151
|
Song Y, Ye Y, Su SH, Stephens A, Cai T, Chung MT, Han M, Newstead MW, Yessayan L, Frame D, Humes D, Singer BH, Kurabayashi K. A digital protein microarray for COVID-19 cytokine storm monitoring. LAB ON A CHIP 2021; 21:331-343. [PMID: 33211045 PMCID: PMC7855944 DOI: 10.1039/d0lc00678e] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Despite widespread concern regarding cytokine storms leading to severe morbidity in COVID-19, rapid cytokine assays are not routinely available for monitoring critically ill patients. We report the clinical application of a digital protein microarray platform for rapid multiplex quantification of cytokines from critically ill COVID-19 patients admitted to the intensive care unit (ICU) at the University of Michigan Hospital. The platform comprises two low-cost modules: (i) a semi-automated fluidic dispensing/mixing module that can be operated inside a biosafety cabinet to minimize the exposure of the technician to the virus infection and (ii) a 12-12-15 inch compact fluorescence optical scanner for the potential near-bedside readout. The platform enabled daily cytokine analysis in clinical practice with high sensitivity (<0.4 pg mL-1), inter-assay repeatability (∼10% CV), and rapid operation providing feedback on the progress of therapy within 4 hours. This test allowed us to perform serial monitoring of two critically ill patients with respiratory failure and to support immunomodulatory therapy using the selective cytopheretic device (SCD). We also observed clear interleukin-6 (IL-6) elevations after receiving tocilizumab (IL-6 inhibitor) while significant cytokine profile variability exists across all critically ill COVID-19 patients and to discover a weak correlation between IL-6 to clinical biomarkers, such as ferritin and C-reactive protein (CRP). Our data revealed large subject-to-subject variability in patients' response to COVID-19, reaffirming the need for a personalized strategy guided by rapid cytokine assays.
Collapse
Affiliation(s)
- Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yuxuan Ye
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Andrew Stephens
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Tao Cai
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Meng-Ting Chung
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Meilan Han
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Michael W. Newstead
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Lenar Yessayan
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - David Frame
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, United States
| | - David Humes
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Benjamin H. Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, United States
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, United States
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, United States
| |
Collapse
|
152
|
Pum A, Ennemoser M, Adage T, Kungl AJ. Cytokines and Chemokines in SARS-CoV-2 Infections-Therapeutic Strategies Targeting Cytokine Storm. Biomolecules 2021; 11:91. [PMID: 33445810 PMCID: PMC7828218 DOI: 10.3390/biom11010091] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 01/08/2023] Open
Abstract
The recently identified severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, the cause of coronavirus disease (COVID-19) and the associated ongoing pandemic, frequently leads to severe respiratory distress syndrome and pneumonia with fatal consequences. Although several factors of this infection and its consequences are not completely clear, the presence and involvement of specific chemokines is undoubtedly crucial for the development and progression of COVID-19. Cytokine storm and the often-resulting cytokine release syndrome (CRS) are pathophysiological hallmarks in COVID-19 infections related to its most severe and fatal cases. In this hyperinflammatory event, chemokines and other cytokines are highly upregulated and are therefore not fulfilling their beneficial function in the host response anymore but causing harmful effects. Here, we present the recent views on the involvement of chemokines and selected cytokines in COVID-19 and the therapeutics currently in clinical development targeting or interfering with them, discussing their potentials in the treatment of COVID-19 infections.
Collapse
Affiliation(s)
- Alexandra Pum
- Institute Of Pharmaceutical Sciences, Karl-Franzens-University Graz, Schubertstrasse 1, 8010 Graz, Austria; (A.P.); (M.E.)
| | - Maria Ennemoser
- Institute Of Pharmaceutical Sciences, Karl-Franzens-University Graz, Schubertstrasse 1, 8010 Graz, Austria; (A.P.); (M.E.)
| | - Tiziana Adage
- Brain Implant Therapeutics (BIT) Pharma, Leonhardstrasse 109, 8010 Graz, Austria;
| | - Andreas J. Kungl
- Institute Of Pharmaceutical Sciences, Karl-Franzens-University Graz, Schubertstrasse 1, 8010 Graz, Austria; (A.P.); (M.E.)
- Antagonis Biotherapeutics GmbH, Strasserhofweg 77, 8045 Graz, Austria
| |
Collapse
|
153
|
Castelnovo L, Tamburello A, Lurati A, Zaccara E, Marrazza MG, Olivetti M, Mumoli N, Mastroiacovo D, Colombo D, Ricchiuti E, Vigano’ P, Paola F, Mazzone A. Anti-IL6 treatment of serious COVID-19 disease: A monocentric retrospective experience. Medicine (Baltimore) 2021; 100:e23582. [PMID: 33429732 PMCID: PMC7793456 DOI: 10.1097/md.0000000000023582] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 01/05/2023] Open
Abstract
ABSTRACT COVID-19 is causing a high influx of patients suffering from serious respiratory complications leading the necessity to find effective therapies. These patients seem to present with cytokine perturbation and high levels of IL6. Tocilizumab and sarilumab could be effective in this condition.We retrospectively collected data about 112 consecutive hospitalized in a single center.Fifty (IL6 group) treated with tocilizumab (8 mg/kg intravenously [IV], 2 infusions 12 hours apart) or sarilumab 400 mg IV once and 62 treated with the standard of care but not anti-cytokine drugs (CONTROL group).To determine whether anti-IL6 drugs are effective in improving prognosis and reducing hospitalization times and mortality in COVID-19 pneumonia.To date 84% (42/50) of IL6 group patients have already been discharged and only 2/50 are still recovered and intubated in intensive care. Six/fifty patients (12%) died: 5/6 due to severe respiratory failure within a framework of severe acute respiratory distress syndrome (ARDS), 1 suffered an acute myocardial infarction, and 1 died of massive pulmonary thromboembolism. There were no adverse treatment events or infectious complications. Compared to the CONTROL group they showed a lower mortality rate (12% versus 43%), for the same number of complications and days of hospitalization.Anti-IL6 drugs seem to be effective in the treatment of medium to severe forms of COVID-19 pneumonia reducing the risk of mortality due to multi-organ failure, acting at the systemic level and reducing inflammation levels and therefore microvascular complications. However, it is essential to identify the best time for treatment, which, if delayed, is rendered useless as well as counterproductive. Further studies and ongoing clinical trials will help us to better define patients eligible as candidates for more aggressive intervention.
Collapse
Affiliation(s)
- Laura Castelnovo
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Legnano
| | - Antonio Tamburello
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Legnano
| | | | - Eleonora Zaccara
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Legnano
| | | | - Micol Olivetti
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Magenta
| | - Nicola Mumoli
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Magenta
| | | | - Daniele Colombo
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Legnano
| | | | - Paolo Vigano’
- Department of Infectious Diseases, ASST Ovest Milanese Ospedale di Legnano, Italy
| | - Faggioli Paola
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Legnano
| | - Antonino Mazzone
- Department of Internal Medicine, ASST Ovest Milanese Ospedale di Legnano
| |
Collapse
|
154
|
Tocilizumab for Fulminant Programmed Death 1 Inhibitor-Associated Myocarditis. J Thorac Oncol 2021; 15:e31-e32. [PMID: 32093854 DOI: 10.1016/j.jtho.2019.09.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 11/24/2022]
|
155
|
George JA, Mayne ES. The Novel Coronavirus and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1321:127-138. [PMID: 33656719 DOI: 10.1007/978-3-030-59261-5_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The SARS-CoV-2 virus which causes COVID-19 disease was initially described in the Hubei Province of China and has since spread to more than 200 countries and territories of the world. Severe cases of the disease are characterised by release of high levels of pro-inflammatory cytokines and other inflammatory mediators in a process characterised as a cytokine storm. These inflammatory mediators are associated with pathological leukocyte activation states with tissue damage. Here, we review these effects with a focus on their potential use in diagnosis, patient stratification and prognosis, as well as new drug targets.
Collapse
Affiliation(s)
- J A George
- Department of Chemical Pathology, National Health Laboratory Services and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - E S Mayne
- Department of Immunology, National Health Laboratory Services and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
156
|
Chen HJ, Meng T, Gao PJ, Ruan CC. The Role of Brown Adipose Tissue Dysfunction in the Development of Cardiovascular Disease. Front Endocrinol (Lausanne) 2021; 12:652246. [PMID: 34113316 PMCID: PMC8185286 DOI: 10.3389/fendo.2021.652246] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT), consisted of brown adipocytes and stromal vascular fraction, which includes endothelial cells, lymphocytes, fibroblasts and stem cells, plays a vital role in regulating cardiovascular health and diseases. As a thermogenic organ, BAT can influence body through strengthening energy expenditure by promoting glucose and lipid metabolism. In addition, BAT is also an endocrine organ which is able to secret adipokines in an autocrine and/or paracrine fashion. BAT plays a protective role in cardiovascular system through attenuating cardiac remodeling and suppressing inflammatory response. In this review, we summarize the advances from the discovery of BAT to the present and provide an overview on the role of BAT dysfunction in cardiovascular diseases.
Collapse
Affiliation(s)
- Hong-Jin Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Meng
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Chao Ruan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- *Correspondence: Cheng-Chao Ruan,
| |
Collapse
|
157
|
Abstract
PURPOSE OF REVIEW Immunotherapy with gene-engineered chimeric antigen receptor (CAR)-T cells has curative potential in advanced malignancies and undergoes a surging preclinical and clinical development. Here, we present a selection of new targets and technologies that illustrate the progress that is being made with the aspiration to make CAR-T cell therapy a universally applicable and effective treatment in cancer medicine. RECENT FINDINGS There is a rich pipeline of new target antigens for CAR-T cells in hematology and oncology that are rated based on uniformity but also stability of expression on tumor cells under therapeutic pressure. New technologies in CAR-T cell engineering are directed at neutralizing inhibitory ligands and factors in the tumor microenvironment, preventing CAR-T cell exhaustion and enhancing selectivity for tumor cells with 'smart' CAR designs. The manufacture of CAR-T cells using virus-free protocols is anticipated to reduce supply-chain complexity and to improve patient access. SUMMARY CD19 CAR-T cell therapy is an approved treatment for B-cell leukemia and -lymphoma and considering the current 'target and technology' pipeline, we anticipate that additional CAR-T cell products will accomplish their 'breakthrough' and clinical proof-of-concept in other indications in hematology and in oncology. Technologies to enhance therapeutic index and facilitate manufacturing will be key for assuring availability and accessibility of CAR-T cell products and their implementation into routine clinical practice.
Collapse
|
158
|
McBride DA, Kerr MD, Dorn NC, Ogbonna DA, Santos EC, Shah NJ. Triggers, Timescales, and Treatments for Cytokine-Mediated Tissue Damage. ACTA ACUST UNITED AC 2020; 5:52-62. [PMID: 34013158 DOI: 10.33590/emjinnov/20-00203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Inflammation is an essential cytokine-mediated process for generating a neutralizing immune response against pathogens and is generally protective. However, aberrant or excessive production of pro-inflammatory cytokines is associated with uncontrolled local and systemic inflammation, resulting in cell death and often irreversible tissue damage. Uncontrolled inflammation can manifest over timescales spanning hours to years and is primarily dependent on the triggering event. Rapid and potentially lethal increase in cytokine production, or a 'cytokine storm,' develops in hours to days and is associated with cancer cell-based immunotherapies, such as CAR-T cell therapy. On the other hand, some bacterial and viral infections with high microbial replication or highly potent antigens elicit immune responses that result in supraphysiological systemic cytokine concentrations which manifest over days to weeks. Immune dysregulation in autoimmune diseases can lead to chronic cytokine-mediated tissue damage spanning months to years, which often occurs episodically. While the initiating events and cellular participants may differ in these disease processes, many of the cytokines that drive disease progression are shared. For example, upregulation of IL-1, IL-6, IFN-γ, TNF, and GM-CSF frequently coincides with cytokine storm, sepsis, and autoimmune disease. Targeted inhibition of these pro-inflammatory molecules via antagonist monoclonal antibodies has improved clinical outcomes, but the complexity of the underlying immune dysregulation results in high variability. Rather than a "one size fits all" treatment approach, an identification of disease endotypes may permit the development of effective therapeutic strategies that address the contributors of disease progression. Here, we present a literature review of the cytokine-associated etiology of acute and chronic cytokine-mediated tissue damage, describe successes and challenges in developing clinical treatments, and highlight advancements in preclinical therapeutic strategies for mitigating pathological cytokine production.
