51
|
Gullickson M, Nichols L, Scheibe M. A Novel Therapy for a Rare Condition: Continuous Anakinra Infusion for a Patient With Macrophage Activation Syndrome. Cureus 2023; 15:e42968. [PMID: 37667692 PMCID: PMC10475328 DOI: 10.7759/cureus.42968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 09/06/2023] Open
Abstract
Macrophage activation syndrome (MAS) is a type of hemophagocytic lymphohistiocytosis (HLH), which occurs due to excessive stimulation of the immune system. Common precipitants of MAS include disseminated infection or underlying rheumatologic disorders such as adult-onset Still's disease which is characterized as an inflammatory arthritis with daily fevers and a salmon-colored rash. We present a case of a patient with probable adult-onset Still's disease and subsequent disseminated cytomegalovirus (CMV) infection, who met the criteria for MAS based on the presence of a fever, cytopenia in multiple cell lines, elevated ferritin, presence of hemophagocytosis on bone marrow, low fibrinogen, and mild splenomegaly on physical exam. The patient responded to treatment with continuous anakinra infusion and ganciclovir for treatment of CMV. Though cytotoxic medications such as etoposide have traditionally been considered first-line treatment for HLH/MAS, interleukin-1 inhibitors such as anakinra are emerging as aless cytotoxic alternative.
Collapse
Affiliation(s)
| | - Laura Nichols
- Internal Medicine, University of North Dakota School of Medicine and Health Sciences, Fargo, USA
- Internal Medicine, Sanford Health, Fargo, USA
| | | |
Collapse
|
52
|
Talasaz AH, Sculthorpe R, Pak M, Lipinski M, Roberts C, Markley R, Trankle CR, Canada JM, Wohlford GF, Golino M, Dixon D, Van Tassell BW, Abbate A. Comparison of Safety and Biological Efficacy of Anakinra (Kineret) Dispensed in Polycarbonate Plastic versus Borosilicate Glass Syringes: A Patient-Level Analysis of VCUART2 and VCUART3 Clinical Trials. J Pharmacol Exp Ther 2023; 386:138-142. [PMID: 36868827 PMCID: PMC10353132 DOI: 10.1124/jpet.122.001404] [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: 08/10/2022] [Revised: 02/09/2023] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Anakinra is a recombinant human interleukin-1 receptor antagonist approved for the treatment of inflammatory diseases. Kineret is available as a solution prepared in a borosilicate glass syringe. For implementing a placebo-controlled double-blind randomized clinical trial, anakinra is commonly transferred into plastic syringes. However, there is limited data on anakinra's stability in polycarbonate syringes. We described the results of our previous studies on the use of anakinra in glass (VCUART3) versus plastic syringes (VCUART2) compared with placebo. These studies were conducted in patients with ST-segment elevation myocardial infarction (STEMI), and we assessed the anti-inflammatory effects of anakinra versus placebo by comparing the area under the curve for high-sensitivity cardiac reactive protein (AUC-CRP) levels during the first 14 days of STEMI, its clinical effects on heart failure (HF) hospitalization, cardiovascular death, or new diagnosis of HF as well as adverse events profile between groups. The levels of AUC-CRP were 75 (50-255 mg·day/l) for anakinra in plastic syringes versus 255 (116-592 mg·day/l) in placebo and 60 (24-139 mg·day/l) and 86 (43-123 mg·day/l) for anakinra once and twice daily in glass syringes, respectively, compared with placebo 214 (131-394 mg·day/l). The rate of adverse events was also comparable between groups. There were no differences in the rate of HF hospitalization or cardiovascular death in patients who received anakinra in plastic or glass syringes. Fewer cases of new-onset heart failure occurred in patients receiving anakinra in plastic or glass syringes compared with placebo. Anakinra stored in plastic (polycarbonate) syringes provides comparable biologic and clinical effect to glass (borosilicate) syringes. SIGNIFICANCE STATEMENT: Anakinra (Kineret) 100 mg administered subcutaneously in patients with ST-segment elevation myocardial infarction (STEMI) for a duration of up to 14 days appears to have comparable safety and biological efficacy signals when delivered in prefilled glass or transferred into plastic polycarbonate syringes. This may have important implications for the feasibility of designing clinical trials in STEMI and other clinical conditions.
Collapse
Affiliation(s)
- Azita H Talasaz
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Robin Sculthorpe
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Mary Pak
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Michael Lipinski
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Charlotte Roberts
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Roshanak Markley
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Cory R Trankle
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Justin M Canada
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - George F Wohlford
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Michele Golino
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Dave Dixon
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Benjamin W Van Tassell
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Antonio Abbate
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| |
Collapse
|
53
|
Zhang L, Jiang Y, He J, Chen J, Qi R, Yuan L, Shao T, Zhao H, Chen C, Chen Y, Wang X, Lei X, Gao Q, Zhuang C, Zhou M, Ma J, Liu W, Yang M, Fu R, Wu Y, Chen F, Xiong H, Nie M, Chen Y, Wu K, Fang M, Wang Y, Zheng Z, Huang S, Ge S, Cheng SC, Zhu H, Cheng T, Yuan Q, Wu T, Zhang J, Chen Y, Zhang T, Li C, Qi H, Guan Y, Xia N. Intranasal influenza-vectored COVID-19 vaccine restrains the SARS-CoV-2 inflammatory response in hamsters. Nat Commun 2023; 14:4117. [PMID: 37433761 DOI: 10.1038/s41467-023-39560-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/19/2023] [Indexed: 07/13/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants and "anatomical escape" characteristics threaten the effectiveness of current coronavirus disease 2019 (COVID-19) vaccines. There is an urgent need to understand the immunological mechanism of broad-spectrum respiratory tract protection to guide broader vaccines development. Here we investigate immune responses induced by an NS1-deleted influenza virus vectored intranasal COVID-19 vaccine (dNS1-RBD) which provides broad-spectrum protection against SARS-CoV-2 variants in hamsters. Intranasal delivery of dNS1-RBD induces innate immunity, trained immunity and tissue-resident memory T cells covering the upper and lower respiratory tract. It restrains the inflammatory response by suppressing early phase viral load post SARS-CoV-2 challenge and attenuating pro-inflammatory cytokine (Il6, Il1b, and Ifng) levels, thereby reducing excess immune-induced tissue injury compared with the control group. By inducing local cellular immunity and trained immunity, intranasal delivery of NS1-deleted influenza virus vectored vaccine represents a broad-spectrum COVID-19 vaccine strategy to reduce disease burden.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yao Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Jinhang He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Junyu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Ruoyao Qi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Tiange Shao
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Hui Zhao
- National Institute for Food and Drug Control, 102629, Beijing, China
| | - Congjie Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yaode Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Xijing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Xing Lei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Qingxiang Gao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Chunlan Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Ming Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Jian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Wei Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Man Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Rao Fu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yangtao Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Feng Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Hualong Xiong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Meifeng Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yiyi Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Kun Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Mujin Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Yingbin Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Zizheng Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Shoujie Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Shengxiang Ge
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Shih Chin Cheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, 515063, Shantou, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China
| | - Ting Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| | - Changgui Li
- National Institute for Food and Drug Control, 102629, Beijing, China.
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, 100084, Beijing, China.
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China.
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, 515063, Shantou, China.
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health & School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, 361102, Xiamen, Fujian, China.
| |
Collapse
|
54
|
Phoophiboon V, Brown P, Burns KEA. Systemic lupus erythematosus associated with development of macrophage activation syndrome and disseminated aspergillosis. Can J Anaesth 2023; 70:1255-1260. [PMID: 37349668 DOI: 10.1007/s12630-023-02506-2] [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: 05/13/2022] [Revised: 01/05/2023] [Accepted: 01/14/2023] [Indexed: 06/24/2023] Open
Abstract
PURPOSE Macrophage activation syndrome (MAS) is a rare illness, especially in critically ill adults. The diagnosis of MAS is challenging, requiring the expertise of multiple specialists, and treatments for MAS can be associated with catastrophic complications. CLINICAL FEATURES We describe the case of a 31-yr-old Vietnamese student who was diagnosed with cutaneous systemic lupus erythematosus (SLE) in November 2020 and was initiated on treatment with low-dose corticosteroids and hydroxychloroquine as an outpatient. Ten days later, she presented to hospital with decreased consciousness, fever, periorbital swelling, and hypotension necessitating intubation. Computed tomography angiography (CTA) and lumbar puncture did not show a stroke or central nervous system infection. Serology and clinical presentation were consistent with MAS. She was initially treated with 4.5 g pulse methylprednisolone and subsequently with the interleukin-1 receptor antagonist, anakinra, and maintenance corticosteroids because of persistently elevated inflammatory markers. Her intensive care unit stay was complicated by aspiration, airway obstruction due to fungal tracheobronchitis necessitating extracorporeal membrane oxygenation (ECMO), and ring-enhancing cerebral lesions, and, ultimately, massive hemoptysis resulting in death. CONCLUSIONS Four features of this case merit discussion, including the: 1) infrequent association of SLE with MAS; 2) short interval between SLE diagnosis and critical illness; 3) manifestation of fungal tracheobronchitis with airway obstruction; and 4) lack of response to antifungal treatment while receiving ECMO.
Collapse
Affiliation(s)
- Vorakamol Phoophiboon
- Interdepartmental Division of Critical Care Medicine, Temerty School of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Paula Brown
- Department of Pharmacy, University Health Network, Toronto, ON, Canada
| | - Karen E A Burns
- Division of Critical Care Medicine, Unity Health Toronto - St. Michael's Hospital, Li Ka Shing Knowledge Institute, 30 Bond Street, Office 4-045 Donnelly Wing, Toronto, ON, M5B 1W8, Canada
| |
Collapse
|
55
|
Reiff DD, Cron RQ. Cytokine Storm Syndrome Triggered by Extracorporeal Membrane Oxygenation in Pediatric Patients. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1052. [PMID: 37371283 DOI: 10.3390/children10061052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
Cytokine storm syndrome (CSS) is a serious and potentially life-threatening condition caused by severe systemic inflammation, immune activation, and a positive feedback loop of cytokine release. Typically triggered by systemic infection, malignancy, monogenic or rheumatic disease, similar patterns of hyper-inflammation have been seen in patients undergoing cardiopulmonary bypass (CPB) and in patients treated with extracorporeal membrane oxygenation (ECMO). Typical treatments used for the prevention and treatment of CPB/ECMO-induced hyper-inflammation have not been shown to be substantially effective. Two patients suffering from ECMO-related CSS were identified by their severe hyper-inflammatory profile and life-threatening sequelae of vasodilatory shock and respiratory failure. Anakinra, an interleukin-1 receptor antagonist, was employed as specific cytokine-directed therapy for the treatment of CSS in these two patients to good effect, with significant improvement in hyper-inflammation and cardiorespiratory status. The use of cytokine-directed therapies in CPB/ECMO-related CSS has great potential to improve the treatment and outcomes of this serious condition.
Collapse
Affiliation(s)
- Daniel D Reiff
- Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Randy Q Cron
- Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
56
|
De Benedetti F, Grom AA, Brogan PA, Bracaglia C, Pardeo M, Marucci G, Eleftheriou D, Papadopoulou C, Schulert GS, Quartier P, Antón J, Laveille C, Frederiksen R, Asnaghi V, Ballabio M, Jacqmin P, de Min C. Efficacy and safety of emapalumab in macrophage activation syndrome. Ann Rheum Dis 2023; 82:857-865. [PMID: 37001971 PMCID: PMC10314091 DOI: 10.1136/ard-2022-223739] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/02/2023] [Indexed: 04/03/2023]
Abstract
OBJECTIVES Macrophage activation syndrome (MAS) is a severe, life-threatening complication of systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD). The objective of this study was to confirm the adequacy of an emapalumab dosing regimen in relation to interferon-γ (IFNγ) activity by assessing efficacy and safety. The efficacy outcome was MAS remission by week 8, based on clinical and laboratory criteria. METHODS We studied emapalumab, a human anti-IFNγ antibody, administered with background glucocorticoids, in a prospective single-arm trial involving patients who had MAS secondary to sJIA or AOSD and had previously failed high-dose glucocorticoids, with or without anakinra and/or ciclosporin. The study foresaw 4-week treatment that could be shortened or prolonged based on investigator's assessment of response. Patients entered a long-term (12 months) follow-up study. RESULTS Fourteen patients received emapalumab. All patients completed the trial, entered the long-term follow-up and were alive at the end of follow-up. The investigated dosing regimen, based on an initial loading dose followed by maintenance doses, was appropriate, as shown by rapid neutralisation of IFNγ activity, demonstrated by a prompt decrease in serum C-X-C motif chemokine ligand 9 (CXCL9) levels. By week 8, MAS remission was achieved in 13 of the 14 patients at a median time of 25 days. Viral infections and positive viral tests were observed. CONCLUSIONS Neutralisation of IFNγ with emapalumab was efficacious in inducing remission of MAS secondary to sJIA or AOSD in patients who had failed high-dose glucocorticoids. Screening for viral infections should be performed, particularly for cytomegalovirus. TRIAL REGISTRATION NUMBER NCT02069899 and NCT03311854.
