152 results on '"Immunotherapy, Adoptive"'
Search Results
2. CAR-T-Zell-Therapie beim multiplen Myelom.
- Author
-
Zhou, X., Einsele, H., and Danhof, S.
- Abstract
Copyright of Der Internist is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2021
- Full Text
- View/download PDF
3. Multiples Myelom – bald heilbar?
- Author
-
Goldschmidt, Hartmut
- Abstract
Copyright of Der Internist is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2021
- Full Text
- View/download PDF
4. CAR-T-Zellen als Arzneimittel für neuartige Therapien (Advanced Therapy Medicinal Products).
- Author
-
Köhl, Ulrike and Abken, Hinrich
- Abstract
Copyright of Der Internist is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2021
- Full Text
- View/download PDF
5. T-Zellen mit chimärem Antigenrezeptor (CAR) bei akuter B‑Zell-Leukämie.
- Author
-
Hudecek, M.
- Abstract
Copyright of Best Practice Onkologie is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2019
- Full Text
- View/download PDF
6. [CAR-T cell therapy - personalized cellular immunotherapy in 2022]
- Author
-
Janina, Niebling, Wolfgang, Bethge, and Claudia, Lengerke
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Humans ,Immunologic Factors ,Immunotherapy ,Immunotherapy, Adoptive - Abstract
Chimeric antigen receptor T cells (CAR T cells) are genetically modified autologous or allogeneic T lymphocytes that target specific antigens and can be used to combat tumor cells. The approval studies show a high response and remission rate in heavily pretreated patients with aggressive B-cell lymphoma, acute lymphoblastic leukemia or multiple myeloma. The article presents the indications for approval, the clinical implementation and the management of side effects of CAR cell therapy.
- Published
- 2022
7. [Imaging findings of CAR-T cell therapy associated complications]
- Author
-
Karolin, Baumgartner, Wolfgang, Bethge, Christian Philipp, Reinert, Benjamin, Bender, Konstantin, Nikolaou, and Marius, Horger
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Immunotherapy, Adoptive - Published
- 2022
8. Adjuvante Immuntherapie mit dendritischen Zellen beim Rektumkarzinom : eine retrospektive Analyse
- Author
-
Shtein, Viktoriya, Poch, Bertram, and Kleger, Alexander
- Subjects
Dendritische Zelle ,Immuntherapie ,Rectal neoplasms ,ddc:610 ,DDC 610 / Medicine & health ,Dendritic cells ,Immunotherapy, Adoptive ,Mastdarmkrebs - Abstract
Trotz neuer, verbesserter Möglichkeiten in der Diagnostik und etablierter Vorsorgeprogramme wird auch heutzutage das Rektumkarzinom häufig erst im fortgeschrittenen Stadium erkannt. In der vorliegenden retrospektiven Analyse wurde untersucht, inwieweit eine adjuvante Immuntherapie mit autologen dendritischen Zellen die Rezidiv-/Metastasierungsraten und die krebsbedingte 5-Jahres-Überlebenszeit beeinflusst. Zwischen März 2002 und Mai 2011 wurden 95 Patienten mit Rektumkarzinom in der adjuvanten Situation im GPS-Zentrum für Chirurgie behandelt. Neben der S3-Leitlinien-konformen adjuvanten Standardtherapie wurde allen Patienten eine additive Immuntherapie mit DC (dendritischen Zellen) im Rahmen eines individuellen Heilversuches angeboten. Alle Patienten wurden über die Chancen und möglichen Risiken einer additiven Immuntherapie mit dendritischen Zellen aufgeklärt. 