Collapse
Affiliation(s)
- David A McBride
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA.,Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.,Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew D Kerr
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA.,Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.,Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicholas C Dorn
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA.,Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dora A Ogbonna
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA.,Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Evan C Santos
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA.,Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nisarg J Shah
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA.,Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.,Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA 92093, USA.,Program in Immunology, University of California, San Diego, La Jolla, CA 92093, USA.,San Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
159
|
Hamilton JA, Henry CJ. Aging and immunotherapies: New horizons for the golden ages. AGING AND CANCER 2020; 1:30-44. [PMID: 35874875 PMCID: PMC9307207 DOI: 10.1002/aac2.12014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The life expectancy of the world’s elderly population (65 and older) continues to reach new milestones with older individuals currently comprising greater than 8.5% (617 million) of the world’s population. This percentage is predicted to approach 20% of the world’s population by 2050 (representing 1.6 billion people). Despite this amazing feat, many healthcare systems are not equipped to handle the multitude of diseases that commonly manifest with age, including most types of cancers. As the world’s aging population grows, cancer treatments continue to evolve. Immunotherapies are a new drug class that has revolutionized our ability to treat previously intractable cancers; however, their efficacy in patients with compromised immune systems remains unclear. In this review, we will discuss how aging-associated losses in immune homeostasis impact the efficacy and safety of immunotherapy treatment in preclinical models of aging. We will also discuss how these findings translate to elderly patients receiving immunotherapy treatment for refractory and relapsed cancers, as well as, strategies that could be explored to improve the efficacy of immunotherapies in aged patients.
Collapse
Affiliation(s)
- Jamie A.G. Hamilton
- Department of Pediatrics Emory University School of Medicine Atlanta Georgia USA
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Atlanta Georgia USA
| | - Curtis J. Henry
- Department of Pediatrics Emory University School of Medicine Atlanta Georgia USA
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Atlanta Georgia USA
| |
Collapse
|
160
|
Salvati L, Occhipinti M, Gori L, Ciani L, Mazzoni A, Maggi L, Capone M, Parronchi P, Liotta F, Miele V, Annunziato F, Lavorini F, Cosmi L. Pulmonary vascular improvement in severe COVID-19 patients treated with tocilizumab. Immunol Lett 2020; 228:122-128. [PMID: 33161002 PMCID: PMC7644186 DOI: 10.1016/j.imlet.2020.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
As of October 2020 management of Coronavirus disease 2019 (COVID-19) is based on supportive care and off-label or compassionate-use therapies. On March 2020 tocilizumab - an anti-IL-6 receptor monoclonal antibody - was suggested as immunomodulatory treatment in severe COVID-19 because hyperinflammatory syndrome occurs in many patients similarly to the cytokine release syndrome that develops after CAR-T cell therapy. In our retrospective observational study, 20 severe COVID-19 patients requiring intensive care were treated with tocilizumab in addition to standard-of-care therapy (SOC) and compared with 13 COVID-19 patients receiving only SOC. Clinical respiratory status, inflammatory markers and vascular radiologic score improved after one week from tocilizumab administration. On the contrary, these parameters were stable or worsened in patients receiving only SOC. Despite major study limitations, improvement of alveolar-arterial oxygen gradient as well as vascular radiologic score after one week may account for improved pulmonary vascular perfusion and could explain the more rapid recovery of COVID-19 patients receiving tocilizumab compared to controls.
Collapse
Affiliation(s)
- Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mariaelena Occhipinti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Leonardo Gori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luca Ciani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paola Parronchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Vittorio Miele
- Department of Emergency Radiology, Careggi University Hospital, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Flow Cytometry Diagnostic Center and Immunotherapy (CDCI), Careggi University Hospital, Florence, Italy
| | - Federico Lavorini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Pneumology and Intensive Care Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
161
|
Gupta S, Madhyastha R, Hamed F, Balkis M, El Nekidy W, Attallah N. Tocilizumab Use in a Chronic Hemodialysis Patient for the Management of COVID-19-Associated Pneumonia and Acute Respiratory Distress Syndrome. Case Rep Nephrol 2020; 2020:8829309. [PMID: 33299621 PMCID: PMC7682475 DOI: 10.1155/2020/8829309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/09/2020] [Accepted: 10/24/2020] [Indexed: 12/26/2022] Open
Abstract
Novel coronavirus disease 2019 (COVID-19) is a highly infectious, rapidly spreading viral disease. As of writing this article, there are over 4.4 million people affected by COVID-19, and unfortunately, 300,000 have succumbed to the infection. In this article, we address a particularly more susceptible group of the population of end-stage renal disease (ESRD) patients on dialysis who may potentially benefit from being treated with tocilizumab. The use of tocilizumab has not been reported widely in ESRD patients on dialysis to treat COVID-19. In this case report, we describe a patient with ESRD on hemodialysis who was admitted to the intensive care unit, with severe pneumonia secondary to COVID-19 infection. This patient was treated with tocilizumab 400 mg intravenous and had a favorable outcome with no apparent adverse events.
Collapse
Affiliation(s)
- Sudeendra Gupta
- Medical Sub-specialities Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Rakesh Madhyastha
- Medical Sub-specialities Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Fadi Hamed
- Critical Care Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Maher Balkis
- Medical Sub-specialities Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Wasim El Nekidy
- Department of Pharmacy, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Nizar Attallah
- Medical Sub-specialities Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| |
Collapse
|
162
|
Reply to Wang et al.: Tocilizumab treatment should be used in a timely manner, at suitable dose, and in suitable patients. Proc Natl Acad Sci U S A 2020; 117:30898-30899. [PMID: 33203670 DOI: 10.1073/pnas.2017204117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
163
|
Rauf A, Abu-Izneid T, Olatunde A, Ahmed Khalil A, Alhumaydhi FA, Tufail T, Shariati MA, Rebezov M, Almarhoon ZM, Mabkhot YN, Alsayari A, Rengasamy KRR. COVID-19 Pandemic: Epidemiology, Etiology, Conventional and Non-Conventional Therapies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:8155. [PMID: 33158234 PMCID: PMC7662254 DOI: 10.3390/ijerph17218155] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/31/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), which reported in an outbreak in 2019 in Wuhan, Hubei province, China, is caused by the SARS-CoV-2 virus. The virus belongs to the beta-coronavirus class, along with the Middle East Respiratory Syndrome coronavirus and Severe Acute Respiratory Syndrome coronavirus. Interestingly, the virus binds with angiotensin-converting enzyme-2 found in host cells, through the spike (S) protein that exists on its surface. This binding causes the entry of the virus into cells of the host organism. The actual mechanism used by the COVID-19 virus to induce disease is still speculative. A total of 44,322,504 cases, a 1,173,189 death toll and 32,486,703 recovery cases have been reported in 217 countries globally as of 28 October 2020. Symptoms from the infection of the virus include chest pain, fever, fatigue, nausea, and others. Acute respiratory stress syndrome, arrhythmia, and shock are some of the chronic manifestations recorded in severe COVID-19. Transmission is majorly by individual-to-individual through coughing, sneezing, etc. The lack of knowledge regarding the mechanism of and immune response to the virus has posed a challenge in the development of a novel drug and vaccine. Currently, treatment of the disease involves the use of anti-viral medications such as lopinavir, remdesivir, and other drugs. These drugs show some efficacy in the management of COVID-19. Studies are still on-going for the development of an ideal and novel drug for treatment. In terms of natural product intervention, Traditional Chinese Medicines (TCM) have been employed to alleviate the clinical manifestation and severity of the disease and have shown some efficacy. This review presents an updated detailed overview of COVID-19 and the virus, concerning its structure, epidemiology, symptoms and transmission, immune responses, and current interventions, and highlights the potential of TCM. It is anticipated that this review will further add to the understanding of COVID-19 and the virus, hence opening new research perspectives.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar 23430, Khyber Pakhtunkhwa, Pakistan;
| | - Tareq Abu-Izneid
- Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain Campus 64141, UAE;
| | - Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauch 740272, Nigeria;
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore 54000, Pakistan; (A.A.K.); (T.T.)
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Tabussam Tufail
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore 54000, Pakistan; (A.A.K.); (T.T.)
| | - Mohammad Ali Shariati
- K.G. Razumovsky Moscow State University of Technologies and Management (the First Cossack University), 73 Zemlyanoy Val, 109004 Moscow, Russian;
| | - Maksim Rebezov
- V.M. Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, 109316 Moscow, Russian;
- Prokhorov General Physics Institute of the Russian Academy of Science, 119991 Moscow, Russian
| | - Zainab M. Almarhoon
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Yahia N. Mabkhot
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia;
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Kannan R. R. Rengasamy
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
- Faculty of Environment and Chemical Engineering, Duy Tan University, Da Nang 550000, Vietnam
- Indigenous Knowledge Systems Centre, Faculty of Natural and Agricultural Sciences, North-West University, Private Bag X2046, Mmabatho 2745, North West Province, South Africa
| |
Collapse
|
164
|
Lewis TC, Adhikari S, Tatapudi V, Holub M, Kunichoff D, Troxel AB, Montgomery RA, Sterman DH. A Propensity-Matched Cohort Study of Tocilizumab in Patients With Coronavirus Disease 2019. Crit Care Explor 2020; 2:e0283. [PMID: 33225307 PMCID: PMC7671881 DOI: 10.1097/cce.0000000000000283] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
To determine the impact of tocilizumab, a monoclonal antibody against the interleukin 6 receptor, on survival in patients with coronavirus disease 2019. DESIGN Observational cohort study of patients hospitalized with coronavirus disease 2019 between March 1, 2020, and April 24, 2020. A propensity-matched (1:1) analysis was used to compare patients who received tocilizumab to controls who did not. Competing risk survival analysis was used to determine the primary outcome of time to mortality, and adjusted log-linear and logistic regression for secondary outcomes. SETTING Three hospitals within the NYU Langone Health system in New York. PATIENTS Consecutive adult patients hospitalized with coronavirus disease 2019. INTERVENTION Tocilizumab 400-mg IV once in addition to standard of care or standard of care alone. MEASUREMENTS AND MAIN RESULTS Data from 3,580 severe acute respiratory syndrome coronavirus 2 positive qualifying hospitalized patients were included, of whom 497 (13.9%) were treated with tocilizumab. In the analysis of tocilizumab-treated patients and matched controls, fewer tocilizumab-treated patients died (145/497, 29.2%) than did controls (211/497, 42.4%). In the adjusted competing risk regression model, tocilizumab therapy was associated with improved survival relative to controls (hazard ratio = 0.24, 95% CI = 0.18-0.33, p < 0.001). Tocilizumab-treated patients and controls had similar adjusted time to discharge from hospital (hazard ratio = 0.96, 95% CI = 0.78-1.17, p = 0.67). However, they had longer adjusted ICU length of stay (rate ratio = 3.1, 95% CI = 2.5-3.7, p < 0.001) and a higher adjusted infection rate (odds ratio = 4.18, 95% CI = 2.72-6.52, p < 0.001) than controls. CONCLUSIONS Tocilizumab therapy was associated with significantly improved survival in coronavirus disease 2019 patients. This survival benefit was associated with increased ICU length of stay and increased infection rate, even as more patients in the tocilizumab group were rescued from rapid death. A prospective, randomized, placebo-controlled trial is needed to confirm these findings.
Collapse
Affiliation(s)
- Tyler C Lewis
- Department of Pharmacy, NYU Langone Health, New York, NY
- Transplant Institute, NYU Langone Health, New York, NY
| | | | | | - Meredith Holub
- Department of Pulmonary and Critical Care Medicine, NYU Langone Health, New York, NY
| | | | | | | | - Daniel H Sterman
- Department of Pulmonary and Critical Care Medicine, NYU Langone Health, New York, NY
| |
Collapse
|
165
|
Nasonov E, Samsonov M. The role of Interleukin 6 inhibitors in therapy of severe COVID-19. Biomed Pharmacother 2020; 131:110698. [PMID: 32920514 PMCID: PMC7455113 DOI: 10.1016/j.biopha.2020.110698] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokine storm syndrome (CSS) is a severe complication of inflammatory immune diseases or treatment of malignancies; it may also appear during the progression of COVID-19. CSS is caused by dysregulation of the synthesis of cytokines, including pro-inflammatory, regulatory, and anti-inflammatory cytokines and chemokines, leading to pathologic activation of innate and adaptive (Th1 and Th17 mediated) immunity. Interleukin-6 (IL-6) plays an important role in the pathogenesis of CSS. The significant role of IL-6 in pathogenesis of COVID-19 was confirmed in a range of studies, which showed that the plasma concentration of IL-6 was increased in patients with severe COVID-19. Currently, IL-6 inhibitor therapeutics are not yet approved for the treatment of COVID-19; however, these medicines, including tocilizumab (TCZ) are used off-label for the treatment of patients with severe COVID-19, including life-threatening conditions. The role of IL-6 in the pathogenesis of CSS during COVID-19 is important however, a number of related issues are not yet clear. These issues include the indications for treatment with IL-6 inhibitors, as well as the estimation of risk associated with the disease, outcomes, treatment options, and adverse drug reactions. The development of personalized immunomodulatory therapy, with respect to the role of cytokines in pathogenesis, requires the studies that aimed to find other relevant therapeutic targets for the treatment of CSS in patients with COVID-19. These therapeutic targets include inhibition of IL-1, IL-6, TNFα, GM-CSF, IFNγ, IL-17, IL-18, and also activation of the complement system. The challenge of CSS in patients with COVID-19 is identifying the correct scientific targets and developing clinical trials aimed to evaluate the pathogenesis and treat immune-mediated inflammatory diseases (IMIDs). Hopefully, the significant efforts of scientists and physicians across the globe will improve the prognosis in COVID-19 patients and provide useful information on IMIDs required to support the struggle for treating potential viral outbreaks, and treatment of well-known IMIDs.
