551
|
Podoplanin as an Attractive Target of CAR T Cell Therapy. Cells 2020; 9:cells9091971. [PMID: 32858947 PMCID: PMC7564405 DOI: 10.3390/cells9091971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022] Open
Abstract
To date, various kinds of cancer immunotherapy methods have been developed, but T cell immunotherapy is one of the most promising strategies. In general, T cell receptor (TCR) or chimeric antigen receptor (CAR) is used to modify the antigen specificity of T cells. CARs possess an underlying potential with treatment efficacy to treat a broad range of cancer patients compared with TCRs. Although a variety of CAR molecules have been developed so far, the clinical application for solid tumors is limited partly due to its adverse effect known as “on-target off-tumor toxicity”. Therefore, it is very important for CAR T cell therapy to target specific antigens exclusively expressed by malignant cells. Here, we review the application of T cell immunotherapy using specific antigen receptor molecules and discuss the possibility of the clinical application of podoplanin-targeted CAR derived from a cancer-specific monoclonal antibody (CasMab).
Collapse
|
552
|
Muccioli L, Pensato U, Cani I, Guarino M, Cortelli P, Bisulli F. COVID-19-Associated Encephalopathy and Cytokine-Mediated Neuroinflammation. Ann Neurol 2020; 88:860-861. [PMID: 32715524 DOI: 10.1002/ana.25855] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/12/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Lorenzo Muccioli
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Umberto Pensato
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Ilaria Cani
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Guarino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Francesca Bisulli
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
553
|
Pilotto A, Padovani A. Reply to the Letter "COVID-19-Associated Encephalopathy and Cytokine-Mediated Neuroinflammation". Ann Neurol 2020; 88:861-862. [PMID: 32737995 PMCID: PMC7436670 DOI: 10.1002/ana.25856] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Parkinson's Disease Rehabilitation Centre, FERB ONLUS - S. Isidoro Hospital, Trescore Balneario (BG), Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | -
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
554
|
Mu W, Carrillo MA, Kitchen SG. Engineering CAR T Cells to Target the HIV Reservoir. Front Cell Infect Microbiol 2020; 10:410. [PMID: 32903563 PMCID: PMC7438537 DOI: 10.3389/fcimb.2020.00410] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022] Open
Abstract
The HIV reservoir remains to be a difficult barrier to overcome in order to achieve a therapeutic cure for HIV. Several strategies have been developed to purge the reservoir, including the “kick and kill” approach, which is based on the notion that reactivating the latent reservoir will allow subsequent elimination by the host anti-HIV immune cells. However, clinical trials testing certain classes of latency reactivating agents (LRAs) have so far revealed the minimal impact on reducing the viral reservoir. A robust immune response to reactivated HIV expressing cells is critical for this strategy to work. A current focus to enhance anti-HIV immunity is through the use of chimeric antigen receptors (CARs). Currently, HIV-specific CARs are being applied to peripheral T cells, NK cells, and stem cells to boost recognition and killing of HIV infected cells. In this review, we summarize current developments in engineering HIV directed CAR-expressing cells to facilitate HIV elimination. We also summarize current LRAs that enhance the “kick” strategy and how new generation and combinations of LRAs with HIV specific CAR T cell therapies could provide an optimal strategy to target the viral reservoir and achieve HIV clearance from the body.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayra A Carrillo
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Scott G Kitchen
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
555
|
Schmidts A, Wehrli M, Maus MV. Toward Better Understanding and Management of CAR-T Cell-Associated Toxicity. Annu Rev Med 2020; 72:365-382. [PMID: 32776808 DOI: 10.1146/annurev-med-061119-015600] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adoptive transfer of T cells modified with chimeric antigen receptors (CAR-T cells) has changed the therapeutic landscape of hematological malignancies, particularly for acute lymphoblastic leukemia and large B cell lymphoma, where two different CAR-T products are now considered standard of care. Furthermore, intense research efforts are under way to expand the clinical application of CAR-T cell therapy for the benefit of patients suffering from other types of cancers. Nevertheless, CAR-T cell treatment is associated with toxicities such as cytokine release syndrome, which can range in severity from mild flu-like symptoms to life-threatening vasodilatory shock, and a neurological syndrome termed ICANS (immune effector cell-associated neurotoxicity syndrome), which can also range in severity from a temporary cognitive deficit lasting only a few hours to lethal cerebral edema. In this review, we provide an in-depth discussion of different types of CAR-T cell-associated toxicities, including an overview of clinical presentation and grading, pathophysiology, and treatment options. We also address future perspectives and opportunities, with a special focus on hematological malignancies.
Collapse
Affiliation(s)
- Andrea Schmidts
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02129, USA; .,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Marc Wehrli
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02129, USA; .,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02129, USA; .,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
556
|
Joly F, Castel H, Tron L, Lange M, Vardy J. Potential Effect of Immunotherapy Agents on Cognitive Function in Cancer Patients. J Natl Cancer Inst 2020; 112:123-127. [PMID: 31504664 DOI: 10.1093/jnci/djz168] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/02/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022] Open
Abstract
A paradigm shift is occurring in cancer therapy, where instead of targeting tumor cells, immunotherapy agents (IA) target the immune system to overcome cancer tolerance and to stimulate an antitumor immune response. IA using immune checkpoint inhibitors (CPI) or chimeric antigen receptor T-cells have emerged as the most encouraging approaches to treat cancer patients. CPI are reported to induce moderate-to-severe neurologic immune-related adverse events in less than 1% of patients, whereas chimeric antigen receptor T-cell therapy is associated with frequent neurological toxicities that can be severe or even fatal. Cognitive difficulties have been described following chemotherapy and targeted therapy, but not specifically explored in patients receiving IA. The aim of this review is to establish a picture of the first published studies suggesting some biological and physiopathological effects of IA on cognitive functions among cancer patients. The first results originate from a preclinical study evaluating the role of CPI associated with peripheral radiation on cognitive dysfunction and the recent discovery of the central nervous lymphatic system allowing leukocytes to penetrate the central nervous system. Evaluating possible side effects of IA on cognitive function will be an important challenge for future clinical trials and for better understanding the underlying mechanisms through preclinical animal models.
Collapse
Affiliation(s)
- Florence Joly
- Clinical Research Department, Centre François Baclesse, Caen, France.,Normandie University, UNICAEN, INSERM, ANTICIPE, Caen, France.,Cancer and Cognition Platform, Ligue Nationale contre le Cancer, Caen, France.,University Hospital of Caen, Caen, France
| | - Hélène Castel
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, Caen, France.,Normandie Univ, UNIROUEN, INSERM U1239, DC2N, Rouen, France.,Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Laure Tron
- Normandie University, UNICAEN, INSERM, ANTICIPE, Caen, France.,Cancer and Cognition Platform, Ligue Nationale contre le Cancer, Caen, France.,University Hospital of Caen, Caen, France
| | - Marie Lange
- Clinical Research Department, Centre François Baclesse, Caen, France.,Normandie University, UNICAEN, INSERM, ANTICIPE, Caen, France.,Cancer and Cognition Platform, Ligue Nationale contre le Cancer, Caen, France
| | - Janette Vardy
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Concord Cancer Centre, Concord Hospital, Concord, New South Wales, Australia
| |
Collapse
|
557
|
Greenbaum U, Kebriaei P, Srour SA, Olson A, Bashir Q, Neelapu SS, Rezvani K, Shpall EJ. Chimeric antigen receptor T-cell therapy toxicities. Br J Clin Pharmacol 2020; 87:2414-2424. [PMID: 32463929 DOI: 10.1111/bcp.14403] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer immunotherapy has greatly advanced in recent years, with chimeric antigen receptor (CAR) T cells emerging as an innovative technology that harnesses the immune system to fight malignant diseases. These genetically engineered T-cells have shown encouraging results for B-cell lymphoid malignancies and are now being explored for other cancer types. However, this novel adoptive cell therapy is associated with a new spectrum of immune-mediated adverse events and toxicities. As CAR T cells recognize and engage tumour cells, cytokines are secreted and activate other immune cells, frequently leading to rapid development of cytokine release syndrome, which can result in acute deterioration of the patient's clinical condition. In many patients, cytokine release syndrome is mild and easy to manage, but others experience persistent fevers accompanied by hypotension and hypoxia, which require management with immune-modulatory agents. Another deleterious effect of cytokines released by effector cells is immune effector cell-associated neurotoxicity syndrome. This syndrome, caused by a disruption of the blood-brain barrier as a consequence of the immune process, can result in rapid deterioration in cognitive function. This is often associated with subtle changes in handwriting, often progressing to loss of memory and concentration and reduced ability to name objects or follow commands. In some cases, the neurological state is further compromised by seizures and in rare instances with fulminant life-threatening cerebral oedema. In this review, we discuss these toxicities, as well as other CAR T-cell-related immune phenomenon, and address their clinical manifestations, grading, and management options.
