51
|
Oluwole OO, Bouabdallah K, Muñoz J, De Guibert S, Vose JM, Bartlett NL, Lin Y, Deol A, McSweeney PA, Goy AH, Kersten MJ, Jacobson CA, Farooq U, Minnema MC, Thieblemont C, Timmerman JM, Stiff P, Avivi I, Tzachanis D, Kim JJ, Bashir Z, McLeroy J, Zheng Y, Rossi JM, Johnson L, Goyal L, van Meerten T. Prophylactic corticosteroid use in patients receiving axicabtagene ciloleucel for large B-cell lymphoma. Br J Haematol 2021; 194:690-700. [PMID: 34296427 PMCID: PMC8457222 DOI: 10.1111/bjh.17527] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/21/2021] [Indexed: 01/22/2023]
Abstract
ZUMA‐1 (NCT02348216) examined the safety and efficacy of axicabtagene ciloleucel (axi‐cel), an autologous CD19‐directed chimaeric antigen receptor (CAR)‐T cell therapy, in refractory large B‐cell lymphoma. To reduce treatment‐related toxicity, several exploratory safety management cohorts were added to ZUMA‐1. Specifically, cohort 6 investigated management of cytokine release syndrome (CRS) and neurologic events (NEs) with prophylactic corticosteroids and earlier corticosteroid and tocilizumab intervention. CRS and NE incidence and severity were primary end‐points. Following leukapheresis, patients could receive optional bridging therapy per investigator discretion. All patients received conditioning chemotherapy (days −5 through −3), 2 × 106 CAR‐T cells/kg (day 0) and once‐daily oral dexamethasone [10 mg, day 0 (before axi‐cel) through day 2]. Forty patients received axi‐cel. CRS occurred in 80% of patients (all grade ≤2). Any grade and grade 3 or higher NEs occurred in 58% and 13% of patients respectively. Sixty‐eight per cent of patients did not experience CRS or NEs within 72 h of axi‐cel. With a median follow‐up of 8·9 months, objective and complete response rates were 95% and 80% respectively. Overall, prophylactic corticosteroids and earlier corticosteroid and/or tocilizumab intervention resulted in no grade 3 or higher CRS, a low rate of grade 3 or higher NEs and high response rates in this study population.
Collapse
Affiliation(s)
| | - Krimo Bouabdallah
- Service d'Hématologie et Thérapie Cellulaire, CHU Bordeaux, Bordeaux, France
| | - Javier Muñoz
- Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | | | - Julie M Vose
- University of Nebraska Medical Center, Omaha, NE, USA
| | - Nancy L Bartlett
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yi Lin
- Mayo Clinic, Rochester, MN, USA
| | - Abhinav Deol
- Karmanos Cancer Center, Wayne State University, Detroit, MI, USA
| | | | - Andre H Goy
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Marie José Kersten
- Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands, on behalf of HOVON/LLPC
| | | | | | - Monique C Minnema
- University Medical Center Utrecht, Utrecht, Netherlands, on behalf of HOVON/LLPC
| | - Catherine Thieblemont
- Université de Paris, AP-HP, Hôpital Saint-Louis, Hemato-oncology, DMU HI; Research Unit NF-kappaB, Différenciation et Cancer, Paris, France
| | | | - Patrick Stiff
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Irit Avivi
- Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Jenny J Kim
- Kite, a Gilead Company, Santa Monica, CA, USA
| | | | | | - Yan Zheng
- Kite, a Gilead Company, Santa Monica, CA, USA
| | | | | | | | - Tom van Meerten
- University Medical Center Groningen, Groningen, Netherlands, on behalf of HOVON/PPLC
| |
Collapse
|
52
|
Raja K, Daniel N, Morrison S, Patel R, Gerges J, Nadeem K, Chen B, Kang S, Patel M, Beggs D, Attalla M, Ballem N, Philips M. Impact of Tocilizumab on Clinical Outcomes in COVID-19-Associated Cytokine Release Syndrome: A Single-Center Experience. J Pharm Pract 2021; 36:213-220. [PMID: 34231415 DOI: 10.1177/08971900211028208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tocilizumab is an interleukin-6 receptor antagonist hypothesized to blunt the uncontrolled immune response, cytokine release syndrome, in severe COVID-19 and prevent attributable morbidity and mortality. Objective: The objective of this study was to assess the impact of tocilizumab on clinical outcomes in COVID-19-associated cytokine release syndrome. METHODS Single-center, retrospective cohort study assessing sixty-nine adult patients receiving tocilizumab for suspected COVID-19 cytokine release syndrome. The primary outcome was change in WHO clinical status scale on day seven post-dose analyzed using the Wilcoxon signed rank test. Secondary outcomes assessed impact of timing of administration on clinical outcome. Safety analyses included development of neutropenia, thrombocytopenia, transaminitis, and sepsis within 7 days post-dose. Statistical analyses were conducted using Microsoft Excel. RESULTS No aggregate clinical change was found between day 0 and day 7. Eleven patients improved, twenty-seven worsened, and thirty-one showed no change. Clinical outcomes were weakly correlated with time from symptom onset (rs = 0.21; p = 0.08) or hospital admission (rs = -0.08; p = 0.49) to dose. In-hospital mortality was 63%. Sepsis was diagnosed in 21 patients, five of which were post-dose. Transaminitis, neutropenia, and thrombocytopenia occurred in seven, one, and six patients, respectively. CONCLUSION Tocilizumab did not appear to influence clinical outcomes in our study population, irrespective of timing of administration. Adverse events were not considered drug-related.
Collapse
Affiliation(s)
- Karan Raja
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Nicole Daniel
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Susan Morrison
- Department of Medicine, Clara Maass Medical Center, Belleville, NJ, USA
| | - Ruben Patel
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Jessica Gerges
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Komal Nadeem
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Brandon Chen
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Soo Kang
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Mitesh Patel
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Donald Beggs
- Department of Medicine, Clara Maass Medical Center, Belleville, NJ, USA
| | - Mark Attalla
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| | - Naveen Ballem
- Department of Medicine, Clara Maass Medical Center, Belleville, NJ, USA
| | - Mona Philips
- Department of Pharmacy, Clara Maass Medical Center, Belleville, NJ, USA
| |
Collapse
|
53
|
Meng Y, Deng B, Rong L, Li C, Song W, Ling Z, Xu J, Duan J, Wang Z, Chang AH, Feng X, Xiong X, Chen X, Pan J. Short-Interval Sequential CAR-T Cell Infusion May Enhance Prior CAR-T Cell Expansion to Augment Anti-Lymphoma Response in B-NHL. Front Oncol 2021; 11:640166. [PMID: 34277400 PMCID: PMC8279746 DOI: 10.3389/fonc.2021.640166] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy emerges as a new treatment for refractory or relapsed (r/r) B-cell non-Hodgkin lymphoma (B-NHL); however, the overall response rate (ORR) of which in the B-NHL patients is much lower compared to the patients with r/r B acute lymphoblastic leukemia (B-ALL). We previously confirmed that sequential infusions of CD20 and CD22 CAR-T cells significantly improved the prognosis of the B-NHL patients, while some advanced patients still progressed to death during these CAR-T cell treatments. In this study, we showed that timely sequential administration of the second CAR-T cells could enhance expansion of prior CAR-T cells with stronger tumor-killing capacity in vitro and in vivo. We further conducted compassionate treatments on two advanced B-NHL patients with short-interval sequential infusions of CD19/22/20 CAR-T cells. Disease progression was observed in both patients after primary CAR-T cell infusion but robust re-expansion of prior CAR-T cells and anti-tumor effects was induced by infusion of a secondary CAR-T cells. These results indicate sequential infusions of CAR-T cells with a short interval may improve therapeutic efficacy in the B-NHL patients by promoting expansion of prior CAR-T cells.
Collapse
Affiliation(s)
- Yuan Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Biping Deng
- Cytology Laboratory, Beijing Boren Hospital, Beijing, China
| | - Luan Rong
- Cytology Laboratory, Beijing Boren Hospital, Beijing, China
| | - Chuo Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Weiliang Song
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Zhuojun Ling
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Jinlong Xu
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Jiajia Duan
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Zelin Wang
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Alex H Chang
- Clinical Translational Research Center, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiujuan Xiong
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, China
| | - Xiaoli Chen
- Ganzhou Key Laboratory of Molecular Medicine, the Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Jing Pan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,State Key Laboratory of Experimental Hematology, Boren Clinical Translational Center, Department of Hematology, Beijing Boren Hospital, Beijing, China
| |
Collapse
|
54
|
Yang E, Choi H, Park JS, Noh YW, Choi CM, Lee WJ, Ko JW, Kim J. A first-in-human study of KMRC011, a potential treatment for acute radiation syndrome, to explore tolerability, pharmacokinetics, and pharmacodynamics. Clin Transl Sci 2021; 14:2161-2170. [PMID: 34080313 PMCID: PMC8604209 DOI: 10.1111/cts.13073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 11/27/2022] Open
Abstract
KMRC011 is a novel Toll-like receptor 5 agonist under development as a treatment for acute radiation syndrome (ARS). The aim of this first-in-human study was to investigate the tolerability, pharmacokinetics, and pharmacodynamics of a single intramuscular dose of KMRC011 in healthy subjects. A randomized, single-blind, placebo-controlled, single dose-escalation study was conducted with the starting dose of 5 μg. Eight (4 only for 5 μg cohort) subjects per cohort were randomly assigned to KMRC011 or placebo in a 3:1 ratio. Dose-limiting toxicity (DLT) was assessed throughout the study. Serum concentrations of KMRC011, granulocyte colony-stimulating factor (G-CSF), and interleukin-6 (IL-6) were measured up to 48 h postdose. Based on safety review, the dose of KMRC011 escalated up to 20 μg, and consequently, a total of 4 dose levels (5, 10, 15, and 20 μg) were explored. The most common adverse event was injection site reaction, showing no dose-related trend. Three DLTs (2 cases of hepatic enzyme increased and 1 of pyrexia) were observed; 1 in the 15 μg cohort and 2 in the 20 μg cohort. A developed method could not detect any KMRC011 in serum. KMRC011 15 μg and 20 μg showed significant increases of G-CSF, IL-6, and absolute neutrophil counts, compared with the placebo. A single intramuscular administration of KMRC011 ranging from 5 to 15 μg was tolerated in healthy subjects. Doses of KMRC011 equal to or greater than 15 μg exerted TLR5 agonist-like activities by increasing serum G-CSF and IL-6. It suggests that KMRC011 has the potential for a treatment for ARS.