Collapse
Affiliation(s)
| | - Alexei A Grom
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul A Brogan
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Claudia Bracaglia
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Manuela Pardeo
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giulia Marucci
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Despina Eleftheriou
- University College London Great Ormond Street Institute of Child Health, London, UK
| | | | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pierre Quartier
- Pediatric Immuno-Hematology and Rheumatology Unit, RAISE Rare Disease Reference Centre, Hopital Universitaire Necker-Enfants Malades, Assistance Publique-Hopitaux de Paris, Paris, France
- Université Paris-Cité, Paris, France
| | - Jordi Antón
- Pediatric Rheumatology, Hospital Sant Joan de Deu, Barcelona, Spain
- Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
57
|
Saunders BN, Kuijpers MV, Sloan JJ, Gertner E. Continuous IV infusion of anakinra. Front Pharmacol 2023; 14:1162742. [PMID: 37229265 PMCID: PMC10203171 DOI: 10.3389/fphar.2023.1162742] [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: 02/09/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Objective: A review of the use of continuous IV infusion of anakinra; a description of the protocol for continuous IV infusion of anakinra in the treatment of cytokine storm developed over the past 4 years at a tertiary level academic medical center in the United States. Methods: We reviewed published reports of continuous IV infusion of anakinra in cytokine storm and summarize this method of treatment in other diseases. As well, over the past 4 years, continuous IV infusions of anakinra were administered at our tertiary level academic medical center in the United States (Regions Hospital, St. Paul, Minnesota) for approximately 400 patient days of treatment primarily for the cytokine storm associated with macrophage activation syndrome (MAS) in adults. This updated protocol is presented. While this a single center protocol, it may serve as an initial guide for further refinement of protocols in MAS and other conditions. Conclusion:Continuous IV infusion of anakinra has advantages over subcutaneous infusions and may be important in controlling severe life-threatening cytokine storm as seen in macrophage activation syndrome. This has the potential to be an important therapy for other syndromes including Cytokine Release Syndrome related to CAR T-cell therapy. Close collaboration between Rheumatology, Pharmacy and Nursing allows this treatment to be delivered rapidly and efficiently.
Collapse
|
58
|
Lee PY, Cron RQ. The Multifaceted Immunology of Cytokine Storm Syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1015-1024. [PMID: 37011407 PMCID: PMC10071410 DOI: 10.4049/jimmunol.2200808] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 04/05/2023]
Abstract
Cytokine storm syndromes (CSSs) are potentially fatal hyperinflammatory states that share the underpinnings of persistent immune cell activation and uninhibited cytokine production. CSSs can be genetically determined by inborn errors of immunity (i.e., familial hemophagocytic lymphohistiocytosis) or develop as a complication of infections, chronic inflammatory diseases (e.g., Still disease), or malignancies (e.g., T cell lymphoma). Therapeutic interventions that activate the immune system such as chimeric Ag receptor T cell therapy and immune checkpoint inhibition can also trigger CSSs in the setting of cancer treatment. In this review, the biology of different types of CSSs is explored, and the current knowledge on the involvement of immune pathways and the contribution of host genetics is discussed. The use of animal models to study CSSs is reviewed, and their relevance for human diseases is discussed. Lastly, treatment approaches for CSSs are discussed with a focus on interventions that target immune cells and cytokines.
Collapse
Affiliation(s)
- Pui Y. Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Randy Q. Cron
- Division of Pediatric Rheumatology, Children’s of Alabama, University of Alabama Heersink School of Medicine, Birmingham, AL
- Department of Pediatrics, University of Alabama Heersink School of Medicine, Birmingham, AL
| |
Collapse
|
59
|
Lee JC, Logan AC. Diagnosis and Management of Adult Malignancy-Associated Hemophagocytic Lymphohistiocytosis. Cancers (Basel) 2023; 15:1839. [PMID: 36980725 PMCID: PMC10046521 DOI: 10.3390/cancers15061839] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a syndrome of severe, dysregulated inflammation driven by the inability of T cells to clear an antigenic target. When associated with malignancy (mHLH), the HLH syndrome is typically associated with extremely poor survival. Here, we review the diagnosis of secondary HLH (sHLH) syndromes in adults, with emphasis on the appropriate workup and treatment of mHLH. At present, the management of HLH in adults, including most forms of mHLH, is based on the use of corticosteroids and etoposide following the HLH-94 regimen. In some cases, this therapeutic approach may be cohesively incorporated into malignancy-directed therapy, while in other cases, the decision about whether to treat HLH prior to initiating other therapies may be more complicated. Recent studies exploring the efficacy of other agents in HLH, in particular ruxolitinib, offer hope for better outcomes in the management of mHLH. Considerations for the management of lymphoma-associated mHLH, as well as other forms of mHLH and immunotherapy treatment-related HLH, are discussed.
Collapse
Affiliation(s)
- Jerry C. Lee
- Hematology, Blood and Marrow Transplantation, and Cellular Therapy Program, Division of Hematology/Oncology, University of California, San Francisco, CA 94143, USA;
| | | |
Collapse
|
60
|
sCD25 as an independent adverse prognostic factor in adult patients with HLH: results of a multicenter retrospective study. Blood Adv 2023; 7:832-844. [PMID: 35973195 PMCID: PMC9986715 DOI: 10.1182/bloodadvances.2022007953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/11/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rare but often fatal hyperinflammatory syndrome caused by an inborn or acquired error of immunity. In adults, the underlying immunodeficiency generally arises alongside severe infections, malignancies, autoimmune diseases, and immunosuppressive treatment. To analyze risk factors and outcome in adults, we conducted a multicenter retrospective study. A total of 62 adult (age ≥18 years) patients met at least one of the following inclusion criteria: (1) ≥5 of 8 HLH-2004 criteria, (2) HScore ≥ 200 plus 4 HLH-2004 criteria, or (3) mutation compatible with an HLH diagnosis. Most patients (65%) were male, and the median age at diagnosis was 53.5 years (range, 19-81 years). All patients were assigned to 4 etiologic subgroups based on their most likely HLH trigger. The survival probability of the 4 etiologic subgroups differed significantly (P = .004, log-rank test), with patients with an underlying malignancy having the worst clinical outcome (1-year survival probability of 21%). The parameters older age, malignant trigger, elevated serum levels of aspartate transferase, creatinine, international normalized ratio, lactate dehydrogenase, sCD25, and a low albumin level and platelet count at treatment initiation were significantly (P < .1) associated with worse overall survival in the univariate Cox regression model. In multivariate analysis, sCD25 remained the only significant prognostic factor (P = .005). Our results suggest that sCD25 could be a useful marker for the prognosis of patients with HLH that might help to stratify therapeutic interventions.
Collapse
|
61
|
Bindoli S, Galozzi P, Doria A, Sfriso P. Intravenous anakinra to curb cytokine storm in adult-onset Still's disease and in macrophage activation syndrome: A case series. Joint Bone Spine 2023; 90:105524. [PMID: 36623798 DOI: 10.1016/j.jbspin.2023.105524] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Adult-onset Still's disease (AOSD) is an auto-inflammatory polygenic disorder, for which the diagnosis is essentially clinical. The exclusion of mimickers [such as common bacterial and viral infections, hematologic malignancies, and, more recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] is necessary to confirm the diagnosis. Anti-interleukin (IL)-1 therapy is considered a treatment milestone for AOSD. Herein, we present a short series of newly-diagnosed AOSD or upcoming macrophage activation syndrome (MAS) cases who received intravenous (IV) anakinra, an IL-1 receptor blocker. METHODS Four patients with newly-diagnosed AOSD or upcoming MAS were treated with IV anakinra at the Rheumatology Unit of Padova University Hospital, Italy. We obtained informed consent from the patients for use of their cases and medical images for publication purposes. RESULTS All patients presented with AOSD or MAS during the COVID-19 pandemic, making diagnosis challenging due to similar immunological and clinical characteristics across both pathologies. All patients presented with hyperpyrexia and elevated inflammatory markers; two patients had a skin rash typically seen in AOSD. IV anakinra slowed down AOSD progression in all patients, prevented severe outcomes and mitigated the risk of multiorgan failure. All cases improved within 24hours of anakinra administration. CONCLUSION We found that administration of anakinra in patients with newly-diagnosed AOSD and/or upcoming MAS reduced hyperinflammation and prevented life-threatening complications. The IV route appears to be preferable in the hospital setting, where comorbidities such as coagulopathies and thrombocytopenia can complicate the use of other routes of administration.
Collapse
Affiliation(s)
- Sara Bindoli
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Paola Galozzi
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Paolo Sfriso
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, via Giustiniani 2, 35128 Padova, Italy.
| |
Collapse
|
62
|
Hughes SA, Lin M, Weir A, Huang B, Xiong L, Chua NK, Pang J, Santavanond JP, Tixeira R, Doerflinger M, Deng Y, Yu C, Silke N, Conos SA, Frank D, Simpson DS, Murphy JM, Lawlor KE, Pearson JS, Silke J, Pellegrini M, Herold MJ, Poon IKH, Masters SL, Li M, Tang Q, Zhang Y, Rashidi M, Geng L, Vince JE. Caspase-8-driven apoptotic and pyroptotic crosstalk causes cell death and IL-1β release in X-linked inhibitor of apoptosis (XIAP) deficiency. EMBO J 2023; 42:e110468. [PMID: 36647737 PMCID: PMC9975961 DOI: 10.15252/embj.2021110468] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
Genetic lesions in X-linked inhibitor of apoptosis (XIAP) pre-dispose humans to cell death-associated inflammatory diseases, although the underlying mechanisms remain unclear. Here, we report that two patients with XIAP deficiency-associated inflammatory bowel disease display increased inflammatory IL-1β maturation as well as cell death-associated caspase-8 and Gasdermin D (GSDMD) processing in diseased tissue, which is reduced upon patient treatment. Loss of XIAP leads to caspase-8-driven cell death and bioactive IL-1β release that is only abrogated by combined deletion of the apoptotic and pyroptotic cell death machinery. Namely, extrinsic apoptotic caspase-8 promotes pyroptotic GSDMD processing that kills macrophages lacking both inflammasome and apoptosis signalling components (caspase-1, -3, -7, -11 and BID), while caspase-8 can still cause cell death in the absence of both GSDMD and GSDME when caspase-3 and caspase-7 are present. Neither caspase-3 and caspase-7-mediated activation of the pannexin-1 channel, or GSDMD loss, prevented NLRP3 inflammasome assembly and consequent caspase-1 and IL-1β maturation downstream of XIAP inhibition and caspase-8 activation, even though the pannexin-1 channel was required for NLRP3 triggering upon mitochondrial apoptosis. These findings uncouple the mechanisms of cell death and NLRP3 activation resulting from extrinsic and intrinsic apoptosis signalling, reveal how XIAP loss can co-opt dual cell death programs, and uncover strategies for targeting the cell death and inflammatory pathways that result from XIAP deficiency.
Collapse
|
63
|
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: 5] [Impact Index Per Article: 2.5] [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
|
64
|
Lee J, Pham B, Karanjawala ZE, Adesina O. Postpartum fevers, a rare presentation of secondary hemophagocytic lymphohistiocytosis. Clin Case Rep 2023; 11:e7070. [PMID: 36941837 PMCID: PMC10023518 DOI: 10.1002/ccr3.7070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 01/05/2023] [Accepted: 02/13/2023] [Indexed: 03/20/2023] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rare disease of excessive immune system activation. We report a case of HLH in a 20-year-old primigravid woman who presented with postpartum fevers. She was successfully treated with dexamethasone and anakinra, a deviation from the HLH-94 protocol, to preserve her ability to breastfeed.
