251
|
Abstract
Advances in cancer therapies have significantly improved patient outcomes. However, with improvements in survival, the toxicities associated with cancer therapy have become of paramount importance and oncologists are faced with the challenge of establishing therapeutic efficacy while minimizing toxicity. Cardiovascular disease represents a significant risk to survivors of childhood cancer and is a major cause of morbidity and mortality. This article outlines the current state of knowledge regarding cardiotoxicity in children undergoing cancer therapies, including the impact of specific oncologic therapies, recommendations for cardiovascular screening, the management of established cardiac disease, and the evolving field of pediatric cardio-oncology.
Collapse
Affiliation(s)
- Thomas D Ryan
- Department of Pediatrics, Division of Pediatric Cardiology, Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 2003, Cincinnati, OH 45229, USA.
| | - Rajaram Nagarajan
- Department of Pediatrics, Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 7018, Cincinnati, OH 45229, USA
| | - Justin Godown
- Department of Pediatrics, Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Suite 5230 DOT, Nashville, TN 37232, USA
| |
Collapse
|
252
|
Thakar MS, Kearl TJ, Malarkannan S. Controlling Cytokine Release Syndrome to Harness the Full Potential of CAR-Based Cellular Therapy. Front Oncol 2020; 9:1529. [PMID: 32076597 PMCID: PMC7006459 DOI: 10.3389/fonc.2019.01529] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 12/18/2019] [Indexed: 01/25/2023] Open
Abstract
Chimeric Antigen Receptor (CAR)-based therapies offer a promising, targeted approach to effectively treat relapsed or refractory B cell malignancies. However, the treatment-related toxicity defined as cytokine-release syndrome (CRS) often develops in patients, and if uncontrolled, can be fatal. Grading systems have now been developed to further characterize and objectify clinical findings in order to provide algorithm-based guidance on CRS-related treatment decisions. The pharmacological treatments associated with these algorithms suppress inflammation through a variety of mechanisms and are paramount to improving the safety profile of CAR-based therapies. However, fatalities are still occurring, and there are downsides to these treatments, including the possibility of disrupting CAR-T cell persistence. This review article will describe the clinical presentation and current management of CRS, and through our now deeper understanding of downstream signaling pathways, will provide a molecular framework to formulate new hypotheses regarding clinical applications to contain proinflammatory cytokines responsible for CRS.
Collapse
Affiliation(s)
- Monica S Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tyce J Kearl
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Center of Excellence in Prostate Cancer, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
253
|
Multimodal Therapeutic Approach of Cytokine Release Syndrome Developing in a Child Given Chimeric Antigen Receptor-Modified T Cell Infusion. Crit Care Explor 2020; 2:e0071. [PMID: 32166291 PMCID: PMC7063902 DOI: 10.1097/cce.0000000000000071] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Objectives: To describe a pediatric case of cytokine release syndrome secondary to chimeric antigen receptor-modified T cells associated with acute respiratory distress syndrome. Design: Case report. Setting: PICU. Patients: A 14-year-old boy with refractory B cell precursor acute lymphoblastic leukemia given chimeric antigen receptor cells developed severe cytokine release syndrome 7 days after the drug product infusion with progressive respiratory failure. He was admitted to PICU with a clinical picture of acute respiratory distress syndrome, requiring mechanical ventilation, and secondary hemophagocytic lymphohistiocytosis. Interventions: Hemoadsorption with cartridge column (Cytosorb) in combination with continuous renal replacement therapy was associated to the anti-cytokine therapy (tocilizumab, a monoclonal antibody targeting interleukin-6 receptor). Measurements and Main Results: Decrease of the inflammatory biomarkers (ferritin, interleukin-6, interleukin-10) in the first 96 hours associated with a progressive improvement of acute respiratory distress syndrome (Pao2/Fio2 ratio) 7 day after the start of the multimodal treatment. Conclusions: This case suggests that hemoadsorption with cartridge column in combination with continuous renal replacement therapy and tocilizumab is safe and potentially effective in pediatric patients with severe cytokine release syndrome.
Collapse
|
254
|
Gupta S, Seethapathy H, Strohbehn IA, Frigault MJ, O'Donnell EK, Jacobson CA, Motwani SS, Parikh SM, Curhan GC, Reynolds KL, Leaf DE, Sise ME. Acute Kidney Injury and Electrolyte Abnormalities After Chimeric Antigen Receptor T-Cell (CAR-T) Therapy for Diffuse Large B-Cell Lymphoma. Am J Kidney Dis 2020; 76:63-71. [PMID: 31973908 DOI: 10.1053/j.ajkd.2019.10.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023]
Abstract
RATIONALE & OBJECTIVE Cytokine release syndrome is a well-known complication of chimeric antigen receptor T-cell (CAR-T) therapy and can lead to multiorgan dysfunction. However, the nephrotoxicity of CAR-T therapy is unknown. We aimed to characterize the occurrence, cause, and outcomes of acute kidney injury (AKI), along with the occurrence of electrolyte abnormalities, among adults with diffuse large B-cell lymphoma receiving CAR-T therapy. STUDY DESIGN Case series. SETTING & PARTICIPANTS We reviewed the course of 78 adults receiving CAR-T therapy with axicabtagene ciloleucel or tisagenlecleucel at 2 major cancer centers between October 2017 and February 2019. Baseline demographics, comorbid conditions, medications, and laboratory values were obtained from electronic health records. AKI was defined using KDIGO (Kidney Disease: Improving Global Outcomes) criteria. The cause, clinical course, and outcome of AKI events and electrolyte abnormalities in the first 30 days after CAR-T infusion were characterized using data contained in electronic health records. RESULTS Among 78 patients receiving CAR-T therapy, cytokine release syndrome occurred in 85%, of whom 62% were treated with tocilizumab. AKI occurred in 15 patients (19%): 8 had decreased kidney perfusion, 6 developed acute tubular necrosis, and 1 patient had urinary obstruction related to disease progression. Those with acute tubular necrosis and obstruction had the longest lengths of stay and highest 60-day mortality. Electrolyte abnormalities were common; hypophosphatemia, hypokalemia, and hyponatremia occurred in 75%, 56%, and 51% of patients, respectively. LIMITATIONS Small sample size; AKI adjudicated by retrospective chart review; lack of biopsy data. CONCLUSIONS In this case series of patients with diffuse large B-cell lymphoma receiving CAR-T therapy, AKI and electrolyte abnormalities occurred commonly in the context of cytokine release syndrome.
Collapse
Affiliation(s)
- Shruti Gupta
- Division of Renal Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Harish Seethapathy
- Renal Division, Department of Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Ian A Strohbehn
- Renal Division, Department of Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Matthew J Frigault
- Oncology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Elizabeth K O'Donnell
- Oncology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Shveta S Motwani
- Division of Renal Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA; Dana Farber Cancer Institute, Boston, MA
| | - Samir M Parikh
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Gary C Curhan
- Division of Renal Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Kerry L Reynolds
- Oncology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - David E Leaf
- Division of Renal Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Meghan E Sise
- Renal Division, Department of Internal Medicine, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
255
|
Sabahi M, Jabbari P, Alizadeh Haghighi M, Soltani S, Soudi S, Rahmani F, Rezaei N. Proposing a tandem AND-gate CAR T cell targeting glioblastoma multiforme. Med Hypotheses 2020; 137:109559. [PMID: 31962251 DOI: 10.1016/j.mehy.2020.109559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/30/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022]
Abstract
CAR T cell therapy is suggested as an effective method to treat hematological malignancies. However, high recurrence rates and in vivo toxicities have limited their widespread use. In order to reduce toxicity and improve tumor specificity, we propose a CAR T cell targeting glioblastoma multiforme utilizing the synNotch receptor pathway linked to a tandem CAR T cell. The extracellular domain of the synNotch receptor is replaced by a single chain fragment variable specific for the EGF receptor variant III (scfv-EGFRvIII), and covalently bonded to a IL-13Rα2-CD133-tandem CAR. This would produce an AND-gate CAR-T cell, which requires activation of both signals from synNotch receptor binding to EGFRvIII and then binding of the tandem CAR to either of the two IL-13Rα2 or CD133 ligands-specific antigens for glioblastoma stem cells. SynNotch receptor activation along with the 4-1BB costimulatory domain results in CAR T cell expression under the TRE promoter, culminating in a tri-specific and effective tumor stem cell recognition and elimination of glioblastoma multiforme.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Neurosurgery Research Group (NRG), Student Reaserch Committee, Hamadan University of Medical Sciences, Hamadan, Iran; Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Jabbari
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Milad Alizadeh Haghighi
- Neurosurgery Research Group (NRG), Student Reaserch Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Setare Soltani
- Student Reaserch Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farzaneh Rahmani
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
256
|
The renal adverse effects of cancer immunotherapy. J Nephrol 2020; 33:467-481. [PMID: 31912436 DOI: 10.1007/s40620-019-00691-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022]
Abstract
Over the past decade, the development and clinical use of immunotherapy agents has increased exponentially. As clinical experience builds with these agents so too does our understanding of the associated adverse effects. In particular, the effects of immunotherapy on the kidneys, individual nephrons, and kidney function remain less well described than the adverse effects on barrier organ systems such as the gastrointestinal tract and skin. However, phase IV post-marketing surveillance and clinical case studies together with basic research has begun to reveal mechanisms by which immunotherapy mediates renal adverse effects. This work may lead to improvements in treatment guidelines and therapy. These advances are particularly important as post-cancer survival increases leaving patients to cope with the consequences of not only the cancer, but the short- and long-term adverse effects of treatment. Here we discuss the major renal adverse effects encountered with individual immunotherapeutic agents, putative mechanisms, their current management, and how cancer survivorship programs can help patients who have been treated with immunotherapy.
Collapse
|
257
|
Nicolaysen A. Nephrotoxic Chemotherapy Agents: Old and New. Adv Chronic Kidney Dis 2020; 27:38-49. [PMID: 32147000 DOI: 10.1053/j.ackd.2019.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
In the last several decades, advancements in chemotherapy have improved the overall survival of cancer patients. These agents, however, are associated with adverse effects, including various kidney lesions. This review summarizes the nephrotoxic potential of chemotherapy agents, old and new, as well as the different factors that contribute to kidney injury. Provided for each class of chemotherapy agent is the associated kidney lesion and a brief discussion of clinical manifestation, mechanism of action, and possible treatment when available. Understanding the nephrotoxic potential of these agents have on the kidneys is imperative for both the oncologist and the nephrologist to properly care for cancer patients and ensure their best outcomes.