Collapse
Affiliation(s)
- E Nasonov
- V.A. Nasonova Research Institute of Rheumatology, Moscow, Russia; I.M. Sechenov First Moscow State Medical University, MOH, Moscow, Russia; Kashirskoye roadway, 34А, 1115522, Moscow, Russia; Trubetskaya str, 8, bdg. 2, 2119991, Moscow, Russia.
| | - M Samsonov
- RPharm JSC, Leninsky prospect 111, 11942, Moscow, Russia.
| |
Collapse
|
166
|
Tocilizumab Treatment in COVID-19–induced Cytokine Release Syndrome. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2020. [DOI: 10.1097/ipc.0000000000000940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
167
|
Zhang J, Hao Y, Ou W, Ming F, Liang G, Qian Y, Cai Q, Dong S, Hu S, Wang W, Wei S. Serum interleukin-6 is an indicator for severity in 901 patients with SARS-CoV-2 infection: a cohort study. J Transl Med 2020; 18:406. [PMID: 33121497 PMCID: PMC7594951 DOI: 10.1186/s12967-020-02571-x] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Interleukin-6 (IL-6) was proposed to be associated with the severity of coronavirus disease 2019 (COVID-19). The present study aimed to explore the kinetics of IL-6 levels, validate this association in COVID-19 patients, and report preliminary data on the efficacy of IL-6 receptor blockade. METHODS We conducted a retrospective single-institutional study of 901 consecutive confirmed cases. Serum IL-6 concentrations were tested on admission and/or during hospital stay. Tocilizumab was given to 16 patients with elevated IL-6 concentration. RESULTS 366 patients were defined as common cases, 411 patients as severe, and 124 patients as critical according to the Chinese guideline on diagnosis and treatment of COVID-19. The median concentration of IL-6 was < 1.5 pg/ml (IQR < 1.50-2.15), 1.85 pg/ml (IQR < 1.50-5.21), and 21.55 pg/ml (IQR 6.47-94.66) for the common, severe, and critical groups respectively (P < 0.001). The follow-up kinetics revealed serum IL-6 remained high in critical patients even when cured. An IL-6 concentration higher than 37.65 pg/ml was predictive of in-hospital death (AUC 0.97 [95% CI 0.95-0.99], P < 0.001) with a sensitivity of 91.7% and a specificity of 95.7%. In the 16 patients who received tocilizumab, IL-6 concentrations were significantly increased after administration, and survival outcome was not significantly different from that of propensity-score matched counterparts (n = 53, P = 0.12). CONCLUSION Serum IL-6 should be included in diagnostic work-up to stratify disease severity, but the benefit of tocilizumab needs further confirmation. Trial registration retrospectively registered.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Yiqun Hao
- Department of Rheumatology and Immunology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wuling Ou
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Fei Ming
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Gai Liang
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Yu Qian
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Qian Cai
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Weida Wang
- Department of Hematological Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Yuexiu District, Guangzhou, 510060, Guangdong, China.
| | - Shaozhong Wei
- Department of Gastrointestinal Surgery & Colorectal Cancer Center of Hubei Province, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, Hubei, China.
| |
Collapse
|
168
|
Photoswitchable CAR-T Cell Function In Vitro and In Vivo via a Cleavable Mediator. Cell Chem Biol 2020; 28:60-69.e7. [PMID: 33113407 DOI: 10.1016/j.chembiol.2020.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 10/07/2020] [Indexed: 01/20/2023]
Abstract
Chimeric antigen receptor (CAR)-T-based therapeutics are a breakthrough in cancer treatment; however, they are hampered by constitutive activation, which leads to worrisome side effects. Engineering CAR-T cells to be as tightly controllable as possible remains a topic of ongoing investigation. Here, we report a photoswitchable approach that uses a mediator for the at-will regulation of CAR-T cells. This mediator carries dual folate and fluorescein isothiocyanate moieties tethered by an ortho-nitrobenzyl ester photocleavable linker. CAR-T cells were shown to be highly cytotoxic to targeted cells only in the presence of the mediator and acted in a dose-dependent manner. The toxicity of CAR-T cells can be rapidly terminated by cleavage of the mediator, and the effects of CAR-T cells can be activated again by resupplementation with the mediator without compromising tumor therapy. The approach described here provides a direction for enhancing the controllability of CAR-T cells and can likely be applied in other immunotherapies.
Collapse
|
169
|
Hu W, Wang G, Wang Y, Riese MJ, You M. Uncoupling Therapeutic Efficacy from Immune-Related Adverse Events in Immune Checkpoint Blockade. iScience 2020; 23:101580. [PMID: 33083746 PMCID: PMC7554032 DOI: 10.1016/j.isci.2020.101580] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy with monoclonal antibodies targeting immune checkpoint molecules, including programmed death-1 (PD-1), PD ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen (CTLA)-4, has become prominent in the treatment of many types of cancer. However, a significant number of patients treated with immune checkpoint inhibitors (ICIs) develop immune-related adverse events (irAEs). irAEs can affect any organ system, and although most are clinically manageable, irAEs can result in mortality or long-term morbidity. Factors that can predict irAEs remain elusive. Understanding the etiology of ICI-induced irAEs and ways to limit these adverse events are needed. In this review, we provide basic science and clinical insights on the mechanisms responsible for ICI efficacy and ICI-induced irAEs. We further provide insights into approaches that may uncouple irAEs from the ability of ICIs to kill tumor cells.
Collapse
Affiliation(s)
- Weilei Hu
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Guosheng Wang
- Department of Biomedical Engineering, Binghamton University—SUNY, 4400 Vestal Pkwy E, Binghamton, NY 13902, USA
| | - Yian Wang
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Blood Research Institute, Versiti Inc, Milwaukee, WI 53226, USA
| | - Ming You
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
170
|
Burke KE, Kochar B, Allegretti JR, Winter RW, Lochhead P, Khalili H, Colizzo FP, Hamilton MJ, Chan WW, Ananthakrishnan AN. Immunosuppressive Therapy and Risk of COVID-19 Infection in Patients With Inflammatory Bowel Diseases. Inflamm Bowel Dis 2020; 27:155-161. [PMID: 33089863 PMCID: PMC7665507 DOI: 10.1093/ibd/izaa278] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The effect of immunosuppressive treatment for immune-mediated diseases on risk of the novel coronavirus disease 2019 (COVID-19) has not been established. We aimed to define the effect of targeted biologic and immunomodulator therapy on risk of COVID-19 in a multi-institutional cohort of patients with inflammatory bowel disease (IBD). METHODS We identified patients 18 years and older who received care for IBD at Partners Healthcare between January 2019 and April 2020. The primary outcome was development of COVID-19 defined as a positive polymerase chain reaction test for severe acute respiratory syndrome coronavirus 2. Multivariable regression models were used to examine the effect of immunosuppression on risk of COVID-19 and its outcomes. RESULTS In a cohort of 5302 IBD patients, 39 (0.7%) developed COVID-19. There was no difference in age, sex, or race between IBD patients with and without COVID-19. The rate of COVID-19 was similar between patients treated with immunosuppression (0.8%) compared with those who were not (0.64%; P = 0.55). After adjusting for age, sex, race, and comorbidities, use of immunosuppressive therapy was not associated with an increased risk of COVID-19 (odds ratio, 1.73; 95% confidence interval, 0.82-3.63). The presence of obesity was associated with a higher risk of COVID-19 (odds ratio, 8.29; 95% confidence interval, 3.72-18.47). There were 7 hospitalizations, 3 intensive care unit stays, and 1 death. Older age and obesity but not immunosuppressive treatment were associated with severe COVID-19 infection. CONCLUSIONS The use of systemic immunosuppression was not associated with an increased risk of COVID-19 in a multi-institutional cohort of patients with IBD.
Collapse
Affiliation(s)
- Kristin E Burke
- Crohn’s and Colitis Center, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Bharati Kochar
- Crohn’s and Colitis Center, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Jessica R Allegretti
- Harvard Medical School, Boston, MA,Crohn’s and Colitis Center, Brigham and Women’s Hospital, Boston, MA
| | - Rachel W Winter
- Harvard Medical School, Boston, MA,Crohn’s and Colitis Center, Brigham and Women’s Hospital, Boston, MA
| | - Paul Lochhead
- Crohn’s and Colitis Center, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Hamed Khalili
- Crohn’s and Colitis Center, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Francis P Colizzo
- Crohn’s and Colitis Center, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Matthew J Hamilton
- Harvard Medical School, Boston, MA,Crohn’s and Colitis Center, Brigham and Women’s Hospital, Boston, MA
| | - Walter W Chan
- Harvard Medical School, Boston, MA,Crohn’s and Colitis Center, Brigham and Women’s Hospital, Boston, MA
| | - Ashwin N Ananthakrishnan
- Crohn’s and Colitis Center, Massachusetts General Hospital, Boston, MA,Harvard Medical School, Boston, MA,Address correspondence to: Ashwin N. Ananthakrishnan, MD, MPH, MGH, Crohn’s and Colitis Center, 165 Cambridge Street, 9th Floor, Boston, MA 02114, USA. E-mail:
| |
Collapse
|
171
|
How I treat relapsed acute lymphoblastic leukemia in the pediatric population. Blood 2020; 136:1803-1812. [DOI: 10.1182/blood.2019004043] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/11/2020] [Indexed: 01/04/2023] Open
Abstract
Abstract
Relapsed acute lymphoblastic leukemia (ALL) has remained challenging to treat in children, with survival rates lagging well behind those observed at initial diagnosis. Although there have been some improvements in outcomes over the past few decades, only ∼50% of children with first relapse of ALL survive long term, and outcomes are much worse with second or later relapses. Recurrences that occur within 3 years of diagnosis and any T-ALL relapses are particularly difficult to salvage. Until recently, treatment options were limited to intensive cytotoxic chemotherapy with or without site-directed radiotherapy and allogeneic hematopoietic stem cell transplantation (HSCT). In the past decade, several promising immunotherapeutics have been developed, changing the treatment landscape for children with relapsed ALL. Current research in this field is focusing on how to best incorporate immunotherapeutics into salvage regimens and investigate long-term survival and side effects, and when these might replace HSCT. As more knowledge is gained about the biology of relapse through comprehensive genomic profiling, incorporation of molecularly targeted therapies is another area of active investigation. These advances in treatment offer real promise for less toxic and more effective therapy for children with relapsed ALL, and we present several cases highlighting contemporary treatment decision-making.
Collapse
|
172
|
Hong M, Clubb JD, Chen YY. Engineering CAR-T Cells for Next-Generation Cancer Therapy. Cancer Cell 2020; 38:473-488. [PMID: 32735779 DOI: 10.1016/j.ccell.2020.07.005] [Citation(s) in RCA: 408] [Impact Index Per Article: 81.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
Abstract
T cells engineered to express chimeric antigen receptors (CARs) with tumor specificity have shown remarkable success in treating patients with hematologic malignancies and revitalized the field of adoptive cell therapy. However, realizing broader therapeutic applications of CAR-T cells necessitates engineering approaches on multiple levels to enhance efficacy and safety. Particularly, solid tumors present unique challenges due to the biological complexity of the solid-tumor microenvironment (TME). In this review, we highlight recent strategies to improve CAR-T cell therapy by engineering (1) the CAR protein, (2) T cells, and (3) the interaction between T cells and other components in the TME.
Collapse
Affiliation(s)
- Mihe Hong
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Justin D Clubb
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
173
|
Remy KE, Brakenridge SC, Francois B, Daix T, Deutschman CS, Monneret G, Jeannet R, Laterre PF, Hotchkiss RS, Moldawer LL. Immunotherapies for COVID-19: lessons learned from sepsis. THE LANCET. RESPIRATORY MEDICINE 2020; 8:946-949. [PMID: 32444269 PMCID: PMC7195015 DOI: 10.1016/s2213-2600(20)30217-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Kenneth E Remy
- Departments of Pediatrics and Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610-0108, USA
| | - Bruno Francois
- INSERM CIC-1435, Dupuytren University Hospital, Limoges, France; Intensive Care Unit, Dupuytren University Hospital, Limoges, France; INSERM UMR-1092, Université Limoges, Limoges, France
| | - Thomas Daix
- INSERM CIC-1435, Dupuytren University Hospital, Limoges, France; Intensive Care Unit, Dupuytren University Hospital, Limoges, France; INSERM UMR-1092, Université Limoges, Limoges, France
| | - Clifford S Deutschman
- Cohen Children's Medical Center, Northwell Health, Feinstein Institute for Medical Research, Zucker-Hofstra School of Medicine, Manhasset, NY, USA
| | - Guillaume Monneret
- Cellular Immunology Laboratory, Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Robin Jeannet
- INSERM CIC-1435, Dupuytren University Hospital, Limoges, France; UMR CNRS-7276 INSERM-1262, Limoges University, Limoges, France
| | - Pierre-Francois Laterre
- Department of Critical Care Medicine, St Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - Richard S Hotchkiss
- Departments of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, MO 63110-1093, USA.