Collapse
Affiliation(s)
- Jacqueline Lee
- Department of Internal MedicineUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Brian Pham
- Division of Hematology and OncologyUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Zarir E. Karanjawala
- Division of HematopathologyUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Oyebimpe Adesina
- Division of Hematology and OncologyUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| |
Collapse
|
65
|
Yang L, Lowry S, Heath T. Use of Intravenous Anakinra for Management of Pediatric Cytokine Storm Syndromes at an Academic Medical Center. Hosp Pharm 2023. [DOI: 10.1177/00185787221142470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Background: Off-label intravenous (IV) route of anakinra is increasingly recognized to enable higher and faster maximal plasma concentrations than subcutaneous route for treatment of cytokine storm syndromes. Objective: To describe off-label indications of IV anakinra, corresponding dosing and safety profiles, particularly during the coronavirus disease 19 (COVID-19) pandemic. Methods: A retrospective, single-cohort study was conducted at an academic medical center to evaluate use of IV anakinra in hospitalized pediatric patients (age ≤21 years). Institutional Review Board review was considered exempt. The primary endpoint was the primary indication(s) for IV anakinra. The key secondary endpoints were dosing of IV anakinra, previous immunomodulatory therapies, and adverse events. Results: Of 14 pediatric patients, 8 (57.1%) received IV anakinra for treatment of multisystem inflammatory syndrome in children (MIS-C) associated with COVID-19, whereas 3 and 2 were treated for hemophagocytic lymphohistiocytosis (HLH) and flares of systemic onset juvenile idiopathic arthritis (SoJIA), respectively. The initial dosing regimen of IV anakinra for MIS-C associated with COVID-19 was a median dose of 2.25 mg/kg/dose with a median dosing interval of 12 hours for a median initial treatment duration of 3.5 days. Eleven (78.6%) patients received previous immunomodulatory therapies (IV immune globulin [n = 10; 71.4%] and steroids [n = 9; 64.3%]). No adverse drug events were documented. Conclusion: IV anakinra was used off-label for treatment of MIS-C associated with COVID-19, HLH and SoJIA flares in critically ill patients with no adverse drug events documented. This study helped ascertain the off-label indications of IV anakinra and corresponding patient characteristics.
Collapse
|
66
|
Della Casa F, Petraroli A, Mormile I, Lagnese G, Di Salvatore A, Rossi FW, de Paulis A. Adult-onset macrophage activation syndrome treated by interleukin-1 inhibition. Rheumatol Adv Pract 2023; 7:rkad014. [PMID: 36789245 PMCID: PMC9912699 DOI: 10.1093/rap/rkad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2023] [Indexed: 02/05/2023] Open
Affiliation(s)
- Francesca Della Casa
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Angelica Petraroli
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Center for Basic and Clinical Immunology Research, WAO Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Ilaria Mormile
- Correspondence to: Ilaria Mormile, Department of Translational Medical Sciences, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy. E-mail:
| | - Gianluca Lagnese
- Post-Graduate Program in Clinical Immunology and Allergy, University of Naples Federico II, Naples, Italy
| | - Antonio Di Salvatore
- Post-Graduate Program in Clinical Immunology and Allergy, University of Naples Federico II, Naples, Italy
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Center for Basic and Clinical Immunology Research, WAO Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Center for Basic and Clinical Immunology Research, WAO Center of Excellence, University of Naples Federico II, Naples, Italy
| |
Collapse
|
67
|
Bektaş M, Yüce S, Ay M, Uyar MH, Önder ME, Kılıç Mİ. High-dose intravenous anakinra treatment is safe and effective in severe and critical COVID-19 patients: a propensity score-matched study in a single center. Inflammopharmacology 2023; 31:787-797. [PMID: 36707494 PMCID: PMC9882740 DOI: 10.1007/s10787-023-01138-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND In COVID-19, severe disease course such as need of intensive care unit (ICU) as well as development of mortality is mainly due to cytokine storm. In this study, we aimed to evaluate the high-dose intravenous anakinra treatment response and outcome in patients with severe and critically ill COVID-19 compared to standard of care. METHODS This retrospective observational study was carried out at a tertiary referral center. The study population consisted of two groups as follows: the patients receiving high-dose intravenous anakinra (anakinra group) between 01.09.2021 and 01.02.2022 and the patients treated with standard of care (SoC, control group) as historical control group who were hospitalized between 01.07.2021 and 01.09.2021. RESULTS After the propensity score 1:1 matching, 79 patients in anakinra and 79 patients in SoC matched and were included into the analysis. Mean ± SD patient age was 67.4 ± 16.7 and 67.1 ± 16.3 years in anakinra and SoC groups, respectively (p = 0.9). Male gender was 38 (48.7%) in anakinra and 36 (46.2%) in SoC (p = 0.8). Overall, ICU admission was in 14.1% (n = 11) and 30.8% (n = 24) (p = 0.013; OR 6.2), intubation in 12.8% (n = 10) and 16.7% (n = 13) patients (p = 0.5), and 14.1% (n = 11) and 32.1% (n = 25) patients died in anakinra and control groups, respectively (p = 0.008; OR 7.1). CONCLUSION In our study, mortality was lower in patients receiving anakinra compared to SoC. Intravenous high-dose anakinra is safe and effective treatment in patients with severe and critical COVID-19.
Collapse
Affiliation(s)
- Murat Bektaş
- Division of Rheumatology, Department of Internal Medicine, Aksaray Training and Research Hospital, Yeni Sanayi Mahallesi, 68200 Merkez/Aksaray, Turkey
| | - Servet Yüce
- Department of Public Health and Biostatistics, Istanbul Faculty of Medicine, Çapa, Şehremini, Istanbul, Turkey
| | - Mustafa Ay
- Department of Emergency Medicine, Aksaray Training and Research Hospital, 68200 Merkez/Aksaray, Turkey
| | - Muhammed Hamdi Uyar
- Department of Emergency Medicine, Aksaray Training and Research Hospital, 68200 Merkez/Aksaray, Turkey
| | - Mustafa Erkut Önder
- Division of Rheumatology, Department of Physical Therapy and Rehabilitation, Aksaray Training and Research Hospital, Yeni Sanayi Mahallesi, 68200 Merkez/Aksaray, Turkey
| | - Muhammed İkbal Kılıç
- Department of Internal Medicine, Aksaray Training and Research Hospital, Yeni Sanayi Mahallesi, 68200 Merkez/Aksaray, Turkey
| |
Collapse
|
68
|
Elmekaty EZI, Maklad A, Abouelhassan R, Munir W, Ibrahim MIM, Nair A, Alibrahim R, Iqbal F, Al Bishawi A, Abdelmajid A, Aboukamar M, Hadi HA, Khattab MA, Al Soub H, Al Maslamani M. Evaluation of anakinra in the management of patients with COVID-19 infection: A randomized clinical trial. Front Microbiol 2023; 14:1098703. [PMID: 36778864 PMCID: PMC9910697 DOI: 10.3389/fmicb.2023.1098703] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Background The global COVID-19 pandemic led to substantial clinical and economic outcomes with catastrophic consequences. While the majority of cases has mild to moderate disease, minority of patients progress into severe disease secondary to the stimulation of the immune response. The hyperinflammatory state contributes towards progression into multi-organ failure which necessitates suppressive therapy with variable outcomes. This study aims to explore the safety and efficacy of anakinra in COVID-19 patients with severe disease leading to cytokine release syndromes. Methods In this open-label, multi-center, randomized clinical trial, patients with confirmed COVID-19 infection with evidence of respiratory distress and signs of cytokine release syndrome were randomized in 1:1 ratio to receive either standard of care (SOC) or anakinra (100 mg subcutaneously every 12 h for 3 days then 100 mg subcutaneously once daily for 4 days) in addition to SOC. The primary outcome was treatment success at day 14 as defined by the WHO clinical progression score of ≤3. Primary analysis was based upon intention-to-treat population, with value of p of <0.05. Results Out 327 patients screened for eligibility, 80 patients were recruited for the study. The mean age was 49.9 years (SD = 11.7), with male predominance at 82.5% (n = 66). The primary outcome was not statistically different (87.5% (n = 35) in anakinra group vs. 92.5% (n = 37) in SOC group, p = 0.712; OR = 1.762 (95%CI: 0.39-7.93). The majority of reported adverse events were mild in severity and not related to the study treatment. Elevated aspartate aminotransferase was the only significant adverse event which was not associated with discontinuation of therapy. Conclusion In patients with severe COVID-19 infection, the addition of anakinra to SOC treatment was safe but was not associated with significant improvement according to the WHO clinical progression scale. Further studies are warranted to explore patients' subgroups characteristics that might benefit from administered therapy. Clinical Trial Registration Trial registration at ClinicalTrials.gov, identifier: NCT04643678.
Collapse
Affiliation(s)
- Eman Zeyad I. Elmekaty
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar,*Correspondence: Eman Zeyad I. Elmekaty,
| | - Aya Maklad
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | | | - Waqar Munir
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Arun Nair
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Rim Alibrahim
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Iqbal
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Ahmad Al Bishawi
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Mohamed Aboukamar
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Hamad Abdel Hadi
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Hussam Al Soub
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Muna Al Maslamani
- Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
69
|
Mayne ES, George JA, Louw S. Assessing Biomarkers in Viral Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:159-173. [PMID: 37378766 DOI: 10.1007/978-3-031-28012-2_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Current biomarkers to assess the risk of complications of both acute and chronic viral infection are suboptimal. Prevalent viral infections like human immunodeficiency virus (HIV), hepatitis B and C virus, herpes viruses, and, more recently, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) may be associated with significant sequelae including the risk of cardiovascular disease, other end-organ diseases, and malignancies. This review considers some biomarkers which have been investigated in diagnosis and prognosis of key viral infections including inflammatory cytokines, markers of endothelial dysfunction and activation and coagulation, and the role that more conventional diagnostic markers, such as C-reactive protein and procalcitonin, can play in predicting these secondary complications, as markers of severity and to distinguish viral and bacterial infection. Although many of these are still only available in the research setting, these markers show promise for incorporation in diagnostic algorithms which may assist to predict adverse outcomes and to guide therapy.
Collapse
Affiliation(s)
- Elizabeth S Mayne
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town and National Health Laboratory Service, Cape Town, South Africa.
| | - Jaya A George
- National Health Laboratory Service and Wits Diagnostic Innovation Hub, University of Witwatersrand, Johannesburg, South Africa
| | - Susan Louw
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
70
|
Amikishiyev S, Gunver MG, Bektas M, Aghamuradov S, Ince B, Koca N, Torun ES, Aliyeva N, Sari S, Cetin C, Yalcin-Dulundu BC, Deniz R, Kemik F, Agargun BF, Gulseren UA, Besisik B, Alkan O, Bagriacik C, Tor YB, Senkal N, Catma Y, Durak G, Mese S, Agacfidan A, Kose M, Erelel M, Cagatay AA, Simsek-Yavuz S, Kalayoglu-Besisik S, Esen F, Gül A. Criteria for Hyperinflammation Developing in COVID-19: Analysis of 2 Cohorts From Different Periods of the Pandemic. Arthritis Rheumatol 2022; 75:664-672. [PMID: 36508470 PMCID: PMC9878117 DOI: 10.1002/art.42417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/15/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Hyperinflammation (HI) that develops in week 2 of COVID-19 contributes to a worse outcome. Because week 2 laboratory findings can be relatively mild, the available criteria for classification of hemophagocytic lymphohistiocytosis or macrophage activation syndrome are not helpful. METHODS Our study included a discovery cohort of patients from Turkey with symptomatic COVID-19 who were followed up while hospitalized during the initial wave and a replication cohort of hospitalized patients from a later period, all of whom required oxygen support and received glucocorticoids. Diagnosis of HI was made by an expert panel; most patients with COVID-19-associated HI (HIC) received tocilizumab or anakinra. Clinical and laboratory data from start day of treatment with tocilizumab or anakinra in HIC patients were compared with the data from day 5-6 in patients without HIC. Values maximizing the sensitivity and specificity of each parameter were calculated to determine criteria items. RESULTS The discovery cohort included 685 patients, and the replication cohort included 156 patients, with 150 and 61 patients receiving treatment for HI, respectively. Mortality rate in HI patients in the discovery cohort (23.3%) was higher than the rate in patients without HI (3.7%) and the rate in patients in the overall replication cohort (10.3%). The 12-item criteria that we developed for HIC showed that a score of 35 provided 85.3% sensitivity and 81.7% specificity for identification of HIC. In the replication cohort, the same criteria resulted in 90.0% sensitivity for HIC; however, lower specificity values were observed because of the inclusion of milder cases of HIC responding only to glucocorticoids. CONCLUSION The use of the 12-item criteria for HIC can better define patients with HIC with reasonable sensitivity and specificity and enables an earlier treatment start.