Collapse
|
258
|
Abstract
Lung injury associated with cancer therapeutics is often the limiting factor that trumps otherwise successful cancer therapy. Thoracic radiation as well as cancer pharmacotherapeutics, including conventional chemotherapy, molecular targeted agents, and cancer immunotherapies, have been associated with a unique spectrum of histopathologic injury patterns that may involve the lung parenchyma, pleura, airways, and/or pulmonary vasculature. Injury patterns may be idiosyncratic, unpredictable, and highly variable from one agent class to the next. Variability in lung injury patterns within a specific therapeutic class of drugs also occurs, adding to the conundrum. Drug-induced toxicities to the thoracic cavity are infrequent, and early recognition of clinical clues portends a good outcome in most cases. Failure to recognize early clinical signs, however, may result in irreversible and potentially lethal consequences. This chapter provides an overview of our current knowledge of thoracic complications associated with cancer pharmacotherapies. The review is not intended to be a treatise of all cancer agents that adversely affect the lungs, but rather a discussion of established risk factors and histopathologic patterns of lung injury associated with broad classes of cancer agents. Optimal management strategies, based on existing clinical experience, will also be discussed. Complications associated with thoracic radiation are also reviewed. It is hoped that these discussions will facilitate early recognition and management of treatment-related thoracic complications and, ultimately, better patient outcomes.
Collapse
Affiliation(s)
- Joseph L. Nates
- Department of Critical Care and Respiratory Care, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Kristen J. Price
- Division of Anesthesiology, Critical Care and Pain Medicine, Department of Critical Care and Respiratory Care, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
259
|
Abreu TR, Fonseca NA, Gonçalves N, Moreira JN. Current challenges and emerging opportunities of CAR-T cell therapies. J Control Release 2019; 319:246-261. [PMID: 31899268 DOI: 10.1016/j.jconrel.2019.12.047] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/27/2022]
Abstract
Infusion of chimeric antigen receptor (CAR)-genetically modified T cells (CAR-T cells) have led to remarkable clinical responses and cancer remission in patients suffering from relapsed or refractory B-cell malignancies. This is a new form of adoptive T cell therapy (ACT), whereby the artificial CAR enables the redirection of T cells endogenous antitumor activity towards a predefined tumor-associated antigen, leading to the elimination of a specific tumor. The early success in blood cancers has prompted the US Food and Drug Administration (FDA) to approve the first CAR-T cell therapies for the treatment of CD19-positive leukemias and lymphomas in 2017. Despite the emergence of CAR-T cells as one of the latest breakthroughs of cancer immunotherapies, their wider application has been hampered by specific life-threatening toxicities, and a substantial lack of efficacy in the treatment of solid tumors, owing to the strong immunosuppressive tumor microenvironment and the paucity of reliable tumor-specific targets. Herein, besides providing an overview of the emerging CAR-technologies and current clinical applications, the major hurdles of CAR-T cell therapies will be discussed, namely treatment-related life-threatening toxicities and the obstacles posed by the immunosupressive tumor-microenvironment of solid tumors, as well as the next-generation strategies currently designed to simultaneously improve safety and efficacy of CAR-T cell therapies in vivo.
Collapse
Affiliation(s)
- Teresa R Abreu
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Nuno A Fonseca
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; TREAT U, SA, Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal.
| | - Nélio Gonçalves
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal.
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
260
|
Hughes-Parry HE, Cross RS, Jenkins MR. The Evolving Protein Engineering in the Design of Chimeric Antigen Receptor T Cells. Int J Mol Sci 2019; 21:ijms21010204. [PMID: 31892219 PMCID: PMC6981602 DOI: 10.3390/ijms21010204] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/24/2022] Open
Abstract
The clinical success of chimeric antigen receptor (CAR) T cell immunotherapy in the treatment of haematological cancers has encouraged the extensive development of CAR design to improve their function and increase their applicability. Advancements in protein engineering have seen modifications to both the ecto- and endo-domains of the CAR, with recent designs targeting multiple antigens and including inducible elements. These developments are likely to play an important role in inducing effective CAR T cell responses in a solid tumour context, where clinical responses have not been effective to date. This review highlights the spectrum of novel strategies being employed in CAR design, including for example variations in targeting tumour antigens by utilising different ectodomain designs such as dual chain CARs, natural receptor or ligand-based CARs, and T cell receptor fusion constructs, and also reviews some of the innovative approaches to a "universal" CAR and various multi-antigen targeting CAR strategies. We also explore how choices in the endodomain impact CAR function and how these need to be considered in the overall CAR design.
Collapse
Affiliation(s)
- Hannah E. Hughes-Parry
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (H.E.H.-P.); (R.S.C.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Ryan S. Cross
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (H.E.H.-P.); (R.S.C.)
| | - Misty R. Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (H.E.H.-P.); (R.S.C.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
- Institute of Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
261
|
Slattery K, Gardiner CM. NK Cell Metabolism and TGFβ - Implications for Immunotherapy. Front Immunol 2019; 10:2915. [PMID: 31921174 PMCID: PMC6927492 DOI: 10.3389/fimmu.2019.02915] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
NK cells are innate lymphocytes which play an essential role in protection against cancer and viral infection. Their functions are dictated by many factors including the receptors they express, cytokines they respond to and changes in the external environment. These cell processes are regulated within NK cells at many levels including genetic, epigenetic and expression (RNA and protein) levels. The last decade has revealed cellular metabolism as another level of immune regulation. Specific immune cells adopt metabolic configurations that support their functions, and this is a dynamic process with cells undergoing metabolic reprogramming during the course of an immune response. Upon activation with pro-inflammatory cytokines, NK cells upregulate both glycolysis and oxphos metabolic pathways and this supports their anti-cancer functions. Perturbation of these pathways inhibits NK cell effector functions. Anti-inflammatory cytokines such as TGFβ can inhibit metabolic changes and reduce functional outputs. Although a lot remains to be learned, our knowledge of potential molecular mechanisms involved is growing quickly. This review will discuss our current knowledge on the role of TGFβ in regulating NK cell metabolism and will draw on a wider knowledge base regarding TGFβ regulation of cellular metabolic pathways, in order to highlight potential ways in which TGFβ might be targeted to contribute to the exciting progress that is being made in terms of adoptive NK cell therapies for cancer.
Collapse
Affiliation(s)
- Karen Slattery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Clair M Gardiner
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| |
Collapse
|
262
|
Herrera L, Santos S, Vesga MA, Anguita J, Martin-Ruiz I, Carrascosa T, Juan M, Eguizabal C. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci Rep 2019; 9:18729. [PMID: 31822751 PMCID: PMC6904575 DOI: 10.1038/s41598-019-55239-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
Among hematological cancers, Acute Lymphoblastic Leukemia (ALL) and Chronic Lymphocytic Leukemia (CLL) are the most common leukemia in children and elderly people respectively. Some patients do not respond to chemotherapy treatments and it is necessary to complement it with immunotherapy-based treatments such as chimeric antigen receptor (CAR) therapy, which is one of the newest and more effective treatments against these cancers and B-cell lymphoma. Although complete remission results are promising, CAR T cell therapy presents still some risks for the patients, including cytokine release syndrome (CRS) and neurotoxicity. We proposed a different immune cell source for CAR therapy that might prevent these side effects while efficiently targeting malignant cells. NK cells from different sources are a promising vehicle for CAR therapy, as they do not cause graft versus host disease (GvHD) in allogenic therapies and they are prompt to attack cancer cells without prior sensitization. We studied the efficacy of NK cells from adult peripheral blood (AB) and umbilical cord blood (CB) against different target cells in order to determine the best source for CAR therapy. AB CAR-NK cells are slightly better at killing CD19 presenting target cells and CB NK cells are easier to stimulate and they have more stable number from donor to donor. We conclude that CAR-NK cells from both sources have their advantages to be an alternative and safer candidate for CAR therapy.
Collapse
Affiliation(s)
- L Herrera
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - S Santos
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - M A Vesga
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - J Anguita
- Macrophage and Tick Vaccine Laboratory, CIC bioGUNE, Derio, Biscay, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Biscay, Spain
| | - I Martin-Ruiz
- Macrophage and Tick Vaccine Laboratory, CIC bioGUNE, Derio, Biscay, Spain
| | - T Carrascosa
- Servicio de Hematología, Hospital Galdakao-Usansolo, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - M Juan
- Servei d´Immunologia, Hospital Clínic de Barcelona, Hospital Sant Joan de Déu, Institut d'Investigacions Biomèdiques August Pi i Sunyer Hospital, Universitat de Barcelona, Barcelona, Spain
| | - C Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain. .,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain.
| |
Collapse
|
263
|
Zhao Z, Zheng L, Chen W, Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: recent advances and future perspectives. J Hematol Oncol 2019; 12:126. [PMID: 31779642 PMCID: PMC6883629 DOI: 10.1186/s13045-019-0817-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy has become an emerging strategy for the treatment of cancer. Immunotherapeutic drugs have been increasing for clinical treatment. Despite significant advances in immunotherapy, the clinical application of immunotherapy for cancer patients has some challenges associated with safety and efficacy, including autoimmune reactions, cytokine release syndrome, and vascular leak syndrome. Novel strategies, particularly improved delivery strategies, including nanoparticles, scaffolds, and hydrogels, are able to effectively target tumors and/or immune cells of interest, increase the accumulation of immunotherapies within the lesion, and reduce off-target effects. Here, we briefly describe five major types of cancer immunotherapy, including their clinical status, strengths, and weaknesses. Then, we introduce novel delivery strategies, such as nanoparticle-based delivery of immunotherapy, implantable scaffolds, injectable biomaterials for immunotherapy, and matrix-binding molecular conjugates, which can improve the efficacy and safety of immunotherapies. Also, the limitations of novel delivery strategies and challenges of clinical translation are discussed.
Collapse
Affiliation(s)
- Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Liyun Zheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Wei Weng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Jingjing Song
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China. .,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China. .,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Affiliated Lishui Hospital of Zhejiang University, The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.
| |
Collapse
|
264
|
Lesch S, Benmebarek MR, Cadilha BL, Stoiber S, Subklewe M, Endres S, Kobold S. Determinants of response and resistance to CAR T cell therapy. Semin Cancer Biol 2019; 65:80-90. [PMID: 31705998 DOI: 10.1016/j.semcancer.2019.11.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 11/03/2019] [Indexed: 12/27/2022]
Abstract
The remarkable success of chimeric antigen receptor (CAR)-engineered T cells in pre-B cell acute lymphoblastic leukemia (ALL) and B cell lymphoma led to the approval of anti-CD19 CAR T cells as the first ever CAR T cell therapy in 2017. However, with the number of CAR T cell-treated patients increasing, observations of tumor escape and resistance to CAR T cell therapy with disease relapse are demonstrating the current limitations of this therapeutic modality. Mechanisms hampering CAR T cell efficiency include limited T cell persistence, caused for example by T cell exhaustion and activation-induced cell death (AICD), as well as therapy-related toxicity. Furthermore, the physical properties, antigen heterogeneity and immunosuppressive capacities of solid tumors have prevented the success of CAR T cells in these entities. Herein we review current obstacles of CAR T cell therapy and propose strategies in order to overcome these hurdles and expand CAR T cell therapy to a broader range of cancer patients.
