Chemical Aspects of Metal Ion Chelation in the Synthesis and Application Antibody-Based Radiotracers

Total Page:16

File Type:pdf, Size:1020Kb

Chemical Aspects of Metal Ion Chelation in the Synthesis and Application Antibody-Based Radiotracers Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2018 Chemical aspects of metal ion chelation in the synthesis and application antibody-based radiotracers Boros, Eszter ; Holland, Jason P Abstract: Radiometals are becoming increasingly accessible and are utilized frequently in the design of ra- diotracers for imaging and therapy. Nuclear properties ranging from the emission of ฀‐rays and ฀+‐particles (imaging) to Auger electron and ฀− and ฀‐particles (therapy) in combination with long half‐lives are ide- ally matched with the relatively long biological half‐life of monoclonal antibodies in vivo. Radiometal labeling of antibodies requires the incorporation of a metal chelate onto the monoclonal antibody. This chelate must coordinate the metal under mild conditions required for the handling of antibodies, as well as provide high kinetic, thermodynamic, and metabolic stability once the metal ion is coordinated to prevent release of the radionuclide before the target site is reached in vivo. Herein, we review the role of different radiometals that have found applications of the design of radiolabeled antibodies for imaging and radioimmunotherapy. Each radionuclide is described regarding its nuclear synthesis, coordinative preference, and radiolabeling properties with commonly used and novel chelates, as well as examples of their preclinical and clinical applications. An overview of recent trends in antibody‐based radiopharma- ceuticals is provided to spur continued development of the chemistry and application of radiometals for imaging and therapy. DOI: https://doi.org/10.1002/jlcr.3590 Posted at the Zurich Open Repository and Archive, University of Zurich ZORA URL: https://doi.org/10.5167/uzh-167582 Journal Article Accepted Version Originally published at: Boros, Eszter; Holland, Jason P (2018). Chemical aspects of metal ion chelation in the synthesis and application antibody-based radiotracers. Journal of Labelled Compounds and Radiopharmaceuticals, 61(9):652-671. DOI: https://doi.org/10.1002/jlcr.3590 Chemical aspects of metal ion chelation in the synthesis and application antibody-based radiotracers Eszter Boros1* and Jason P. Holland2* 1 Stony Brook University, Department of Chemistry, 100 Nicolls road, 11790 Stony Brook, NY, United States 2 University of Zurich, Department of Chemistry, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland * Correspondence: Eszter Boros Tel: +1.631.632.8572 E-mail: [email protected] Jason P. Holland Tel: +41.44.63.53990 E-mail: [email protected] Running Title: Metal coordination chemistry for radiolabelling antibodies Word count (Main text): Page 1 Abstract Radiometals are becoming increasingly accessible and are utilized frequently in the design of radiotracers for imaging and therapy. Nuclear properties ranging from the emission of g-ray and b+-particles (imaging) to Auger electron, b- and a-particles (therapy) in combination with long half-lives are ideally matched with the relatively long biological half-life of monoclonal antibodies in vivo. Radiometal labelling of antibodies requires the incorporation of a metal chelate onto the monoclonal antibody. This chelate must coordinate the metal under mild conditions required for the handling of antibodies, as well as provide high kinetic, thermodynamic and metabolic stability once the metal ion is coordinated in order to prevent in vivo release of the radionuclide before the target site is reached. Herein, we review the role of different radiometals that have found applications the design of radiolabelled antibodies for imaging and radioimmunotherapy. Each radionuclide is described with regard to its nuclear synthesis, coordinative preference, and radiolabelling properties with commonly used chelates. A summary of recent developments in the area of chelate synthesis, as well as examples of their preclinical and clinical applications is presented. The aims of review are to provide an overview of recent trends in antibody-based radiopharmaceuticals and to spur continued development of the chemistry and application of radiometals for imaging and therapy. Key words Radiometals, chelates, coordination chemistry, immuno-PET, radioimmunotherapy Page 2 Introduction In the pharmaceutical industry, biological products (biologics) and related biosimilar compounds represent one of the fastest growth sectors. In 2016, the size of the biologics market was estimated to be ~$200 billion USD. Recent forecasts predict continued growth at around 10% per annum with the market reaching ~$480 billion USD by 2024. Biologics include recombinant hormones and proteins, as well as cellular- and gene-based therapies, but the majority of this growth is driven by new vaccines and monoclonal antibodies (mAbs). Monoclonal antibodies or antibody-drug conjugates (ADCs) represent the single most important branch of biologics.1 Between 2011 and August 2017, the United States Federal Drug Administration (US-FDA) approved a total of 235 New Molecular Entities (NMEs), of which 37 were either mAbs or ADCs for therapy (Figure 1). This growing trend is supported by historical data which suggest that biologics, and mAbs in particular, have received higher approval rates than conventional small-molecule drugs.2 With prescription costs for mAb-based treatments estimated to be around 22-times more than small- molecule therapies, and profit margins often over 40%, it is easy to see the commercial attraction of biologics. However, this success is partially offset by the longer time frames required from initial clinical trials to full approval, more stringent manufacturing regulations, and the associated increase in costs. Recent changes to US legislation, implemented as part of the Patient Protection and Affordable Care Act of 2010, are also helping to create an abbreviated licensure pathway for NMEs that are demonstrated to be “biosimilar” to, or “interchangeable” with, an FDA-licensed biological product. This new legal framework will speed-up the discovery and clinical translation of mAb-based agents. In nuclear medicine, desirable biochemical properties including high target affinity, specificity and selectivity, pre-optimised pharmacokinetics, and relative ease of selection using Page 3 various library technologies mean that antibody-based radiotracers are attractive platforms for developing diagnostic imaging and radioimmunotherapy (RIT) agents.3–6 Nevertheless, production and validation of radiolabelled mAbs for use as immuno-single photon emission computed tomography (immuno-SPECT), immuno-positron emission tomography (immuno-PET) or RIT agents is non-trivial.3 Synthesis of radiolabelled mAbs requires chemical reactions on the protein (Figure 2). Functionalisation usually involves post-translational modification of amino-acid side chains (particularly peptide bond formation using the primary amine group of lysine residues), derivatisation of cysteine sulfhydryl groups, or site-specific labelling of glycans using chemical and/or enzymatic methods.7,8 More recent protein engineering routes have also exploited site- specific enzymatic ligation with prominent methods including transglutaminase derivatisation, sortase coupling and formylglycine reactions to produce a range of ADCs.7–9 While the chemical nature of the linker group plays an important role in determining the metabolic stability and pharmacokinetics of a radiolabelled mAb, an equally important decision revolves around the choice of the radiometal ion and the chelation chemistry used to produce thermodynamically and kinetically stable radiometal ion complexes.10 In this review, we discuss the chemical requirements for chelation of various radiometal ions and highlight selected applications of radiometal chemistry in the development of radiotracers for immuno-PET, immune-SPECT and RIT.11 1. Radionuclides for antibody imaging and therapy In this issue of the Journal of Labelled Compounds and Radiopharmaceuticals, Engle and co- workers present an overview of the emerging radionuclides for use in combination with mAbs for diagnostic and RIT applications. Pertinent physical decay characteristics of 11 radiometal ions, and for comparison three different iodine radionuclides are presented in Table 1.12 When selecting Page 4 a radionuclide for labelling antibodies or antibody-related constructs, the most crucial decision is to ensure that the physical half-life of the radionuclide is a reasonable match to the expected 10 68 biological half-life of the radiotracer in vivo. For example, Ga (t1/2 = 67.7 min.) is an excellent radionuclide for producing PET radiotracers based on small-molecules and peptides but would be an inappropriate choice for developing immuno-PET radiotracers based on full-sized mAbs (molecular weight ~150 kDa). Experience