47 Patienten mit Rektumkarzinom entschieden sich für eine additive Immuntherapie mit dendritischen Zellen, 48 Patienten dagegen. Die Beobachtungszeit bei allen Patienten lag über 5 Jahre. 5/47 Patienten, die eine adjuvante DC-Therapie erhalten hatten, verstarben innerhalb des Beobachtungszeitraumes an nicht-rektumkarzinom-bedingten Ursachen, sodass die Daten von 42 Patienten ausgewertet werden konnten. Bei 6/42 Patienten trat ein Rezidiv der Erkrankung auf (14,3 %), wohingegen 36/42 (85,7 %) der Patienten rezidiv-/ metastasenfrei blieben. Bei den Patienten im UICC (Union for International Cancer Control) Stadium III (Lymphknoten bereits befallen) zeigte sich dieser Trend ebenfalls. Lediglich 4/17 Patienten (23,5 %) entwickelten ein Lokalrezidiv/Metastasen im Nachbeobachtungszeitraum von 5 Jahren, wohingegen bei den ausschließlich mit Standardtherapie behandelten Patienten bereits 14/19 Patienten (73,7 %) ein Lokalrezidiv/Metastasen auftraten, was dem Literaturvergleich entspricht. Bei den 5-Jahres-Überlebenszeiten ergab sich bei allen additiv mit dendritischen Zellen behandelten Patienten eine krankheitsbedingte 5-Jahres-Überlebensrate von 92,9 % (39/42), bei den UICC III Patienten 82,4 % (14/17). Bei den ausschließlich standardtherapierten Patienten zeigte sich eine krankheitsbedingte 5-Jahres-Überlebensrate von 73,9 % (34/46) und bei den Patienten im UICC Stadium III eine 5-Jahres-Überlebensrate von 47,4 % (9/19). Diese Daten – erhoben im Rahmen einer retrospektiven Analyse von Patienten, die im Rahmen eines individuellen Heilversuches behandelt wurden – deuten auf einen positiven Effekt einer additiven Immuntherapie mit dendritischen Zellen hin. Um diesen positiven Eindruck zu untermauern sind jedoch weitere Untersuchungen im Rahmen der prospektiven und randomisierten multizentrischen Studien mit einer großen Anzahl an Patienten notwendig.
- Published
- 2021
9. [Practical aspects of the application of CAR T cells and management of their toxicities]
- Author
-
Veit, Bücklein, Viktoria, Blumenberg, and Marion, Subklewe
- Subjects
Cytotoxicity, Immunologic ,Biological Products ,Receptors, Chimeric Antigen ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Antineoplastic Agents, Immunological ,Treatment Outcome ,Neoplasms ,Cytokines ,Humans ,Neurotoxicity Syndromes ,Leukapheresis ,Cytokine Release Syndrome - Abstract
CD19 CAR T cells induce - in part long-lasting - remissions in heavily pretreated patients with relapsed/refractory B-cell malignancies. However, they are associated with unique toxicities, and patient management therefore requires specific expertise.In this review, we outline the basics of their mode of action and present the currently available data on their efficacy in various B-cell and plasma cell malignancies. Currently approved therapies (Tisagenlecleucel, Axicabtagene ciloleucel, Brexucabtagene autoleucel) for patients are outlined as well as indications where approvals are expected in the near future. We discuss practical aspects of CAR T cell therapy from the patient's initial presentation, over leukapheresis, to CAR T cell transfusion. Additionally, we highlight the pathophysiology and principles of the management of the most common toxicities (cytokine release syndrome [CRS], immune cell associated neurotoxicity syndrome [ICANS] and cytopenias).
