GM-CSF: a Promising Target in Inflammation and Autoimmunity

Total Page:16

File Type:pdf, Size:1020Kb

GM-CSF: a Promising Target in Inflammation and Autoimmunity ImmunoTargets and Therapy Dovepress open access to scientific and medical research Open Access Full Text Article REVIEW GM-CSF: A Promising Target in Inflammation and Autoimmunity This article was published in the following Dove Press journal: ImmunoTargets and Therapy Kevin MC Lee 1 Abstract: The cytokine, granulocyte macrophage-colony stimulating factor (GM-CSF), was Adrian A Achuthan 1 firstly identified as being able to induce in vitro the proliferation and differentiation of bone John A Hamilton 1,2 marrow progenitors into granulocytes and macrophages. Much preclinical data have indi­ cated that GM-CSF has a wide range of functions across different tissues in its action on 1Department of Medicine, Royal Melbourne Hospital, The University of myeloid cells, and GM-CSF deletion/depletion approaches indicate its potential as an Melbourne, Melbourne, VIC, 3050, important therapeutic target in several inflammatory and autoimmune disorders, for example, 2 Australia; Australian Institute for rheumatoid arthritis. In this review, we discuss briefly the biology of GM-CSF, raise some Musculoskeletal Science (AIMSS), The University of Melbourne and Western current issues and questions pertaining to this biology, summarize the results from preclinical Health, Melbourne, VIC, Australia models of a range of inflammatory and autoimmune disorders and list the latest clinical trials evaluating GM-CSF blockade in such disorders. Keywords: GM-CSF, inflammation, autoimmunity, therapeutic Introduction Granulocyte macrophage-colony stimulating factor (GM-CSF, CSF2) was origin­ ally defined as a hemopoietic growth factor due to its ability to form colonies of granulocytes and macrophages in vitro by proliferation and differentiation of bone marrow progenitor cells.1 A number of reports have revealed that GM-CSF and the GM-CSF receptor (GM-CSFR) levels are elevated and correlated with disease severity in many inflammatory/autoimmune diseases, for example, rheumatoid arthritis (RA).2 In addition, there is much preclinical data providing a strong rationale for the involvement of GM-CSF in such diseases. As a result, GM-CSF and GM-CSFR have both attracted great interest as potential therapeutic targets. This review provides a brief overview of the pleiotropic biology of GM-CSF and outlines some of the most recent preclinical findings and in particular the resultant clinical studies using GM-CSF- or GM-CSFR-targeting monoclonal antibodies (mAbs) in various diseases. It also summarizes some of the contentious issues and outstanding questions pertaining to GM-CSF biology. This review cannot obviously cover all aspect of the broad topic and further background information on GM-CSF biology and targeting can be found in earlier reviews (see, for example,2–6). GM-CSF Biology GM-CSF Receptor and Signaling GM-CSF binds to the multimeric GM-CSFR, comprising a specificlow-affinity ligand- Correspondence: John A Hamilton Email [email protected] binding α subunit (GM-CSFRα) and a signal-transducing β subunit (GM-CSFRβ), the submit your manuscript | www.dovepress.com ImmunoTargets and Therapy 2020:9 225–240 225 DovePress © 2020 Lee et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php http://doi.org/10.2147/ITT.S262566 and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php). Lee et al Dovepress latter shared with the interleukin-3 (IL-3) and IL-5 receptors. hematopoietic, for example, T and B cells, and non- The activation of GM-CSFR triggers (i) phosphorylation of hematopoietic, for example, tissue resident cells. the GM-CSFRβ subunit, which commonly leads to the bind­ ing of signal transducer and activator of transcription 5 T Cells (STAT5) thereby initiating Janus Kinase (JAK) 2 GM-CSF has been reported to be produced by TH1 and TH 7–9 signaling and (ii) activation of the MEK/ERK, phosphati­ 17 cells via STAT signaling33–35 and shown to be crucial 10,11 dylinositol 3 kinase (PI3K) and NFκB pathways. for encephalitogenicity.33 A distinct subset of T cells, The hemopoietic-specific