has found that mAbs reach optimum target (usually tumour) uptake and present maximal tumour-to-background contrast ratios in mouse models at around 24 – 96 h post-administration. Hence, in this pharmacokinetic situation, 68Ga is likely to undergo complete decay before the biodistribution of a radiolabelled mAb reaches the optimum imaging window. Notably, in humans the pharmacokinetic profiles of radiolabelled mAbs are often slower than in rodents, with optimum target accumulation and image contrast achieved at time points beyond 72 h post-administration of the radiotracer. Such prolonged circulation times necessitate the use of radionuclides that have physical half-lives ranging from ca. 10 hours
Recommended publications
  • For Acute Myeloid Leukemia
    Published OnlineFirst September 21, 2010; DOI: 10.1158/1078-0432.CCR-10-0382 Published OnlineFirst on September 21, 2010 as 10.1158/1078-0432.CCR-10-0382 Clinical Therapy Clinical Cancer Research Sequential Cytarabine and α-Particle Immunotherapy with Bismuth-213–Lintuzumab (HuM195) for Acute Myeloid Leukemia Todd L. Rosenblat1, Michael R. McDevitt1, Deborah A. Mulford1, Neeta Pandit-Taskar1, Chaitanya R. Divgi1, Katherine S. Panageas1, Mark L. Heaney1, Suzanne Chanel1, Alfred Morgenstern2, George Sgouros1, Steven M. Larson1, David A. Scheinberg1, and Joseph G. Jurcic1 Abstract Purpose: Lintuzumab (HuM195), a humanized anti-CD33 antibody, targets myeloid leukemia cells and has modest single-agent activity against acute myeloid leukemia (AML). To increase the potency of the antibody without the nonspecific cytotoxicity associated with β-emitters, the α-particle–emitting ra- dionuclide bismuth-213 (213Bi) was conjugated to lintuzumab. This phase I/II trial was conducted to determine the maximum tolerated dose (MTD) and antileukemic effects of 213Bi-lintuzumab, the first targeted α-emitter, after partially cytoreductive chemotherapy. Experimental Design: Thirty-one patients with newly diagnosed (n = 13) or relapsed/refractory (n = 18) AML (median age, 67 years; range, 37-80) were treated with cytarabine (200 mg/m2/d) for 5 days followed by 213Bi-lintuzumab (18.5-46.25 MBq/kg). Results: The MTD of 213Bi-lintuzumab was 37 MB/kg; myelosuppression lasting >35 days was dose limiting. Extramedullary toxicities were primarily limited to grade ≤2 events, including infusion-related reactions. Transient grade 3/4 liver function abnormalities were seen in five patients (16%). Treatment- related deaths occurred in 2 of 21 (10%) patients who received the MTD.
    [Show full text]
  • USAN Naming Guidelines for Monoclonal Antibodies |
    Monoclonal Antibodies In October 2008, the International Nonproprietary Name (INN) Working Group Meeting on Nomenclature for Monoclonal Antibodies (mAb) met to review and streamline the monoclonal antibody nomenclature scheme. Based on the group's recommendations and further discussions, the INN Experts published changes to the monoclonal antibody nomenclature scheme. In 2011, the INN Experts published an updated "International Nonproprietary Names (INN) for Biological and Biotechnological Substances—A Review" (PDF) with revisions to the monoclonal antibody nomenclature scheme language. The USAN Council has modified its own scheme to facilitate international harmonization. This page outlines the updated scheme and supersedes previous schemes. It also explains policies regarding post-translational modifications and the use of 2-word names. The council has no plans to retroactively change names already coined. They believe that changing names of monoclonal antibodies would confuse physicians, other health care professionals and patients. Manufacturers should be aware that nomenclature practices are continually evolving. Consequently, further updates may occur any time the council believes changes are necessary. Changes to the monoclonal antibody nomenclature scheme, however, should be carefully considered and implemented only when necessary. Elements of a Name The suffix "-mab" is used for monoclonal antibodies, antibody fragments and radiolabeled antibodies. For polyclonal mixtures of antibodies, "-pab" is used. The -pab suffix applies to polyclonal pools of recombinant monoclonal antibodies, as opposed to polyclonal antibody preparations isolated from blood. It differentiates polyclonal antibodies from individual monoclonal antibodies named with -mab. Sequence of Stems and Infixes The order for combining the key elements of a monoclonal antibody name is as follows: 1.