- Published
- 2021
10. [CAR T-cell therapy for malignant B-cell lymphoma : A new treatment paradigm]
- Author
-
H, Balke-Want and P, Borchmann
- Subjects
Lymphoma, B-Cell ,T-Lymphocytes ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Child ,Immunotherapy, Adoptive ,Leukemia, Lymphocytic, Chronic, B-Cell - Abstract
Following the first demonstration of efficacy of anti-CD19-directed chimeric antigen receptor (CAR) T cells in a patient with relapsed chronic lymphocytic leukemia (CLL) in 2011, pivotal studies for this innovative therapy were initially conducted in multiple relapsed or refractory (r/r) childhood and young adult acute B‑cell leukemia and in aggressive adult B‑cell lymphoma. The studies demonstrated efficacy even in chemotherapy-refractory disease, resulting in the first approval of autologous and genetically engineered T cells for the treatment of r/r B‑cell acute lymphoblastic leukemia (B-ALL) in the US for the product tisagenlecleucel (Kymriah®, Novartis) back in 2018. Approval for the treatment of r/r aggressive B‑cell lymphoma followed shortly thereafter for tisagenlecleucel and axicabtagene ciloleucel (Yescarta, Kite/Gilead). This review focuses on the treatment of aggressive B‑cell lymphoma and other CD19 positive B‑cell lymphomas by summarizing the study results of clinically tested CAR T cells, discussing possible resistance mechanisms, and providing an outlook on ongoing studies with new target antigens for the treatment of B‑cell lymphomas.Nachdem 2011 erstmalig die Wirksamkeit von gegen CD19 gerichteten CAR-T-Zellen (CAR chimärer Antigenrezeptor) bei einem Patienten mit rezidivierter chronischer lymphatischer Leukämie (CLL) gezeigt werden konnte, wurden die Zulassungsstudien für diese innovative Therapie zunächst bei der mehrfach rezidivierten oder refraktären (r/r) akuten B‑Zell-Leukämie im Kindes- und jungen Erwachsenenalter und beim aggressiven B‑Zell-Lymphom erwachsener Patienten durchgeführt. Die Studien zeigten Wirksamkeit sogar bei chemotherapierefraktären Krankheitsverläufen, sodass mit dem Produkt Tisagenlecleucel (Kymriah®, Novartis) die ersten autologen und gentechnisch modifizierten T‑Zellen bereits 2018 für die Behandlung der r/r akuten lymphatischen B‑Zell-Leukämie (B-ALL) in den USA zugelassen wurden. Die Zulassung zur Behandlung r/r aggressiver B‑Zell-Lymphome erfolgte kurz darauf für Tisagenlecleucel und Axicabtagen Ciloleucel (Yescarta, Kite/Gilead). Die vorliegende Übersicht konzentriert sich auf die Behandlung des aggressiven B‑Zell-Lymphoms und anderer CD19-positiver B‑Zell-Lymphome. Dafür werden die Studienergebnisse klinisch geprüfter CAR-T-Zellen zusammengefasst, mögliche Resistenzmechanismen erörtert und ein Ausblick auf laufende Studien mit neuen Zielantigenen für die Behandlung von B‑Zell-Lymphomen gegeben.
- Published
- 2021
11. [Toxicity after chimeric antigen receptor T-cell therapy : Overview and management of early and late onset side effects]
- Author
-
Jorge, Garcia Borrega, Katrin, Heindel, Yasemin, Göreci, Clemens, Warnke, Oezguer A, Onur, Matthias, Kochanek, Natalie, Schub, Francis, Ayuk, Dominic, Wichmann, and Boris, Böll
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Humans ,Cytokine Release Syndrome ,Immunotherapy, Adoptive ,United States - Abstract
The transfusion of chimeric antigen receptor (CAR) T‑cells has become established as a new treatment option in oncology; however, this is regularly associated with immune-mediated side effects, which can also run a severe course and necessitate a specific treatment and intensive medical treatment.A literature review was carried out on CAR T-cell therapy, toxicities and the management of side effects.The cytokine release syndrome (CRS) and the immune effector cell-associated neurotoxicity syndrome (ICANS) regularly occur shortly after CAR T-cell treatment. The symptoms of CRS can range from mild flu-like symptoms to multiorgan failure. In addition to mild symptoms, such as disorientation and aphasia, ICANS can also lead to convulsive seizures and brain edema. The management of CRS and ICANS is based on the severity according to the grading of the American Society for Transplantation and Cellular Therapy (ASTCT). Tocilizumab and corticosteroids are recommended for CRS and corticosteroids are used for ICANS. In the further course persisting hypogammaglobulinemia and cytopenia are frequent even months after the