Collapse
Affiliation(s)
- Eunsol Yang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Hyejung Choi
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Jin-Sol Park
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju-si, Korea
| | | | - Woo-Jong Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon-si, Korea
| | - Jae-Wook Ko
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Jungryul Kim
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea.,Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
55
|
Bayat M, Asemani Y, Mohammadi MR, Sanaei M, Namvarpour M, Eftekhari R. An overview of some potential immunotherapeutic options against COVID-19. Int Immunopharmacol 2021; 95:107516. [PMID: 33765610 PMCID: PMC7908848 DOI: 10.1016/j.intimp.2021.107516] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
After the advent of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) in the late 2019, the resulting severe and pernicious syndrome (COVID-19) immediately was deployed all around the world. To date, despite relentless efforts to control the disease by drug repurposing, there is no approved specific therapy for COVID-19. Given the role of innate and acquired immune components in the control and elimination of viral infections and inflammatory mutilations during SARS-CoV2 pathogenesis, immunotherapeutic strategies appear to be beneficent. Passive immunotherapies such as convalescent plasma, which has received much attention especially in severe cases, as well as suppressing inflammatory cytokines, interferon administration, inhibition of kinases and complement cascade, virus neutralization with key engineered products, cell-based therapies, immunomodulators and anti-inflammatory drugs are among the key immunotherapeutic approaches to deal with COVID-19, which is discussed in this review. Also, details of leading COVID-19 vaccine candidates as the most potent immunotherapy have been provided. However, despite salient improvements, there is still a lack of completely assured vaccines for universal application. Therefore, adopting proper immunotherapies according to the cytokine pattern and involved immune responses, alongside engineered biologics specially ACE2-Fc to curb SARS-CoV2 infection until achieving a tailored vaccine is probably the best strategy to better manage this pandemic. Therefore, gaining knowledge about the mechanism of action, potential targets, as well as the effectiveness of immune-based approaches to confront COVID-19 in the form of a well-ordered review study is highly momentous.
Collapse
Affiliation(s)
- Maryam Bayat
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yahya Asemani
- Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Mohammadi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahsa Sanaei
- Department of Environmental, Polymer and Organic Chemistry, School of Chemistry, Damghan University, Damghan, Iran
| | - Mozhdeh Namvarpour
- Department of Immunology, Shahid Sadoughi University of Medical Science and services, Yazd, Iran
| | - Reyhaneh Eftekhari
- Department of Microbiology, Faculty of Biology, Semnan University, Semnan, Iran
| |
Collapse
|
56
|
Frayberg M, Yung A, Zubiri L, Zlotoff DA, Reynolds KL. What the Cardiologist Needs to Know About Cancer Immunotherapies and Complications. Curr Treat Options Oncol 2021; 22:53. [PMID: 34037918 DOI: 10.1007/s11864-021-00844-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Immunotherapies have transformed the current landscape for cancer treatment and demonstrated unparalleled improvements in survival rates. Now, a third of cancer patients are eligible for treatment with the most widely used class of immunotherapy, immune checkpoint inhibitors (ICIs). As more patients are treated with these novel agents, it is critical for both oncologists and subspecialists to establish a better understanding of the adverse events which can occur. The incidence of myocarditis associated with ICI therapy has been reported to be between 0.27 and 1.14%, 5 times that of myocarditis from other cancer therapies, and, of those patients, 20-50% develop a fulminant form. However, because of unclear risk factors, a broad clinical spectrum, and lack of specific noninvasive studies for diagnosis, the care of patients with ICI-associated cardiotoxicity can be challenging. Here, we have provided a brief overview of the current immunotherapy agents with a focus on the emerging evidence regarding diagnosis and management of cardiac adverse events.
Collapse
Affiliation(s)
- Marina Frayberg
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Anthony Yung
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Leyre Zubiri
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A Zlotoff
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerry L Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
57
|
Bhagali R, Prabhudesai NP, Prabhudesai MN. Post COVID-19 opportunistic candida retinitis: A case report. Indian J Ophthalmol 2021; 69:987-989. [PMID: 33727474 PMCID: PMC8012955 DOI: 10.4103/ijo.ijo_3047_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A 42-year-old male patient presented with profound impairment of vision in both eyes, just as he was recovering from COVID-19. A known diabetic and hypertensive, he suffered from COVID-19 pneumonia further complicated by ARDS, septicaemia and acute kidney injury. His vision on presentation was finger counting close to face bilaterally with multiple, yellowish lesions at the posterior pole. Based on the clinical findings and previous blood culture report, it was diagnosed as candida retinitis and treated with oral and intravitreal anti-fungals. The lesions were regressing at follow-up. This is a post COVID-19 presumed candida retinitis case report.
Collapse
|
58
|
Roth P, Winklhofer S, Müller AMS, Dummer R, Mair MJ, Gramatzki D, Le Rhun E, Manz MG, Weller M, Preusser M. Neurological complications of cancer immunotherapy. Cancer Treat Rev 2021; 97:102189. [PMID: 33872978 DOI: 10.1016/j.ctrv.2021.102189] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy has emerged as a powerful therapeutic approach in many areas of clinical oncology and hematology. The approval of ipilimumab, a monoclonal antibody targeting the immune cell receptor CTLA-4, has marked the beginning of the era of immune checkpoint inhibitors. In the meantime, numerous antibodies targeting the PD-1 pathway have expanded the class of clinically approved immune checkpoint inhibitors. Furthermore, novel antibodies directed against other immune checkpoints are currently in clinical evaluation. More recently, bispecific antibodies, which link T cells directly to tumor cells as well as adoptive T cell transfer with immune cells engineered to express a chimeric antigen receptor, have been approved in certain indications. Neurological complications associated with the use of these novel immunotherapeutic concepts have been recognized more and more frequently. Immune checkpoint inhibitors may cause various neurological deficits mainly by alterations of the peripheral nervous system's integrity. These include radiculopathies, neuropathies, myopathies as well as myasthenic syndromes. Side effects involving the central nervous system are less frequent but may result in severe clinical symptoms and syndromes. The administration of chimeric antigen receptor (CAR) T cell is subject to rigorous patient selection and their use is frequently associated with neurological complications including encephalopathy and seizures, which require immediate action and appropriate therapeutic measures. Close clinical monitoring for neurological symptoms is key for early recognition of immunotherapy-related side effects. Comprehensive diagnostic work-up and adequate therapeutic measures are essential to avoid further clinical deterioration and residual neurological deficits.
Collapse
Affiliation(s)
- Patrick Roth
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Sebastian Winklhofer
- Department of Neuroradiology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Antonia M S Müller
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Austria
| | - Dorothee Gramatzki
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emilie Le Rhun
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland; Department of Neurosurgery, University Hospital and University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Austria
| |
Collapse
|
59
|
Burns EA, Gentille C, Trachtenberg B, Pingali SR, Anand K. Cardiotoxicity Associated with Anti-CD19 Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Recognition, Risk Factors, and Management. Diseases 2021; 9:20. [PMID: 33802788 PMCID: PMC8006027 DOI: 10.3390/diseases9010020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor T-cells (CAR-T) are improving outcomes in pediatric and adult patients with relapsed or refractory B-cell acute lymphoblastic leukemias and subtypes of non-Hodgkin Lymphoma. As this treatment is being increasingly utilized, a better understanding of the unique toxicities associated with this therapy is warranted. While there is growing knowledge on the diagnosis and treatment of cytokine release syndrome (CRS), relatively little is known about the associated cardiac events that occur with CRS that may result in prolonged length of hospital stay, admission to the intensive care unit for pressor support, or cardiac death. This review focuses on the various manifestations of cardiotoxicity, potential risk factors, real world and clinical trial data on prevalence of reported cardiotoxicity events, and treatment recommendations.
Collapse
Affiliation(s)
- Ethan A. Burns
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA; (C.G.); (S.R.P.)
| | - Cesar Gentille
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA; (C.G.); (S.R.P.)
| | - Barry Trachtenberg
- Houston Methodist DeBakey Heart and Vascular Center, 6565 Fannin St, Houston, TX 77030, USA;
| | - Sai Ravi Pingali
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA; (C.G.); (S.R.P.)
| | - Kartik Anand
- Callahan Cancer Center, Great Plains Health, 601 W Leota St, North Platte, NE 69101, USA
| |
Collapse
|
60
|
Luo Y, Song G, Liang S, Li F, Liu K. Research advances in chimeric antigen receptor-modified T-cell therapy (Review). Exp Ther Med 2021; 21:484. [PMID: 33790993 PMCID: PMC8005741 DOI: 10.3892/etm.2021.9915] [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: 08/11/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells are T-cells that have been genetically engineered to express CAR molecules to target specific surface antigens on tumor cells. CAR T-cell therapy, a novel cancer immunotherapy, has been attracting increasing attention, since it exhibited notable efficacy in the treatment of hematological tumors in clinical trials. However, for this type of therapy, challenges must be overcome in the treatment of solid tumors. Furthermore, certain side effects associated with CAR T-cell therapy, including cytokine release syndrome, immune effector cell-related neurotoxicity syndrome, tumor lysis syndrome and on-target off-tumor toxicity, must be taken into consideration. The present study provides a systematic review of the principle, clinical application, current challenges, possible solutions and future perspectives for CAR T-cell therapy.