Collapse
Affiliation(s)
- Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Marion Subklewe
- German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany; Department of Medicine III, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany.
| |
Collapse
|
265
|
Hallaj S, Meshkini F, Chaleshtari MG, Ghorbani A, Namdar A, Soleimanpour H, Jadidi-niaragh F. Conjugated CAR T cell one step beyond conventional CAR T cell for a promising cancer immunotherapy. Cell Immunol 2019; 345:103963. [DOI: 10.1016/j.cellimm.2019.103963] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/07/2019] [Accepted: 08/08/2019] [Indexed: 02/04/2023]
|
266
|
Murthy H, Iqbal M, Chavez JC, Kharfan-Dabaja MA. Cytokine Release Syndrome: Current Perspectives. Immunotargets Ther 2019; 8:43-52. [PMID: 31754614 PMCID: PMC6825470 DOI: 10.2147/itt.s202015] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/07/2019] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor T cell (CART) therapy represents a novel and a paradigm-shifting approach to treating cancer. Recent clinical successes have widened the applicability of CD19 CART cells for the treatment of relapsed/refractory B-cell NHL, namely tisagenleclucel and axicabtagene ciloleucel. Tisagenleclucel is also approved for relapsed and/or refractory B-ALL up to age 25. CART therapy is associated with unique and potentially life-threatening toxicities, notably cytokine release syndrome (CRS). A better understanding of the pathogenesis of CRS is crucial to ensure proper management. In this review, CRS definitions, profiles, risk factors and grading systems are discussed. Finally, current and novel investigational approaches and therapies for CRS are summarized.
Collapse
Affiliation(s)
- Hemant Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
267
|
The Conundrum of Septic Shock Imitators in Patients with Hematologic Cancers: Case Presentation and Possible Differential Diagnoses. Case Rep Crit Care 2019; 2019:6561018. [PMID: 31583137 PMCID: PMC6754887 DOI: 10.1155/2019/6561018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/04/2019] [Accepted: 07/15/2019] [Indexed: 11/27/2022] Open
Abstract
The authors describe the case of a patient treated with several cycles of chemotherapy due to an advanced stage non-Hodgkin lymphoma. One daafter the last cycle, he was admitted to our Intensive Care Unit with a septic shock-like clinical picture which didn't respond to the aggressive treatment and the patient died a few hours later. The autoptical findings cast some doubts on the diagnosis, and demonstrated the presence of other factors imitating its symptoms. In this article, the mimickers of septic shock are reviewed and discussed, as some of them require an aggressive immunosuppression instead of the recommended treatment for septic shock.
Collapse
|
268
|
Diagnosis and Management of Immune Related Adverse Events (irAEs) in Cancer Immunotherapy. Biomed Pharmacother 2019; 120:109437. [PMID: 31590992 DOI: 10.1016/j.biopha.2019.109437] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint inhibitors (ICPIs) and chimeric antigen receptor (CAR) T-cell therapy are two main promising methods of immunotherapy, which have become increasingly important in cancer treatment. After the wider application of these medicine in clinic, a range of immune related adverse events (irAEs) covering almost any system arouse the concern for being randomness and unpredictability. Even if most adverse events are mild and controllable after thoughtful management, the occurrence of life-threatening toxicities should not be ignored because of the insidious and atypical symptoms, which makes the early diagnosis even more challenging. In this review, a brief introduction of immunotherapy and mechanisms underlying irAEs is involved. We mainly focus on the early diagnostic method and recommended management of toxicities of different systems separately, and consequently maximized effectiveness of immunotherapy can be achieved.
Collapse
|
269
|
Witkowski MT, Lasry A, Carroll WL, Aifantis I. Immune-Based Therapies in Acute Leukemia. Trends Cancer 2019; 5:604-618. [PMID: 31706508 PMCID: PMC6859901 DOI: 10.1016/j.trecan.2019.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022]
Abstract
Treatment resistance remains a leading cause of acute leukemia-related deaths. Thus, there is an unmet need to develop novel approaches to improve outcome. New immune-based therapies with chimeric antigen receptor (CAR) T cells, bi-specific T cell engagers (BiTEs), and immune checkpoint blockers (ICBs) have emerged as effective treatment options for chemoresistant B cell acute lymphoblastic leukemia (B-ALL) and acute myeloid leukemia (AML). However, many patients show resistance to these immune-based approaches. This review describes crucial lessons learned from immune-based approaches targeting high-risk B-ALL and AML, such as the leukemia-intrinsic (e.g., target antigen loss, tumor heterogeneity) and -extrinsic (e.g., immunosuppressive microenvironment) mechanisms that drive treatment resistance, and discusses alternative approaches to enhance the effectiveness of these immune-based treatment regimens.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Disease Susceptibility
- Humans
- Immunity
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Molecular Targeted Therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Matthew T Witkowski
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| | - Audrey Lasry
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - William L Carroll
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; Department of Pediatrics, New York University School of Medicine, New York, NY 10016, USA
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
270
|
Kang S, Tanaka T, Narazaki M, Kishimoto T. Targeting Interleukin-6 Signaling in Clinic. Immunity 2019; 50:1007-1023. [PMID: 30995492 DOI: 10.1016/j.immuni.2019.03.026] [Citation(s) in RCA: 546] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine with roles in immunity, tissue regeneration, and metabolism. Rapid production of IL-6 contributes to host defense during infection and tissue injury, but excessive synthesis of IL-6 and dysregulation of IL-6 receptor signaling is involved in disease pathology. Therapeutic agents targeting the IL-6 axis are effective in rheumatoid arthritis, and applications are being extended to other settings of acute and chronic inflammation. Recent studies reveal that selective blockade of different modes of IL-6 receptor signaling has different outcomes on disease pathology, suggesting novel strategies for therapeutic intervention. However, some inflammatory diseases do not seem to respond to IL-6 blockade. Here, we review the current state of IL-6-targeting approaches in the clinic and discuss how to apply the growing understanding of the immunobiology of IL-6 to clinical decisions.
Collapse
Affiliation(s)
- Sujin Kang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita City, Osaka, Japan
| | - Toshio Tanaka
- Medical Affairs Bureau, Osaka Habikino Medical Center, 3-7-1 Habikino, Habikino City, Osaka, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita City, Osaka, Japan
| | - Tadamitsu Kishimoto
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita City, Osaka, Japan.
| |
Collapse
|
271
|
Yu S, Yi M, Qin S, Wu K. Next generation chimeric antigen receptor T cells: safety strategies to overcome toxicity. Mol Cancer 2019; 18:125. [PMID: 31429760 PMCID: PMC6701025 DOI: 10.1186/s12943-019-1057-4] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is an emerging and effective cancer immunotherapy. Especially in hematological malignancies, CAR-T cells have achieved exciting results. Two Anti-CD19 CAR-T therapies have been approved for the treatment of CD19-positive leukemia or lymphoma. However, the application of CAR-T cells is obviously hampered by the adverse effects, such as cytokines release syndrome and on-target off-tumor toxicity. In some clinical trials, patients quitted the treatment of CAR-T cells due to life-threatening toxicity. Seeking to alleviate these toxicities or prevent the occurrence, researchers have developed a number of safety strategies of CAR-T cells, including suicide genes, synthetic Notch receptor, on-switch CAR, combinatorial target-antigen recognition, bispecific T cell engager and inhibitory CAR. This review summarized the preclinical studies and clinical trials of the safety strategies of CAR-T cells and their respective strengths and weaknesses.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
272
|
Erber R, Hartmann A, Beckmann MW, Mackensen A, Kremer A, Reimann H, Hübner H, Hein A, Lux MP, Jud S, Häberle L, Gaß P, Volz B, Schulz-Wendtland R, Rübner M, Fasching PA. [TILGen study-immunological targets in patients with breast cancer : Influence of tumor-infiltrating lymphocytes]. DER PATHOLOGE 2019; 39:236-240. [PMID: 30406831 DOI: 10.1007/s00292-018-0526-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The interaction of our immune system with breast cancer (BC) cells prompted the investigation of tumor-infiltrating lymphocytes (TILs) and targeted, tumor antigen-specific immunotherapy. OBJECTIVES Correlation between TILs and pathological complete response (pCR) after neoadjuvant systemic therapy (NACT). Tumor-specific antigens (TSAs) in HER2+ and triple negative BC and establishment of TSA-specific therapies within the interdisciplinary TILGen study. METHODS Illustration of the TILGen study design. Assessment of TILs and correlation with pCR within this BC study. RESULTS pCR was achieved in 38.4% (56/146) and associated with estrogen receptor/progesterone receptor negative (ER-/PR-) and HER2+ tumors. Lymphocytic predominant BC (LPBC) was found in 16.4% (24/146), particularly in ER-/PR- (ER-: 27.3% vs. ER+: 9.9%, PR-: 22.3% vs. PR+: 8.2%), large, and poorly differentiated BC. TILs were significantly correlated with pCR in multivariate analysis. In LPBC, pCR was achieved in 66.7%, whereas it was 32.8% in non-LPBC. CONCLUSIONS First results confirm the influence of the human immune system on the response to NACT in HER2+ and triple negative BC. TSA-specific immunotherapy might improve the outcome in BC patients but there is an urgent need for comprehensive studies to further investigate this issue.
Collapse
Affiliation(s)
- R Erber
- Pathologisches Institut, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 8-10, 91054, Erlangen, Deutschland.
| | - A Hartmann
- Pathologisches Institut, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 8-10, 91054, Erlangen, Deutschland
| | - M W Beckmann
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - A Mackensen
- Medizinische Klinik 5, Hämatologie und Internistische Onkologie, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - A Kremer
- Medizinische Klinik 5, Hämatologie und Internistische Onkologie, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - H Reimann
- Medizinische Klinik 5, Hämatologie und Internistische Onkologie, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - H Hübner
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - A Hein
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - M P Lux
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - S Jud
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - L Häberle
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - P Gaß
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - B Volz
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - R Schulz-Wendtland
- Radiologisches Institut/Gynäkologische Radiologie, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - M Rübner
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| | - P A Fasching
- Frauenklinik, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Deutschland
| |
Collapse
|
273
|
Chimeric Antigen Receptor T-Cell Therapy Clinical Results in Pediatric and Young Adult B-ALL. Hemasphere 2019; 3:e279. [PMID: 31723849 PMCID: PMC6745916 DOI: 10.1097/hs9.0000000000000279] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 06/02/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cell therapy has revolutionized the care of patients with relapsed and refractory B-cell acute lymphoblastic leukemia (B-ALL). Results from clinical trials across multiple institutions report remarkable remission rates with CD19-directed CAR-modified T-cell therapy. These remissions are also proving to be durable in many patients with a relapse-free survival (RFS) of approximately 50% to 60% at 1 year across several trials and institutions in this population that has been historically very difficult to treat. In addition, new products are being developed to enhance upon the original CAR T-cell products, which include a humanized CAR, allogeneic CARs, and both CD22 and biallelic CD19 and CD22 constructs. Toxicity after CAR-modified T-cell therapy is characterized by cytokine release syndrome (CRS) and neurotoxicity in the acute post-infusion period and B-cell aplasia as a long-term consequence of treatment. This review will summarize the published data thus far on the use of CAR-modified T-cell therapy in pediatric B-ALL and outline the various CAR products now being developed for this population. Delivery of this therapy and the decision to pursue hematopoietic stem cell transplant (HSCT) after treatment will be discussed.