Collapse
Affiliation(s)
- Shirkhan Amikishiyev
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - M Guven Gunver
- Department of Medical Statistics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Murat Bektas
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sarvan Aghamuradov
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Burak Ince
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nevzat Koca
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ege Sinan Torun
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Numune Aliyeva
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Selma Sari
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cigdem Cetin
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Banu C Yalcin-Dulundu
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Rabia Deniz
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Fatih Kemik
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Besim Fazil Agargun
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ubeyde Ayse Gulseren
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Beliz Besisik
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Onur Alkan
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ceren Bagriacik
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yavuz B Tor
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Naci Senkal
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yunus Catma
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Gorkem Durak
- Department of Radiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sevim Mese
- Division of Virology and Fundamental Immunology, Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ali Agacfidan
- Division of Virology and Fundamental Immunology, Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Murat Kose
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mustafa Erelel
- Department of Chest Diseases, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - A Atahan Cagatay
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Serap Simsek-Yavuz
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sevgi Kalayoglu-Besisik
- Division of Hematology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Figen Esen
- Department of Anesthesiology and Reanimation, Intensive Care Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ahmet Gül
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
71
|
Tang JP, Peters CW, Quiros C, Wang X, Klomhaus AM, Yamada RE, Timmerman JM, Moore TB, Nowicki TS. Hypophosphatemia Due to Increased Effector Cell Metabolic Activity Is Associated with Neurotoxicity Symptoms in CD19-Targeted CAR T-cell Therapy. Cancer Immunol Res 2022; 10:1433-1440. [PMID: 36259217 PMCID: PMC9722515 DOI: 10.1158/2326-6066.cir-22-0418] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/27/2022] [Accepted: 09/15/2022] [Indexed: 01/10/2023]
Abstract
A major complication of chimeric antigen receptor (CAR) T-cell therapy is immune effector cell-associated neurotoxicity syndrome (ICANS), which presents as aphasia, confusion, weakness, somnolence, seizures, and coma. This is similar to the neurologic manifestations of hypophosphatemia, which can result from sudden increases in metabolic demand for phosphorylated intermediates (e.g., refeeding syndrome and sepsis). Given these similarities, we investigated whether CAR T-cell effector metabolic activity is associated with increased extracellular phosphate consumption and a possible association between hypophosphatemia and ICANS. In vitro 4-1BB and CD28 CD19-targeted CAR T-cell effector activity was found to be associated with increased consumption of media phosphorus, which was temporally associated with increased single-cell effector secretomic activity and increased phosphorus-dependent metabolic demand of the CAR T cells. A clinical cohort of 77 patients treated with CD19-targeted CAR T-cell therapy demonstrated a significant anticorrelation between serum phosphorus and ICANS incidence and severity, with earlier onset of hypophosphatemia after CAR T-cell infusion more likely to result in neurotoxicity. These results imply phosphorous level monitoring could alert to the development of ICANS in clinical scenarios. See related Spotlight by Tobin et al., p. 1422.
Collapse
Affiliation(s)
- Jack Pengfei Tang
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Cole W. Peters
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Crystal Quiros
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, California
| | - Alexandra M. Klomhaus
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, California
| | - Reiko E. Yamada
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, California
| | - John M. Timmerman
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California
| | - Theodore B. Moore
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Theodore S. Nowicki
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California
| |
Collapse
|
72
|
Summerlin J, Wells DA, Anderson MK, Halford Z. A Review of Current and Emerging Therapeutic Options for Hemophagocytic Lymphohistiocytosis. Ann Pharmacother 2022:10600280221134719. [DOI: 10.1177/10600280221134719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Objective: To provide an overview of clinical sequelae and emerging treatment options for hemophagocytic lymphohistiocytosis (HLH). Data Sources: A literature search was conducted using the search terms “hemophagocytic lymphohistiocytosis,” “hemophagocytic syndrome,” “macrophage activation syndrome,” and “treatment” on Ovid and PubMed from January 1, 2017, through September 28, 2022. Study Selection and Data Extraction: Relevant clinical trials, meta-analyses, case reports, review articles, package inserts, and guidelines to identify current and emerging therapeutic options for the management of HLH. Data Synthesis: Genetic disorders and secondary causes may trigger HLH in both children and adults. Notable improvements in the diagnosis of HLH were seen with implementation of the HLH-2004 standard diagnostic criteria; however, timely and accurate identification of HLH remain significant barriers to optimal management. Multiagent immunochemotherapy are the backbone of aggressive therapy for acutely ill patients with HLH. Relevance to Patient Care and Clinical Practice: The global coronavirus 2019 (COVID-19) pandemic and emerging immune effector cell therapies have served to highlight the concerns with immune dysregulation and subsequent HLH precipitation. Without prompt identification and treatment, HLH can be fatal. Historically, the clinician’s armamentarium for managing HLH was sparse, with etoposide-based protocols serving as the standard of care. Relapsed or refractory disease portends a poor prognosis and requires additional treatment options. Second- or subsequent-line options now include hematopoietic stem cell transplantation, emapalumab, alemtuzumab, anakinra, ruxolitinib, and tocilizumab. Conclusions: Improvements in diagnostic methods and novel immunosuppressive treatment strategies, including noncytotoxic immunochemotherapy, have transformed the therapeutic landscape. Unfortunately, many unanswered questions remain. Additional studies are required to optimize dosing, schedules, treatment sequences, and indications for novel treatment options.
Collapse
Affiliation(s)
- Jenna Summerlin
- Division of Pharmacy Practice, The University of Texas at Austin College of Pharmacy, Austin, TX, USA
| | - Drew A. Wells
- Internal Medicine, Department of Pharmacy, Methodist University Hospital, Memphis, TN, USA
| | | | | |
Collapse
|
73
|
Nazerian Y, Ghasemi M, Yassaghi Y, Nazerian A, Mahmoud Hashemi S. Role of SARS-CoV-2-induced Cytokine Storm in Multi-Organ Failure: Molecular Pathways and Potential Therapeutic Options. Int Immunopharmacol 2022; 113:109428. [PMID: 36379152 PMCID: PMC9637536 DOI: 10.1016/j.intimp.2022.109428] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
Coronavirus disease 2019 (COVID-19) outbreak has become a global public health emergency and has led to devastating results. Mounting evidence proposes that the disease causes severe pulmonary involvement and influences different organs, leading to a critical situation named multi-organ failure. It is yet to be fully clarified how the disease becomes so deadly in some patients. However, it is proven that a condition called “cytokine storm” is involved in the deterioration of COVID-19. Although beneficial, sustained production of cytokines and overabundance of inflammatory mediators causing cytokine storm can lead to collateral vital organ damages. Furthermore, cytokine storm can cause post-COVID-19 syndrome (PCS), an important cause of morbidity after the acute phase of COVID-19. Herein, we aim to explain the possible pathophysiology mechanisms involved in COVID-19-related cytokine storm and its association with multi-organ failure and PCS. We also discuss the latest advances in finding the potential therapeutic targets to control cytokine storm wishing to answer unmet clinical demands for treatment of COVID-19.
Collapse
Affiliation(s)
- Yasaman Nazerian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Ghasemi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Younes Yassaghi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Seyed Mahmoud Hashemi
- Medical nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding author at: Medical nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran / Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
74
|
Gál P, Brábek J, Holub M, Jakubek M, Šedo A, Lacina L, Strnadová K, Dubový P, Hornychová H, Ryška A, Smetana K. Autoimmunity, cancer and COVID-19 abnormally activate wound healing pathways: critical role of inflammation. Histochem Cell Biol 2022; 158:415-434. [PMID: 35867145 PMCID: PMC9305064 DOI: 10.1007/s00418-022-02140-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 11/30/2022]
Abstract
Recent evidence indicates that targeting IL-6 provides broad therapeutic approaches to several diseases. In patients with cancer, autoimmune diseases, severe respiratory infections [e.g. coronavirus disease 2019 (COVID-19)] and wound healing, IL-6 plays a critical role in modulating the systemic and local microenvironment. Elevated serum levels of IL-6 interfere with the systemic immune response and are associated with disease progression and prognosis. As already noted, monoclonal antibodies blocking either IL-6 or binding of IL-6 to receptors have been used/tested successfully in the treatment of rheumatoid arthritis, many cancer types, and COVID-19. Therefore, in the present review, we compare the impact of IL-6 and anti-IL-6 therapy to demonstrate common (pathological) features of the studied diseases such as formation of granulation tissue with the presence of myofibroblasts and deposition of new extracellular matrix. We also discuss abnormal activation of other wound-healing-related pathways that have been implicated in autoimmune disorders, cancer or COVID-19.
Collapse
Affiliation(s)
- Peter Gál
- Department of Pharmacology, Pavol Jozef Šafárik University, Košice, Slovak Republic
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic
- Prague Burn Centre, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, 160 00 Prague, Czech Republic
| | - Milan Jakubek
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Aleksi Šedo
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 120 00 Praha 2, Czech Republic
| | - Lukáš Lacina
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Karolína Strnadová
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Petr Dubový
- Institute of Anatomy, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Helena Hornychová
- The Fingerland Department of Pathology, Faculty of Medicine Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Aleš Ryška
- The Fingerland Department of Pathology, Faculty of Medicine Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| |
Collapse
|
75
|
Variation of Diagnostic Approaches and Treatment Practices for Hemophagocytic Lymphohistiocytosis/Macrophage Activation Syndrome Among Pediatric Subspecialists. J Pediatr 2022; 255:65-71.e6. [PMID: 36328191 DOI: 10.1016/j.jpeds.2022.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/17/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To assess the diagnostic and treatment practices among a variety of subspecialists at pediatric institutions in the US. STUDY DESIGN Using a web-based survey, we assessed the consultation, diagnostic, and treatment preferences of providers from the different pediatric subspecialties who care for pediatric patients with hemophagocytic lymphohistiocytosis (HLH)/macrophage activating syndrome (MAS). Domains included demographics, provider training level and specialty, experience and comfort level with the diagnosis and treatment of HLH/MAS, and institutional approaches toward the diagnosis and management of HLH/MAS. Participants also were given 2 case scenarios: one describing Epstein-Barr virus-associated HLH and another describing an underlying rheumatologic condition with MAS. RESULTS Of 263 respondents, 23%, 29%, 39%, and 7% identified as hematology/oncology, rheumatology, general pediatrics/critical care/hospitalist, and allergy/immunology, respectively. For Epstein-Barr virus/HLH, hematology/oncology was the preferred first consultant by most respondents other than rheumatologists, of whom only 47% agreed. For MAS, 92% of respondents from all specialties favored a rheumatology consultation. Preferred diagnostic tests varied by subspecialty, with hematology/oncology more likely than rheumatology to order an infectious workup, natural killer cell function, soluble interleukin-2 receptor, bone marrow biopsy, and genetic testing. First-line therapy also varied, with hematology/oncology preferring dexamethasone and etoposide and rheumatology more often preferring methylprednisolone and anakinra. One-half of respondents were unaware of institutional algorithms for diagnosis and treatment of HLH/MAS. Most (85.6%) favored the development of treatment algorithms for HLH/MAS, and 90% supported a multidisciplinary approach. CONCLUSIONS Current consulting patterns, diagnostic workup, and treatment approaches of HLH/MAS vary by specialty, highlighting the need for standardized management algorithms and institutional multidisciplinary HLH/MAS teams.
Collapse
|
76
|
Green EA, Metz D, Galinsky R, Atkinson R, Skuza EM, Clark M, Gunn AJ, Kirkpatrick CM, Hunt RW, Berger PJ, Nold-Petry CA, Nold MF. Anakinra Pilot - a clinical trial to demonstrate safety, feasibility and pharmacokinetics of interleukin 1 receptor antagonist in preterm infants. Front Immunol 2022; 13:1022104. [PMID: 36389766 PMCID: PMC9647081 DOI: 10.3389/fimmu.2022.1022104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/10/2022] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD), its complication pulmonary hypertension (BPD-PH) and preterm brain and gut injury lead to significant morbidity and mortality in infants born extremely prematurely. There is extensive evidence that the pro-inflammatory cytokine interleukin 1 (IL-1) plays a key role in the pathophysiology of these illnesses. Two decades of clinical use in paediatric and adult medicine have established an excellent safety and efficacy record for IL-1 blockade with IL-1 receptor antagonist (IL-1Ra, medication name anakinra). Building on robust pre-clinical evidence, the Anakinra Pilot trial aims to demonstrate safety and feasibility of administering anakinra to preterm infants, and to establish pharmacokinetics in this population. Its ultimate goal is to facilitate large studies that will test whether anakinra can ameliorate early-life inflammation, thus alleviating multiple complications of prematurity. METHODS AND ANALYSIS Anakinra Pilot is an investigator-initiated, single arm, safety and feasibility dose-escalation trial in extremely preterm infants born between 24 weeks 0 days (240) and 276 weeks of gestational age (GA). Enrolled infants will receive anakinra intravenously over the first 21 days after birth, starting in the first 24 h after birth. In the first phase, dosing is 1 mg/kg every 48 h, and dosage will increase to 1.5 mg/kg every 24 h in the second phase. Initial anakinra dosing was determined through population pharmacokinetic model simulations. During the study, there will be a interim analysis to confirm predictions before undertaking dose assessment. Anakinra therapy will be considered safe if the frequency of adverse outcomes/events does not exceed that expected in infants born at 240-276 weeks GA. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/, identifier NCT05280340.