    [Show full text]
  • Challenges and Approaches for the Development of Safer Immunomodulatory Biologics
    REVIEWS Challenges and approaches for the development of safer immunomodulatory biologics Jean G. Sathish1*, Swaminathan Sethu1*, Marie-Christine Bielsky2, Lolke de Haan3, Neil S. French1, Karthik Govindappa1, James Green4, Christopher E. M. Griffiths5, Stephen Holgate6, David Jones2, Ian Kimber7, Jonathan Moggs8, Dean J. Naisbitt1, Munir Pirmohamed1, Gabriele Reichmann9, Jennifer Sims10, Meena Subramanyam11, Marque D. Todd12, Jan Willem Van Der Laan13, Richard J. Weaver14 and B. Kevin Park1 Abstract | Immunomodulatory biologics, which render their therapeutic effects by modulating or harnessing immune responses, have proven their therapeutic utility in several complex conditions including cancer and autoimmune diseases. However, unwanted adverse reactions — including serious infections, malignancy, cytokine release syndrome, anaphylaxis and hypersensitivity as well as immunogenicity — pose a challenge to the development of new (and safer) immunomodulatory biologics. In this article, we assess the safety issues associated with immunomodulatory biologics and discuss the current approaches for predicting and mitigating adverse reactions associated with their use. We also outline how these approaches can inform the development of safer immunomodulatory biologics. Immunomodulatory Biologics currently represent more than 30% of licensed The high specificity of the interactions of immu- biologics pharmaceutical products and have expanded the thera- nomodulatory biologics with their relevant immune Biotechnology-derived peutic options available
    [Show full text]
  • New Biological Therapies: Introduction to the Basis of the Risk of Infection
    New biological therapies: introduction to the basis of the risk of infection Mario FERNÁNDEZ RUIZ, MD, PhD Unit of Infectious Diseases Hospital Universitario “12 de Octubre”, Madrid ESCMIDInstituto de Investigación eLibraryHospital “12 de Octubre” (i+12) © by author Transparency Declaration Over the last 24 months I have received honoraria for talks on behalf of • Astellas Pharma • Gillead Sciences • Roche • Sanofi • Qiagen Infections and biologicals: a real concern? (two-hour symposium): New biological therapies: introduction to the ESCMIDbasis of the risk of infection eLibrary © by author Paul Ehrlich (1854-1915) • “side-chain” theory (1897) • receptor-ligand concept (1900) • “magic bullet” theory • foundation for specific chemotherapy (1906) • Nobel Prize in Physiology and Medicine (1908) (together with Metchnikoff) Infections and biologicals: a real concern? (two-hour symposium): New biological therapies: introduction to the ESCMIDbasis of the risk of infection eLibrary © by author 1981: B-1 antibody (tositumomab) anti-CD20 monoclonal antibody 1997: FDA approval of rituximab for the treatment of relapsed or refractory CD20-positive NHL 2001: FDA approval of imatinib for the treatment of chronic myelogenous leukemia Infections and biologicals: a real concern? (two-hour symposium): New biological therapies: introduction to the ESCMIDbasis of the risk of infection eLibrary © by author Functional classification of targeted (biological) agents • Agents targeting soluble immune effector molecules • Agents targeting cell surface receptors
    [Show full text]
  • Study Protocol
    PROTOCOL SYNOPSIS A Multicentre, Randomised, Double-blind, Placebo-controlled, Phase 3 Study Evaluating the Efficacy and Safety of Two Doses of Anifrolumab in Adult Subjects with Active Systemic Lupus Erythematosus International Coordinating Investigator Study site(s) and number of subjects planned Approximately 450 subjects are planned at approximately 173 sites. Study period Phase of development Estimated date of first subject enrolled Q2 2015 3 Estimated date of last subject completed Q2 2018 Study design This is a Phase 3, multicentre, multinational, randomised, double-blind, placebo-controlled study to evaluate the efficacy and safety of an intravenous treatment regimen of anifrolumab (150 mg or 300 mg) versus placebo in subjects with moderately to severely active, autoantibody-positive systemic lupus erythematosus (SLE) while receiving standard of care (SOC) treatment. The study will be performed in adult subjects aged 18 to 70 years of age. Approximately 450 subjects receiving SOC treatment will be randomised in a 1:2:2 ratio to receive a fixed intravenous dose of 150 mg anifrolumab, 300 mg anifrolumab, or placebo every 4 weeks (Q4W) for a total of 13 doses (Week 0 to Week 48), with the primary endpoint evaluated at the Week 52 visit. Investigational product will be administered as an intravenous (IV) infusion via an infusion pump over a minimum of 30 minutes, Q4W. Subjects must be taking either 1 or any combination of the following: oral corticosteroids (OCS), antimalarial, and/or immunosuppressants. Randomisation will be stratified using the following factors: SLE Disease Activity Index 2000 (SLEDAI-2K) score at screening (<10 points versus ≥10 points); Week 0 (Day 1) OCS dose 2(125) Revised Clinical Study Protocol Drug Substance Anifrolumab (MEDI-546) Study Code D3461C00005 Edition Number 5 Date 18 May 2016 (<10 mg/day versus ≥10 mg/day prednisone or equivalent); and results of a type 1 interferon (IFN) test (high versus low).