Collapse
|
274
|
Approach to the Adult Acute Lymphoblastic Leukemia Patient. J Clin Med 2019; 8:jcm8081175. [PMID: 31390838 PMCID: PMC6722778 DOI: 10.3390/jcm8081175] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
During recent decades, understanding of the molecular mechanisms of acute lymphoblastic leukemia (ALL) has improved considerably, resulting in better risk stratification of patients and increased survival rates. Age, white blood cell count (WBC), and specific genetic abnormalities are the most important factors that define risk groups for ALL. State-of-the-art diagnosis of ALL requires cytological and cytogenetical analyses, as well as flow cytometry and high-throughput sequencing assays. An important aspect in the diagnostic characterization of patients with ALL is the identification of the Philadelphia (Ph) chromosome, which warrants the addition of tyrosine kinase inhibitors (TKI) to the chemotherapy backbone. Data that support the benefit of hematopoietic stem cell transplantation (HSCT) in high risk patient subsets or in late relapse patients are still questioned and have yet to be determined conclusive. This article presents the newly published data in ALL workup and treatment, putting it into perspective for the attending physician in hematology and oncology.
Collapse
|
275
|
Kotch C, Barrett D, Teachey DT. Tocilizumab for the treatment of chimeric antigen receptor T cell-induced cytokine release syndrome. Expert Rev Clin Immunol 2019; 15:813-822. [PMID: 31219357 PMCID: PMC7936577 DOI: 10.1080/1744666x.2019.1629904] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 06/06/2019] [Indexed: 01/08/2023]
Abstract
Introduction: Cancer-directed immunotherapies are transforming the landscape in oncology as new and exciting therapies move from the laboratory to the bedside. Chimeric antigen receptor T (CAR-T) cells are one of these novel therapies, demonstrating impressive efficacy against B-cell malignancies. With the development of new therapies, it is not uncommon to identify new and unanticipated toxicities. CAR-T cells cause unique toxicities not typically found with traditional cytotoxic chemotherapy or small molecule inhibitors. Areas covered: CAR-T cell associated toxicities include cytokine release syndrome (CRS) and CAR-T cell-related encephalopathy syndrome (CRES), alternatively known as immune effector cell-associated neurotoxicity syndrome (ICANS). Prompt identification and management of CRS and CRES are imperative for the prevention of life-threatening complications of these innovative therapies. This literature review describes the seminal trials of CD19-directed immunotherapy and the pathophysiology and management of the toxicities found with CAR-T cells. In addition, the use of the interleukin-6 receptor antibody tocilizumab for CRS is reviewed. Expert opinion: This review describes the recommended management of CRS and CRES and examines the current limitations in management. Alternative therapies for the treatment of CAR-T cell related toxicities are also explored. Furthermore, the review proposes future directions for research.
Collapse
Affiliation(s)
- Chelsea Kotch
- a Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine , Philadelphia , PA , USA
| | - David Barrett
- a Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine , Philadelphia , PA , USA
| | - David T Teachey
- a Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine , Philadelphia , PA , USA
| |
Collapse
|
276
|
Karches CH, Benmebarek MR, Schmidbauer ML, Kurzay M, Klaus R, Geiger M, Rataj F, Cadilha BL, Lesch S, Heise C, Murr R, Vom Berg J, Jastroch M, Lamp D, Ding J, Duewell P, Niederfellner G, Sustmann C, Endres S, Klein C, Kobold S. Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy. Clin Cancer Res 2019; 25:5890-5900. [PMID: 31285373 DOI: 10.1158/1078-0432.ccr-18-3927] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/25/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Genetically engineered T cells are powerful anticancer treatments but are limited by safety and specificity issues. We herein describe an MHC-unrestricted modular platform combining autologous T cells, transduced with a targetable synthetic agonistic receptor (SAR), with bispecific antibodies (BiAb) that specifically recruit and activate T cells for tumor killing. EXPERIMENTAL DESIGN BiAbs of different formats were generated by recombinant expression. T cells were retrovirally transduced with SARs. T-cell activation, proliferation, differentiation, and T-cell-induced lysis were characterized in three murine and human tumor models in vitro and in vivo. RESULTS Murine T cells transduced with SAR composed of an extracellular domain EGFRvIII fused to CD28 and CD3ζ signaling domains could be specifically recruited toward murine tumor cells expressing EpCAM by anti-EGFRvIII × anti-EpCAM BiAb. BiAb induced selective antigen-dependent activation, proliferation of SAR T cells, and redirected tumor cell lysis. Selectivity was dependent on the monovalency of the antibody for EGFRvIII. We identified FAS ligand as a major mediator of killing utilized by the T cells. Similarly, human SAR T cells could be specifically redirected toward mesothelin-expressing human pancreatic cancer cells. In vivo, treatment with SAR T cells and BiAb mediated antitumoral activity in three human pancreatic cancer cell xenograft models. Importantly, SAR activity, unlike CAR activity, was reversible in vitro and in vivo. CONCLUSIONS We describe a novel ACT platform with antitumor activity in murine and human tumor models with a distinct mode of action that combines adoptive T-cell therapy with bispecific antibodies.
Collapse
Affiliation(s)
- Clara H Karches
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Moritz L Schmidbauer
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Mathias Kurzay
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Richard Klaus
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | | | - Felicitas Rataj
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Constanze Heise
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Ramona Murr
- Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Martin Jastroch
- Helmholtz Diabetes Center and German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | - Daniel Lamp
- Helmholtz Diabetes Center and German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | - Jian Ding
- TCR Therapeutics, Cambridge, Massachusetts
| | - Peter Duewell
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | | | | | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | | | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL).
| |
Collapse
|
277
|
Oved JH, Barrett DM, Teachey DT. Cellular therapy: Immune-related complications. Immunol Rev 2019; 290:114-126. [PMID: 31355491 PMCID: PMC7041800 DOI: 10.1111/imr.12768] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 12/17/2022]
Abstract
The advent of chimeric antigen receptor T (CAR-T) and the burgeoning field of cellular therapy has revolutionized the treatment of relapsed/refractory leukemia and lymphoma. This personalized "living therapy" is highly effective against a number of malignancies, but this efficacy is tempered by side effects relatively unique to immunotherapies, including CAR-T. The overwhelming release of cytokines and chemokines by activated CAR-T and other secondarily activated immune effector cells can lead to cytokine release syndrome (CRS), which can have clinical and pathophysiology similarities to systemic inflammatory response syndrome and macrophage activating syndrome/hemophagocytic lymphohistiocytosis. Tocilizumab, an anti-IL6 receptor antibody, was recently FDA approved for treatment of CRS after CAR-T based on its ability to mitigate CRS in many patients. Unfortunately, some patients are refractory and additional therapies are needed. Patients treated with CAR-T can also develop neurotoxicity and, as the biology is poorly understood, current therapeutic interventions are limited to supportive care. Nevertheless, a number of recent studies have shed new light on the pathophysiology of CAR-T-related neurotoxicity, which will hopefully lead to effective treatments. In this review we discuss some of the mechanistic contributions intrinsic to the CAR-T construct, the tumor being treated, and the individual patient that impact the development and severity of CRS and neurotoxicity. As CAR-T and cellular therapy have redefined the concept of personalized medicine, so too will personalization be necessary in managing the unique side effects of these therapies.
Collapse
Affiliation(s)
- Joseph H. Oved
- Divisions of Hematology and Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - David M. Barrett
- Divisions of Hematology and Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - David T. Teachey
- Divisions of Hematology and Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| |
Collapse
|
278
|
Sarkar RR, Gloude NJ, Schiff D, Murphy JD. Cost-Effectiveness of Chimeric Antigen Receptor T-Cell Therapy in Pediatric Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia. J Natl Cancer Inst 2019; 111:719-726. [PMID: 30551196 PMCID: PMC6624167 DOI: 10.1093/jnci/djy193] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/18/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR-T) therapy is a promising new class of cancer therapy but has a high up-front cost. We evaluated the cost-effectiveness of CAR-T therapy among pediatric patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). METHODS We built a microsimulation model for pediatric patients with relapsed/refractory B-ALL receiving either CAR-T therapy or standard of care. Outcomes included costs, quality of life (health utility), complications, and survival. We measured cost-effectiveness with the incremental cost-effectiveness ratio (ICER), with ICERs under $100 000 per quality-adjusted life-year (QALY) considered cost effective. One-way and probabilistic sensitivity analyses were used to test model uncertainty. RESULTS Compared to standard of care, CAR-T therapy increased overall cost by $528 200 and improved effectiveness by 8.18 QALYs, resulting in an ICER of $64 600/QALY. The model was sensitive to assumptions about long-term CAR-T survival, the complete remission rate of CAR-T patients, and the health utility of long-term survivors. The base model assumed a 76.0% one-year survival with CAR-T, although if this decreased to 57.8%, then CAR-T was no longer cost effective. If the complete remission rate of CAR-T recipients decreased from 81% to 56.2%, or if the health utility of disease-free survivors decreased from 0.94 to 0.66, then CAR-T was no longer cost effective. Probabilistic sensitivity analysis found that CAR-T was cost effective in 94.8% of iterations at a willingness to pay of $100 000/QALY. CONCLUSION CAR-T therapy may represent a cost-effective option for pediatric relapsed/refractory B-ALL, although longer follow-up of CAR-T survivors is required to confirm validity of these findings.
Collapse
Affiliation(s)
- Reith R Sarkar
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA
| | - Nicholas J Gloude
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Rady Children’s Hospital, San Diego, CA
| | - Deborah Schiff
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Rady Children’s Hospital, San Diego, CA
| | - James D Murphy
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA
| |
Collapse
|
279
|
Metzinger MN, Verghese C, Hamouda DM, Lenhard A, Choucair K, Senzer N, Brunicardi FC, Dworkin L, Nemunaitis J. Chimeric Antigen Receptor T-Cell Therapy: Reach to Solid Tumor Experience. Oncology 2019; 97:59-74. [PMID: 31261152 DOI: 10.1159/000500488] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/12/2019] [Indexed: 11/19/2022]
Abstract
Chimeric antigen receptor (CAR) modified T-cell therapy, a unique platform technology highlighting precision medicine through utilization of molecular biology and cell-based therapeutics has shown unprecedented rates in patients with hematological malignancies such as acute lymphocyte leukemia, non-Hodgkin's lymphoma and multiple myeloma (MM). With the approval of CD19-targeted CAR T-cells by the Food and Drug Administration in acute lymphoblastic leukemia (ALL) and NHL, this technology is positioned for aggressive expansion to combination therapeutic opportunities and proof of principle towards utility in other malignant disorders. However, despite the impressive results seen with hematological malignancies, CAR T-cells have shown limited efficacy in solid tumors with several unsuccessful preclinical studies. Regardless, these attempts have provided us with a better understanding of the imminent challenges specific to solid tumors even if they have not so far led to expanded clinical treatment opportunities outside ALL/NHL/MM. This review summarizes our current understanding of CAR T-cell mechanism of action, while presenting the major limitations of CAR T-cell derived treatments in solid tumors. We further discuss recent findings and present new potential strategies to overcome the challenges facing solid tumor targeting by CAR T-cell platforms.