Collapse
Affiliation(s)
- Yuxi Luo
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China.,The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Guiqin Song
- Department of Biology, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shichu Liang
- The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Feifei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
61
|
Tedesco VE, Mohan C. Biomarkers for Predicting Cytokine Release Syndrome following CD19-Targeted CAR T Cell Therapy. THE JOURNAL OF IMMUNOLOGY 2021; 206:1561-1568. [PMID: 33692146 DOI: 10.4049/jimmunol.2001249] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/27/2021] [Indexed: 12/23/2022]
Abstract
Chimeric Ag receptor (CAR) T cell therapy has shown astonishing potency in treating a variety of hematological malignancies in recent years. Along with this lifesaving potential comes the life-threatening toxicities of cytokine release syndrome (CRS) and neurotoxicity. This work seeks to consolidate biomarker candidates with the potential to predict the severity of CRS and neurotoxicity in patients receiving CD19-targeted CAR T cell therapy. In this systematic review, 33 clinical trials were evaluated for biomarkers that can predict the severity of posttreatment CRS and neurotoxicity. CRS and neurotoxicity occurred in 73.4 and 37% of the reviewed patients, respectively. Identified biomarker candidates included tumor burden, platelet count, C-reactive protein, ferritin, IFN-γ, IL-2, IL-6, IL-8, IL-10, IL-15, and TGF-β. Combinatorial algorithms based on cytokine levels and clinical parameters show excellent promise in predicting CAR-T-cell-therapy-associated toxicities, with improved accuracy over the component biomarkers.
Collapse
Affiliation(s)
- Victor E Tedesco
- Department of Biomedical Engineering, University of Houston, Houston, TX 77004
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX 77004
| |
Collapse
|
62
|
Byar KL, Lunning M. Addressing the Challenges of Aggressive Lymphomas. J Adv Pract Oncol 2021; 11:316-320. [PMID: 33598333 PMCID: PMC7857314 DOI: 10.6004/jadpro.2020.11.3.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Katherine L. Byar, MSN, ANP-BC, BMTCN®, and Matthew Lunning, DO, FACP, covered how to tackle challenges in the treatment of aggressive lymphomas by understanding how to apply emerging data, reviewing optimal therapies and treatment recommendations, and discovering how to manage associated adverse events in this disease state at JADPRO Live 2019.
Collapse
|
63
|
Razavi A, Hamblin MR, Rezaei N. COVID-19 in patients with cancer: Risks and precautions. Am J Emerg Med 2021; 48:357-360. [PMID: 33546958 PMCID: PMC7840397 DOI: 10.1016/j.ajem.2021.01.067] [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] [Received: 10/26/2020] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a member of the coronavirus family, which causes coronavirus disease 2019 (COVID-19). The phenotype of the disease varies from asymptomatic, to a mild phenotype, through to the severe form of acute respiratory distress syndrome (ARDS), which often leads to death, especially in those with underlying diseases. It has been reported that those who suffer from cancer (especially lung cancer and hematological malignancies) are at higher risk of serious complications and death from COVID-19. Some cancer treatments such as CAR T cell therapy can produce a cytokine storm, which is also a hallmark of severe COVID-19. Therefore, patients receiving CAR T cells are at higher risk if they become infected with COVID-19, and could be treated with anti-cytokine approaches.
Collapse
Affiliation(s)
- AzadehSadat Razavi
- Department of Animal Biology, Faculty of Biology Sciences, University of Kharazmi, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Doornfontein, South Africa
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
64
|
Myeloid cell and cytokine interactions with chimeric antigen receptor-T-cell therapy: implication for future therapies. Curr Opin Hematol 2021; 27:41-48. [PMID: 31764168 DOI: 10.1097/moh.0000000000000559] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor (CAR)-T-cell therapy is a revolutionary tool in the treatment of cancer. CAR-T cells exhibit their effector functions through the recognition of their specific antigens on tumor cells and recruitment of other immune cells. However, this therapy is limited by the development of severe toxicities and modest antitumor activity in solid tumors. The host and tumor microenvironment interactions with CAR-T cells play an important role in orchestrating CAR-T-cell functions. Specifically, myeloid lineage cells and their cytokines critically influence the behavior of CAR-T cells. Here, we review the specific effects of myeloid cell interactions with CAR-T cells, their impact on CAR-T-cell response and toxicities, and potential efforts to modulate myeloid cell effects to enhance CAR-T-cell therapy efficacy and reduce toxicities. RECENT FINDINGS Independent studies and correlative science from clinical trials indicate that inhibitory myeloid cells and cytokines contribute to the development of CAR-T-cell-associated toxicities and impairment of their effector functions. SUMMARY These findings illuminate a novel way to reduce CAR-T-cell-associated toxicities and enhance their efficacy through the modulation of myeloid lineage cells and inhibitory cytokines.
Collapse
|
65
|
Maus MV, Alexander S, Bishop MR, Brudno JN, Callahan C, Davila ML, Diamonte C, Dietrich J, Fitzgerald JC, Frigault MJ, Fry TJ, Holter-Chakrabarty JL, Komanduri KV, Lee DW, Locke FL, Maude SL, McCarthy PL, Mead E, Neelapu SS, Neilan TG, Santomasso BD, Shpall EJ, Teachey DT, Turtle CJ, Whitehead T, Grupp SA. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer 2020; 8:jitc-2020-001511. [PMID: 33335028 PMCID: PMC7745688 DOI: 10.1136/jitc-2020-001511] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Immune effector cell (IEC) therapies offer durable and sustained remissions in significant numbers of patients with hematological cancers. While these unique immunotherapies have improved outcomes for pediatric and adult patients in a number of disease states, as 'living drugs,' their toxicity profiles, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), differ markedly from conventional cancer therapeutics. At the time of article preparation, the US Food and Drug Administration (FDA) has approved tisagenlecleucel, axicabtagene ciloleucel, and brexucabtagene autoleucel, all of which are IEC therapies based on genetically modified T cells engineered to express chimeric antigen receptors (CARs), and additional products are expected to reach marketing authorization soon and to enter clinical development in due course. As IEC therapies, especially CAR T cell therapies, enter more widespread clinical use, there is a need for clear, cohesive recommendations on toxicity management, motivating the Society for Immunotherapy of Cancer (SITC) to convene an expert panel to develop a clinical practice guideline. The panel discussed the recognition and management of common toxicities in the context of IEC treatment, including baseline laboratory parameters for monitoring, timing to onset, and pharmacological interventions, ultimately forming evidence- and consensus-based recommendations to assist medical professionals in decision-making and to improve outcomes for patients.
Collapse
Affiliation(s)
- Marcela V Maus
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Sara Alexander
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael R Bishop
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | - Colleen Callahan
- Cancer Immunotherapy Program, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marco L Davila
- Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Claudia Diamonte
- Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew J Frigault
- Bone Marrow Transplant and Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Terry J Fry
- Pediatric Hematology/Oncology/BMT, Children's Hospital Colorado and University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
| | - Jennifer L Holter-Chakrabarty
- Department of Hematology/Oncology/Bone Marrow Transplant and Cellular Therapy, The University of Oklahoma Stephenson Cancer Center, Oklahoma City, Oklahoma, USA
| | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Daniel W Lee
- Department of Pediatrics, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Shannon L Maude
- Cancer Immunotherapy Program, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Philip L McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Elena Mead
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tomas G Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bianca D Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David T Teachey
- Cancer Center, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cameron J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| | - Tom Whitehead
- Emily Whitehead Foundation, Phillipsburg, Pennsylvania, USA
| | - Stephan A Grupp
- Cancer Immunotherapy Program, Division of Oncology, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
66
|
Pro-Inflammatory Cytokines in the Formation of the Pre-Metastatic Niche. Cancers (Basel) 2020; 12:cancers12123752. [PMID: 33322216 PMCID: PMC7764404 DOI: 10.3390/cancers12123752] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The formation of the pre-metastatic niche, a favorable microenvironment in an organ distant from a primary tumor, is critical for tumor metastasis. We review the role of a key player, a class of proteins named pro-inflammatory cytokines secreted from both tumor cells and other cells in tissues, in helping to build the pre-metastatic niche. Various drugs have been developed to target pro-inflammatory cytokines, and their effects on tumor metastases are under investigation. Future clinical studies should focus on combining those drugs and applying them during cancer surgery, a critical moment for the establishment of the pre-metastatic niche. Abstract In the presence of a primary tumor, the pre-metastatic niche is established in secondary organs as a favorable microenvironment for subsequent tumor metastases. This process is orchestrated by bone marrow-derived cells, primary tumor-derived factors, and extracellular matrix. In this review, we summarize the role of pro-inflammatory cytokines including interleukin (IL)-6, IL-1β, CC-chemokine ligand 2 (CCL2), granulocyte-colony stimulating factor (G-CSF), granulocyte–macrophage colony-stimulating factor (GM-CSF), stromal cell-derived factor (SDF)-1, macrophage migration inhibitory factor (MIF), and Chemokine (C–X–C motif) ligand 1 (CXCL1) in the formation of the pre-metastatic niche according to the most recent studies. Pro-inflammatory cytokines released from tumor cells or stromal cells act in both autocrine and paracrine manners to induce phenotype changes in tumor cells, recruit bone marrow-derived cells, and form an inflammatory milieu, all of which prime a secondary organ’s microenvironment for metastatic cell colonization. Considering the active involvement of pro-inflammatory cytokines in niche formation, clinical strategies targeting them offer ways to inhibit the establishment of the pre-metastatic niche and therefore attenuate metastatic progression. We review clinical trials targeting different inflammatory cytokines in patients with metastatic cancers. Due to the pleiotropy and redundancy of pro-inflammatory cytokines, combined therapies should be designed in the future.
