1
|
Westover AJ, Humes HD, Pino CJ. Immunomodulatory effects of a cell processing device to ameliorate dysregulated hyperinflammatory disease states. Sci Rep 2024; 14:12747. [PMID: 38830924 PMCID: PMC11148190 DOI: 10.1038/s41598-024-63121-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 05/24/2024] [Indexed: 06/05/2024] Open
Abstract
Cell directed therapy is an evolving therapeutic approach to treat organ dysfunction arising from hyperinflammation and cytokine storm by processing immune cells in an extracorporeal circuit. To investigate the mechanism of action of the Selective Cytopheretic Device (SCD), in vitro blood circuits were utilized to interrogate several aspects of the immunomodulatory therapy. SCD immunomodulatory activity is due to its effects on circulating neutrophils and monocytes in a low ionized calcium (iCa, Ca2+) blood circuit. Activated neutrophils adhere to the SCD fibers and degranulate with release of the constituents of their exocytotic vesicles. Adhered neutrophils in the low iCa environment display characteristics of apoptotic senescence. These neutrophils are subsequently released and returned back to circulation, demonstrating a clear potential for in vivo feedback. For monocytes, SCD treatment results in the selective adhesion of more pro-inflammatory subsets of the circulating monocyte pool, as demonstrated by both cell surface markers and cytokine secretory rates. Once bound, over time a subset of monocytes are released from the membrane with a less inflammatory functional phenotype. Similar methods to interrogate mechanism in vitro have been used to preliminarily confirm comparable findings in vivo. Therefore, the progressive amelioration of circulating leukocyte activation and immunomodulation of excessive inflammation observed in SCD clinical trials to date is likely due to this continuous autologous leukocyte processing.
Collapse
Affiliation(s)
- Angela J Westover
- Nephrology/Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Innovative BioTherapies, Ann Arbor, MI, 48108, USA
| | - H David Humes
- Nephrology/Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Innovative BioTherapies, Ann Arbor, MI, 48108, USA.
| | - Christopher J Pino
- Nephrology/Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Innovative BioTherapies, Ann Arbor, MI, 48108, USA
| |
Collapse
|
2
|
Iyer SPN, Pino CJ, Yessayan LT, Goldstein SL, Weir MR, Westover AJ, Catanzaro DA, Chung KK, Humes HD. Increasing Eligibility to Transplant Through the Selective Cytopheretic Device: A Review of Case Reports Across Multiple Clinical Conditions. Transplant Direct 2024; 10:e1627. [PMID: 38769980 PMCID: PMC11104718 DOI: 10.1097/txd.0000000000001627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 05/22/2024] Open
Abstract
A stable, minimum physiological health status is required for patients to qualify for transplant or artificial organ support eligibility to ensure the recipient has enough reserve to survive the perioperative transplant period. Herein, we present a novel strategy to stabilize and improve patient clinical status through extracorporeal immunomodulation of systemic hyperinflammation with impact on multiple organ systems to increase eligibility and feasibility for transplant/device implantation. This involves treatment with the selective cytopheretic device (SCD), a cell-directed extracorporeal therapy shown to adhere and immunomodulate activated neutrophils and monocytes toward resolution of systemic inflammation. In this overview, we describe a case series of successful transition of pediatric and adult patients with multiorgan failure to successful transplant/device implantation procedures by treatment with the SCD in the following clinical situations: pediatric hemophagocytic lymphohistiocytosis, and adult hepatorenal and cardiorenal syndromes. Application of the SCD in these cases may represent a novel paradigm in increasing clinical eligibility of patients to successful transplant outcomes.
Collapse
Affiliation(s)
| | - Christopher J. Pino
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Lenar T. Yessayan
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Stuart L. Goldstein
- Division of Nephrology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Matthew R. Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Angela J. Westover
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | | | - Kevin K. Chung
- Department of Medical Affairs, SeaStar Medical, Denver, CO
| | - H. David Humes
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI
| |
Collapse
|
3
|
Yessayan LT, Sharma P, Westover AJ, Szamosfalvi B, Humes HD. Extracorporeal Immunomodulation Therapy in Acute Chronic Liver Failure With Multiorgan Failure: First in Human Use. ASAIO J 2024; 70:e53-e56. [PMID: 37643314 PMCID: PMC10902198 DOI: 10.1097/mat.0000000000002033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
Two patients presented with acute on chronic liver failure and multiorgan failure and, as typical for this disorder, they presented with hyperinflammation and anticipated high mortality rates. Both cases were diagnosed with hepatorenal syndrome (HRS). Under a FDA approved Investigational Device Exemption clinical trial, they underwent treatment with an extracorporeal cell-directed immunomodulatory device, called selective cytopheretic device. Both patients showed rapid clinical improvement associated with a decline in elevated blood cytokine concentrations and diminution of activation levels of circulating leukocytes. On follow-up, one patient was alive at day 90 after treatment and undergoing liver transplantation evaluation and the other patient had a successful liver transplantation 6 days after selective cytopheretic device therapy ended. These cases represent the first in human evaluation of extracorporeal cell-directed immunomodulation therapy in acute on chronic liver failure with successful clinical outcomes in a disorder with dismal prognosis.
Collapse
Affiliation(s)
- Lenar T Yessayan
- From the Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | |
Collapse
|
4
|
Teixeira JP, Zeidman A, Beaubien-Souligny W, Cerdá J, Connor MJ, Eggleston K, Juncos LA, da Silva JR, Wells C, Yessayan L, Barker AB, McConville W, Speer R, Wille KM, Neyra JA, Tolwani A. Proceedings of the 2022 UAB CRRT Academy: Non-Invasive Hemodynamic Monitoring to Guide Fluid Removal with CRRT and Proliferation of Extracorporeal Blood Purification Devices. Blood Purif 2023; 52:857-879. [PMID: 37742622 DOI: 10.1159/000533573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/29/2023] [Indexed: 09/26/2023]
Abstract
In 2022, we celebrated the 15th anniversary of the University of Alabama at Birmingham (UAB) Continuous Renal Replacement Therapy (CRRT) Academy, a 2-day conference attended yearly by an international audience of over 100 nephrology, critical care, and multidisciplinary trainees and practitioners. This year, we introduce the proceedings of the UAB CRRT Academy, a yearly review of select emerging topics in the field of critical care nephrology that feature prominently in the conference. First, we review the rapidly evolving field of non-invasive hemodynamic monitoring and its potential to guide fluid removal by renal replacement therapy (RRT). We begin by summarizing the accumulating data associating fluid overload with harm in critical illness and the potential for harm from end-organ hypoperfusion caused by excessive fluid removal with RRT, underscoring the importance of accurate, dynamic assessment of volume status. We describe four applications of point-of-care ultrasound used to identify patients in need of urgent fluid removal or likely to tolerate fluid removal: lung ultrasound, inferior vena cava ultrasound, venous excess ultrasonography, and Doppler of the left ventricular outflow track to estimate stroke volume. We briefly introduce other minimally invasive hemodynamic monitoring technologies before concluding that additional prospective data are urgently needed to adapt these technologies to the specific task of fluid removal by RRT and to learn how best to integrate them into practical fluid-management strategies. Second, we focus on the growth of novel extracorporeal blood purification devices, starting with brief reviews of the inflammatory underpinnings of multiorgan dysfunction and the specific applications of pathogen, endotoxin, and/or cytokine removal and immunomodulation. Finally, we review a series of specific adsorptive technologies, several of which have seen substantial clinical use during the COVID-19 pandemic, describing their mechanisms of target removal, the limited existing data supporting their efficacy, ongoing and future studies, and the need for additional prospective trials.