Collapse
Affiliation(s)
- Uri Greenbaum
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Samer A Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
558
|
Freyer CW, Porter DL. Cytokine release syndrome and neurotoxicity following CAR T-cell therapy for hematologic malignancies. J Allergy Clin Immunol 2020; 146:940-948. [PMID: 32771558 DOI: 10.1016/j.jaci.2020.07.025] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
Chimeric antigen receptor T cells are a new and exciting immunotherapeutic approach to managing cancer, with impressive efficacy but potentially life-threatening inflammatory toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Patients with severe CRS may develop capillary leak syndrome and disseminated intravascular coagulation, with a cytokine signature similar to that of macrophage activation syndrome/hemophagocytic lymphohistiocytosis. Moderate-to-severe CRS is managed with the IL-6 receptor antagonist tocilizumab with or without corticosteroids, with questions remaining regarding the optimal management of nonresponders. ICANS is an inflammatory neurotoxicity typically occurring after CRS and characterized by impaired blood-brain barrier integrity. Symptoms of encephalopathy range from mild confusion and aphasia to somnolence, obtundation, and in some cases seizures and cerebral edema. ICANS is currently managed with corticosteroids; however, the optimal dose and duration remain to be determined. Little information is available to guide the management of patients with steroid-refractory ICANS. Numerous cytokine-targeted therapies have been proposed to manage these inflammatory toxicities, but few clinical data are available. Management of inflammatory toxicities of chimeric antigen receptor T cells often requires multidisciplinary management and intensive care, during which allergists and immunologists may encounter patients with these unique toxicities.
Collapse
Affiliation(s)
- Craig W Freyer
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pa.
| | - David L Porter
- Division of Cellular Therapy and Stem Cell Transplant, Hospital of the University of Pennsylvania, Philadelphia, Pa; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| |
Collapse
|
559
|
Rubinstein JD, Krupski C, Nelson AS, O'Brien MM, Davies SM, Phillips CL. Chimeric Antigen Receptor T Cell Therapy in Patients with Multiply Relapsed or Refractory Extramedullary Leukemia. Biol Blood Marrow Transplant 2020; 26:e280-e285. [PMID: 32755637 DOI: 10.1016/j.bbmt.2020.07.036] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022]
Abstract
Autologous CD19-directed chimeric antigen receptor T lymphocyte (CAR-T) therapy is an approved and effective treatment for the management of patients with refractory and multiply relapsed B cell precursor acute lymphoblastic leukemia (B-ALL). Experience using this therapy in pediatric patients with extramedullary (EM) disease is limited, in part because these patients have frequently been excluded from clinical trials owing to concerns for an increased risk of immune effector cell-associated neurotoxicity syndrome (ICANS). We infused 7 patients with refractory or multiply relapsed B-ALL who presented with isolated EM relapse with tisagenlecleucel. Six patients had isolated central nervous system (CNS) leukemia, and 1 patient had an isolated testicular relapse. An initial complete response was seen in all patients, with 5 patients remaining in CAR-T-induced remission at a median of 18 months from first infusion. Reversible ICANS was seen in 1 patient with CNS leukemia. Durable B cell aplasia occurred in 3 patients, with a median time to B cell recovery of 6.5 months in the other patients. These data suggest that CAR-T therapy has promising safety and efficacy in treating EM leukemia, although definitive conclusions are limited by the small size of the cohort and limited follow-up period.
Collapse
Affiliation(s)
- Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - Christa Krupski
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam S Nelson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maureen M O'Brien
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine L Phillips
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
560
|
Gutierrez C, Brown ART, Herr MM, Kadri SS, Hill B, Rajendram P, Duggal A, Turtle CJ, Patel K, Lin Y, May HP, Gallo de Moraes A, Maus MV, Frigault MJ, Brudno JN, Athale J, Shah NN, Kochenderfer JN, Dharshan A, Beitinjaneh A, Arias AS, McEvoy C, Mead E, Stephens RS, Nates JL, Neelapu SS, Pastores SM. The chimeric antigen receptor-intensive care unit (CAR-ICU) initiative: Surveying intensive care unit practices in the management of CAR T-cell associated toxicities. J Crit Care 2020; 58:58-64. [PMID: 32361219 PMCID: PMC7321897 DOI: 10.1016/j.jcrc.2020.04.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE A task force of experts from 11 United States (US) centers, sought to describe practices for managing chimeric antigen receptor (CAR) T-cell toxicity in the intensive care unit (ICU). MATERIALS AND METHODS Between June-July 2019, a survey was electronically distributed to 11 centers. The survey addressed: CAR products, toxicities, targeted treatments, management practices and interventions in the ICU. RESULTS Most centers (82%) had experience with commercial and non-FDA approved CAR products. Criteria for ICU admission varied between centers for patients with Cytokine Release Syndrome (CRS) but were similar for Immune Effector Cell Associated Neurotoxicity Syndrome (ICANS). Practices for vasopressor support, neurotoxicity and electroencephalogram monitoring, use of prophylactic anti-epileptic drugs and tocilizumab were comparable. In contrast, fluid resuscitation, respiratory support, methods of surveillance and management of cerebral edema, use of corticosteroid and other anti-cytokine therapies varied between centers. CONCLUSIONS This survey identified areas of investigation that could improve outcomes in CAR T-cell recipients such as fluid and vasopressor selection in CRS, management of respiratory failure, and less common complications such as hemophagocytic lymphohistiocytosis, infections and stroke. The variability in specific treatments for CAR T-cell toxicities, needs to be considered when designing future outcome studies of critically ill CAR T-cell patients.
Collapse
Affiliation(s)
- Cristina Gutierrez
- Department of Critical Care, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America.
| | - Anne Rain T Brown
- Clinical Pharmacy Specialist in Critical Care, Department of Pharmacy, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Megan M Herr
- Transplant and Cellular Therapy Program, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Sameer S Kadri
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, United States of America
| | - Brian Hill
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, United States of America
| | - Prabalini Rajendram
- Department of Critical Care, Cleveland Clinic, Cleveland Clinic Lerner School of Medicine, Cleveland, OH, United States of America
| | - Abhijit Duggal
- Medical Intensive Care Unit, Cleveland Clinic and Assistant Professor of Medicine, Lerner School of Medicine, Cleveland Clinic, Cleveland, OH, United States of America
| | - Cameron J Turtle
- Anderson Family Endowed Chair for Immunotherapy, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, United States of America
| | - Kevin Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle Cancer Alliance, Seattle, WA, United States of America
| | - Yi Lin
- Division of Hematology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, United States of America
| | - Heather P May
- Mayo Clinic College of Medicine and Science, Critical Care Clinical Pharmacist, Department of Pharmacy, Mayo Clinic, Rochester, MN, United States of America
| | - Alice Gallo de Moraes
- Department of Medicine, Division of Pulmonary and Critical Care, Mayo Clinic, Rochester, MN, United States of America
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Mathew J Frigault
- Cellular Immunotherapy Program, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Jennifer N Brudno
- Assistant Research Physician, Surgery Branch, National Cancer Institute, National Institutes of Health, United States of America
| | - Janhavi Athale
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, United States of America
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, National Institute of Health, United States of America
| | - James N Kochenderfer
- Surgery Branch of the National Cancer Institute, National Cancer Institute, National Institute of Health, United States of America
| | - Ananda Dharshan
- Intensive Care Unit, Roswell Park Comprehensive Cancer Center, Department of Anesthesiology, Jacobs School of Medicine & Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, United States of America
| | - Amer Beitinjaneh
- Department of Medicine, Division of Transplantation and Cellular Therapy, University of Miami, Miami, FL, United States of America
| | - Alejandro S Arias
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Miami, Miami, FL, United States of America
| | - Colleen McEvoy
- Stem Cell Transplant and Oncology Intensive Care Unit, Assistant Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Elena Mead
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States of America
| | - R Scott Stephens
- Oncology and Bone Marrow Transplant Critical Care, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Joseph L Nates
- Surgical and Medical Intensive Care Units, Division of Anesthesiology and Critical Care, Department of Critical Care, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Stephen M Pastores
- Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, United States of America
| |
Collapse
|
561
|
The model of cytokine release syndrome in CAR T-cell treatment for B-cell non-Hodgkin lymphoma. Signal Transduct Target Ther 2020; 5:134. [PMID: 32728035 PMCID: PMC7388484 DOI: 10.1038/s41392-020-00256-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/12/2020] [Accepted: 07/15/2020] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor T (CAR T) cell therapy has demonstrated efficacy in the treatment of haematologic malignancies. However, the accompanying adverse events, the most common of which is cytokine release syndrome (CRS), substantially limit its wide application. Due to its unique physiological characteristics, CRS in CAR T-cell treatment for B-cell non-Hodgkin lymphoma (B-NHL) may exhibit some special features. Although existing guidelines had greatly promoted the recognition and management of CRS, many recommendations are not fully applicable to B-NHL. Therefore, it is imperative to identify responses that are specific to CRS observed following CAR T treatment for B-NHL. Based on underlying biological processes and known pathophysiological mechanisms, we tentatively propose a new model to illustrate the occurrence and evolution of CAR T-cell-therapy-related CRS in B-NHL. In this model, tumour burden and bone marrow suppression are considered determinants of CRS. Novel phenomena after CAR T-cell infusion (such as local inflammatory response) are further identified. The proposed model will help us better understand the basic biology of CRS and recognize and manage it more rationally.