Collapse
|
67
|
Zhou Z, Price CC. Overview on the use of IL-6 agents in the treatment of patients with cytokine release syndrome (CRS) and pneumonitis related to COVID-19 disease. Expert Opin Investig Drugs 2020; 29:1407-1412. [DOI: 10.1080/13543784.2020.1840549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zeyun Zhou
- Program of Advanced Professional Master of Public Health, Global Health, Yale School of Public Health, New Haven, CT, USA
| | - Christina C. Price
- Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT, USA
- Department of Allergy and Immunology, VA Medical Center, West Haven, CT, USA
| |
Collapse
|
68
|
Zahid A, Siegler EL, Kenderian SS. CART Cell Toxicities: New Insight into Mechanisms and Management. Clin Hematol Int 2020; 2:149-155. [PMID: 33409484 PMCID: PMC7785104 DOI: 10.2991/chi.k.201108.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
T cells genetically engineered with chimeric antigen receptors (CART) have become a potent class of cancer immunotherapeutics. Numerous clinical trials of CART cells have revealed remarkable remission rates in patients with relapsed or refractory hematologic malignancies. Despite recent clinical success, CART cell therapy has also led to significant morbidity and occasional mortality from associated toxicities. Cytokine release syndrome (CRS) and Immune effector cell-associated neurotoxicity syndrome (ICANS) present barriers to the extensive use of CART cell therapy in the clinic. CRS can lead to fever, hypoxia, hypotension, coagulopathies, and multiorgan failure, and ICANS can result in cognitive dysfunction, seizures, and cerebral edema. The mechanisms of CRS and ICANS are becoming clearer, but many aspects remain unknown. Disease type and burden, peak serum CART cell levels, CART cell dose, CAR structure, elevated pro-inflammatory cytokines, and activated myeloid and endothelial cells all contribute to CART cell toxicity. Current guidelines for the management of toxicities associated with CART cell therapy vary between clinics, but are typically comprised of supportive care and treatment with corticosteroids or tocilizumab, depending on the severity of the symptoms. Acquiring a deeper understanding of CART cell toxicities and developing new management and prevention strategies are ongoing. In this review, we present findings in the mechanisms and management of CART cell toxicities.
Collapse
Affiliation(s)
- Anas Zahid
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, U.A.E
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
69
|
Dal’bo N, Patel R, Parikh R, Shah SP, Guha A, Dani SS, Ganatra S. Cardiotoxicity of Contemporary Anticancer Immunotherapy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020; 22:62. [PMID: 33162729 PMCID: PMC7605901 DOI: 10.1007/s11936-020-00867-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Contemporary anticancer immunotherapy, particularly immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cell therapy, has changed the landscape of treatment for patients with a variety of malignancies who historically had a poor prognosis. However, both immune checkpoint inhibitors and CAR T cell therapy are associated with serious cardiovascular adverse effects. As immunotherapy evolves to include high-risk patients with preexisting cardiovascular risk factors and disease, the risk and relevance of its associated cardiotoxicity will be even higher. RECENT FINDINGS ICI can cause myocarditis, which usually occurs early after initiation, can be fulminant, and prompt treatment with high-dose corticosteroids is crucial. CAR T cell therapy frequently leads to cytokine release syndrome, which is associated with cardiomyopathy or arrhythmia development and may also result in circulatory collapse. Supportive treatment, as well as tocilizumab, an anti-interleukin-6 receptor antibody, is the cornerstone of treatment. Recent findings suggest that preexisting cardiovascular risk factors and disease may increase the risk of such cardiotoxicity, and prompt recognition, as well as treatment, may favorably alter the outcomes. SUMMARY ICI and CAR T cell therapy have improved cancer-related outcomes; however, they both are associated with potentially therapy-limiting cardiotoxicity. Cardio-oncologists are required to play an important role in patient selection, pretherapy cardiovascular optimization, and prompt recognition and treatment of cardiotoxicity.
Collapse
Affiliation(s)
- Natalie Dal’bo
- Division of Internal Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Rushin Patel
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Rohan Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Sachin P. Shah
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Avirup Guha
- Harrington Heart and Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, USA
| | - Sourbha S. Dani
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805 USA
| |
Collapse
|
70
|
Kim MY, Brennan DC, Shah P. General Approach to the Clinical Care of Solid Organ Transplant Recipients with COVID-19 Infection: Management for Transplant Recipients. CURRENT TRANSPLANTATION REPORTS 2020; 7:366-378. [PMID: 33145146 PMCID: PMC7594940 DOI: 10.1007/s40472-020-00305-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Insufficient knowledge about COVID-19 and the potential risks of COVID-19 are limiting organ transplantation in wait-listed candidates and deferring essential health care in solid organ transplant recipients. In this review, we expand the understanding and present an overview of the optimized management of COVID-19 in solid organ transplant recipients. RECENT FINDINGS Transplant recipients are at an increased risk of severe COVID-19. The unique characteristics of transplant recipients can make it more difficult to identify COVID-19. Based on the COVID-19 data to date and our experience, we present testing, management, and prevention methods for COVID-19. Comprehensive diagnostic tests should be performed to determine disease severity, phase of illness, and identify other comorbidities in transplant recipients diagnosed with COVID-19. Outpatients should receive education for preventative measures and optimal health care delivery minimizing potential infectious exposures. Multidisciplinary interventions should be provided to hospitalized transplant recipients for COVID-19 because of the complexity of caring for transplant recipients. SUMMARY Transplant recipients should strictly adhere to infection prevention measures. Understanding of the transplant specific pathophysiology and development of effective treatment strategies for COVID-19 should be prioritized.
Collapse
Affiliation(s)
- Min Young Kim
- Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Daniel C. Brennan
- Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Pali Shah
- Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
- Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
71
|
The incidence of cytokine release syndrome and neurotoxicity of CD19 chimeric antigen receptor-T cell therapy in the patient with acute lymphoblastic leukemia and lymphoma. Cytotherapy 2020; 22:214-226. [PMID: 32305113 DOI: 10.1016/j.jcyt.2020.01.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/24/2022]
Abstract
Our objective was to summarize the side effect of chimeric antigen receptor (CAR)-T cell therapy in patients with acute lymphocytic leukemia (ALL) and lymphoma. Two independent reviewers extracted relevant data. A total of 35 hematologic malignancy studies with CD19 CAR-T cell were included (1412 participants). Severe cytokine release syndrome (sCRS) proportion was experienced by 18.5% (95% confidence interval [CI], 0.128-0.259; P = 0.000) of 982 patients with the National Cancer Institute/Lee/common terminology criteria for adverse events grading system. The pooled neurotoxicity proportion was 21.7% (95% CI, 0.167-0.287; P = 0.000) of 747 patients with the same grading system. For all of the 25 clinical trials with the same grading system, subgroup analysis was performed. Based on the different disease type, a pooled prevalence of 35.7% was observed with event rate (ER) of 0.358 (95% CI, 0.289-0.434; P = 0.000) for ALL in 12 clinical trials. For lymphoma, a pooled prevalence of 13% was observed with ER of 0.073 (95% CI, 0.028-0.179; P = 0.000) in eight clinical trials. It was demonstrated that the patients who were older than 18 years of age have the lower sCRS incidence of 16.1% (95% CI, 0.110-0.250; P = 0.000) compared with 28.6% of the remaining population who were younger than 18 years of age (95% CI, 0.117-0.462: P = 0.023) in our analysis. Based on the different co-stimulatory domain, the sCRS of 16.5% was observed with ER of 0.175 (95% CI, 0.090-0.312; P = 0.000) for 4-1BB. The sCRS of 22.2% was observed with ER of 0.193 (95% CI, 0.107-0.322; P = 0.000) for CD28. For both the CD28 and 4-1BB, the sCRS of 17.3% was observed with ER of 0.170 (95% CI, 0.067-0.369; P = 0.003). Sub-analysis sCRS of the impact with cell dose and specific disease indication were also demonstrated. Limitations include heterogeneity of study populations, as well as high risk of bias of included studies. These results are helpful for physicians, patients and the other stakeholders to understand the adverse events and to further promote the improvement of CAR-T cell therapy in the future.
Collapse
|
72
|
Gutierrez C, Rajendram P, Pastores SM. Toxicities Associated with Immunotherapy and Approach to Cardiotoxicity with Novel Cancer Therapies. Crit Care Clin 2020; 37:47-67. [PMID: 33190775 DOI: 10.1016/j.ccc.2020.08.003] [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: 12/12/2022]
Abstract
In recent years, major advances in oncology especially the advent of targeted agents and immunotherapies (immune checkpoint inhibitors [ICIs] and chimeric antigen receptor [CAR] T-cell therapy) have led to improved quality of life and survival rates in patients with cancer. This article focuses on the clinical features, and grading and management of toxicities associated with ICIs and CAR T-cell therapy. In addition, because cardiotoxicity is one of the most harmful effects of anticancer therapeutics, we describe the risk factors and mechanisms of cardiovascular injury associated with newer agents, screening technologies for at-risk patients, and preventive and treatment strategies.
Collapse
Affiliation(s)
- Cristina Gutierrez
- Department of Critical Care Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Prabalini Rajendram
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Stephen M Pastores
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue C-1179, New York, NY 10065, USA.
| |
Collapse
|
73
|
Schubert ML, Schmitt M, Wang L, Ramos CA, Jordan K, Müller-Tidow C, Dreger P. Side-effect management of chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol 2020; 32:34-48. [PMID: 33098993 DOI: 10.1016/j.annonc.2020.10.478] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells directed against the B-cell marker CD19 are currently changing the landscape for treatment of patients with refractory and/or relapsed B-cell malignancies. Due to the nature of CAR T cells as living drugs, they display a unique toxicity profile. As CAR T-cell therapy is extending towards other diseases and being more broadly employed in hematology and oncology, optimal management strategies of side-effects associated with CAR T-cell therapy are of high relevance. Cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and cytopenias constitute challenges in the treatment of patients with CAR T cells. This review summarizes the current understanding of CAR T-cell toxicity and its management.
Collapse
Affiliation(s)
- M-L Schubert
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| | - M Schmitt
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - L Wang
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - C A Ramos
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - K Jordan
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - C Müller-Tidow
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - P Dreger
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
74
|
Boyiadzis MM, Aksentijevich I, Arber DA, Barrett J, Brentjens RJ, Brufsky J, Cortes J, De Lima M, Forman SJ, Fuchs EJ, Fukas LJ, Gore SD, Litzow MR, Miller JS, Pagel JM, Waller EK, Tallman MS. The Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of acute leukemia. J Immunother Cancer 2020; 8:jitc-2020-000810. [PMID: 33077513 PMCID: PMC7574947 DOI: 10.1136/jitc-2020-000810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Acute leukemia is a constellation of rapidly progressing diseases that affect a wide range of patients regardless of age or gender. Traditional treatment options for patients with acute leukemia include chemotherapy and hematopoietic cell transplantation. The advent of cancer immunotherapy has had a significant impact on acute leukemia treatment. Novel immunotherapeutic agents including antibody-drug conjugates, bispecific T cell engagers, and chimeric antigen receptor T cell therapies have efficacy and have recently been approved by the US Food and Drug Administration (FDA) for the treatment of patients with acute leukemia. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop a clinical practice guideline composed of consensus recommendations on immunotherapy for the treatment of acute lymphoblastic leukemia and acute myeloid leukemia.