transcription factor, interferon known as GM-CSF-producing TH cells, has recently been regulatory factor 4 (IRF4), is a key signaling molecule for 35 identified. GM-CSF-producing TH cells express unde­ the adoption of dendritic cell (DC)-like properties in GM-CSF tectable T-bet, GATA-3, or RORγt and do not express 12–16 -treated precursors, such as monocytes. Recently, it was a cytokine signature that other T cell subsets express.35,36 reported that the GM-CSF stimulation of monocytes/macro­ In humans, GM-CSF-producing TH cells can be identified phages in vitro leads to the formation of CCL17 via IRF4 as an as CCR10+ CCR4+ CXCR3− CCR6− cells.36 GM-CSF- important pathway, termed the GM-CSF/CCL17 axis (see producing TH cells have been implicated in autoimmune below).17 Mechanistically, GM-CSF up-regulates IRF4 brain disease as fewer GM-CSF-producing TH cells corre­ 17 expression by enhancing JMJD3 demethylase activity. lated with less severe experimental autoimmune encepha­ Additionally, GM-CSF-IRF4 signaling favours the polariza­ lomyelitis (EAE).35 tion of pro-inflammatory macrophages and increased antigen presentation capability (ie increased MHC class II expression) during in vivo inflammation.18 In contrast, some literature Innate Lymphoid Cells The innate lymphoid cell (ILC) family encompasses the indicates IRF5, but not IRF4, to be important for GM-CSF- classic cytotoxic natural killer (NK) cells and the non- induced macrophage polarization19,20 and IRF4 has been con­ cytotoxic ILCs.37,38 NK cells have been recently reported sidered to have an anti-inflammatory role in macrophages (for to produce GM-CSF when infiltrating into joints for the example, enhanced interleukin (IL)-10 and reduced TNF maintenance of inflammatory arthritis.39 Among the sub­ production).21–23 sets of ILCs, type 3 ILCs have been shown to secrete GM- CSF in intestinal inflammation.40–42 In spondyloarthritis, GM-CSF and the Lung type 3 ILCs are found to be enriched in the inflamed joint While GM-CSF appears to be dispensable for steady state and are the predominate source of GM-CSF.43 myeloid cell development in vivo,24 GM-CSF directly reg­ ulates the differentiation of liver-derived fetal monocytes B Cells into immature alveolar macrophages during embryonic A subset of B cells, namely innate response activator (IRA) development25 and also promotes the development of func­ B cells, resides in nonlymphoid sites, such as the peritoneal tional alveolar macrophages via PU.1.25,26 GM-CSF gene- and pleural cavities, and provides a firstline of defense against deficient mice develop pulmonary alveolar proteinosis,24,27 infection.44 In a model of Toll-like receptor (TLR)-induced which can also occur in humans due to genetic mutations or sepsis, IRA B cells, which produce GM-CSF and also reported endogenous neutralizing antibody (53).28 This pathology to express the GM-CSFR, mediate a GM-CSF-dependent IgM results from compromised alveolar macrophage functions.29 protective mechanism against septic shock.44–46 Mixed chi­ In addition, GM-CSF deficientmice have been reported to be meric mice with B cell-restricted GM-CSF deficiency had more susceptible to lung infections.30,31 Inhaled GM-CSF high bacterial titer morbidity after infection but did not can protect mice from such infections30,32 via enhancing develop alveolar proteinosis,44 indicating that B cell-derived macrophage and DC function.30,31 These studies suggest GM-CSF is dispensable for surfactant clearance by alveolar that the availability of excess GM-CSF could be beneficial macrophages. Memory B cells isolated from multiple sclerosis in certain circumstances. (MS) patients produce high levels of GM-CSF, termed GM- CSF+ B cells, and co-culture of these cells with macrophage- Sources of GM-CSF in Inflammation colony stimulating factor (M-CSF)-generated, human blood A wide range of cells can produce GM-CSF.2–6 During monocyte-derived macrophages initiates proinflammatory inflammation, major sources of GM-CSF are both responses.47 Interestingly, dimethyl fumarate ameliorates MS 226 submit your manuscript | www.dovepress.com ImmunoT argets and Therapy 2020:9 DovePress Dovepress Lee et al and depletes