Collapse
|
562
|
Rodríguez-Lobato LG, Ganzetti M, Fernández de Larrea C, Hudecek M, Einsele H, Danhof S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front Oncol 2020; 10:1243. [PMID: 32850376 PMCID: PMC7399644 DOI: 10.3389/fonc.2020.01243] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/16/2020] [Indexed: 01/24/2023] Open
Abstract
Despite recent therapeutic advances, the prognosis of multiple myeloma (MM) patients remains poor. Thus, new strategies to improve outcomes are imperative. Chimeric antigen receptor (CAR) T-cell therapy has changed the treatment landscape of B-cell malignancies, providing a potentially curative option for patients who are refractory to standard treatment. Long-term remissions achieved in patients with acute lymphoblastic leukemia and Non-Hodgkin Lymphoma encouraged its further development in MM. B-cell maturation antigen (BCMA)-targeted CAR T-cells have established outstanding results in heavily pre-treated patients. However, several other antigens such as SLAMF7 and CD44v6 are currently under investigation with promising results. Idecabtagene vicleucel is expected to be approved soon for clinical use. Unfortunately, relapses after CAR T-cell infusion have been reported. Hence, understanding the underlying mechanisms of resistance is essential to promote prevention strategies and to enhance CAR T-cell efficacy. In this review we provide an update of the most recent clinical and pre-clinical data and we elucidate both, the potential and the challenges of CAR T-cell therapy in the future.
Collapse
Affiliation(s)
- Luis Gerardo Rodríguez-Lobato
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maya Ganzetti
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlos Fernández de Larrea
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michael Hudecek
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
563
|
Azoulay E, Darmon M, Valade S. Acute life-threatening toxicity from CAR T-cell therapy. Intensive Care Med 2020; 46:1723-1726. [PMID: 32705293 DOI: 10.1007/s00134-020-06193-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/19/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Elie Azoulay
- Médecine Intensive Et Réanimation, APHP, Hôpital Saint-Louis, Paris Diderot Sorbonne University, Paris, France.
| | - Michael Darmon
- Médecine Intensive Et Réanimation, APHP, Hôpital Saint-Louis, Paris Diderot Sorbonne University, Paris, France
| | - Sandrine Valade
- Médecine Intensive Et Réanimation, APHP, Hôpital Saint-Louis, Paris Diderot Sorbonne University, Paris, France
| |
Collapse
|
564
|
Bald T, Krummel MF, Smyth MJ, Barry KC. The NK cell-cancer cycle: advances and new challenges in NK cell-based immunotherapies. Nat Immunol 2020; 21:835-847. [PMID: 32690952 DOI: 10.1038/s41590-020-0728-z] [Citation(s) in RCA: 310] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells belong to the innate immune system and contribute to protecting the host through killing of infected, foreign, stressed or transformed cells. Additionally, via cellular cross-talk, NK cells orchestrate antitumor immune responses. Hence, significant efforts have been undertaken to exploit the therapeutic properties of NK cells in cancer. Current strategies in preclinical and clinical development include adoptive transfer therapies, direct stimulation, recruitment of NK cells into the tumor microenvironment (TME), blockade of inhibitory receptors that limit NK cell functions, and therapeutic modulation of the TME to enhance antitumor NK cell function. In this Review, we introduce the NK cell-cancer cycle to highlight recent advances in NK cell biology and to discuss the progress and problems of NK cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Tobias Bald
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Matthew F Krummel
- Department of Pathology, ImmunoX Initiative, and Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.
| | - Kevin C Barry
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
565
|
Faramand R, Jain M, Staedtke V, Kotani H, Bai R, Reid K, Lee SB, Spitler K, Wang X, Cao B, Pinilla J, Lazaryan A, Khimani F, Shah B, Chavez JC, Nishihori T, Mishra A, Mullinax J, Gonzalez R, Hussaini M, Dam M, Brandjes BD, Bachmeier CA, Anasetti C, Locke FL, Davila ML. Tumor Microenvironment Composition and Severe Cytokine Release Syndrome (CRS) Influence Toxicity in Patients with Large B-Cell Lymphoma Treated with Axicabtagene Ciloleucel. Clin Cancer Res 2020; 26:4823-4831. [PMID: 32669372 DOI: 10.1158/1078-0432.ccr-20-1434] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/14/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE One of the challenges of adoptive T-cell therapy is the development of immune-mediated toxicities including cytokine release syndrome (CRS) and neurotoxicity (NT). We aimed to identify factors that place patients at high risk of severe toxicity or treatment-related death in a cohort of 75 patients with large B-cell lymphoma treated with a standard of care CD19 targeted CAR T-cell product (axicabtagene ciloleucel). EXPERIMENTAL DESIGN Serum cytokine and catecholamine levels were measured prior to lymphodepleting chemotherapy, on the day of CAR T infusion and daily thereafter while patients remained hospitalized. Tumor biopsies were taken within 1 month prior to CAR T infusion for evaluation of gene expression. RESULTS We identified an association between pretreatment levels of IL6 and life-threatening CRS and NT. Because the risk of toxicity was related to pretreatment factors, we hypothesized that the tumor microenvironment (TME) may influence CAR T-cell toxicity. In pretreatment patient tumor biopsies, gene expression of myeloid markers was associated with higher toxicity. CONCLUSIONS These results suggest that a proinflammatory state and an unfavorable TME preemptively put patients at risk for toxicity after CAR T-cell therapy. Tailoring toxicity management strategies to patient risk may reduce morbidity and mortality.
Collapse
Affiliation(s)
- Rawan Faramand
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michael Jain
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Verena Staedtke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hiroshi Kotani
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Renyuan Bai
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kayla Reid
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sae Bom Lee
- Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Kristen Spitler
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Biwei Cao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Javier Pinilla
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aleksander Lazaryan
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Farhad Khimani
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Bijal Shah
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julio C Chavez
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Asmita Mishra
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - John Mullinax
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Sarcoma, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Ricardo Gonzalez
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Sarcoma, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mohammad Hussaini
- Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marian Dam
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brigett D Brandjes
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Christina A Bachmeier
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marco L Davila
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. .,Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
566
|
Analysis of IL-6 serum levels and CAR T cell-specific digital PCR in the context of cytokine release syndrome. Exp Hematol 2020; 88:7-14.e3. [PMID: 32673688 DOI: 10.1016/j.exphem.2020.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Chimeric antigen receptor T-cell (CAR-T) therapies are increasingly used to treat relapsed B-cell lymphomas and acute lymphoblastic leukemia. Considering the frequency of cytokine release syndrome and CAR-T-related encephalopathy syndrome (CRS/CRES) after CAR-T administration, strategies enabling timely prediction of impending CRS/CRES are a clinical need. METHODS We evaluated the dynamics of serum interleukin (IL)-6 levels and CAR-T transgene copy numbers by digital droplet polymerase chain reaction in the peripheral blood of 11 consecutive patients with aggressive B-cell malignancies. RESULTS Four of 11 patients developed CRS, and 3 patients had CRES (33%), 2 of them had previous CRS. IL-6 levels increased on the day of clinical manifestation of CRS. All CRS patients had increased IL-6 peak levels (median IL-6 peak 606 pg/mL in CRS patients vs. 22 pg/mL in non-CRS patients, p = 0.0061). Different patterns emerged from the dynamics of CAR-T/µg genomic DNA: "rapid increase and rapid decrease with complete disappearance," "rapid increase and slow decrease with higher persistence," "rapid increase and rapid decrease with lower persistence," and "slow increase and rapid decrease with almost disappearance." Patients with the pattern "rapid increase and slow decrease with higher persistence" of CAR-T/µg genomic DNA concentration seemed to be at higher risk of developing CRS/CRES. CONCLUSION Thus, the dynamics of CAR-T transgene copy numbers merits further evaluation for a possible association with manifestation of CRS. Increased IL-6 serum levels at CRS manifestation may contribute to the interpretation of symptoms.
Collapse
|
567
|
Frontal encephalopathy related to hyperinflammation in COVID-19. J Neurol 2020; 268:16-19. [PMID: 32654063 PMCID: PMC7353824 DOI: 10.1007/s00415-020-10057-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/24/2022]
|
568
|
Critical Care Management of Toxicities Associated With Targeted Agents and Immunotherapies for Cancer. Crit Care Med 2020; 48:10-21. [PMID: 31725440 DOI: 10.1097/ccm.0000000000004087] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To describe the most common serious adverse effects and organ toxicities associated with emerging therapies for cancer that may necessitate admission to the ICU. DATA SOURCES AND STUDY SELECTION PubMed and Medline search of relevant articles in English on the management of adverse effects of immunotherapy for cancer. DATA EXTRACTION AND DATA SYNTHESIS Targeted therapies including tyrosine kinase inhibitors, monoclonal antibodies, checkpoint inhibitors, and immune effector cell therapy have improved the outcome and quality of life of patients with cancer. However, severe and life-threatening side effects can occur. These toxicities include infusion or hypersensitivity reactions, cytokine release syndrome, pulmonary, cardiac, renal, hepatic, and neurologic toxicities, hemophagocytic lymphohistiocytosis, opportunistic infections, and endocrinopathies. Cytokine release syndrome is the most common serious toxicity after administration of monoclonal antibodies and immune effector cell therapies. Most of the adverse events from immunotherapy results from an exaggerated T-cell response directed against normal tissue, resulting in the generation of high levels of proinflammatory cytokines. Toxicities from targeted therapies are usually secondary to "on target toxicities." Management is largely supportive and may include discontinuation of the specific agent, corticosteroids, and other immune suppressing agents for severe (grade 3 or 4) immune-related adverse events like neurotoxicity and pneumonitis. CONCLUSIONS The complexity of toxicities associated with modern targeted and immunotherapeutic agents for cancer require a multidisciplinary approach among ICU staff, oncologists, and organ specialists and adoption of standardized treatment protocols to ensure the best possible patient outcomes.
