1
|
Feng Z, Wang L, Yang J, Li T, Liao X, Kang Y, Xiao F, Zhang W. Sepsis: the evolution of molecular pathogenesis concepts and clinical management. MedComm (Beijing) 2025; 6:e70109. [PMID: 39991626 PMCID: PMC11847631 DOI: 10.1002/mco2.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/29/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025] Open
Abstract
The mortality rate of sepsis is approximately 22.5%, accounting for 19.7% of the total global mortality. Since Lewis Thomas proposed in 1972 that "it is our response that makes the disease (sepsis)" rather than the invading microorganisms, numerous drugs have been developed to suppress the "overwhelming" inflammatory response, but none of them has achieved the desired effect. Continued failure has led investigators to question whether deaths in septic patients are indeed caused by uncontrolled inflammation. Here, we review the history of clinical trials based on evolving concepts of sepsis pathogenesis over the past half century, summarize the factors that led to the failure of these historical drugs and the prerequisites for the success of future drugs, and propose the basic principles of preclinical research to ensure successful clinical translation. The strategy of targeting inflammatory factors are like attempting to eliminate invaders by suppressing the host's armed forces, which is logically untenable. Sepsis may not be that complex; rather, sepsis may be the result of a failure to fight microbes when the force of an invading pathogen overwhelms our defenses. Thus, strengthening the body's defense forces instead of suppressing them may be the correct strategy to overcome sepsis.
Collapse
Affiliation(s)
- Zhongxue Feng
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Lijun Wang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jing Yang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tingting Li
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xuelian Liao
- Department of Critical Care MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yan Kang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Fei Xiao
- Department of Intensive Care Unit of Gynecology and ObstetricsWest China Second University Hospital, Sichuan UniversityChengduSichuanChina
| | - Wei Zhang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
2
|
Wiesinger A, Bigger B, Giugliani R, Lampe C, Scarpa M, Moser T, Kampmann C, Zimmermann G, Lagler FB. Development of a novel tool for individual treatment trials in mucopolysaccharidosis. J Inherit Metab Dis 2025; 48:e12816. [PMID: 39572375 PMCID: PMC11670214 DOI: 10.1002/jimd.12816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 12/28/2024]
Abstract
Mucopolysaccharidosis (MPS) encompasses a group of genetic lysosomal storage disorders, linked to reduced life expectancy and a significant lack of effective treatment options. Immunomodulatory drugs could have the potential to be a relevant medical approach, as the accumulation of undegraded substances initiates an innate immune response, which leads to inflammation and clinical deterioration. However, immunomodulators are not licensed for this indication. Consequently, we aim to provide evidence advocating fast access to innovative individual treatment trials (ITTs) with immunomodulatory drugs and high-quality evaluation of drug effects by implementing a risk-benefit model tailored for MPS. The iterative methodology of our novel decision analysis framework (DAF) involves three key steps: (i) literature review on promising treatment targets and immunomodulators in MPS; (ii) quantitative risk-benefit assessment (RBA) of selected molecules; (iii) assigning phenotypic profiles and quantitative evaluations. The results facilitate a personalized application of the model and are based on published evidence as well as interdisciplinary experts' consensus and patient perspectives. Four promising immunomodulators have been identified: adalimumab, abatacept, anakinra, and cladribine. An improvement in mobility is most likely with adalimumab, while anakinra is anticipated as a treatment of choice for neuronopathic MPS patients. Nevertheless, a comprehensive RBA should always be completed on an individual basis. Our evidence-based DAF tool for ITTs directly addresses the substantial unmet medical need in MPS and characterizes an initial stride toward precision medicine with immunomodulators.