Collapse
Affiliation(s)
- Michael M Boyiadzis
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - John Barrett
- Stem Cell Allotransplantation Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jill Brufsky
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jorge Cortes
- Department of Medicine, Division of Hematology/Oncology, Georgia Cancer Center, Augusta, Georgia, USA
| | - Marcos De Lima
- Division of Hematology, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephen J Forman
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Ephraim J Fuchs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linda J Fukas
- Clinical Research Services, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Steven D Gore
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey S Miller
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - John M Pagel
- Center for Blood Disorders and Stem Cell Transplantation, Swedish Cancer Institute, Seattle, Washington, USA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Martin S Tallman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
75
|
Abstract
Cell-based immunotherapies using T cells that are engineered to express a chimeric antigen receptor (CAR-T cells) are an effective treatment option for several B cell malignancies. Compared with most drugs, CAR-T cell products are highly complex, as each cell product is composed of a heterogeneous mixture of millions of cells. The biodistribution and kinetics of CAR-T cells, following administration, are unique given the ability of T cells to actively migrate as well as replicate within the patient. CAR-T cell therapies also have multiple mechanisms of action that contribute to both their antitumor activity and their toxicity. This review provides an overview of the unique pharmacology of CAR-T cells, with a focus on CD19-targeting and B cell maturation antigen (BCMA)-targeting CAR-T cells.
Collapse
Affiliation(s)
- Edward Z Song
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; ,
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; ,
| |
Collapse
|
76
|
Abstract
Despite considerable advances in treatment approaches in the past two decades, multiple myeloma remains an incurable disease. Treatments for myeloma continue to evolve with many emerging immunotherapies. The first immunotherapy used to treat hematologic cancers, including multiple myeloma, was an allogeneic stem cell transplant. In the mid-2000s, immunomodulatory drugs thalidomide, lenalidomide, and subsequently pomalidomide were proven to be effective in multiple myeloma and substantially improved survival. The next wave of immunotherapies for multiple myeloma included the monoclonal antibodies daratumumab and elotuzumab, which were approved by the Food and Drug Administration in 2015. Subsequently, a variety of immunotherapies have been developed for multiple myeloma, including chimeric antigen receptor T cells, bispecific antibodies, antibody drug conjugates, and checkpoint inhibitors. Many of these emerging treatments target the B cell maturation antigen, which is expressed on plasma cells, although several other novel receptors are also being studied. This review summarizes the evidence of these various immunotherapies, their mechanism of action, and data from clinical trials regarding the treatments' safety and efficacy.
Collapse
Affiliation(s)
- Urvi A Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| |
Collapse
|
77
|
Implications of Oxidative Stress and Potential Role of Mitochondrial Dysfunction in COVID-19: Therapeutic Effects of Vitamin D. Antioxidants (Basel) 2020; 9:antiox9090897. [PMID: 32967329 PMCID: PMC7555731 DOI: 10.3390/antiox9090897] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
Due to its high degree of contagiousness and like almost no other virus, SARS-CoV-2 has put the health of the world population on alert. COVID-19 can provoke an acute inflammatory process and uncontrolled oxidative stress, which predisposes one to respiratory syndrome, and in the worst case, death. Recent evidence suggests the mechanistic role of mitochondria and vitamin D in the development of COVID-19. Indeed, mitochondrial dynamics contribute to the maintenance of cellular homeostasis, and its uncoupling involves pathological situations. SARS-CoV-2 infection is associated with altered mitochondrial dynamics with consequent oxidative stress, pro-inflammatory state, cytokine production, and cell death. Furthermore, vitamin D deficiency seems to be associated with increased COVID-19 risk. In contrast, vitamin D can normalize mitochondrial dynamics, which would improve oxidative stress, pro-inflammatory state, and cytokine production. Furthermore, vitamin D reduces renin–angiotensin–aldosterone system activation and, consequently, decreases ROS generation and improves the prognosis of SARS-CoV-2 infection. Thus, the purpose of this review is to deepen the knowledge about the role of mitochondria and vitamin D directly involved in the regulation of oxidative stress and the inflammatory state in SARS-CoV-2 infection. As future prospects, evidence suggests enhancing the vitamin D levels of the world population, especially of those individuals with additional risk factors that predispose to the lethal consequences of SARS-CoV-2 infection.
Collapse
|
78
|
|
79
|
Goossens GH, Dicker D, Farpour-Lambert NJ, Frühbeck G, Mullerova D, Woodward E, Holm JC. Obesity and COVID-19: A Perspective from the European Association for the Study of Obesity on Immunological Perturbations, Therapeutic Challenges, and Opportunities in Obesity. Obes Facts 2020; 13:439-452. [PMID: 32791497 PMCID: PMC7490508 DOI: 10.1159/000510719] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence suggests that obesity is a major risk factor for the initiation, progression, and outcomes of coronavirus disease 2019 (COVID-19). The European Association for the Study of Obesity (EASO), as a scientific and medical society dedicated to the promotion of health and well-being, is greatly concerned about the concomitant obesity and COVID-19 pandemics and their impact on health and society at large. In this perspective, we will address the inherent immunological perturbations and alterations in the renin-angiotensin-aldosterone system in patients with obesity and COVID-19, and discuss how these impairments may underlie the increased susceptibility and more detrimental outcomes of COVID-19 in people with obesity. Clearly, this has important implications for preventive measures, vaccination, and future therapeutic strategies to combat COVID-19. Furthermore, we will highlight important knowledge gaps and provide suggestions for future research and recommendations for policy actions. Since many new reports on COVID-19 rapidly appear, the present perspective should be seen as a focus for discussion to drive forward further understanding, research initiatives, and clinical management of COVID-19.
Collapse
Affiliation(s)
- Gijs H Goossens
- European Association for the Study of Obesity, Teddington, United Kingdom,
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands,
| | - Dror Dicker
- European Association for the Study of Obesity, Teddington, United Kingdom
- Department of Internal Medicine D and Obesity Clinic, Hasharon Hospital, Rabin Medical Center, Petah Tikva, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nathalie J Farpour-Lambert
- European Association for the Study of Obesity, Teddington, United Kingdom
- Obesity Prevention and Care Program Contrepoids, Service of Endocrinology, Diabetology, Nutrition, and Therapeutic Patient Education, Department of Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Gema Frühbeck
- European Association for the Study of Obesity, Teddington, United Kingdom
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, University of Navarra, IdiSNA, CIBEROBN, Spanish Health Institute Carlos III, Pamplona, Spain
| | - Dana Mullerova
- European Association for the Study of Obesity, Teddington, United Kingdom
- Department of Public Health and Preventive Medicine, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Euan Woodward
- European Association for the Study of Obesity, Teddington, United Kingdom
- European Association for the Study of Obesity Secretariat, Teddington, United Kingdom
| | - Jens-Christian Holm
- European Association for the Study of Obesity, Teddington, United Kingdom
- The Children's Obesity Clinic, Department of Pediatrics, Holbæk University Hospital, Holbæk, Denmark
| |
Collapse
|
80
|
Quartuccio L, Sonaglia A, Pecori D, Peghin M, Fabris M, Tascini C, De Vita S. Higher levels of IL-6 early after tocilizumab distinguish survivors from nonsurvivors in COVID-19 pneumonia: A possible indication for deeper targeting of IL-6. J Med Virol 2020; 92:2852-2856. [PMID: 32515499 PMCID: PMC7301025 DOI: 10.1002/jmv.26149] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 01/01/2023]
Abstract
Introduction The most serious COVID‐19 deriving from severe acute respiratory syndrome coronavirus 2 causes a cytokine release storm and it is associated with worse outcomes. In COVID‐19 patients, interleukin‐6 (IL‐6) levels are significantly elevated. Blocking IL‐6 preliminarily resulted in the improvement of this hyperinflammatory state. It is unknown which patients could require higher doses of tocilizumab to get out of the cytokine storm. Materials and Methods Twenty‐four patients affected by COVID‐19 pneumonia were included. All the patients underwent tocilizumab 8 mg/kg intravenously and were tested for serum IL‐6 24 to 48 hours before and 12 to 48 hours after tocilizumab infusion. Comparisons between survivors and nonsurvivors were performed. Results Eighteen patients were discharged, while six patients died, with no clinical or laboratory differences between the two groups at baseline. IL‐6 was not different at baseline (P = .41), while 24 to 48 hours post‐tocilizumab IL‐6 serum levels were significantly higher in nonsurvivors than in survivors (2398.5 [430.5‐9372] vs 290.5 [58.5‐1305.5] pg/mL, P = .022). Serum IL‐6 post‐tocilizumab showed a good predictive ability to discriminate survivors from nonsurvivors (area under the curve, 0.815; 95% confidence interval, 0.63‐0.99, P = .02). Conclusion Repeated measurement of the serum level of IL‐6 early after tocilizumab may distinguish nonsurvivors from survivors and support the choice of deeper targeting IL‐6 in COVID‐19 pneumonia. IL‐6 could be a useful biomarker for severity of COVID‐19. The highest burden of inflammation may be revealed by IL‐6 levels after tocilizumab. IL‐6 levels before and after tocilizumab may indicate how to improve IL‐6 targeting therapy in COVID‐19.