Collapse
|
569
|
Hunter BD, Jacobson CA. CAR T-Cell Associated Neurotoxicity: Mechanisms, Clinicopathologic Correlates, and Future Directions. J Natl Cancer Inst 2020; 111:646-654. [PMID: 30753567 DOI: 10.1093/jnci/djz017] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/12/2018] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary new form of immunotherapy for the treatment of hematologic malignancies. The two primary toxicities associated with CAR T-cell therapy include cytokine-release syndrome and neurotoxicity. Cytokine-release syndrome is generally self-limited but high-grade toxicities like hypotension and hypoxemia can be managed with agents that block the effects of IL-6, like tocilizumab, and/or corticosteroids. Although CAR T-cell therapy-associated neurotoxicity is a well-described clinical phenomenon, its pathophysiology remains inadequately understood; treatments and preventive strategies remain elusive. Animal models and clinical trial experience suggest the centrality of monocytes, endothelial dysfunction, and the blood-brain barrier in the development of CAR T-cell-associated neurotoxicity. Here we report what is known from preclinical models, clinical trials, and histopathologic studies regarding the pathophysiology of neurotoxicity, predictors of its incidence, and potential targets for the treatment and prevention of neurotoxicity.
Collapse
|
570
|
|
571
|
Shah N, Aiello J, Avigan DE, Berdeja JG, Borrello IM, Chari A, Cohen AD, Ganapathi K, Gray L, Green D, Krishnan A, Lin Y, Manasanch E, Munshi NC, Nooka AK, Rapoport AP, Smith EL, Vij R, Dhodapkar M. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of multiple myeloma. J Immunother Cancer 2020; 8:e000734. [PMID: 32661116 PMCID: PMC7359060 DOI: 10.1136/jitc-2020-000734] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
Outcomes in multiple myeloma (MM) have improved dramatically in the last two decades with the advent of novel therapies including immunomodulatory agents (IMiDs), proteasome inhibitors and monoclonal antibodies. In recent years, immunotherapy for the treatment of MM has advanced rapidly, with the approval of new targeted agents and monoclonal antibodies directed against myeloma cell-surface antigens, as well as maturing data from late stage trials of chimeric antigen receptor CAR T cells. Therapies that engage the immune system to treat myeloma offer significant clinical benefits with durable responses and manageable toxicity profiles, however, the appropriate use of these immunotherapy agents can present unique challenges for practicing physicians. Therefore, the Society for Immunotherapy of Cancer convened an expert panel, which met to consider the current role of approved and emerging immunotherapy agents in MM and provide guidance to the oncology community by developing consensus recommendations. As immunotherapy evolves as a therapeutic option for the treatment of MM, these guidelines will be updated.
Collapse
Affiliation(s)
- Nina Shah
- Division of Hematology-Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jack Aiello
- Patient Empowerment Network, San Jose, California, USA
| | - David E Avigan
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jesus G Berdeja
- Department of Medicine, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Ivan M Borrello
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center of Johns Hopkins, Baltimore, Maryland, USA
| | - Ajai Chari
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adam D Cohen
- Department of Medicine, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karthik Ganapathi
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lissa Gray
- University of California San Francisco, San Francisco, CA, USA
| | - Damian Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Multiple Myeloma Center for Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elisabet Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nikhil C Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ajay K Nooka
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia, USA
| | - Aaron P Rapoport
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Eric L Smith
- Myeloma Service and Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ravi Vij
- Division of Medical Oncology, Siteman Cancer Center, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Madhav Dhodapkar
- School of Medicine, Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
572
|
Thakkar JP, Prabhu VC, Rouse S, Lukas RV. Acute Neurological Complications of Brain Tumors and Immune Therapies, a Guideline for the Neuro-hospitalist. Curr Neurol Neurosci Rep 2020; 20:32. [PMID: 32596758 DOI: 10.1007/s11910-020-01056-0] [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: 10/24/2022]
Abstract
PURPOSE OF REVIEW Patients with brain tumors presenting to the emergency room with acute neurologic complications may warrant urgent investigations and emergent management. As the neuro-hospitalist will likely encounter this complex patient population, an understanding of the acute neurologic issues will have value. RECENT FINDINGS We discuss updated information and management regarding various acute neurologic complications among neuro-oncology patients and neurologic complications of immunotherapy. Understanding of the acute neurologic complications associated with central nervous system tumors and with common contemporary cancer treatments will facilitate the neuro-hospitalist management of these patient populations. While there are aspects analogous to the diagnosis and management in the non-oncologic population, a number of unique features discussed in this review should be considered.
Collapse
Affiliation(s)
- Jigisha P Thakkar
- Department of Neurology, Stritch School of Medicine, Loyola University Medical Center, 2160 S. 1st Avenue, Bldg 105, Room 2700, Maywood, IL, 60153, USA. .,Department of Neurosurgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Avenue, Bldg 105, Room 1900, Maywood, IL, 60153, USA.
| | - Vikram C Prabhu
- Department of Neurosurgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Avenue, Bldg 105, Room 1900, Maywood, IL, 60153, USA
| | - Stasia Rouse
- Department of Neurology, Stritch School of Medicine, Loyola University Medical Center, 2160 S. 1st Avenue, Bldg 105, Room 2700, Maywood, IL, 60153, USA
| | - Rimas V Lukas
- Department of Neurology, Northwestern University Feinberg School of Medicine, 710 N. Lake Shore Drive, Abbott Hall 1114, Chicago, IL, 60611, USA.,Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
573
|
Sievers S, Watson G, Johncy S, Adkins S. Recognizing and Grading CAR T-Cell Toxicities: An Advanced Practitioner Perspective. Front Oncol 2020; 10:885. [PMID: 32670871 PMCID: PMC7327099 DOI: 10.3389/fonc.2020.00885] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/05/2020] [Indexed: 12/30/2022] Open
Abstract
Over the past decade, chimeric antigen receptor (CAR) T-cell therapy has significantly improved the outlook for many patients with relapsed and/or refractory B-cell malignancies. The use of CAR T-cell therapy and other therapeutic immune effector cells will likely continue to expand with the development of other targets and use in solid tumors. Although these therapies have shown significant promise in the treatment of some malignancies, they can be associated with unique toxicities including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome which can be fatal if not identified early and treated appropriately. An understanding of how best to manage the toxicities associated with CAR T-cell therapy is continually evolving. Institutions providing CAR T-cell therapy have undergone changes in infrastructure and staffing models in order to safely care for patients receiving this novel therapy. As members of a multi-disciplinary health care team, advanced practice providers play significant roles in caring for this patient population and must be well-versed in the recognition, grading, and appropriate management of CAR T-cell therapy-related toxicities as these providers care for patients in multiple settings across the continuum of care.
Collapse
Affiliation(s)
- Savannah Sievers
- Department of Physician Assistant Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Grace Watson
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Swapna Johncy
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sherry Adkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
574
|
Kang L, Tang X, Zhang J, Li M, Xu N, Qi W, Tan J, Lou X, Yu Z, Sun J, Wang Z, Dai H, Chen J, Lin G, Wu D, Yu L. Interleukin-6-knockdown of chimeric antigen receptor-modified T cells significantly reduces IL-6 release from monocytes. Exp Hematol Oncol 2020; 9:11. [PMID: 32523801 PMCID: PMC7278071 DOI: 10.1186/s40164-020-00166-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/19/2020] [Indexed: 02/17/2023] Open
Abstract
Background T cells expressing a chimeric antigen receptor (CAR) engineered to target CD19 can treat leukemia effectively but also increase the risk of complications such as cytokine release syndrome (CRS) and CAR T cell related encephalopathy (CRES) driven by interleukin-6 (IL-6). Here, we investigated whether IL-6 knockdown in CART-19 cells can reduce IL-6 secretion from monocytes, which may reduce the risk of adverse events. Methods Supernatants from cocultures of regular CART-19 cells and B lymphoma cells were added to monocytes in vitro, and the IL-6 levels in monocyte supernatants were measured 24 h later. IL-6 expression was knocked down in regular CART-19 cells by adding a short hairpin RNA (shRNA) (termed ssCART-19) expression cassette specific for IL-6 to the conventional CAR vector. Transduction efficiency and cell proliferation were measured by flow cytometry, and cytotoxicity was measured by evaluating the release of lactate dehydrogenase into the medium. Gene expression was assessed by qRT-PCR and RNA sequencing. A xenograft leukemia mouse model was established by injecting NOD/SCID/γc-/- mice with luciferase-expressing B lymphoma cells, and then the animals were treated with regular CART-19 cells or ssCART-19. Tumor growth was assessed by bioluminescence imaging. Results Both recombinant IL-6 and CART-19 derived IL-6 significantly triggered IL-6 release by monocytes. IL-6 knockdown in ssCART-19 cells dramatically reduced IL-6 release from monocytes in vitro stduy. In vivo study further demonstrated that the mice bearing Raji cells treated with ssCART-19 cells showed significant lower IL-6 levels in serum than those treated with regular CART-19 cells, but comparable anti-tumor efficacy between the animal groups. Conclusion CAR T-derived IL-6 is one of the most important initiators to amplify release of IL-6 from monocytes that further drive sCRS development. IL-6 knockdown in ssCART-19 cells by shRNA technology provide a promising strategy to improve the safety of CAR T cell therapy.