Collapse
Affiliation(s)
- Matthew N Metzinger
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Cherian Verghese
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Danae M Hamouda
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Amanda Lenhard
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Khalil Choucair
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Neil Senzer
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | | | - Lance Dworkin
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - John Nemunaitis
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA, .,ProMedica Health System, Toledo, Ohio, USA,
| |
Collapse
|
280
|
Klener P, Etrych T, Klener P. Biological Therapy of Hematologic Malignancies: Toward a Chemotherapy- free Era. Curr Med Chem 2019; 26:1002-1018. [PMID: 28990505 DOI: 10.2174/0929867324666171006144725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
Abstract
Less than 70 years ago, the vast majority of hematologic malignancies were untreatable diseases with fatal prognoses. The development of modern chemotherapy agents, which had begun after the Second World War, was markedly accelerated by the discovery of the structure of DNA and its role in cancer biology and tumor cell division. The path travelled from the first temporary remissions observed in children with acute lymphoblastic leukemia treated with single-agent antimetabolites until the first cures achieved by multi-agent chemotherapy regimens was incredibly short. Despite great successes, however, conventional genotoxic cytostatics suffered from an inherently narrow therapeutic index and extensive toxicity, which in many instances limited their clinical utilization. In the last decade of the 20th century, increasing knowledge on the biology of certain malignancies resulted in the conception and development of first molecularly targeted agents designed to inhibit specific druggable molecules involved in the survival of cancer cells. Advances in technology and genetic engineering enabled the production of structurally complex anticancer macromolecules called biologicals, including therapeutic monoclonal antibodies, antibody-drug conjugates and antibody fragments. The development of drug delivery systems (DDSs), in which conventional drugs were attached to various types of carriers including nanoparticles, liposomes or biodegradable polymers, represented an alternative approach to the development of new anticancer agents. Despite the fact that the antitumor activity of drugs attached to DDSs was not fundamentally different, the improved pharmacokinetic profiles, decreased toxic side effects and significantly increased therapeutic indexes resulted in their enhanced antitumor efficacy compared to conventional (unbound) drugs. Approval of the first immune checkpoint inhibitor for the treatment of cancer in 2011 initiated the era of cancer immunotherapy. Checkpoint inhibitors, bispecific T-cell engagers, adoptive T-cell approaches and cancer vaccines have joined the platform so far, represented mainly by recombinant cytokines, therapeutic monoclonal antibodies and immunomodulatory agents. In specific clinical indications, conventional drugs have already been supplanted by multi-agent, chemotherapy-free regimens comprising diverse immunotherapy and/or targeted agents. The very distinct mechanisms of the anticancer activity of new immunotherapy approaches not only call for novel response criteria, but might also change fundamental treatment paradigms of certain types of hematologic malignancies in the near future.
Collapse
Affiliation(s)
- Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| | - Tomas Etrych
- Department of biomedical polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, 162 06 Prague, Czech Republic
| | - Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic
| |
Collapse
|
281
|
Lee YG, Chu H, Lu Y, Leamon CP, Srinivasarao M, Putt KS, Low PS. Regulation of CAR T cell-mediated cytokine release syndrome-like toxicity using low molecular weight adapters. Nat Commun 2019; 10:2681. [PMID: 31213606 PMCID: PMC6581910 DOI: 10.1038/s41467-019-10565-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 05/14/2019] [Indexed: 12/31/2022] Open
Abstract
Although chimeric antigen receptor (CAR) T cell therapies have demonstrated considerable success in treating hematologic malignancies, they have simultaneously been plagued by a cytokine release syndrome (CRS) that can harm or even kill the cancer patient. We describe a CAR T cell strategy in which CAR T cell activation and cancer cell killing can be sensitively regulated by adjusting the dose of a low molecular weight adapter that must bridge between the CAR T cell and cancer cell to initiate tumor eradication. By controlling the concentration and dosing schedule of adapter administration, we document two methods that can rapidly terminate (<3 h) a pre-existing CRS-like toxicity and two unrelated methods that can pre-emptively prevent a CRS-like toxicity that would have otherwise occurred. Because all four methods concurrently enhance CAR T cell potency, we conclude that proper use of bispecific adapters could potentially avoid a life-threatening CRS while enhancing CAR T cell tumoricidal activity.
Collapse
Affiliation(s)
- Yong Gu Lee
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Haiyan Chu
- Endocyte Inc., 3000 Kent Ave, West Lafayette, IN, 47906, USA
| | - Yingjuan Lu
- Endocyte Inc., 3000 Kent Ave, West Lafayette, IN, 47906, USA
| | | | - Madduri Srinivasarao
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Karson S Putt
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
282
|
Pediatric Cardio-Oncology: Development of Cancer Treatment-Related Cardiotoxicity and the Therapeutic Approach to Affected Patients. Curr Treat Options Oncol 2019; 20:56. [PMID: 31129800 DOI: 10.1007/s11864-019-0658-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OPINION STATEMENT The past 5 decades have seen significant improvements in outcomes for pediatric patients with cancer. Unfortunately, children and adolescents who have been treated for cancer are five to six times more likely to develop cardiovascular disease as a result of their therapies. Cardiovascular disease may manifest in a plethora of ways, from asymptomatic ventricular dysfunction to end-stage heart failure, hypertension, arrhythmia, valvular disease, early coronary artery disease, or peripheral vascular disease. A number of treatment modalities are implicated in pediatric and adult populations, including anthracyclines, radiation therapy, alkylating agents, targeted cancer therapies (small molecules and antibody therapies), antimetabolites, antimicrotubule agents, immunotherapy, interleukins, and chimeric antigen receptor T cells. For some therapies, such as anthracyclines, the mechanism of injury is elucidated, but for many others it is not. While a few protective strategies exist, in many cases, observation and close monitoring is the only defense against developing end-stage cardiovascular disease. Because of the variety of potential outcomes after cancer therapy, a one-size-fits-all approach is not appropriate. Rather, a good working relationship between oncology and cardiology to assess the risks and benefits of various therapies and planning for appropriate surveillance is the best model. When disease is identified, any of a number of therapies may be appropriate; however, in the pediatric and adolescent population supportive data are limited.
Collapse
|
283
|
Stoiber S, Cadilha BL, Benmebarek MR, Lesch S, Endres S, Kobold S. Limitations in the Design of Chimeric Antigen Receptors for Cancer Therapy. Cells 2019; 8:cells8050472. [PMID: 31108883 PMCID: PMC6562702 DOI: 10.3390/cells8050472] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer therapy has entered a new era, transitioning from unspecific chemotherapeutic agents to increasingly specific immune-based therapeutic strategies. Among these, chimeric antigen receptor (CAR) T cells have shown unparalleled therapeutic potential in treating refractory hematological malignancies. In contrast, solid tumors pose a much greater challenge to CAR T cell therapy, which has yet to be overcome. As this novel therapeutic modality matures, increasing effort is being invested to determine the optimal structure and properties of CARs to facilitate the transition from empirical testing to the rational design of CAR T cells. In this review, we highlight how individual CAR domains contribute to the success and failure of this promising treatment modality and provide an insight into the most notable advances in the field of CAR T cell engineering.
Collapse
Affiliation(s)
- Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
- German Center for Translational Cancer Research (DKTK), 80337 Munich, Germany.
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
- German Center for Translational Cancer Research (DKTK), 80337 Munich, Germany.
| |
Collapse
|
284
|
Baeuerle PA, Ding J, Patel E, Thorausch N, Horton H, Gierut J, Scarfo I, Choudhary R, Kiner O, Krishnamurthy J, Le B, Morath A, Baldeviano GC, Quinn J, Tavares P, Wei Q, Weiler S, Maus MV, Getts D, Schamel WW, Hofmeister R. Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response. Nat Commun 2019; 10:2087. [PMID: 31064990 PMCID: PMC6504948 DOI: 10.1038/s41467-019-10097-0] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/08/2019] [Indexed: 12/30/2022] Open
Abstract
T cells expressing CD19-targeting chimeric antigen receptors (CARs) reveal high efficacy in the treatment of B cell malignancies. Here, we report that T cell receptor fusion constructs (TRuCs) comprising an antibody-based binding domain fused to T cell receptor (TCR) subunits can effectively reprogram an intact TCR complex to recognize tumor surface antigens. Unlike CARs, TRuCs become a functional component of the TCR complex. TRuC-T cells kill tumor cells as potently as second-generation CAR-T cells, but at significant lower cytokine release and despite the absence of an extra co-stimulatory domain. TRuC-T cells demonstrate potent anti-tumor activity in both liquid and solid tumor xenograft models. In several models, TRuC-T cells are more efficacious than respective CAR-T cells. TRuC-T cells are shown to engage the signaling capacity of the entire TCR complex in an HLA-independent manner.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Cell Line, Tumor
- Female
- Humans
- Immunotherapy, Adoptive/methods
- Mice
- Mice, Inbred NOD
- Neoplasms/immunology
- Neoplasms/therapy
- Primary Cell Culture
- Protein Domains
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Artificial/genetics
- Receptors, Artificial/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- T-Lymphocytes/immunology
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | - Jian Ding
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Ekta Patel
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Niko Thorausch
- Department of Immunology, Faculty of Biology, BIOSS Center for Biological Signalling Studies, CIBSS-Centre for Integrative Biological Signalling Studies and Centre for Chronic Immunodeficiency CCI, University of Freiburg, Schänzlestraβe 18, Freiburg, 79104, Germany
| | - Holly Horton
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Jessica Gierut
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Irene Scarfo
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Bldg. 149 13th Street, Charlestown, MA, USA
| | - Rashmi Choudhary
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Olga Kiner
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | | | - Bonnie Le
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Anna Morath
- Department of Immunology, Faculty of Biology, BIOSS Center for Biological Signalling Studies, CIBSS-Centre for Integrative Biological Signalling Studies and Centre for Chronic Immunodeficiency CCI, University of Freiburg, Schänzlestraβe 18, Freiburg, 79104, Germany
| | | | - Justin Quinn
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Patrick Tavares
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Qi Wei
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Solly Weiler
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Bldg. 149 13th Street, Charlestown, MA, USA
| | - Daniel Getts
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA
| | - Wolfgang W Schamel
- Department of Immunology, Faculty of Biology, BIOSS Center for Biological Signalling Studies, CIBSS-Centre for Integrative Biological Signalling Studies and Centre for Chronic Immunodeficiency CCI, University of Freiburg, Schänzlestraβe 18, Freiburg, 79104, Germany
| | - Robert Hofmeister
- TCR² Therapeutics, Inc., 100 Binney Street, Cambridge, MA, 02142, USA.