Collapse
Affiliation(s)
- Elys A. Green
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - David Metz
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Children’s Hospital, Melbourne, VIC, Australia
- Murdoch Children’s Research Institute, Melbourne, VIC, Australia
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Rebecka Atkinson
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Elizbeth M. Skuza
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Megan Clark
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
- Faculty of Pharmacy and Pharmaceutical Science, Monash University, Melbourne, VIC, Australia
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Carl M. Kirkpatrick
- Monash Institute for Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Rod W. Hunt
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Philip J. Berger
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Claudia A. Nold-Petry
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Marcel F. Nold
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
77
|
Macovei LA, Burlui A, Bratoiu I, Rezus C, Cardoneanu A, Richter P, Szalontay A, Rezus E. Adult-Onset Still's Disease-A Complex Disease, a Challenging Treatment. Int J Mol Sci 2022; 23:12810. [PMID: 36361602 PMCID: PMC9655522 DOI: 10.3390/ijms232112810] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 12/02/2022] Open
Abstract
Adult-onset Still's disease (AOSD) is a systemic inflammatory disorder with an unknown cause characterized by high-spiking fever, lymphadenopathy, hepatosplenomegaly, hyperferritinemia, and leukocytosis. The clinical course can be divided into three significant patterns, each with a different prognosis: Self-limited or monophasic, intermittent or polycyclic systemic, and chronic articular. Two criteria sets have been validated. The Yamaguchi criteria are the most generally used, although the Fautrel criteria offer the benefit of adding ferritin and glycosylated ferritin values. AOSD's pathogenesis is not yet completely understood. Chemokines and pro-inflammatory cytokines, including interferon (IFN)-γ, tumor necrosis factor α (TNFα), interleukin (IL)-1, IL-6, IL-8, and IL-18, play a crucial role in the progression of illness, resulting in the development of innovative targeted therapeutics. There are no treatment guidelines for AOSD due to its rarity, absence of controlled research, and lack of a standard definition for remission and therapy objectives. Non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids (CS), and conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) are used in AOSD treatment. Biological therapy, including IL-1, IL-6, IL-18, and IL-17 inhibitors, as well as TNFα or Janus-kinases (JAKs) inhibitors, is administered to patients who do not react to CS and csDMARDs or achieve an inadequate response.
Collapse
Affiliation(s)
- Luana Andreea Macovei
- Department of Rheumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Alexandra Burlui
- Department of Rheumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Patricia Richter
- Department of Rheumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Andreea Szalontay
- Department of Psychiatry, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Psychiatry “Socola”, 700282 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
78
|
Khani E, Shahrabi M, Rezaei H, Pourkarim F, Afsharirad H, Solduzian M. Current evidence on the use of anakinra in COVID-19. Int Immunopharmacol 2022; 111:109075. [PMID: 35905562 PMCID: PMC9296834 DOI: 10.1016/j.intimp.2022.109075] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/07/2023]
Abstract
Despite the progressing knowledge in COVID-19 management, remdesivir is the only agent that got approval to inhibit viral replication. However, there are limited data about effective immunomodulatory agents to prevent cytokine release in COVID-19. Cytokine release syndrome in COVID-19 resembles secondary hemophagocytic lymphohistiocytosis, in which interleukin-1 (IL-1) plays a key role. Anakinra is the first recombinant IL-1 receptor antagonist studied for off-label use in COVID-19 treatment. This study reviews the current clinical evidence on the role of interleukin-1 in COVID-19-related cytokine storm, therapeutic effects, significant clinical concerns, and pros and cons of anakinra administration in the management of COVID-19 patients. In this review, four items are shown to be important for achieving the optimal therapeutic effects of anakinra in COVID-19 patients. These items include duration of treatment ≥ 10 days, doses ≥ 100 mg, intravenous administration, and early initiation of therapy. Also, anakinra might be more beneficial in the early stages of the disease when higher levels of cytokines are yet to be observed, which could prevent progression to severe illness and mechanical ventilation. Further studies are required to address the SARS-CoV-2 induced cytokine release syndrome and the role of anakinra in identifying ideal treatment approaches for COVID-19 patients based on their clinical status.
Collapse
Affiliation(s)
- Elnaz Khani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marzieh Shahrabi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Rezaei
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Pourkarim
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Afsharirad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Solduzian
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
79
|
van Arkel C, Boeree M, Magis-Escurra C, Hoefsloot W, Carpaij N, van Ingen J, Pegge S, Wielders P, Smeenk F, Aarnoutse R, Netea MG, van Crevel R, van Laarhoven A. Interleukin-1 receptor antagonist anakinra as treatment for paradoxical responses in HIV-negative tuberculosis patients: A case series. MED 2022; 3:603-611.e2. [PMID: 36041428 DOI: 10.1016/j.medj.2022.07.001] [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: 02/22/2022] [Revised: 05/19/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Paradoxical inflammatory responses can occur during microbiologically successful antituberculous therapy. Optimal treatment is unknown, but corticosteroids are used most often. It is likely that interleukin-1 (IL-1) plays a central role in the development of these paradoxical responses, and if corticosteroids fail or are undesirable because of adverse effects, anti-IL-1 therapy may therefore be a rational choice. METHODS We present seven HIV-negative tuberculosis patients with paradoxical responses, two with exclusively pulmonary and five with extrapulmonary tuberculosis. All had received corticosteroids, with unsatisfactory effect. Patients were treated with the IL-1 receptor antagonist anakinra and monitored for reduction of fever and inflammatory markers, imaging evidence of stabilization or regression of lesions, and respiratory improvement. FINDINGS Six patients had anemia and four patients had lymphopenia at the start of the antituberculosis treatment. Fever was present in six patients at the moment of paradoxical response. Anakinra resulted in the decrease of fever within days, followed by resolution of symptoms and radiological improvement in five patients. Anakinra induced neutropenia, necessitating its cessation in two patients, who recovered quickly afterward. CONCLUSION Anakinra can be considered in HIV-negative tuberculosis patients with paradoxical responses when steroids fail or are undesired. Given its favorable safety profile and reversible side effects, it is conceivable that anakinra might also be used as first-line adjuvant treatment for paradoxical responses. FUNDING A.v.L. and R.v.C. are supported by National Institutes of Health (R01AI145781).
Collapse
Affiliation(s)
- Cynthia van Arkel
- Department of Pulmonary Diseases, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Pulmonary Diseases, Catharina Hospital, 5623 EJ Eindhoven, the Netherlands
| | - Martin Boeree
- Department of Pulmonary Diseases, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Cécile Magis-Escurra
- Department of Pulmonary Diseases, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Wouter Hoefsloot
- Department of Pulmonary Diseases, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Neeltje Carpaij
- Department of Pulmonary Diseases, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Sjoert Pegge
- Department of Radiology, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Pascal Wielders
- Department of Pulmonary Diseases, Catharina Hospital, 5623 EJ Eindhoven, the Netherlands
| | - Frank Smeenk
- Department of Pulmonary Diseases, Catharina Hospital, 5623 EJ Eindhoven, the Netherlands
| | - Rob Aarnoutse
- Department of Pharmacy, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Reinout van Crevel
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Arjan van Laarhoven
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
80
|
Cron RQ. Biologic disease-modifying antirheumatic drugs to treat multisystem inflammatory syndrome in children. Curr Opin Rheumatol 2022; 34:274-279. [PMID: 35791863 DOI: 10.1097/bor.0000000000000889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Multisystem inflammatory syndrome in children (MIS-C) is a postinfectious complication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection primarily affecting children. MIS-C shares features with Kawasaki disease (KD) and cytokine storm syndrome (CSS) frequently requiring intensive care support. Although intravenous immunoglobulin (IVIg) and glucocorticoids (GCs) are effective therapeutics for most, refractory MIS-C is treated with various biologic disease-modifying antirheumatic drugs (bDMARDs). Understanding the clinical features, inflammatory cytokines, and genetic associations provides rationale for bDMARD in treating severe MIS-C. RECENT FINDINGS Children with MIS-C have clinical KD features and often present in hypovolemic and cardiogenic shock requiring volume repletion (gastrointestinaI losses) and cardiac pressor support (epinephrine). Investigation of MIS-C serum reveals elevated pro-inflammatory cytokines [interleukin (IL)-1, IL-6, IL-18, interferon gamma (IFNγ), tumor necrosis factor (TNF)], but to a lesser extent than other established CSS. Gene sequencing of MIS-C children identifies heterozygous mutations in CSS associated genes. Treatment of refractory (IVIg and GC) MIS-C with bDMARDs to IL-1, IL-6, and TNF is efficacious for survival as well as resolving cardiac and coronary artery inflammation. SUMMARY MIS-C is a postinfectious complication of SARS-CoV-2 resembling KD and CSS, both genetically and by pro-inflammatory cytokines. MIS-C that is refractory to IVIg and GC is routinely responsive to bDMARDs targeting IL-1, IL-6, and TNF.
Collapse
Affiliation(s)
- Randy Q Cron
- Division of Pediatric Rheumatology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
81
|
Audemard-Verger A, Le Gouge A, Pestre V, Courjon J, Langlois V, Vareil MO, Devaux M, Bienvenu B, Leroy V, Goulabchand R, Colombain L, Bigot A, Guimard T, Douadi Y, Urbanski G, Faucher JF, Maulin L, Lioger B, Talarmin JP, Groh M, Emmerich J, Deriaz S, Ferreira-Maldent N, Cook AR, Lengellé C, Bourgoin H, Mekinian A, Aouba A, Maillot F, Caille A. Efficacy and safety of anakinra in adults presenting deteriorating respiratory symptoms from COVID-19: A randomized controlled trial. PLoS One 2022; 17:e0269065. [PMID: 35925914 PMCID: PMC9351999 DOI: 10.1371/journal.pone.0269065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/12/2022] [Indexed: 01/08/2023] Open
Abstract
Objective We aimed to investigate whether anakinra, an interleukin-1receptor inhibitor, could improve outcome in moderate COVID-19 patients. Methods In this controlled, open-label trial, we enrolled adults with COVID-19 requiring oxygen. We randomly assigned patients to receive intravenous anakinra plus optimized standard of care (oSOC) vs. oSOC alone. The primary outcome was treatment success at day 14 defined as patient alive and not requiring mechanical ventilation or extracorporeal membrane oxygenation. Results Between 27th April and 6th October 2020, we enrolled 71 patients (240 patients planned to been enrolled): 37 were assigned to the anakinra group and 34 to oSOC group. The study ended prematurely by recommendation of the data and safety monitoring board due to safety concerns. On day 14, the proportion of treatment success was significantly lower in the anakinra group 70% (n = 26) vs. 91% (n = 31) in the oSOC group: risk difference—21 percentage points (95% CI, -39 to -2), odds ratio 0.23 (95% CI, 0.06 to 0.91), p = 0.027. After a 28-day follow-up, 9 patients in the anakinra group and 3 in the oSOC group had died. Overall survival at day 28 was 75% (95% CI, 62% to 91%) in the anakinra group versus 91% (95% CI, 82% to 100%) (p = 0.06) in the oSOC group. Serious adverse events occurred in 19 (51%) patients in the anakinra group and 18 (53%) in the oSOC group (p = 0·89). Conclusion This trial did not show efficacy of anakinra in patients with COVID-19. Furthermore, contrary to our hypothesis, we found that anakinra was inferior to oSOC in patients with moderate COVID-19 pneumonia.