Collapse
Affiliation(s)
- Liqing Kang
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, No, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Xiaowen Tang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jian Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Minghao Li
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, No, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Nan Xu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, No, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Wei Qi
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, No, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Jingwen Tan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, No, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Xiaoyan Lou
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| | - Zhou Yu
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| | - Juanjuan Sun
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| | - Zhenkun Wang
- Central Laboratory of Hematology and Oncology, First Affiliated Hospital, Harbin Medical University, Harbin, 150001 Heilongjiang China
| | - Haiping Dai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Guoqing Lin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical College, Huai'an Second People's Hospital, Huai'an, 223002 China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, No, 3663 North Zhongshan Road, Shanghai, 200065 China.,Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| |
Collapse
|
575
|
|
576
|
Zhao JY, Yan JY, Qu JM. Interpretations of "Diagnosis and Treatment Protocol for Novel Coronavirus Pneumonia (Trial Version 7)". Chin Med J (Engl) 2020; 133:1347-1349. [PMID: 32301757 PMCID: PMC7289291 DOI: 10.1097/cm9.0000000000000866] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Indexed: 01/22/2023] Open
Affiliation(s)
- Jing-Ya Zhao
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | | | | |
Collapse
|
577
|
Neill L, Rees J, Roddie C. Neurotoxicity-CAR T-cell therapy: what the neurologist needs to know. Pract Neurol 2020; 20:285-293. [PMID: 32503897 DOI: 10.1136/practneurol-2020-002550] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 11/04/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is one of the most innovative therapies for haematological malignancies to emerge in a generation. Clinical studies have shown that a single dose of CAR T-cells can deliver durable clinical remissions for some patients with B-cell cancers where conventional therapies have failed.A significant complication of CAR therapy is the immune effector cell-associated neurotoxicity syndrome (ICANS). This syndrome presents a continuum from mild tremor to cerebral oedema and in a minority of cases, death. Management of ICANS is mainly supportive, with a focus on seizure prevention and attenuation of the immune system, often using corticosteroids. Parallel investigation to exclude other central nervous system pathologies (infection, disease progression) is critical. In this review, we discuss current paradigms around CAR T-cell therapy, with a focus on appropriate investigation and management of ICANS.
Collapse
Affiliation(s)
- Lorna Neill
- University College London Hospitals NHS Foundation Trust, London, UK.,University College London, London, UK
| | - Jeremy Rees
- Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | | |
Collapse
|
578
|
Abstract
The introduction of chimeric antigen receptor T-cell (CAR T) therapy has resulted in a paradigm shift in the management of relapsed/refractory B-cell malignancies. Patients with acute lymphoblastic leukemia and non-Hodgkin's lymphoma who had exhausted all meaningful treatment options now have an opportunity for long-term remission and possibly cure. CAR T is rapidly expanding into the treatment paradigm for multiple myeloma with approvals expected in the near future. CAR T for chronic lymphocytic leukemia may not be far behind, while CAR T studies in Hodgkin lymphoma and acute myeloid leukemia are ongoing. Such therapeutic success brings challenges in toxicity management related to cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Our understanding of these unique syndromes is evolving, with predictive models and additional treatment strategies on the horizon. This review aims to summarize the progress of CAR T therapeutics within malignant hematology thus far and highlight ongoing advances in the field.
Collapse
Affiliation(s)
- Craig W Freyer
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David L Porter
- Division of Hematology/Oncology, Cell Therapy and Transplant, Perelman School of Medicine and the Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
579
|
Chou CK, Turtle CJ. Assessment and management of cytokine release syndrome and neurotoxicity following CD19 CAR-T cell therapy. Expert Opin Biol Ther 2020; 20:653-664. [PMID: 32067497 PMCID: PMC7393694 DOI: 10.1080/14712598.2020.1729735] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/11/2020] [Indexed: 12/14/2022]
Abstract
Introduction: The success of CD19 chimeric antigen receptor (CAR)-T cell therapy for treatment of CD19 positive malignancies has led to the FDA approval of two CD19 CAR-T cell products, tisagenlecleucel and axicabtagene ciloleucel, and ongoing clinical trials of new products. Cytokine release syndrome (CRS) and neurotoxicity are common toxicities associated with CD19 CAR-T cell therapies.Areas covered: This review will discuss CRS and neurotoxicity associated with CD19 CAR-T cell therapies, including clinical presentation, risk factors, pathophysiology, and therapeutic or prophylactic interventions.Expert opinion: In conjunction with improved understanding of the pathophysiology of CRS and neurotoxicity, we expect that the recent development of consensus guidelines for the evaluation of these toxicities will enhance management of patients undergoing CD19 CAR-T cell therapies.
Collapse
Affiliation(s)
- Cassie K. Chou
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Cameron J. Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
580
|
Gupta R, Roach C, Hryniewicki AT, Vilke GM, Shatsky RA, Coyne CJ. Management of Chimeric Antigen Receptor (CAR) T-Cell Toxicities: A Review and Guideline for Emergency Providers. J Emerg Med 2020; 59:61-74. [PMID: 32473867 DOI: 10.1016/j.jemermed.2020.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy is an adoptive cellular immunotherapy that is being utilized more frequently due to its initial success in advanced-stage cancers. Unfortunately, CAR T-cell therapy is often associated with acute systemic toxicities, including cytokine release syndrome (CRS) and CAR T-cell-associated neurotoxicity (neurotoxicity). OBJECTIVE We created a review that addresses the potential common emergency department (ED) presentations associated with CAR T-cell therapy. We reviewed the relevant research and clinical guidelines to develop a guide tailored toward addressing the needs of the emergency medicine community to manage these complications. In addition, a case is presented and the evaluation and management of CRS and neurotoxicity are reviewed in detail. DISCUSSION Despite CAR T-cell designs showing promising results, the risk of acquiring an acute toxicity is high, with CRS and neurotoxicity reported most often. The systemic toxicities associated with these adverse events can lead to end-organ damage and compromise the patient acutely or jeopardize the continuation in treatment of their underlying malignancy. Depending on the severity of the toxicity, treatment typically starts with vigilant supportive care, but may include administration of tocilizumab and possibly high-dose corticosteroids if the toxicity is deemed of high severity. CONCLUSIONS With the increasing administration of CAR T-cell therapy, emergency physicians will likely encounter more patients with associated adverse events, including CRS and neurotoxicity. It is increasingly important that emergency physicians are aware of these potential toxicities in order to rapidly diagnose and treat patients undergoing CAR T-cell therapy.
Collapse
Affiliation(s)
- Rishi Gupta
- Department of Emergency Medicine, University of California, San Diego, San Diego, California
| | - Colin Roach
- Department of Emergency Medicine, University of California, San Diego, San Diego, California
| | - Adam T Hryniewicki
- Department of Emergency Medicine, University of California, San Diego, San Diego, California
| | - Gary M Vilke
- Department of Emergency Medicine, University of California, San Diego, San Diego, California
| | - Rebecca A Shatsky
- Department of Emergency Medicine, University of California, San Diego, San Diego, California
| | - Christopher J Coyne
- Department of Emergency Medicine, University of California, San Diego, San Diego, California
| |
Collapse
|
581
|
Gavriilaki E, Sakellari I, Gavriilaki M, Anagnostopoulos A. A New Era in Endothelial Injury Syndromes: Toxicity of CAR-T Cells and the Role of Immunity. Int J Mol Sci 2020; 21:E3886. [PMID: 32485958 PMCID: PMC7312228 DOI: 10.3390/ijms21113886] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy with chimeric antigen receptor T (CAR-T cells) has been recently approved for patients with relapsed/refractory B-lymphoproliferative neoplasms. Along with great efficacy in patients with poor prognosis, CAR-T cells have been also linked with novel toxicities in a significant portion of patients. Cytokine release syndrome (CRS) and neurotoxicity present with unique clinical phenotypes that have not been previously observed. Nevertheless, they share similar characteristics with endothelial injury syndromes developing post hematopoietic cell transplantation (HCT). Evolution in complement therapeutics has attracted renewed interest in these life-threatening syndromes, primarily concerning transplant-associated thrombotic microangiopathy (TA-TMA). The immune system emerges as a key player not only mediating cytokine responses but potentially contributing to endothelial injury in CAR-T cell toxicity. The interplay between complement, endothelial dysfunction, hypercoagulability, and inflammation seems to be a common denominator in these syndromes. As the indications for CAR-T cells and patient populations expand, there in an unmet clinical need of better understanding of the pathophysiology of CAR-T cell toxicity. Therefore, this review aims to provide state-of-the-art knowledge on cellular therapies in clinical practice (indications and toxicities), endothelial injury syndromes and immunity, as well as potential therapeutic targets.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| | - Ioanna Sakellari
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| | - Maria Gavriilaki
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Achilles Anagnostopoulos
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| |
Collapse
|
582
|
Direct Delivery of piggyBac CD19 CAR T Cells Has Potent Anti-tumor Activity against ALL Cells in CNS in a Xenograft Mouse Model. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:37-46. [PMID: 32637579 PMCID: PMC7321814 DOI: 10.1016/j.omto.2020.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/22/2020] [Indexed: 12/29/2022]
Abstract
The anti-CD19 chimeric antigen receptor (CAR) T cells showed excellent effect against acute lymphoblastic leukemia (ALL) in bone marrow (BM) in clinical trials. However, it remains to be elucidated whether the CD19 CAR T cell therapy is effective for ALL cells in central nervous system (CNS) because the patients with isolated or advanced CNS disease were excluded from clinical trials of systemic intravenous (i.v.) delivery of CAR T cells. Therefore, the preclinical evaluation for the efficacy of CAR T cell therapy against ALL cells in CNS is essential for clinical application. We evaluated the effect and adverse reaction of CD19 CAR T cells against ALL in CNS using a xenograft mouse model by i.v. or intra-cerebroventricular (i.c.v.) delivery of CAR T cells. Injection of piggyBac CD19 CAR T cells by i.v. had partial effects, whereas all CAR T i.c.v.-delivered mice had eliminated ALL in CNS. Although some CAR T i.c.v.-delivered mice showed transient changes of clinical symptoms during the first few days after treatment, none of CAR T i.c.v.-delivered mice displayed fatal adverse events. In this study, we demonstrated that direct delivery into CNS of CAR T cells is a possible therapeutic approach with the xenograft mouse model.