    [Show full text]
  • Monoclonal Antibody Playbook
    Federal Response to COVID-19: Monoclonal Antibody Clinical Implementation Guide Outpatient administration guide for healthcare providers 2 SEPTEMBER 2021 1 Introduction to COVID-19 Monoclonal Antibody Therapy 2 Overview of Emergency Use Authorizations 3 Site and Patient Logistics Site preparation Patient pathways to monoclonal administration 4 Team Roles and Responsibilities Leadership Administrative Clinical Table of 5 Monoclonal Antibody Indications and Administration Indications Contents Preparation Administration Response to adverse events 6 Supplies and Resources Infrastructure Administrative Patient Intake Administration 7 Examples: Sites of Administration and Staffing Patterns 8 Additional Resources 1 1. Introduction to Monoclonal Therapy 2 As of 08/13/21 Summary of COVID-19 Therapeutics 1 • No Illness . Health, no infections • Exposed Asymptomatic Infected . Scope of this Implementation Guide . Not hospitalized, no limitations . Monoclonal Antibodies for post-exposure prophylaxis (Casirivimab + Imdevimab (RGN)) – EUA Issued. • Early Symptomatic . Scope of this Implementation Guide . Not hospitalized, with limitations . Monoclonal Antibodies for treatment (EUA issued): Bamlanivimab + Etesevimab1 (Lilly) Casirivimab + Imdevimab (RGN) Sotrovimab (GSK/Vir) • Hospital Adminission. Treated with Remdesivir (FDA Approved) or Tocilizumab (EUA Issued) . Hospitalized, no acute medical problems . Hospitalized, not on oxygen . Hospitlaized, on oxygen • ICU Admission . Hospitalized, high flow oxygen, non-invasive ventilation
    [Show full text]
  • REGENERON PHARMACEUTICALS, INC. (Exact Name of Registrant As Specified in Charter)
    UNITED STATES SECURITIES AND EXCHANGE COMMISSION Washington, DC 20549 FORM 8-K CURRENT REPORT Pursuant to Section 13 or 15(d) of the Securities Exchange Act of 1934 Date of Report (Date of earliest event reported): February 9, 2017 (February 9, 2017) REGENERON PHARMACEUTICALS, INC. (Exact Name of Registrant as Specified in Charter) New York 000-19034 13-3444607 (State or other jurisdiction (Commission (IRS Employer of Incorporation) File No.) Identification No.) 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707 (Address of principal executive offices, including zip code) (914) 847-7000 (Registrant's telephone number, including area code) Check the appropriate box below if the Form 8-K filing is intended to simultaneously satisfy the filing obligation of the registrant under any of the following provisions: ☐ Written communications pursuant to Rule 425 under the Securities Act (17 CFR 230.425) ☐ Soliciting material pursuant to Rule 14a-12 under the Exchange Act (17 CFR 240.14a-12) ☐ Pre-commencement communications pursuant to Rule 14d-2(b) under the Exchange Act (17 CFR 240.14d-2(b)) ☐ Pre-commencement communications pursuant to Rule 13e-4(c) under the Exchange Act (17 CFR 240.13e-4(c)) Item 2.02 Results of Operations and Financial Condition. On February 9, 2017, Regeneron Pharmaceuticals, Inc. issued a press release announcing its financial and operating results for the quarter and year ended December 31, 2016. A copy of the press release is being furnished to the Securities and Exchange Commission as Exhibit 99.1 to this Current Report on Form 8-K and is incorporated by reference to this Item 2.02.
    [Show full text]
  • The Future of Antibodies As Cancer Drugs
    REVIEWS Drug Discovery Today Volume 17, Numbers 17/18 September 2012 The biopharmaceutical industry’s pipeline of anticancer antibodies includes 165 candidates with substantial diversity in composition, targets and mechanisms of action that hold promise to be the cancer drugs of the future. Reviews FOUNDATION REVIEW Foundation review: The future of antibodies as cancer drugs 1 2 Dr Janice Reichert Janice M. Reichert and Eugen Dhimolea is Research Assistant Professor at Tufts 1 Center for the Study of Drug Development, Tufts University School of Medicine, 75 Kneeland Street, University’s Center for the Study of Drug Development Suite 1100, Boston, MA 02111, USA 2 (CSDD). She is also Founder Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, 77 Louis Pasteur Ave., and Editor-in-Chief of mAbs, Harvard Institutes of Medicine, Room 309, Boston, MA 02215, USA a peer-reviewed, PubMed- indexed biomedical journal that focuses on topics relevant to antibody research Targeted therapeutics such as monoclonal antibodies (mAbs) have proven and development; President of the board of directors of The Antibody Society; and a member of the board successful as cancer drugs. To profile products that could be marketed in of the Peptide Therapeutics Foundation. At CSDD, the future, we examined the current commercial clinical pipeline of mAb Dr Reichert studies innovation in the pharmaceutical and biotechnology industries. Her work focuses on candidates for cancer. Our analysis revealed trends toward development of strategic analyses of investigational candidates and marketed products, with an emphasis on the clinical a variety of noncanonical mAbs, including antibody–drug conjugates development and approval of new therapeutics and (ADCs), bispecific antibodies, engineered antibodies and antibody vaccines.
