101
|
A biomarker panel for risk of early respiratory failure following hematopoietic cell transplantation. Blood Adv 2022; 6:1866-1878. [PMID: 35139145 PMCID: PMC8941462 DOI: 10.1182/bloodadvances.2021005770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/22/2022] [Indexed: 11/20/2022] Open
Abstract
This study identified and validated ST2, WFDC2, IL-6, and TNFR1 as risk biomarkers for RF and related mortality post-HCT.
Plasma biomarkers associated with respiratory failure (RF) following hematopoietic cell transplantation (HCT) have not been identified. Therefore, we aimed to validate early (7 and 14 days post-HCT) risk biomarkers for RF. Using tandem mass spectrometry, we compared plasma obtained at day 14 post-HCT from 15 patients with RF and 15 patients without RF. Six candidate proteins, from this discovery cohort or identified in the literature, were measured by enzyme-linked immunosorbent assay in day-7 and day-14 post-HCT samples from the training (n = 213) and validation (n = 119) cohorts. Cox proportional-hazard analyses with biomarkers dichotomized by Youden’s index, as well as landmark analyses to determine the association between biomarkers and RF, were performed. Of the 6 markers, Stimulation-2 (ST2), WAP 4-disulfide core domain protein 2 (WFDC2), interleukin-6 (IL-6), and tumor necrosis factor receptor 1 (TNFR1), measured at day 14 post-HCT, had the most significant association with an increased risk for RF in the training cohort (ST2: hazard ratio [HR], 4.5, P = .004; WFDC2: HR, 4.2, P = .010; IL-6: HR, 6.9, P < .001; and TFNR1: HR, 6.1, P < .001) and in the validation cohort (ST2: HR, 23.2, P = .013; WFDC2: HR, 18.2, P = .019; IL-6: HR, 12.2, P = .014; and TFNR1: HR, 16.1, P = .001) after adjusting for the conditioning regimen. Using cause-specific landmark analyses, including days 7 and 14, high plasma levels of ST2, WFDC2, IL-6, and TNFR1 were associated with an increased HR for RF in the training and validation cohorts. These biomarkers were also predictive of mortality from RF. ST2, WFDC2, IL-6 and TNFR1 levels measured early posttransplantation improve risk stratification for RF and its related mortality.
Collapse
|
102
|
Kumar A, Sharma A, Tirpude NV, Sharma S, Padwad YS, Kumar S. Pharmaco-immunomodulatory interventions for averting cytokine storm-linked disease severity in SARS-CoV-2 infection. Inflammopharmacology 2022; 30:23-49. [PMID: 35048262 PMCID: PMC8769772 DOI: 10.1007/s10787-021-00903-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
The year 2020 is characterised by the COVID-19 pandemic that has quelled more than half a million lives in recent months. We are still coping with the negative repercussions of COVID-19 pandemic in 2021, in which the 2nd wave in India resulted in a high fatality rate. Regardless of emergency vaccine approvals and subsequent meteoric global vaccination drives in some countries, hospitalisations for COVID-19 will continue to occur due to the propensity of mutation in SARS-CoV-2 virus. The immune response plays a vital role in the control and resolution of infectious diseases. However, an impaired immune response is responsible for the severity of the respiratory distress in many diseases. The severe COVID-19 infection persuaded cytokine storm that has been linked with acute respiratory distress syndrome (ARDS), culminates into vital organ failures and eventual death. Thus, safe and effective therapeutics to treat hospitalised patients remains a significant unmet clinical need. In that state, any clue of possible treatments, which save patients life, can be treasured for this time point. Many cohorts and clinical trial studies demonstrated that timely administration of immunomodulatory drugs on severe COVID-19 patients may mitigate the disease severity, hospital stay and mortality. This article addresses the severity and risk factors of hypercytokinemia in COVID-19 patients, with special emphasis on prospective immunomodulatory therapies.
Collapse
Affiliation(s)
- Arbind Kumar
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, Himachal Pradesh India
| | - Aashish Sharma
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, Himachal Pradesh India
| | - Narendra Vijay Tirpude
- Animal Facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, Himachal Pradesh India
| | - Suresh Sharma
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, Himachal Pradesh India
| | - Yogendra S. Padwad
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, Himachal Pradesh India
| | - Sanjay Kumar
- CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, Himachal Pradesh India
| |
Collapse
|
103
|
Zhou W, Chen W, Wan X, Luo C, Du X, Li X, Chen Q, Gao R, Zhang X, Xie M, Wang M. Benefits of Chimeric Antigen Receptor T-Cell Therapy for B-Cell Lymphoma. Front Genet 2022; 12:815679. [PMID: 35126471 PMCID: PMC8811184 DOI: 10.3389/fgene.2021.815679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Objective: The aim was to study the benefits and risks of anti-CD19 chimeric antigen receptor (CAR) T-cells in adults with B-cell lymphoma. Methods: From October 2015 to October 2021, we treated five patients with B-cell lymphoma, comprising two with mantle cell lymphoma, one case of Burkitt lymphoma, one case of diffuse large B-cell lymphoma, and one case of chronic lymphocytic leukemia/small lymphocytic lymphoma. The patients were given the FC regimen 5 days before the infusion of anti-CD19 CAR T-cells. The median total number of CAR T-cells infusions was 350*10^6 (88*10^6–585*10^6). Results: 1) Patients who received CAR T-cell induction therapy achieved complete remission (CR) in Case 1 and Case 3 and partial remission (PR) in Case 2. Case 3’s ATM and D13S25 gene deletions were negative 42 days after CAR T-cell therapy, and molecular biology CR (mCR) and minimal residual disease (MRD) were negative for 5 years and 6 months. The patient in Case 3 was cured. 2) Case 4 patient’s TP53 gene mutation became negative 1 month after CAR T-cell therapy. MRD was negative after CAR T-cell therapy at 41 and 42 months in Cases 4 and 5, respectively. 3) Case 1∼Case 3 patients developed cytokine release syndrome (CRS) without encephalopathy syndrome, accompanied with serious adverse events. CRS can be effectively managed with tocilizumab, etanercept, glucocorticoids, and plasmapheresis. Conclusion: Anti-CD19 CAR T-cell therapy is effective in treating relapsed/refractory B-cell lymphoma, and the side effects of CAR T-cell therapy can be properly managed. CAR T-cell therapy has high efficacy and presented no side effects in the treatment of MRD in B-cell lymphoma (NCT03685786, NCT02456350).
Collapse
Affiliation(s)
- Wenyujing Zhou
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
| | - Weihong Chen
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
- *Correspondence: Weihong Chen,
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changru Luo
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xin Du
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xiaoqing Li
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
| | - Qian Chen
- Shenzhen BinDeBioTech Co., Shenzhen, China
| | - Ruiwen Gao
- Research Management and Supporting Department, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaohan Zhang
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
| | - Mei Xie
- Department of Hematology, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People’s Hospital, Shenzhen, China
| | - Mingjun Wang
- Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| |
Collapse
|
104
|
Talwar D, Kumar S, Acharya S, Raisinghani N, Madaan S, Hulkoti V, Akhilesh A, Khanna S, Shah D, Nimkar S. Interleukin 6 and Its Correlation with COVID-19 in Terms of Outcomes in an Intensive Care Unit of a Rural Hospital: A Cross-sectional Study. Indian J Crit Care Med 2022; 26:39-42. [PMID: 35110842 PMCID: PMC8783243 DOI: 10.5005/jp-journals-10071-24075] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Methods Result Conclusion How to cite this article
Collapse
Affiliation(s)
- Dhruv Talwar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Sunil Kumar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
- Sunil Kumar, Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India, e-mail:
| | - Sourya Acharya
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Nitin Raisinghani
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Sparsh Madaan
- Department of Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Vidyashree Hulkoti
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Annadatha Akhilesh
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Shivam Khanna
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Divit Shah
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| | - Shubham Nimkar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Medical Sciences (Deemed to be University), Wardha, Maharashtra, India
| |
Collapse
|
105
|
The differential effects of tumor burdens on predicting the net benefits of ssCART-19 cell treatment on r/r B-ALL patients. Sci Rep 2022; 12:378. [PMID: 35013456 PMCID: PMC8748521 DOI: 10.1038/s41598-021-04296-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
The tumor burden (TB) is significantly related to the severity of cytokine release syndrome (CRS) caused by CAR-T cells, but its correlation with therapeutic efficacy has not been systematically studied. This study focused on the effects of the TB level on both the safety and efficacy of ssCART-19 as a treatment for r/r B-ALL. Taking the 5% tumor burden as the boundary, the study participants were divided into 2 groups, high and low tumor burden groups. Under this grouping strategy, the impacts of differential r/r B-ALL TBs on the clinical therapeutic efficacy (CR rate and long-term survival) and safety profiles after ssCART-19 cell treatment were analysed. 78 patients were reported in this study. The differential B-ALL TBs significantly affected the complete remission (CR) rates of patients treated with ssCART-19, with rates of 93.94% and 75.56% in the low and high TB groups, respectively (P = 0.0358). The effects of TBs on long-term therapeutic efficacy were further studied based on event-free survival (EFS) and overall survival (OS) profiles; both the OS and EFS of the low TB group were better than those of the high TB group, but the differences were not statistically significant. Importantly, the time points of TB measurement did not significantly affect the OS and EFS profiles regardless of whether the TBs were measured before or after fludarabine-cyclophosphamide (FC) preconditional chemotherapy. On the other hand, the severity of CRS was significantly correlated with the TB level (P = 0.0080), and the incidence of sCRS was significantly related to the TB level (the sCRS incidence increased as the TB level increased, P = 0.0224). Unexpectedly, the ssCART-19 cell expansion peaks were not significantly different (P = 0.2951) between the study groups. Patients with a low r/r B-ALL TB yield more net benefits from CAR-T treatment than those with a high TB in terms of safety and CR rate. These findings are critical and valuable for determining the optimal CAR-T cell treatment window for r/r B-ALL patients and will further the development of comprehensive and reasonable CAR-T cell treatment plans for r/r B-ALL patients with differential TBs.Trial registration: ClinicalTrials.gov identifier, NCT03919240.
Collapse
|
106
|
Mirzaee Godarzee M, Mahmud Hussen B, Razmara E, Hakak‐Zargar B, Mohajerani F, Dabiri H, Fatih Rasul M, Ghazimoradi MH, Babashah S, Sadeghizadeh M. Strategies to overcome the side effects of chimeric antigen receptor T cell therapy. Ann N Y Acad Sci 2022; 1510:18-35. [DOI: 10.1111/nyas.14724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/05/2021] [Accepted: 10/22/2021] [Indexed: 11/26/2022]
Affiliation(s)
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy Hawler Medical University Erbil Iraq
| | - Ehsan Razmara
- Australian Regenerative Medicine Institute Monash University, Clayton, Victoria, Australia, 3800
| | | | - Fatemeh Mohajerani
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| | - Hamed Dabiri
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| | - Mohammed Fatih Rasul
- Department of Medical Analysis, Faculty of Sciences Tishk International University Erbil Iraq
| | | | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| |
Collapse
|
107
|
de Groot PM, Arevalo O, Shah K, Strange CD, Shroff GS, Ahuja J, Truong MT, de Groot JF, Vlahos I. Imaging Primer on Chimeric Antigen Receptor T-Cell Therapy for Radiologists. Radiographics 2022; 42:176-194. [PMID: 34990326 DOI: 10.1148/rg.210065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a recently approved breakthrough treatment that has become a new paradigm in treatment of recurrent or refractory B-cell lymphomas and pediatric or adult acute lymphoid leukemia. CAR T cells are a type of cellular immunotherapy that artificially enhances T cells to boost eradication of malignancy through activation of the native immune system. The CAR construct is a synthetically created functional cell receptor grafted onto previously harvested patient T cells, which bind to preselected tumor-associated antigens and thereby activate host immune signaling cascades to attack tumor cells. Advantages include a single treatment episode of 2-3 weeks and durable disease elimination, with remission rates of over 80%. Responses to therapy are more rapid than with conventional chemotherapy or immunotherapy, with intervening short-interval edema. CAR T-cell administration is associated with therapy-related toxic effects in a large percentage of patients, notably cytokine release syndrome, immune effect cell-associated neurotoxicity syndrome, and infections related to immunosuppression. Knowledge of the expected evolution of therapy response and potential adverse events in CAR T-cell therapy and correlation with the timeline of treatment are important to optimize patient care. Some toxic effects are radiologically evident, and familiarity with their imaging spectrum is key to avoiding misinterpretation. Other clinical toxic effects may be occult at imaging and are diagnosed on the basis of clinical assessment. Future directions for CAR T-cell therapy include new indications and expanded tumor targets, along with novel ways to capture T-cell activation with imaging. An invited commentary by Ramaiya and Smith is available online. Online supplemental material is available for this article. ©RSNA, 2022.