Collapse
Affiliation(s)
- Alexandra Audemard-Verger
- Department of Internal Medicine and Clinical Immunology, CHRU Tours, Tours, France
- University of Tours, Tours, France
- * E-mail:
| | | | - Vincent Pestre
- Department of Internal Medicine and Infectious Diseases, CH Avignon, Avignon, France
| | - Johan Courjon
- Department of Infectious Diseases, Université Côte d’Azur, CHU Nice, Nice, France
| | | | | | - Mathilde Devaux
- Department of Internal Medicine, CH de Poissy, Poissy, France
| | - Boris Bienvenu
- Department of Internal Medicine, Hôpital Saint Joseph, Marseille, France
| | - Vincent Leroy
- Department of Infectious Diseases, Clinique Tessier, Valenciennes, France
| | - Radjiv Goulabchand
- Internal Medicine Department & Department of Infectious and Tropical Diseases, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Léa Colombain
- Department of Infectious Diseases, CH de Perpignan, Perpignan, France
| | - Adrien Bigot
- Department of Internal Medicine and Clinical Immunology, CHRU Tours, Tours, France
| | - Thomas Guimard
- Department of Infectious Diseases, CH la Roche sur Yon, La Roche sur Yon, France
| | - Youcef Douadi
- Department of Infectious Diseases, CH Saint Quentin, Saint Quentin, France
| | - Geoffrey Urbanski
- Department of Internal Medicine and Clinical Immunology, CHU Angers, Angers France
| | | | - Laurence Maulin
- Department of Infectious Diseases, CH Aix en Provence, Aix en Provence, France
| | | | | | - Matthieu Groh
- Department of Internal Medicine, Hôpital Foch, Suresnes, France
| | - Joseph Emmerich
- Department of Vascular Medicine, GH Saint Joseph and Université de Paris, INSERM CRESS 1153, Paris, France
| | - Sophie Deriaz
- Department of Internal Medicine and Clinical Immunology, CHRU Tours, Tours, France
| | | | - Ann-Rose Cook
- Department of Internal Medicine and Clinical Immunology, CHRU Tours, Tours, France
| | | | | | - Arsène Mekinian
- Department of Internal Medicine, Hôpital Saint Antoine, Sorbonne Université, Paris, France
| | - Achille Aouba
- Department of Internal Medicine, CHU de Caen, Caen, France
| | - François Maillot
- Department of Internal Medicine and Clinical Immunology, CHRU Tours, Tours, France
- University of Tours, Tours, France
| | - Agnès Caille
- University of Tours, Tours, France
- INSERM CIC1415, CHRU Tours, Tours, France
| |
Collapse
|
82
|
Dafni M, Karampeli M, Michelakis I, Manta A, Spanoudaki A, Mantzos D, Krontira S, Georgiadou V, Lioni A, Tzavara V. Treatment with 3-day methylprednisolone pulses in severe cases of COVID-19 compared with the standard regimen protocol of dexamethasone. J Investig Med 2022; 70:1423-1428. [PMID: 35379701 PMCID: PMC9002257 DOI: 10.1136/jim-2021-002274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 01/08/2023]
Abstract
Since the outbreak of COVID-19, research has been focused on establishing effective treatments, especially for patients with severe pneumonia and hyperinflammation. The role and dose of corticosteroids remain obscure. We evaluated 58 patients with severe COVID-19 during two periods. 24 patients who received methylprednisolone pulses (250 mg/day intravenously for 3 days) were compared with 34 patients treated according to the standard dexamethasone protocol of 6 mg/day. Among non-intubated patients, the duration of hospitalization was shorter for those who received methylprednisolone pulses (9.5 vs 13.5, p<0.001). In a subgroup analysis of patients who required intubation, those treated with the dexamethasone protocol demonstrated a relative risk=1.89 (p=0.09) for dying, in contrast to the other group which showed a tendency towards extubation and discharge from the hospital. A 'delayed' need for intubation was also observed (6 vs 2 days, p=0.06). Treatment with methylprednisolone pulses significantly reduced hospitalization time. Although there was no statistically significant influence on the necessity for intubation, methylprednisolone pulses revealed a tendency to delay intubation and hospital discharges. This treatment could benefit patients in the hyperinflammatory phase of the disease.
Collapse
Affiliation(s)
- Maria Dafni
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Maria Karampeli
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Ioannis Michelakis
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Aspasia Manta
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Anastasia Spanoudaki
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Dionysios Mantzos
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Sofia Krontira
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Victoria Georgiadou
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Athina Lioni
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| | - Vasiliki Tzavara
- 1st Department of Internal Medicine, Korgialenio-Benakio Red Cross General Hospital, Athens, Greece
| |
Collapse
|
83
|
Bertoni A, Penco F, Mollica H, Bocca P, Prigione I, Corcione A, Cangelosi D, Schena F, Del Zotto G, Amaro A, Paladino N, Pontali E, Feasi M, Signa S, Bustaffa M, Caorsi R, Palmeri S, Contini P, De Palma R, Pfeffer U, Uva P, Rubartelli A, Gattorno M, Volpi S. Spontaneous NLRP3 inflammasome-driven IL1-β secretion is induced in severe COVID-19 patients and responds to anakinra treatment. J Allergy Clin Immunol 2022; 150:796-805. [PMID: 35835255 PMCID: PMC9272569 DOI: 10.1016/j.jaci.2022.05.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 05/03/2022] [Accepted: 05/18/2022] [Indexed: 10/27/2022]
Abstract
BACKGROUND SARS-CoV-2 infection may result in a severe pneumonia associated to elevation of blood inflammatory parameters, reminiscent of cytokine storm syndrome. Steroidal anti-inflammatory therapies have shown efficacy in reducing mortality in critically ill patients, however the mechanisms by which SARS-CoV2 virus triggers such an extensive inflammation remain unexplained. OBJECTIVES To dissect the mechanisms underlying SARS-CoV-2 associated inflammation in severe COVID-19 patients we studied the role of IL-1β, a pivotal cytokine driving inflammatory phenotypes, whose maturation and secretion are regulated by inflammasomes. METHODS We analyzed NLRP3 pathway activation by means of confocal microscopy, plasma cytokine measurement, cytokine secretion following in vitro stimulation of blood circulating monocytes and whole blood RNA sequencing. The role of ORF3a SARS-CoV2 protein was assessed by confocal microscopy analysis following nucleofection of a monocytic cell line. RESULTS We found that circulating monocytes from COVID-19 patients display ASC specks that colocalize with NLRP3 inflammasome and spontaneously secrete IL-1β in vitro. This spontaneous activation reverts following patient's treatment with the IL-1 receptor antagonist anakinra. Transfection of a monocytic cell line with cDNA coding for the ORF3a SARS-CoV2 protein, resulted in ASC speck formation CONCLUSIONS: These results provide further evidence that IL-1β targeting could represent an effective strategy in this disease and suggest a mechanistic explanation for the strong inflammatory manifestations associated to COVID-19.
Collapse
Affiliation(s)
- Arinna Bertoni
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy; DINOGMI, Università degli Studi di Genova, Genova, Italy
| | - Federica Penco
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Hilaria Mollica
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy; DINOGMI, Università degli Studi di Genova, Genova, Italy
| | - Paola Bocca
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Ignazia Prigione
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Anna Corcione
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Davide Cangelosi
- Clinical Bioinformatics Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Francesca Schena
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Genny Del Zotto
- Department of Research and Diagnostics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Adriana Amaro
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Noemi Paladino
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | | | | - Sara Signa
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy; DINOGMI, Università degli Studi di Genova, Genova, Italy
| | - Marta Bustaffa
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Roberta Caorsi
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Serena Palmeri
- DINOGMI, Università degli Studi di Genova, Genova, Italy
| | - Paola Contini
- Department of Internal Medicine, University of Genoa and IRCCS IST-Ospedale San Martino, Genoa, Italy
| | - Raffaele De Palma
- Department of Internal Medicine, University of Genoa and IRCCS IST-Ospedale San Martino, Genoa, Italy
| | | | - Paolo Uva
- Clinical Bioinformatics Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy; Italian Institute of Technology, Genova, Italy
| | | | - Marco Gattorno
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy.
| | - Stefano Volpi
- UOSD Centro per le Malattie Autoinfiammatorie e Immunodeficienze, IRCCS Istituto Giannina Gaslini, Genova, Italy; DINOGMI, Università degli Studi di Genova, Genova, Italy
| |
Collapse
|
84
|
Zhou J, Wu ZQ, Qiao T, Xu HG. Development of Laboratory Parameters-Based Formulas in Predicting Short Outcomes for Adult Hemophagocytic Lymphohistiocytosis Patients with Different Underlying Diseases. J Clin Immunol 2022; 42:1000-1008. [PMID: 35386042 DOI: 10.1007/s10875-022-01263-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Hemophagocytic lymphohistiocytosis (HLH) is a severe disease with high mortality. The purpose of this investigation was to build models to predict 30-day death in total and subgroup HLH patients based on available and cheap laboratory parameters. METHOD The research contained 431 adults HLH patients from January 2015 to September 2021 in the hospital. Logistic regression and receiver operating characteristic (ROC) were utilized to build models. RESULTS Results suggested that age, ferritin, lymphocyte (LY), international normalized ratio (INR), thrombin time (TT), globulin, uric acid (UA), chloride, activated partial thromboplastin time (APTT), aspartate aminotransferase (AST), triglycerides (TG), total bilirubin (TB), and indirect bilirubin (IB) were independent factors in HLH and subgroups. Then, models adapted to patients with different underlying diseases were established based on these factors. Area under curve (AUC) of these models was excellent: HLH patients: 0.838 (p < 0.001); infection-associated HLH (I-HLH) patients: 0.913 (p < 0.001); malignancy-associated HLH (M-HLH): 0.921 (p < 0.001) and 0.809 (p < 0.001) for two or more different etiologies-associated HLH (Mix-HLH patients). In addition, UA, TT, and chloride were firstly confirmed as independent factors in adult HLH. CONCLUSION Four models depending on biomarkers that available and affordable in clinical practice were built. With these models, high-risk patients with different underlying diseases could be easily identified.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, Jiangsu, China
| | - Zhi-Qi Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, Jiangsu, China
| | - Tengfei Qiao
- Department of Laboratory Medicine, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Hua-Guo Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
85
|
Zhang X, Zhu L, Zhang H, Chen S, Xiao Y. CAR-T Cell Therapy in Hematological Malignancies: Current Opportunities and Challenges. Front Immunol 2022; 13:927153. [PMID: 35757715 PMCID: PMC9226391 DOI: 10.3389/fimmu.2022.927153] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy represents a major breakthrough in cancer treatment, and it has achieved unprecedented success in hematological malignancies, especially in relapsed/refractory (R/R) B cell malignancies. At present, CD19 and BCMA are the most common targets in CAR-T cell therapy, and numerous novel therapeutic targets are being explored. However, the adverse events related to CAR-T cell therapy might be serious or even life-threatening, such as cytokine release syndrome (CRS), CAR-T-cell-related encephalopathy syndrome (CRES), infections, cytopenia, and CRS-related coagulopathy. In addition, due to antigen escape, the limited CAR-T cell persistence, and immunosuppressive tumor microenvironment, a considerable proportion of patients relapse after CAR-T cell therapy. Thus, in this review, we focus on the progress and challenges of CAR-T cell therapy in hematological malignancies, such as attractive therapeutic targets, CAR-T related toxicities, and resistance to CAR-T cell therapy, and provide some practical recommendations.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Zhu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Shanshan Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Xiao
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
86
|
Diorio C, Vatsayan A, Talleur AC, Annesley C, Jaroscak JJ, Shalabi H, Ombrello AK, Hudspeth M, Maude SL, Gardner RA, Shah NN. Anakinra utilization in refractory pediatric CAR T-cell associated toxicities. Blood Adv 2022; 6:3398-3403. [PMID: 35395068 PMCID: PMC9198909 DOI: 10.1182/bloodadvances.2022006983] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Caroline Diorio
- Pediatric Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Anant Vatsayan
- Division of BMT, Children’s National Hospital, Washington, DC
| | - Aimee C. Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Colleen Annesley
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA
| | - Jennifer J. Jaroscak
- Pediatric Hematology/Oncology, Medical University of South Carolina, Charleston, SC
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; and
| | - Amanda K. Ombrello
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Michelle Hudspeth
- Pediatric Hematology/Oncology, Medical University of South Carolina, Charleston, SC
| | - Shannon L. Maude
- Pediatric Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | | | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; and
| |
Collapse
|
87
|
Cefai E, Caruana D, Sillato MG, Mercieca C. Haemophagocytic lymphohistiocytosis secondary to parvovirus B19 infection: an unexpected case of multiorgan failure. Br J Hosp Med (Lond) 2022; 83:1-4. [DOI: 10.12968/hmed.2021.0619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- E Cefai
- Rheumatology Department, Mater Dei Hospital, Msida, Malta
| | - D Caruana
- Rheumatology Department, Mater Dei Hospital, Msida, Malta
| | | | - C Mercieca
- Rheumatology Department, Mater Dei Hospital, Msida, Malta
| |
Collapse
|
88
|
Sil A, Das A, Datta D. Mucocutaneous manifestations of COVID-19 related multisystem inflammatory syndrome in adults (MIS-A): an update. Clin Exp Dermatol 2022; 47:1774-1780. [PMID: 35603763 PMCID: PMC9348323 DOI: 10.1111/ced.15271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2022] [Indexed: 11/29/2022]
Abstract
Multisystem inflammatory syndrome in adults (MIS‐A) is an inflammatory condition that affects multiple extrapulmonary organ systems (cardiac, gastrointestinal tract, dermatological and/or neurological), attributed to a postinfectious and atypical complication occurring weeks to months after infection with severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2). The diagnosis is primarily based on findings encompassing persistent fever, elevated inflammatory markers, multiorgan involvement and a temporal relationship with COVID‐19 infection. The existing literature on MIS‐A, although growing, is limited to case reports and small case series. It is imperative that dermatologists be aware of this entity and aid the critical care team to ensure timely diagnosis and early therapeutic intervention. In this review, we concisely highlight the varied presentations, pathogenesis and treatment options in MIS‐A.