| |
Collapse
|
285
|
Jiang H, Liu L, Guo T, Wu Y, Ai L, Deng J, Dong J, Mei H, Hu Y. Improving the safety of CAR-T cell therapy by controlling CRS-related coagulopathy. Ann Hematol 2019; 98:1721-1732. [PMID: 31055613 DOI: 10.1007/s00277-019-03685-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 04/06/2019] [Indexed: 12/12/2022]
Abstract
The CD19-targeted chimeric antigen receptor T cell (CAR-T) therapy has been widely proved effective on relapsed and refractory (r/r) B cell acute lymphoblastic leukemia (B-ALL). Meanwhile, CAR-T therapy-related toxicities, including cytokine release syndrome (CRS) and neurological toxicities, are drawing researchers' attention. In addition, our research team notices that coagulopathy and even disseminated intravascular coagulation (DIC) are common problems during CAR-T therapy. In our phase 1/2 clinical trial (NCT02965092), 53 r/r B-ALL patients underwent leukapheresis on day - 11 and received lymphodepleting chemotherapy on day - 7 to day - 5. Finally, they received split infusions of anti-CD19 CAR-T cells on day 0 to day 2. Plasma concentrations of tissue factor (TF) and platelet endothelial cell adhesion molecular-1 (PECAM-1) were also measured to identify the mechanism of coagulation disorders. The overall 1-month remission rate of the 53 patients was 88.7%. During the treatment course, 19 patients experienced grade 3-4 CRS, 8 patients developed grade 2-3 neurological toxicities. Beyond that, 30 patients (30/53, 56.6%) suffered from coagulation disorders, and half of them should be diagnosed as DIC. Benefiting from replacement and anticoagulant therapy, 14 patients successfully got out of the conditions of DIC. Remarkably, the severity of coagulopathy was positively correlated with CRS grade. What is more, plasma TF and PECAM-1 levels indicated that vascular endothelial factors played key roles in the process of CRS-related coagulopathy. To conclude, coagulation disorders frequently happen during CAR-T therapy. TF and PECAM-1 are of great importance in the etiology and pathogenesis of coagulation problems. Early and proper interventions targeted at CRS-related coagulopathy contribute a lot to the control of side effects in CAR-T therapy.
Collapse
Affiliation(s)
- Huiwen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Lin Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yaohui Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Jian Dong
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China. .,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China. .,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
286
|
Hirayama AV, Turtle CJ. Toxicities of CD19 CAR-T cell immunotherapy. Am J Hematol 2019; 94:S42-S49. [PMID: 30784102 DOI: 10.1002/ajh.25445] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/30/2022]
Abstract
CD19-targeted chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy has demonstrated impressive results in B-cell malignancies, and CAR-T cell therapies targeting other antigens are in development for other cancers. Cytokine release syndrome (CRS) and neurotoxicity can be life-threatening in a subset of patients. The severity of CRS and neurotoxicity can be impacted by the disease burden, lymphodepletion regimen, and CAR-T cell dose. Tocilizumab and corticosteroids have been used to manage these toxicities, enabling CD19 CAR-T cells to be administered without obvious compromise in efficacy. Consensus criteria for grading and managing toxicities will facilitate the widespread application of this treatment modality.
Collapse
Affiliation(s)
- Alexandre V. Hirayama
- Clinical Research Division and Integrated Immunotherapy Research CenterFred Hutchinson Cancer Research Center Seattle Washington
| | - Cameron J. Turtle
- Clinical Research Division and Integrated Immunotherapy Research CenterFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| |
Collapse
|
287
|
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor -(CAR) T-cell therapy has become a commonly used immunotherapy originally used in the treatment of B-cell leukemias but which are now applied broadly across tumor classes. Although high rates of remission are associated with CAR T-cell therapy, toxicities associated with these novel treatment regimens can be lethal if not recognized in a timely manner. RECENT FINDINGS Cytokine release syndrome and neurotoxicity are the two most common toxicities associated with CAR T-cell therapy. Cytokine release syndrome is characterized by a flu-like illness accompanied by significant hemodynamic instability; treatments include administration of tocilizumab and corticosteroids. Neurotoxicity is associated with nonpattern-specific neurological changes and can rapidly progress to a comatose state from cerebral edema and death. Other potential toxicities from CAR T-cell therapy include tumor lysis syndrome, B-cell aplasia, graft versus host disease, and dermatological eruptions. SUMMARY Clinical awareness of CAR T-cell toxicities is important because prompt treatment leads to improved survival and remission rates.
Collapse
|
288
|
Castañeda S, Martínez-Quintanilla D, Martín-Varillas JL, García-Castañeda N, Atienza-Mateo B, González-Gay MA. Tocilizumab for the treatment of adult-onset Still’s disease. Expert Opin Biol Ther 2019; 19:273-286. [DOI: 10.1080/14712598.2019.1590334] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Santos Castañeda
- Rheumatology Division, Hospital de La Princesa, IIS-Princesa, Catedra de EPID-Futuro, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Dolores Martínez-Quintanilla
- Rheumatology Division, Hospital de La Princesa, IIS-Princesa, Catedra de EPID-Futuro, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José L. Martín-Varillas
- Division of Rheumatology and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Noelia García-Castañeda
- Rheumatology Division, Hospital de La Princesa, IIS-Princesa, Catedra de EPID-Futuro, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Belén Atienza-Mateo
- Division of Rheumatology and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Miguel A. González-Gay
- Division of Rheumatology and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
- Department of Medicine, University of Cantabria, Santander, Spain
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
289
|
Abstract
BACKGROUND Pemphigus diseases are a heterogeneous group of potentially life-threatening autoimmune bullous disorders. Therefore, rapidly acting and effective therapeutic approaches are essential. OBJECTIVES In this review, current therapeutic options in line with available guidelines are presented and new therapeutic approaches are discussed. METHODS A literature search was performed using PubMed. RESULTS Treatment of pemphigus is based on systemic glucocorticosteroids, frequently combined with potentially corticosteroid-sparing immunosuppressants such as azathioprine and mycophenolate mofetil/mycophenolic acid. Recently, the impressive efficacy of the anti-CD20 antibody rituximab has been shown in a prospective randomized trial. In severe or treatment-refractory cases, immunoadsorption or high-dose intravenous immunoglobulins (IVIG) are recommended. Adjuvant immunoadsorption also seems to be useful within the first 8-12 weeks of therapy in patients with very high autoantibody levels. A variety of new therapeutic approaches is currently evaluated in phase IIa studies. CONCLUSION Therapy of pemphigus has been greatly improved by the employment of rituximab. The use of glucocorticosteroids, associated with a high number of adverse events and elevated mortality, could be reduced by the additional use of rituximab. After approval of rituximab for the treatment of pemphigus by the US Food and Drug Administration in 2018, licensing in Europe is expected in 2019.
Collapse
Affiliation(s)
- N van Beek
- Klinik für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Deutschland.
| | - D Zillikens
- Klinik für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Deutschland
| | - E Schmidt
- Klinik für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Deutschland.,Lübecker Institut für Experimentelle Dermatologie (LIED), Universität zu Lübeck, Lübeck, Deutschland
| |
Collapse
|
290
|
Leahy AB, Elgarten CW, Grupp SA, Maude SL, Teachey DT. Tisagenlecleucel for the treatment of B-cell acute lymphoblastic leukemia. Expert Rev Anticancer Ther 2019; 18:959-971. [PMID: 30111196 DOI: 10.1080/14737140.2018.1512411] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Cure rates for pediatric and young adult patients with refractory or recurrently relapsed acute lymphoblastic leukemia (ALL) are dismal. Survival from time of relapse is typically measured in weeks to months, and standard chemotherapy and currently approved targeted therapy achieve remission in less than a third of affected patients. To date, the only definitive curative therapy has been allogeneic hematopoietic stem cell transplant (HSCT). Advances in immunotherapy, with the introduction of chimeric antigen receptor T-cell therapies and the development of tisagenlecleucel, have changed the landscape. Areas covered: This review will describe the pharmacology of tisagenlecleucel and summarize the clinical evidence for its use in the treatment of multiple-relapsed or refractory B-cell ALL (B-ALL). Also discussed are other immunotherapies for B-ALL as well as the most commonly-encountered toxicities and corresponding management strategies. Expert commentary: Early phase trials indicate that tisagenlecleucel significantly improves survival for patients with B-ALL that is refractory or in second or later relapse. In responding patients, remissions have been reported on the order of years, and thus, tisagenlecleucel may herald a dramatic shift in the treatment paradigm of this largely fatal disease.
Collapse
Affiliation(s)
- Allison Barz Leahy
- a Divisions of Pediatric Hematology and Oncology , Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine , Philadelphia , PA , USA
| | - Caitlin W Elgarten
- a Divisions of Pediatric Hematology and Oncology , Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine , Philadelphia , PA , USA
| | - Stephan A Grupp
- a Divisions of Pediatric Hematology and Oncology , Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine , Philadelphia , PA , USA
| | - Shannon L Maude
- a Divisions of Pediatric Hematology and Oncology , Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine , Philadelphia , PA , USA
| | - David T Teachey
- a Divisions of Pediatric Hematology and Oncology , Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine , Philadelphia , PA , USA
| |
Collapse
|
291
|
Acharya UH, Dhawale T, Yun S, Jacobson CA, Chavez JC, Ramos JD, Appelbaum J, Maloney DG. Management of cytokine release syndrome and neurotoxicity in chimeric antigen receptor (CAR) T cell therapy. Expert Rev Hematol 2019; 12:195-205. [PMID: 30793644 DOI: 10.1080/17474086.2019.1585238] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cell immunotherapy has demonstrated remarkable anti-tumor activity in B-cell malignancies and is under investigation in other hematologic malignancies and solid tumors. While highly efficacious, post-infusion T cell activity often results in massive cytokine release precipitating cytokine release syndrome (CRS), the signature toxicity of CAR T cells. This toxicity is characterized by systemic immune activation resulting in fever, hypotension, respiratory insufficiency and capillary leak. Either in conjunction with or in the absence of CRS, a subset of patients may also develop mild to severe neurotoxicity. Although the precise pathogenesis of CRS and neurotoxicity aren't fully elucidated, risk factors and mitigation strategies have been reported. Areas covered: This manuscript provides an in-depth overview of the pathogenesis, clinical characteristics, current toxicity management strategies, and future perspectives pertaining to CRS and neurotoxicity. Expert Opinion: As CAR T cell based therapies gain popularity in the management of various malignancies, the complimentary toxicities of CRS and neurotoxicity pose a clinical challenge in practice. Risk adaptive modeling incorporating disease profile, patient demographics, lymphodepletion, cell dosing, CAR T construct, and potentially cytokine gene polymorphisms may be instructive to assess individualized risk and optimal CRS/neurotoxicity management.