Collapse
Affiliation(s)
- Anna‐Maria Wiesinger
- Institute of Congenital Metabolic DiseasesParacelsus Medical UniversitySalzburgAustria
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
| | - Brian Bigger
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Institute for Regeneration and RepairThe University of EdinburghEdinburghUK
| | - Roberto Giugliani
- Department of Genetics, UFRGS, Medical Genetics Service and Biodiscovery LaboratoryHCPA, INAGEMP, DASA, and Casa dos RarosPorto AlegreBrazil
| | - Christina Lampe
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Department of Child Neurology, Epileptology and Social Pediatrics, Center of Rare DiseasesUniversity Hospital GiessenMarburgGermany
| | - Maurizio Scarpa
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Regional Coordinating Center for Rare DiseasesUniversity Hospital UdineUdineItaly
| | - Tobias Moser
- Department of NeurologyChristian Doppler University Hospital, Paracelsus Medical UniversitySalzburgAustria
| | | | - Georg Zimmermann
- Team Biostatistics and Big Medical DataIDA Lab Salzburg, Paracelsus Medical UniversitySalzburgAustria
- Department of Artificial Intelligence and Human Interfaces, Faculty of Digital and Analytical SciencesParis Lodron UniversitySalzburgAustria
| | - Florian B. Lagler
- Institute of Congenital Metabolic DiseasesParacelsus Medical UniversitySalzburgAustria
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
| |
Collapse
|
3
|
Jones OY. Single Center-Based Real-World Experience on Anti-IL 1 Biological Response Modifiers: A Case Series and Literature Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1146. [PMID: 39334678 PMCID: PMC11430789 DOI: 10.3390/children11091146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND This communication summarizes our single-center experience with the use of anti-IL-1 biologic response modifiers for treating autoimmune and autoinflammatory conditions in children. METHODS We outline our rationale for the off-label use of anakinra and discuss emerging treatment paradigms that necessitate further research and validation. RESULTS Anakinra has enabled personalized treatment, whether used as a single agent on an as-needed basis, as part of a background treatment regimen, or in combination with colchicine. Our data also highlight the significance of anakinra in treating post-infectious inflammatory diseases, demonstrating its high efficacy in novel applications such as rheumatic fever and post-viral arthritis. Canakinumab, on the other hand, has provided long-term remission. Both medications were well-tolerated, with no serious adverse effects reported. CONCLUSIONS Based on our observations and successful outcomes, we advocate for future collaborative efforts to improve access to anti-IL-1 medications to better manage excessive and harmful inflammation in children.
Collapse
Affiliation(s)
- Olcay Y Jones
- Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Department Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department Rheumatology, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
4
|
Dubinsky S, Hamadeh A, Imburgia C, McKnite A, Porter Hunt J, Wong K, Rice C, Rower J, Watt K, Edginton A. Physiologically Based Pharmacokinetic Modelling in Critically Ill Children Receiving Anakinra While on Extracorporeal Life Support. Clin Pharmacokinet 2024; 63:1343-1356. [PMID: 39331235 DOI: 10.1007/s40262-024-01424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND OBJECTIVE Because of the pathophysiological changes associated with critical illness and the use of extracorporeal life support (ECLS) such as continuous renal replacement therapy (CRRT) and extracorporeal membrane oxygenation (ECMO), the pharmacokinetics of drugs are often altered. The objective of this study was to develop a physiologically based pharmacokinetic (PBPK) model for anakinra in children that accounts for the physiological changes associated with critical illness and ECLS technology to guide appropriate pharmacotherapy. METHODS A PBPK model for anakinra was first developed in healthy individuals prior to extrapolating to critically ill children receiving ECLS. To account for the impact of anakinra clearance by the dialysis circuit, a CRRT compartment was added to the pediatric PBPK model and parameterized using data from a previously published ex-vivo study. Additionally, an ECMO compartment was added to the whole-body structure to create the final anakinra ECLS-PBPK model. The final model structure was validated by comparing predicted concentrations with observed patient data. Due to limited information in guiding anakinra dosing in this population, in-silico dose simulations were conducted to provide baseline recommendations. RESULTS By accounting for changes in physiology and the addition of ECLS compartments, the final ECLS-PBPK model predicted the observed plasma concentrations in an adolescent receiving subcutaneous anakinra. Furthermore, dosing simulations suggest that anakinra exposure in adolescents receiving ECLS is similar to that in healthy counterparts. CONCLUSION The anakinra ECLS-PBPK model developed in this study is the first to predict plasma concentrations in a population receiving simultaneous CRRT and ECMO. Dosing simulations provided may be used to inform anakinra use in critically ill children and guide future clinical trial planning.