Collapse
Affiliation(s)
- Abheek Sil
- Department of Dermatology, Venereology, and Leprosy; RG Kar Medical College & Hospital, Kolkata, India
| | - Anupam Das
- Department of Dermatology, Venereology, and Leprosy; KPC Medical College & Hospital, Kolkata, India
| | - Debatri Datta
- Consultant Dermatologist, Oliva Skin and Hair Clinic, Kolkata, India
| |
Collapse
|
89
|
Dreyzin A, Jacobsohn D, Angiolillo A, Wistinghausen B, Schore RJ, Perez E, Wells E, Terao J, Bonifant C, Rohatgi R, Dave H, Vatsayan A. Intravenous anakinra for tisagenlecleucel-related toxicities in children and young adults. Pediatr Hematol Oncol 2022; 39:370-378. [PMID: 34672243 DOI: 10.1080/08880018.2021.1988012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Alexandra Dreyzin
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - David Jacobsohn
- Division of Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Anne Angiolillo
- Division of Oncology, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Birte Wistinghausen
- Division of Oncology, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Reuven J Schore
- Division of Oncology, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Evelio Perez
- Division of Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Elizabeth Wells
- Neuroscience and Behavioral Medicine Center, Children's National Hospital, Washington, DC, USA
| | - Joshua Terao
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Challice Bonifant
- Division of Oncology, Johns Hopkins Medical Institutions Campus, Baltimore, MD, USA
| | - Radha Rohatgi
- Division of Pharmacy Services, Children's National Hospital, Washington, DC, USA
| | - Hema Dave
- Division of Oncology, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - Anant Vatsayan
- Division of Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| |
Collapse
|
90
|
Mannion ML, Cron RQ. Therapeutic strategies for treating juvenile idiopathic arthritis. Curr Opin Pharmacol 2022; 64:102226. [PMID: 35461129 DOI: 10.1016/j.coph.2022.102226] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
Recent development of new medications has changed the juvenile idiopathic arthritis (JIA) treatment goal to inactive disease. With numerous options, how does a clinician choose which medication to use? Treatment options may depend on the clinical classification and a new paradigm considers the JIA subtypes in reference to categories of adult inflammatory arthritis; poligo JIA, spondyloarthritis JIA, and systemic JIA that can help guide a clinician in determining treatment options. Treatment strategies such as consensus treatment plans can provide guidance on treatment escalation. However, a treat-to-target strategy using frequent standardized disease activity measurements, shared decision making with the patient, and treatment escalation to achieve the disease activity target can provide a personalized approach to managing JIA.
Collapse
Affiliation(s)
- Melissa L Mannion
- University of Alabama at Birmingham, Pediatric Rheumatology, 1600 7th Ave S, CPPN G10, Birmingham, AL, 35233, USA.
| | - Randy Q Cron
- University of Alabama at Birmingham, Pediatric Rheumatology, 1600 7th Ave S, CPPN G10, Birmingham, AL, 35233, USA
| |
Collapse
|
91
|
Ailioaie LM, Ailioaie C, Litscher G. Implications of SARS-CoV-2 Infection in Systemic Juvenile Idiopathic Arthritis. Int J Mol Sci 2022; 23:4268. [PMID: 35457086 PMCID: PMC9029451 DOI: 10.3390/ijms23084268] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/30/2022] [Accepted: 04/10/2022] [Indexed: 12/15/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is a serious multifactorial autoinflammatory disease with a significant mortality rate due to macrophage activation syndrome (MAS). Recent research has deepened the knowledge about the pathophysiological mechanisms of sJIA-MAS, facilitating new targeted treatments, and biological disease-modifying antirheumatic drugs (bDMARDs), which significantly changed the course of the disease and prognosis. This review highlights that children are less likely to suffer severe COVID-19 infection, but at approximately 2-4 weeks, some cases of multisystem inflammatory syndrome in children (MIS-C) have been reported, with a fulminant course. Previous established treatments for cytokine storm syndrome (CSS) have guided COVID-19 therapeutics. sJIA-MAS is different from severe cases of COVID-19, a unique immune process in which a huge release of cytokines will especially flood the lungs. In this context, MIS-C should be reinterpreted as a special MAS, and long-term protection against SARS-CoV-2 infection can only be provided by the vaccine, but we do not yet have sufficient data. COVID-19 does not appear to have a substantial impact on rheumatic and musculoskeletal diseases (RMDs) activity in children treated with bDMARDs, but the clinical features, severity and outcome in these patients under various drugs are not yet easy to predict. Multicenter randomized controlled trials are still needed to determine when and by what means immunoregulatory products should be administered to patients with sJIA-MAS with a negative corticosteroid response or contraindications, to optimize their health and safety in the COVID era.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania; (L.M.A.); (C.A.)
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania; (L.M.A.); (C.A.)
| | - Gerhard Litscher
- Research Unit of Biomedical Engineering in Anesthesia and Intensive Care Medicine, Research Unit for Complementary and Integrative Laser Medicine, Traditional Chinese Medicine (TCM) Research Center Graz, Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 39, 8036 Graz, Austria
| |
Collapse
|
92
|
Huemer C. Aktuelles aus der pädiatrischen Rheumatologie. RHEUMA PLUS 2022; 21:54-55. [PMID: 35401847 PMCID: PMC8978499 DOI: 10.1007/s12688-022-00523-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Christian Huemer
- Abteilung für Kinder- und Jugendheilkunde, Landeskrankenhaus Bregenz, Akademisches Lehrkrankenhaus der UniversitätenWien, Innsbruck & Graz, Carl Pedenzstr. 2, 6900 Bregenz, Österreich
| |
Collapse
|
93
|
Cirillo E, Esposito C, Giardino G, Azan G, Fecarotta S, Pittaluga S, Ruggiero L, Barretta F, Frisso G, Notarangelo LD, Pignata C. Case Report: Severe Rhabdomyolysis and Multiorgan Failure After ChAdOx1 nCoV-19 Vaccination. Front Immunol 2022; 13:845496. [PMID: 35371100 PMCID: PMC8968726 DOI: 10.3389/fimmu.2022.845496] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/28/2022] [Indexed: 12/18/2022] Open
Abstract
Background Severe skeletal muscle damage has been recently reported in patients with SARS-CoV-2 infection and as a rare vaccination complication. Case summary On Apr 28, 2021 a 68-year-old man who was previously healthy presented with an extremely severe rhabdomyolysis that occurred nine days following the first dose of SARS-CoV-2 ChAdOx1 nCov-19 vaccination. He had no risk factors, and denied any further assumption of drugs except for fermented red rice, and berberine supplement. The clinical scenario was complicated by a multi organ failure involving bone marrow, liver, lung, and kidney. For the rapid increase of the inflammatory markers, a cytokine storm was suspected and multi-target biologic immunosuppressive therapy was started, consisting of steroids, anakinra, and eculizumab, which was initially successful resulting in close to normal values of creatine phosphokinase after 17 days of treatment. Unfortunately, 48 days after the vaccination an accelerated phase of deterioration, characterized by severe multi-lineage cytopenia, untreatable hypotensive shock, hypoglycemia, and dramatic increase of procalcitonin (PCT), led to patient death. Conclusion Physicians should be aware that severe and fatal rhabdomyolysis may occur after SARS-CoV2 vaccine administration.
Collapse
Affiliation(s)
- Emilia Cirillo
- Departments of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Ciro Esposito
- Department of Transplants, A. Cardarelli Hospital, Naples, Italy
| | - Giuliana Giardino
- Departments of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Gaetano Azan
- Department of Transplants, A. Cardarelli Hospital, Naples, Italy
| | - Simona Fecarotta
- Departments of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Stefania Pittaluga
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Lucia Ruggiero
- Department of Neuroscience, Reproductive and Odontostomatological Science, Federico II University of Naples, Naples, Italy
| | - Ferdinando Barretta
- Department of Molecular Medicine and Medical Biotechnology , Federico II University of Naples, Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnology , Federico II University of Naples, Naples, Italy
| | - Luigi Daniele Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Claudio Pignata
- Departments of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
- *Correspondence: Claudio Pignata,
| |
Collapse
|
94
|
Cooray S, Sabanathan S, Hacohen Y, Worth A, Eleftheriou D, Hemingway C. Treatment Strategies for Central Nervous System Effects in Primary and Secondary Haemophagocytic Lymphohistiocytosis in Children. Curr Treat Options Neurol 2022. [DOI: 10.1007/s11940-022-00705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Abstract
Purpose of Review
This review presents an appraisal of current therapeutic options for the treatment of central nervous system haemophagocytic lymphohistiocytosis (CNS-HLH) in the context of systemic disease, as well as when CNS features occur in isolation. We present the reader with a diagnostic approach to CNS-HLH and commonly used treatment protocols. We discuss and evaluate newer treatments on the horizon.
Recent Findings
Mortality is high in patients who do not undergo HSCT, and while larger studies are required to establish benefit in many treatments, a number of new treatments are currently being evaluated. Alemtuzumab is being used as a first-line treatment for CNS-HLH in a phase I/II multicentre prospective clinical trial as an alternative to traditional HLH-1994 and 2004 protocols. It has also been used successfully as a second-line agent for the treatment of isolated CNS-HLH that is refractory to standard treatment. Ruxolitinib and emapalumab are new immunotherapies that block the Janus kinase—Signal Transducer and Activator of Transcription (JAK-STAT) pathway that have shown efficacy in refractory HLH, including for CNS-HLH disease.
Summary
Treatment of CNS-HLH often requires HLH-94 or 2004 protocols followed by haematopoietic stem cell transplantation (HSCT) to maintain remission, although relapse can occur, particularly with reduced intensity conditioning if donor chimerism falls. CNS features have been shown to improve or stabilise following HSCT in CNS-HLH in the context of systemic disease and in isolated CNS-HLH. Encouraging reports of early cohort studies suggest alemtuzumab and the Janus kinase (JAK) inhibitor ruxolitinib offer potential salvage therapy for relapsed and refractory CNS-HLH. Newer immunotherapies such as tocilizumab and natalizumab have been shown to be beneficial in sporadic cases. CNS-HLH due to primary gene defects may be amenable to gene therapy in the future.
Collapse
|
95
|
Stubbs LA, Szafron V, Forbes LR, Musick MA, Gillispie AE, Sauer HE, Smith VR, Fasipe TA, Munoz FM, Tejtel KS, Silva-Carmona M, Vogel TP, Muscal E. Severe Pediatric COVID-19 Pneumonia Treated With Adjuvant Anakinra. Hosp Pediatr 2022; 12:e162-e170. [PMID: 35237791 DOI: 10.1542/hpeds.2021-006376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To compare prior hemophagocytic lymphohistiocytosis (HLH) criteria to adult COVID-19-associated hyperinflammatory syndrome (cHIS) criteria for the diagnosis of hyperinflammation in pediatric patients with COVID-19. The secondary objective was to assess treatment response to intravenous (IV) anakinra in these patients. METHODS This case series included children admitted to the pediatric intensive care unit (PICU) for COVID-19 pneumonia with hyperinflammation and treated with IV anakinra between July 2020 to April 2021. Hyperinflammatory criteria were determined for each patient. Clinical course, chest imaging, and inflammatory marker trends were assessed pre- and post-anakinra treatment. RESULTS All patients had a cHIS criteria score of >5. Two patients met 2004-HLH criteria. Only the patient that required extracorporeal membrane oxygenation (ECMO) met the H-Score cut-off value. All but one patient had a decrease in their inflammatory markers and improvement in clinical status with early initiation of adjunctive IV anakinra. CONCLUSIONS In this case series, adult cHIS criteria were successfully used to identify pediatric COVID-19 patients with hyperinflammation. Ferritin levels decreased following early initiation of IV anakinra.