Collapse
Affiliation(s)
- Utkarsh H Acharya
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA.,c Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Tejaswini Dhawale
- d Division of Hematology, Department of Medicine , University of Washington , Seattle , WA , USA
| | - Seongseok Yun
- e Department of Malignant Hematology , H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Caron A Jacobson
- c Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Julio C Chavez
- e Department of Malignant Hematology , H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Jorge D Ramos
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| | - Jacob Appelbaum
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,d Division of Hematology, Department of Medicine , University of Washington , Seattle , WA , USA
| | - David G Maloney
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| |
Collapse
|
292
|
Gong F, Meng Q, Liu C, Zhao Y. Efficacy and association analysis of high-dose methotrexate in the treatment of children with acute lymphoblastic leukemia. Oncol Lett 2019; 17:4423-4428. [PMID: 30988812 PMCID: PMC6447950 DOI: 10.3892/ol.2019.10128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/15/2019] [Indexed: 01/24/2023] Open
Abstract
Effect of high-dose methotrexate (MTX) on children with acute lymphoblastic leukemia (ALL) with different subtypes and disease courses was investigated. A retrospective analysis of 207 children with ALL who were admitted to the People's Hospital of Pingyi County from March 2014 to June 2017 was carried out. According to the subtype of the disease, the children were divided into two groups. B-lineage group: ALL occurred in B-lineage lymphocytes (n=128); T-lineage group: ALL occurred in T-lineage lymphocytes (n=79). According to the disease course, the children were divided into three groups. High-risk group: disease course >15 days (n=67); moderate-risk group: disease course >8 and <15 days (n=58); low-risk group: disease course <8 days (n=82). The plasma concentration, calcium formyltetrahydrofolate (CF) rescue times and adverse reactions were compared at 12 h (T1), 48 h (T2), and 72 h (T3) after MTX infusion. The plasma concentration in B-lineage group was significantly higher than that in the T-lineage group at T2 and T3 (P<0.05). The incidence of adverse reactions in children with ALL in the B-lineage group was significantly higher than that in the T-lineage group (P<0.05). The CF rescue times in high-risk group were more than that in moderate- and low-risk groups (P<0.05). The incidence of adverse reactions in the high-risk group was significantly higher than that in the moderate- and low-risk groups (P<0.05), and in the moderate-risk group was significantly higher than that in the low-risk group (P<0.05). Compared with T-lineage ALL children, high-dose MTX causes more toxic injury to B-lineage ALL children. During clinical application of MTX in the treatment of ALL, close attention should be paid to the changes of the vital signs of patients, and timely CF rescue should be performed.
Collapse
Affiliation(s)
- Fangwei Gong
- Ward 2, Department of Pediatrics, The People's Hospital of Pingyi County, Linyi, Shandong 273300, P.R. China
| | - Qingjun Meng
- Ward 2, Department of Pediatrics, The People's Hospital of Pingyi County, Linyi, Shandong 273300, P.R. China
| | - Chengjuan Liu
- Ward 2, Department of Pediatrics, The People's Hospital of Pingyi County, Linyi, Shandong 273300, P.R. China
| | - Yeqi Zhao
- Ward 6, Department of Pediatrics, The People's Hospital of Pingyi County, Linyi, Shandong 273300, P.R. China
| |
Collapse
|
293
|
Abstract
Immunotherapy has become a powerful clinical strategy for treating cancer. The number of immunotherapy drug approvals has been increasing, with numerous treatments in clinical and preclinical development. However, a key challenge in the broad implementation of immunotherapies for cancer remains the controlled modulation of the immune system, as these therapeutics have serious adverse effects including autoimmunity and nonspecific inflammation. Understanding how to increase the response rates to various classes of immunotherapy is key to improving efficacy and controlling these adverse effects. Advanced biomaterials and drug delivery systems, such as nanoparticles and the use of T cells to deliver therapies, could effectively harness immunotherapies and improve their potency while reducing toxic side effects. Here, we discuss these research advances, as well as the opportunities and challenges for integrating delivery technologies into cancer immunotherapy, and we critically analyse the outlook for these emerging areas.
Collapse
Affiliation(s)
- Rachel S Riley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
294
|
Sachdeva M, Duchateau P, Depil S, Poirot L, Valton J. Granulocyte-macrophage colony-stimulating factor inactivation in CAR T-cells prevents monocyte-dependent release of key cytokine release syndrome mediators. J Biol Chem 2019; 294:5430-5437. [PMID: 30804212 DOI: 10.1074/jbc.ac119.007558] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/19/2019] [Indexed: 01/22/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapy has been shown to be clinically effective for managing a variety of hematological cancers. However, CAR T-cell therapy is associated with multiple adverse effects, including neurotoxicity and cytokine release syndrome (CRS). CRS arises from massive cytokine secretion and can be life-threatening, but it is typically managed with an anti-IL-6Ra mAb or glucocorticoid administration. However, these treatments add to a patient's medication burden and address only the CRS symptoms. Therefore, alternative strategies that can prevent CRS and neurotoxicity associated with CAR T-cell treatment are urgently needed. Here, we explored a therapeutic route aimed at preventing CRS rather than limiting its consequences. Using a cytokine-profiling assay, we show that granulocyte-macrophage colony-stimulating factor (GMCSF) is a key CRS-promoting protein. Through a combination of in vitro experiments and gene-editing technology, we further demonstrate that antibody-mediated neutralization or TALEN-mediated genetic inactivation of GMCSF in CAR T-cells drastically decreases available GMCSF and abolishes macrophage-dependent secretion of CRS biomarkers, including monocyte chemoattractant protein 1 (MCP-1), interleukin (IL) 6, and IL-8. Of note, we also found that the genetic inactivation of GMCSF does not impair the antitumor function or proliferative capacity of CAR T-cells in vitro We conclude that it is possible to prevent CRS by using "all-in-one" GMCSF-knockout CAR T-cells. This approach may eliminate the need for anti-CRS treatment and may improve the overall safety of CAR T-cell therapies for cancer patients.
Collapse
Affiliation(s)
- Mohit Sachdeva
- From Cellectis, Inc., 430 East 29th St., New York, New York 10016 and
| | | | | | | | - Julien Valton
- From Cellectis, Inc., 430 East 29th St., New York, New York 10016 and
| |
Collapse
|
295
|
Abstract
AbstractChimeric antigen receptor T (CAR-T) cell therapy is a new pillar in cancer therapeutics; however, its application is limited by the associated toxicities. These include cytokine release syndrome (CRS) and neurotoxicity. Although the IL-6R antagonist tocilizumab is approved for treatment of CRS, there is no approved treatment of neurotoxicity associated with CD19-targeted CAR-T (CART19) cell therapy. Recent data suggest that monocytes and macrophages contribute to the development of CRS and neurotoxicity after CAR-T cell therapy. Therefore, we investigated neutralizing granulocyte-macrophage colony-stimulating factor (GM-CSF) as a potential strategy to manage CART19 cell–associated toxicities. In this study, we show that GM-CSF neutralization with lenzilumab does not inhibit CART19 cell function in vitro or in vivo. Moreover, CART19 cell proliferation was enhanced and durable control of leukemic disease was maintained better in patient-derived xenografts after GM-CSF neutralization with lenzilumab. In a patient acute lymphoblastic leukemia xenograft model of CRS and neuroinflammation (NI), GM-CSF neutralization resulted in a reduction of myeloid and T cell infiltration in the central nervous system and a significant reduction in NI and prevention of CRS. Finally, we generated GM-CSF–deficient CART19 cells through CRISPR/Cas9 disruption of GM-CSF during CAR-T cell manufacturing. These GM-CSFk/o CAR-T cells maintained normal functions and had enhanced antitumor activity in vivo, as well as improved overall survival, compared with CART19 cells. Together, these studies illuminate a novel approach to abrogate NI and CRS through GM-CSF neutralization, which may potentially enhance CAR-T cell function. Phase 2 studies with lenzilumab in combination with CART19 cell therapy are planned.
Collapse
|
296
|
Grigor EJM, Fergusson D, Kekre N, Montroy J, Atkins H, Seftel MD, Daugaard M, Presseau J, Thavorn K, Hutton B, Holt RA, Lalu MM. Risks and Benefits of Chimeric Antigen Receptor T-Cell (CAR-T) Therapy in Cancer: A Systematic Review and Meta-Analysis. Transfus Med Rev 2019; 33:98-110. [PMID: 30948292 DOI: 10.1016/j.tmrv.2019.01.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/21/2019] [Accepted: 01/26/2019] [Indexed: 12/27/2022]
Abstract
Promising efficacy results of chimeric antigen receptor (CAR) T-cell therapy have been tempered by safety considerations. Our objective was to comprehensively summarize the efficacy and safety of CAR-T cell therapy in patients with relapsed or refractory hematologic or solid malignancies. MEDLINE, Embase, and the Cochrane Register of Controlled Trials (inception - November 21, 2017). Interventional studies investigating CAR-T cell therapy in patients with malignancies were included. Our primary outcome of interest was complete response (defined as the absence of detectable cancer). Two independent reviewers extracted relevant data, assessed risk of bias, and graded the quality of evidence using established methods. A total of 42 hematological malignancy studies and 18 solid tumor studies met were included (913 participants). Of 486 evaluable hematologic patients, 54.4% [95% CI, 42.5%-65.9%] experienced complete response in 27 CD19 CAR-T cell therapy studies. Of 65 evaluable hematologic patients, 24.4% [95% CI, 9.4%-50.3%] experienced complete response in seven non-CD19 CAR-T cell therapy studies. Cytokine release syndrome was experienced by 55.3% [95% CI, 40.3%-69.4%] of patients and neurotoxicity 37.2% [95% CI, 28.6%-46.8%] of patients with hematologic malignancies. Of 86 evaluable solid tumor patients, 4.1% [95% CI, 1.6%-10.6%] experienced complete response in eight CAR-T cell therapy studies. Limitations include heterogeneity of study populations, as well as high risk of bias of included studies. There was a strong signal for efficacy of CAR-T cell therapy in patients with CD19+ hematologic malignancies and no overall signal in solid tumor trials published to date. These results will help inform patients, physicians, and other stakeholders of the benefits and risks associated with CAR-T cell therapy.