Collapse
Affiliation(s)
- Patricia M de Groot
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Octavio Arevalo
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Komal Shah
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Chad D Strange
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Girish S Shroff
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Jitesh Ahuja
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Mylene T Truong
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - John F de Groot
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Ioannis Vlahos
- From the Departments of Thoracic Imaging (P.M.d.G., C.D.S., G.S.S., J.A., M.T.T., I.V.), Neuroradiology (O.A., K.S.), and Neuro-oncology (J.F.d.G.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| |
Collapse
|
108
|
Gu T, Hu K, Si X, Hu Y, Huang H. Mechanisms of immune effector cell-associated neurotoxicity syndrome after CAR-T treatment. WIREs Mech Dis 2022; 14:e1576. [PMID: 35871757 PMCID: PMC9787013 DOI: 10.1002/wsbm.1576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 06/05/2022] [Accepted: 06/22/2022] [Indexed: 12/30/2022]
Abstract
Chimeric antigen receptor T-cell (CAR-T) treatment has revolutionized the landscape of cancer therapy with significant efficacy on hematologic malignancy, especially in relapsed and refractory B cell malignancies. However, unexpected serious toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) still hamper its broad application. Clinical trials using CAR-T cells targeting specific antigens on tumor cell surface have provided valuable information about the characteristics of ICANS. With unclear mechanism of ICANS after CAR-T treatment, unremitting efforts have been devoted to further exploration. Clinical findings from patients with ICANS strongly indicated existence of overactivated peripheral immune response followed by endothelial activation-induced blood-brain barrier (BBB) dysfunction, which triggers subsequent central nervous system (CNS) inflammation and neurotoxicity. Several animal models have been built but failed to fully replicate the whole spectrum of ICANS in human. Hopefully, novel and powerful technologies like single-cell analysis may help decipher the precise cellular response within CNS from a different perspective when ICANS happens. Moreover, multidisciplinary cooperation among the subjects of immunology, hematology, and neurology will facilitate better understanding about the complex immune interaction between the peripheral, protective barriers, and CNS in ICANS. This review elaborates recent findings about ICANS after CAR-T treatment from bed to bench, and discusses the potential cellular and molecular mechanisms that may promote effective management in the future. This article is categorized under: Cancer > Biomedical Engineering Immune System Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Tianning Gu
- Bone Marrow Transplantation Centerthe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310003China,Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina,Institute of HematologyZhejiang UniversityHangzhou310058China,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Kejia Hu
- Bone Marrow Transplantation Centerthe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310003China,Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina,Institute of HematologyZhejiang UniversityHangzhou310058China,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Xiaohui Si
- Bone Marrow Transplantation Centerthe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310003China,Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina,Institute of HematologyZhejiang UniversityHangzhou310058China,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Yongxian Hu
- Bone Marrow Transplantation Centerthe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310003China,Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina,Institute of HematologyZhejiang UniversityHangzhou310058China,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - He Huang
- Bone Marrow Transplantation Centerthe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310003China,Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina,Institute of HematologyZhejiang UniversityHangzhou310058China,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| |
Collapse
|
109
|
Natrajan K, Raval P, Panakkal B, Joshi D, Shukla A, Dubey G, Sahoo S, Bajpai S. A retrospective analysis of moderate and severe COVID-19 pneumonia with special reference to the use of Adjuvant Tocilizumab. ARCHIVES OF MEDICINE AND HEALTH SCIENCES 2022. [DOI: 10.4103/amhs.amhs_266_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
110
|
Gatti M, Pea F. The Cytokine Release Syndrome and/or the Proinflammatory Cytokines as Underlying Mechanisms of Downregulation of Drug Metabolism and Drug Transport: A Systematic Review of the Clinical Pharmacokinetics of Victim Drugs of this Drug-Disease Interaction Under Different Clinical Conditions. Clin Pharmacokinet 2022; 61:1519-1544. [PMID: 36059001 PMCID: PMC9441320 DOI: 10.1007/s40262-022-01173-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVE An ever-growing body of evidence supports the impact of cytokine modulation on the patient's phenotypic drug response. The aim of this systematic review was to analyze the clinical studies that assessed the pharmacokinetics of victim drugs of this drug-disease interaction in the presence of different scenarios of cytokine modulation in comparison with baseline conditions. METHODS We conducted a systematic review by searching the PubMed-MEDLINE database from inception until February 2022 to retrieve prospective and/or retrospective observational studies, population pharmacokinetic studies, phase I studies, and/or case series/reports that investigated the impact of cytokine modulation on the pharmacokinetic behavior of victim drugs. Only studies providing quantitative pharmacokinetic data of victim drugs by comparing normal status versus clinical conditions with documented cytokine modulation or by assessing the influence of anti-inflammatory biological agents on metabolism and/or transport of victim drugs were included. RESULTS Overall, 26 studies were included. Rheumatoid arthritis (6/26; 23.1%) and sepsis (5/26; 19.2%) were the two most frequently investigated pro-inflammatory clinical scenarios. The victim drug most frequently assessed was midazolam (14/26; 53.8%; as a probe for cytochrome P450 [CYP] 3A4). Cytokine modulation showed a moderate inhibitory effect on CYP3A4-mediated metabolism (area under the concentration-time curve increase and/or clearance decrease between 1.98-fold and 2.59-fold) and a weak-to-moderate inhibitory effect on CYP1A2, CYP2C9, and CYP2C19-mediated metabolism (in the area under the concentration-time curve increase or clearance decrease between 1.29-fold and 1.97-fold). Anti-interleukin-6 agents showed remarkable activity in counteracting downregulation of CYP3A4-mediated activity (increase in the area under the concentration-time curve between 1.75-fold and 2.56-fold). CONCLUSIONS Cytokine modulation may cause moderate or weak-to-moderate downregulation of metabolism/transport of victim drugs, and this may theoretically have relevant clinical consequences.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy ,Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy ,Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| |
Collapse
|
111
|
Castillo RL, Gonzaléz-Candia A, Candia AA. Pathophysiology of Acute Respiratory Failure by CoV-2 Infection: Role of Oxidative Stress, Endothelial Dysfunction and Obesity. Open Respir Med J 2021. [DOI: 10.2174/1874306402115010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) due to CoV-2 (coronavirus type 2) virus possess a particular risk of developing acute respiratory distress syndrome (ARDS) or SARS (severe acute respiratory syndrome coronavirus 2)-CoV2 in people with pre-existing conditions related to endothelial dysfunction and increased pro-inflammatory and pro-oxidant state. In between these conditions, chronic systemic inflammation related to obese patients is associated with the development of atherosclerosis, type 2 diabetes, and hypertension, comorbidities that adversely affect the clinical outcome in critical patients with COVID-19. Obesity affects up to 40% of the general population in the USA and more than 30% of the adult population in Chile. Until April 2021, 1,019,478 people have been infected, with 23,524 deaths. Given the coexistence of this worldwide obesity epidemic, COVID-19 negative outcomes are seriously enhanced in the current scenario. On the other hand, obesity is characterized by endothelial dysfunction observed in different vascular beds, an alteration which can be associated with impaired vasodilation, oxidative stress, and inflammatory events. Emerging evidence shows that obesity-related conditions such as endothelial dysfunction are associated with detrimental outcomes for COVID-19 evolution, especially if the patient derives to Intensive Care Units (ICU). This implies the need to understand the pathophysiology of the infection in the obese population, in order to propose therapeutic alternatives and public health policies, especially if the virus remains in the population. In this review, we summarize evidence about the pathogeny of Cov-2 infection in obese individuals and discuss how obesity-associated inflammatory and prooxidant status increase the severity of COVID-19.
Collapse
|
112
|
Dracocephalum moldavica Ethanol Extract Suppresses LPS-Induced Inflammatory Responses through Inhibition of the JNK/ERK/NF-κB Signaling Pathway and IL-6 Production in RAW 264.7 Macrophages and in Endotoxic-Treated Mice. Nutrients 2021; 13:nu13124501. [PMID: 34960054 PMCID: PMC8706341 DOI: 10.3390/nu13124501] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
The excessive synthesis of interleukin-6 (IL-6) is related to cytokine storm in COVID-19 patients. Moreover, blocking IL-6 has been suggested as a treatment strategy for inflammatory diseases such as sepsis. Sepsis is a severe systemic inflammatory response syndrome with high mortality. In the present study, we investigated the anti-inflammatory and anti-septic effects and the underlying mechanisms of Dracocephalum moldavica ethanol extract (DMEE) on lipopolysaccharide (LPS)-induced inflammatory stimulation in RAW 264.7 macrophages along with septic mouse models. We found that DMEE suppressed the release of inflammatory mediators NO and PGE2 and inhibited both the mRNA and protein expression levels of iNOS and COX-2, respectively. In addition, DMEE reduced the release of proinflammatory cytokines, mainly IL-6 and IL-1β, in RAW 264.7 cells by inhibiting the phosphorylation of JNK, ERK and p65. Furthermore, treatment with DMEE increased the survival rate and decreased the level of IL-6 in plasma in LPS-induced septic shock mice. Our findings suggest that DMEE elicits an anti-inflammatory effect in LPS-stimulated RAW 264.7 macrophages and an anti-septic effect on septic mouse model through the inhibition of the ERK/JNK/NF-κB signaling cascades and production of IL-6.
Collapse
|
113
|
Barbara I, Silvia M, Dario T, Giuseppe C, Stallone G. Case Report: Tocilizumab for Acute Kidney Graft Dysfunction in Patient Affected by COVID-19. Front Med (Lausanne) 2021; 8:732792. [PMID: 34901054 PMCID: PMC8655874 DOI: 10.3389/fmed.2021.732792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was isolated in January 2020 and, on March, the WHO declared the status of a pandemic. It causes a cytokine release syndrome, called “cytokine storm,” characterized by systemic inflammation involving elevated levels of cytokines and hyperactivation of immune cell; this profound alteration in the immune system led to an overshooting inflammatory response contributing to morbidity and mortality. Solid organ transplant recipients are at particularly higher risk of developing critical coronavirus disease 2019 (COVID-19) due to chronic immunosuppression; in fact, establishing the balance between infection and rejection in any transplant recipient is the principal aim when prescribing immunosuppression. Tocilizumab, a humanized monoclonal antibody against interleukin-6 (IL-6) receptor widely adopted in adult rheumatoid arthritis, is used as rescue therapy for chronic antibody-mediated rejection in kidney transplantation. Data about the use of tocilizumab for treating acute kidney graft dysfunction in a setting of kidney-transplanted patients affected by COVID-19 are lacking. In this case study, we discuss the case of kidney transplant recipient with proven SARS-CoV-2 infection that develops acute graft dysfunction and the management of immunosuppression with concomitant tocilizumab administration.