Collapse
|
583
|
Chen M, Xu J, Zhou Y, Zhang S, Zhu D. CRISPR-Cas9 genome editing for cancer immunotherapy: opportunities and challenges. Brief Funct Genomics 2020; 19:183-190. [PMID: 31788683 DOI: 10.1093/bfgp/elz027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/19/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Cancer immunotherapy, consisting of antibodies, adoptive T-cell transfer, vaccines and cytokines, is a novel strategy for fighting cancer by artificially stimulating the immune system. It has developed rapidly in recent years, and its efficacy in hematological malignancies and solid tumors has been remarkable. It is regarded as one of the most promising methods for cancer therapy. The current trend in immunotherapy research seeks to improve its efficacy and to ensure the safety of cancer immunotherapy through the use of gene editing technologies. As it is an efficient and simple technology, the CRISPR-Cas9 system is highly anticipated to dramatically strengthen cancer immunotherapy. Intensive research on the CRISPR-Cas9 system has provided increasing confidence to clinicians that this system can be put into clinical use in the near future. This paper reviews the application and challenges of CRISPR-Cas9 in this field, based on various strategies including adaptive cell therapy and antibody therapy, and also highlights the function of CRISPR/Cas9 in the screening of new cancer targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Laboratory Medicine, Sixth Affiliated Hospital of Yangzhou University, Taizhou, Jiangsu, China, 225400.,Department of Laboratory Medicine, Affiliated Taixing Hospital of Bengbu Medical College, Taizhou, Jiangsu, China, 225400
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China, 223001
| | - Yang Zhou
- Department of Pharmacology, Minhang Hospital and School of Pharmacy, Fudan University, Shanghai, China 201203
| | - Silin Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China, 430060
| | - Di Zhu
- Department of Pharmacology, Minhang Hospital and School of Pharmacy, Fudan University, Shanghai, China 201203.,Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China, 201203
| |
Collapse
|
584
|
Qiu L, Zhu F, Wei G, Wu W, Yang L, Hu Y, Huang H. Idiopathic thrombocytopenic purpura treatment in a relapsed/refractory multiple myeloma patient after chimeric antigen receptor T cell therapy. Regen Ther 2020; 14:271-274. [PMID: 32455157 PMCID: PMC7232090 DOI: 10.1016/j.reth.2020.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 10/27/2022] Open
Abstract
The adoptive transfer of CAR-T cells, which are modified T cells expressing chimeric antigen receptors (CARs), to target B cell maturation antigen (BCMA) has demonstrated impressive results in treating relapsed/refractory multiple myeloma. Although BCMA CAR-T therapy induces certain complications in some patients, idiopathic thrombocytopenic purpura (ITP) has not been reported as one of them. To the best of our knowledge, this is the first report of the successful treatment of ITP that arose in a relapsed/refractory multiple myeloma patient following anti-BCMA CAR-T cell infusion. Herein, we describe this relatively uncommon complication and provide guidance on its treatment.
Collapse
Affiliation(s)
- Lei Qiu
- Department of Hematology, Zhoushan Hospital, Zhoushan, 310000, China.,Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Feng Zhu
- Department of Hematology, Zhoushan Hospital, Zhoushan, 310000, China.,Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Guoqing Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310000, China.,Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Wenjun Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310000, China.,Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Luxin Yang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310000, China.,Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310000, China.,Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310000, China.,Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| |
Collapse
|
585
|
Cerrano M, Ruella M, Perales MA, Vitale C, Faraci DG, Giaccone L, Coscia M, Maloy M, Sanchez-Escamilla M, Elsabah H, Fadul A, Maffini E, Pittari G, Bruno B. The Advent of CAR T-Cell Therapy for Lymphoproliferative Neoplasms: Integrating Research Into Clinical Practice. Front Immunol 2020; 11:888. [PMID: 32477359 PMCID: PMC7235422 DOI: 10.3389/fimmu.2020.00888] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/17/2020] [Indexed: 01/13/2023] Open
Abstract
Research on CAR T cells has achieved enormous progress in recent years. After the impressive results obtained in relapsed and refractory B-cell acute lymphoblastic leukemia and aggressive B-cell lymphomas, two constructs, tisagenlecleucel and axicabtagene ciloleucel, were approved by FDA. The role of CAR T cells in the treatment of B-cell disorders, however, is rapidly evolving. Ongoing clinical trials aim at comparing CAR T cells with standard treatment options and at evaluating their efficacy earlier in the disease course. The use of CAR T cells is still limited by the risk of relevant toxicities, most commonly cytokine release syndrome and neurotoxicity, whose management has nonetheless significantly improved. Some patients do not respond or relapse after treatment, either because of poor CAR T-cell expansion, lack of anti-tumor effects or after the loss of the target antigen on tumor cells. Investigators are trying to overcome these hurdles in many ways: by testing constructs which target different and/or multiple antigens or by improving CAR T-cell structure with additional functions and synergistic molecules. Alternative cell sources including allogeneic products (off-the-shelf CAR T cells), NK cells, and T cells obtained from induced pluripotent stem cells are also considered. Several trials are exploring the curative potential of CAR T cells in other malignancies, and recent data on multiple myeloma and chronic lymphocytic leukemia are encouraging. Given the likely expansion of CAR T-cell indications and their wider availability over time, more and more highly specialized clinical centers, with dedicated clinical units, will be required. Overall, the costs of these cell therapies will also play a role in the sustainability of many health care systems. This review will focus on the major clinical trials of CAR T cells in B-cell malignancies, including those leading to the first FDA approvals, and on the new settings in which these constructs are being tested. Besides, the most promising approaches to improve CAR T-cell efficacy and early data on alternative cell sources will be reviewed. Finally, we will discuss the challenges and the opportunities that are emerging with the advent of CAR T cells into clinical routine.
Collapse
Affiliation(s)
- Marco Cerrano
- Department of Oncology/Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Marco Ruella
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
| | - Candida Vitale
- Department of Oncology/Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Danilo Giuseppe Faraci
- Department of Oncology/Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Luisa Giaccone
- Department of Oncology/Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Marta Coscia
- Department of Oncology/Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Molly Maloy
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
| | - Miriam Sanchez-Escamilla
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
- Department of Hematological Malignancies and Stem Cell Transplantation, Research Institute of Marques de Valdecilla (IDIVAL), Santander, Spain
| | - Hesham Elsabah
- Department of Medical Oncology, Hematology/BMT Service, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Afraa Fadul
- Department of Medical Oncology, Hematology/BMT Service, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Enrico Maffini
- Hematology and Stem Cell Transplant Unit, Romagna Transplant Network, Ravenna, Italy
| | - Gianfranco Pittari
- Department of Medical Oncology, Hematology/BMT Service, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Benedetto Bruno
- Department of Oncology/Hematology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| |
Collapse
|
586
|
Du M, Hari P, Hu Y, Mei H. Biomarkers in individualized management of chimeric antigen receptor T cell therapy. Biomark Res 2020; 8:13. [PMID: 32426136 PMCID: PMC7216329 DOI: 10.1186/s40364-020-00190-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
The development of chimeric antigen receptor (CAR) T cell immunotherapy has achieved promising results, both in clinical studies and in commercial products for patients with hematologic malignancies. Despite high remission rates of CAR-T cell therapy in previously untreatable, refractory and/or relapsed patients, several challenges in CAR-T therapy remain to be overcome, especially in integrating such therapies into personalized disease management approaches. Given the unique characteristics of CAR-T therapy, it is particularly urgent to identify biomarkers to maximize their clinical benefits. This systematic review summarizes clinically relevant biomarkers that may help individualized disease management in patients receiving CAR-T cell therapy in terms of toxicity warning, efficacy prediction and relapse monitoring. We summarize data from 18 clinical trials, including traditional indicators like cytokines, biochemical proteins, tumor burden, as well as potential novel indicators such as CAR-T cell expansion and persistency. The establishment of a biomarker-based system aimed at individualized management is recommended to guide better clinical application of CAR-T products.
Collapse
Affiliation(s)
- Mengyi Du
- 1Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Parameswaran Hari
- 2Division of Hematology/Oncology, Medical College of Wisconsin (MCW), Milwaukee, WI USA
| | - Yu Hu
- 1Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Heng Mei
- 1Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
587
|
Hong KT. Immunotherapy in Pediatric Hematologic Malignant Neoplasms. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2020. [DOI: 10.15264/cpho.2020.27.1.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Kyung Taek Hong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Seoul, Korea
| |
Collapse
|
588
|
Futami M, Suzuki K, Kato S, Ohmae S, Tahara Y, Nojima M, Imai Y, Mimura T, Watanabe Y, Tojo A. The novel multi-cytokine inhibitor TO-207 specifically inhibits pro-inflammatory cytokine secretion in monocytes without affecting the killing ability of CAR T cells. PLoS One 2020; 15:e0231896. [PMID: 32320454 PMCID: PMC7176125 DOI: 10.1371/journal.pone.0231896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy using chimeric antigen receptor–armed T (CAR T) cells have been shown to improve outcomes significantly in patients with hematological malignancies. However, cytokine release syndrome (CRS) remains a risk. CRS is characterized by the excessive activation of CAR T cells and macrophages. Signs and symptoms of CRS are usually resolved after steroid administration, but steroids abrogate the expansion and persistence of CAR T cell populations. Tocilizumab is a humanized monoclonal antibody (mAb) that attenuates CRS without significant loss of CAR T cell activity. However, interleukin-6 (IL-6)/IL-6 receptor (IL-6R) blockade alone cannot relieve CRS symptoms fully, and novel treatments are needed to prevent or cure CRS. TO-207 is an N-benzoyl-L-phenylalanine derivative that significantly inhibits inflammatory cytokine production in human monocyte and macrophage-specific manner. We investigated whether TO-207 could inhibit cytokine production without impairing CAR T cell function in a CRS-simulating co-culture system.