Collapse
Affiliation(s)
- Luca Quartuccio
- Clinic of Rheumatology, Department of Medicine (DAME), ASUFC, University of Udine, Udine, Italy
| | - Arianna Sonaglia
- Clinic of Rheumatology, Department of Medicine (DAME), ASUFC, University of Udine, Udine, Italy
| | - Davide Pecori
- Infectious Diseases Unit, Department of Medicine (DAME), ASUFC, Udine, Italy
| | - Maddalena Peghin
- Infectious Diseases Unit, Department of Medicine (DAME), ASUFC, Udine, Italy
| | - Martina Fabris
- Institute of Clinical Pathology, Department of Medicine (DAME), ASUFC, Udine, Italy
| | - Carlo Tascini
- Infectious Diseases Unit, Department of Medicine (DAME), ASUFC, Udine, Italy
| | - Salvatore De Vita
- Clinic of Rheumatology, Department of Medicine (DAME), ASUFC, University of Udine, Udine, Italy
| |
Collapse
|
81
|
Tocchetti CG, Ameri P, de Boer RA, D’Alessandra Y, Russo M, Sorriento D, Ciccarelli M, Kiss B, Bertrand L, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Linke WA, Mayr M, van der Velden J, Zacchigna S, Ghigo A, Hirsch E, Lyon AR, Görbe A, Ferdinandy P, Madonna R, Heymans S, Thum T. Cardiac dysfunction in cancer patients: beyond direct cardiomyocyte damage of anticancer drugs: novel cardio-oncology insights from the joint 2019 meeting of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc Res 2020; 116:1820-1834. [DOI: 10.1093/cvr/cvaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/17/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
In western countries, cardiovascular (CV) disease and cancer are the leading causes of death in the ageing population. Recent epidemiological data suggest that cancer is more frequent in patients with prevalent or incident CV disease, in particular, heart failure (HF). Indeed, there is a tight link in terms of shared risk factors and mechanisms between HF and cancer. HF induced by anticancer therapies has been extensively studied, primarily focusing on the toxic effects that anti-tumour treatments exert on cardiomyocytes. In this Cardio-Oncology update, members of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart discuss novel evidence interconnecting cardiac dysfunction and cancer via pathways in which cardiomyocytes may be involved but are not central. In particular, the multiple roles of cardiac stromal cells (endothelial cells and fibroblasts) and inflammatory cells are highlighted. Also, the gut microbiota is depicted as a new player at the crossroads between HF and cancer. Finally, the role of non-coding RNAs in Cardio-Oncology is also addressed. All these insights are expected to fuel additional research efforts in the field of Cardio-Oncology.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, AB31, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Yuri D’Alessandra
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine Surgery and Odontology, University of Salerno, Salerno, Italy
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
| | - Luc Bertrand
- IREC Institute, Pole of Cardiovascular Research, Université Catholique de Louvain, Brussels, Belgium
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Ines Falcao-Pires
- Unidade de Investigação e Desenvolvimento Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr Universität Bochum, Bochum, Germany
- Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Witten, Germany
| | | | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Imperial College London, London, UK
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| |
Collapse
|
82
|
Zhong J, Tang J, Ye C, Dong L. The immunology of COVID-19: is immune modulation an option for treatment? THE LANCET. RHEUMATOLOGY 2020; 2:e428-e436. [PMID: 32835246 PMCID: PMC7239618 DOI: 10.1016/s2665-9913(20)30120-x] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In December, 2019, an outbreak of COVID-19 emerged in Wuhan, China and quickly spread globally. As of May 7, 2020, there were 3 672 238 confirmed infections and 254 045 deaths attributed to COVID-19. Evidence has shown that there are asymptomatic carriers of COVID-19 who can transmit the disease to others. The virus incubation time shows a wide range (0-24 days) and the virus displays a high infectivity. It is therefore urgent to develop an effective therapy to treat patients with COVID-19 and to control the spread of the causative agent, severe respiratory syndrome coronavirus 2. Repurposing of approved drugs is widely adopted to fight newly emerged diseases such as COVID-19, as these drugs have known pharmacokinetic and safety profiles. As pathological examination has confirmed the involvement of immune hyperactivation and acute respiratory distress syndrome in fatal cases of COVID-19, several disease-modifying anti-rheumatic drugs (DMARDS), such as hydroxychloroquine and tocilizumab, have been proposed as potential therapies for the treatment of COVID-19. In this Review, we discuss the immunological aspects of COVID-19 and the potential implication of DMARDs in treating this disease.
Collapse
Affiliation(s)
- Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jungen Tang
- Department of Rheumatology and Immunology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cong Ye
- Department of Rheumatology and Immunology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
83
|
Lundh S, Maji S, Melenhorst JJ. Next-generation CAR T cells to overcome current drawbacks. Int J Hematol 2020; 114:532-543. [PMID: 32594314 DOI: 10.1007/s12185-020-02923-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 12/26/2022]
Abstract
As a rapidly emerging treatment in the oncology field, adoptive transfer of autologous, genetically modified chimeric antigen receptor (CAR) T cells has shown striking efficacy and is curative in certain relapsed/refractory patients with hematologic malignancy. This treatment modality of using a "living drug" offers many tantalizing and novel therapeutic strategies for cancer patients whose remaining treatment options may have otherwise been limited. Despite the early success of CAR T cells in hematologic malignancies, many barriers remain for widespread adoption. General barriers include cellular manufacturing limitations, baseline quality of the T cells, adverse events post-infusion such as cytokine release syndrome (CRS) and neurotoxicity, and host rejection of non-human CARs. Additionally, each hematologic disease presents unique mechanisms of relapse which have to be addressed in future clinical trials if we are to augment the efficacy of CAR T treatment. In this review, we will describe current barriers to hindering efficacy of CAR T-cell treatment for hematologic malignancies in a disease-specific manner and review recent innovations aimed at enhancing the potency and applicability of CAR T cells, with the overall goal of building a framework to begin incorporating this form of therapy into the standard medical management of blood cancers.
Collapse
Affiliation(s)
- Stefan Lundh
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sayantan Maji
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J Joseph Melenhorst
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA. .,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. .,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, South Pavilion Expansion, Room 9-105, 3400 Civic Center Blvd., Bldg. 421, Philadelphia, PA, 19104, USA.
| |
Collapse
|
84
|
Convertino I, Tuccori M, Ferraro S, Valdiserra G, Cappello E, Focosi D, Blandizzi C. Exploring pharmacological approaches for managing cytokine storm associated with pneumonia and acute respiratory distress syndrome in COVID-19 patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:331. [PMID: 32527304 PMCID: PMC7289228 DOI: 10.1186/s13054-020-03020-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Abstract
Sars-CoV-2 complications include pneumonia and acute respiratory distress syndrome (ARDS), which require intensive care unit admission. These conditions have rapidly overwhelmed healthcare systems, with detrimental effects on the quality of care and increased mortality. Social isolation strategies have been implemented worldwide with the aim of reducing hospital pressure. Among therapeutic strategies, the use of immunomodulating drugs, to improve prognosis, seems promising. Particularly, since pneumonia and ARDS are associated with a cytokine storm, drugs belonging to therapeutic classes as anti-IL-6, anti-TNF, and JAK inhibitors are currently studied. In this article, we discuss the potential advantages of the most promising pharmacological approaches.
Collapse
Affiliation(s)
- Irma Convertino
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco Tuccori
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy. .,Unit of Adverse Drug Reactions Monitoring, Pisa University Hospital, Pisa, Italy.
| | - Sara Ferraro
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giulia Valdiserra
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Emiliano Cappello
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Unit of Adverse Drug Reactions Monitoring, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
85
|
Ky B, Mann DL. COVID-19 Clinical Trials: A Primer for the Cardiovascular and Cardio-Oncology Communities. JACC CardioOncol 2020; 2:254-269. [PMID: 32313885 PMCID: PMC7164888 DOI: 10.1016/j.jaccao.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease-2019 (COVID-19) pandemic has resulted in a proliferation of clinical trials designed to slow the spread of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Many therapeutic agents that are being used to treat patients with COVID-19 are repurposed treatments for influenza, Ebola, or for malaria that were developed decades ago and are unlikely to be familiar to the cardiovascular and cardio-oncology communities. Here, we provide a foundation for cardiovascular and cardio-oncology physicians on the front line providing care to patients with COVID-19, so that they may better understand the emerging cardiovascular epidemiology and the biological rationale for the clinical trials that are ongoing for the treatment of patients with COVID-19.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- ACE2
- AT1R, angiotensin II type 1 receptor
- CI, confidence interval
- COVID-19
- COVID-19, coronavirus disease-2019
- CoV, coronavirus
- FDA, Food and Drug Administration
- IFN, interferon
- IL, interleukin
- IQR, interquartile range
- MERS, Middle East respiratory syndrome
- RAS, renin-angiotensin system
- RNA, ribonucleic acid
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome-coronavirus-2
- TMPRSS2, transmembrane protease serine 2
- clinical trials
- renin angiotensin system
- sACE2, soluble angiotensin-converting enzyme 2
Collapse
Affiliation(s)
- Bonnie Ky
- Department of Medicine, Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Douglas L. Mann
- Department of Medicine, Division of Cardiology, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
86
|
Morena V, Milazzo L, Oreni L, Bestetti G, Fossali T, Bassoli C, Torre A, Cossu MV, Minari C, Ballone E, Perotti A, Mileto D, Niero F, Merli S, Foschi A, Vimercati S, Rizzardini G, Sollima S, Bradanini L, Galimberti L, Colombo R, Micheli V, Negri C, Ridolfo AL, Meroni L, Galli M, Antinori S, Corbellino M. Off-label use of tocilizumab for the treatment of SARS-CoV-2 pneumonia in Milan, Italy. Eur J Intern Med 2020; 76:36-42. [PMID: 32448770 PMCID: PMC7241995 DOI: 10.1016/j.ejim.2020.05.011] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Tocilizumab, a humanized monoclonal antibody, targets IL-6 receptors blocking downstream pro-inflammatory effects of IL-6. In preliminary reports it was suggested to be beneficial in patients with severe COVID-19. METHODS In this open-label prospective study we describe clinical characteristics and outcome of 51 patients hospitalized with confirmed and severe COVID-19 pneumonia treated with tocilizumab intravenously. All patients had elevated IL-6 plasma level (>40 pg/mL) and oxygen saturation <93% in ambient air. Clinical outcomes, oxygen support, laboratory data and adverse events were collected over a follow-up of 30 days. RESULTS Forty-five patients (88%) were on high-flow oxygen supplementation, six of whom with invasive ventilation. From baseline to day 7 after tocilizumab we observed a dramatic drop of body temperature and CRP value with a significant increase in lymphocyte count (p<0.001). Over a median follow-up time of 34 days from tocilizumab, 34 patients (67%) showed an improvement in their clinical severity class; 31 were discharged; 17 (33%) showed a worsening of their clinical status, of these 14 died (27%). The mortality rate was significantly associated with mechanical ventilation at baseline (83.3% vs 20% of patients on non-invasive oxygen support; p=0.0001). The most frequent side effects were an increase of hepatic enzymes (29%), thrombocytopenia (14%), and serious bacterial and fungal infections (27%). CONCLUSION Tocilizumab exerts a rapidly beneficial effect on fever and inflammatory markers, although no significant impact on the clinical outcome can be inferred by our results. Critically ill patients seem to have a high risk of serious infections with this drug.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antiviral Agents/adverse effects
- Betacoronavirus/drug effects
- Betacoronavirus/isolation & purification
- COVID-19
- Coronavirus Infections/blood
- Coronavirus Infections/epidemiology
- Coronavirus Infections/physiopathology
- Coronavirus Infections/therapy
- Female
- Fever/diagnosis
- Fever/drug therapy
- Humans
- Italy/epidemiology
- Lymphocyte Count/methods
- Male
- Middle Aged
- Outcome and Process Assessment, Health Care
- Pandemics
- Pneumonia, Viral/blood
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/etiology
- Pneumonia, Viral/physiopathology
- Pneumonia, Viral/therapy
- Receptors, Interleukin-6/antagonists & inhibitors
- Respiration, Artificial/methods
- Respiratory Insufficiency/etiology
- Respiratory Insufficiency/therapy
- Retrospective Studies
- SARS-CoV-2
Collapse
Affiliation(s)
- Valentina Morena
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Italy
| | - Laura Milazzo
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy.