Collapse
Affiliation(s)
- Infante Barbara
- Nephrology Dialysis and Transplantation Unit, University of Foggia, Foggia, Italy
| | - Mercuri Silvia
- Nephrology Dialysis and Transplantation Unit, University of Foggia, Foggia, Italy
| | - Troise Dario
- Nephrology Dialysis and Transplantation Unit, University of Foggia, Foggia, Italy
| | - Castellano Giuseppe
- Nephrology Dialysis and Transplantation Unit, University of Foggia, Foggia, Italy.,Unit of Nephrology, Dialysis and Renal Transplantation-Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca'Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, University of Foggia, Foggia, Italy
| |
Collapse
|
114
|
Chakraborty C, Sharma AR, Bhattacharya M, Agoramoorthy G, Lee SS. The Drug Repurposing for COVID-19 Clinical Trials Provide Very Effective Therapeutic Combinations: Lessons Learned From Major Clinical Studies. Front Pharmacol 2021; 12:704205. [PMID: 34867318 PMCID: PMC8636940 DOI: 10.3389/fphar.2021.704205] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 has spread across the globe in no time. In the beginning, people suffered due to the absence of efficacious drugs required to treat severely ill patients. Nevertheless, still, there are no established therapeutic molecules against the SARS-CoV-2. Therefore, repurposing of the drugs started against SARS-CoV-2, due to which several drugs were approved for the treatment of COVID-19 patients. This paper reviewed the treatment regime for COVID-19 through drug repurposing from December 8, 2019 (the day when WHO recognized COVID-19 as a pandemic) until today. We have reviewed all the clinical trials from RECOVERY trials, ACTT-1 and ACTT-2 study group, and other major clinical trial platforms published in highly reputed journals such as NEJM, Lancet, etc. In addition to single-molecule therapy, several combination therapies were also evaluated to understand the treatment of COVID-19 from these significant clinical trials. To date, several lessons have been learned on the therapeutic outcomes for COVID-19. The paper also outlines the experiences gained during the repurposing of therapeutic molecules (hydroxychloroquine, ritonavir/ lopinavir, favipiravir, remdesivir, ivermectin, dexamethasone, camostatmesylate, and heparin), immunotherapeutic molecules (tocilizumab, mavrilimumab, baricitinib, and interferons), combination therapy, and convalescent plasma therapy to treat COVID-19 patients. We summarized that anti-viral therapeutic (remdesivir) and immunotherapeutic (tocilizumab, dexamethasone, and baricitinib) therapy showed some beneficial outcomes. Until March 2021, 4952 clinical trials have been registered in ClinicalTrials.gov toward the drug and vaccine development for COVID-19. More than 100 countries have participated in contributing to these clinical trials. Other than the registered clinical trials (medium to large-size), several small-size clinical trials have also been conducted from time to time to evaluate the treatment of COVID-19. Four molecules showed beneficial therapeutic to treat COVID-19 patients. The short-term repurposing of the existing drug may provide a successful outcome for COVID-19 patients. Therefore, more clinical trials can be initiated using potential anti-viral molecules by evaluating in different phases of clinical trials.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | | | | | - Sang-Soo Lee
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| |
Collapse
|
115
|
Cozzi-Lepri A, Smith C, Mussini C. Signals were broadly positive for months, but never definitive: the tocilizumab story. Clin Microbiol Infect 2021; 28:371-374. [PMID: 34768021 PMCID: PMC8576060 DOI: 10.1016/j.cmi.2021.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022]
Abstract
Background Most treatment guidelines for coronavirus disease 2019 (COVID-19) currently recommend tocilizumab in combination with dexamethasone in critically ill patients who are exhibiting rapid respiratory decompensation. Aims To produce a critical review and summary of the pathway which led to the repurposing of tocilizumab for COVID-19 treatment, from in vitro observations to guidelines recommendations. Sources All studies evaluating the effectiveness of tocilizumab to treat COVID-19 disease published between July 2020 and July 2021. Content Two large and methodologically well conducted observational studies, the TESEO and the STOP COVID cohorts, showed a reduction in the risk of invasive mechanical ventilation or death in patients treated with tocilizumab as compared to standard of care in 2020. Concomitantly, and up to February 2021, a number of randomized trials (RCTs) with small sample sizes were showing discrepant results. These RCTs had a number of issues: small sample size, various designs and inclusion criteria, and different dosages of tocilizumab used. The confidence interval of the meta-analytic estimate for the RCT results was consistent with the hypothesis of no efficacy of tocilizumab. In our opinion, this was mainly because the meta-analysis included small and heterogeneous studies. These results led to a delay in the inclusion of tocilizumab in guidelines which occurred only in the summer of 2021. Implications Although observational studies are unable to control for unmeasured confounding, they can be put together quickly during a pandemic and promptly provide important information. The large sample size allows us to investigate effect measure modifiers and to better target interventions. It is key that the effect size is somewhat large (RR > 2), all sources of bias are properly accounted for, and the direct evidence is weighted against these factors. It appears to us that for tocilizumab, not having dismissed the results of carefully designed and analysed observational studies in 2020 could have prevented many deaths over those months.
Collapse
Affiliation(s)
- Alessandro Cozzi-Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation (CREME), Institute for Global Health, UCL, London, UK.
| | - Colette Smith
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation (CREME), Institute for Global Health, UCL, London, UK
| | - Cristina Mussini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| |
Collapse
|
116
|
Tvedt THA, Vo AK, Bruserud Ø, Reikvam H. Cytokine Release Syndrome in the Immunotherapy of Hematological Malignancies: The Biology behind and Possible Clinical Consequences. J Clin Med 2021; 10:jcm10215190. [PMID: 34768710 PMCID: PMC8585070 DOI: 10.3390/jcm10215190] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cytokine release syndrome (CRS) is an acute systemic inflammatory syndrome characterized by fever and multiple organ dysfunction associated with (i) chimeric antigen receptor (CAR)-T cell therapy, (ii) therapeutic antibodies, and (iii) haploidentical allogeneic stem cell transplantation (haplo-allo-HSCT). Severe CRS can be life-threatening in some cases and requires prompt management of those toxicities and is still a great challenge for physicians. The pathophysiology of CRS is still not fully understood, which also applies to the identifications of predictive biomarkers that can forecast these features in advance. However, a broad range of cytokines are involved in the dynamics of CRS. Treatment approaches include both broad spectrum of immunosuppressant, such as corticosteroids, as well as more specific inhibition of cytokine release. In the present manuscript we will try to review an update regarding pathophysiology, etiology, diagnostics, and therapeutic options for this serious complication.
Collapse
Affiliation(s)
| | - Anh Khoi Vo
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
- Clinic for Medicine, Haukeland University Hospital, 5020 Bergen, Norway
| | - Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
- Clinic for Medicine, Haukeland University Hospital, 5020 Bergen, Norway
- Correspondence:
| |
Collapse
|
117
|
Gatto L, Franceschi E, Di Nunno V, Maggio I, Lodi R, Brandes AA. Engineered CAR-T and novel CAR-based therapies to fight the immune evasion of glioblastoma: gutta cavat lapidem. Expert Rev Anticancer Ther 2021; 21:1333-1353. [PMID: 34734551 DOI: 10.1080/14737140.2021.1997599] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The field of cancer immunotherapy has achieved great advancements through the application of genetically engineered T cells with chimeric antigen receptors (CAR), that have shown exciting success in eradicating hematologic malignancies and have proved to be safe with promising early signs of antitumoral activity in the treatment of glioblastoma (GBM). AREAS COVERED We discuss the use of CAR T cells in GBM, focusing on limitations and obstacles to advancement, mostly related to toxicities, hostile tumor microenvironment, limited CAR T cells infiltration and persistence, target antigen loss/heterogeneity and inadequate trafficking. Furthermore, we introduce the refined strategies aimed at strengthening CAR T activity and offer insights in to novel immunotherapeutic approaches, such as the potential use of CAR NK or CAR M to optimize anti-tumor effects for GBM management. EXPERT OPINION With the progressive wide use of CAR T cell therapy, significant challenges in treating solid tumors, including central nervous system (CNS) tumors, are emerging, highlighting early disease relapse and cancer cell resistance issues, owing to hostile immunosuppressive microenvironment and tumor antigen heterogeneity. In addition to CAR T cells, there is great interest in utilizing other types of CAR-based therapies, such as CAR natural killer (CAR NK) or CAR macrophages (CAR M) cells for CNS tumors.
Collapse
Affiliation(s)
- Lidia Gatto
- Medical Oncology Department, Azienda USL, Bologna, Italy
| | - Enrico Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, Bologna, Italy
| | | | - Ilaria Maggio
- Medical Oncology Department, Azienda USL, Bologna, Italy
| | - Raffaele Lodi
- IrcssIstituto di Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alba Ariela Brandes
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, Bologna, Italy
| |
Collapse
|
118
|
Kyriakopoulos C, Ntritsos G, Gogali A, Milionis H, Evangelou E, Kostikas K. Tocilizumab administration for the treatment of hospitalized patients with COVID-19: A systematic review and meta-analysis. Respirology 2021; 26:1027-1040. [PMID: 34605114 PMCID: PMC8661720 DOI: 10.1111/resp.14152] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022]
Abstract
Tocilizumab has been repurposed against the ‘cytokine storm’ in the setting of coronavirus disease 2019 (COVID‐19). Our aim was to evaluate the efficacy of tocilizumab in the management of hospitalized COVID‐19 patients. We searched MEDLINE, CENTRAL and medRxiv for studies of tocilizumab in hospitalized COVID‐19 patients. Primary objective was the effectiveness of tocilizumab on mortality. Secondary objectives included the need for invasive mechanical ventilation (IMV), composite endpoints of mortality or IMV and intensive care unit (ICU) admission or IMV, length of hospitalization and differences in mortality in subgroups (ICU and non‐ICU patients and patients receiving or not receiving concomitant corticosteroids). We included 52 studies (nine randomized controlled trials [RCTs] and 43 observational) with a total of 27,004 patients. In both RCTs and observational studies, the use of tocilizumab was associated with a reduction in mortality; 11% in RCTs (risk ratio [RR] 0.89, 95% CI 0.82 to 0.96) and 31% in observational studies (RR 0.69, 95% CI 0.58 to 0.83). The need for IMV was reduced by 19% in RCTs (RR 0.81, 95% CI 0.71 to 0.93), while no significant reduction was observed in observational studies. Both RCTs and observational studies showed a benefit from tocilizumab on the composite endpoint of mortality or IMV. Tocilizumab improved mortality both in ICU and non‐ICU patients. Reduction in mortality was evident in observational studies regardless of the use of systemic corticosteroids, while that was not the case in the RCTs. Tocilizumab was associated with lower mortality and other clinically relevant outcomes in hospitalized patients with moderate‐to‐critical COVID‐19.
Collapse
Affiliation(s)
- Christos Kyriakopoulos
- Respiratory Medicine Department, University of Ioannina Faculty of Medicine, Ioannina, Greece
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, University of Ioannina Faculty of Medicine, Ioannina, Greece
| | - Athena Gogali
- Respiratory Medicine Department, University of Ioannina Faculty of Medicine, Ioannina, Greece
| | - Haralampos Milionis
- Internal Medicine Department, University of Ioannina Faculty of Medicine, Ioannina, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Faculty of Medicine, Ioannina, Greece
| | - Konstantinos Kostikas
- Respiratory Medicine Department, University of Ioannina Faculty of Medicine, Ioannina, Greece
| |
Collapse
|
119
|
Collins KL, Younis US, Tanyaratsrisakul S, Polt R, Hay M, Mansour HM, Ledford JG. Angiotensin-(1-7) Peptide Hormone Reduces Inflammation and Pathogen Burden during Mycoplasma pneumoniae Infection in Mice. Pharmaceutics 2021; 13:1614. [PMID: 34683907 PMCID: PMC8539524 DOI: 10.3390/pharmaceutics13101614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
The peptide hormone, angiotensin (Ang-(1-7)), produces anti-inflammatory and protective effects by inhibiting production and expression of many cytokines and adhesion molecules that are associated with a cytokine storm. While Ang-(1-7) has been shown to reduce inflammation and airway hyperreactivity in models of asthma, little is known about the effects of Ang-(1-7) during live respiratory infections. Our studies were developed to test if Ang-(1-7) is protective in the lung against overzealous immune responses during an infection with Mycoplasma pneumonia (Mp), a common respiratory pathogen known to provoke exacerbations in asthma and COPD patients. Wild type mice were treated with infectious Mp and a subset of was given either Ang-(1-7) or peptide-free vehicle via oropharyngeal delivery within 2 h of infection. Markers of inflammation in the lung were assessed within 24 h for each set of animals. During Mycoplasma infection, one high dose of Ang-(1-7) delivered to the lungs reduced neutrophilia and Muc5ac, as well as Tnf-α and chemokines (Cxcl1) associated with acute respiratory distress syndrome (ARDS). Despite decreased inflammation, Ang-(1-7)-treated mice also had significantly lower Mp burden in their lung tissue, indicating decreased airway colonization. Ang-(1-7) also had an impact on RAW 264.7 cells, a commonly used macrophage cell line, by dose-dependently inhibiting TNF-α production while promoting Mp killing. These new findings provide additional support to the protective role(s) of Ang1-7 in controlling inflammation, which we found to be highly protective against live Mp-induced lung inflammation.
Collapse
Affiliation(s)
- Katie L. Collins
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA;
| | - Usir S. Younis
- Asthma and Airway Disease Research Center, Tucson, AZ 85724, USA; (U.S.Y.); (S.T.)
| | | | - Robin Polt
- Departments of Chemistry and Biochemistry, College of Science, The University of Arizona, Tucson, AZ 85721, USA;
| | - Meredith Hay
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA;
| | - Heidi M. Mansour
- BIO5 Institute, The University of Arizona, Tucson, AZ 85719, USA;
- Department of Medicine, Division of Translational & Regenerative Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Departments of Pharmacology/Toxicology and Pharmaceutical Sciences, College of Pharmacy, The University of Arizona, Tucson, AZ 85724, USA
| | - Julie G. Ledford
- Asthma and Airway Disease Research Center, Tucson, AZ 85724, USA; (U.S.Y.); (S.T.)