Collapse
Affiliation(s)
- Samuel Dubinsky
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Abdullah Hamadeh
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Carina Imburgia
- Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Autumn McKnite
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - J Porter Hunt
- Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kristy Wong
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Cassandra Rice
- Center for Human Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Joseph Rower
- Center for Human Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Kevin Watt
- Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Andrea Edginton
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
5
|
Zeng R, He L, Kuang Z, Jian Y, Qiu M, Liu Y, Hu M, Ye Y, Wu L. Clinical characteristics, immunological alteration and distinction of MOG-IgG-associated disorders and GFAP-IgG-associated disorders. J Neuroimmunol 2024; 393:578398. [PMID: 39002186 DOI: 10.1016/j.jneuroim.2024.578398] [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: 05/09/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024]
Abstract
The classification of autoimmune encephalitis (AE) is based on the presence of different types of antibodies. Currently, the clinical manifestations and treatment regimens of patients with all types of AE exhibit similarities. However, the presence of immunological distinctions among different types of AE remains uncertain. In this study, we prospectively collected clinical data, as well as blood and cerebrospinal fluid (CSF) samples from patients diagnosed with MOG antibody-associated disease (MOGAD) or GFAP astrocytopathy (GFAP-A), in order to assess changes in inflammatory biomarkers such as immunoglobulin oligoclonal bands, cytokines in serum and CSF, as well as peripheral blood lymphocyte subtypes within different subsets. To further distinguish the immune response in patients with MOGAD and GFAP-A from that of healthy individuals, we prospectively recruited 20 hospitalized patients diagnosed with AE. Among them, 15 (75%) tested positive for MOG antibodies, 4 (20%) tested positive for GFAP antibodies, and 1 (5%) tested positive for both MOG and GFAP antibodies. These patients were then followed up for a period of 18 months. Compared to healthy controls (HC), AE patients exhibited elevated levels of MIP-1beta, SDF-1alpha, IL-12p70, IL-5, IL-1RA, IL-8 and decreased levels of IL-23, IL-31, IFN-alpha, IL-7, TNF-beta and TNF-alpha in serum. The CSF of AE patients showed increased levels of IL-1RA, IL-6 and IL-2 while decreased levels of RANTES, IL-18,IL-7,TNF-beta,TNF-alpha,RANTES,Eotaxin,and IL-9. The level of MCP-1 in the CSF of GFAP-A patients was found to be lower compared to that of MOGAD patients, while RANTES levels were higher. And the levels of IL-17A, Eotaxin, GRO-alpha, IL-8, IL-1beta, MIP-1beta were higher in the CSF of patients with epilepsy. The presence of intrathecal immune responses is also observed in patients with spinal muscular atrophy (SMA). However, no biomarker was found to be associated with disease severity in patients with AE. Among the 17 patients, recovery was observed, while 2 patients experienced persistent symptoms after an 18-month follow-up period. Additionally, within one year of onset, 8 patients had a single recurrence. Therefore, the immunological profiles of MOGAD and GFAP-A patients differ from those of normal individuals, and the alterations in cytokine levels may also exhibit a causal association with the clinical presentations, such as seizure.
Collapse
Affiliation(s)
- Rongrong Zeng
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | - Lu He
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | - Zhuo Kuang
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | - Yiemin Jian
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | | | - Yuting Liu
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | - Mengdie Hu
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | - Yizhi Ye
- The School of Pediatrics, Hengyang Medical School, University of South China (Hunan Children's Hospital), China
| | - Liwen Wu
- Hunan Children's Hospital, China.
| |
Collapse
|
6
|
Ozdemir Isik O, Karadag DT, Tekeoglu S, Yazici A, Cefle K, Cefle A. Long-term efficacy of canakinumab in hyperimmunoglobulin D syndrome. Int J Rheum Dis 2024; 27:e14857. [PMID: 37578023 DOI: 10.1111/1756-185x.14857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/17/2023] [Accepted: 07/22/2023] [Indexed: 08/15/2023]
Abstract
Hyperimmunoglobulin D syndrome (HIDS) is a rare autoinflammatory disorder with autosomal recessive inheritance. It is caused by specific mutations in the mevalonate kinase gene (MVK). No treatment specific to HIDS has been approved to date; however, nonsteroidal anti-inflammatory drugs, steroids, colchicine, tumor necrosis factor-α inhibitors, and anti-interleukin-1 treatments are used, based on case reports and observational studies. Herein, we report a case with recurrent fever and arthritis attacks who did not respond to anakinra and was successfully treated with canakinumab. Long-term remission was achieved without any side effects with 300 mg canakinumab treatment every 4 weeks for 5 years.
Collapse
Affiliation(s)
- Ozlem Ozdemir Isik
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Duygu Temiz Karadag
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Senem Tekeoglu
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Ayten Yazici
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Kıvanc Cefle
- Division of Medical Genetics, Department of Internal Medicine, School of Medicine, Istanbul University, Istanbul, Turkey
| | - Ayse Cefle
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
7
|
Li Z, Jing X, Zhang S, Liu T, Guo Q. Tumor necrosis factor receptor-associated cycle syndrome: a case report and literature review. Front Pediatr 2023; 11:1296487. [PMID: 38155741 PMCID: PMC10753791 DOI: 10.3389/fped.2023.1296487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
Recurring episodes of fever characterize tumor necrosis factor receptor-associated periodic syndrome (TRAPS) which is autosomal dominant. The primary symptoms of patients with TRAPS include prolonged fever, abdominal pain, muscle pain, and skin rashes. The prevalence of TRAPS appeared higher in Western countries than in Asian countries. Herein, we present the case of a 13-year-old girl who experienced intermittent fever for 8 years, with episodes that occur every 2 years. The patient demonstrated periodic fever, headache, vomiting, rash, and elevated inflammatory marker levels during the disease course. A heterozygous C55Y mutation was identified via a direct DNA sequencing of her genomic DNA. This mutation is located in exon 4 of TNFRSF1A. Genetic studies of her sister and mother revealed that they possessed the C55Y heterozygous mutation without demonstrating any clinical signs, while the father did not. Further, we conducted a thorough assessment of the literature and compiled the information from the eight TRAPS case series.