Collapse
Affiliation(s)
- Leigh A Stubbs
- Department of Pediatrics, Division of Rheumatology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Vibha Szafron
- Department of Pediatrics, Division of Allergy and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Lisa R Forbes
- Department of Pediatrics, Division of Allergy and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Matthew A Musick
- Department of Pediatrics, Division of Critical Care Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | | | - Hannah E Sauer
- Department of Pharmacy, Texas Children's Hospital, Houston, TX
| | - Valeria R Smith
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Titilope A Fasipe
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Flor M Munoz
- Department of Pediatrics, Division of Infectious Diseases, and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Kristen Sexson Tejtel
- Department of Pediatrics, Division of Cardiology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Manuel Silva-Carmona
- Department of Pediatrics, Division of Pulmonology Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston,TX
| | - Tiphanie P Vogel
- Department of Pediatrics, Division of Rheumatology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Eyal Muscal
- Department of Pediatrics, Division of Rheumatology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| |
Collapse
|
96
|
Hyper-inflammation after COVID-19 mARN vaccination: at the crossroads of multisystem inflammatory disease and adult-onset Still's disease. Does terminology matter? ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2022; 60:3-5. [PMID: 34487678 DOI: 10.2478/rjim-2021-0035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Indexed: 01/20/2023]
|
97
|
Hayden PJ, Roddie C, Bader P, Basak GW, Bonig H, Bonini C, Chabannon C, Ciceri F, Corbacioglu S, Ellard R, Sanchez-Guijo F, Jäger U, Hildebrandt M, Hudecek M, Kersten MJ, Köhl U, Kuball J, Mielke S, Mohty M, Murray J, Nagler A, Rees J, Rioufol C, Saccardi R, Snowden JA, Styczynski J, Subklewe M, Thieblemont C, Topp M, Ispizua ÁU, Chen D, Vrhovac R, Gribben JG, Kröger N, Einsele H, Yakoub-Agha I. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol 2022; 33:259-275. [PMID: 34923107 DOI: 10.1016/j.annonc.2021.12.003] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several commercial and academic autologous chimeric antigen receptor T-cell (CAR-T) products targeting CD19 have been approved in Europe for relapsed/refractory B-cell acute lymphoblastic leukemia, high-grade B-cell lymphoma and mantle cell lymphoma. Products for other diseases such as multiple myeloma and follicular lymphoma are likely to be approved by the European Medicines Agency in the near future. DESIGN The European Society for Blood and Marrow Transplantation (EBMT)-Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association collaborated to draft best practice recommendations based on the current literature to support health care professionals in delivering consistent, high-quality care in this rapidly moving field. RESULTS Thirty-six CAR-T experts (medical, nursing, pharmacy/laboratory) assembled to draft recommendations to cover all aspects of CAR-T patient care and supply chain management, from patient selection to long-term follow-up, post-authorisation safety surveillance and regulatory issues. CONCLUSIONS We provide practical, clinically relevant recommendations on the use of these high-cost, logistically complex therapies for haematologists/oncologists, nurses and other stakeholders including pharmacists and health sector administrators involved in the delivery of CAR-T in the clinic.
Collapse
Affiliation(s)
- P J Hayden
- Department of Haematology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - C Roddie
- UCL Cancer Institute, London, UK; University College London Hospital NHS Foundation Trust, London, UK.
| | - P Bader
- Clinic for Children and Adolescents, University Children's Hospital, Frankfurt, Germany
| | - G W Basak
- Medical University of Warsaw, Department of Hematology, Transplantation and Internal Medicine, Warsaw, Poland
| | - H Bonig
- Institute for Transfusion Medicine and Immunohematology of Goethe University and German Red Cross Blood Service, Frankfurt, Germany
| | - C Bonini
- Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | - C Chabannon
- Aix-Marseille université, Inserm CBT-1409, Institut Paoli-Calmettes, centre de thérapie cellulaire, unité de transplantation et de thérapie cellulaire, département de biologie du cancer, Marseille, France
| | - F Ciceri
- Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - S Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Franz-Josef-Strauss-Allee 11, University Hospital of Regensburg, Regensburg, Germany
| | - R Ellard
- Royal Marsden Hospital, Fulham Rd, London, UK
| | - F Sanchez-Guijo
- IBSAL-Hospital Universitario de Salamanca, CIC, Universidad de Salamanca, Salamanca, Spain
| | - U Jäger
- Clinical Department for Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - M Hildebrandt
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital Grosshadern, Munich
| | - M Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - M J Kersten
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, the Netherlands
| | - U Köhl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI) and Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany; Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - J Kuball
- Department of Hematology and Centre for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Mielke
- Karolinska Institutet and University Hospital, Department of Laboratory Medicine/Department of Cell Therapy and Allogeneic Stem Cell Transplantation (CAST), Stockholm, Sweden
| | - M Mohty
- Hôpital Saint-Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - J Murray
- Christie Hospital NHS Trust, Manchester, UK
| | - A Nagler
- The Chaim Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - J Rees
- University College London Hospital NHS Foundation Trust, London, UK; UCL Institute of Neurology, University College of London Hospitals NHS Foundation Trust, London, UK
| | - C Rioufol
- Hospices Civils de Lyon, UCBL1, EMR 3738 CICLY, Lyon, France
| | - R Saccardi
- Cell Therapy and Transfusion Medicine Department, Careggi University Hospital, Florence, Italy
| | - J A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - J Styczynski
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | - M Subklewe
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Thieblemont
- AP-HP, Saint-Louis Hospital, Hemato-oncology, University of Paris, Paris, France
| | - M Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Á U Ispizua
- Department of Hematology, ICMHO, Hospital Clínic de Barcelona, Barcelona, Spain
| | - D Chen
- University College London Hospital NHS Foundation Trust, London, UK; Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - R Vrhovac
- Department of Haematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - J G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - N Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg, Germany
| | - H Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - I Yakoub-Agha
- CHU de Lille, Univ Lille, INSERM U1286, Infinite, Lille, France
| |
Collapse
|
98
|
Kim DW, Bukhari A, Lutfi F, Zafforoni F, Merechi F, Mustafa Ali MK, Gottlieb D, Lee ST, Kocoglu MH, Hardy NM, Yared J, Rapoport AP, Dahiya S, Law JY. Low utility of the H-Score and HLH-2004 criteria to identify patients with secondary hemophagocytic lymphohistiocytosis after CAR-T cell therapy for relapsed/refractory diffuse large B-Cell lymphoma. Leuk Lymphoma 2022; 63:1339-1347. [PMID: 35045791 DOI: 10.1080/10428194.2021.2024817] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Secondary hemophagocytic lymphohistiocytosis (HLH) is a life-threatening immune dysregulation disorder. Use of chimeric antigen receptor T-cell therapy (CAR-T) is associated with cytokine release syndrome (CRS), Immune Effector Cell Associated Neurotoxicity Syndrome (ICANS) and secondary HLH. However, application of HLH scoring systems (H-score, HLH-2004 criteria) are not validated in this setting. We analyzed the utility of applying the H-score and the HLH-2004 criteria to identify patients with possible HLH post-CAR-T for Relapsed/Refractory Diffuse Large B-cell Lymphoma. Only two of four patients with post CAR-T HLH met five or more of the diagnostic criteria for HLH by HLH 2004 criteria. In contrast all four post CAR-T HLH patients had a high H-score (>169); however, an additional ten patients that did not have HLH also had a high H-score. Thus, in this patient population, both scoring systems were demonstrated to have low prognostic significance in differentiating between high grade CRS and HLH.
Collapse
Affiliation(s)
- Dong Won Kim
- Department of Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Ali Bukhari
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Forat Lutfi
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Facundo Zafforoni
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Fikru Merechi
- Department of Pathology, University of Maryland Medical Center, Baltimore, MD, USA
| | - Moaath K Mustafa Ali
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - David Gottlieb
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Seung T Lee
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Mehmet H Kocoglu
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Nancy M Hardy
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jean Yared
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Aaron P Rapoport
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Saurabh Dahiya
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jennie Y Law
- Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| |
Collapse
|
99
|
Wehrli M, Gallagher K, Chen YB, Leick MB, McAfee SL, El-Jawahri AR, DeFilipp Z, Horick N, O'Donnell P, Spitzer T, Dey B, Cook D, Trailor M, Lindell K, Maus MV, Frigault MJ. Single-center experience using anakinra for steroid-refractory immune effector cell-associated neurotoxicity syndrome (ICANS). J Immunother Cancer 2022; 10:jitc-2021-003847. [PMID: 34996813 PMCID: PMC8744112 DOI: 10.1136/jitc-2021-003847] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/18/2022] Open
Abstract
In addition to remarkable antitumor activity, chimeric antigen receptor (CAR) T-cell therapy is associated with acute toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Current treatment guidelines for CRS and ICANS include use of tocilizumab, a monoclonal antibody that blocks the interleukin (IL)-6 receptor, and corticosteroids. In patients with refractory CRS, use of several other agents as third-line therapy (including siltuximab, ruxolitinib, anakinra, dasatinib, and cyclophosphamide) has been reported on an anecdotal basis. At our institution, anakinra has become the standard treatment for the management of steroid-refractory ICANS with or without CRS, based on recent animal data demonstrating the role of IL-1 in the pathogenesis of ICANS/CRS. Here, we retrospectively analyzed clinical and laboratory parameters, including serum cytokines, in 14 patients at our center treated with anakinra for steroid-refractory ICANS with or without CRS after standard treatment with tisagenlecleucel (Kymriah) or axicabtagene ciloleucel (Yescarta) CD19-targeting CAR T. We observed statistically significant and rapid reductions in fever, inflammatory cytokines, and biomarkers associated with ICANS/CRS after anakinra treatment. With three daily subcutaneous doses, anakinra did not have a clear, clinically dramatic effect on neurotoxicity, and its use did not result in rapid tapering of corticosteroids; although neutropenia and thrombocytopenia were common at the time of anakinra dosing, there were no clear delays in hematopoietic recovery or infections that were directly attributable to anakinra. Anakinra may be useful adjunct to steroids and tocilizumab in the management of CRS and/or steroid-refractory ICANs resulting from CAR T-cell therapies, but prospective studies are needed to determine its efficacy in these settings.
Collapse
Affiliation(s)
- Marc Wehrli
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Medical School, Boston, MA, USA
| | - Kathleen Gallagher
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Medical School, Boston, MA, USA
| | - Yi-Bin Chen
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Mark B Leick
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Steven L McAfee
- Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Areej R El-Jawahri
- Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Zachariah DeFilipp
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Nora Horick
- Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Paul O'Donnell
- Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Thomas Spitzer
- Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Bimal Dey
- Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Daniella Cook
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Michael Trailor
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Kevin Lindell
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA .,Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Medical School, Boston, MA, USA.,Hematopoietic Cell Transplant & Cell Therapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
100
|
FRAGILE-COLCOVID19: A Clinical Trial Based on Early Administration of an Oral Combination of Colchicine and Prednisone in Elderly Patients with COVID-19 in Geriatric Facilities. Clin Drug Investig 2022; 42:949-964. [PMID: 36173596 PMCID: PMC9521010 DOI: 10.1007/s40261-022-01201-2] [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] [Accepted: 09/05/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Unprotected and fragile elderly people in nursing homes experienced the highest mortality rates during the initial coronavirus disease 2019 (COVID-19) pandemic. OBJECTIVE Our aim was to study the role of two oral anti-inflammatory drugs, colchicine and prednisone, in elderly patients with COVID-19 in geriatric centers. METHODS A phase II/III, randomized, controlled, multicenter clinical trial was performed in a geriatric population comparing the efficacy and safety of an oral combination of prednisone (60 mg/day for 3 days) and colchicine (at loading doses of 1-1.5 mg/day for 3 days, followed by 0.5 mg/day for 11 days) with the standard treatment, based on intravenous dexamethasone. Primary endpoints assessed the efficacy in reducing death or the modified endpoint death/therapeutic failure to the study drugs over a 28-day period, while secondary endpoints included safety, laboratory changes, and additional therapies used. RESULTS Fifty-four patients (35 female/19 male) were enrolled, 25 (46.3%) of whom were allocated to the experimental arm and 29 (53.7%) to the control arm. At day 28, no differences in deaths were observed. The combination of mortality or therapeutic failure occurred in 12 (45.13%) patients receiving dexamethasone and 6 (28.13%) patients receiving colchicine/prednisone, resulting in a reduction of risk difference (RD) of - 17% (p = 0.17), with an average reduction of 39% (risk ratio [RR] 0.61) in patients receiving colchicine/prednisone (p = 0.25). Control patients received higher amounts of additional glucocorticoids (p = 0.0095) over a longer time frame (p = 0.0003). Colchicine/prednisone significantly reduced ferritin levels at day 14, as well as D-dimer and lactate dehydrogenase (LDH) levels at day 28. Adverse events were similar in both groups. CONCLUSIONS The combination colchicine/prednisone compared with intravenous dexamethasone has shown a remarkable trend to increase disease survival over a 28-day period in elderly patients requiring oxygen therapy in geriatric centers, without safety issues. CLINICAL TRIAL REGISTRY Clinical Trials Registration Number: NCT04492358.
Collapse
|