Collapse
Affiliation(s)
- J Pedro Teixeira
- Division of Nephrology and Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Amanda Zeidman
- Division of Nephrology, Department of Medicine, Institute for Critical Care Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Jorge Cerdá
- Department of Medicine, Nephrology, Albany Medical College, Albany, New York, USA
| | - Michael J Connor
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine and Division of Renal Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | | | - Luis A Juncos
- Division of Nephrology, Department of Internal Medicine, Central Arkansas Veterans' Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Catherine Wells
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lenar Yessayan
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew B Barker
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Wendy McConville
- School of Nursing, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Rajesh Speer
- Division of Nephrology, Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Keith M Wille
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Javier A Neyra
- Division of Nephrology, Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Ashita Tolwani
- Division of Nephrology, Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
5
|
Hambrick HR, Short K, Askenazi D, Krallman K, Pino C, Yessayan L, Westover A, Humes HD, Goldstein SL. Hemolytic Uremic Syndrome-Induced Acute Kidney Injury Treated via Immunomodulation with the Selective Cytopheretic Device. Blood Purif 2023; 52:812-820. [PMID: 37607519 DOI: 10.1159/000531963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023]
Abstract
INTRODUCTION Shiga-toxin associated-hemolytic uremic syndrome (STEC-HUS) is a severe cause of acute kidney injury (AKI) in children. Although most children recover, about 5% die and 30% develop chronic renal morbidity. HUS pathophysiology includes activated neutrophils damaging vascular endothelial cells. Therapeutic immunomodulation of activated neutrophils may alter the progression of disease. We present 3 pediatric patients treated with the selective cytopheretic device (SCD). METHODS We describe a 12 y.o. (patient 1) and two 2 y.o. twins (patients 2 and 3) with STEC-HUS requiring continuous renal replacement therapy (CRRT) who were enrolled in two separate studies of the SCD. RESULTS Patient 1 presented with STEC-HUS causing AKI and multisystem organ failure and received 7 days of SCD and CRRT treatment. After SCD initiation, the patient had gradual recovery of multi-organ dysfunction, with normal kidney and hematologic parameters at 60-day follow-up. Patients 2 and 3 presented with STEC-HUS with AKI requiring dialysis. Each received 24 h of SCD therapy. Thereafter, both gradually improved, with normalization (patient 2) and near-normalization (patient 3) of kidney function at 60-day follow-up. CONCLUSION Immunomodulatory treatment with the SCD was associated with improvements in multisystem stigmata of STEC-HUS-induced AKI and was well-tolerated without any device-related adverse events.
Collapse
Affiliation(s)
- H Rhodes Hambrick
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Acute Care Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kara Short
- Pediatric and Infant Center for Acute Nephrology, Children's of Alabama, Birmingham, Alabama, USA
| | - David Askenazi
- Pediatric and Infant Center for Acute Nephrology, Children's of Alabama, Birmingham, Alabama, USA
| | - Kelli Krallman
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Acute Care Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Christopher Pino
- Division of Nephrology, University of Michigan Department of Medicine, Ann Arbor, Michigan, USA
- Innovative BioTherapies Inc., Ann Arbor, Michigan, USA
| | - Lenar Yessayan
- Division of Nephrology, University of Michigan Department of Medicine, Ann Arbor, Michigan, USA
- Innovative BioTherapies Inc., Ann Arbor, Michigan, USA
| | - Angela Westover
- Division of Nephrology, University of Michigan Department of Medicine, Ann Arbor, Michigan, USA
- Innovative BioTherapies Inc., Ann Arbor, Michigan, USA
| | - H David Humes
- Division of Nephrology, University of Michigan Department of Medicine, Ann Arbor, Michigan, USA
- Innovative BioTherapies Inc., Ann Arbor, Michigan, USA
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Acute Care Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
6
|
Humes HD, Aaronson KD, Buffington DA, Sabbah HN, Westover AJ, Yessayan LT, Szamosfalvi B, Pagani FD. Translation of immunomodulatory therapy to treat chronic heart failure: Preclinical studies to first in human. PLoS One 2023; 18:e0273138. [PMID: 37023139 PMCID: PMC10079025 DOI: 10.1371/journal.pone.0273138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Inflammation has been associated with progression and complications of chronic heart failure (HF) but no effective therapy has yet been identified to treat this dysregulated immunologic state. The selective cytopheretic device (SCD) provides extracorporeal autologous cell processing to lessen the burden of inflammatory activity of circulating leukocytes of the innate immunologic system. AIM The objective of this study was to evaluate the effects of the SCD as an extracorporeal immunomodulatory device on the immune dysregulated state of HF. HF. METHODS AND RESULTS SCD treatment in a canine model of systolic HF or HF with reduced ejection fraction (HFrEF) diminished leukocyte inflammatory activity and enhanced cardiac performance as measured by left ventricular (LV) ejection fraction and stroke volume (SV) up to 4 weeks after treatment initiation. Translation of these observations in first in human, proof of concept clinical study was evaluated in a patient with severe HFrEFHFrEF ineligible for cardiac transplantation or LV LV assist device (LVAD) due to renal insufficiency and right ventricular dysfunction. Six hour SCD treatments over 6 consecutive days resulted in selective removal of inflammatory neutrophils and monocytes and reduction in key plasma cytokines, including tumor necrosis factor-alpha (TNF-α),), interleukin (IL)-6, IL-8, and monocyte chemoattractant protein (MCP)-1. These immunologic changes were associated with significant improvements in cardiac power output, right ventricular stroke work index, cardiac index and LVSV index…. Stabilization of renal function with progressive volume removal permitted successful LVAD implantation. CONCLUSION This translational research study demonstrates a promising immunomodulatory approach to improve cardiac performance in HFrEFHFrEF and supports the important role of inflammation in the progression of HFHF.