Collapse
Affiliation(s)
- Ziwei Li
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoping Jing
- Department of Traditional Chinese Medicine, School of Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuya Zhang
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tiantian Liu
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qingyin Guo
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
8
|
Foiadelli T, Santangelo A, Costagliola G, Costa E, Scacciati M, Riva A, Volpedo G, Smaldone M, Bonuccelli A, Clemente AM, Ferretti A, Savasta S, Striano P, Orsini A. Neuroinflammation and status epilepticus: a narrative review unraveling a complex interplay. Front Pediatr 2023; 11:1251914. [PMID: 38078329 PMCID: PMC10703175 DOI: 10.3389/fped.2023.1251914] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/11/2023] [Indexed: 12/09/2024] Open
Abstract
Status epilepticus (SE) is a medical emergency resulting from the failure of the mechanisms involved in seizure termination or from the initiation of pathways involved in abnormally prolonged seizures, potentially leading to long-term consequences, including neuronal death and impaired neuronal networks. It can eventually evolve to refractory status epilepticus (RSE), in which the administration of a benzodiazepine and another anti-seizure medications (ASMs) had been ineffective, and super-refractory status epilepticus (SRSE), which persists for more than 24 h after the administration of general anesthesia. Objective of the present review is to highlight the link between inflammation and SE. Several preclinical and clinical studies have shown that neuroinflammation can contribute to seizure onset and recurrence by increasing neuronal excitability. Notably, microglia and astrocytes can promote neuroinflammation and seizure susceptibility. In fact, inflammatory mediators released by glial cells might enhance neuronal excitation and cause drug resistance and seizure recurrence. Understanding the molecular mechanisms of neuroinflammation could be crucial for improving SE treatment, wich is currently mainly addressed with benzodiazepines and eventually phenytoin, valproic acid, or levetiracetam. IL-1β signal blockade with Anakinra has shown promising results in avoiding seizure recurrence and generalization in inflammatory refractory epilepsy. Inhibiting the IL-1β converting enzyme (ICE)/caspase-1 is also being investigated as a possible target for managing drug-resistant epilepsies. Targeting the ATP-P2X7R signal, which activates the NLRP3 inflammasome and triggers inflammatory molecule release, is another avenue of research. Interestingly, astaxanthin has shown promise in attenuating neuroinflammation in SE by inhibiting the ATP-P2X7R signal. Furthermore, IL-6 blockade using tocilizumab has been effective in RSE and in reducing seizures in patients with febrile infection-related epilepsy syndrome (FIRES). Other potential approaches include the ketogenic diet, which may modulate pro-inflammatory cytokine production, and the use of cannabidiol (CBD), which has demonstrated antiepileptic, neuroprotective, and anti-inflammatory properties, and targeting HMGB1-TLR4 axis. Clinical experience with anti-cytokine agents such as Anakinra and Tocilizumab in SE is currently limited, although promising. Nonetheless, Etanercept and Rituximab have shown efficacy only in specific etiologies of SE, such as autoimmune encephalitis. Overall, targeting inflammatory pathways and cytokines shows potential as an innovative therapeutic option for drug-resistant epilepsies and SE, providing the chance of directly addressing its underlying mechanisms, rather than solely focusing on symptom control.
Collapse
Affiliation(s)
- T. Foiadelli
- Clinica Pediatrica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A. Santangelo
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - G. Costagliola
- Pediatric Oncology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - E. Costa
- Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - M. Scacciati
- Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - A. Riva
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health, IRCCS Istituto “G. Gaslini”, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - G. Volpedo
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health, IRCCS Istituto “G. Gaslini”, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - M. Smaldone
- Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - A. Bonuccelli
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - A. M. Clemente
- Clinica Pediatrica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A. Ferretti
- Pediatrics Unit, Neuroscience, Mental Health and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - S. Savasta
- Pediatric Clinic and Rare Disease Microcitemico Hospital, University of Cagliari, Cagliari, Italy
| | - P. Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health, IRCCS Istituto “G. Gaslini”, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - A. Orsini