GM-CSF-producing B cells in these patients48,49 alleviate experimental colitis.67 As a result, it has been consistent with a pathogenic role for these cells. recommended that the M1/M2 polarization terminology not be applied to GM-CSF action.2,17,61,72 Tissue Resident Cells GM-CSF also promotes the development of migratory Tissue resident cells can also be a potential source of GM- CD103+ CD11b+ DCs,40,73 while negatively regulating the CSF during inflammation. For example, it has been shown development of resident CD8+ DCs.74 GM-CSF, often in that fibroblast-like synoviocytes are important for the combination with IL-4, is widely used to generate in vitro initiation of experimental autoimmune arthritis via their murine and human DC populations from bone marrow GM-CSF production.50 GM-CSF has been reported to be precursors and blood monocytes,
Recommended publications
  • Presentation
    Strategies to support the COVID-19 response in LMICs A virtual seminar series Therapeutics Landscape for COVID-19 Natasha Mubeen Chida, MD MSPH Associate Program Director, Infectious Disease Fellowship Program Assistant Professor, Division of Infectious Diseases Johns Hopkins University School of Medicine Objectives • Review biological plausibility of Remdesivir for SARS-CoV-2 treatment • Review clinical data on Remdesivir • Review biological plausibility of Hydroxychloroquine for SARS-CoV-2 treatment • Review clinical data on Hydroxychloroquine • Discuss cytokine release syndrome in COVID-19 • Discuss use of anti IL-6 blockade for management of CRS • State ongoing clinical trials for Remdesivir, hydroxychloroquine, IL-6 blockade Sample of COVID-19 Therapeutic Landscape Antivirals Immune Modulators Other Baloxavir Anakinra ACEI/ARB Chloroquine/Hydroxychloroquine Convalescent Plasma Ascorbic Acid DAS-181 Corticosteroids Azithromycin Favipiravir IVIG Epoprostenol Interferon Lenzilumab Indomethacin Lopinavir/Ritonavir Ruxolitinib Ivermectin Neuraminidase inhibitors Sarilumab Niclosamide Remdesivir Sirolimus Nitazoxanide Ribavarin Tocilizumab Statins Umifenovir Acalabrutinib SARS-CoV-2 Liu C, et al. ACS Cent Sci. doi: 10.1021/acscentsci.0c00272 (2020). Jiang S, Hillyer C, Du L. Trends Immunol doi: 10.1016/j.it.2020.03.007 (2020). “Antivirals” Remdesivir • 2013 Ebola outbreak • CDC/USAMRIDD/Gilead Sciences identified nucleoside lead à prodrug, RDV • Metabolized to active form, adenosine nucleoside analog • Interferes with RNA polymerase • Evades
    [Show full text]
  • (COVID-19) in the Era of Cardiac Vigilance: a Systematic Review
    Journal of Clinical Medicine Review Repurposing Immunomodulatory Therapies against Coronavirus Disease 2019 (COVID-19) in the Era of Cardiac Vigilance: A Systematic Review Courtney M. Campbell 1,* , Avirup Guha 2 , Tamanna Haque 3, Tomas G. Neilan 4 and Daniel Addison 1,5 1 Cardio-Oncology Program, Division of Cardiology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43210, USA; [email protected] 2 Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, OH 44106, USA; [email protected] 3 Division of Hematology/Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43210, USA; [email protected] 4 Cardio-Oncology Program, Division of Cardiology, Department of Internal Medicine, Massachusetts General Hospital, Boston, MA 02144, USA; [email protected] 5 Division of Cancer Prevention and Control, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA * Correspondence: [email protected] Received: 23 July 2020; Accepted: 8 September 2020; Published: 11 September 2020 Abstract: The ongoing coronavirus disease 2019 (COVID-19) pandemic has resulted in efforts to identify therapies to ameliorate adverse clinical outcomes. The recognition of the key role for increased inflammation in COVID-19 has led to a proliferation of clinical trials targeting inflammation. The purpose of this review is to characterize the current state of immunotherapy trials in COVID-19, and focuses on associated cardiotoxicities, given the importance of pharmacovigilance. The search terms related to COVID-19 were queried in ClinicalTrials.gov. A total of 1621 trials were identified and screened for interventional trials directed at inflammation.