Collapse
Affiliation(s)
- Emma J M Grigor
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada; Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada; Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Blood and Marrow Transplant Program, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Joshua Montroy
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Harold Atkins
- Blood and Marrow Transplant Program, The Ottawa Hospital, Ottawa, Ontario, Canada; Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Matthew D Seftel
- Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba
| | - Mads Daugaard
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Justin Presseau
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Kednapa Thavorn
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada; Institute for Clinical Evaluative Sciences, Ottawa, Ontario, Canada
| | - Brian Hutton
- School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada; Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Robert A Holt
- Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Manoj M Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada; Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
297
|
Del Bufalo F, Merli P, Alessi I, Locatelli F. B-cell depleting immunotherapies: therapeutic opportunities and toxicities. Expert Rev Clin Immunol 2019; 15:497-509. [PMID: 30681371 DOI: 10.1080/1744666x.2019.1573672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The last few years have witnessed what can certainly be defined as a 'period of renaissance' for immunotherapy in the field of hematological malignancies. In particular, antibody-mediated and cell-mediated immunotherapy have significantly changed the treatment approach of patients with B-cell lymphoproliferative disorders. These therapies, initially employed in patients with refractory/relapsed disease, are now integrated in the treatment of newly diagnosed patients. Together with the therapeutic success, we have also learnt that these innovative therapies can induce relevant, sometimes life-threatening or even fatal, side effects. Areas covered: In this review article, we analyzed the applicative therapeutic scenario and the peculiar toxicities associated with approaches of immunotherapy, paying particular attention to the new emerging side effects, substantially unknown before the introduction of these therapies. Expert commentary: Both monoclonal antibodies and cell therapy with lymphocytes genetically modified to be redirected against leukemia targets through the transduction with chimeric antigen receptors (CARs) have obtained unprecedented success in rescuing patients with resistant B-cell malignancies. Complications, such as neurotoxicity, cytokine release syndrome or persistent B-cell lymphopenia, must always be taken into consideration and diagnosed in a timely manner in patients with B-cell neoplasms to guarantee optimal management, thus avoiding they blunting the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Francesca Del Bufalo
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Pietro Merli
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Iside Alessi
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Franco Locatelli
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy.,b Department of Pediatrics , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
298
|
Jacobson CA. CD19 Chimeric Antigen Receptor Therapy for Refractory Aggressive B-Cell Lymphoma. J Clin Oncol 2019; 37:328-335. [DOI: 10.1200/jco.18.01457] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Anti-CD19–directed chimeric antigen receptor (CAR) T-cell therapy has had a resounding effect on the treatment of chemotherapy-insensitive aggressive B-cell non-Hodgkin lymphoma (B-NHL). There are now two US Food and Drug Administration (FDA)–approved products available for treating these patients, and a third product is expected to be approved in the coming months. The question remains: Is there a preferred or best product for my patient? However, answering that question is more complicated than simply balancing the reported efficacy and toxicity results. DESIGN This review outlines potential confounding factors involving the three products and their pivotal clinical trials and highlights additional considerations of manufacturing reliability and overall cost that must be considered when weighing one product against another. It will also review the directions in which the field is moving and strategies being examined to improve efficacy as well as toxicity. CONCLUSION Because a randomized three-arm clinical trial is unlikely, a product may have to be chosen on the basis of results from treatment centers that have experience with all three products. But by the time those results are available, they are likely to be outdated because, given the rapid evolution of the field, the next product will probably have been identified.
Collapse
|
299
|
Yakoub-Agha I, Moreau AS, Ahmad I, Borel C, Hadhoum N, Masouridi-Levrat S, Naudin J, Nicolas-Virelizier E, Ouachée-Chardin M, Platon L, Quessar A, Roth-Guepin G, Beauvais D, Baruchel A, Cornillon J. [Management of cytokine release syndrome in adult and pediatric patients undergoing CAR-T cell therapy for hematological malignancies: Recommendation of the French Society of Bone Marrow and cellular Therapy (SFGM-TC)]. Bull Cancer 2019; 106:S102-S109. [PMID: 30661749 DOI: 10.1016/j.bulcan.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/02/2018] [Indexed: 12/12/2022]
Abstract
The cytokine release syndrome (CRS) is the most common complication after adoptive immunotherapies such as chimeric antigen receptor T cells (CAR-T). The incidence varies from 30 to 100% depending on the CAR-T construct, cell doses and the underlying disease. Severe cases may involve 10 to 30% of patients. The triggering event is the activation of the CAR-T, after meeting with their target. The T cell activation leads to the release of effector cytokines, such as IFNγ, TNFα and IL2, that are responsible for the activating of monocyte/macrophage system, resulting in the production of pro-inflammatory cytokines, (including IL6, IFN-γ, IL10, MCP1) and associated with a significant elevation of CRP and ferritin. The CRS usually appears between 1 and 14days after the infusion of the cells and can last from 1 to 10days. Rare fatal cases have been reported in the literature. The first symptom is often a fever, sometimes very high, which must alert and reinforce the surveillance. In moderate forms, one can find fatigue, headache, rash, arthralgia and myalgia. T cell-related encephalopathy (CRES) syndrome may occur concomitantly. In case of aggravation, a vasoplegic shock associating capillary leakage and respiratory distress can occur. Close clinical monitoring is essential right from the injection to quickly detect the first symptoms. The treatment of severe forms, in addition to symptomatic management involves monoclonal antibodies targeting the IL6 or IL6 receptor, and sometimes steroids. Close cooperation with intensive care units is essential since 20 to 50% of patients require intensive care unit transfer.
Collapse
Affiliation(s)
| | - Anne-Sophie Moreau
- CHU de Lille, hôpital Salengro, centre de réanimation, 1, rue Émile-Laine, 59000 Lille, France
| | - Imran Ahmad
- Université de Montréal, hôpital Maisonneuve-Rosemont, service d'hématologie et d'oncologie médicale, 5415, boulevard de l'Assomption, Montréal, Québec H1T 2M4, Canada
| | - Cécile Borel
- Institut universitaire du cancer de Toulouse, service d'hématologie, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France
| | - Nawal Hadhoum
- CHU de Lille, hôpital Roger-Salengro, neurologie D/pathologies neuro-inflammatoires, 59037 Lille cedex, France
| | - Stavroula Masouridi-Levrat
- Hôpitaux universitaires de Genève, département d'oncologie, service d'hématologie, 4, rue Gabrielle-Perret-Gentil, 1205 Genève, Suisse
| | - Jérôme Naudin
- AP-HP, hôpital Robert-Debré, service de réanimation et surveillance continue pédiatrique, 48, boulevard Serrurier, 75019 Paris, France
| | | | - Marie Ouachée-Chardin
- IHOPe, service d'immuno-hématologie pédiatrie, 1, place Joseph-Renault, 69008 Lyon, France
| | - Lara Platon
- CHU Lapeyronie, service de réanimation médicale et médecine intensive, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France
| | - Asmaa Quessar
- CHU Ibn Rochd, hôpital 20-Août, service d'hématologie et d'oncologie pédiatrique, rue Lahcen Al Arjoun, Casablanca 20000, Maroc
| | | | - Davis Beauvais
- CHU de Lille, maladies du sang, 2, avenue Oscar-Lambret, 59037 Lille, France
| | - André Baruchel
- AP-HP, hôpital universitaire Robert-Debré, hématologie-immunologie pédiatrique, 48, boulevard Sérurier, 75935 Paris cedex 19, France
| | - Jérôme Cornillon
- Institut de cancérologie Lucien-Neuwirth, département d'hématologie clinique, 108 Bis, avenue Albert-Raimond, 42271 Saint-Priest-en-Jarez, France.
| |
Collapse
|
300
|
Mahadeo KM, Khazal SJ, Abdel-Azim H, Fitzgerald JC, Taraseviciute A, Bollard CM, Tewari P, Duncan C, Traube C, McCall D, Steiner ME, Cheifetz IM, Lehmann LE, Mejia R, Slopis JM, Bajwa R, Kebriaei P, Martin PL, Moffet J, McArthur J, Petropoulos D, O'Hanlon Curry J, Featherston S, Foglesong J, Shoberu B, Gulbis A, Mireles ME, Hafemeister L, Nguyen C, Kapoor N, Rezvani K, Neelapu SS, Shpall EJ. Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy. Nat Rev Clin Oncol 2019; 16:45-63. [PMID: 30082906 PMCID: PMC7096894 DOI: 10.1038/s41571-018-0075-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In 2017, an autologous chimeric antigen receptor (CAR) T cell therapy indicated for children and young adults with relapsed and/or refractory CD19+ acute lymphoblastic leukaemia became the first gene therapy to be approved in the USA. This innovative form of cellular immunotherapy has been associated with remarkable response rates but is also associated with unique and often severe toxicities, which can lead to rapid cardiorespiratory and/or neurological deterioration. Multidisciplinary medical vigilance and the requisite health-care infrastructure are imperative to ensuring optimal patient outcomes, especially as these therapies transition from research protocols to standard care. Herein, authors representing the Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network Hematopoietic Stem Cell Transplantation (HSCT) Subgroup and the MD Anderson Cancer Center CAR T Cell Therapy-Associated Toxicity (CARTOX) Program have collaborated to provide comprehensive consensus guidelines on the care of children receiving CAR T cell therapy.
Collapse
Affiliation(s)
- Kris M Mahadeo
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Sajad J Khazal
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hisham Abdel-Azim
- Department of Pediatrics, Blood and Marrow Transplantation Program, Keck School of Medicine, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Division of Critical Care, University of Pennsylvania Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Agne Taraseviciute
- Department of Pediatrics, Division of Hematology-Oncology, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research and Department of Pediatrics, Children's National and The George Washington University, Washington DC, USA
| | - Priti Tewari
- Department of Pediatrics, Stem Cell Transplantation, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Christine Duncan
- Pediatric Hematology-Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA, USA
| | - Chani Traube
- Department of Pediatric Critical Care, Weil Cornell Medical College, New York Presbyterian Hospital, New York, NY, USA
| | - David McCall
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marie E Steiner
- Department of Pediatrics, Division of Critical Care, University of Minnesota, Masonic Children's Hospital, University of Minnesota, Minneapolis, MN, USA
| | - Ira M Cheifetz
- Department of Pediatrics, Division of Critical Care, Duke Children's Hospital, Duke University, Durham, NC, USA
| | - Leslie E Lehmann
- Pediatric Hematology-Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA, USA
| | - Rodrigo Mejia
- Department of Pediatrics, Critical Care, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Slopis
- Department of Pediatrics, Neurology, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajinder Bajwa
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Nationwide Children's Hospital, the Ohio State University, Columbus, OH, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul L Martin
- Department of Pediatrics, Division of Blood and Marrow Transplant, Duke Children's Hospital, Duke University, Durham, NC, USA
| | - Jerelyn Moffet
- Department of Pediatrics, Division of Blood and Marrow Transplant, Duke Children's Hospital, Duke University, Durham, NC, USA
| | - Jennifer McArthur
- Department of Pediatrics, Division of Critical Care, St. Jude's Children's Research Hospital, Memphis, TN, USA
| | - Demetrios Petropoulos
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joan O'Hanlon Curry
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah Featherston
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jessica Foglesong
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Basirat Shoberu
- Department of Pharmacy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alison Gulbis
- Department of Pharmacy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria E Mireles
- Department of Pharmacy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisa Hafemeister
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cathy Nguyen
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neena Kapoor
- Department of Pediatrics, Blood and Marrow Transplantation Program, Keck School of Medicine, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|