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| |
Collapse
|
9
|
Martins FR, Azevedo AC, Ganhão S, Aguiar F, Rodrigues M, Brito I. Anakinra and hepatotoxicity in pediatric rheumatology: a case series. Pediatr Rheumatol Online J 2023; 21:112. [PMID: 37803456 PMCID: PMC10559407 DOI: 10.1186/s12969-023-00891-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/04/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Anakinra is a recombinant interleukin-1 (IL-1) receptor antagonist used in systemic juvenile idiopathic arthritis (sJIA), refractory Kawasaki disease (KD) and cryopyrin-associated autoinflammatory syndrome (CAPS). Anakinra associated hepatotoxicity, while rare, has been described in several cases in daily practice. In this case series the authors describe three pediatric patients with this side effect in the setting of severe macrophage activation syndrome (MAS) in KD and sJIA. CASE PRESENTATION The first patient was a 12-year-old boy who presented with fever, maculo-papular exanthema and polyarthralgia. Tonsillitis, distal limb induration and tender cervical lymph nodes were observed. Erythrocyte-sedimentation rate (ESR), C-reactive protein (CRP), ferritin (11,975 ng/mL), D-dimers (5,98 mg/L FEU) and soluble CD25 (3645 pg/mL) levels were elevated. Exclusion of sepsis / toxic shock syndrome warranted introduction of IV methylprednisolone and immunoglobulin (IG IV), with partial response. A MAS secondary to KD was assumed, and anakinra 2 mg/kg/day was introduced. Twenty days later he developed new-onset nausea and severe cyto-cholestasis, normalizing after 2 months of drug discontinuation. Posterior onset of polyarthritis and evanescent lead to a final diagnosis of sJIA. The second patient was a 2-year-old boy with a 10-day history of fevers, generalized rash, hepatosplenomegaly and strawberry tongue. Leucocytosis with neutrophilia and elevated CRP were observed. Initial treatment with IVIG in the setting of incomplete KD was ineffective. Mild anaemia, leukopenia and very high serum ferritin (maximum 26,128 ng/mL) ensued. Presumptive sJIA associated MAS was treated with IV methylprednisolone and anakinra 2 mg/kg/day, with prompt response. Four weeks later transaminitis was detected, and temporary anakinra suspension led to normalisation of laboratorial values. The third case related to a 4-year-old boy presenting with fever, maculopapular rash and cervical lymphadenopathy. CRP and ESR were elevated, and KD was diagnosed. IVIG and methylprednisolone were initiated with clinical worsening, warranting for anakinra introduction at 2 mg/kg/day. After three weeks, liver enzymes progressively elevated, resolving on 2 weeks of anakinra discontinuation. CONCLUSIONS To the best of our knowledge, this is the first case series describing anakinra associated hepatotoxicity in pediatric patients with rheumatic diseases other than sJIA, bringing additional insight to therapeutic monitoring in patients undergoing this treatment.
Collapse
Affiliation(s)
| | - André Costa Azevedo
- Pediatrics Department, Local Health Unit Alto Minho, Viana do Castelo, Portugal
| | - Sara Ganhão
- Pediatric and Young Adult Rheumatology Unit, University Hospital Centre São João, Porto, Portugal
| | - Francisca Aguiar
- Pediatric and Young Adult Rheumatology Unit, University Hospital Centre São João, Porto, Portugal
| | - Mariana Rodrigues
- Pediatric and Young Adult Rheumatology Unit, University Hospital Centre São João, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Iva Brito
- Pediatric and Young Adult Rheumatology Unit, University Hospital Centre São João, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Zhao J, Huh Y, Bortsov A, Diatchenko L, Ji RR. Immunotherapies in chronic pain through modulation of neuroimmune interactions. Pharmacol Ther 2023; 248:108476. [PMID: 37307899 PMCID: PMC10527194 DOI: 10.1016/j.pharmthera.2023.108476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/18/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
It is generally believed that immune activation can elicit pain through production of inflammatory mediators that can activate nociceptive sensory neurons. Emerging evidence suggests that immune activation may also contribute to the resolution of pain by producing distinct pro-resolution/anti-inflammatory mediators. Recent research into the connection between the immune and nervous systems has opened new avenues for immunotherapy in pain management. This review provides an overview of the most utilized forms of immunotherapies (e.g., biologics) and highlight their potential for immune and neuronal modulation in chronic pain. Specifically, we discuss pain-related immunotherapy mechanisms that target inflammatory cytokine pathways, the PD-L1/PD-1 pathway, and the cGAS/STING pathway. This review also highlights cell-based immunotherapies targeting macrophages, T cells, neutrophils and mesenchymal stromal cells for chronic pain management.