    [Show full text]
  • Q3 2020 Results
    Q3 2020 Results 28 October 2020 Cautionary statement regarding forward-looking statements This presentation may contain forward-looking statements. Forward-looking statements give the Group’s current expectations or forecasts of future events. An investor can identify these statements by the fact that they do not relate strictly to historical or current facts. They use words such as ‘anticipate’, ‘estimate’, ‘expect’, ‘intend’, ‘will’, ‘project’, ‘plan’, ‘believe’, ‘target’ and other words and terms of similar meaning in connection with any discussion of future operating or financial performance. In particular, these include statements relating to future actions, prospective products or product approvals, future performance or results of current and anticipated products, sales efforts, expenses, the outcome of contingencies such as legal proceedings, dividend payments and financial results. Other than in accordance with its legal or regulatory obligations (including under the Market Abuse Regulations, UK Listing Rules and the Disclosure Guidance and Transparency Rules of the Financial Conduct Authority), the Group undertakes no obligation to update any forward-looking statements, whether as a result of new information, future events or otherwise. Investors should, however, consult any additional disclosures that the Group may make in any documents which it publishes and/or files with the US Securities and Exchange Commission (SEC). All investors, wherever located, should take note of these disclosures. Accordingly, no assurance can be given that any particular expectation will be met and investors are cautioned not to place undue reliance on the forward-looking statements. Forward-looking statements are subject to assumptions, inherent risks and uncertainties, many of which relate to factors that are beyond the Group’s control or precise estimate.
    [Show full text]
  • Q3 2019 Results
    Q3 2019 Results 30 October 2019 Cautionary statement regarding forward-looking statements This presentation may contain forward-looking statements. Forward-looking statements give the Group’s current expectations or forecasts of future events. An investor can identify these statements by the fact that they do not relate strictly to historical or current facts. They use words such as ‘anticipate’, ‘estimate’, ‘expect’, ‘intend’, ‘will’, ‘project’, ‘plan’, ‘believe’, ‘target’ and other words and terms of similar meaning in connection with any discussion of future operating or financial performance. In particular, these include statements relating to future actions, prospective products or product approvals, future performance or results of current and anticipated products, sales efforts, expenses, the outcome of contingencies such as legal proceedings, dividend payments and financial results. Other than in accordance with its legal or regulatory obligations (including under the Market Abuse Regulations, UK Listing Rules and the Disclosure Guidance and Transparency Rules of the Financial Conduct Authority), the Group undertakes no obligation to update any forward-looking statements, whether as a result of new information, future events or otherwise. Investors should, however, consult any additional disclosures that the Group may make in any documents which it publishes and/or files with the US Securities and Exchange Commission (SEC). All investors, wherever located, should take note of these disclosures. Accordingly, no assurance can be given that any particular expectation will be met and investors are cautioned not to place undue reliance on the forward-looking statements. Forward-looking statements are subject to assumptions, inherent risks and uncertainties, many of which relate to factors that are beyond the Group’s control or precise estimate.
    [Show full text]
  • Inflammatory Response in Children & Adults
    INFLAMMATORY RESPONSE IN CHILDREN & ADULTS JUNE 3, 2020 WEBINAR SERIES PARTNERS American Academy of Clinical Toxicology (AACT) American Academy of Emergency Medicine (AAEM) American Academy of Emergency Nurse Practitioners (AAENP) American Association of Poison Control Centers (AAPCC) American College of Medical Toxicology (ACMT) Asia Pacific Association of Medical Toxicologists (APAMT) European Association of Poison Centers and Clinical Toxicologists (EAPCCT) Middle East & North Africa Clinical Toxicology Association (MENATOX) ON-DEMAND RESOURCES All webinars are recorded and www.acmt.net/covid19web Questions? posted to the ACMT website Write to: [email protected] Q&A will be at end of the Webinar Please type your questions into the Q&A or Chat function during the webinar and we will get to as many as we can We monitor all platforms, including YouTube and Facebook, for questions MODERATORS Paul M. Wax, MD FACMT ¡ Executive Director, American College of Medical Toxicology (ACMT) Ziad Kazzi, MD, FACMT ¡ Board Member, American College of Medical Toxicology (ACMT) ¡ President, Middle East & North Africa Clinical Toxicology Association (MENATOX) MULTISYSTEM INFLAMMATORY SYNDROME IN CHILDREN (MIS-C) MEDICAL AND PUBLIC HEALTH CONSIDERATIONS OF COVID-19 James Schneider, MD, FAAP, FCCP Chief, Pediatric Critical Care Medicine, Associate Professor of Pediatrics, Cohen Children’s Medical Center, New Hyde Park, NY [email protected] CONFLICT OF THIS SPEAKER DOES NOT HAVE ANY CONFLICTS OF INTEREST INTEREST TO DISCLOSE Recognition of New disease?