- BIO5 Institute, The University of Arizona, Tucson, AZ 85719, USA;
- Department of Cellular and Molecular Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
120
|
Meyer A, Yan S, Golumba-Nagy V, Esser RL, Barbarino V, Blakemore SJ, Rusyn L, Nikiforov A, Seeger-Nukpezah T, Grüll H, Pallasch CP, Kofler DM. Kinase activity profiling reveals contribution of G-protein signaling modulator 2 deficiency to impaired regulatory T cell migration in rheumatoid arthritis. J Autoimmun 2021; 124:102726. [PMID: 34555678 DOI: 10.1016/j.jaut.2021.102726] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/27/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022]
Abstract
The ability of regulatory T (Treg) cells to migrate into inflammatory sites is reduced in autoimmune diseases, including rheumatoid arthritis (RA). The reasons for impaired Treg cell migration remain largely unknown. We performed multiplex human kinase activity arrays to explore possible differences in the post-translational phosphorylation status of kinase related proteins that could account for altered Treg cell migration in RA. Results were verified by migration assays and Western blot analysis of CD4+ T cells from RA patients and from mice with collagen type II induced arthritis. Kinome profiling of CD4+ T cells from RA patients revealed significantly altered post-translational phosphorylation of kinase related proteins, including G-protein-signaling modulator 2 (GPSM2), protein tyrosine kinase 6 (PTK6) and vitronectin precursor (VTNC). These proteins have not been associated with RA until now. We found that GPSM2 expression is reduced in CD4+ T cells from RA patients and is significantly downregulated in experimental autoimmune arthritis following immunization of mice with collagen type II. Interestingly, GPSM2 acts as a promoter of Treg cell migration in healthy individuals. Treatment of RA patients with interleukin-6 receptor (IL-6R) blocking antibodies restores GPSM2 expression, thereby improving Treg cell migration. Our study highlights the potential of multiplex kinase activity arrays as a tool for the identification of RA-related proteins which could serve as targets for novel treatments.
Collapse
Affiliation(s)
- Anja Meyer
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Shuaifeng Yan
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Viktoria Golumba-Nagy
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ruth L Esser
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Verena Barbarino
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Stuart J Blakemore
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lisa Rusyn
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anastasia Nikiforov
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tamina Seeger-Nukpezah
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Grüll
- Institute of Diagnostic and Interventional Radiology, University Hospital Cologne, Germany
| | - Christian P Pallasch
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - David M Kofler
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany.
| |
Collapse
|
121
|
Endothelial Dysfunction, Inflammation, and Oxidative Stress in COVID-19-Mechanisms and Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8671713. [PMID: 34457119 PMCID: PMC8397545 DOI: 10.1155/2021/8671713] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023]
Abstract
The outbreak of the COVID-19 pandemic represents an ongoing healthcare emergency responsible for more than 3.4 million deaths worldwide. COVID-19 is the disease caused by SARS-CoV-2, a virus that targets not only the lungs but also the cardiovascular system. COVID-19 can manifest with a wide range of clinical manifestations, from mild symptoms to severe forms of the disease, characterized by respiratory failure due to severe alveolar damage. Several studies investigated the underlying mechanisms of the severe lung damage associated with SARS-CoV-2 infection and revealed that the respiratory failure associated with COVID-19 is the consequence not only of acute respiratory distress syndrome but also of macro- and microvascular involvement. New observations show that COVID-19 is an endothelial disease, and the consequent endotheliopathy is responsible for inflammation, cytokine storm, oxidative stress, and coagulopathy. In this review, we show the central role of endothelial dysfunction, inflammation, and oxidative stress in the COVID-19 pathogenesis and present the therapeutic targets deriving from this endotheliopathy.
Collapse
|
122
|
Chandrasekaran S, Funk CR, Kleber T, Paulos CM, Shanmugam M, Waller EK. Strategies to Overcome Failures in T-Cell Immunotherapies by Targeting PI3K-δ and -γ. Front Immunol 2021; 12:718621. [PMID: 34512641 PMCID: PMC8427697 DOI: 10.3389/fimmu.2021.718621] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
PI3K-δ and PI3K-γ are critical regulators of T-cell differentiation, senescence, and metabolism. PI3K-δ and PI3K-γ signaling can contribute to T-cell inhibition via intrinsic mechanisms and regulation of suppressor cell populations, including regulatory T-cells and myeloid derived suppressor cells in the tumor. We examine an exciting new role for using selective inhibitors of the PI3K δ- and γ-isoforms as modulators of T-cell phenotype and function in immunotherapy. Herein we review the current literature on the implications of PI3K-δ and -γ inhibition in T-cell biology, discuss existing challenges in adoptive T-cell therapies and checkpoint blockade inhibitors, and highlight ongoing efforts and future directions to incorporate PI3K-δ and PI3K-γ as synergistic T-cell modulators in immunotherapy.
Collapse
Affiliation(s)
- Sanjay Chandrasekaran
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Christopher Ronald Funk
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Troy Kleber
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Chrystal M. Paulos
- Department of Surgery/Microbiology & Immunology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| |
Collapse
|
123
|
Montini L, DE Pascale G, Bello G, Grieco DL, Grasselli G, Pesenti A, Ranieri VM, Cecconi M, D'Arcangelo E, Gremese E, Antonelli M. Compassionate use of anti-IL6 receptor antibodies in critically ill patients with acute respiratory distress syndrome due to SARS-COV-2. Minerva Anestesiol 2021; 87:1080-1090. [PMID: 34337913 DOI: 10.23736/s0375-9393.21.15609-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND COVID-19 patients developing the acute respiratory distress syndrome (ARDS) show increased production of pro-inflammatory cytokines, including interleukin-6 (IL-6). The use of humanized monoclonal antibody against interleukin-6 receptor (IL-6R) may represent a potential treatment strategy. We analyzed the effects of compassionate use of Tocilizumab and Sarilumab on clinical outcome of patients affected by ARDS due COVID-19. METHODS This single-center, observational, exploratory study was performed during the acute phase of COVID-19 outbreak, between March 7th and April 21st, 2020 in a University Hospital in Rome, Italy. All consecutive adult patients admitted to the intensive care unit with laboratory-confirmed COVID-19 and fulfilling ARDS criteria were enrolled. Patients who were treated with anti-IL-6R therapy were compared to those who were not, as per clinical decision. Inverse probability weights were applied to weight individual's contribution to survival curves and in the multivariate regression model. RESULTS Among 105 ARDS patients, 65 received compassionate treatment with anti-IL-6R therapy [43 (66%) Tocilizumab and 22 (34%) Sarilumab, respectively], with oxygenation improvement. In the multivariable Cox proportional regression hazards model with propensity score inverse probability weighting, patients who received anti-IL-6R treatment had lower risk of death compared to those who did not, with a hazard ration of 0.34 [95% confidence interval 0.17-0.74], p=0.001. CONCLUSIONS Our data suggest that immune modulator therapy based on anti-human IL-6 receptor monoclonal antibodies might lead to improved outcome in patients with ARDS due to COVID-19. These data support the need for confirmatory randomized trials to assess the effect of immune modulator therapies on mortality.
Collapse
Affiliation(s)
- Luca Montini
- Department of Intensive Care Medicine and Anesthesiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gennaro DE Pascale
- Department of Intensive Care Medicine and Anesthesiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Bello
- Department of Intensive Care Medicine and Anesthesiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico L Grieco
- Department of Intensive Care Medicine and Anesthesiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy - .,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giacomo Grasselli
- Dipartimento di Anestesia, Rianimazione ed Emergenza-Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Antonio Pesenti
- Dipartimento di Anestesia, Rianimazione ed Emergenza-Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Vito M Ranieri
- Anesthesia and Intensive Care Medicine, Policlinico di Sant'Orsola, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Maurizio Cecconi
- Department of Anaesthesia and Intensive Care Medicine, Humanitas Clinical and Research Centre-IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Enzo D'Arcangelo
- Department of Statistical Sciences, Sapienza University of Rome, Rome, Italy
| | - Elisa Gremese
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimo Antonelli
- Department of Intensive Care Medicine and Anesthesiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | | |
Collapse
|
124
|
Shang L, Lye DC, Cao B. Contemporary narrative review of treatment options for COVID-19. Respirology 2021; 26:745-767. [PMID: 34240518 PMCID: PMC8446994 DOI: 10.1111/resp.14106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is ongoing and many drugs have been studied in clinical trials. From a pathophysiological perspective, anti-viral drugs may be more effective in the early stage while immunomodulators may be more effective in severe patients in later stages of infection. While drugs such as lopinavir-ritonavir, hydroxychloroquine and azithromycin have proved to be ineffective in randomized controlled trials, corticosteroids, neutralizing monoclonal antibodies, remdesivir, tocilizumab and baricitinib have been reported to benefit certain groups of patients with COVID-19. In this review, we will present the key clinical evidence and progress in promising COVID-19 therapeutics, as well as summarize the experience and lessons learned from the development of the current therapeutics.
Collapse
Affiliation(s)
- Lianhan Shang
- Beijing University of Chinese MedicineBeijingChina
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory MedicineChina‐Japan Friendship HospitalBeijingChina
- Institute of Respiratory MedicineChinese Academy of Medical SciencesBeijingChina
| | - David Chien Lye
- Department of Infectious DiseasesTan Tock Seng HospitalSingapore
- National Centre for Infectious DiseasesSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Bin Cao
- Beijing University of Chinese MedicineBeijingChina
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory MedicineChina‐Japan Friendship HospitalBeijingChina
- Institute of Respiratory MedicineChinese Academy of Medical SciencesBeijingChina
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Respiratory MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
125
|
Constantinescu C, Pasca S, Tat T, Teodorescu P, Vlad C, Iluta S, Dima D, Tomescu D, Scarlatescu E, Tanase A, Sigurjonsson OE, Colita A, Einsele H, Tomuleasa C. Continuous renal replacement therapy in cytokine release syndrome following immunotherapy or cellular therapies? J Immunother Cancer 2021; 8:jitc-2020-000742. [PMID: 32474415 PMCID: PMC7264828 DOI: 10.1136/jitc-2020-000742] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Recently, an increasing number of novel drugs were approved in oncology and hematology. Nevertheless, pharmacology progress comes with a variety of side effects, of which cytokine release syndrome (CRS) is a potential complication of some immunotherapies that can lead to multiorgan failure if not diagnosed and treated accordingly. CRS generally occurs with therapies that lead to highly activated T cells, like chimeric antigen receptor T cells or in the case of bispecific T-cell engaging antibodies. This, in turn, leads to a proinflammatory state with subsequent organ damage. To better manage CRS there is a need for specific therapies or to repurpose strategies that are already known to be useful in similar situations. Current management strategies for CRS are represented by anticytokine directed therapies and corticosteroids. Based on its pathophysiology and the resemblance of CRS to sepsis and septic shock, as well as based on the principles of initiation of continuous renal replacement therapy (CRRT) in sepsis, we propose the rationale of using CRRT therapy as an adjunct treatment in CRS where all the other approaches have failed in controlling the clinically significant manifestations.
Collapse
Affiliation(s)
- Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Anesthesia - Intensive Care, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Tiberiu Tat
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Catalin Vlad
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Dana Tomescu
- Department of Anesthesia - Intensive Care, Carol Davila University of Medicine and Pharmacy, Bucuresti, Romania.,Department of Anesthesia - Intensive Care, Fundeni Clinical Institute, Bucuresti, Romania
| | - Ecaterina Scarlatescu
- Department of Stem Cell Transplantation, Clinical Institute Fundeni, Bucuresti, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Clinical Institute Fundeni, Bucuresti, Romania
| | - Olafur Eysteinn Sigurjonsson
- University of Reykjavik, Reykjavik, Iceland.,Bloodbank, Landspitali National University Hospital of Iceland, Reykjavik, Iceland
| | - Anca Colita
- Department of Stem Cell Transplantation, Clinical Institute Fundeni, Bucuresti, Romania
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Wurzburg, Bayern, Germany
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| |
Collapse
|
126
|
Wu C, Lin D, Ma F, Jiang F, Wang Y. New progress in elucidating the relationship between cancer therapy and cardiovascular toxicity. Biosci Trends 2021; 15:211-218. [PMID: 34305102 DOI: 10.5582/bst.2021.01278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Onco-cardiology is an emerging field linking cancer with cardiovascular injury. Understanding the mechanism of cardiac injury helps improve the quality of life of cancer survivors. A series of studies on adverse reactions to cancer or oncological treatments has indicated that adverse cardiovascular events related to cancer treatments may occur over a longer period of survival, and even years after therapy has concluded. Current cancer therapies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, have been found to have latent cardiovascular toxicity. These toxic effects are often progressive and irreversible and ultimately lead to cardiovascular events such as heart failure, hypertension, coronary heart diseases, arrhythmia, and thromboembolism. Therefore, more emphasis should be placed on revealing the mechanism of cancer treatment-related cardiovascular toxicity. This would help to guide prevention, diagnosis, and treatment of CVDs in cancer survivors.