initial treatment and promote infections even months after CAR T‑cell therapy.Potentially severe complications regularly occur after CAR T-cell therapy. An interdisciplinary cooperation between intensive care physicians, hematologists, neurologists and specialists in other disciplines is of decisive importance for the optimal care of patients after CAR T‑cell therapy.HINTERGRUND: Die Transfusion von Chimären-Antigenrezeptor(CAR)-T-Zellen hat sich als neue Therapieform in der Onkologie etabliert, geht jedoch regelhaft mit immunvermittelten Nebenwirkungen einher, die auch schwer verlaufen können sowie eine spezifische und intensivmedizinische Behandlung erfordern.Literaturrecherche zu CAR-T-Zell-Therapie, Toxizitäten und Nebenwirkungsmanagement.Das „cytokine release syndrome“ (CRS) und das „immune effector cell-associated neurotoxicity syndrome“ (ICANS) treten regelhaft kurz nach einer Therapie mit CAR-T-Zellen auf. Das CRS kann von einer milden grippeähnlichen Symptomatik bis zum Multiorganversagen führen. Beim ICANS kann es neben milden Symptomen wie Verwirrtheit und Aphasie auch zu Krampfanfällen und Hirnödem kommen. Das Management von CRS und ICANS orientiert sich am Schweregrad nach dem Grading der American Society for Transplantation and Cellular Therapy (ASTCT). Beim CRS werden Tocilizumab und Kortikosteroide empfohlen; beim ICANS werden Kortikosteroide eingesetzt. Im weiteren Verlauf sind eine persistierende Hypogammaglobulinämie und Zytopenien auch Monate nach der Therapie häufig; diese begünstigen Infektionen auch Monate nach CAR-T-Zell-Therapie.Nach CAR-T-Zell-Therapie kommt es regelhaft zu potenziell schweren Komplikationen. Eine interdisziplinäre Zusammenarbeit zwischen Intensivmediziner*innen, Hämatolog*innen, Neurolog*innen und Ärzt*innen anderer Fachabteilungen ist für die optimale Versorgung von CAR-T-Zell-Patient*innen von entscheidender Bedeutung.
- Published
- 2021
12. [CAR T-cell therapy for multiple myeloma]
- Author
-
X, Zhou, H, Einsele, and S, Danhof
- Subjects
Receptors, Chimeric Antigen ,Chimäre Antigenrezeptoren ,T‑lymphocytes ,Receptors, chimeric antigen ,Unerwünschte Wirkungen der CAR-T-Zell-Therapie ,Immunotherapy, adoptive ,Immunotherapy, Adoptive ,„B-cell maturation antigen“ ,CAR T‑cell therapy/adverse events ,Schwerpunkt: Zelluläre Immuntherapien in der Onkologie: CAR-T-Zellen ,Humans ,T‑Lymphozyten ,B-Cell Maturation Antigen ,Neoplasm Recurrence, Local ,Adoptive Immuntherapie ,Multiple Myeloma ,B‑cell maturation antigen - Abstract
Chimeric antigen receptor (CAR) T-cell treatment is a novel immunotherapy utilizing the patient's own immune system as a "weapon against tumor cells". In patients with multiple myeloma (MM), CAR T-cell therapy has been investigated in clinical trials. The current data on B‑cell maturation antigen (BCMA)-targeted CAR T-cells have shown impressive efficacy, and official approval is expected shortly. However, the majority of patients relapse after CAR T-cell therapy. Moreover, the treatment can cause severe adverse events such as cytokine release syndrome and neurotoxicity with lethal outcome. The cost-benefit ratio of this treatment also needs to be optimized. Despite these limitations, CAR T-cell therapy represents an attractive option for patients with MM and has the potential to be incorporated into the standard of care.Die Behandlung mit CAR-T-Zellen (CAR chimärer Antigenrezeptor) ist eine neuartige Strategie der zellulären Immuntherapie, die das patienteneigene Immunsystem als „Waffe gegen Tumorzellen“ benutzt. Bei Patienten mit multiplem Myelom werden CAR-T-Zell-Therapien im Rahmen klinischer Studien getestet. Die aktuellen Studiendaten der gegen das „B-cell maturation antigen“ (BCMA) gerichteten CAR-T-Zell-Therapien zeigen eine beachtliche Wirksamkeit, die eine baldige Zulassung erwarten lässt. Allerdings erleiden weiterhin die meisten Patienten nach einer Behandlung mit CAR-T-Zellen ein Rezidiv. Hinzu kommt, dass CAR-T-Zell-Therapien zu schwerwiegenden Nebenwirkungen wie Zytokinfreisetzungssyndrom und Neurotoxizität mit teilweise auch letalem Ausgang führen können. Ein angemessenes Kosten-Nutzen-Verhältnis der CAR-T-Zell-Therapie stellt eine weitere Herausforderung dar. Trotz dieser Limitationen erscheint die CAR-T-Zell-Therapie eine attraktive Option für Patienten mit Myelom, sodass diese Therapie das Potenzial hat, in die Standardbehandlung integriert zu werden.