    [Show full text]
  • Immunomodulators Under Evaluation for the Treatment of COVID-19
    Immunomodulators Under Evaluation for the Treatment of COVID-19 Last Updated: August 4, 2021 Summary Recommendations See Therapeutic Management of Hospitalized Adults with COVID-19 for the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations on the use of the following immunomodulators for patients according to their disease severity: • Baricitinib with dexamethasone • Dexamethasone • Tocilizumab with dexamethasone Additional Recommendations There is insufficient evidence for the Panel to recommend either for or against the use of the following immunomodulators for the treatment of COVID-19: • Colchicine for nonhospitalized patients • Fluvoxamine • Granulocyte-macrophage colony-stimulating factor inhibitors for hospitalized patients • Inhaled budesonide • Interleukin (IL)-1 inhibitors (e.g., anakinra) • Interferon beta for the treatment of early (i.e., <7 days from symptom onset) mild to moderate COVID-19 • Sarilumab for patients who are within 24 hours of admission to the intensive care unit (ICU) and who require invasive mechanical ventilation, noninvasive ventilation, or high-flow oxygen (>0.4 FiO2/30 L/min of oxygen flow) The Panel recommends against the use of the following immunomodulators for the treatment of COVID-19, except in a clinical trial: • Baricitinib with tocilizumab (AIII) • Interferons (alfa or beta) for the treatment of severely or critically ill patients with COVID-19 (AIII) • Kinase inhibitors: • Bruton’s tyrosine kinase inhibitors (e.g., acalabrutinib, ibrutinib, zanubrutinib) (AIII) • Janus kinase inhibitors other than baricitinib (e.g., ruxolitinib, tofacitinib) (AIII) • Non-SARS-CoV-2-specific intravenous immunoglobulin (IVIG) (AIII). This recommendation should not preclude the use of IVIG when it is otherwise indicated for the treatment of complications that arise during the course of COVID-19.
    [Show full text]
  • Promising Therapeutic Targets for Treatment of Rheumatoid Arthritis
    REVIEW published: 09 July 2021 doi: 10.3389/fimmu.2021.686155 Promising Therapeutic Targets for Treatment of Rheumatoid Arthritis † † Jie Huang 1 , Xuekun Fu 1 , Xinxin Chen 1, Zheng Li 1, Yuhong Huang 1 and Chao Liang 1,2* 1 Department of Biology, Southern University of Science and Technology, Shenzhen, China, 2 Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China Rheumatoid arthritis (RA) is a systemic poly-articular chronic autoimmune joint disease that mainly damages the hands and feet, which affects 0.5% to 1.0% of the population worldwide. With the sustained development of disease-modifying antirheumatic drugs (DMARDs), significant success has been achieved for preventing and relieving disease activity in RA patients. Unfortunately, some patients still show limited response to DMARDs, which puts forward new requirements for special targets and novel therapies. Understanding the pathogenetic roles of the various molecules in RA could facilitate discovery of potential therapeutic targets and approaches. In this review, both Edited by: existing and emerging targets, including the proteins, small molecular metabolites, and Trine N. Jorgensen, epigenetic regulators related to RA, are discussed, with a focus on the mechanisms that Case Western Reserve University, result in inflammation and the development of new drugs for blocking the various United States modulators in RA. Reviewed by: Åsa Andersson, Keywords: rheumatoid arthritis, targets, proteins, small molecular metabolites, epigenetic regulators Halmstad University, Sweden Abdurrahman Tufan, Gazi University, Turkey *Correspondence: INTRODUCTION Chao Liang [email protected] Rheumatoid arthritis (RA) is classified as a systemic poly-articular chronic autoimmune joint † disease that primarily affects hands and feet.