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610-0108, USA.
| |
Collapse
|
174
|
Jasinski S, De Los Reyes FA, Yametti GC, Pierro J, Raetz E, Carroll WL. Immunotherapy in Pediatric B-Cell Acute Lymphoblastic Leukemia: Advances and Ongoing Challenges. Paediatr Drugs 2020; 22:485-499. [PMID: 32860590 PMCID: PMC7537790 DOI: 10.1007/s40272-020-00413-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leukemia, most commonly B-cell acute lymphoblastic leukemia (B-ALL), accounts for about 30% of childhood cancer diagnoses. While there have been dramatic improvements in childhood ALL outcomes, certain subgroups-particularly those who relapse-fare poorly. In addition, cure is associated with significant short- and long-term side effects. Given these challenges, there is great interest in novel, targeted approaches to therapy. A number of new immunotherapeutic agents have proven to be efficacious in relapsed or refractory disease and are now being investigated in frontline treatment regimens. Blinatumomab (a bispecific T-cell engager that targets cluster of differentiation [CD]-19) and inotuzumab ozogamicin (a humanized antibody-drug conjugate to CD22) have shown the most promise. Chimeric antigen receptor T (CAR-T) cells, a form of adoptive immunotherapy, rely on the transfer of genetically modified effector T cells that have the potential to persist in vivo for years, providing ongoing long-term disease control. In this article, we discuss the clinical biology and treatment of B-ALL with an emphasis on the role of immunotherapy in overcoming the challenges of conventional cytotoxic therapy. As immunotherapy continues to move into the frontline of pediatric B-ALL therapy, we also discuss strategies to address unique side effects associated with these agents and efforts to overcome mechanisms of resistance to immunotherapy.
Collapse
Affiliation(s)
- Sylwia Jasinski
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | | | - Gloria Contreras Yametti
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Joanna Pierro
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - Elizabeth Raetz
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA
| | - William L Carroll
- Perlmutter Cancer Center, Smilow 1211, Division of Pediatric Hematology/Oncology, Department of Pediatrics, NYU Langone Health, 560 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
175
|
Trujillo H, Caravaca-Fontán F, Sevillano Á, Gutiérrez E, Fernández-Ruiz M, López-Medrano F, Hernández A, Aguado JM, Praga M, Andrés A. Tocilizumab use in Kidney Transplant Patients with COVID-19. Clin Transplant 2020; 34:e14072. [PMID: 32862472 DOI: 10.1111/ctr.14072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023]
Abstract
A potential benefit of immunomodulatory agents such as tocilizumab (TCZ) has been reported in patients with coronavirus disease 2019 (COVID-19) and severe pulmonary involvement. However, this therapy has been scarcely studied in kidney transplant (KT) recipients. Herein, we describe the clinical course and outcome of 10 KT patients with severe COVID-19 that were treated with TCZ. Mean age of the study group was 54 ± 10 years (70% females), and 30% of the cases were within 6 months from transplant. Mycophenolate mofetil was discontinued in all cases upon admission, whereas baseline steroids were maintained and tacrolimus dose was reduced. Initial treatment included hydroxychloroquine, antibiotics, and prophylactic anticoagulation. Before treatment with TCZ, 3 patients were receiving high-flow oxygen, 4 patients low-flow oxygen and 1 case non-invasive ventilation. All patients received a single dose of intravenous TCZ within a mean time of 7 ± 4 days since admission. During a median follow-up of 16 days (IQR: 10-29), 7 patients (70%) gradually improved and were finally discharged while three cases (30%) did not exhibited clinical improvement and ultimately died. In conclusion, although treatment with TCZ could be associated with improved clinical outcomes in a subset of KT recipients with COVID-19, further studies are warranted before drawing firm conclusions.
Collapse
Affiliation(s)
| | - Fernando Caravaca-Fontán
- Department of Nephrology, Hospital Universitario, Madrid, Spain.,Research Institute Hospital, Madrid, Spain
| | - Ángel Sevillano
- Department of Nephrology, Hospital Universitario, Madrid, Spain
| | | | - Mario Fernández-Ruiz
- Research Institute Hospital, Madrid, Spain.,Unit of Infectious Diseases, Hospital Universitario, Madrid, Spain
| | - Francisco López-Medrano
- Research Institute Hospital, Madrid, Spain.,Unit of Infectious Diseases, Hospital Universitario, Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Hernández
- Department of Nephrology, Hospital Universitario, Madrid, Spain
| | - José María Aguado
- Research Institute Hospital, Madrid, Spain.,Unit of Infectious Diseases, Hospital Universitario, Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital Universitario, Madrid, Spain.,Research Institute Hospital, Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Amado Andrés
- Department of Nephrology, Hospital Universitario, Madrid, Spain.,Research Institute Hospital, Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
176
|
Favorable Outcome of COVID-19 Pneumonia in a Kidney Transplant Recipient Treated with Tocilizumab. Case Rep Infect Dis 2020; 2020:8830626. [PMID: 32963854 PMCID: PMC7495166 DOI: 10.1155/2020/8830626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
The presentation of COVID-19 pneumonia in kidney transplant recipients is similar to that of the general population. However, in the former, it may have a worse clinical course. We report a kidney transplant patient affected by COVID-19 pneumonia whose condition worsened 9 days after the initial presentation. As no therapeutic guidelines on the subject are currently available, here we share our approach in the management of the immunosuppressive medications and the antiviral therapy and compare them to the scarce available data. We also expose the use of tocilizumab in our patient with excellent results.
Collapse
|
177
|
Streicher C, Engalenc X, Gaudin M, Vignaud G, Daulange A, Abraham B. Could Tocilizumab be an Attractive Therapeutic Option for Elderly Patients with Severe COVID-19? A Case Report. Clin Drug Investig 2020; 40:1085-1088. [PMID: 32951190 PMCID: PMC7502217 DOI: 10.1007/s40261-020-00969-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Caroline Streicher
- Unité de préparation et de contrôle pharmaceutique (UPCP), Centre Hospitalier de Brive La Gaillarde, 1 boulevard du Dr Verlhac, 19312, Brive la Gaillarde Cedex, France.
| | - Xavier Engalenc
- Médecine interne et Maladies infectieuses, Centre Hospitalier de Brive La Gaillarde, 1 boulevard du Dr Verlhac, 19312, Brive la Gaillarde Cedex, France
| | - Marion Gaudin
- Médecine interne et Maladies infectieuses, Centre Hospitalier de Brive La Gaillarde, 1 boulevard du Dr Verlhac, 19312, Brive la Gaillarde Cedex, France
| | - Guillaume Vignaud
- Médecine interne et Maladies infectieuses, Centre Hospitalier de Brive La Gaillarde, 1 boulevard du Dr Verlhac, 19312, Brive la Gaillarde Cedex, France
| | - Annick Daulange
- Unité de préparation et de contrôle pharmaceutique (UPCP), Centre Hospitalier de Brive La Gaillarde, 1 boulevard du Dr Verlhac, 19312, Brive la Gaillarde Cedex, France
| | - Bruno Abraham
- Médecine interne et Maladies infectieuses, Centre Hospitalier de Brive La Gaillarde, 1 boulevard du Dr Verlhac, 19312, Brive la Gaillarde Cedex, France
| |
Collapse
|
178
|
Ataca Atilla P, McKenna MK, Tashiro H, Srinivasan M, Mo F, Watanabe N, Simons BW, McLean Stevens A, Redell MS, Heslop HE, Mamonkin M, Brenner MK, Atilla E. Modulating TNFα activity allows transgenic IL15-Expressing CLL-1 CAR T cells to safely eliminate acute myeloid leukemia. J Immunother Cancer 2020; 8:jitc-2020-001229. [PMID: 32938629 PMCID: PMC7497527 DOI: 10.1136/jitc-2020-001229] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Background C-type lectin-like molecule 1 (CLL-1) is highly expressed in acute myeloid leukemia (AML) but is absent in primitive hematopoietic progenitors, making it an attractive target for a chimeric antigen receptor (CAR) T-cell therapy. Here, we optimized our CLL-1 CAR for anti-leukemic activity in mouse xenograft models of aggressive AML. Methods First, we optimized the CLL-1 CAR using different spacer, transmembrane and costimulatory sequences. We used a second retroviral vector to coexpress transgenic IL15. We measured the effects of each construct on T cell phenotype and sequential (recursive) co culture assays with tumor cell targets to determine the durability of the anti tumor activity by flow cytometry. We administered CAR T cells to mice engrafted with patient derived xenografts (PDX) and AML cell line and determined anti tumor activity by bioluminescence imaging and weekly bleeding, measured serum cytokines by multiplex analysis. After euthanasia, we examined formalin-fixed/paraffin embedded sections. Unpaired two-tailed Student’s t-tests were used and values of p<0.05 were considered significant. Survival was calculated using Mantel-Cox log-rank test. Results In vitro, CLL-1 CAR T cells with interleukin-15 (IL15) were less terminally differentiated (p<0.0001) and had superior expansion compared with CD28z-CD8 CAR T cells without IL15 (p<0.001). In both AML PDX and AML cell line animal models, CLL-1 CAR T coexpressing transgenic IL15 initially expanded better than CD28z-CD8 CAR T without IL15 (p<0.0001), but produced severe acute toxicity associated with high level production of human tumor necrosis factor α (TNFα), IL15 and IL2. Histopathology showed marked inflammatory changes with tissue damage in lung and liver. This acute toxicity could be managed by two strategies, individually or in combination. The excessive TNF alpha secretion could be blocked with anti-TNF alpha antibody, while excessive T cell expansion could be arrested by activation of an inducible caspase nine safety switch by administration of dimerizing drug. Both strategies successfully prolonged tumor-free survival. Conclusion Combinatorial treatment with a TNFα blocking antibody and subsequent activation of the caspase-9 control switch increased the expansion, survival and antileukemic potency of CLL-1 CAR T-cells expressing transgenic IL15 while avoiding the toxicities associated with excessive cytokine production and long-term accumulation of activated T-cells.
Collapse
Affiliation(s)
- Pinar Ataca Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Haruko Tashiro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | | - Feiyan Mo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Brian Wesley Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Alexandra McLean Stevens
- Division of Pediatric Hematology/Oncology, Texas Children's Hospital, Houston, Texas, USA.,Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Michele S Redell
- Division of Pediatric Hematology/Oncology, Texas Children's Hospital, Houston, Texas, USA.,Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Erden Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
179
|
Abdin SM, Elgendy SM, Alyammahi SK, Alhamad DW, Omar HA. Tackling the cytokine storm in COVID-19, challenges and hopes. Life Sci 2020; 257:118054. [PMID: 32663575 PMCID: PMC7832727 DOI: 10.1016/j.lfs.2020.118054] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
The outbreak of Coronavirus disease 2019 (COVID-19) is the current world health concern, presenting a public health dilemma with ascending morbidity and mortality rates exceeding any previous viral spread, without a standard effective treatment yet. SARS-CoV-2 infection is distinguished with multiple epidemiological and pathological features, one of them being the elevated levels of cytokine release, which in turn trigger an aberrant uncontrolled response known as "cytokine storm". This phenomenon contributes to severe acute respiratory distress syndrome (ARDS), leading to pneumonia and respiratory failure, which is considered a major contributor to COVID-19-associated fatality rates. Taking into account that the vast majority of the COVID-19 cases are aggravated by the respiratory and multiorgan failure triggered by the sustained release of cytokines, implementing therapeutics that alleviate or diminish the upregulated inflammatory response would provide a therapeutic advantage to COVID-19 patients. Indeed, dexamethasone, a widely available and inexpensive corticosteroid with anti-inflammatory effects, has shown a great promise in reducing mortality rates in COVID-19 patients. In this review, we have critically compared the clinical impact of several potential therapeutic agents that could block or interfere with the cytokine storm, such as IL-1 inhibitors, IL-6 inhibitors, mast cell targeting agents, and corticosteroids. This work focused on highlighting and contrasting the current success and limitations towards the involvement of these agents in future treatment protocols.