Collapse
Affiliation(s)
- H. David Humes
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Innovative Biotherapies, Ann Arbor, Michigan, United States of America
| | - Keith D. Aaronson
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Deborah A. Buffington
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hani N. Sabbah
- Department of Medicine, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Angela J. Westover
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Innovative Biotherapies, Ann Arbor, Michigan, United States of America
| | - Lenar T. Yessayan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Balazs Szamosfalvi
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Francis D. Pagani
- Department of Cardiovascular Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
7
|
Goldstein SL, Yessayan LT, Krallman KA, Collins M, Benoit S, Westover A, Humes HD. Use of extracorporeal immunomodulation in a toddler with hemophagocytic lymphohistiocytosis and multisystem organ failure. Pediatr Nephrol 2023; 38:927-931. [PMID: 35869162 PMCID: PMC9307428 DOI: 10.1007/s00467-022-05692-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Hemophagocytic lymphohistiocytosis (HLH) is a dysregulated immune disorder in children, associated with Epstein-Barr virus (EBV) infection or malignancies. In severe forms, HLH presents with signs and symptoms of hyperinflammation that progress to life-threatening multiorgan failure. Intervention with an extracorporeal immunomodulatory treatment utilizing a selective cytopheretic device (SCD) could be beneficial. The SCD with regional citrate anticoagulation selectively binds the most highly activated circulating neutrophils and monocytes and deactivates them before release to the systemic circulation. Multiple clinical studies, including a multicenter study in children, demonstrate SCD therapy attenuates hyperinflammation, resolves ongoing tissue injury and allows progression to functional organ recovery. We report the first case of SCD therapy in a patient with HLH and multi-organ failure. CASE DIAGNOSIS/TREATMENT A previously healthy 22-month-old toddler presented with fever, abdominal distension, organomegaly, pancytopenia, and signs of hyperinflammation. EBV PCR returned at > 25 million copies. The clinical and laboratory pictures were consistent with systemic EBV-positive T-cell lymphoma with symptoms secondary to HLH. The patient met inclusion criteria for an ongoing study of integration of the SCD with a continuous kidney replacement therapy (CKRT) as part of standard of care. The patient received CKRT-SCD for 4 days with normalization of serum markers of sepsis and inflammation. The patient underwent hematopoietic stem cell transplantation 52 days after presentation and has engrafted with normal kidney function 8 months later. CONCLUSIONS SCD treatment resulted in improvement of poor tissue perfusion reflected by rapid decline in serum lactate levels, lessened systemic capillary leak with discontinuation of vasoactive agents, and repair and recovery of lung and kidney function with extubation and removal of hemodialysis support.
Collapse
Affiliation(s)
- Stuart L Goldstein
- Center for Acute Care Nephrology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7022, Cincinnati, Ohio, 45229, USA.
| | - Lenar T Yessayan
- University of Michigan Medical Center, University of Michigan Hospital, Ann Arbor, MI, USA
| | - Kelli A Krallman
- Center for Acute Care Nephrology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7022, Cincinnati, Ohio, 45229, USA
| | - Michaela Collins
- Center for Acute Care Nephrology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7022, Cincinnati, Ohio, 45229, USA
| | - Stefanie Benoit
- Cincinnati Children's Hospital Medical Center Burnet Campus, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Angela Westover
- University of Michigan Medical Center, University of Michigan Hospital, Ann Arbor, MI, USA
| | - H David Humes
- University of Michigan Medical Center, University of Michigan Hospital, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Chang K, Li Y, Qin Z, Zhang Z, Wang L, Yang Q, Geng J, Deng N, Chen S, Su B. Effect of extracorporeal hemoadsorption in critically ill patients with COVID-19: A narrative review. Front Immunol 2023; 14:1074465. [PMID: 36817416 PMCID: PMC9936071 DOI: 10.3389/fimmu.2023.1074465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
COVID-19 has been affecting the world unprecedentedly and will remain widely prevalent due to its elusive pathophysiological mechanism and the continuous emergence of new variants. Critically ill patients with COVID-19 are commonly associated with cytokine storm, multiple organ dysfunction, and high mortality. To date, growing evidence has shown that extracorporeal hemoadsorption can exert its adjuvant effect to standard of care by regulating immune homeostasis, reducing viremia, and decreasing endotoxin activity in critically ill COVID-19 cases. However, the selection of various hemofilters, timing of initiation and termination of hemoadsorption therapy, anticoagulation management of extracorporeal circuits, identification of target subgroups, and ultimate survival benefit remain controversial. The purpose of this narrative review is to comprehensively summarize the rationale for the use of hemoadsorption in critically ill patients with COVID-19 and to gather the latest clinical evidence in this field.
Collapse
Affiliation(s)
- Kaixi Chang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Yupei Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Qin
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuyun Zhang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Liya Wang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Qinbo Yang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiwen Geng
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Ningyue Deng
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Shanshan Chen
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China.,Med-X Center for Materials, Sichuan University, Chengdu, China.,Med+ Biomaterial Institute of West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Lemoine S, Penny J, Fraser DD, Salerno FR, Dorie J, Tamasi T, Arntfield R, House A, Slessarev M, McIntyre CW. Initial evaluation of extracorporeal immunomodulatory therapy for the treatment of critically ill COVID-19 infected patients. Sci Rep 2022; 12:20236. [PMID: 36424380 PMCID: PMC9684905 DOI: 10.1038/s41598-022-21944-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 10/06/2022] [Indexed: 11/27/2022] Open
Abstract
Severe COVID-19 infection results in significant immune dysregulation resulting from excessive recruitment and activation of neutrophils. The aim of this study was to confirm feasibility, initial safety and detect signal of efficacy of a non-propriety device delivered using an intermittent extra-corporeal system (LMOD) allowing leucocytes modulation in the setting of Severe COVID-19 infection. Twelve patients were recruited. Inclusion criteria were > 18 years age, confirmed COVID-19, acute respiratory distress syndrome requiring mechanical support and hypotension requiring vasopressor support. Primary end point was vasopressor requirements (expressed as epinephrine dose equivalents) and principle secondary endpoints related to safety, ability to deliver the therapy and markers of inflammation assessed over five days after treatment initiation. LMOD treatment appeared safe, defined by hemodynamic stability and no evidence of white cell number depletion from blood. We demonstrated a significant decrease in vasopressor doses (-37%, p = 0.02) in patients receiving LMOD therapy (despite these patients having to tolerate an additional extracorporeal intermittent therapy). Vasopressor requirements unchanged/increasing in control group (+ 10%, p = 0.48). Although much about the use of this therapy in the setting of severe COVID-19 infection remains to be defined (e.g. optimal dose and duration), this preliminary study supports the further evaluation of this novel extracorporeal approach.