Collapse
Affiliation(s)
- Muneyoshi Futami
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| | - Keisuke Suzuki
- Research laboratories, Torii Pharmaceutical., Sakura-shi, Japan
| | - Satomi Kato
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Saori Ohmae
- Research laboratories, Torii Pharmaceutical., Sakura-shi, Japan
| | - Yoshio Tahara
- Research laboratories, Torii Pharmaceutical., Sakura-shi, Japan
| | - Masanori Nojima
- Center for Translational Research, The Institute of Medical Science Hospital, The University of Tokyo, Tokyo, Japan
| | - Yoichi Imai
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takayuki Mimura
- Research laboratories, Torii Pharmaceutical., Sakura-shi, Japan
| | - Yoshihiro Watanabe
- Research laboratories, Torii Pharmaceutical., Sakura-shi, Japan
- Innovative Clinical Research Center, Kanazawa University, Kanazawa, Japan
| | - Arinobu Tojo
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
589
|
Tan AHJ, Vinanica N, Campana D. Chimeric antigen receptor-T cells with cytokine neutralizing capacity. Blood Adv 2020; 4:1419-1431. [PMID: 32271901 PMCID: PMC7160280 DOI: 10.1182/bloodadvances.2019001287] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
Infusion of T lymphocytes expressing chimeric antigen receptors (CARs) can produce extraordinary antitumor activity in patients with leukemia, lymphoma, and myeloma. The signaling mechanisms activating T cells and provoking tumor cell killing also trigger cytokine secretion and macrophage activation, leading to cytokine release syndrome (CRS). CRS is a serious side effect of CAR-T cells, and proinflammatory interleukin-6 (IL-6) is central to its pathogenesis. To endow T cells with anti-CRS activity, we designed a nonsignaling membrane-bound IL-6 receptor (mbaIL6) constituted by a single chain variable fragment derived from an anti-IL-6 antibody linked to a transmembrane anchoring peptide. We found that mbaIL6 expressed on the surface of T cells could rapidly remove IL-6 from the culture supernatant. IL-6 removal was proportional to the number of mbaIL6+ cells, increased with T-cell proliferation, and neutralized IL-6 signaling and function. A construct encoding for mbaIL6 and an anti-CD19-41BB-CD3ζ CAR allowed simultaneous expression of both receptors. T cells with mbaIL6 and CAR neutralized macrophage-derived IL-6 while exerting powerful antitumor activity. Cytotoxicity and proliferation were identical to those of cells expressing CAR alone in vitro, and CAR-T cells were effective in xenograft models regardless of mbaIL6 expression. Levels of human IL-6 in mice, however, were greatly reduced if T cells expressed both receptors instead of CAR alone. Thus, CAR-T cells with on-board capacity to extinguish IL-6 represent a new approach to prevent CRS and suppress its severity without affecting the antitumor potential of CAR-T cells.
Collapse
Affiliation(s)
- Adrian H J Tan
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Natasha Vinanica
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Dario Campana
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
590
|
Long B, Brém E, Koyfman A. Oncologic Emergencies: Immune-Based Cancer Therapies and Complications. West J Emerg Med 2020; 21:566-580. [PMID: 32421502 PMCID: PMC7234690 DOI: 10.5811/westjem.2020.1.45898] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer therapies have undergone several recent advancements. Current cancer treatments include immune-based therapies comprised of checkpoint inhibitors, and adoptive immunotherapy; each treatment has the potential for complications that differ from chemotherapy and radiation. This review evaluates immune-based therapies and their complications for emergency clinicians. Therapy complications include immune-related adverse events (irAE), cytokine release syndrome (CRS), autoimmune toxicity, and chimeric antigen receptor (CAR) T-cell-related encephalopathy syndrome (CRES). Immune-related adverse events are most commonly encountered with checkpoint inhibitors and include dermatologic complications, pneumonitis, colitis/diarrhea, hepatitis, and endocrinopathies. Less common irAEs include nephritis, myocardial injury, neurologic toxicity, ocular diseases, and musculoskeletal complications. CRS and CRES are more commonly associated with CAR T-cell therapy. CRS commonly presents with flu-like illness and symptoms resembling sepsis, but severe myocardial and pulmonary disease may occur. Critically ill patients require resuscitation, broad-spectrum antibiotics, and hematology/oncology consultation.
Collapse
Affiliation(s)
- Brit Long
- Brooke Army Medical Center, Department of Emergency Medicine, Fort Sam Houston, Texas
| | - Elizabeth Brém
- University of California, Irvine Health, Division of Hematology/Oncology, Orange, California
| | - Alex Koyfman
- The University of Texas Southwestern Medical Center, Department of Emergency Medicine, Dallas, Texas
| |
Collapse
|
591
|
Extracorporeal cytokine adsorption for treating severe refractory cytokine release syndrome (CRS). Bone Marrow Transplant 2020; 55:2052-2055. [PMID: 32277144 DOI: 10.1038/s41409-020-0896-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023]
|
592
|
Natural Killer Cell Responses in Hepatocellular Carcinoma: Implications for Novel Immunotherapeutic Approaches. Cancers (Basel) 2020; 12:cancers12040926. [PMID: 32283827 PMCID: PMC7226319 DOI: 10.3390/cancers12040926] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) still represents a significant complication of chronic liver disease, particularly when cirrhosis ensues. Current treatment options include surgery, loco-regional procedures and chemotherapy, according to specific clinical practice guidelines. Immunotherapy with check-point inhibitors, aimed at rescuing T-cells from exhaustion, has been applied as second-line therapy with limited and variable success. Natural killer (NK) cells are an essential component of innate immunity against cancer and changes in phenotype and function have been described in patients with HCC, who also show perturbations of NK activating receptor/ligand axes. Here we discuss the current status of NK cell treatment of HCC on the basis of existing evidence and ongoing clinical trials on adoptive transfer of autologous or allogeneic NK cells ex vivo or after activation with cytokines such as IL-15 and use of antibodies to target cell-expressed molecules to promote antibody-dependent cellular cytotoxicity (ADCC). To this end, bi-, tri- and tetra-specific killer cell engagers are being devised to improve NK cell recognition of tumor cells, circumventing tumor immune escape and efficiently targeting NK cells to tumors. Moreover, the exciting technique of chimeric antigen receptor (CAR)-engineered NK cells offers unique opportunities to create CAR-NK with multiple specificities along the experience gained with CAR-T cells with potentially less adverse effects.
Collapse
|
593
|
Rubinstein JD, Nelson AS, Krupski C, O'Brien W, Taylor JM, Badgett TC, Huang M, Davies SM, Phillips CL. Chimeric antigen receptor T-cell therapy in patients with neurologic comorbidities. Pediatr Blood Cancer 2020; 67:e28199. [PMID: 32020723 DOI: 10.1002/pbc.28199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 11/07/2022]
Abstract
Chimeric antigen receptor T cells (CAR-T) are an effective and potentially durable treatment for refractory and multiply relapsed B-cell acute lymphoblastic leukemia. Neurotoxicity is frequent after CAR-T cell therapy. Mechanisms driving neurotoxicity are incompletely understood, and symptoms can range from transient and mild to severe and life-threatening. Providers have exercised caution in providing CAR-T to patients with neurological comorbidities or extramedullary disease. Here, we report three patients with prior significant neurologic morbidity who safely tolerated CAR-T cell infusion after bridging therapy with conventional chemotherapy.
Collapse
Affiliation(s)
- Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Oncology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam S Nelson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christa Krupski
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - William O'Brien
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - J Michael Taylor
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tom C Badgett
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Michael Huang
- Pediatric Cancer and Blood Disorders, University of Louisville School of Medicine, Louisville, Kentucky
| | - Stella M Davies
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine L Phillips
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Oncology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
594
|
Wang J, Mou N, Yang Z, Li Q, Jiang Y, Meng J, Liu X, Deng Q. Efficacy and safety of humanized anti-CD19-CAR-T therapy following intensive lymphodepleting chemotherapy for refractory/relapsed B acute lymphoblastic leukaemia. Br J Haematol 2020; 191:212-222. [PMID: 32232846 PMCID: PMC7687133 DOI: 10.1111/bjh.16623] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/09/2020] [Indexed: 12/27/2022]
Abstract
We studied the efficacy and safety of humanized CAR-T therapy following intensive chemotherapy for refractory/relapsed (R/R) acute lymphoblastic leukaemia (B-ALL). Twenty-three patients with R/R B-ALL were pretreated with intensive chemotherapy (fludarabine combined with medium-dose cytarabine) 12 days before CAR-T therapy. Adverse events (AEs), curative effects, infection indicators and cytokine release syndrome (CRS) were monitored. Each of the 23 patients received a dose of 1·0 × 106 cells/kg CAR-T cell infusion on day 0. After 14 days, 19 patients (82·61%) achieved complete response (CR) or CR with incomplete count recovery. No survival benefit was achieved with consolidative haematopoietic stem-cell transplantation (HSCT), with a median follow-up of 14·0 months (range, 1·5-21·0 months). The notable AEs were grade 1-2 CRS in 18 patients, while the other five patients were grade 3 CRS. No patients died of CRS. Only one patient died of respiratory failure due to cytomegalovirus infection 24 days after infusion. The proportion of leukaemic cells in bone marrow on infusion day and the peaks of IL-6, TNF-α and IL-8 levels were correlated with CRS levels. A lower disease burden was achieved by intensive lymphodepleting chemotherapy, and the subsequent CAR-T therapy had a high response and manageable toxicity. Trial registration: The patients were enrolled in a clinical trial of ChiCTR-ONN-16009862, and ChiCTR1800019622.