Collapse
Affiliation(s)
- Chen Wu
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dawei Lin
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Ma
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Jiang
- Clinical Research & Innovation Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Yaosheng Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Clinical Research & Innovation Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
127
|
Yin Z, Zhang Y, Wang X. Advances in chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Biomark Res 2021; 9:58. [PMID: 34256851 PMCID: PMC8278776 DOI: 10.1186/s40364-021-00309-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
B-cell non-Hodgkin lymphoma (B-NHL) is a group of heterogeneous disease which remains incurable despite developments of standard chemotherapy regimens and new therapeutic agents in decades. Some individuals could have promising response to standard therapy while others are unresponsive to standard chemotherapy or relapse after autologous hematopoietic stem-cell transplantation (ASCT), which indicates the necessity to develop novel therapies for refractory or relapsed B-NHLs. In recent years, a novel cell therapy, chimeric antigen receptor T-cell therapy (CAR-T), was invented to overcome the limitation of traditional treatments. Patients with aggressive B-NHL are considered for CAR-T cell therapy when they have progressive lymphoma after second-line chemotherapy, relapse after ASCT, or require a third-line therapy. Clinical trials of anti-CD19 CAR-T cell therapy have manifested encouraging efficacy in refractory or relapsed B-NHL. However, adverse effects of this cellular therapy including cytokine release syndrome, neurotoxicity, tumor lysis syndrome and on-target, off-tumor toxicities should attract our enough attention despite the great anti-tumor effects of CAR-T cell therapy. Although CAR-T cell therapy has shown remarkable results in patients with B-NHL, the outcomes of patients with B-NHL were inferior to patients with acute lymphoblastic leukemia. The inferior response rate may be associated with physical barrier of lymphoma, tumor microenvironment and low quality of CAR-T cells manufactured from B-NHL patients. Besides, some patients relapsed after anti-CD19 CAR-T cell therapy, which possibly were due to limited CAR-T cells persistence, CD19 antigen escape or antigen down-regulation. Quite a few new antigen-targeted CAR-T products and new-generation CAR-T, for example, CD20-targeted CAR-T, CD79b-targeted CAR-T, CD37-targeted CAR-T, multi-antigen-targeted CAR-T, armored CAR-T and four-generation CAR-T are developing rapidly to figure out these deficiencies.
Collapse
Affiliation(s)
- Zixun Yin
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.,School of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
128
|
Cardiotoxicity associated with immune checkpoint inhibitors and CAR T-cell therapy. Am J Emerg Med 2021; 50:51-58. [PMID: 34274878 DOI: 10.1016/j.ajem.2021.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION The expanding use of immunotherapy and the growing population of patients with cancer has led to an increase in the reporting of immune related adverse events (irAEs). The emergency clinician should be aware of these emerging toxicities, some of which can be fatal. In this review we discuss the cardiotoxic side effects of immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell (CAR T-cell) therapy. DISCUSSION Recognizing the possible presentations of cardiotoxic irAEs is of utmost important as the diagnosis of cardiotoxicity associated with ICI and CAR T-cell can be difficult to make in the emergency department. The emergency clinician will have to presume the diagnosis and treat it without final confirmation in most cases. For this reason, if the diagnosis is suspected, early involvement of the cardiologist and oncologist is important to help guide management. Most irAEs will be treated with glucocorticoids, but in the case of CAR T-cell cardiotoxicity, Tocilizumab should be used as first line. CONCLUSION Although cardiotoxicity is rare, it is often life-threatening. Treatment should be initiated as soon as the diagnosis is suspected, and early involvement of the cardiologist and oncologist is imperative for optimal treatment.
Collapse
|
129
|
Hafezi B, Chan L, Knapp JP, Karimi N, Alizadeh K, Mehrani Y, Bridle BW, Karimi K. Cytokine Storm Syndrome in SARS-CoV-2 Infections: A Functional Role of Mast Cells. Cells 2021; 10:1761. [PMID: 34359931 PMCID: PMC8308097 DOI: 10.3390/cells10071761] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/27/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Cytokine storm syndrome is a cascade of escalated immune responses disposing the immune system to exhaustion, which might ultimately result in organ failure and fatal respiratory distress. Infection with severe acute respiratory syndrome-coronavirus-2 can result in uncontrolled production of cytokines and eventually the development of cytokine storm syndrome. Mast cells may react to viruses in collaboration with other cells and lung autopsy findings from patients that died from the coronavirus disease that emerged in 2019 (COVID-19) showed accumulation of mast cells in the lungs that was thought to be the cause of pulmonary edema, inflammation, and thrombosis. In this review, we present evidence that a cytokine response by mast cells may initiate inappropriate antiviral immune responses and cause the development of cytokine storm syndrome. We also explore the potential of mast cell activators as adjuvants for COVID-19 vaccines and discuss the medications that target the functions of mast cells and could be of value in the treatment of COVID-19. Recognition of the cytokine storm is crucial for proper treatment of patients and preventing the release of mast cell mediators, as impeding the impacts imposed by these mediators could reduce the severity of COVID-19.
Collapse
Affiliation(s)
- Bahareh Hafezi
- Department of Clinical Science, School of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad 9177948974, Iran; (B.H.); (N.K.)
| | - Lily Chan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Jason P. Knapp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Negar Karimi
- Department of Clinical Science, School of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad 9177948974, Iran; (B.H.); (N.K.)
| | - Kimia Alizadeh
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA;
| | - Yeganeh Mehrani
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| |
Collapse
|
130
|
Karki R, Kanneganti TD. The 'cytokine storm': molecular mechanisms and therapeutic prospects. Trends Immunol 2021; 42:681-705. [PMID: 34217595 DOI: 10.1016/j.it.2021.06.001] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022]
Abstract
Cytokine storm syndrome (CSS) has generally been described as a collection of clinical manifestations resulting from an overactivated immune system. Cytokine storms (CSs) are associated with various pathologies, as observed in infectious diseases, certain acquired or inherited immunodeficiencies and autoinflammatory diseases, or following therapeutic interventions. Despite the role of CS in tissue damage and multiorgan failure, a systematic understanding of its underlying molecular mechanisms is lacking. Recent studies demonstrate a positive feedback loop between cytokine release and cell death pathways; certain cytokines, pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), can activate inflammatory cell death, leading to further cytokine secretion. Here, we discuss recent progress in innate immunity and inflammatory cell death, providing insights into the cellular and molecular mechanisms of CSs and therapeutics that might quell ensuing life-threatening effects.
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
131
|
Kulanthaivel S, Kaliberdenko VB, Balasundaram K, Shterenshis MV, Scarpellini E, Abenavoli L. Tocilizumab in SARS-CoV-2 Patients with the Syndrome of Cytokine Storm: A Narrative Review. Rev Recent Clin Trials 2021; 16:138-145. [PMID: 32940187 DOI: 10.2174/1574887115666200917110954] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/15/2020] [Accepted: 08/11/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Corona virus is a group of viruses that cause diseases in mammals and birds. In humans, these families of viruses can cause respiratory infections from a mild form to fatal. It is preferably called coronavirus. Formally, it is known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or 2019 novel coronavirus (2019-nCoV) and this disease is called coronavirus disease 2019 (COVID-19). SARS-CoV-2 is infectious in humans and the world health organization has announced COVID-19 as pandemic disease. Tocilizumab is a biological agent that inhibits the cytokine, interleukin 6 (IL-6 inhibitor). As SARS-CoV-2 infection leads to the development of cytokine storm syndrome, the drug, tocilizumab, seems to have a positive effect on patients with COVID-19. AIM To analyze and review the possible effects and efficacy of the tocilizumab (monoclonal antibody against IL-6 receptors) in SARS-CoV-2 patients. MATERIALS AND METHODS A search was carried out for all recent review articles, which were used to study the SARS-CoV-2 disease and their characteristics. Furthermore, we have analyzed the most recent research articles on monoclonal antibody against IL-6 receptors (tocilizumab) and their possible clinical effects in COVID-19 and its' clinical trials. RESULTS COVID-19 is a disease caused by SARS-CoV-2 infection. It is a life threatening condition, which can give rise to fatal outcomes if left untreated. However, there are no approved treatments for COVID-19 globally. Furthermore, we can conclude that SARS-CoV-2 is associated with the worsening of lung conditions, characterized by interstitial pneumonia with acute respiratory distress syndrome as a result of cytokine storm syndrome. According to available research data, tocilizumab, a recombinant humanized anti-human monoclonal antibody of IgG1τ (gamma 1, kappa), can improve patient's condition from cytokine storm syndrome by inhibiting the IL-6 (Interleukin 6) receptors. CONCLUSION The rational use of the tocilizumab in severe and critically ill COVID-19 patients can prevent the development of irreversible lung injury and death of the patient. Three retrospective studies of Xiaoling Xu et al., Pan luo et al., and Paola Tonaiti et al. have shown the efficacy of tocilizumab in severe and critically ill COVID-19 patients. However, we need more randomized research studies with a significant number of patients which can confirm the promising results on tocilizumab treatment in COVID-19 patients. Moreover, ongoing clinical trails such as TOSCA, COVACTA results have not been published yet which are expected to give better and more significant results on tocilizumab's effectiveness and safety.
Collapse
Affiliation(s)
- Shanmugaraj Kulanthaivel
- Department of Internal Medicine No.2, V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Vitalii B Kaliberdenko
- Department of Internal Medicine No.2, V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Keerthanaa Balasundaram
- Department of Internal Medicine No.2, V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - Michael V Shterenshis
- Science Research Department, Alexander Muss Institute for Israel Education (AMIIE), Hod Hasharon, Israel
| | - Emidio Scarpellini
- Clinical Nutrition Unit and Internal Medicine Unit, Madonna del Soccorso General Hospital, Via Luciano Manara 7, 63074 San Benedetto del Tronto (AP), Italy
| | - Ludovico Abenavoli
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
132
|
Fischer JW, Bhattarai N. CAR-T Cell Therapy: Mechanism, Management, and Mitigation of Inflammatory Toxicities. Front Immunol 2021; 12:693016. [PMID: 34220853 PMCID: PMC8250150 DOI: 10.3389/fimmu.2021.693016] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Engineered T cell therapies such as chimeric antigen receptor (CAR) expressing T cells (CAR-T cells) have great potential to treat many human diseases; however, inflammatory toxicities associated with these therapies present safety risks and can greatly limit its widespread use. This article briefly reviews our current understanding of mechanisms for inflammatory toxicities during CAR T-cell therapy, current strategies for management and mitigation of these risks and highlights key areas of knowledge gap for future research.
Collapse
Affiliation(s)
- Joseph W Fischer
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Nirjal Bhattarai
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
133
|
Kobak S. The Perfect Storm: A Rheumatologist's Point of View on COVID-19 Infection. Curr Rheumatol Rev 2021; 17:141-152. [PMID: 33121412 DOI: 10.2174/1573397116666201029155105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/20/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022]
Abstract
The new coronavirus infection (Covid-19) is a pandemic that has affected the whole world and progresses with high morbidity and mortality. It has a high contagion rate and a course capable of rapid lung involvement with severe acute respiratory distress syndrome (ARDS) and pulmonary insufficiency. A severe clinical picture develops as a result of a "perfect cytokine storm" which results from possible immunological mechanisms triggered by the viral infection. Immune system dysregulation and possible autoinflammatory and autoimmune mechanisms are responsible for a higher amount of cytokines release from immune cells. Although no clear treatment of Covid-19 infection has emerged yet, it is argued that some disease-modifying anti-rheumatic drugs (DMARDs) may be effective in addition to anti-viral treatments. These drugs (anti-malarial drugs, colchicum dispert, biologics) have been well known to rheumatologists for years because they are used in the treatment of many inflammatory rheumatologic diseases. Another important issue is whether DMARDs, which can cause severe immunosuppression, pose a risk for Covid-19 infection and whether they have been discontinued beforehand. Although there are insufficient data on this subject, considering the risk of disease reactivation, patients may continue their DMARDs treatment under the supervision of a rheumatologist. In this article, the possible immunological mechanisms in the pathogenesis of Covid-19 infection and the efficacy and safety of various DMARDs used in the treatment are discussed from a rheumatologist's perspective in the light of recent literature data.