Collapse
Affiliation(s)
- Sandrine Lemoine
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Jarrin Penny
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
- Division of Nephrology, Schulich School of Medicine & Dentistry, Western Ontario, London, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Douglas D Fraser
- Lawson Health Research Institute, London, ON, Canada
- Departments of Pediatrics, Clinical Neurological Sciences and Physiology and Pharmacology, Western University, London, ON, Canada
| | - Fabio R Salerno
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada
- Robarts Research Institute, Western University, London, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Justin Dorie
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Tanya Tamasi
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Robert Arntfield
- Lawson Health Research Institute, London, ON, Canada
- Departments of Critical Care Medicine, Victoria Hospital, London, Canada
| | - Andrew House
- Division of Nephrology, Schulich School of Medicine & Dentistry, Western Ontario, London, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Marat Slessarev
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada
- Lawson Health Research Institute, London, ON, Canada
- Departments of Critical Care Medicine, Victoria Hospital, London, Canada
| | - Christopher W McIntyre
- The Lilibeth Caberto Kidney Clinical Research Unit (KCRU), Kidney Clinical Research Unit Room ELL-101, London Health Sciences, University of Western Ontario, Centre 800 Commissioners Rd E, London, ON, N6A5W9, Canada.
- Robarts Research Institute, Western University, London, Canada.
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada.
- Division of Nephrology, Schulich School of Medicine & Dentistry, Western Ontario, London, Canada.
- Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
10
|
Niazi NS, Nassar TI, Stewart IJ, Honore PM, Sharma K, Chung KK. A Review of Extracorporeal Blood Purification Techniques for the Treatment of Critically Ill Coronavirus Disease 2019 Patients. ASAIO J 2022; 68:1219-1227. [PMID: 35417433 PMCID: PMC9521577 DOI: 10.1097/mat.0000000000001761] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In late 2019, a novel betacoronavirus, later termed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was discovered in patients with an unknown respiratory illness in Wuhan, China. SARS-CoV-2 and the disease caused by the novel coronavirus, coronavirus disease 2019 (COVID-19), spread rapidly and resulted in the World Health Organization declaring a pandemic in March 2020. In a minority of patients infected with SARS-CoV-2, severe illness develops characterized by a dysregulated immune response, acute respiratory distress syndrome, and multisystem organ failure. Despite the development of antiviral and multiple immunomodulatory therapies, outcomes of severe illness remain poor. In response, the Food and Drug Administration in the United States authorized the emergency use of several extracorporeal blood purification (EBP) devices for critically ill patients with COVID-19. Extracorporeal blood purification devices target various aspects of the host response to infection to reduce immune dysregulation. This review highlights the underlying technology, currently available literature on use in critically ill COVID-19 patients, and future studies involving four EBP platforms: 1) oXiris filter, 2) CytoSorb filter, 3) Seraph 100 Microbind blood affinity filter, and 4) the Spectra Optia Apheresis System with the Depuro D2000 Adsorption Cartridge.
Collapse
Affiliation(s)
- Nicholas S. Niazi
- From the Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Tareq I. Nassar
- From the Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ian J. Stewart
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Patrick M. Honore
- ICU Department, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Kumar Sharma
- From the Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Kevin K. Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
11
|
Mohseni Afshar Z, Barary M, Babazadeh A, Tavakoli Pirzaman A, Hosseinzadeh R, Alijanpour A, Allahgholipour A, Miri SR, Sio TT, Sullman MJM, Carson‐Chahhoud K, Ebrahimpour S. The role of cytokines and their antagonists in the treatment of COVID-19 patients. Rev Med Virol 2022; 33:e2372. [PMID: 35621229 PMCID: PMC9347599 DOI: 10.1002/rmv.2372] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 01/28/2023]
Abstract
The coronavirus disease 2019 (COVID-19) has various presentations, of which immune dysregulation or the so-called cytokine storm syndrome (COVID-CSS) is prominent. Even though cytokines are vital regulators of body immunoinflammatory responses, their exaggerated release can be harmful. This hyperinflammatory response is more commonly observed during severe COVID-19 infections, caused by the excessive release of pro-inflammatory cytokines, such as interleukin-1 (IL-1), IL-6, IL-8, tumour necrosis factor, granulocyte-macrophage colony-stimulating factor, and interferon-gamma, making their blockers and antagonists of great interest as therapeutic options in this condition. Thus, the pathophysiology of excessive cytokine secretion is outlined, and their most important blockers and antagonists are discussed, mainly focussing on tocilizumab, an interleukin-6 receptor blocker approved to treat severe COVID-19 infections.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development CenterImam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
| | - Mohammad Barary
- Student Research CommitteeVirtual School of Medical Education and ManagementShahid Beheshti University of Medical SciencesTehranIran,Students' Scientific Research Center (SSRC)Tehran University of Medical SciencesTehranIran
| | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research CenterHealth Research InstituteBabol University of Medical SciencesBabolIran
| | | | | | | | - Amirreza Allahgholipour
- Student Research CommitteeSchool of Nursing and MidwiferyShahid Beheshti University of Medical SciencesTehranIran
| | - Seyed Rouhollah Miri
- Cancer Research CenterCancer Institute of IranTehran University of Medical ScienceTehranIran
| | - Terence T. Sio
- Department of Radiation OncologyMayo ClinicPhoenixArizonaUSA
| | - Mark J. M. Sullman
- Department of Social SciencesUniversity of NicosiaNicosiaCyprus,Department of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| | | | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research CenterHealth Research InstituteBabol University of Medical SciencesBabolIran
| |
Collapse
|
12
|
|
13
|
Yang Y, Xiaohui Y, Miao S, Yingshuo Z. The causal relationship between circulating cytokines and critically ill COVID-19: A bidirectional Mendelian randomization analysis. J Glob Health 2022; 12:05010. [PMID: 35356648 PMCID: PMC8934535 DOI: 10.7189/jogh.12.05010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background In this study, we performed a bidirectional mendelian randomization analysis on circulating cytokines and critically ill COVID-19. Methods Both the exposure and outcome data were obtained from public genome wide association study (GWAS) database. We extracted independent instrumental variables from exposure at genome level significance (P < 5 × 10−8). Wald ratio or inverse variance weighted (IVW) method were used for estimating the causal relationships between circulating cytokines and critically ill COVID-19. Results Only IL5 (cytokines to critically ill COVID-19 direction) and bNGF, IL8 (critically ill COVID-19 to cytokines direction) showed suggestive causal relations. However, these associations lost significance after FDR correction. Another validation data set of critically ill COVID-19 did not confirm these associations, either. Conclusions Our Mendelian randomization did not find causal relationships between analyzable circulating cytokines and critically ill COVID-19.