Collapse
Affiliation(s)
- Shifaa M Abdin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sara M Elgendy
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Shatha K Alyammahi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Dima W Alhamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
180
|
Omolo CA, Soni N, Fasiku VO, Mackraj I, Govender T. Update on therapeutic approaches and emerging therapies for SARS-CoV-2 virus. Eur J Pharmacol 2020; 883:173348. [PMID: 32634438 PMCID: PMC7334944 DOI: 10.1016/j.ejphar.2020.173348] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 02/09/2023]
Abstract
The global pandemic of coronavirus disease 2019 (COVID-19), caused by novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in over 7,273,958 cases with almost over 413,372 deaths worldwide as per the WHO situational report 143 on COVID-19. There are no known treatment regimens with proven efficacy and vaccines thus far, posing an unprecedented challenge to identify effective drugs and vaccines for prevention and treatment. The urgency for its prevention and cure has resulted in an increased number of proposed treatment options. The high rate and volume of emerging clinical trials on therapies for COVID-19 need to be compared and evaluated to provide scientific evidence for effective medical options. Other emerging non-conventional drug discovery techniques such as bioinformatics and cheminformatics, structure-based drug design, network-based methods for prediction of drug-target interactions, artificial intelligence (AI) and machine learning (ML) and phage technique could provide alternative routes to discovering potent Anti-SARS-CoV2 drugs. While drugs are being repurposed and discovered for COVID-19, novel drug delivery systems will be paramount for efficient delivery and avoidance of possible drug resistance. This review describes the proposed drug targets for therapy, and outcomes of clinical trials that have been reported. It also identifies the adopted treatment modalities that are showing promise, and those that have failed as drug candidates. It further highlights various emerging therapies and future strategies for the treatment of COVID-19 and delivery of Anti-SARS-CoV2 drugs.
Collapse
Affiliation(s)
- Calvin A Omolo
- United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya; Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| | - Nikki Soni
- United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya
| | - Victoria Oluwaseun Fasiku
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Irene Mackraj
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Thirumala Govender
- United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya.
| |
Collapse
|
181
|
Atallah B, El Nekidy W, Mallah SI, Cherfan A, AbdelWareth L, Mallat J, Hamed F. Thrombotic events following tocilizumab therapy in critically ill COVID-19 patients: a Façade for prognostic markers. Thromb J 2020; 18:22. [PMID: 32922212 PMCID: PMC7479301 DOI: 10.1186/s12959-020-00236-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hospitals in the Middle East Gulf region have experienced an influx of COVID-19 patients to their medical wards and intensive care units. The hypercoagulability of these patients has been widely reported on a global scale. However, many of the experimental treatments used to manage the various complications of COVID-19 have not been widely studied in this context. The effect of the current treatment protocols on patients' diagnostic and prognostic biomarkers may thus impact the validity of the algorithms adopted. CASE PRESENTATION In this case series, we report four cases of venous thromboembolism and 1 case of arterial thrombotic event, in patients treated with standard or intensified prophylactic doses of unfractionated heparin or low molecular weight heparin at our institution. Tocilizumab has been utilized as an add-on therapy to the standard of care to treat patients with SARS-CoV-2 associated acute respiratory distress syndrome, in order to dampen the hyperinflammatory response. It is imperative to be aware that this drug may be masking the inflammatory markers (e.g. IL6, CRP, fibrinogen, and ferritin), without reducing the risk of thrombotic events in this population, creating instead a façade of an improved prognostic outcome. However, the D-dimer levels remained prognostically reliable in these cases, as they were not affected by the drug and continued to be at the highest level until event occurrence. CONCLUSIONS In the setting of tocilizumab therapy, traditional prognostic markers of worsening infection and inflammation, and thus potential risk of acute thrombosis, should be weighed carefully as they may not be reliable for prognosis and may create a façade of an improved prognostic outcome insteasd. Additionally, the fact that thrombotic events continued to be observed despite decrease in inflammatory markers and the proactive anticoagulative approach adopted, raises more questions about the coagulative mechanisms at play in COVID-19, and the appropriate management strategy.
Collapse
Affiliation(s)
- Bassam Atallah
- Department of Pharmacy Services, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi, United Arab Emirates
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH USA
| | - Wasim El Nekidy
- Department of Pharmacy Services, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi, United Arab Emirates
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH USA
| | - Saad I. Mallah
- School of Medicine, Royal College of Surgeons in Ireland–Bahrain, Al Sayh, Bahrain
| | - Antoine Cherfan
- Department of Pharmacy Services, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi, United Arab Emirates
| | - Laila AbdelWareth
- Department of Clinical Pathology, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi, United Arab Emirates
| | - Jihad Mallat
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH USA
- Critical Care Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi, United Arab Emirates
- Normandy University, UNICAEN ED 497, Caen, France
| | - Fadi Hamed
- Critical Care Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi, United Arab Emirates
| |
Collapse
|
182
|
Guadarrama-Ortiz P, Choreño-Parra JA, Sánchez-Martínez CM, Pacheco-Sánchez FJ, Rodríguez-Nava AI, García-Quintero G. Neurological Aspects of SARS-CoV-2 Infection: Mechanisms and Manifestations. Front Neurol 2020; 11:1039. [PMID: 33013675 PMCID: PMC7499054 DOI: 10.3389/fneur.2020.01039] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/10/2020] [Indexed: 01/08/2023] Open
Abstract
The human infection of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a public health emergency of international concern that has caused more than 16.8 million new cases and 662,000 deaths as of July 30, 2020. Although coronavirus disease 2019 (COVID-19), which is associated with this virus, mainly affects the lungs, recent evidence from clinical and pathological studies indicates that this pathogen has a broad infective ability to spread to extrapulmonary tissues, causing multiorgan failure in severely ill patients. In this regard, there is increasing preoccupation with the neuroinvasive potential of SARS-CoV-2 due to the observation of neurological manifestations in COVID-19 patients. This concern is also supported by the neurotropism previously documented in other human coronaviruses, including the 2002-2003 SARS-CoV-1 outbreak. Hence, in the current review article, we aimed to summarize the spectrum of neurological findings associated with COVID-19, which include signs of peripheral neuropathy, myopathy, olfactory dysfunction, meningoencephalitis, Guillain-Barré syndrome, and neuropsychiatric disorders. Furthermore, we analyze the mechanisms underlying such neurological sequela and discuss possible therapeutics for patients with neurological findings associated with COVID-19. Finally, we describe the host- and pathogen-specific factors that determine the tissue tropism of SARS-CoV-2 and possible routes employed by the virus to invade the nervous system from a pathophysiological and molecular perspective. In this manner, the current manuscript contributes to increasing the current understanding of the neurological aspects of COVID-19 and the impact of the current pandemic on the neurology field.
Collapse
Affiliation(s)
- Parménides Guadarrama-Ortiz
- Departament of Neurosurgery, Centro Especializado en Neurocirugía y Neurociencias México (CENNM), Mexico City, Mexico
| | - José Alberto Choreño-Parra
- Departament of Neurosurgery, Centro Especializado en Neurocirugía y Neurociencias México (CENNM), Mexico City, Mexico
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Francisco Javier Pacheco-Sánchez
- Internado Medico de Pregrado, Centro Especializado en Neurocirugía y Neurociencias México (CENNM), Mexico City, Mexico
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Alberto Iván Rodríguez-Nava
- Internado Medico de Pregrado, Centro Especializado en Neurocirugía y Neurociencias México (CENNM), Mexico City, Mexico
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Gabriela García-Quintero
- Internado Medico de Pregrado, Centro Especializado en Neurocirugía y Neurociencias México (CENNM), Mexico City, Mexico
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
183
|
Khiali S, Khani E, Entezari‐Maleki T. A Comprehensive Review of Tocilizumab in COVID-19 Acute Respiratory Distress Syndrome. J Clin Pharmacol 2020; 60:1131-1146. [PMID: 32557541 PMCID: PMC7323169 DOI: 10.1002/jcph.1693] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023]
Abstract
Currently, the world is facing the pandemic of a novel strain of beta-coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Acute respiratory distress syndrome (ARDS) is the most devastating complication of SARS-CoV-2. It was indicated that cytokine-release syndrome and dominantly interleukin (IL)-6 play a central role in the pathophysiology of ARDS related to the novel 2019 coronavirus disease (COVID-19). Despite the global emergency of the disease, at this time, there are no proven therapies for the management of the disease. Tocilizumab is a potential recombinant monoclonal antibody against IL-6 and currently is under investigation for the management of ARDS in patients with COVID-19. Given these points, we reviewed the current evidence regarding the potential therapeutic role of tocilizumab and its important clinical issues in the treatment of ARDS related to COVID-19.
Collapse
Affiliation(s)
- Sajad Khiali
- Department of Clinical PharmacyFaculty of PharmacyTabriz University of Medical SciencesTabrizIran
| | - Elnaz Khani
- Department of Clinical PharmacyFaculty of PharmacyTabriz University of Medical SciencesTabrizIran
| | - Taher Entezari‐Maleki
- Department of Clinical PharmacyFaculty of PharmacyTabriz University of Medical SciencesTabrizIran
- Cardiovascular Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
184
|
Shamim S, Khan M, Kharaba ZJ, Ijaz M, Murtaza G. Potential strategies for combating COVID-19. Arch Virol 2020; 165:2419-2438. [PMID: 32778950 PMCID: PMC7416802 DOI: 10.1007/s00705-020-04768-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022]
Abstract
Coronavirus disease 2019, also known as COVID-19, is caused by a novel coronavirus named severe acute respiratory syndrome coronavirus 2, or SARS-CoV-2. The infection has now catapulted into a full-blown pandemic across the world, which has affected more than 2 million people and has led to approximately 150,000 fatalities all over the world (WHO). In this review, we elaborate all currently available data that shed light on possible methods for treatment of COVID-19, such as antiviral drugs, corticosteroids, convalescent plasma, and potentially effective vaccines. Additionally, ongoing and discontinued clinical trials that have been carried out for validating probable treatments for COVID-19 are discussed. The review also elaborates the prospective approach and the possible advantages of using convalescent plasma and stem cells for the improvement of clinical symptoms and meeting the demand for an instantaneous cure.
Collapse
Affiliation(s)
- Saba Shamim
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Maryam Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Islamabad, 54000, Pakistan.
| |
Collapse
|
185
|
Shchelkanov MY, Kolobukhina LV, Burgasova OA, Kruzhkova IS, Maleev VV. COVID-19: etiology, clinical picture, treatment. ACTA ACUST UNITED AC 2020. [DOI: 10.15789/2220-7619-cec-1473] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Whereas the XX century marked the history of acute respiratory disease investigation as a period for generating in-depth system of combating influenza viruses (Articulavirales: Orthomyxoviridae, Alpha-/Betainfluenzavirus) (based on environmental and virological monitoring of influenza A virus in its natural reservoir — aquatic and semi-aquatic birds — to supervising epidemic influenza), a similar system is necessary to build up in the XXI century with regard to especially dangerous betacoronaviruses (Nidovirales: Coronaviridae, Betacoronavirus): Severe acute respiratory syndrome-related coronavirus (SARS-CoV) (subgenus Sarbecovirus), Severe acute respiratory syndrome-related coronavirus 2 (SARSCoV-2) (Sarbecovirus), Middle East respiratory syndrome-related coronavirus (MERS-CoV) (Merbecovirus). This became particularly evident after pandemic potential has been revealed in 2020 by the SARS-CoV-2. This review provides an insight into the historic timeline of discovering this virus, its current taxonomy, ecology, virion morphology, life cycle, molecular biology, pathogenesis and clinical picture of the etiologically related COVID-19 (Coronavirus disease 2019) as well as data available in the scientific literature on the anti-SARS-CoV-2-effectiveness of passive immunotherapy and most debated drugs used to treat COVID-19: Chloroquine, Hydroxychloroquine, Nitazoxanide, Ivermectin, Lopinavir and Ritonavir, Camostat mesilate, Remdesivir, Ribavirin, Tocilizumab, Anakinra, corticosteroids, and type I interferons. The pathogenesis of SARS-CoV-2 infection implicates decreased efficacy of artificial respiration, which, in this case might be replaced by more efficient extracorporeal membrane blood oxygenation supplemented with nitrogen oxide and/or Heliox inhalations.
Collapse
Affiliation(s)
- M. Yu. Shchelkanov
- International Scientific and Educational Center for Biological Security of Rospotrebnadzor;
Federal Scientific Center of East Asia Terrestrial Biodiversity, Far Eastern Branch of RAS;
Center of Hygiene and Epidemiology in the Primorsky Territory
| | - L. V. Kolobukhina
- D.I. Ivanovsky Institute of Virology of the N.F. Gamaleya National Scientific Center of Epidemiology and Microbiology, Russian Ministry of Public Health
| | | | - I. S. Kruzhkova
- D.I. Ivanovsky Institute of Virology of the N.F. Gamaleya National Scientific Center of Epidemiology and Microbiology, Russian Ministry of Public Health
| | - V. V. Maleev
- Central Research Institute of Epidemiology and Microbiology of Rospotrebnadzor
| |
Collapse
|
186
|
Guo C, Li B, Ma H, Wang X, Cai P, Yu Q, Zhu L, Jin L, Jiang C, Fang J, Liu Q, Zong D, Zhang W, Lu Y, Li K, Gao X, Fu B, Liu L, Ma X, Weng J, Wei H, Jin T, Lin J, Qu K. Single-cell analysis of two severe COVID-19 patients reveals a monocyte-associated and tocilizumab-responding cytokine storm. Nat Commun 2020; 11:3924. [PMID: 32764665 PMCID: PMC7413381 DOI: 10.1038/s41467-020-17834-w] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
Abstract
Several studies show that the immunosuppressive drugs targeting the interleukin-6 (IL-6) receptor, including tocilizumab, ameliorate lethal inflammatory responses in COVID-19 patients infected with SARS-CoV-2. Here, by employing single-cell analysis of the immune cell composition of two severe-stage COVID-19 patients prior to and following tocilizumab-induced remission, we identify a monocyte subpopulation that contributes to the inflammatory cytokine storms. Furthermore, although tocilizumab treatment attenuates the inflammation, immune cells, including plasma B cells and CD8+ T cells, still exhibit robust humoral and cellular antiviral immune responses. Thus, in addition to providing a high-dimensional dataset on the immune cell distribution at multiple stages of the COVID-19, our work also provides insights into the therapeutic effects of tocilizumab, and identifies potential target cell populations for treating COVID-19-related cytokine storms.