    [Show full text]
  • 2017 Immuno-Oncology Medicines in Development
    2017 Immuno-Oncology Medicines in Development Adoptive Cell Therapies Drug Name Organization Indication Development Phase ACTR087 + rituximab Unum Therapeutics B-cell lymphoma Phase I (antibody-coupled T-cell receptor Cambridge, MA www.unumrx.com immunotherapy + rituximab) AFP TCR Adaptimmune liver Phase I (T-cell receptor cell therapy) Philadelphia, PA www.adaptimmune.com anti-BCMA CAR-T cell therapy Juno Therapeutics multiple myeloma Phase I Seattle, WA www.junotherapeutics.com Memorial Sloan Kettering New York, NY anti-CD19 "armored" CAR-T Juno Therapeutics recurrent/relapsed chronic Phase I cell therapy Seattle, WA lymphocytic leukemia (CLL) www.junotherapeutics.com Memorial Sloan Kettering New York, NY anti-CD19 CAR-T cell therapy Intrexon B-cell malignancies Phase I Germantown, MD www.dna.com ZIOPHARM Oncology www.ziopharm.com Boston, MA anti-CD19 CAR-T cell therapy Kite Pharma hematological malignancies Phase I (second generation) Santa Monica, CA www.kitepharma.com National Cancer Institute Bethesda, MD Medicines in Development: Immuno-Oncology 1 Adoptive Cell Therapies Drug Name Organization Indication Development Phase anti-CEA CAR-T therapy Sorrento Therapeutics liver metastases Phase I San Diego, CA www.sorrentotherapeutics.com TNK Therapeutics San Diego, CA anti-PSMA CAR-T cell therapy TNK Therapeutics cancer Phase I San Diego, CA www.sorrentotherapeutics.com Sorrento Therapeutics San Diego, CA ATA520 Atara Biotherapeutics multiple myeloma, Phase I (WT1-specific T lymphocyte South San Francisco, CA plasma cell leukemia www.atarabio.com
    [Show full text]
  • Looking for Therapeutic Antibodies in Next Generation Sequencing Repositories
    bioRxiv preprint doi: https://doi.org/10.1101/572958; this version posted March 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Title: Looking for Therapeutic Antibodies in Next Generation Sequencing Repositories. Authors: Konrad Krawczyk1*, Matthew Raybould2, Aleksandr Kovaltsuk2, Charlotte M. Deane2 1 NaturalAntibody, Hamburg, Germany 2 Oxford University Department of Statistics, Oxford, UK *Correspondence to [email protected] Abstract: Recently it has become possible to query the great diversity of natural antibody repertoires using Next Generation Sequencing (NGS). These methods are capable of producing millions of sequences in a single experiment. Here we compare Clinical Stage Therapeutic antibodies to the ~1b sequences from 60 independent sequencing studies in the Observed Antibody Space Database. Of the 242 post Phase I antibodies, we find 16 with sequence identity matches of 95% or better for both heavy and light chains. There are also 54 perfect matches to therapeutic CDR-H3 regions in the NGS outputs, suggesting a nontrivial amount of convergence between naturally observed sequences and those developed artificially. This has potential implications for both the discovery of antibody therapeutics and the legal protection of commercial antibodies. Introduction Antibodies are proteins in jawed vertebrates that recognize noxious molecules (antigens) for elimination. An organism expresses millions of diverse antibodies to increase the chances that some of them will be able to bind the foreign antigen, initiating the adaptive immune response.