Collapse
Affiliation(s)
- Yu Yang
- Endocrinology Department, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Yang Xiaohui
- Endocrinology Department, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Sui Miao
- Endocrinology Department, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Zhong Yingshuo
- Endocrinology Department, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
14
|
Go YY, Ju WM, Lee CM, Chae SW, Song JJ. Different Transcutaneous Auricular Vagus Nerve Stimulation Parameters Modulate the Anti-Inflammatory Effects on Lipopolysaccharide-Induced Acute Inflammation in Mice. Biomedicines 2022; 10:biomedicines10020247. [PMID: 35203459 PMCID: PMC8869637 DOI: 10.3390/biomedicines10020247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Vagus nerve stimulation (VNS) is considered a potential method for anti-inflammation due to the involvement of the VN in the cholinergic anti-inflammatory pathway (CAP) formation of a connection between the central nervous system and peripheral immune cells that help relieve inflammation. However, whether a non-invasive transcutaneous auricular VNS (taVNS) modulates the inflammation levels via altering the parameter of taVNS is poorly understood. This study aimed to determine the differential inhibitory effects of taVNS on lipopolysaccharide (LPS)-induced systemic inflammation using electrical stimulation parameters such as pulse frequency and time. The taVNS-promoted CAP activity significantly recovered LPS-induced tissue injuries (lung, spleen, and intestine) and decreased inflammatory cytokine levels and tissue-infiltrated immune cells. Interestingly, the anti-inflammatory capacity of taVNS with 15 Hz was much higher than that of taVNS with 25 Hz. When a cytokine array was used to investigate the changes of inflammation and immune response-related cytokines/chemokines expression in taVNS with 15 Hz or 25 Hz treatment in LPS-induced endotoxemia in mice, most of the expression of cytokines/chemokines associated with pro-inflammation was severely decreased in taVNS with 15 Hz compared to 25 Hz. This study demonstrated that the taVNS parameter could differentially modulate the inflammation levels of animals, suggesting the importance of taVNS parameter selection for use in feasible interventions for acute inflammation treatment.
Collapse
Affiliation(s)
- Yoon-Young Go
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul 08308, Korea; (Y.-Y.G.); (W.-M.J.); (C.-M.L.); (S.-W.C.)
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul 08308, Korea
| | - Won-Min Ju
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul 08308, Korea; (Y.-Y.G.); (W.-M.J.); (C.-M.L.); (S.-W.C.)
| | - Chan-Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul 08308, Korea; (Y.-Y.G.); (W.-M.J.); (C.-M.L.); (S.-W.C.)
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul 08308, Korea; (Y.-Y.G.); (W.-M.J.); (C.-M.L.); (S.-W.C.)
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul 08308, Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul 08308, Korea; (Y.-Y.G.); (W.-M.J.); (C.-M.L.); (S.-W.C.)
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul 08308, Korea
- Neurive Institute, Neurive Co., Ltd., Seoul 08308, Korea
- Correspondence: or ; Tel.: +82-2-2626-3186; Fax: +82-2-2626-0475
| |
Collapse
|
15
|
Raina R, Sethi SK, Chakraborty R, Singh S, Teo S, Khooblall A, Montini G, Bunchman T, Topaloglu R, Yap HK. Blood Filters in Children with COVID-19 and AKI: A Review. Ther Apher Dial 2022; 26:566-582. [PMID: 34997670 DOI: 10.1111/1744-9987.13793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/11/2021] [Accepted: 01/03/2022] [Indexed: 11/28/2022]
Abstract
COVID-19 has challenged the global healthcare system through rapid proliferation and lack of existing treatment resulting in over 180 million cases and 3.8 million deaths since December 2019. Although pediatric patients only comprise 1-2% of diagnosed cases, their incidence of acute kidney injury ranges from 8.2% to 18.2% compared to 49% in adults. Severe infection, initiated by dysregulated host response, can lead to multiorgan failure. In this review, we focus on the use of various blood filters approved for use in pediatric kidney replacement therapy to mitigate adverse effects of severe illness. Therapeutic effects of these blood filters range from cytokine removal (CytoSorb, HA330, HCO/MCO), endotoxin removal (Toraymyxin, CPFA), both cytokine and endotoxin removal (oXiris), and non-specific removal of proteins (PMMA) that have already been established and can be used to mitigate the various effects of the cytokine storm syndrome in COVID-19.
Collapse
Affiliation(s)
- Rupesh Raina
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH.,Department of Nephrology, Akron Children's Hospital, Akron, OH
| | - Sidharth Kumar Sethi
- Pediatric Nephrology, Kidney Institute, Medanta, The Medicity Hospital, Gurgaon, Haryana, India
| | - Ronith Chakraborty
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH.,Department of Nephrology, Akron Children's Hospital, Akron, OH
| | - Siddhartha Singh
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH.,Department of Nephrology, Akron Children's Hospital, Akron, OH
| | - Sharon Teo
- Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Amrit Khooblall
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH.,Department of Nephrology, Akron Children's Hospital, Akron, OH
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda IRCCS, Policlinico di Milano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Timothy Bunchman
- Pediatric Nephrology & Transplantation, Children's Hospital of Richmond at VCU, Richmond, VA
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Hui Kim Yap
- Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore, Singapore.,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
16
|
Gao Z, Song Y, Hsiao TY, He J, Wang C, Shen J, MacLachlan A, Dai S, Singer BH, Kurabayashi K, Chent P. Machine-Learning-Assisted Microfluidic Nanoplasmonic Digital Immunoassay for Cytokine Storm Profiling in COVID-19 Patients. ACS NANO 2021; 15:18023-18036. [PMID: 34714639 PMCID: PMC8577373 DOI: 10.1021/acsnano.1c06623] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/25/2021] [Indexed: 05/08/2023]
Abstract
Cytokine storm, known as an exaggerated hyperactive immune response characterized by elevated release of cytokines, has been described as a feature associated with life-threatening complications in COVID-19 patients. A critical evaluation of a cytokine storm and its mechanistic linkage to COVID-19 requires innovative immunoassay technology capable of rapid, sensitive, selective detection of multiple cytokines across a wide dynamic range at high-throughput. In this study, we report a machine-learning-assisted microfluidic nanoplasmonic digital immunoassay to meet the rising demand for cytokine storm monitoring in COVID-19 patients. Specifically, the assay was carried out using a facile one-step sandwich immunoassay format with three notable features: (i) a microfluidic microarray patterning technique for high-throughput, multiantibody-arrayed biosensing chip fabrication; (ii) an ultrasensitive nanoplasmonic digital imaging technology utilizing 100 nm silver nanocubes (AgNCs) for signal transduction; (iii) a rapid and accurate machine-learning-based image processing method for digital signal analysis. The developed immunoassay allows simultaneous detection of six cytokines in a single run with wide working ranges of 1-10,000 pg mL-1 and ultralow detection limits down to 0.46-1.36 pg mL-1 using a minimum of 3 μL serum samples. The whole chip can afford a 6-plex assay of 8 different samples with 6 repeats in each sample for a total of 288 sensing spots in less than 100 min. The image processing method enhanced by convolutional neural network (CNN) dramatically shortens the processing time ∼6,000 fold with a much simpler procedure while maintaining high statistical accuracy compared to the conventional manual counting approach. The immunoassay was validated by the gold-standard enzyme-linked immunosorbent assay (ELISA) and utilized for serum cytokine profiling of COVID-19 positive patients. Our results demonstrate the nanoplasmonic digital immunoassay as a promising practical tool for comprehensive characterization of cytokine storm in patients that holds great promise as an intelligent immunoassay for next generation immune monitoring.