Collapse
Affiliation(s)
- Senol Kobak
- Department of Internal Medicine and Rheumatology, Faculty of Medicine, Istinye University, Liv Hospital, Istanbul, Turkey
| |
Collapse
|
134
|
Gu R, Mao T, Lu Q, Tianjiao Su T, Wang J. Myeloid dysregulation and therapeutic intervention in COVID-19. Semin Immunol 2021; 55:101524. [PMID: 34823995 PMCID: PMC8576142 DOI: 10.1016/j.smim.2021.101524] [Citation(s) in RCA: 10] [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: 09/05/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022]
Abstract
The dysregulation of myeloid cell responses is increasingly demonstrated to be a major mechanism of pathogenesis for COVID-19. The pathological cellular and cytokine signatures associated with this disease point to a critical role of a hyperactivated innate immune response in driving pathology. Unique immunopathological features of COVID-19 include myeloid-cell dominant inflammation and cytokine release syndrome (CRS) alongside lymphopenia and acute respiratory distress syndrome (ARDS), all of which correlate with severe disease. Studies suggest a range of causes mediating myeloid hyperactivation, such as aberrant innate sensing, asynchronized immune cellular responses, as well as direct viral protein/host interactions. These include the recent identification of new myeloid cell receptors that bind SARS-CoV-2, which drive myeloid cell hyperinflammatory responses independently of lung epithelial cell infection via the canonical receptor, angiotensin-converting enzyme 2 (ACE2). The spectrum and nature of myeloid cell dysregulation in COVID-19 also differs from, at least to some extent, what is observed in other infectious diseases involving myeloid cell activation. While much of the therapeutic effort has focused on preventative measures with vaccines or neutralizing antibodies that block viral infection, recent clinical trials have also targeted myeloid cells and the associated cytokines as a means to resolve CRS and severe disease, with promising but thus far modest effects. In this review, we critically examine potential mechanisms driving myeloid cell dysregulation, leading to immunopathology and severe disease, and discuss potential therapeutic strategies targeting myeloid cells as a new paradigm for COVID-19 treatment.
Collapse
Affiliation(s)
- Runxia Gu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Qiao Lu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, 10016, USA
| | - Tina Tianjiao Su
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, 10016, USA.
| |
Collapse
|
135
|
Mazzoni A, Salvati L, Maggi L, Annunziato F, Cosmi L. Hallmarks of immune response in COVID-19: Exploring dysregulation and exhaustion. Semin Immunol 2021; 55:101508. [PMID: 34728121 PMCID: PMC8547971 DOI: 10.1016/j.smim.2021.101508] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023]
Abstract
One and half year following the occurrence of COVID-19 pandemic, significant efforts from laboratories all over the world generated a huge amount of data describing the prototypical features of immunity in the course of SARS-CoV-2 infection. In this Review, we rationalize and organize the main observations, trying to define a "core" signature of immunity in COVID-19. We identified six hallmarks describing the main alterations occurring in the early infection phase and in the course of the disease, which predispose to severe illness. The six hallmarks are dysregulated type I IFN activity, hyperinflammation, lymphopenia, lymphocyte impairment, dysregulated myeloid response, and heterogeneous adaptive immunity to SARS-CoV-2. Dysregulation and exhaustion came out as the trait d'union, connecting abnormalities affecting both innate and adaptive immunity, humoral and cellular responses.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
136
|
Hahn VS, Zhang KW, Sun L, Narayan V, Lenihan DJ, Ky B. Heart Failure With Targeted Cancer Therapies: Mechanisms and Cardioprotection. Circ Res 2021; 128:1576-1593. [PMID: 33983833 DOI: 10.1161/circresaha.121.318223] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oncology has seen growing use of newly developed targeted therapies. Although this has resulted in dramatic improvements in progression-free and overall survival, challenges in the management of toxicities related to longer-term treatment of these therapies have also become evident. Although a targeted approach often exploits the differences between cancer cells and noncancer cells, overlap in signaling pathways necessary for the maintenance of function and survival in multiple cell types has resulted in systemic toxicities. In particular, cardiovascular toxicities are of important concern. In this review, we highlight several targeted therapies commonly used across a variety of cancer types, including HER2 (human epidermal growth factor receptor 2)+ targeted therapies, tyrosine kinase inhibitors, immune checkpoint inhibitors, proteasome inhibitors, androgen deprivation therapies, and MEK (mitogen-activated protein kinase kinase)/BRAF (v-raf murine sarcoma viral oncogene homolog B) inhibitors. We present the oncological indications, heart failure incidence, hypothesized mechanisms of cardiotoxicity, and potential mechanistic rationale for specific cardioprotective strategies.
Collapse
Affiliation(s)
- Virginia S Hahn
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD (V.S.H.)
| | - Kathleen W Zhang
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Lova Sun
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Vivek Narayan
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Bonnie Ky
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Cardiovascular Medicine (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Biostatistics (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
137
|
Sanjay S, Kawali A, Mahendradas P, Shetty R. Lockdown effects on a patient receiving immunosuppression for unilateral HLA- B27 associated uveitis during COVID-19 pandemic. Indian J Ophthalmol 2021; 69:1351-1353. [PMID: 33913911 PMCID: PMC8186619 DOI: 10.4103/ijo.ijo_3504_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Srinivasan Sanjay
- Department of Uveitis and Ocular Immunology, Narayana Nethralaya Super Speciality Hospital and Post-Graduate Institute of Ophthalmology, Bangalore, Karnataka, India
| | - Ankush Kawali
- Department of Uveitis and Ocular Immunology, Narayana Nethralaya Super Speciality Hospital and Post-Graduate Institute of Ophthalmology, Bangalore, Karnataka, India
| | - Padmamalini Mahendradas
- Department of Uveitis and Ocular Immunology, Narayana Nethralaya Super Speciality Hospital and Post-Graduate Institute of Ophthalmology, Bangalore, Karnataka, India
| | - Rohit Shetty
- Department of Neuro-Ophthalmology, Cornea and Refractive Surgery, Narayana Nethralaya Super Speciality Hospital and Post-Graduate Institute of Ophthalmology, Bangalore, Karnataka, India
| |
Collapse
|
138
|
Venuti A, Donzelli S, Nisticò P, Blandino G, Ciliberto G. Does Interleukin-6 Bridge SARS-CoV-2 With Virus-Associated Cancers? JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:79-85. [PMID: 35663529 PMCID: PMC9153257 DOI: 10.36401/jipo-20-27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/26/2020] [Accepted: 09/10/2021] [Indexed: 06/15/2023]
Abstract
To date SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), a member of the Coronaviridae family, has infected more than 40 million people worldwide. A second wave of SARS-CoV-2 infection is aggressively surging. The clinical worsening of SARS-CoV-2 infection appears to be strictly associated with comorbidities, which can be used to establish an intrinsic patient network whose molecular profile is pivotal for identifying and successfully treating populations at risk. Herein, we focus on the direct interaction between SARS-CoV-2 and virus-associated cancers, exploring the critical role of interleukin-6 (IL-6) as a mediator of this complex cross talk. IL-6 production is enhanced in diverse viral infections ranging from human papilloma virus (HPV) to hepatitis B virus (HBV), human immunodeficiency virus (HIV), and SARS-CoV-2 infection. High systemic levels of IL-6 are associated with viral persistence and poor clinical outcomes in SARS-CoV-2-infected patients. Blockade of IL-6/IL-6R, using specific molecules, is under investigation in active clinical trials for the treatment of patients with SARS-CoV-2. Although the data are as yet inconclusive, they pave the way for selective targeting of crucial cytokine-activated aberrant signaling in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Aldo Venuti
- HPV-Unit, UOSD (Simple Departmental Operational Unit) Tumor Immunology and Immunotherapy, IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Regina Elena National Cancer Institute, Rome, Italy
- UOSD Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- UOSD Tumor Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
139
|
Mieszkowski J, Borkowska A, Stankiewicz B, Kochanowicz A, Niespodziński B, Surmiak M, Waldziński T, Rola R, Petr M, Antosiewicz J. Single High-Dose Vitamin D Supplementation as an Approach for Reducing Ultramarathon-Induced Inflammation: A Double-Blind Randomized Controlled Trial. Nutrients 2021; 13:nu13041280. [PMID: 33924645 PMCID: PMC8069287 DOI: 10.3390/nu13041280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose: A growing number of studies indicate the importance of vitamin D supplementation for sports performance. However, the effects of a single high-dose vitamin D supplementation on ultramarathon-induced inflammation have not been investigated. We here analyzed the effect of a single high-dose vitamin D supplementation on the inflammatory marker levels in ultramarathon runners after an ultramarathon run (maximal run 240 km). Methods: In the study, 35 runners (amateurs) were assigned into two groups: single high-dose vitamin D supplementation group, administered vitamin D (150,000 IU) in vegetable oil 24 h before the start of the run (n = 16); and placebo group (n = 19). Blood was collected for analysis 24 h before, immediately after, and 24 h after the run. Results: Serum 25(OH)D levels were significantly increased after the ultramarathon in both groups. The increase was greater in the vitamin D group than in the control group. Based on post-hoc and other analyses, the increase in interleukin 6 and 10, and resistin levels immediately after the run was significantly higher in runners in the control group than that in those in the supplementation group. Leptin, oncostatin M, and metalloproteinase tissue inhibitor levels were significantly decreased in both groups after the run, regardless of the supplementation. Conclusions: Ultramarathon significantly increases the serum 25(OH)D levels. Attenuation of changes in interleukin levels upon vitamin D supplementation confirmed that vitamin D has anti-inflammatory effect on exercise-induced inflammation.
Collapse
Affiliation(s)
- Jan Mieszkowski
- Department of Gymnastics and Dance, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland;
- Faculty of Physical Education and Sport, Charles University, 162 52 Prague, Czech Republic;
- Correspondence: (J.M.); (J.A.); Tel.: +48-501-619-669 (J.M.); +48-513-046-412 (J.A.)
| | - Andżelika Borkowska
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, 85-064 Gdańsk, Poland;
| | - Błażej Stankiewicz
- Department of Biomedical Basis of Physical Education, Kazimierz Wielki University, Institute of Physical Education, 85-064 Bydgoszcz, Poland;
| | - Andrzej Kochanowicz
- Department of Gymnastics and Dance, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland;
| | - Bartłomiej Niespodziński
- Department of Anatomy and Biomechanics, Institute of Physical Education, Kazimierz Wielki University, 85-064 Bydgoszcz, Poland;
| | - Marcin Surmiak
- Department of Internal Medicine, Jagiellonian University Medical College, 31-007 Krakow, Poland;
| | - Tomasz Waldziński
- Faculty of Health Sciences, Łomża State University of Applied Science, 18-400 Łomża, Poland;
| | - Rafał Rola
- Masdiag Sp. Z O.O., 01-882 Warsaw, Poland;
- Chair of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, 87-100 Toruń, Poland
| | - Miroslav Petr
- Faculty of Physical Education and Sport, Charles University, 162 52 Prague, Czech Republic;
| | - Jędrzej Antosiewicz
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, 85-064 Gdańsk, Poland;
- Correspondence: (J.M.); (J.A.); Tel.: +48-501-619-669 (J.M.); +48-513-046-412 (J.A.)
| |
Collapse
|
140
|
Aldea M, Michot JM, Danlos FX, Ribas A, Soria JC. Repurposing of Anticancer Drugs Expands Possibilities for Antiviral and Anti-Inflammatory Discovery in COVID-19. Cancer Discov 2021; 11:1336-1344. [PMID: 33846172 DOI: 10.1158/2159-8290.cd-21-0144] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023]
Abstract
In 2020, the COVID-19 pandemic led to an unprecedented destabilization of the world's health and economic systems. The rapid spread and life-threatening consequences of COVID-19 have imposed testing of repurposed drugs, by investigating interventions already used in other indications, including anticancer drugs. The contours of anticancer drug repurposing have been shaped by similarities between the pathogenesis of COVID-19 and malignancies, including abnormal inflammatory and immunologic responses. In this review, we discuss the salient positive and negative points of repurposing anticancer drugs to advance treatments for COVID-19. SIGNIFICANCE: Targeting anti-inflammatory pathways with JAK/STAT inhibitors or anticytokine therapies aiming to curb COVID-19-related cytokine storm, using antiangiogenic drugs to reduce vascular abnormalities or immune-checkpoint inhibitors to improve antiviral defenses, could be of value in COVID-19. However, conflicting data on drug efficacy point to the need for better patient selection and biomarker studies.