Collapse
Affiliation(s)
- Junli Zhao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Andrey Bortsov
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC H3A 0G4, Canada; Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0G4, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
11
|
Dizon BLP, Redmond C, Gotschlich EC, Sule S, Ronis T, Vazzana KM, Sherman MA, Connor R, Bosk A, Dham N, Harahsheh AS, Wells E, DeBiasi R, Srinivasalu H. Clinical outcomes and safety of anakinra in the treatment of multisystem inflammatory syndrome in children: a single center observational study. Pediatr Rheumatol Online J 2023; 21:76. [PMID: 37525200 PMCID: PMC10388456 DOI: 10.1186/s12969-023-00858-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Evidence for the treatment of multisystem inflammatory syndrome in children (MIS-C) is lacking. Anakinra, which targets IL-1-mediated inflammation, is reserved for refractory cases of MIS-C; however, its use in the treatment of MIS-C is not clearly established. PATIENTS AND METHODS To examine a role for anakinra in MIS-C, we performed a single center observational cohort study of all MIS-C patients diagnosed at our children's hospital from May 15 to November 15, 2020. Demographics, clinical features, diagnostic testing, and cardiac function parameters were compared between MIS-C patients treated with intravenous immunoglobulin (IVIG) monotherapy and IVIG with anakinra (IVIG + anakinra). RESULTS Among 46 patients with confirmed MIS-C, 32 (70%) were in the IVIG + anakinra group, of which 9 (28%) were also given corticosteroids (CS). No patients were treated with anakinra alone. MIS-C patients in the IVIG + anakinra group were enriched in a CV shock phenotype (p = 0.02), and those with CV shock were treated with higher doses of anakinra for a longer duration. Furthermore, MIS-C patients in the IVIG + anakinra group exhibited improvements in fever and cardiac function with or without CS. No significant adverse events were observed, and no differences in IL-1β levels were found among MIS-C patients in the IVIG + anakinra group. CONCLUSIONS Anakinra treatment, which was co-administered with IVIG primarily in patients with severe MIS-C, was associated with improvements in fever and cardiac function, and demonstrated a favorable side-effect profile. These findings suggest a role for adjunctive anakinra in the treatment of severe MIS-C.
Collapse
Affiliation(s)
- Brian L P Dizon
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
- Rheumatology Fellowship and Training Branch, The National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Christopher Redmond
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
- Rheumatology Fellowship and Training Branch, The National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Emily C Gotschlich
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
| | - Sangeeta Sule
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Tova Ronis
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Kathleen M Vazzana
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
- Department of Pediatric Rheumatology, Arnold Palmer Hospital for Children, Orlando, FL, USA
| | - Matthew A Sherman
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
| | - Rachael Connor
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
| | - Abigail Bosk
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Niti Dham
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
| | - Ashraf S Harahsheh
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
| | - Elizabeth Wells
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
- Division of Neurology, Children's National Hospital, Washington, DC, USA
| | - Roberta DeBiasi
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
- Division of Infectious Diseases, Children's National Hospital, Washington, DC, USA
| | - Hemalatha Srinivasalu
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA.
- Department of Pediatrics, George Washington University School of Medicine & Health Sciences, Washington, DC, USA.
| |
Collapse
|
12
|
Mahmoudi Azar L, Karaman E, Beyaz B, Göktan I, Eyüpoğlu AE, Kizilel S, Erman B, Gül A, Uysal S. Expression and characterization of recombinant IL-1Ra in Aspergillus oryzae as a system. BMC Biotechnol 2023; 23:15. [PMID: 37340430 PMCID: PMC10283290 DOI: 10.1186/s12896-023-00785-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND The interleukin-1 receptor antagonist (IL-1Ra) is a crucial molecule that counteracts the effects of interleukin-1 (IL-1) by binding to its receptor. A high concentration of IL-1Ra is required for complete inhibition of IL-1 activity. However, the currently available Escherichia coli-expressed IL-1Ra (E. coli IL-1Ra, Anakinra) has a limited half-life. This study aims to produce a cost-effective, functional IL-1Ra on an industrial scale by expressing it in the pyrG auxotroph Aspergillus oryzae. RESULTS We purified A. oryzae-expressed IL-1Ra (Asp. IL-1Ra) using ion exchange and size exclusion chromatography (53 mg/L). Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis revealed that Asp. IL-1Ra is N-glycosylated and approximately 17 kDa in size. We conducted a comparative study of the bioactivity, binding kinetics, and half-life between Asp. IL-1Ra and E. coli IL-1Ra. Asp. IL-1Ra showed good bioactivity even at a low concentration of 0.5 nM. The in vitro half-life of Asp. IL-1Ra was determined for different time points (0, 24, 48, 72, and 96 h) and showed higher stability than E. coli IL-1Ra, despite exhibiting a 100-fold lower binding affinity (2 nM). CONCLUSION This study reports the production of a functional Asp. IL-1Ra with advantageous stability, without extensive downstream processing. To our knowledge, this is the first report of a recombinant functional and stable IL-1Ra expressed in A. oryzae. Our results suggest that Asp. IL-1Ra has potential for industrial-scale production as a cost-effective alternative to E. coli IL-1Ra.