Collapse
Affiliation(s)
- Zhuangqiang Gao
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Te Yi Hsiao
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Jiacheng He
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Chuanyu Wang
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Jialiang Shen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Alana MacLachlan
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Siyuan Dai
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Benjamin H. Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Pengyu Chent
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
17
|
Rapid single-molecule digital detection of protein biomarkers for continuous monitoring of systemic immune disorders. Blood 2021; 137:1591-1602. [PMID: 33275650 DOI: 10.1182/blood.2019004399] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Digital protein assays have great potential to advance immunodiagnostics because of their single-molecule sensitivity, high precision, and robust measurements. However, translating digital protein assays to acute clinical care has been challenging because it requires deployment of these assays with a rapid turnaround. Herein, we present a technology platform for ultrafast digital protein biomarker detection by using single-molecule counting of immune-complex formation events at an early, pre-equilibrium state. This method, which we term "pre-equilibrium digital enzyme-linked immunosorbent assay" (PEdELISA), can quantify a multiplexed panel of protein biomarkers in 10 µL of serum within an unprecedented assay incubation time of 15 to 300 seconds over a 104 dynamic range. PEdELISA allowed us to perform rapid monitoring of protein biomarkers in patients manifesting post-chimeric antigen receptor T-cell therapy cytokine release syndrome, with ∼30-minute sample-to-answer time and a sub-picograms per mL limit of detection. The rapid, sensitive, and low-input volume biomarker quantification enabled by PEdELISA is broadly applicable to timely monitoring of acute disease, potentially enabling more personalized treatment.
Collapse
|
18
|
Paisey C, Patvardhan C, Mackay M, Vuylsteke A, Bhagra SK. Continuous hemadsorption with cytokine adsorber for severe COVID-19: A case series of 15 patients. Int J Artif Organs 2021; 44:664-674. [PMID: 34128416 DOI: 10.1177/03913988211023782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hyperinflammation and cytokine release has been associated with severe Covid-19. Hemadsorption cartridges may have a potential role in treatment of cytokine storm associated with the development of severe Covid-19. METHODS We retrospectively examined the case records of patients with severe Covid-19 receiving adjunctive hemadsorption (HA) in our ICU. We analyzed inflammatory biomarkers pre- and post- HA. RESULTS Fifteen patients received HA during the study period. All were intubated, ventilated and required renal replacement therapy. 11/15 were supported on ECMO. Mean ferritin 2652 (±3286) ng/ml, mean CRP 154 (±92) mg/ml, median D-dimer 3071 (±2689) ng/ml, mean troponin 236 (±461) ng/L. We found significant difference in pre-and post- treatment ferritin 3622 ng/ml versus 1682 ng/ml (p = 0.022), CRP 222 mg/ml versus 103 mg/ml (p = 0.008, 95% CI 22.4-126.5), lactate 2 mmol/L versus 1.3 mmol/L (p = 0.017), and procalcitonin 15.3 ng/ml versus 4.2 ng/ml (p = 0.023). No significant difference in pre- and post- treatment IL-6 14 pg/ml versus 43 pg/ml (p = 0.32), IL-10 3.4 pg/ml versus 2.6 pg/ml (p = 0.31), IL1 β 0.37 pg/ml versus 0.77 pg/ml (p = 0.75), TNF α 12.77 pg/ml versus 12.49 pg/ml (p = 0.75), VIS 10.04 versus 6.01 (p = 0.31, 95% CI 5.98-17.16) was seen. CONCLUSIONS The use of HA as adjunctive treatment in a critically unwell group of COVID-19 patients lead to a reduction in ferritin, CRP, procalcitonin and lactate with no significant change in other parameters. The use of HA in the treatment of severe COVID-19 requires further larger randomized studies.
Collapse
Affiliation(s)
- Christopher Paisey
- Department of Intensive Care Medicine, Royal Papworth Hospital, Cambridge, UK
| | - Chinmay Patvardhan
- Department of Intensive Care Medicine, Royal Papworth Hospital, Cambridge, UK
| | - Michael Mackay
- Department of Intensive Care Medicine, Royal Papworth Hospital, Cambridge, UK
| | - Alain Vuylsteke
- Department of Intensive Care Medicine, Royal Papworth Hospital, Cambridge, UK
| | | |
Collapse
|
19
|
Dilogo IH, Aditianingsih D, Sugiarto A, Burhan E, Damayanti T, Sitompul PA, Mariana N, Antarianto RD, Liem IK, Kispa T, Mujadid F, Novialdi N, Luviah E, Kurniawati T, Lubis AMT, Rahmatika D. Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: A randomized controlled trial. Stem Cells Transl Med 2021; 10:1279-1287. [PMID: 34102020 PMCID: PMC8242692 DOI: 10.1002/sctm.21-0046] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
One of the main causes of acute respiratory distress syndrome in coronavirus disease 2019 (COVID-19) is cytokine storm, although the exact cause is still unknown. Umbilical cord mesenchymal stromal cells (UC-MSCs) influence proinflammatory T-helper 2 (Th2 ) cells to shift to an anti-inflammatory agent. To investigate efficacy of UC-MSC administration as adjuvant therapy in critically ill patients with COVID-19, we conducted a double-blind, multicentered, randomized controlled trial at four COVID-19 referral hospitals in Jakarta, Indonesia. This study included 40 randomly allocated critically ill patients with COVID-19; 20 patients received an intravenous infusion of 1 × 106 /kg body weight UC-MSCs in 100 ml saline (0.9%) solution (SS) and 20 patients received 100 ml 0.9% SS as the control group. All patients received standard therapy. The primary outcome was measured by survival rate and/or length of ventilator usage. The secondary outcome was measured by clinical and laboratory improvement, with serious adverse events. Our study showed the survival rate in the UC-MSCs group was 2.5 times higher than that in the control group (P = .047), which is 10 patients and 4 patients in the UC-MSCs and control groups, respectively. In patients with comorbidities, UC-MSC administration increased the survival rate by 4.5 times compared with controls. The length of stay in the intensive care unit and ventilator usage were not statistically significant, and no adverse events were reported. The application of infusion UC-MSCs significantly decreased interleukin 6 in the recovered patients (P = .023). Therefore, application of intravenous UC-MSCs as adjuvant treatment for critically ill patients with COVID-19 increases the survival rate by modulating the immune system toward an anti-inflammatory state.