Collapse
Affiliation(s)
- Mihaela Aldea
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | | | - Francois-Xavier Danlos
- INSERM U1015, Gustave Roussy, Villejuif, France.,Paris Saclay University, Saint-Aubin, France
| | - Antoni Ribas
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angles, Los Angeles, California
| | - Jean-Charles Soria
- Drug Development Department, Gustave Roussy, Villejuif, France. .,Paris Saclay University, Saint-Aubin, France
| |
Collapse
|
141
|
López-Medrano F, Pérez-Jacoiste Asín MA, Fernández-Ruiz M, Carretero O, Lalueza A, Maestro de la Calle G, Caro JM, de la Calle C, Catalán M, García-García R, Martínez-López J, Origüen J, Ripoll M, San Juan R, Trujillo H, Sevillano Á, Gutiérrez E, de Miguel B, Aguilar F, Gómez C, Silva JT, García-Ruiz de Morales D, Saro-Buendía M, Marrero-Sánchez Á, Chiara-Graciani G, Bueno H, Paz-Artal E, Lumbreras C, Pablos JL, Aguado JM. Combination therapy with tocilizumab and corticosteroids for aged patients with severe COVID-19 pneumonia: A single-center retrospective study. Int J Infect Dis 2021; 105:487-494. [PMID: 33647515 PMCID: PMC7908857 DOI: 10.1016/j.ijid.2021.02.099] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The role of combination immunomodulatory therapy with systemic corticosteroids and tocilizumab (TCZ) for aged patients with COVID-19-associated cytokine release syndrome remains unclear. METHODS A retrospective single-center study was conducted on consecutive patients aged ≥65 years who developed severe COVID-19 between 03 March and 01 May 2020 and were treated with corticosteroids at various doses (methylprednisolone 0.5mg/kg/12h to 250mg/24h), either alone (CS group) or associated with intravenous tocilizumab (400-600mg, one to three doses) (CS-TCZ group). The primary outcome was all-cause mortality by day +14, whereas secondary outcomes included mortality by day +28 and clinical improvement (discharge and/or a ≥2 point decrease on a 6-point ordinal scale) by day +14. Propensity score (PS)-based adjustment and inverse probability of treatment weights (IPTW) were applied. RESULTS Totals of 181 and 80 patients were included in the CS and CS-TCZ groups, respectively. All-cause 14-day mortality was lower in the CS-TCZ group, both in the PS-adjusted (hazard ratio [HR]: 0.34; 95% confidence interval [CI]: 0.17-0.68; P=0.002) and IPTW-weighted models (odds ratio [OR]: 0.38; 95% CI: 0.21-0.68; P=0.001). This protective effect was also observed for 28-day mortality (PS-adjusted HR: 0.38; 95% CI: 0.21-0.72; P=0.003). Clinical improvement by day +14 was higher in the CS-TCZ group with IPTW analysis only (OR: 2.26; 95% CI: 1.49-3.41; P<0.001). The occurrence of secondary infection was similar between both groups. CONCLUSIONS The combination of corticosteroids and TCZ was associated with better outcomes among patients aged ≥65 years with severe COVID-19.
Collapse
Affiliation(s)
- Francisco López-Medrano
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain; Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Spain.
| | - María Asunción Pérez-Jacoiste Asín
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Mario Fernández-Ruiz
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Octavio Carretero
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Antonio Lalueza
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Guillermo Maestro de la Calle
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José Manuel Caro
- Department of Pharmacy, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Cristina de la Calle
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Mercedes Catalán
- Department of Intensive Care Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Rocío García-García
- Department of Pneumology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Joaquín Martínez-López
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Spain; Department of Hematology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Julia Origüen
- Department of Emergency Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Mar Ripoll
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain; Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Spain
| | - Hernando Trujillo
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Ángel Sevillano
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Eduardo Gutiérrez
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Borja de Miguel
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Fernando Aguilar
- Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Carlos Gómez
- Department of Oncology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José Tiago Silva
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Daniel García-Ruiz de Morales
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Miguel Saro-Buendía
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Ángel Marrero-Sánchez
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Guillermo Chiara-Graciani
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Héctor Bueno
- Department of Cardiology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Carlos Lumbreras
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Spain; Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José L Pablos
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Spain; Department of Rheumatology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Department of Internal Medicine, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain; Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Spain
| |
Collapse
|
142
|
Ghosn L, Chaimani A, Evrenoglou T, Davidson M, Graña C, Schmucker C, Bollig C, Henschke N, Sguassero Y, Nejstgaard CH, Menon S, Nguyen TV, Ferrand G, Kapp P, Riveros C, Ávila C, Devane D, Meerpohl JJ, Rada G, Hróbjartsson A, Grasselli G, Tovey D, Ravaud P, Boutron I. Interleukin-6 blocking agents for treating COVID-19: a living systematic review. Cochrane Database Syst Rev 2021; 3:CD013881. [PMID: 33734435 PMCID: PMC8406988 DOI: 10.1002/14651858.cd013881] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Interleukin 6 (IL-6) blocking agents have been used for treating severe coronavirus disease 2019 (COVID-19). Their immunosuppressive effect might be valuable in patients with COVID-19 characterised by substantial immune system dysfunction by controlling inflammation and promoting disease tolerance. OBJECTIVES To assess the effect of IL-6 blocking agents compared to standard care alone or with placebo on efficacy and safety outcomes in COVID-19. We will update this assessment regularly. SEARCH METHODS We searched the World Health Organization (WHO) International Clinical Trials Registry Platform (up to 11 February 2021) and the L-OVE platform, and Cochrane COVID-19 Study Register to identify trials up to 26 February 2021. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating IL-6 blocking agents compared with standard care alone or with placebo for people with COVID-19, regardless of disease severity. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. The protocol was amended to reduce the number of outcomes considered. Two review authors independently collected data and assessed the risk of bias with the Cochrane Risk of Bias 2 tool. We rated the certainty of evidence with the GRADE approach for the critical outcomes such as clinical improvement (defined as hospital discharge or improvement on the scale used by trialists to evaluate clinical progression or recovery) (day (D) 28 / ≥ D60); WHO Clinical Progression Score of level 7 or above (i.e. the proportion of participants with mechanical ventilation +/- additional organ support OR death) (D28 / ≥ D60); all-cause mortality (D28 / ≥ D60); incidence of any adverse events; and incidence of serious adverse events. MAIN RESULTS We identified 10 RCTs with available data including one platform trial comparing tocilizumab and sarilumab with standard of care. These trials evaluated tocilizumab (nine RCTs including two platform trials; seven were reported as peer-reviewed articles, two as preprints; 6428 randomised participants); and two sarilumab (one platform trial reported as peer reviewed article, one reported as preprint, 880 randomised participants). All trials included were multicentre trials. They were conducted in Brazil, China, France, Italy, UK, USA, and four were multi-country trials. The mean age range of participants ranged from 56 to 65 years; 4572 (66.3%) of trial participants were male. Disease severity ranged from mild to critical disease. The reported proportion of participants on oxygen at baseline but not intubated varied from 56% to 100% where reported. Five trials reported the inclusion of intubated patients at baseline. We identified a further 20 registered RCTs of tocilizumab compared to placebo/standard care (five completed without available results, five terminated without available results, eight ongoing, two not recruiting); 11 RCTs of sarilumab (two completed without results, three terminated without available results, six ongoing); six RCTs of clazakisumab (five ongoing, one not recruiting); two RCTs of olokizumab (one completed, one not recruiting); one of siltuximab (ongoing) and one RCT of levilimab (completed without available results). Of note, three were cancelled (2 tocilizumab, 1 clazakisumab). One multiple-arm RCT evaluated both tocilizumab and sarilumab compared to standard of care, one three-arm RCT evaluated tocilizumab and siltuximab compared to standard of care and consequently they appear in each respective comparison. Tocilizumab versus standard care alone or with placebo a. Effectiveness of tocilizumab for patients with COVID-19 Tocilizumab probably results in little or no increase in the outcome of clinical improvement at D28 (RR 1.06, 95% CI 1.00 to 1.13; I2 = 40.9%; 7 RCTs, 5585 participants; absolute effect: 31 more with clinical improvement per 1000 (from 0 fewer to 67 more); moderate-certainty evidence). However, we cannot exclude that some subgroups of patients could benefit from the treatment. We did not obtain data for longer-term follow-up (≥ D60). The effect of tocilizumab on the proportion of participants with a WHO Clinical Progression Score of level of 7 or above is uncertain at D28 (RR 0.99, 95% CI 0.56 to 1.74; I2 = 64.4%; 3 RCTs, 712 participants; low-certainty evidence). We did not obtain data for longer-term follow-up (≥ D60). Tocilizumab reduces all-cause mortality at D28 compared to standard care alone or placebo (RR 0.89, 95% CI 0.82 to 0.97; I2 = 0.0%; 8 RCTs, 6363 participants; absolute effect: 32 fewer deaths per 1000 (from 52 fewer to 9 fewer); high-certainty evidence). The evidence suggests uncertainty around the effect on mortality at ≥ D60 (RR 0.86, 95% CI 0.53 to 1.40; I2 = 0.0%; 2 RCTs, 519 participants; low-certainty evidence). b. Safety of tocilizumab for patients with COVID-19 The evidence is very uncertain about the effect of tocilizumab on adverse events (RR 1.23, 95% CI 0.87 to 1.72; I2 = 86.4%; 7 RCTs, 1534 participants; very low-certainty evidence). Nevertheless, tocilizumab probably results in slightly fewer serious adverse events than standard care alone or placebo (RR 0.89, 95% CI 0.75 to 1.06; I2 = 0.0%; 8 RCTs, 2312 participants; moderate-certainty evidence). Sarilumab versus standard care alone or with placebo The evidence is uncertain about the effect of sarilumab on all-cause mortality at D28 (RR 0.77, 95% CI 0.43 to 1.36; 2 RCTs, 880 participants; low certainty), on all-cause mortality at ≥ D60 (RR 1.00, 95% CI 0.50 to 2.0; 1 RCT, 420 participants; low certainty), and serious adverse events (RR 1.17, 95% CI 0.77 to 1.77; 2 RCTs, 880 participants; low certainty). It is unlikely that sarilumab results in an important increase of adverse events (RR 1.05, 95% CI 0.88 to 1.25; 1 RCT, 420 participants; moderate certainty). However, an increase cannot be excluded No data were available for other critical outcomes. AUTHORS' CONCLUSIONS On average, tocilizumab reduces all-cause mortality at D28 compared to standard care alone or placebo and probably results in slightly fewer serious adverse events than standard care alone or placebo. Nevertheless, tocilizumab probably results in little or no increase in the outcome clinical improvement (defined as hospital discharge or improvement measured by trialist-defined scales) at D28. The impact of tocilizumab on other outcomes is uncertain or very uncertain. With the data available, we were not able to explore heterogeneity. Individual patient data meta-analyses are needed to be able to identify which patients are more likely to benefit from this treatment. Evidence for an effect of sarilumab is uncertain and evidence for other anti-IL6 agents is unavailable. Thirty-nine RCTs of IL-6 blocking agents with no results are currently registered, of which nine are completed and seven trials were terminated with no results available. The findings of this review will be updated as new data are made available on the COVID-NMA platform (covid-nma.com).
Collapse
Affiliation(s)
- Lina Ghosn
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Anna Chaimani
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Theodoros Evrenoglou
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Mauricia Davidson
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Carolina Graña
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Christine Schmucker
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Bollig
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | - Camilla Hansen Nejstgaard
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Sonia Menon
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Thu Van Nguyen
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Gabriel Ferrand
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Philipp Kapp
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Carolina Riveros
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | | | - Declan Devane
- HRB-Trials Methodology Research Network, National University of Ireland Galway, Galway, Ireland
- Evidence Synthesis Ireland and Cochrane Ireland, Galway, Ireland
| | - Joerg J Meerpohl
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gabriel Rada
- Epistemonikos Foundation, Santiago, Chile
- UC Evidence Center, Cochrane Chile Associated Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Asbjørn Hróbjartsson
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Giacomo Grasselli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Anesthesia, Intensive Care and Emergency, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Philippe Ravaud
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Isabelle Boutron
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| |
Collapse
|
143
|
Quek E, Tahir H, Kumar P, Hastings R, Jha R. Treatment of COVID-19: a review of current and prospective pharmacotherapies. Br J Hosp Med (Lond) 2021; 82:1-9. [PMID: 33792391 DOI: 10.12968/hmed.2021.0112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread and have grave health and socioeconomic consequences worldwide. Researchers have raced to understand the pathophysiological mechanisms underpinning the disease caused by SARS-CoV-2 so that effective therapeutic targets can be discovered. This review summarises the key pharmacotherapies that are being investigated for treatment of COVID-19, including antiviral, immunomodulator and anticoagulation strategies.