Collapse
Affiliation(s)
- Chuang Guo
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Bin Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Huan Ma
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Xiaofang Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Pengfei Cai
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Qiaoni Yu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Lin Zhu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Liying Jin
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Chen Jiang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Jingwen Fang
- HanGene Biotech, Xiaoshan Innovation Polis, 31200, Hangzhou, Zhejiang, China
| | - Qian Liu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Dandan Zong
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Wen Zhang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Yichen Lu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Kun Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Xuyuan Gao
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Binqing Fu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
- CAS Center for Excellence in Molecular Cell Sciences, the CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
| | - Xiaoling Ma
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences of Medicine, University of Science and Technology of China, 230026, Hefei, China
| | - Haiming Wei
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China
- CAS Center for Excellence in Molecular Cell Sciences, the CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Tengchuan Jin
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China.
- CAS Center for Excellence in Molecular Cell Sciences, the CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| | - Jun Lin
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China.
- CAS Center for Excellence in Molecular Cell Sciences, the CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| | - Kun Qu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230021, Hefei, Anhui, China.
- CAS Center for Excellence in Molecular Cell Sciences, the CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, 230027, Hefei, Anhui, China.
- School of Data Science, University of Science and Technology of China, 230026, Hefei, China.
| |
Collapse
|
187
|
Montesarchio V, Parrela R, Iommelli C, Bianco A, Manzillo E, Fraganza F, Palumbo C, Rea G, Murino P, De Rosa R, Atripaldi L, D'Abbraccio M, Curvietto M, Mallardo D, Celentano E, Grimaldi AM, Palla M, Trojaniello C, Vitale MG, Million-Weaver SL, Ascierto PA. Outcomes and biomarker analyses among patients with COVID-19 treated with interleukin 6 (IL-6) receptor antagonist sarilumab at a single institution in Italy. J Immunother Cancer 2020; 8:e001089. [PMID: 32784217 PMCID: PMC7418768 DOI: 10.1136/jitc-2020-001089] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The inflammatory pathology observed in severe COVID-19 disease caused by the 2019 novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is characterized by elevated serum levels of C reactive protein (CRP) and cytokines, including interferon gamma, interleukin 8 (IL-8), and interleukin 6 (IL-6). Initial reports from the outbreak in Italy, China and the USA have provided anecdotal evidence of improved outcomes with the administration of anti-IL-6 agents, and large-scale trials evaluating these therapies are ongoing. STUDY DESCRIPTION In this retrospective case series, clinical outcomes and correlates of response to treatment with the IL-6 receptor antagonist sarilumab are described for 15 patients with COVID-19 from a single institution in Southern Italy. Among 10 patients whose symptoms improved after sarilumab treatment, rapid decreases in CRP levels corresponded with clinical improvement. Lower levels of IL-6 at baseline as well as lower neutrophil to lymphocyte ratio as compared with patients whose COVID-19 did not improve with treatment were associated with sarilumab-responsive disease. CONCLUSIONS This observation may reflect a possible clinical benefit regarding early intervention with IL-6-modulatory therapies for COVID-19 and that CRP could be a potential biomarker of response to treatment.
Collapse
Affiliation(s)
- Vincenzo Montesarchio
- Department of Medical Oncology, Azienda Ospedaliera dei Colli-Monaldi Hospital, Napoli, Italy
| | - Roberto Parrela
- Department of Infectious Diseases and Infectious Diseases Emergencies, Azienda Ospedaliera dei Colli-Cotugno Hospital, Napoli, Italy
| | - Chiara Iommelli
- Department of Medical Oncology, Azienda Ospedaliera dei Colli-Monaldi Hospital, Napoli, Italy
| | - Antonella Bianco
- Department of Medical Oncology, Azienda Ospedaliera dei Colli-Monaldi Hospital, Napoli, Italy
| | - Elio Manzillo
- Department of Infectious Diseases and Infectious Diseases Emergencies, Azienda Ospedaliera dei Colli-Cotugno Hospital, Napoli, Italy
| | - Fiorentino Fraganza
- Critical Area Department, Azienda Ospedaliera dei Colli-Cotugno and Monaldi Hospital, Napoli, Italy
| | - Cristiana Palumbo
- Department of Health Services, Azienda Ospedaliera dei Colli-Cotugno and Monaldi Hospital, Napoli, Italy
| | - Gaetano Rea
- Department of Health Services, Azienda Ospedaliera dei Colli-Cotugno and Monaldi Hospital, Napoli, Italy
| | - Patrizia Murino
- Critical Area Department, Azienda Ospedaliera dei Colli-Cotugno and Monaldi Hospital, Napoli, Italy
| | - Rosanna De Rosa
- Critical Area Department, Azienda Ospedaliera dei Colli-Cotugno and Monaldi Hospital, Napoli, Italy
| | - Luigi Atripaldi
- Department of Health Services, Azienda Ospedaliera dei Colli-Cotugno and Monaldi Hospital, Napoli, Italy
| | - Maurizio D'Abbraccio
- Department of Infectious Diseases and Infectious Diseases Emergencies, Azienda Ospedaliera dei Colli-Cotugno Hospital, Napoli, Italy
| | - Marcello Curvietto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Domenico Mallardo
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Egidio Celentano
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Antonio Maria Grimaldi
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Marco Palla
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Claudia Trojaniello
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Maria Grazia Vitale
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | | |
Collapse
|
188
|
Slimano F, Baudouin A, Zerbit J, Toulemonde-Deldicque A, Thomas-Schoemann A, Chevrier R, Daouphars M, Madelaine I, Pourroy B, Tournamille JF, Astier A, Ranchon F, Cazin JL, Bardin C, Rioufol C. Cancer, immune suppression and Coronavirus Disease-19 (COVID-19): Need to manage drug safety (French Society for Oncology Pharmacy [SFPO] guidelines). Cancer Treat Rev 2020; 88:102063. [PMID: 32623296 PMCID: PMC7308737 DOI: 10.1016/j.ctrv.2020.102063] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
The Coronavirus disease (COVID-19) pandemic is disrupting our health environment. As expected, studies highlighted the great susceptibility of cancer patients to COVID-19 and more severe complications, leading oncologists to deeply rethink patient cancer care. This review is dedicated to the optimization of care pathways and therapeutics in cancer patients during the pandemic and aims to discuss successive issues. First we focused on the international guidelines proposing adjustments and alternative options to cancer care in order to limit hospital admission and cytopenic treatment in cancer patients, most of whom are immunocompromised. In addition cancer patients are prone to polypharmacy, enhancing the risk of drug-related problems as adverse events and drug-drug interactions. Due to increased risk in case of COVID-19, we reported a comprehensive review of all the drug-related problems between COVID-19 and antineoplastics. Moreover, in the absence of approved drug against COVID-19, infected patients may be included in clinical trials evaluating new drugs with a lack of knowledge, particularly in cancer patients. Focusing on the several experimental drugs currently being evaluated, we set up an original data board helping oncologists and pharmacists to identify promptly drug-related problems between antineoplastics and experimental drugs. Finally additional and concrete recommendations are provided, supporting oncologists and pharmacists in their efforts to manage cancer patients and to optimize their treatments in this new era related to COVID-19.
Collapse
Affiliation(s)
- Florian Slimano
- Department of Pharmacy, CHU Reims, France; Faculty of Pharmacy, Université de Reims Champagne-Ardenne, 51100 Reims, France.
| | - Amandine Baudouin
- Department of Pharmacy, Groupement Hospitalier Sud - Hospices Civils de Lyon, Lyon, France.
| | - Jérémie Zerbit
- Department of Clinical Pharmacy, CHU Paris Centre Cochin, AP-HP, 75 014 Paris, France.
| | | | - Audrey Thomas-Schoemann
- Department of Pharmacy, Groupement Hospitalier Sud - Hospices Civils de Lyon, Lyon, France; UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, Paris Descartes University, PRES Sorbonne Paris Cité, 75006 Paris, France.
| | - Régine Chevrier
- Department of Pharmacy, Jean Perrin Cancer Center, 63011 Clermont Ferrand, France.
| | - Mikaël Daouphars
- Department of Pharmacy, Henri Becquerel Cancer Center, 76038 Rouen, France.
| | - Isabelle Madelaine
- Department of Pharmacy, Saint Louis University Teaching Hospital, Assistance Publique - Hôpitaux de Paris, 75010 Paris, France.
| | - Bertrand Pourroy
- Oncopharma Unit, La Timone University Teaching Hospital, Assistance Publique - Hôpitaux de Marseille, 13005 Marseille, France.
| | | | - Alain Astier
- Department of Pharmacy, Henri Mondor University Hospitals, 94010 Créteil, France.
| | - Florence Ranchon
- Department of Pharmacy, Groupement Hospitalier Sud - Hospices Civils de Lyon, Lyon, France; EA 3738 CICLY, UCBL1 Université de Lyon, Lyon, France.
| | - Jean-Louis Cazin
- Center of Pharmacology and Clinical Pharmacy in Oncology, Centre Oscar Lambret, 59020 Lille, France; Pharmacology and Clinical Pharmacy, Faculté de Pharmacie, Université de Lille, 59000 Lille, France.
| | - Christophe Bardin
- Department of Clinical Pharmacy, CHU Paris Centre Cochin, AP-HP, 75 014 Paris, France.
| | - Catherine Rioufol
- Department of Pharmacy, Groupement Hospitalier Sud - Hospices Civils de Lyon, Lyon, France; EA 3738 CICLY, UCBL1 Université de Lyon, Lyon, France.
| |
Collapse
|
189
|
Kastritis E, Wechalekar A, Schönland S, Sanchorawala V, Merlini G, Palladini G, Minnema M, Roussel M, Jaccard A, Hegenbart U, Kumar S, Cibeira MT, Blade J, Dimopoulos MA. Challenges in the management of patients with systemic light chain (AL) amyloidosis during the COVID-19 pandemic. Br J Haematol 2020; 190:346-357. [PMID: 32480420 PMCID: PMC7300844 DOI: 10.1111/bjh.16898] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/29/2020] [Indexed: 01/08/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-associated coronavirus disease 2019 (COVID-19) is primarily manifested as a respiratory tract infection, but may affect and cause complications in multiple organ systems (cardiovascular, gastrointestinal, kidneys, haematopoietic and immune systems), while no proven specific therapy exists. The challenges associated with COVID-19 are even greater for patients with light chain (AL) amyloidosis, a rare multisystemic disease affecting the heart, kidneys, liver, gastrointestinal and nervous system. Patients with AL amyloidosis may need to receive chemotherapy, which probably increases infection risk. Management of COVID-19 may be particularly challenging in patients with AL amyloidosis, who often present with cardiac dysfunction, nephrotic syndrome, neuropathy, low blood pressure and gastrointestinal symptoms. In addition, patients with AL amyloidosis may be more susceptible to toxicities of drugs used to manage COVID-19. Access to health care may be difficult or limited, diagnosis of AL amyloidosis may be delayed with detrimental consequences and treatment administration may need modification. Both patients and treating physicians need to adapt in a new reality.
