining Huang
- Bioproduct Research and Development, Lilly Research Laboratories , Eli Lilly and Company , Indianapolis , Indiana 46285 , United States
| |
Collapse
|