Collapse
Affiliation(s)
- Eleanor Quek
- Department of General Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, UK
| | - Hasan Tahir
- Department of Rheumatology and General Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, UK.,Division of Medicine, University College London, London, UK
| | - Poornima Kumar
- Department of Clinical Haematology, Barnet Hospital, Royal Free London NHS Foundation Trust, London, UK
| | - Robert Hastings
- Department of Respiratory Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, UK
| | - Rajeev Jha
- Department of Intensive Care, Barnet Hospital, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
144
|
Bhandari S, Rankawat G, Singh A. Tocilizumab: An Effective Therapy for Severely and Critically Ill COVID-19 Patients. Indian J Crit Care Med 2021; 25:260-266. [PMID: 33790504 PMCID: PMC7991771 DOI: 10.5005/jp-journals-10071-23747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Tocilizumab (TCZ), a monoclonal antibody against the most prevalent cytokine interleukin-6 (IL-6), is an emerging therapeutic option for COVID-19 infections. The present study was undertaken to assess the therapeutic response of TCZ therapy in severely or critically ill COVID-19 patients and its role as an effective modality of management. Methods: The present retrospective observational study included 30 admitted severely or critically ill COVID-19 patients, treated with TCZ therapy on behalf of raised IL-6 levels. The patients' data concerning medical history, clinical manifestation, arterial blood gas analysis, mode of oxygenation, radiological imaging, and outcome were extracted from their medical records and compared pre- and post-TCZ infusion. Results: All patients of the study group had symptomatic presentations with a mean PaO2/FiO2 (P/F) ratio of 205.41 before TCZ infusion. All patients had a raised IL-6 level (mean value 206.56 pg/mL) that was extremely elevated in 90% of patients. Infusion of TCZ dramatically reduced mean body temperature (100.78-99.32°F) and the requirement for supplemental oxygen (68-48%) and improved mean SpO2 (86-89%) and mean P/F ratio (208-240) within 24 hours. Three patients on noninvasive ventilation were weaned off after TCZ infusion. Serum levels of IL-6 were raised initially but declined within 3-5 days of post-TCZ infusion. Conclusion: TCZ appears to be an effective therapeutic option in severely or critically ill COVID-19 patients with raised IL-6 levels. TCZ immediately improves the clinical status of patients by a probable mechanism of inhibition of cytokine storm and reduces COVID-19-related mortalities. How to cite this article: Bhandari S, Rankawat G, Singh A. Tocilizumab: An Effective Therapy for Severely and Critically Ill COVID-19 Patients. Indian J Crit Care Med 2021;25(3):260-266.
Collapse
Affiliation(s)
- Sudhir Bhandari
- Department of General Medicine, SMS Medical College and Hospital, Jaipur, Rajasthan, India
| | - Govind Rankawat
- Department of General Medicine, SMS Medical College and Hospital, Jaipur, Rajasthan, India
| | - Ajeet Singh
- Department of General Medicine, SMS Medical College and Hospital, Jaipur, Rajasthan, India
| |
Collapse
|
145
|
Thng ZX, De Smet MD, Lee CS, Gupta V, Smith JR, McCluskey PJ, Thorne JE, Kempen JH, Zierhut M, Nguyen QD, Pavesio C, Agrawal R. COVID-19 and immunosuppression: a review of current clinical experiences and implications for ophthalmology patients taking immunosuppressive drugs. Br J Ophthalmol 2021; 105:306-310. [PMID: 32532764 PMCID: PMC7316101 DOI: 10.1136/bjophthalmol-2020-316586] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/04/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 2019 in Wuhan city, Hubei province, China. This is the third and largest coronavirus outbreak since the new millennium after SARS in 2002 and Middle East respiratory syndrome (MERS) in 2012. Over 3 million people have been infected and the COVID-19 has caused more than 217 000 deaths. A concern exists regarding the vulnerability of patients who have been treated with immunosuppressive drugs prior or during this pandemic. Would they be more susceptible to infection by the SARS-CoV-2 and how would their clinical course be altered by their immunosuppressed state? This is a question the wider medical fraternity-including ophthalmologists, rheumatologists, gastroenterologist and transplant physicians among others-must answer. The evidence from the SARS and MERS outbreak offer some degree of confidence that immunosuppression is largely safe in the current COVID-19 pandemic. Preliminary clinical experiences based on case reports, small series and observational studies show the morbidity and mortality rates in immunosuppressed patients may not differ largely from the general population. Overwhelmingly, current best practice guidelines worldwide recommended the continuation of immunosuppression treatment in patients who require them except for perhaps high-dose corticosteroid therapy and in patients with associated risk factors for severe COVID-19 disease.
Collapse
Affiliation(s)
- Zheng Xian Thng
- Department of Ophthalmology, National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | | | - Cecilia S Lee
- Ophthalmology, University of Washington, Seattle, Washington, USA
| | - Vishali Gupta
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Advanced Eye Centre, Chandigarh, India
| | - Justine R Smith
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Peter J McCluskey
- Department of Medical Retina and Uveitis, Sydney Eye Hospital, Sydney, New South Wales, Australia
- Save Sight Institute, Sydney, New South Wales, Australia
| | - Jennifer E Thorne
- Divsion of Ocular Immunology, Johns Hopkins Wilmer Eye Institute, Baltimore, Maryland, USA
| | - John H Kempen
- Department of Ophthalmology, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, USA
- MyungSung Christian Medical Center (MCM) Eye Unit, MCM General Hospital and Myung Sung Medical College, Addis Ababa, Ethiopia
| | | | - Quan Dong Nguyen
- Byers Eye Institute, Stanford University, Palo Alto, California, USA
| | - Carlos Pavesio
- Department of Medical Retina and Uveitis, Moorfields Eye Hospital NHS Foundation Trust, London, London, UK
| | - Rupesh Agrawal
- Department of Ophthalmology, National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
- Department of Medical Retina and Uveitis, Moorfields Eye Hospital NHS Foundation Trust, London, London, UK
| |
Collapse
|
146
|
Lipe DN, Shafer S. CAR-T and checkpoint inhibitors: toxicities and antidotes in the emergency department. Clin Toxicol (Phila) 2021; 59:376-385. [DOI: 10.1080/15563650.2021.1880008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Demis N. Lipe
- Department of Emergency Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah Shafer
- Department of Emergency Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
147
|
Khodashahi R, Naderi H, Bojdy A, Khodashahi M. Effectiveness of Antiviral and Immunomodulatory Agents in the Treatment of COVID-19: A Systematic Review. CURRENT RESPIRATORY MEDICINE REVIEWS 2021. [DOI: 10.2174/1573398x16999201202121247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome
coronavirus 2 (SARS-CoV-2) posed a severe threat to global health. Therefore, new findings on effective
treatment for symptomatic patients with COVID-19 are considered among emergency issues.
This systematic review investigated the effectiveness of pharmacologic interventions in the
management of patients with COVID-19. All the articles published in three electronic databases, including
Google Scholar, PubMed, and Web of Science, were searched from September 15 to
September 30, 2020. Eventually, 24 papers published till September 30 remained to be included in
this review. The effectiveness of immunomodulatory and antiviral agents in the treatment of patients
with COVID-19 was assessed in this review. The obtained results of the current review rejected
the potential of HCQ for the treatment of COVID; however, there was a clinical improvement in
patients treated with ruxolitinib in comparison to that reported for the control group. Methylprednisolone,
dexamethasone, and calcifediol were suggested as beneficial treatments for patients with
COVID-19. The potential efficacy of these antiviral drugs against the SARS-CoV-2 virus is controversial;
nevertheless, the triple combination of antiviral and immunomodulatory agents is effective
in suppressing the shedding of SARS-CoV-2. There have been no supportive data on the superiority
of favipiravir and LPV/r to standard care in the treatment of COVID-19. In addition, no difference
was observed between favipiravir and arbidol for the treatment of these patients. There was an
association between remdesivir treatment and a reduction of 5 days in clinical improvement among
COVID-19 patients. It is required to carry out further RCTs with an in-depth research basis on
COVID-19.
Collapse
Affiliation(s)
- Rozita Khodashahi
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamidreza Naderi
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Bojdy
- Department of Infectious Diseases and Tropical Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mandana Khodashahi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
148
|
Fordham N, Baker E, Forton D, Klammer M, Patel K, Qadir D, Reyal Y, Willis F, Koh MB. First reported case of safe and efficacious use of tocilizumab for treatment of hyperinflammatory syndrome associated with COVID‐19 in an allogeneic stem cell transplant patient. EJHAEM 2021; 2:143-146. [PMID: 35846103 PMCID: PMC9175733 DOI: 10.1002/jha2.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Nicholas Fordham
- Haematology Department St George's University Hospitals NHS Foundation Trust London UK
- St Catherine's College University of Oxford Oxford UK
| | - Emma Baker
- Institute of Infection and Immunity St George's University of London UK
| | - Daniel Forton
- Department of Gastroenterology and Hepatology St George's University Hospitals NHS Foundation Trust London UK
| | - Matthias Klammer
- Haematology Department St George's University Hospitals NHS Foundation Trust London UK
| | - Kamal Patel
- Department of Gastroenterology and Hepatology St George's University Hospitals NHS Foundation Trust London UK
| | - Dara Qadir
- Haematology Department St George's University Hospitals NHS Foundation Trust London UK
| | - Yasmin Reyal
- Haematology Department St George's University Hospitals NHS Foundation Trust London UK
| | - Fenella Willis
- Haematology Department St George's University Hospitals NHS Foundation Trust London UK
| | - Mickey B.C. Koh
- Haematology Department St George's University Hospitals NHS Foundation Trust London UK
- Cell Therapy Programme Health Sciences Authority Singapore
| |
Collapse
|
149
|
Bacca E, Digaetano M, Meschiari M, Franceschini E, Menozzi M, Cuomo G, Mussini C. Immunomodulation for the management of severe SARS-CoV2 infections. State of the art and review of the literature. Biochem Biophys Res Commun 2021; 538:151-155. [PMID: 33303188 PMCID: PMC7699161 DOI: 10.1016/j.bbrc.2020.11.084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022]
Abstract
This Mini Review of the literature aimed to assess the role of tocilizumab for the treatment of severe coronavirus disease 2019 (COVID-19). Based on the available scientific evidence, it is not clear to date what is the best therapeutic strategy for the treatment of COVID-19. Since SARS-CoV-2 infection stimulates a vigorous proinflammatory response and may cause the so-called "cytokine storm", immunomodulator drugs have been investigated as potential treatment for severe COVID-19 pneumonia. Among immunomodulators, tocilizumab, a recombinant humanized monoclonal antibody directed against IL-6 receptor, seems to be promising. An increasing number of clinical trials are exploring the role of tocilizumab in COVID-19, focusing on outcomes like mortality, risk of intensive care unit admission and the need for mechanical ventilation. At the moment, there is no conclusive evidence that tocilizumab would be proper outright in all patients with COVID-19 pneumonia, but some studies suggest that its use may be beneficial in selected categories of patients.
Collapse
Affiliation(s)
- Erica Bacca
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy,Corresponding author. Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, 41124, Italy
| | - Margherita Digaetano
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Erica Franceschini
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Marianna Menozzi
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Gianluca Cuomo
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
150
|
Stein-Merlob AF, Rothberg MV, Holman P, Yang EH. Immunotherapy-Associated Cardiotoxicity of Immune Checkpoint Inhibitors and Chimeric Antigen Receptor T Cell Therapy: Diagnostic and Management Challenges and Strategies. Curr Cardiol Rep 2021; 23:11. [PMID: 33483873 PMCID: PMC7821837 DOI: 10.1007/s11886-021-01440-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Immunotherapies have demonstrated robust clinical efficacy in treating malignancies with increasing use and FDA approvals. We review the epidemiology, risk factors, diagnosis, and treatment of immunotherapy-associated cardiovascular toxicities. RECENT FINDINGS Cardiotoxicity is reported in patients receiving immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cell therapies. The incidence of ICI-related cardiotoxicity is above 1% and includes myocarditis, pericardial disease, arrhythmia, acute coronary syndrome, and vasculitis. The incidence of CAR T cell-associated cardiotoxicities was shown to be as high as 26% and thought to be primarily mediated by cytokine release syndrome. The presentations of cardiotoxicities are variable but are associated with significant morbidity and mortality and benefit from prompt initiation of immunosuppressive therapy. There is increasing evidence for cardiotoxicities following cancer immunotherapy. Available evidence suggests that pretreatment evaluation, close monitoring, and early intervention may reduce cardiovascular morbidity and improve outcomes in the cancer immunotherapy population.
Collapse
Affiliation(s)
- Ashley F. Stein-Merlob
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Michael V. Rothberg
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Patrick Holman
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA USA
- UCLA-Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA 90095 USA
| |
Collapse
|