Collapse
Affiliation(s)
- Efstathios Kastritis
- Plasma Cell Dyscrasia UnitDepartment of Clinical TherapeuticsNational and Kapodistrian University of AthensAthensGreece
| | | | - Stefan Schönland
- Medical Department V, Amyloidosis CentreUniversity Hospital HeidelbergHeidelbergGermany
| | - Vaishali Sanchorawala
- Amyloidosis CenterSchool of Medicine and Boston Medical CenterBoston UniversityBostonMAUSA
| | - Giampaolo Merlini
- Department of Molecular MedicineAmyloidosis Research and Treatment CenterFoundation “Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo”University of PaviaPaviaItaly
| | - Giovanni Palladini
- Department of Molecular MedicineAmyloidosis Research and Treatment CenterFoundation “Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo”University of PaviaPaviaItaly
| | - Monique Minnema
- Department of HematologyUMC Utrecht Cancer CenterUtrechtThe Netherlands
| | | | | | - Ute Hegenbart
- Medical Department V, Amyloidosis CentreUniversity Hospital HeidelbergHeidelbergGermany
| | - Shaji Kumar
- Division of HematologyDepartment of Internal MedicineMayo ClinicRochesterMNUSA
| | - Maria T. Cibeira
- Hematology DepartmentAmyloidosis and Myeloma UnitHospital Clínic of BarcelonaUniversity of BarcelonaIDIBAPSBarcelonaSpain
| | - Joan Blade
- Hematology DepartmentAmyloidosis and Myeloma UnitHospital Clínic of BarcelonaUniversity of BarcelonaIDIBAPSBarcelonaSpain
| | - Meletios A. Dimopoulos
- Plasma Cell Dyscrasia UnitDepartment of Clinical TherapeuticsNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
190
|
Fernández-Ruiz M, López-Medrano F, Pérez-Jacoiste Asín MA, Maestro de la Calle G, Bueno H, Caro-Teller JM, Catalán M, de la Calle C, García-García R, Gómez C, Laguna-Goya R, Lizasoáin M, Martínez-López J, Origüen J, Pablos JL, Ripoll M, San Juan R, Trujillo H, Lumbreras C, Aguado JM. Tocilizumab for the treatment of adult patients with severe COVID-19 pneumonia: A single-center cohort study. J Med Virol 2020; 93:831-842. [PMID: 32672860 PMCID: PMC7404673 DOI: 10.1002/jmv.26308] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/13/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID‐19) can lead to a massive cytokine release. The use of the anti‐interleukin‐6 receptor monoclonal antibody tocilizumab (TCZ) has been proposed in this hyperinflammatory phase, although supporting evidence is limited. We retrospectively analyzed 88 consecutive patients with COVID‐19 pneumonia that received at least one dose of intravenous TCZ in our institution between 16 and 27 March 2020. Clinical status from day 0 (first TCZ dose) through day 14 was assessed by a 6‐point ordinal scale. The primary outcome was clinical improvement (hospital discharge and/or a decrease of ≥2 points on the 6‐point scale) by day 7. Secondary outcomes included clinical improvement by day 14 and dynamics of vital signs and laboratory values. Rates of clinical improvement by days 7 and 14 were 44.3% (39/88) and 73.9% (65/88). Previous or concomitant receipt of subcutaneous interferon‐β (adjusted odds ratio [aOR]: 0.23; 95% confidence interval [CI]: 0.06‐0.94; P = .041) and serum lactate dehydrogenase more than 450 U/L at day 0 (aOR: 0.25; 95% CI: 0.06‐0.99; P = .048) were negatively associated with clinical improvement by day 7. All‐cause mortality was 6.8% (6/88). Body temperature and respiratory and cardiac rates significantly decreased by day 1 compared to day 0. Lymphocyte count and pulse oximetry oxygen saturation/FiO2 ratio increased by days 3 and 5, whereas C‐reactive protein levels dropped by day 2. There were no TCZ‐attributable adverse events. In this observational single‐center study, TCZ appeared to be useful and safe as immunomodulatory therapy for severe COVID‐19 pneumonia. COVID‐19 can lead to a hyperinflammatory state that mirrors the cytokine release syndrome. The off‐labeluse of the anti‐interleukin‐6 receptor monoclonal antibody tocilizumab has been proposed to abrogate this deleterious inflammatory response, although the supporting evidence is scarce. In the present single‐centre study comprising 88 consecutive patients with COVID‐19 pneumonia that received at least one dose of intravenous tocilizumab between March 16 and 27, 2020, the rates of clinical improvement (defined by discharge to home and/or a decrease of = 2 points on a six‐point ordinal scale) were 44.3% (39/88) and 73.9% (65/88) by days 7 and 14, respectively. The previous or concomitant use of interferon‐β and baseline serum lactate dehydrogenase levels >450 U/L were negatively associated with clinical improvement by day 7. All‐cause mortality was 6.8%, with no tocilizumab‐attributable adverse events.
Collapse
Affiliation(s)
- Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - María Asunción Pérez-Jacoiste Asín
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Guillermo Maestro de la Calle
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Héctor Bueno
- Department of Cardiology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José Manuel Caro-Teller
- Department of Pharmacy, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Mercedes Catalán
- Department of Intensive Care Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Cristina de la Calle
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Rocío García-García
- Department of Pneumology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Carlos Gómez
- Department of Medical Oncology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Rocío Laguna-Goya
- Department of Immunology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Manuel Lizasoáin
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Joaquín Martínez-López
- Department of Hematology, Centro Nacional de Investigaciones Oncológicas (CNIO), Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Universidad Complutense, Madrid, Spain
| | - Julia Origüen
- Department of Emergency Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José Luis Pablos
- Department of Rheumatology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Mar Ripoll
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Hernando Trujillo
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Carlos Lumbreras
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| |
Collapse
|
191
|
Altay O, Mohammadi E, Lam S, Turkez H, Boren J, Nielsen J, Uhlen M, Mardinoglu A. Current Status of COVID-19 Therapies and Drug Repositioning Applications. iScience 2020; 23:101303. [PMID: 32622261 PMCID: PMC7305759 DOI: 10.1016/j.isci.2020.101303] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/09/2023] Open
Abstract
The rapid and global spread of a new human coronavirus (SARS-CoV-2) has produced an immediate urgency to discover promising targets for the treatment of COVID-19. Drug repositioning is an attractive approach that can facilitate the drug discovery process by repurposing existing pharmaceuticals to treat illnesses other than their primary indications. Here, we review current information concerning the global health issue of COVID-19 including promising approved drugs and ongoing clinical trials for prospective treatment options. In addition, we describe computational approaches to be used in drug repurposing and highlight examples of in silico studies of drug development efforts against SARS-CoV-2.
Collapse
Affiliation(s)
- Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm 17121, Sweden
| | - Elyas Mohammadi
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm 17121, Sweden; Department of Animal Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Simon Lam
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg, The Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg 41345, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-Gothenburg, 41296, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm 17121, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm 17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK.
| |
Collapse
|
192
|
Knight TE. Severe Acute Respiratory Syndrome Coronavirus 2 and Coronavirus Disease 2019: A Clinical Overview and Primer. Biopreserv Biobank 2020; 18:492-502. [PMID: 32726140 DOI: 10.1089/bio.2020.0066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Following its emergence in December 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused what rapidly became a global pandemic. The precise origin and subsequent path of transmission have not yet been established-but like the other novel coronaviruses that it closely resembles, it appears to have evolved naturally in a bat host. The disease caused by SARS-CoV-2 infection, designated as coronavirus disease 2019 (COVID-19), ranges from asymptomatic, to mild self-limited illness, to progressive pneumonia, respiratory compromise, multiorgan failure, and death. In addition, a hyperinflammatory disease state occurs in a subset of patients, and may be seen either during acute infection or following recovery. The search for effective pharmacological management of COVID-19 continues, but several promising candidates have been identified, including the viral nucleoside analog remdesivir. However, despite the existence of literally thousands of clinical trials, the management of COVID-19 remains challenging, and the development of an optimal, evidence-based therapeutic approach is ongoing. The impact of SARS-CoV-2 and COVID-19 on the biobanking world is evolving and profound-in particular, it is likely that many of mysteries surrounding COVID-19 will be solved via the availability of high-quality, large-scale collection, storage, and analysis of patient specimens. The purpose of this review article is therefore to provide a rapid, comprehensive, and relevant overview and primer on SARS-CoV-2 and COVID-19, with attention to the epidemiology, virology, transmission, clinical features, and major therapeutic options currently existent.
Collapse
Affiliation(s)
- Tristan E Knight
- Division of Haematology and Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| |
Collapse
|
193
|
Fakhouri EW, Peterson SJ, Kothari J, Alex R, Shapiro JI, Abraham NG. Genetic Polymorphisms Complicate COVID-19 Therapy: Pivotal Role of HO-1 in Cytokine Storm. Antioxidants (Basel) 2020; 9:E636. [PMID: 32708430 PMCID: PMC7402116 DOI: 10.3390/antiox9070636] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/15/2022] Open
Abstract
Coronaviruses are very large RNA viruses that originate in animal reservoirs and include severe acute respiratory distress syndrome (SARS) and Middle East respiratory syndrome (MERS) and other inconsequential coronaviruses from human reservoirs like the common cold. SARS-CoV-2, the virus that causes COVID-19 and is believed to originate from bat, quickly spread into a global pandemic. This RNA virus has a special affinity for porphyrins. It invades the cell at the angiotensin converting enzyme-2 (ACE-2) receptor and binds to hemoproteins, resulting in a severe systemic inflammatory response, particularly in high ACE-2 organs like the lungs, heart, and kidney, resulting in systemic disease. The inflammatory response manifested by increased cytokine levels and reactive oxygen species results in inhibition of heme oxygenase (HO-1), with a subsequent loss of cytoprotection. This has been seen in other viral illness like human immunodeficiency virus (HIV), Ebola, and SARS/MERS. There are a number of medications that have been tried with some showing early clinical promise. This illness disproportionately affects patients with obesity, a chronic inflammatory disease with a baseline excess of cytokines. The majority of the medications used in the treatment of COVID-19 are metabolized by cytochrome P450 (CYP) enzymes, primarily CYP2D6. This is further complicated by genetic polymorphisms of CYP2D6, HO-1, ACE, and ACE-2. There is a potential role for HO-1 upregulation to treat/prevent cytokine storm. Current therapy must focus on antivirals and heme oxygenase upregulation. Vaccine development will be the only magic bullet.
Collapse
Affiliation(s)
- Eddie W. Fakhouri
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (E.W.F.); (J.K.)
| | - Stephen J. Peterson
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (E.W.F.); (J.K.)
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Janish Kothari
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (E.W.F.); (J.K.)
| | - Ragin Alex
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA;
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA;
| | - Nader G. Abraham
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA;
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA;
- Department of Medicine, New York Medical College, Valhalla, New York, NY 10595, USA
| |
Collapse
|
194
|
Baker EH, Patel K, Ball J, Edwards S, Harrison TS, Kaul A, Koh M, Krishna S, Leaver S, Kumar V, Forton DM. Insights from compassionate use of tocilizumab for COVID-19 to inform appropriate design of randomised controlled trials. Br J Clin Pharmacol 2020; 87:1584-1586. [PMID: 32656822 PMCID: PMC7405226 DOI: 10.1111/bcp.14466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 11/10/2022] Open
Affiliation(s)
- Emma H Baker
- Institute of Infection and Immunity, St George's, University of London, London, UK.,Pharmacy and Medicines Directorate, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Kamal Patel
- Department of Gastroenterology and Hepatology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Jonathan Ball
- Adult Critical Care Directorate, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Sarah Edwards
- Department of Science and Technology Studies, University College London, London, UK
| | - Thomas S Harrison
- Institute of Infection and Immunity, St George's, University of London, London, UK.,Clinical Infection Unit, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Arvind Kaul
- Department of Rheumatology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Mickey Koh
- Department of Haematology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Sanjeev Krishna
- Institute of Infection and Immunity, St George's, University of London, London, UK.,Clinical Infection Unit, St George's University Hospitals NHS Foundation Trust, London, UK.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Susannah Leaver
- Adult Critical Care Directorate, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Vinodh Kumar
- Pharmacy and Medicines Directorate, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Daniel M Forton
- Institute of Infection and Immunity, St George's, University of London, London, UK.,Department of Gastroenterology and Hepatology, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
195
|
Farooqi F, Dhawan N, Morgan R, Dinh J, Nedd K, Yatzkan G. Treatment of Severe COVID-19 with Tocilizumab Mitigates Cytokine Storm and Averts Mechanical Ventilation During Acute Respiratory Distress: A Case Report and Literature Review. Trop Med Infect Dis 2020; 5:E112. [PMID: 32635353 PMCID: PMC7559384 DOI: 10.3390/tropicalmed5030112] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19, caused by the novel severe acute respiratory coronavirus 2 (SARS-CoV-2), emerged in Wuhan, China, in 2019 and has resulted in the current pandemic. The disease continues to pose a major therapeutic challenge. Patient mortality is ultimately caused by acute respiratory distress syndrome (ARDS). Cytokine release syndrome (or "cytokine storm") is likely to be a contributing factor to ARDS in many patients. Because interleukin 6 (IL-6) is known to play a key role in inflammation, IL-6 receptor inhibitors such as tocilizumab may potentially treat COVID-19 by attenuating cytokine release. We present the case of a 48-year-old male with severe COVID-19, on the verge of meeting intubation requirements, who needed progressive oxygen support for respiratory distress. The patient was treated with a non-weight-based dosage of tocilizumab to prevent the onset of a cytokine storm. We chose to administer an IL-6 inhibitor because of the gradually increasing levels of acute phase reactants identified on serial blood draws, as well as his declining respiratory status. The treatment was well-tolerated in conjunction with standard drug therapies for COVID-19 (hydroxychloroquine, azithromycin, and zinc). The patient subsequently experienced marked improvements in his respiratory symptoms and overall clinical status over the following days. We believe that tocilizumab played a substantial role in his ability to avert clinical decline, particularly the need for mechanical ventilation. Ultimately, the patient was downgraded from the ICU and discharged within days. We highlight the potential of IL-6 inhibitors to prevent the progression of respiratory disease to a point requiring ventilator support. This case underscores the potential importance of early serial measurements of IL-6 and cytokine storm-associated acute phase reactants, such as ferritin, D-dimer, and C-reactive protein, in guiding clinical decision-making in the management of patients with suspected COVID-19. Conclusion: The early, proactive identification of serum acute phase reactants should be implemented in the treatment of COVID-19 in order to screen for a primary contributor to mortality-the cytokine storm. This screening, when followed by aggressive early treatment for cytokine storm, may have optimal therapeutic benefits and obviate the need for mechanical ventilation, thereby decreasing mortality. Additionally, we review current evidence regarding cytokine release syndrome in COVID-19 and the use of IL-6 receptor inhibition as a therapeutic strategy, and examine other reported cases in the literature describing IL-6 antagonist treatment for patients with COVID-19.