- Published
- 2021
13. [Economics and management of CAR T- cell therapy : Status quo and future perspectives]
- Author
-
F, Kron, J, Franz, A, Kron, and M, Hallek
- Subjects
Receptors, Chimeric Antigen ,Germany ,Neoplasms ,Cell- and Tissue-Based Therapy ,Humans ,Immunotherapy, Adoptive ,Diagnosis-Related Groups - Abstract
Virtually no other topic has attracted more attention in oncology in recent years than chimeric antigen receptor (CAR) T‑cell therapy (CAR T). On the one hand it opens up completely new treatment options for cancer patients, while on the other it generates treatment costs exceeding € 300,000 per treatment.The aim of this work is to analyze the economic, procedural and organizational challenges of CAR T‑cell therapy from the perspective of the service provider, the cost-bearer and the pharmaceutical manufacturer.The current German diagnosis-related-group (G-DRG) catalog, the G‑DRG tariff, of the German Federal Joint Committee (G-BA) guidelines and G‑DRG coding principles were used to evaluate the reimbursement and remuneration system in Germany. Practical experiences of medical sites were integrated in the analysis.The findings demonstrate great economic challenges especially from the perspective of a CAR T site. Increasing certification and qualification efforts lead to financial pressure. Insufficient reimbursement and inadequate cost-covering for CAR T treatment result in budget restrictions for hospitals.High drug costs as well as enormous personnel and infrastructural requirements demand transparent and sufficient reimbursement for hospitals. Interaction between hospital and pharmaceutical manufacturer in the CAR T process might enable new means of cooperation.HINTERGRUND: Kaum ein anderes Thema hat in den vergangenen Jahren in onkologischen Fachkreisen für so viel Aufsehen gesorgt wie die CAR-T-Zell-Therapie (CAR chimärer Antigenrezeptor) – sie eröffnet einerseits vollkommen neue Behandlungsmöglichkeiten für Krebspatienten, kostet andererseits jedoch etwa 300.000 € pro Behandlung.In diesem Beitrag werden ökonomische, prozessuale und organisatorische Herausforderungen der CAR-T-Zell-Therapie analysiert. Dabei wird sowohl die Perspektive der Leistungserbringer und Kostenträger als auch die Sicht der pharmazeutischen Hersteller berücksichtigt.Zur Auswertung der aktuellen Vergütungssituation wurden unter anderem der aktuelle Fallpauschalenkatalog (Diagnosis-Related-Groups[DRG]-Katalog), Beschlüsse des Gemeinsamen Bundesausschusses (G-BA) und Codierleitlinien herangezogen. Infrastrukturelle Besonderheiten und Prozessthemen basieren auf Erfahrungswerten aus dem Praxisalltag.Die Analyse zeigt erhebliche ökonomische und prozessuale Herausforderungen insbesondere für die CAR-T-Zell-Zentren. Zusätzliche mit der Produktion in direktem Zusammenhang stehende Aufwendungen werden nicht adäquat vergütet. Erhebliche Qualitäts- und Qualifizierungsaufwände für Krankenhäuser führen zu einem budgetären Druck auf die Zentren.Die hohen Kosten der CAR-T-Zell-Therapie erfordern eine klare und transparente Definition der Abrechnungs- und Vergütungsregeln. Durch die hohe Interaktionsnotwendigkeit im Zentrum und mit dem pharmazeutischen Hersteller entstehen Mehraufwendungen, die das System nicht kostendeckend abbildet.