Collapse
Affiliation(s)
- Ismail Hadisoebroto Dilogo
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Cluster Indonesian Medical Education and Research Institute (IMERI) Universitas Indonesia, Jakarta, Indonesia.,Department of Orthopaedic and Traumatology, Cipto Mangunkusumo General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Dita Aditianingsih
- Department of Anesthesiology and Intensive Care Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia.,Intensive Care Division, Universitas Indonesia Hospital, Depok, Indonesia
| | - Adhrie Sugiarto
- Department of Anesthesiology and Intensive Care Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Erlina Burhan
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine Universitas Indonesia, Persahabatan General Hospital, Jakarta, Indonesia
| | - Triya Damayanti
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine Universitas Indonesia, Persahabatan General Hospital, Jakarta, Indonesia
| | - Pompini Agustina Sitompul
- Directorate of Medical Services, Nursing and Supporting, Sulianti Saroso Infection Disease Hospital, Jakarta, Indonesia
| | - Nina Mariana
- Directorate of Human Resources Development, Education and Operational Sulianti Saroso Infection Disease Hospital, Jakarta, Indonesia
| | - Radiana D Antarianto
- Stem Cell and Tissue Engineering Research Cluster Indonesian Medical Education and Research Institute (IMERI) Universitas Indonesia, Jakarta, Indonesia.,Department of Histology, Universitas Indonesia Fakultas Kedokteran, Jakarta, Indonesia
| | - Isabella Kurnia Liem
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Cluster Indonesian Medical Education and Research Institute (IMERI) Universitas Indonesia, Jakarta, Indonesia.,Department of Anatomy, Universitas Indonesia Fakultas Kedokteran, Jakarta, Indonesia
| | - Tera Kispa
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Fajar Mujadid
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Novialdi Novialdi
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Evah Luviah
- Stem Cell and Tissue Engineering Research Cluster Indonesian Medical Education and Research Institute (IMERI) Universitas Indonesia, Jakarta, Indonesia
| | - Tri Kurniawati
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Andri M T Lubis
- Department of Orthopaedic and Traumatology, Cipto Mangunkusumo General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Installation of Innovation Management and Intellectual Property, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Dina Rahmatika
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
20
|
Azabou E, Bao G, Bounab R, Heming N, Annane D. Vagus Nerve Stimulation: A Potential Adjunct Therapy for COVID-19. Front Med (Lausanne) 2021; 8:625836. [PMID: 34026778 PMCID: PMC8137825 DOI: 10.3389/fmed.2021.625836] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19) through excessive end organ inflammation. Despite improved understanding of the pathophysiology, management, and the great efforts worldwide to produce effective drugs, death rates of COVID-19 patients remain unacceptably high, and effective treatment is unfortunately lacking. Pharmacological strategies aimed at modulating inflammation in COVID-19 are being evaluated worldwide. Several drug therapies targeting this excessive inflammation, such as tocilizumab, an interleukin (IL)-6 inhibitor, corticosteroids, programmed cell death protein (PD)-1/PD-L1 checkpoint inhibition, cytokine-adsorption devices, and intravenous immunoglobulin have been identified as potentially useful and reliable approaches to counteract the cytokine storm. However, little attention is currently paid for non-drug therapeutic strategies targeting inflammatory and immunological processes that may be useful for reducing COVID-19-induced complications and improving patient outcome. Vagus nerve stimulation attenuates inflammation both in experimental models and preliminary data in human. Modulating the activity of cholinergic anti-inflammatory pathways (CAPs) described by the group of KJ Tracey has indeed become an important target of therapeutic research strategies for inflammatory diseases and sepsis. Non-invasive transcutaneous vagal nerve stimulation (t-VNS), as a non-pharmacological adjuvant, may help reduce the burden of COVID-19 and deserve to be investigated. VNS as an adjunct therapy in COVID-19 patients should be investigated in clinical trials. Two clinical trials on this topic are currently underway (NCT04382391 and NCT04368156). The results of these trials will be informative, but additional larger studies are needed.
Collapse
Affiliation(s)
- Eric Azabou
- Clinical Neurophysiology and Neuromodulation Unit, Departments of Physiology and Critical Care Medicine, Raymond Poincaré Hospital, Assistance Publique- Hôpitaux de Paris, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| | - Guillaume Bao
- Clinical Neurophysiology and Neuromodulation Unit, Departments of Physiology and Critical Care Medicine, Raymond Poincaré Hospital, Assistance Publique- Hôpitaux de Paris, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| | - Rania Bounab
- General Intensive Care Unit - Assistance Publique Hôpitaux de Paris, Raymond Poincaré Hospital, Assistance Publique- Hôpitaux de Paris, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| | - Nicholas Heming
- General Intensive Care Unit - Assistance Publique Hôpitaux de Paris, Raymond Poincaré Hospital, Assistance Publique- Hôpitaux de Paris, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| | - Djillali Annane
- General Intensive Care Unit - Assistance Publique Hôpitaux de Paris, Raymond Poincaré Hospital, Assistance Publique- Hôpitaux de Paris, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| |
Collapse
|
21
|
Song Y, Zhao J, Cai T, Stephens A, Su SH, Sandford E, Flora C, Singer BH, Ghosh M, Choi SW, Tewari M, Kurabayashi K. Machine learning-based cytokine microarray digital immunoassay analysis. Biosens Bioelectron 2021; 180:113088. [PMID: 33647790 PMCID: PMC7896497 DOI: 10.1016/j.bios.2021.113088] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 12/15/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022]
Abstract
Serial measurement of a large panel of protein biomarkers near the bedside could provide a promising pathway to transform the critical care of acutely ill patients. However, attaining the combination of high sensitivity and multiplexity with a short assay turnaround poses a formidable technological challenge. Here, the authors develop a rapid, accurate, and highly multiplexed microfluidic digital immunoassay by incorporating machine learning-based autonomous image analysis. The assay has achieved 12-plexed biomarker detection in sample volume <15 μL at concentrations < 5 pg/mL while only requiring a 5-min assay incubation, allowing for all processes from sampling to result to be completed within 40 min. The assay procedure applies both a spatial-spectral microfluidic encoding scheme and an image data analysis algorithm based on machine learning with a convolutional neural network (CNN) for pre-equilibrated single-molecule protein digital counting. This unique approach remarkably reduces errors facing the high-capacity multiplexing of digital immunoassay at low protein concentrations. Longitudinal data obtained for a panel of 12 serum cytokines in human patients receiving chimeric antigen receptor-T (CAR-T) cell therapy reveals the powerful biomarker profiling capability. The assay could also be deployed for near-real-time immune status monitoring of critically ill COVID-19 patients developing cytokine storm syndrome.