    [Show full text]
  • CYRAMZA (Ramucirumab) Injection Label
    1 HIGHLIGHTS OF PRESCRIBING INFORMATION • 500 mg/50 mL (10 mg per mL) solution, single-dose vial (3) These highlights do not include all the information needed to use CYRAMZA safely and effectively. See full prescribing information ------------------------------- CONTRAINDICATIONS ------------------------------ for CYRAMZA. None CYRAMZA (ramucirumab) injection, for intravenous infusion ------------------------ WARNINGS AND PRECAUTIONS ----------------------- Initial U.S. Approval: 2014 • Arterial Thromboembolic Events (ATEs): Serious, sometimes fatal WARNING: HEMORRHAGE ATEs have been reported in clinical trials. Discontinue CYRAMZA for severe ATEs. (5.2) See full prescribing information for complete boxed warning. • Hypertension: Monitor blood pressure and treat hypertension. CYRAMZA increased the risk of hemorrhage, including severe Temporarily suspend CYRAMZA for severe hypertension. and sometimes fatal hemorrhagic events. Permanently Discontinue CYRAMZA for hypertension that cannot be medically discontinue CYRAMZA in patients who experience severe controlled. (5.3) bleeding [see Dosage and Administration (2.3), Warnings and • Infusion-Related Reactions: Monitor for signs and symptoms Precautions (5.1)]. during infusion. (5.4) • Gastrointestinal Perforation: Discontinue CYRAMZA. (5.5) --------------------------- RECENT MAJOR CHANGES -------------------------- • Impaired Wound Healing: Withhold CYRAMZA prior to surgery. Indications and Usage 11/2014 (5.6) • Clinical Deterioration in Patients with Cirrhosis: New onset or Dosage and Administration:
    [Show full text]
  • COVID-19 Immunotherapy: Novel Humanized 47D11 Monoclonal Antibody
    Review Article ISSN: 2574 -1241 DOI: 10.26717/BJSTR.2020.29.004828 COVID-19 Immunotherapy: Novel Humanized 47D11 Monoclonal Antibody Basma H Marghani* Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Egypt *Corresponding author: Basma H Marghani, Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Egypt ARTICLE INFO ABSTRACT Received: August 10, 2020 Published: August 18, 2020 rapidly to the other countries all over the world, caused a novel Coronavirus-2019 (COV- SARS-CoV-2 was appeared firstly in Wuhan, China in December 2019, then spread- gency. After infection, virus entry the host cell through spike proteins (S)- angiotensin Citation: Basma H Marghani. COVID-19 ID-19). The World Health Organization was branded COVID-19 as a global health emer Immunotherapy: Novel Humanized 47D11 converting enzyme 2 (ACE2) cell membrane receptor via their S-Binding domain, then Monoclonal Antibody. Biomed J Sci & Tech virus replication caused acute respiratory disease syndrome (ARDS). Till now there was Res 29(4)-2020. BJSTR. MS.ID.004828. no vaccines or anti-viral drugs for coronavirus infection. One effective treatment is the use of human monoclonal antibodies (mAbs) which are engineered to target and block Keywords: COVID-19; Immunotherapy; cell surface receptor. mAbs could be given to people in the early stages of COVID-19 as a Full-Length Humanized Monoclonal Anti- therapeutic, or used prophylactically to give immediate, long-term immunity to vulnera- bodies; ACE Inhibitor ble people such as healthcare workers. The neutralizing humanized 47D11 monoclonal antibody targets a common epitope on SARS-CoV2 virus and may offer potential for pre- Abbreviations: ACE2: Angiotensin-Con- vention and treatment of COVID-19.89U (Graphical Abstract 1).