- Published
- 2021
14. [Chimeric antigen receptors (CARs): universal tools in adoptive cell therapy]
- Author
-
A, Holzinger and H, Abken
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,Humans ,Immunotherapy, Adoptive - Abstract
The observation that tumor-infiltrating lymphocytes (TIL) after ex vivo amplification can control tumors in the long term led to the concept of redirecting patients' cytolytic T‑cells by a receptor with defined specificity against the tumor.Development of a recombinant receptor-signal molecule (chimeric antigen receptor, CAR) to increase selectivity and enhance anti-tumor immunity.Description of a prototype CAR, overview of the modular composition and further development of CAR technology for use in adoptive immune cell therapy.Intensive research over the last two decades has shown how CAR-mediated T‑cell activation is influenced by factors such as binding affinity, the epitope of the target antigen, its expression density and accessibility on the tumor cells, as well as by the signaling domains and their combination to induce T‑cell activation. The quality and duration of the T‑cell response can be specifically modulated by modifying the modular composition of the CAR; CAR T‑cells can act as "biopharmaceutical factories" (T-cells redirected for unrestricted cytokine-mediated killing, TRUCK) in the tissue by CAR-mediated release of transgenic therapeutic proteins.Adoptive CAR T‑cell therapy has shown clinical efficacy in the treatment of hematological malignancies; the treatment of solid tumors, however, is more challenging. Allogeneic CAR T‑cell technology is aimed at generating "off-the-shelf" CAR T‑cells that are accessible for a large number of patients. A further promising approach is the use of CAR T‑cells for other therapeutic applications such as the treatment of autoimmune diseases.HINTERGRUND: Die Beobachtung, dass tumorinfiltrierende Lymphozyten (TIL) nach Ex-vivo-Amplifikation Tumoren langfristig kontrollieren können, führte zu dem Konzept, zytolytische T‑Zellen des Patienten durch einen Rezeptor mit definierter Spezifität spezifisch gegen den Tumor zu richten.Entwicklung eines rekombinanten Rezeptor-Signal-Moleküls (chimärer Antigenrezeptor, CAR) zur Erhöhung der Wirksamkeit und Selektivität einer T‑Zell-vermittelten Antitumorantwort.Darstellung des Prototyps eines CAR, wesentlicher Aspekte der modularen Struktur und des Entwicklungspotenzials der CAR für die adoptive Immuntherapie.Intensive Forschung in den letzten zwei Dekaden hat aufgezeigt, wie die CAR-vermittelte T‑Zell Aktivierung gezielt beeinflusst werden kann, unter anderem durch die Affinität der Bindung, das Epitop des Zielantigens, dessen Expressionsdichte und Zugänglichkeit auf den Tumorzellen sowie durch die Signaleinheiten und deren Kombination zur Aktivierung der T‑Zellen. Durch Änderung der CAR-Module können Qualität und Dauer der T‑Zell-Antwort weiterhin moduliert werden; durch die CAR-vermittelte Freisetzung transgener, therapeutisch wirksamer Proteine können CAR-T-Zellen als „biopharmazeutische Fabriken“ („T-cells redirected for unrestricted cytokine-mediated killing“, TRUCK) im Gewebe aktiv werden.Die adoptive Immuntherapie mit CAR-T-Zellen wird erfolgreich zur Therapie hämatologischer Tumoren eingesetzt; die Therapie solider Tumoren ist in der Entwicklung. Forschungen zielen unter anderem dahin, allogene CAR-T-Zellen für eine große Anzahl von Patienten „off the shelf“ zur Verfügung zu stellen. Es besteht weiterhin ein erhebliches Entwicklungspotenzial der CAR hin zu anderen therapeutischen Anwendungen, wie für die Therapie von Autoimmunerkrankungen.