Collapse
Affiliation(s)
- Lena Mahmoudi Azar
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, Istanbul, 34820, Turkey
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, 10010, USA
- NYU Pain Research Center, New York University College of Dentistry, New York, 10010, USA
| | - Elif Karaman
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, Istanbul, 34820, Turkey
- Department of Biotechnology, Institute of Health Sciences, Bezmialem Vakif University, Fatih, Istanbul, 34093, Turkey
| | - Burcu Beyaz
- Chemical and Biological Engineering, Koç University, Sariyer, Istanbul, 34450, Turkey
| | - Işılay Göktan
- Chemical and Biological Engineering, Koç University, Sariyer, Istanbul, 34450, Turkey
| | - Alp Ertunga Eyüpoğlu
- Department of of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, Istanbul, Sarıyer, 34450, Turkey
| | - Seda Kizilel
- Chemical and Biological Engineering, Koç University, Sariyer, Istanbul, 34450, Turkey
| | - Batu Erman
- Department of of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bogazici University, Istanbul, Sarıyer, 34450, Turkey
| | - Ahmet Gül
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul, 34134, Turkey
| | - Serdar Uysal
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, Istanbul, 34820, Turkey.
| |
Collapse
|
13
|
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
|
14
|
Adam Z, Šedivá A, Zeman D, Fojtík Z, Petrášová H, Diatková J, Tomíška M, Král Z, Treglerová J, Peřina V, Kamaradová K, Adamová Z, Pour L. Successful treatment of SAPHO syndrome (chronic nonbacterial osteomyelitis and acne) with anakinra and denosumab. Case report and review of therapy. VNITRNI LEKARSTVI 2023; 69:4-14. [PMID: 37827817 DOI: 10.36290/vnl.2023.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
SAPHO is an acronym derived from capital letters of Synovitis, Acne, Pustulosis, Hyperostosis, and Osteitis (SAPHO). SAPHO syndrome is an umbrella term covering a constellation of bone lesions and skin manifestations. A 40-year-old male complained about his jaw and back pain, swelling of multiple joints and weight loss accompanied by physical deterioration and acne type skin lesions. Laboratory tests revealed abnormal elevation of inflammatory markers. Imaging studies illustrated multiple osteolytic bone lesions and paraosseal infiltrates. According to the set of criteria diagnosis of SAPHO syndrome was stated. The patient was treated with glucocorticoids and non-steroidal anti-inflammatory drugs (NSAIDs), but only high dose dexamethasone and prednisone were effective. Daily subcutaneous administration of anakinra at the dose of 100 mg was initiated due to limited response to more classical therapies. Because of planned mandibular osteosynthesis initiation of denosumab was preferred before bisphosphonates. Therapeutic response was confirmed by FDG-PET/MR after 5 months of anakinra and denosumab therapy, showing decreased accumulation of FDG in periosteal and paraosseal infiltrates. Inflammatory markers significantly decreased, bone pain deferred but skin manifestation receded only partially. Therefore the response was evaluated as partial remission.
Collapse
|
15
|
Nasonov EL. [Role of interleukin-1 in human diseases: pharmacotherapy prospects: A review]. TERAPEVT ARKH 2022; 94:999-1005. [PMID: 36286981 DOI: 10.26442/00403660.2022.08.201781] [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: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
According to current concepts, human immunoinflammatory diseases (IIDs), depending on the prevailing mechanisms of immunopathogenesis, are divided into two main categories: autoimmune and autoinflammatory. At the same time, both autoimmune and autoinflammatory mechanisms are involved in the pathogenesis of most IIDs, and the complex interaction of these mechanisms is reflected in the polymorphism of clinical presentation, course variants, outcomes and therapy efficacy. It is suggested that in IIDs, overproduction of cytokines of the interleukin (IL)-1 family, which is one of the key regulators of innate immunity, determines the "crossing" between autoinflammation and autoimmunity mechanisms. Currently, anakinra, a recombinant non-glycosylated analog of the IL-1 receptor antagonist that blocks both IL-1b and IL-1a signaling, and canakinumab, a monoclonal antibody to IL-1b, are used in clinical practice to inhibit the pathological effects of IL-1. Analysis of the treatment outcomes with these drugs suggests that IL-1 inhibition should be considered a promising direction of pharmacotherapy of systemic autoinflammatory diseases and critical conditions associated with hyperinflammation in children and adults.