| | - Letizia Oreni
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Giovanna Bestetti
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Tommaso Fossali
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Cinzia Bassoli
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Italy
| | - Alessandro Torre
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | | | - Caterina Minari
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Elisabetta Ballone
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Andrea Perotti
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Davide Mileto
- Microbiology, Virology and Bioemergence Diagnostics, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Fosca Niero
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Stefania Merli
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Antonella Foschi
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Stefania Vimercati
- Clinical Pathology Laboratory, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Giuliano Rizzardini
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Salvatore Sollima
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Lucia Bradanini
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Italy
| | - Laura Galimberti
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Riccardo Colombo
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Valeria Micheli
- Microbiology, Virology and Bioemergence Diagnostics, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Cristina Negri
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Anna Lisa Ridolfo
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Luca Meroni
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Massimo Galli
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy; Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Italy
| | - Spinello Antinori
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy; Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Italy
| | - Mario Corbellino
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, Milan, Italy
| |
Collapse
|
87
|
Shetty R, Ghosh A, Honavar SG, Khamar P, Sethu S. Therapeutic opportunities to manage COVID-19/SARS-CoV-2 infection: Present and future. Indian J Ophthalmol 2020; 68:693-702. [PMID: 32317431 PMCID: PMC7350468 DOI: 10.4103/ijo.ijo_639_20] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 01/08/2023] Open
Abstract
A severe form of respiratory disease - COVID-19, caused by SARS-CoV-2 infection, has evolved into a pandemic resulting in significant morbidity and mortality. The unabated spread of the disease is due to lack of vaccine and effective therapeutic agents against this novel virus. Hence, the situation demands an immediate need to explore all the plausible therapeutic and prophylactic strategies that can be made available to stem the spread of the disease. Towards this effort, the current review outlines the key aspects of the pathobiology associated with the morbidity and mortality in COVID-19 patients, which includes a viral response phase and an exaggerated host response phase. The review also summarizes therapeutic agents that are currently being explored along with those with potential for consideration. The broad groups of therapeutic agents discussed include those that: (i) block viral entry to host cells, (ii) block viral replication and survival in host cells, and (iii) dampen exaggerated host immune response. The various kinds of pharmaceutical prophylactic options that may be followed to prevent COVID-19 have also been discussed.
Collapse
Affiliation(s)
- Rohit Shetty
- Narayana Nethralaya, Bangalore, Karnataka, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | | | - Pooja Khamar
- Narayana Nethralaya, Bangalore, Karnataka, India
| | - Swaminathan Sethu
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| |
Collapse
|
88
|
Ky B, Mann DL. COVID-19 Clinical Trials: A Primer for the Cardiovascular and Cardio-Oncology Communities. JACC Basic Transl Sci 2020; 5:501-517. [PMID: 32309679 PMCID: PMC7162643 DOI: 10.1016/j.jacbts.2020.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 01/08/2023]
Abstract
The coronavirus disease-2019 (COVID-19) pandemic has resulted in a proliferation of clinical trials designed to slow the spread of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Many therapeutic agents that are being used to treat patients with COVID-19 are repurposed treatments for influenza, Ebola, or for malaria that were developed decades ago and are unlikely to be familiar to the cardiovascular and cardio-oncology communities. Here, the authors provide a foundation for cardiovascular and cardio-oncology physicians on the front line providing care to patients with COVID-19, so that they may better understand the emerging cardiovascular epidemiology and the biological rationale for the clinical trials that are ongoing for the treatment of patients with COVID-19.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- ACE2
- AT1R, angiotensin II type 1 receptor
- CI, confidence interval
- COVID-19
- COVID-19, coronavirus disease-2019
- CoV, coronavirus
- FDA, Food and Drug Administration
- IFN, interferon
- IL, interleukin
- IQR, interquartile range
- MERS, Middle East respiratory syndrome
- RAS, renin-angiotensin system
- RNA, ribonucleic acid
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome-coronavirus-2
- TMPRSS2, transmembrane protease serine 2
- clinical trials
- renin angiotensin system
- sACE2, soluble angiotensin-converting enzyme 2
Collapse
Affiliation(s)
- Bonnie Ky
- Department of Medicine, Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Douglas L. Mann
- Department of Medicine, Division of Cardiology, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
89
|
Frigault MJ, Maus MV. State of the art in CAR T cell therapy for CD19+ B cell malignancies. J Clin Invest 2020; 130:1586-1594. [PMID: 32235098 PMCID: PMC7108913 DOI: 10.1172/jci129208] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cellular therapy for hematologic malignancies is a rapidly evolving field, with new iterations of novel constructs being developed at a rapid pace. Since the initial reports of chimeric antigen receptor T cell (CAR T cell)success in CD19+ B cell malignancies, multiple clinical trials of CAR T cell therapy directed to CD19 have led to the approval of this therapy by the FDA and the European Medicines Agency for specific indications. Despite strikingly similar efficacy, investigators at multiple centers participating in these studies have observed the nuances of each CAR T cell product, including variability in manufacturing, availability, and toxicity profiles. Here we review state-of-the-art clinical data on CD19-directed CAR T cell therapies in B cell hematologic malignancies, advances made in understanding and modeling associated toxicities, and several exciting advances and creative solutions for overcoming challenges with this therapeutic modality.
Collapse
|
90
|
Yadav RK, Ali A, Kumar S, Sharma A, Baghchi B, Singh P, Das S, Singh C, Sharma S. CAR T cell therapy: newer approaches to counter resistance and cost. Heliyon 2020; 6:e03779. [PMID: 32322738 PMCID: PMC7171532 DOI: 10.1016/j.heliyon.2020.e03779] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/05/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
The genetically engineered Chimeric Antigen Receptor bearing T-cell (CAR T cell) therapy has been emerged as the new paradigm of cancer immunotherapy. However, recent studies have reported an increase in the number of relapsed haematological malignancies. This review provides newer insights into how the efficacy of CAR T cells might be increased by the application of new genome editing technologies, monitoring the complexity of tumor types and T cells sub-types. Next, tumor mutation burden along with tumormicroenvironment and epigenetic mechanisms of CAR T cell as well as tumor cell may play a vital role to tackle the cancer resistance mechanisms. These studies highlight the need to consider traditional cancer therapy in conjunction with CAR T cell therapy for relapsed or cases unresponsive to treatment. Of note, this therapy is highly expensive and requires multi-skill for successful implementation, which results in reduction of its accessibility/affordability to the patients. Here, we also propose a model for cost minimization of CAR T cell therapy by a collaboration of academia, hospitals and industry.
Collapse
Affiliation(s)
- Rajesh Kumar Yadav
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Santosh Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Delhi, India
| | - Basab Baghchi
- Department of Medical Oncology/Haematology, All India Institute of Medical Sciences, Patna, India
| | - Pritanjali Singh
- Department of Radiotherapy, All India Institute of Medical Sciences, Patna, India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Patna, India
| | - Chandramani Singh
- Department of Community and Family Medicine, All India Institute of Medical Sciences, Patna, India
| | - Sadhana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
91
|
Atrash S, Bano K, Harrison B, Abdallah AO. CAR-T treatment for hematological malignancies. J Investig Med 2020; 68:956-964. [PMID: 32200355 DOI: 10.1136/jim-2020-001290] [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] [Accepted: 02/25/2020] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has sparked a wave of optimism in hematological malignancies, reflected by the successful results of early clinical trials involving patients with pre-B-cell acute lymphoblastic leukemia, B-cell lymphomas and multiple myeloma. CAR-T-cell therapy is considered to be a novel immunotherapy treatment that has the potential for curing certain hematological cancers. However, as use of CAR-T-cell therapy has grown, new challenges have surfaced. These challenges include the process of manufacturing the CAR-T cells, the mechanisms of resistance that underlie disease relapse, adverse effects and cost. This review describes the published results of clinical trials and expected developments to overcome CAR-T resistance.
Collapse
Affiliation(s)
- Shebli Atrash
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Charlotte, North Carolina, USA
| | - Kulsum Bano
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Charlotte, North Carolina, USA
| | - Bradley Harrison
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Charlotte, North Carolina, USA
| | - Al-Ola Abdallah
- Department of Internal Medicine, Division of Hematological Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
92
|
Nicastri E, Petrosillo N, Ascoli Bartoli T, Lepore L, Mondi A, Palmieri F, D’Offizi G, Marchioni L, Murachelli S, Ippolito G, Antinori A. National Institute for the Infectious Diseases "L. Spallanzani", IRCCS. Recommendations for COVID-19 clinical management. Infect Dis Rep 2020; 12:8543. [PMID: 32218915 PMCID: PMC7097833 DOI: 10.4081/idr.2020.8543] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/23/2022] Open
Abstract
On January 9 2020, the World Health Organization (WHO) declared the identification, by Chinese Health authorities, of a novel coronavirus, further classified as SARS-CoV-2 responsible of a disease (COVID-19) ranging from asymptomatic cases to severe respiratory involvement. On March 9 2020, WHO declared COVID-19 a global pandemic. Italy is the second most affected country by COVID-19 infection after China. The "L. Spallanzani" National Institute for the Infectious Diseases, IRCCS, Rome, Italy, has been the first Italian hospital to admit and manage patients affected by COVID-19. Hereby, we show our recommendations for the management of COVID-19 patients, based on very limited clinical evidences; they should be considered as expert opinions, which may be modified according to newly produced literature data.