Collapse
Affiliation(s)
- Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jingyang Zhao
- Department of Energy Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Tao Cai
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew Stephens
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Erin Sandford
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Christopher Flora
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin H Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Monalisa Ghosh
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sung Won Choi
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Pediatrics, University of Michigan, Ann Arbor, MI, 48109, USA; Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Muneesh Tewari
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, 48109, USA; Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
22
|
Williamson J, Black L, Black A, Koduri G, Kelly C. There are similarities between rheumatic disease with lung involvement and COVID-19 pneumonia. Ir J Med Sci 2021; 191:1-5. [PMID: 33564974 PMCID: PMC7872822 DOI: 10.1007/s11845-021-02545-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/02/2021] [Indexed: 01/08/2023]
Abstract
Introduction There is considerable overlap between the clinical manifestations of covid-19 pneumonia and the acute interstitial lung disease seen in certain rheumatic disorders. In addition, pulmonary fibrosis is increasingly recognised as a potentially serious consequence of both. Methods This review explores this overlap of clinical features, risk factors and causation, offering insights into the immune mechanisms that contribute to both sets of disorders. Results The therapeutic role of immunosuppression and biologic agents in the treatment of covid-19 is explained in the light of this. Discussion We propose how lessons learned from the insights recently gained into each disorder can improve our insight into immunological mechanisms and application of therapeutic interventions in the other.
Collapse
Affiliation(s)
| | | | | | | | - Clive Kelly
- University of Newcastle Upon Tyne, Newcastle upon Tyne, UK.
| |
Collapse
|
23
|
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
|
24
|
Szamosfalvi B, Heung M, Yessayan L. Technology Innovations in Continuous Kidney Replacement Therapy: The Clinician's Perspective. Adv Chronic Kidney Dis 2021; 28:3-12. [PMID: 34389135 DOI: 10.1053/j.ackd.2021.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 11/11/2022]
Abstract
Continuous kidney replacement therapy (CKRT) has improved remarkably since its first implementation as continuous arteriovenous hemofiltration in the 1970s. However, when looking at the latest generation of CKRT machines, one could argue that clinical deployment of breakthrough innovations by device manufacturers has slowed in the last decade. Simultaneously, there has been a steady accumulation of clinical knowledge using CKRT as well as a multitude of therapeutic and diagnostic innovations in the dialysis and broader intensive care unit technology fields adaptable to CKRT. These include multiple different anticlotting measures; cloud-computing for optimized treatment prescribing and delivered therapy data collection and analysis; novel blood purification techniques aimed at improving the severe multiorgan dysfunction syndrome; and real-time sensing of blood and/or filter effluent composition. The authors present a view of how CKRT devices and programs could be reimagined incorporating these innovations to achieve specific measurable clinical outcomes with personalized care and improved simplicity, safety, and efficacy of CKRT therapy.
Collapse
|
25
|
Goldstein SL, Askenazi DJ, Basu RK, Selewski DT, Paden ML, Krallman KA, Kirby CL, Mottes TA, Terrell T, Humes HD. Use of the Selective Cytopheretic Device in Critically Ill Children. Kidney Int Rep 2020; 6:775-784. [PMID: 33732992 PMCID: PMC7938071 DOI: 10.1016/j.ekir.2020.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 11/27/2022] Open
Abstract
Introduction Critically ill children with acute kidney injury (AKI) requiring continuous kidney replacement therapy (CKRT) are at increased risk of death. The selective cytopheretic device (SCD) promotes an immunomodulatory effect when circuit ionized calcium (iCa2+) is maintained at <0.40 mmol/l with regional citrate anticoagulation (RCA). In a randomized trial of adult patients on CRRT, those treated with the SCD maintaining an iCa2+ <0.40 mmol/l had improved survival/dialysis independence. We conducted a US Food and Drug Administration (FDA)–sponsored study to evaluate safety and feasibility of the SCD in 16 critically ill children. Methods Four pediatric intensive care units (ICUs) enrolled children with AKI and multiorgan dysfunction receiving CKRT to receive the SCD integrated post-CKRT membrane. RCA was used to achieve a circuit iCa2+ level <0.40 mmol/l. Subjects received SCD treatment for 7 days or CKRT discontinuation, whichever came first. Results The FDA target enrollment of 16 subjects completed the study from December 2016 to February 2020. Mean age was 12.3 ± 5.1 years, weight was 53.8 ± 28.9 kg, and median Pediatric Risk of Mortality II was 7 (range 2–19). Circuit iCa2+ levels were maintained at <0.40 mmol/l for 90.2% of the SCD therapy time. Median SCD duration was 6 days. Fifteen subjects survived SCD therapy; 12 survived to ICU discharge. All ICU survivors were dialysis independent at 60 days. No SCD-related adverse events (AEs) were reported. Conclusion Our data demonstrate that SCD therapy is feasible and safe in children who require CKRT. Although we cannot make efficacy claims, the 75% survival rate and 100% renal recovery rate observed suggest a possible favorable benefit-to-risk ratio.
Collapse
Affiliation(s)
| | | | - Rajit K Basu
- Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - David T Selewski
- Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Kelli A Krallman
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Cassie L Kirby
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Theresa A Mottes
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tara Terrell
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | |
Collapse
|
26
|
Is It the Missing Piece for Coronavirus Disease 2019, Acute Respiratory Distress Syndrome, and Venovenous Extracorporeal Membrane Oxygenation? ASAIO J 2020; 66:1084-1086. [PMID: 33136593 DOI: 10.1097/mat.0000000000001313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|