Collapse
Affiliation(s)
- Faryal Farooqi
- Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA; (N.D.); (R.M.); (J.D.); (K.N.)
| | - Naveen Dhawan
- Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA; (N.D.); (R.M.); (J.D.); (K.N.)
| | - Richard Morgan
- Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA; (N.D.); (R.M.); (J.D.); (K.N.)
| | - John Dinh
- Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA; (N.D.); (R.M.); (J.D.); (K.N.)
| | - Kester Nedd
- Department of Internal Medicine, Larkin Community Hospital, South Miami, FL 33143, USA; (N.D.); (R.M.); (J.D.); (K.N.)
| | - George Yatzkan
- Department of Pulmonary Medicine, Larkin Community Hospital, South Miami, FL 33143, USA;
| |
Collapse
|
196
|
Cao Y, Wei J, Zou L, Jiang T, Wang G, Chen L, Huang L, Meng F, Huang L, Wang N, Zhou X, Luo H, Mao Z, Chen X, Xie J, Liu J, Cheng H, Zhao J, Huang G, Wang W, Zhou J. Ruxolitinib in treatment of severe coronavirus disease 2019 (COVID-19): A multicenter, single-blind, randomized controlled trial. J Allergy Clin Immunol 2020; 146:137-146.e3. [PMID: 32470486 PMCID: PMC7250105 DOI: 10.1016/j.jaci.2020.05.019] [Citation(s) in RCA: 329] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Accumulating evidence proposed Janus-associated kinase (JAK) inhibitors as therapeutic targets warranting rapid investigation. OBJECTIVE This study evaluated the efficacy and safety of ruxolitinib, a JAK1/2 inhibitor, for coronavirus disease 2019. METHODS We conducted a prospective, multicenter, single-blind, randomized controlled phase II trial involving patients with severe coronavirus disease 2019. RESULTS Forty-three patients were randomly assigned (1:1) to receive ruxolitinib plus standard-of-care treatment (22 patients) or placebo based on standard-of-care treatment (21 patients). After exclusion of 2 patients (1 ineligible, 1 consent withdrawn) from the ruxolitinib group, 20 patients in the intervention group and 21 patients in the control group were included in the study. Treatment with ruxolitinib plus standard-of-care was not associated with significantly accelerated clinical improvement in severe patients with coronavirus disease 2019, although ruxolitinib recipients had a numerically faster clinical improvement. Eighteen (90%) patients from the ruxolitinib group showed computed tomography improvement at day 14 compared with 13 (61.9%) patients from the control group (P = .0495). Three patients in the control group died of respiratory failure, with 14.3% overall mortality at day 28; no patients died in the ruxolitinib group. Ruxolitinib was well tolerated with low toxicities and no new safety signals. Levels of 7 cytokines were significantly decreased in the ruxolitinib group in comparison to the control group. CONCLUSIONS Although no statistical difference was observed, ruxolitinib recipients had a numerically faster clinical improvement. Significant chest computed tomography improvement, a faster recovery from lymphopenia, and favorable side-effect profile in the ruxolitinib group were encouraging and informative to future trials to test efficacy of ruxolitinib in a larger population.
Collapse
Affiliation(s)
- Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Zou
- Department of Hematology, Wuhan No. 1 Hospital, Wuhan, China
| | - Tiebin Jiang
- Hematology Department of The Third Xiangya Hospital Central South University, Changsha, China
| | - Gaoxiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fankai Meng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lifang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Luo
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zekai Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Jing Liu
- Hematology Department of The Third Xiangya Hospital Central South University, Changsha, China
| | - Hui Cheng
- Department of Hematology, Wuhan No. 1 Hospital, Wuhan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clincal Trial and Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
197
|
Lubetzky M, Aull MJ, Craig-Schapiro R, Lee JR, Marku-Podvorica J, Salinas T, Gingras L, Lee JB, Sultan S, Kodiyanplakkal RP, Hartono C, Saal S, Muthukumar T, Kapur S, Suthanthiran M, Dadhania DM. Kidney allograft recipients, immunosuppression, and coronavirus disease-2019: a report of consecutive cases from a New York City transplant center. Nephrol Dial Transplant 2020; 35:1250-1261. [PMID: 32678882 PMCID: PMC7454827 DOI: 10.1093/ndt/gfaa154] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Kidney graft recipients receiving immunosuppressive therapy may be at heightened risk for coronavirus disease 2019 (Covid-19) and adverse outcomes. It is therefore important to characterize the clinical course and outcome of Covid-19 in this population and identify safe therapeutic strategies. METHODS We performed a retrospective chart review of 73 adult kidney graft recipients evaluated for Covid-19 from 13 March to 20 April 2020. Primary outcomes included recovery from symptoms, acute kidney injury, graft failure and case fatality rate. RESULTS Of the 73 patients screened, 54 tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-39 with moderate to severe symptoms requiring hospital admission and 15 with mild symptoms managed in the ambulatory setting. Hospitalized patients were more likely to be male, of Hispanic ethnicity and to have cardiovascular disease. In the hospitalized group, tacrolimus dosage was reduced in 46% of patients and mycophenolate mofetil (MMF) therapy was stopped in 61% of patients. None of the ambulatory patients had tacrolimus reduction or discontinuation of MMF. Azithromycin or doxycycline was prescribed at a similar rate among hospitalized and ambulatory patients (38% versus 40%). Hydroxychloroquine was prescribed in 79% of hospitalized patients. Graft failure requiring hemodialysis occurred in 3 of 39 hospitalized patients (8%) and 7 patients died, resulting in a case fatality rate of 13% among Covid-19-positive patients and 18% among hospitalized Covid-19-positive patients. CONCLUSIONS Data from our study suggest that a strategy of systematic triage to outpatient or inpatient care, early management of concurrent bacterial infections and judicious adjustment of immunosuppressive drugs rather than cessation is feasible in kidney transplant recipients with Covid-19.
Collapse
Affiliation(s)
- Michelle Lubetzky
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | - Meredith J Aull
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
- Division of Transplant Surgery, Weill Cornell Medicine, New York, NY, USA
| | | | - John R Lee
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | | | - Thalia Salinas
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
| | - Laura Gingras
- Department of Internal Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jun B Lee
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | - Samuel Sultan
- Division of Transplant Surgery, Weill Cornell Medicine, New York, NY, USA
| | | | - Choli Hartono
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | - Stuart Saal
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | - Sandip Kapur
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
- Division of Transplant Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Manikkam Suthanthiran
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| | - Darshana M Dadhania
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, USA
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
198
|
Jamilloux Y, Henry T, Belot A, Viel S, Fauter M, El Jammal T, Walzer T, François B, Sève P. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti-cytokine interventions. Autoimmun Rev 2020; 19:102567. [PMID: 32376392 PMCID: PMC7196557 DOI: 10.1016/j.autrev.2020.102567] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 01/08/2023]
Abstract
The coronavirus disease-19 pandemic (COVID-19), which appeared in China in December 2019 and rapidly spread throughout the world, has forced clinicians and scientists to take up extraordinary challenges. This unprecedented situation led to the inception of numerous fundamental research protocols and many clinical trials. It quickly became apparent that although COVID-19, in the vast majority of cases, was a benign disease, it could also develop a severe form with sometimes fatal outcomes. Cytokines are central to the pathophysiology of COVID-19; while some of them are beneficial (type-I interferon, interleukin-7), others appear detrimental (interleukin-1β, -6, and TNF-α) particularly in the context of the so-called cytokine storm. Yet another characteristic of the disease has emerged: concomitant immunodeficiency, notably involving impaired type-I interferon response, and lymphopenia. This review provides an overview of current knowledge on COVID-19 immunopathology. We discuss the defective type-I IFN response, the theoretical role of IL-7 to restore lymphocyte repertoire, as well as we mention the two patterns observed in severe COVID-19 (i.e. interleukin-1β-driven macrophage activation syndrome vs. interleukin-6-driven immune dysregulation). Next, reviewing current evidence drawn from clinical trials, we examine a number of cytokine and anti-cytokine therapies, including interleukin-1, -6, and TNF inhibitors, as well as less targeted therapies, such as corticosteroids, chloroquine, or JAK inhibitors.
Collapse
Affiliation(s)
- Yvan Jamilloux
- Department of Internal Medicine, Lyon University Hospital, Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France.
| | - Thomas Henry
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France; Department of Pediatric rheumatology, nephrology, and dermatology, Lyon University Hospital, Lyon, France; National Referee Centre for Rheumatic and AutoImmune and Systemic diseases in childrEn (RAISE), Lyon, France
| | - Sébastien Viel
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France; Service d'Immunologie Biologique, Centre, Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France; National Referee Centre for Rheumatic and AutoImmune and Systemic diseases in childrEn (RAISE), Lyon, France
| | - Maxime Fauter
- Department of Internal Medicine, Lyon University Hospital, Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France
| | - Thomas El Jammal
- Department of Internal Medicine, Lyon University Hospital, Lyon, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France
| | - Bruno François
- Intensive care unit & Inserm CIC 1435 & Inserm UMR 1092, Dupuytren University Hospital, Limoges, France
| | - Pascal Sève
- Department of Internal Medicine, Lyon University Hospital, Lyon, France
| |
Collapse
|
199
|
Perricone C, Triggianese P, Bartoloni E, Cafaro G, Bonifacio AF, Bursi R, Perricone R, Gerli R. The anti-viral facet of anti-rheumatic drugs: Lessons from COVID-19. J Autoimmun 2020; 111:102468. [PMID: 32317220 PMCID: PMC7164894 DOI: 10.1016/j.jaut.2020.102468] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
The outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has posed the world at a pandemic risk. Coronavirus-19 disease (COVID-19) is an infectious disease caused by SARS-CoV-2, which causes pneumonia, requires intensive care unit hospitalization in about 10% of cases and can lead to a fatal outcome. Several efforts are currently made to find a treatment for COVID-19 patients. So far, several anti-viral and immunosuppressive or immunomodulating drugs have demonstrated some efficacy on COVID-19 both in vitro and in animal models as well as in cases series. In COVID-19 patients a pro-inflammatory status with high levels of interleukin (IL)-1B, IL-1 receptor (R)A and tumor necrosis factor (TNF)-α has been demonstrated. Moreover, high levels of IL-6 and TNF-α have been observed in patients requiring intensive-care-unit hospitalization. This provided rationale for the use of anti-rheumatic drugs as potential treatments for this severe viral infection. Other agents, such as hydroxychloroquine and chloroquine might have a direct anti-viral effect. The anti-viral aspect of immunosuppressants towards a variety of viruses has been known since long time and it is herein discussed in the view of searching for a potential treatment for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Elena Bartoloni
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Angelo F Bonifacio
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Roberto Bursi
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Roberto Gerli
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy.
| |
Collapse
|
200
|
Geng YJ, Wei ZY, Qian HY, Huang J, Lodato R, Castriotta RJ. Pathophysiological characteristics and therapeutic approaches for pulmonary injury and cardiovascular complications of coronavirus disease 2019. Cardiovasc Pathol 2020; 47:107228. [PMID: 32375085 PMCID: PMC7162778 DOI: 10.1016/j.carpath.2020.107228] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) has emerged as a major health crisis, with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) having infected over a million people around the world within a few months of its identification as a human pathogen. Initially, SARS-CoV-2 infects cells in the respiratory system and causes inflammation and cell death. Subsequently, the virus spreads out and damages other vital organs and tissues, triggering a complicated spectrum of pathophysiological changes and symptoms, including cardiovascular complications. Acting as the receptor for SARS-CoV entering mammalian cells, angiotensin converting enzyme-2 (ACE2) plays a pivotal role in the regulation of cardiovascular cell function. Diverse clinical manifestations and laboratory abnormalities occur in patients with cardiovascular injury in COVID-19, characterizing the development of this complication, as well as providing clues to diagnosis and treatment. This review provides a summary of the rapidly appearing laboratory and clinical evidence for the pathophysiology and therapeutic approaches to COVID-19 pulmonary and cardiovascular complications.
Collapse
Affiliation(s)
- Yong-Jian Geng
- Department of Internal Medicine, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Zhi-Yao Wei
- Department of Cardiology, Center for Coronary Heart Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Yan Qian
- Department of Cardiology, Center for Coronary Heart Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Robert Lodato
- Department of Internal Medicine, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Richard J Castriotta
- Department of Internal Medicine, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA; Division of Pulmonary, Critical and Sleep Medicine, University of South California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|