Collapse
Affiliation(s)
- E L Nasonov
- Nasonova Research Institute of Rheumatology
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
16
|
Harahsheh AS, Krishnan A, DeBiasi RL, Olivieri LJ, Spurney C, Donofrio MT, Cross RR, Sharron MP, Frank LH, Berul CI, Christopher A, Dham N, Srinivasalu H, Ronis T, Smith KL, Kline JN, Parikh K, Wessel D, Bost JE, Litt S, Austin A, Zhang J, Sable CA. Cardiac echocardiogram findings of severe acute respiratory syndrome coronavirus-2-associated multi-system inflammatory syndrome in children. Cardiol Young 2022; 32:718-726. [PMID: 34348808 PMCID: PMC8816963 DOI: 10.1017/s1047951121003024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND A novel paediatric disease, multi-system inflammatory syndrome in children, has emerged during the 2019 coronavirus disease pandemic. OBJECTIVES To describe the short-term evolution of cardiac complications and associated risk factors in patients with multi-system inflammatory syndrome in children. METHODS Retrospective single-centre study of confirmed multi-system inflammatory syndrome in children treated from 29 March, 2020 to 1 September, 2020. Cardiac complications during the acute phase were defined as decreased systolic function, coronary artery abnormalities, pericardial effusion, or mitral and/or tricuspid valve regurgitation. Patients with or without cardiac complications were compared with chi-square, Fisher's exact, and Wilcoxon rank sum. RESULTS Thirty-nine children with median (interquartile range) age 7.8 (3.6-12.7) years were included. Nineteen (49%) patients developed cardiac complications including systolic dysfunction (33%), valvular regurgitation (31%), coronary artery abnormalities (18%), and pericardial effusion (5%). At the time of the most recent follow-up, at a median (interquartile range) of 49 (26-61) days, cardiac complications resolved in 16/19 (84%) patients. Two patients had persistent mild systolic dysfunction and one patient had persistent coronary artery abnormality. Children with cardiac complications were more likely to have higher N-terminal B-type natriuretic peptide (p = 0.01), higher white blood cell count (p = 0.01), higher neutrophil count (p = 0.02), severe lymphopenia (p = 0.05), use of milrinone (p = 0.03), and intensive care requirement (p = 0.04). CONCLUSION Patients with multi-system inflammatory syndrome in children had a high rate of cardiac complications in the acute phase, with associated inflammatory markers. Although cardiac complications resolved in 84% of patients, further long-term studies are needed to assess if the cardiac abnormalities (transient or persistent) are associated with major cardiac events.
Collapse
Affiliation(s)
- Ashraf S Harahsheh
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Anita Krishnan
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Roberta L DeBiasi
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Microbiology, Immunology and Tropical Medicine, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Infectious Diseases, Children's National Hospital, Washington, DC, USA
| | - Laura J Olivieri
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Christopher Spurney
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Mary T Donofrio
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Russell R Cross
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Matthew P Sharron
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Critical Care Medicine, Children's National Hospital, Washington, DC, USA
| | - Lowell H Frank
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Charles I Berul
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Adam Christopher
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
| | - Niti Dham
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - Hemalatha Srinivasalu
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
| | - Tova Ronis
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Rheumatology, Children's National Hospital, Washington, DC, USA
| | - Karen L Smith
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Hospitalist Medicine, Children's National Hospital, Washington, DC, USA
| | - Jaclyn N Kline
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
| | - Kavita Parikh
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Hospitalist Medicine, Children's National Hospital, Washington, DC, USA
| | - David Wessel
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| | - James E Bost
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
- Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Sarah Litt
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
| | - Ashley Austin
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
| | - Jing Zhang
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
| | - Craig A Sable
- Division of Cardiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University, School of Medicine & Health Sciences, Washington, DC, USA
| |
Collapse
|
17
|
Fingerhutová Š, Jančová E, Doležalová P. Anakinra in Paediatric Rheumatology and Periodic Fever Clinics: Is the Higher Dose Safe? Front Pediatr 2022; 10:823847. [PMID: 35321008 PMCID: PMC8936593 DOI: 10.3389/fped.2022.823847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/14/2022] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Anakinra has been increasingly used in off-label indications as well as dosing and mode of administration in a variety of inflammatory conditions. We aimed to review our clinical practice and compare treatment outcomes with published data. METHODS Clinical data from electronic records were retrospectively reviewed for patients treated with anakinra over the past 6 years for autoinflammatory diseases (AID). RESULTS From 47 eligible patients (27 female patients), 32 were children. Macrophage activation syndrome (MAS) was the indication for anakinra therapy in 42.6% of patients. Systemic juvenile idiopathic arthritis (SJIA) was the most common underlying diagnosis (19/47) followed by the spectrum of AID. Off-label use was noted in 38.3% patients. Recommended dose was exceeded in 21 children (mean induction dose 5.1, highest dose 29.4 mg/kg/day) and two adults; five patients were treated intravenously. The mean treatment duration for SJIA was 1.4 years, that for AID was 2.2 years, and that for patients with higher anakinra dose was 9.7 (19.3) months. The mean follow-up duration was 2.7 (1.7) years. Treatment was effective in the majority of SJIA and cryopyrinopathy patients as well as those with MAS. Anakinra was well-tolerated without any major adverse effects even in patients with long-term administration of higher than recommended doses including two infants treated with a dose of over 20 mg/kg/day. CONCLUSION Our results support early use of anakinra in the individually tailored dosing. In patients with hyperinflammation, anakinra may be lifesaving and may even allow for corticosteroid avoidance. Further studies are needed in order to set up generally accepted response parameters and define condition-specific optimal dosing regimen.
Collapse
Affiliation(s)
- Šárka Fingerhutová
- Centre for Paediatric Rheumatology and Autoinflammatory Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Eva Jančová
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Pavla Doležalová
- Centre for Paediatric Rheumatology and Autoinflammatory Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|