    [Show full text]
  • Guide to Treatment Role of Antibodies in Acute Leukemia: Focus on Transplant-Eligible Populations
    PRACTICE AID Guide to Treatment Role of Antibodies in Acute Leukemia: Focus on Transplant-Eligible Populations Acute Myeloid Leukemia DRUG STATUS TARGET DOSE EU EMA approved: In combination with daunorubicin and Induction: 3 mg/m2 (up to 5 mg/m2 in EU and 4.5 mg/m2 in US) cytarabine for pts aged ≥15 y with previously untreated, on d 1, 4, and 7 in combination with daunorubicin and cytarabine de novo CD33+ AML, except APL CD33 Consolidation: 3 mg/m2 (up to 5 mg/m2 in EU and 4.5 mg/m2 in US) Gemtuzumab US FDA approved: Newly diagnosed CD33+ AML in adults and 1,2 on d 1 in combination with daunorubicin and cytarabine ozogamicin pediatric pts aged ≥1 month; R/R CD33+ AML in adults and Please consult label for single-agent dosing and dosing in R/R AML. pediatric pts aged 2 y Phase 3 SIERRA study CD45 Adults aged ≥55 y with active R/R AML, adequate organ Dosimetry directed (SIERRA study) Iomab-B3 function, and related/unrelated matched donor (BC8 mAb linked to radioisotope iodine-131) Acute Lymphoblastic Leukemia EU EMA approved: Adults with R/R CD22+ B-cell ALL; adults Cycle 1 dose: 0.8 mg/m2 on d 1; 0.5 mg/m2 on days 8 and 15 for 21 d 2 Inotuzumab with Ph+ R/R B-cell ALL who failed treatment with at least one TKI CD22 Patients in CR/CRi: 0.5 mg/m on days 1, 8, and 15 for 28 d ozogamicin4,5 US FDA approved: Adults with R/R CD22+ B-cell ALL Patients not in CR/CRi: Use cycle 1 dose for 28 d EU Dose: By patient’s weight (≥45 kg = 9 mcg/d on d 1-7 and 28 mcg/d on EU EMA approved: Adults with B-cell ALL who are in d 8-28; <45 kg = 5 mcg/m2/d on
    [Show full text]
  • Whither Radioimmunotherapy: to Be Or Not to Be? Damian J
    Published OnlineFirst April 20, 2017; DOI: 10.1158/0008-5472.CAN-16-2523 Cancer Perspective Research Whither Radioimmunotherapy: To Be or Not To Be? Damian J. Green1,2 and Oliver W. Press1,2,3 Abstract Therapy of cancer with radiolabeled monoclonal antibodies employing multistep "pretargeting" methods, particularly those has produced impressive results in preclinical experiments and in utilizing bispecific antibodies, have greatly enhanced the thera- clinical trials conducted in radiosensitive malignancies, particu- peutic efficacy of radioimmunotherapy and diminished its toxi- larly B-cell lymphomas. Two "first-generation," directly radiola- cities. The dramatically improved therapeutic index of bispecific beled anti-CD20 antibodies, 131iodine-tositumomab and 90yttri- antibody pretargeting appears to be sufficiently compelling to um-ibritumomab tiuxetan, were FDA-approved more than a justify human clinical trials and reinvigorate enthusiasm for decade ago but have been little utilized because of a variety of radioimmunotherapy in the treatment of malignancies, particu- medical, financial, and logistic obstacles. Newer technologies larly lymphomas. Cancer Res; 77(9); 1–6. Ó2017 AACR. "To be, or not to be, that is the question: Whether 'tis nobler in the pembrolizumab (anti-PD-1), which are not directly cytotoxic mind to suffer the slings and arrows of outrageous fortune, or to take for cancer cells but "release the brakes" on the immune system, arms against a sea of troubles, And by opposing end them." Hamlet. allowing cytotoxic T cells to be more effective at recognizing –William Shakespeare. and killing cancer cells. Outstanding results have already been demonstrated with checkpoint inhibiting antibodies even in far Introduction advanced refractory solid tumors including melanoma, lung cancer, Hodgkin lymphoma and are under study for a multi- Impact of monoclonal antibodies on the field of clinical tude of other malignancies (4–6).
    [Show full text]