    [Show full text]
  • Antibodies to Watch in 2021 Hélène Kaplona and Janice M
    MABS 2021, VOL. 13, NO. 1, e1860476 (34 pages) https://doi.org/10.1080/19420862.2020.1860476 PERSPECTIVE Antibodies to watch in 2021 Hélène Kaplona and Janice M. Reichert b aInstitut De Recherches Internationales Servier, Translational Medicine Department, Suresnes, France; bThe Antibody Society, Inc., Framingham, MA, USA ABSTRACT ARTICLE HISTORY In this 12th annual installment of the Antibodies to Watch article series, we discuss key events in antibody Received 1 December 2020 therapeutics development that occurred in 2020 and forecast events that might occur in 2021. The Accepted 1 December 2020 coronavirus disease 2019 (COVID-19) pandemic posed an array of challenges and opportunities to the KEYWORDS healthcare system in 2020, and it will continue to do so in 2021. Remarkably, by late November 2020, two Antibody therapeutics; anti-SARS-CoV antibody products, bamlanivimab and the casirivimab and imdevimab cocktail, were cancer; COVID-19; Food and authorized for emergency use by the US Food and Drug Administration (FDA) and the repurposed Drug Administration; antibodies levilimab and itolizumab had been registered for emergency use as treatments for COVID-19 European Medicines Agency; in Russia and India, respectively. Despite the pandemic, 10 antibody therapeutics had been granted the immune-mediated disorders; first approval in the US or EU in 2020, as of November, and 2 more (tanezumab and margetuximab) may Sars-CoV-2 be granted approvals in December 2020.* In addition, prolgolimab and olokizumab had been granted first approvals in Russia and cetuximab saratolacan sodium was first approved in Japan. The number of approvals in 2021 may set a record, as marketing applications for 16 investigational antibody therapeutics are already undergoing regulatory review by either the FDA or the European Medicines Agency.
    [Show full text]
  • Bispecific T-Cell Redirection Versus Chimeric Antigen Receptor (CAR)-T Cells As Approaches to Kill Cancer Cells
    Bispecific T-Cell Redirection versus Chimeric Antigen Receptor (CAR)-T Cells as Approaches to Kill Cancer Cells William R. Strohl 1,* and Michael Naso 2 1 BiStro Biotech Consulting, LLC, 1086 Tullo Farm Rd., Bridgewater, NJ 08807, USA 2 Century Therapeutics, 3675 Market St., Philadelphia, PA 19104, USA * Correspondence: [email protected]; Tel.: +1-908-745-8576 Received: 27 May 2019; Accepted: 24 June 2019; Published: 3 July 2019 Abstract: The concepts for T-cell redirecting bispecific antibodies (TRBAs) and chimeric antigen receptor (CAR)-T cells are both at least 30 years old but both platforms are just now coming into age. Two TRBAs and two CAR-T cell products have been approved by major regulatory agencies within the last ten years for the treatment of hematological cancers and an additional 53 TRBAs and 246 CAR cell constructs are in clinical trials today. Two major groups of TRBAs include small, short- half-life bispecific antibodies that include bispecific T-cell engagers (BiTE®s) which require continuous dosing and larger, mostly IgG-like bispecific antibodies with extended pharmacokinetics that can be dosed infrequently. Most CAR-T cells today are autologous, although significant strides are being made to develop off-the-shelf, allogeneic CAR-based products. CAR-Ts form a cytolytic synapse with target cells that is very different from the classical immune synapse both physically and mechanistically, whereas the TRBA-induced synapse is similar to the classic immune synapse. Both TRBAs and CAR-T cells are highly efficacious in clinical trials but both also present safety concerns, particularly with cytokine release syndrome and neurotoxicity.
    [Show full text]
  • Cellular Immunology the Role of B Cells in Multiple Sclerosis
    Cellular Immunology 339 (2019) 10–23 Contents lists available at ScienceDirect Cellular Immunology journal homepage: www.elsevier.com/locate/ycimm Research paper ☆ The role of B cells in multiple sclerosis: Current and future therapies T ⁎ Austin Negrona, Rachel R. Robinsona, Olaf Stüveb,c, Thomas G. Forsthubera, a Department of Biology, University of Texas at San Antonio, TX 78249, USA b Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA c Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA ABSTRACT While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies. 1. Introduction oligoclonal bands [12–14]. However, unlike what is observed in NMO and MG, there is significant heterogeneity in the antigen specificity of The contribution of B cells to autoimmune disease pathogenesis has these oligoclonal antibodies, which may target pathogens as well as historically been viewed primarily via the production of pathogenic autoantigens [8].
    [Show full text]