Collapse
Affiliation(s)
- Jia Wang
- Department of HematologyTianjin First Central HospitalTianjinChina
| | - Nan Mou
- Shanghai Genbase Biotechnology Co., LtdShanghaiChina
| | - Zhenxing Yang
- Shanghai Genbase Biotechnology Co., LtdShanghaiChina
| | - Qing Li
- Department of HematologyTianjin First Central HospitalTianjinChina
| | - Yanyu Jiang
- Department of HematologyTianjin First Central HospitalTianjinChina
| | - Juanxia Meng
- Department of HematologyTianjin First Central HospitalTianjinChina
| | - Xuxiang Liu
- Department of PathologyCity of Hope National Medical CenterDuarteCAUSA
| | - Qi Deng
- Department of HematologyTianjin First Central HospitalTianjinChina
| |
Collapse
|
595
|
Petty AJ, Heyman B, Yang Y. Chimeric Antigen Receptor Cell Therapy: Overcoming Obstacles to Battle Cancer. Cancers (Basel) 2020; 12:cancers12040842. [PMID: 32244520 PMCID: PMC7226583 DOI: 10.3390/cancers12040842] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 01/04/2023] Open
Abstract
Chimeric antigen receptors (CAR) are fusion proteins engineered from antigen recognition, signaling, and costimulatory domains that can be used to reprogram T cells to specifically target tumor cells expressing specific antigens. Current CAR-T cell technology utilizes the patient's own T cells to stably express CARs and has achieved exciting clinical success in the past few years. However, current CAR-T cell therapy still faces several challenges, including suboptimal persistence and potency, impaired trafficking to solid tumors, local immunosuppression within the tumor microenvironment and intrinsic toxicity associated with CAR-T cells. This review focuses on recent strategies to improve the clinical efficacy of CAR-T cell therapy and other exciting CAR approaches currently under investigation, including CAR natural killer (NK) and NKT cell therapies.
Collapse
Affiliation(s)
- Amy J. Petty
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Benjamin Heyman
- Division of Regenerative Medicine, Department of Medicine, UC San Diego, La Jolla, CA 92093, USA
- Correspondence: (B.H.); (Y.Y.)
| | - Yiping Yang
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (B.H.); (Y.Y.)
| |
Collapse
|
596
|
Treatment response, survival, safety, and predictive factors to chimeric antigen receptor T cell therapy in Chinese relapsed or refractory B cell acute lymphoblast leukemia patients. Cell Death Dis 2020; 11:207. [PMID: 32231200 PMCID: PMC7105502 DOI: 10.1038/s41419-020-2388-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
This study aimed to evaluate treatment response, survival, safety profiles, and predictive factors to chimeric antigen receptor T cell (CAR-T) therapy in Chinese patients with relapsed or refractory B cell acute lymphoblast leukemia (R/R B-ALL). 39R/R B-ALL patients who underwent CAR-T therapy were included. Baseline data were collected from patients’ electronic medical records. Patients’ peripheral bloods, bone marrow aspirates, and biopsies were obtained for routine examination, and treatment response and survival profiles as well as adverse events were evaluated. The rates of complete remission (CR), CR with minimal residual disease (MRD) negative/positive, and bridging to hematopoietic stem-cell transplantation (HSCT) were 92.3%, 76.9%, 15.4%, and 43.6%, respectively. The median event-free survival (EFS) was 11.6 months (95% confidence interval (CI): 4.0–19.2 months) and median overall survival (OS) was 14.0 months (95% CI: 10.9–17.1 months). Bridging to HSCT independently predicted better EFS and OS, while high bone marrow blasts level independently predicted worse EFS. The incidence of cytokine release syndrome (CRS) was 97.4%, and refractory disease as well as decreased white blood cell independently predicted higher risk of severe CRS. Other common adverse events included hematologic toxicities (grade I: 5.1%, grade II: 7.7%, grade III: 17.9%, grade IV: 69.2%), neurotoxicity (28.2%), infection (38.5%), and admission for intensive care unit (10.3%). In conclusion, CAR-T therapy presents with promising treatment response, survival and safety profiles, and higher disease burden predicts worse survival as well as increased risk of severe CRS in Chinese R/R B-ALL patients.
Collapse
|
597
|
You Have Got a Fast CAR: Chimeric Antigen Receptor NK Cells in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12030706. [PMID: 32192067 PMCID: PMC7140022 DOI: 10.3390/cancers12030706] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022] Open
Abstract
The clinical success stories of chimeric antigen receptor (CAR)-T cell therapy against B-cell malignancies have contributed to immunotherapy being at the forefront of cancer therapy today. Their success has fueled interest in improving CAR constructs, identifying additional antigens to target, and clinically evaluating them across a wide range of malignancies. However, along with the exciting potential of CAR-T therapy comes the real possibility of serious side effects. While the FDA has approved commercialized CAR-T cell therapy, challenges associated with manufacturing, costs, and related toxicities have resulted in increased attention being paid to implementing CAR technology in innate cytotoxic natural killer (NK) cells. Here, we review the current landscape of the CAR-NK field, from successful clinical implementation to outstanding challenges which remain to be addressed to deliver the full potential of this therapy to more patients.
Collapse
|
598
|
Quintás-Cardama A. What CAR Will Win the CD19 Race? Mol Cancer Ther 2020; 18:498-506. [PMID: 30824581 DOI: 10.1158/1535-7163.mct-18-1070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/27/2018] [Accepted: 01/15/2019] [Indexed: 11/16/2022]
Abstract
Adoptive transfer of T cells engineered with synthetic receptors is emerging as a new pillar in the treatment of cancer. The adoptive cell therapy furthest along in clinical development is the engineering of T cells to express chimeric antigen receptors (CAR) against the CD19 antigen. Several platforms have shown remarkable activity in patients with relapsed or refractory B-cell malignancies. In 2017, the FDA approved the first CAR T cell products tisagenlecleucel (Kymriah, Novartis) and axicabtagene ciloleucel (Yescarta, Gilead), and others are expected to follow shortly. Despite their activity, CAR T cell approaches have limitations that will need to be addressed, including excessive toxicity, relapses mediated via antigen escape, difficulties overcoming the suppressive tumor microenvironment, high manufacturing costs and retail prices, and patient access, among others. The CAR T cell product that better addresses those challenges will obtain a critical competitive advantage.
Collapse
|
599
|
McNeer JL, Rau RE, Gupta S, Maude SL, O'Brien MM. Cutting to the Front of the Line: Immunotherapy for Childhood Acute Lymphoblastic Leukemia. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32320280 DOI: 10.1200/edbk_278171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Although many children and young adults with B-cell acute lymphoblastic leukemia (B-ALL) are cured with modern, risk-adapted chemotherapy regimens, 10% to 15% of patients will experience relapse or have refractory disease. Recent efforts to further intensify cytotoxic chemotherapy regimens in the frontline setting have failed as a result of excessive toxicity or lack of improvement in efficacy. As a result, novel approaches will be required to achieve cures in more newly diagnosed patients. Multiple immune-based therapies have demonstrated considerable efficacy in the setting of relapsed or refractory (R/R) disease, including CD19 targeting with blinatumomab and tisagenlecleucel and CD22 targeting with inotuzumab ozogamicin. These agents are now under investigation by the Children's Oncology Group (COG) in clinical trials for newly diagnosed B-ALL, with integration into standard chemotherapy regimens based on clinically and biology-based risk stratification as well as disease response.
Collapse
Affiliation(s)
| | - Rachel E Rau
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Sumit Gupta
- The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Shannon L Maude
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Maureen M O'Brien
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
600
|
Abstract
Cancer immunotherapies, including checkpoint inhibitors and adoptive cell therapy, manipulate the immune system to recognize and attack cancer cells. These therapies have the potential to induce durable responses in multiple solid and hematologic malignancies and thus have transformed treatment algorithms for numerous tumor types. Cancer immunotherapies lead to unique toxicity profiles distinct from the toxicities of other cancer therapies, depending on their mechanism of action. These toxicities often require specific management, which can include steroids and immune-modulating therapy and for which consensus guidelines have been published. This review will focus on the toxicities of checkpoint inhibitors and chimeric antigen receptor T cells, including pathophysiology, diagnosis, and management.
Collapse
Affiliation(s)
| | - April K S Salama
- Division of Medical Oncology, Duke University, Durham, North Carolina
| |
Collapse
|