Collapse
Affiliation(s)
| | - Nicola Petrosillo
- National Institute for Infectious Diseases “L.
Spallanzani”, IRCCS, Rome, Italy
| | | | | | | | | | | | | | | | | | - Andrea Antinori
- National Institute for Infectious Diseases “L.
Spallanzani”, IRCCS, Rome, Italy
| |
Collapse
|
93
|
Stahl K, Schmidt BMW, Hoeper MM, Skripuletz T, Möhn N, Beutel G, Eder M, Welte T, Ganser A, Falk CS, Koenecke C, David S. Extracorporeal cytokine removal in severe CAR-T cell associated cytokine release syndrome. J Crit Care 2020; 57:124-129. [PMID: 32113143 DOI: 10.1016/j.jcrc.2020.02.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/30/2020] [Accepted: 02/17/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Life-threatening complications of CD-19 Chimeric antigen receptor - T (CAR-T) cells such as the cytokine release syndrome (CRS)) have been reported. Treatment is limited to IL-6 blockade and steroids although global removal of elevated soluble inflammatory factors might be more effective. METHODS Clinical course of a CRS patient treated with extracorporeal cytokine adsorption (Cytosorb®). A panel of 48 cytokines, chemokines and endothelial markers has been analyzed longitudinally. Ex vivo stimulation of endothelial cells to visualize (immunocytochemistry) and quantify (ECIS, TER) endothelial barrier effects. RESULTS Following CAR-T cell application a 65 years old male developed grade 4 CRS with refractory shock (3 vasopressors) and severe capillary leakage (+37 L/24 h resuscitation). Treatment included IL-6 blockade, methylprednisolone and additionally Cytosorb hemoperfusion. While multiple soluble inflammatory factors were elevated and most of them decreased by more than 50% following Cytosorb, markers of endothelial injury increased steadily (e.g. Angpt-2/Angpt-1) leading to profound endothelial activation and leakage in ex vivo assays. CONCLUSION This is the first reported use of cytokine adsorption for CRS showing efficacy in absorption of various cytokines but not endothelial growth factors. A randomized controlled trial to evaluate additional Cytosorb treatment in CRS is currently recruiting at our institution (NCT04048434).
Collapse
Affiliation(s)
- Klaus Stahl
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Bernhard M W Schmidt
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| | - Marius M Hoeper
- Department of Respiratory Medicine and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Hannover, Germany.
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany.
| | - Gernot Beutel
- Department of Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover, Germany.
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover, Germany.
| | - Tobias Welte
- Department of Respiratory Medicine and German Centre of Lung Research (DZL), Hannover Medical School, Hannover, Germany.
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover, Germany.
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany.
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover, Germany.
| | - Sascha David
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
94
|
Abstract
As a specifically programmable, living immunotherapeutic drug, chimeric antigen receptor (CAR)-modified T cells are providing an alternative treatment option for a broad variety of diseases including so far refractory cancer. By recognizing a tumor-associated antigen, the CAR triggers an anti-tumor response of engineered patient's T cells achieving lasting remissions in the treatment of leukemia and lymphoma. During the last years, significant progress was made in optimizing the CAR design, in manufacturing CAR-engineered T cells, and in the clinical management of patients showing promise to establish adoptive CAR T cell therapy as an effective treatment option in the forefront.
Collapse
Affiliation(s)
- Astrid Holzinger
- RCI Regensburg Center for Interventional Immunology, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
- Chair Genetic Immunotherapy, RCI c/o University Hospital Regensburg, Regensburg, Germany
| | - Hinrich Abken
- RCI Regensburg Center for Interventional Immunology, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany.
- Chair Genetic Immunotherapy, RCI c/o University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
95
|
Ganatra S, Carver JR, Hayek SS, Ky B, Leja MJ, Lenihan DJ, Lenneman C, Mousavi N, Park JH, Perales MA, Ryan TD, Scherrer-Crosbie M, Steingart RM, Yang EH, Zaha V, Barac A, Liu JE. Chimeric Antigen Receptor T-Cell Therapy for Cancer and Heart: JACC Council Perspectives. J Am Coll Cardiol 2019; 74:3153-3163. [PMID: 31856973 PMCID: PMC8211027 DOI: 10.1016/j.jacc.2019.10.049] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/09/2019] [Accepted: 10/31/2019] [Indexed: 12/23/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has significantly advanced the treatment of patients with relapsed and refractory hematologic malignancies and is increasingly investigated as a therapeutic option of other malignancies. The main adverse effect of CAR T-cell therapy is potentially life-threatening cytokine release syndrome (CRS). Clinical cardiovascular (CV) manifestations of CRS include tachycardia, hypotension, troponin elevation, reduced left ventricular ejection fraction, pulmonary edema, and cardiogenic shock. Although insults related to CRS toxicity might be transient and reversible in most instances in patients with adequate CV reserve, they can be particularly challenging in higher-risk, often elderly patients with pre-existing CV disease. As the use of CAR T-cell therapy expands to include a wider patient population, careful patient selection, pre-treatment cardiac evaluation, and CV risk stratification should be considered within the CAR T-cell treatment protocol. Early diagnosis and management of CV complications in patients with CRS require awareness and multidisciplinary collaboration.
Collapse
Affiliation(s)
- Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, Massachusetts; Cardio-Oncology and Adult Cancer Survivorship Program, Dana Farber Cancer Institute, Boston, Massachusetts; Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Joseph R Carver
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Salim S Hayek
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Bonnie Ky
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Monika J Leja
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Department of Medicine, Washington University, St. Louis, Missouri
| | - Carrie Lenneman
- Division of Cardiovascular Medicine, Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Negaresh Mousavi
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, McGill University Health Center, Montreal, Quebec, Canada
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miguel Angel Perales
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas D Ryan
- Heart Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marielle Scherrer-Crosbie
- Cardio-Oncology Program and Cardiac Ultrasound Laboratory, Division of Cardiology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Richard M Steingart
- Department of Medicine, Weill Cornell Medical College, New York, New York; Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric H Yang
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, California
| | - Vlad Zaha
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, University of Texas Southwestern Medical Center, Divison of Cardiovascular Medicine, Department of Medicine, Dallas, Texas
| | - Ana Barac
- Cardio-Oncology Program, Department of Cardiology, MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC.
| | - Jennifer E Liu
- Department of Medicine, Weill Cornell Medical College, New York, New York; Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
96
|
Abstract
OBJECTIVE To present an overview of novel therapies for the treatment of adult acute lymphoblastic leukemia and to discuss nursing implications for these new therapies. DATA SOURCES Published manuscripts, Web sites, and pharmaceutical package inserts. CONCLUSION Several promising therapies have emerged in the treatment of relapsed/refractory and minimal residual disease acute lymphoblastic leukemia. IMPLICATIONS FOR NURSING PRACTICE With the changing paradigm for hematologic malignancies, nurses must remain current in their knowledge regarding novel therapies, including their administration, toxicity profile, and management of adverse events. This article addresses the clinical benefits of novel agents and nursing implications for those agents.
Collapse
|
97
|
Adkins S. The Role of Advanced Practitioners in Optimizing Clinical Management and Support of Patients With Cytokine Release Syndrome From CAR T-Cell Therapy. J Adv Pract Oncol 2019; 10:833-843. [PMID: 33425467 PMCID: PMC7517761 DOI: 10.6004/jadpro.2019.10.8.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
CAR T-cell therapy is rapidly emerging as a promising treatment for many hematologic malignancies. However, CAR T cells can be associated with unique toxicities, including cytokine release syndrome (CRS), which can be severe or fatal if not recognized promptly and treated appropriately. Therefore, it is essential that advanced practitioners caring for patients who have received CAR T-cell therapy to be knowledgeable regarding the signs and symptoms of CRS and understand how to grade and manage toxicities. Understanding the risk factors that may be associated with the development of toxicities as well as the incidence, severity, and timing of CRS with different CAR T-cell products will allow for earlier recognition and treatment, and therefore improvement of outcomes in patients receiving this novel therapy.
Collapse
Affiliation(s)
- Sherry Adkins
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
98
|
Baer B, Dudley CV, Simons RM. Management Principles Associated With Cytokine Release Syndrome. Semin Oncol Nurs 2019; 35:150931. [PMID: 31477411 DOI: 10.1016/j.soncn.2019.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To discuss current recommendations and resources for nurses to ensure they advocate for patients with cytokine release syndrome (CRS). DATA SOURCES A literature search using key terms: cytokine release syndrome, neurotoxicity, CAR T, adverse events. CONCLUSION Chimeric antigen receptor (CAR) T-cell immunotherapy is a growing and rapidly changing field of research. Prompt recognition and management of the side effects of CAR T-cell therapy is pivotal to the safe outcomes of patients. As patients are treated with these novel therapies, additional recommendations and standards for treating CRS and neurotoxicity will occur. IMPLICATION FOR NURSING PRACTICE Nursing plays a pivotal role in the CAR T patients' treatment course because they are the first line of defense in the care of these patients. Providers and patients both rely on nursing knowledge and training to recognize symptoms of CRS and neurotoxicity. With the early recognition of the signs and symptoms of CRS and neurotoxicity, nursing will help improve the outcomes of the patients receiving CAR T-cell therapy.
Collapse
Affiliation(s)
- Brittney Baer
- Hematology-Oncology Program, Vanderbilt-Ingram Cancer Center, Nashville, TN.
| | | | - Rhea Micci Simons
- Hematology-Oncology Program, Vanderbilt-Ingram Cancer Center, Nashville, TN
| |
Collapse
|