- Published
- 2021
15. [CAR T-cell therapy for children and adolescents with acute lymphatic leukemia]
- Author
-
Peter, Bader and Thomas, Klingebiel
- Subjects
Adolescent ,Acute Disease ,Hematopoietic Stem Cell Transplantation ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Child ,Immunotherapy, Adoptive - Abstract
Acute lymphatic leukemia is the most common malignant disease in children. Despite a good prognosis, new therapeutic concepts are needed for patients with refractory and relapsed disease.The importance of chimeric antigen receptor (CAR) T‑cell therapy for patients with refractory and post-stem cell transplantation.Overview of studies in childhood; presentation of extraction, use, side effects and prognostic factors.In 30 patients with a leukemia burden of5% at the time of lymphocytic chemotherapy, the probability of event-free survival after 12 months was 0.70; in patients with a leukemia burden of5%, the probability of event-free survival was only 0.23.Further improvements in vector design will help to improve the effectiveness of this treatment. It will become important to isolate factors that will make it possible to identify patients who will respond to this therapy in the long term.HINTERGRUND: Die akute lymphatische Leukämie (ALL) ist im Kindesalter die häufigste maligne Erkrankung. Trotz guter Prognose bedarf es neuer Therapiekonzepte für Patienten mit refraktärer und rezidivierter Erkrankung.Stellenwert der CAR-T-Zell-Therapie (CAR chimärer Antigenrezeptor) für Patienten mit refraktärer ALL und nach Stammzelltransplantation MATERIAL UND METHODEN: Übersicht über Studien im Kindesalter; Darstellung von Gewinnung, Einsatz, Nebenwirkungen und Prognosefaktoren ERGEBNISSE: Bei 30 Patienten, die mit CAR-T Zellen behandelt wurden und zum Zeitpunkt der lymphozytendepletierenden Chemotherapie eine Leukämielast5 % aufwiesen, betrug die Wahrscheinlichkeit des ereignisfreien Überlebens nach 12 Monaten 0,70; bei Patienten mit einer Leukämielast5 % nur 0,23.Weitere Verbesserungen im Vektordesign werden dazu beitragen, die Effektivität der CAR-T-Zell-Therapie zu verbessern. Wichtig wird die Identifizierung von Faktoren sein, mit denen sich Patienten bestimmen lassen, die auf diese Therapie langfristig ansprechen.
- Published
- 2021
16. [The critically ill CAR T-cell patient : Relevant toxicities, their management and challenges in critical care]
- Author
-
J, Garcia Borrega, K, Heindel, M, Kochanek, C, Warnke, J, Stemmler, M, von Bergwelt-Baildon, T, Liebregts, and B, Böll
- Subjects
Critical Care ,Critical Illness ,T-Lymphocytes ,Humans ,Cytokine Release Syndrome ,Immunotherapy, Adoptive - Abstract
CAR‑T cell therapy has been implemented as clinical routine treatment option during the last decade. Despite beneficial outcomes in many patients severe side effects and toxicities are seen regularly that can compromise the treatment success.Literature review: CAR T‑cell therapy, toxicities and their management RESULTS: The cytokine release syndrome (CRS) and the immune effector cell-associated neurotoxicity syndrome (ICANS) are seen regularly after CAR T‑cell treatment. CRS symptoms can range from mild flu-like symptoms to severe organ dysfunction requiring vasopressor therapy, mechanical ventilation and other intensive care support. ICANS symptoms usually develop later and can range from disorientation and aphasia to potentially life-threatening brain edema. IL‑6 is a key factor in the pathophysiology of CRS. The pathophysiology of ICANS is not fully understood. The ASTCT consensus grading is recommended to stratify patients for different management options. An interdisciplinary team including hematologist, intensivist, neurologists and other specialties is needed to optimize the treatment.Severe and potentially life-threatening toxicities occur regularly after CAR T‑cell therapy. Treatment strategies for CRS and ICANS still need to be evaluated prospectively. Due to the increasing number of patients treated with CAR T‑cells the number of patients requiring temporary intensive care management due to CRS and ICANS is expected to increase during the next years.HINTERGRUND: Die Therapie mit Chimeric-antigen-receptor(CAR)-T-Zellen hat sich als neue Therapieform in der Onkologie etabliert. Im Rahmen einer Therapie mit CAR-T-Zellen kommt es regelhaft zu schweren Nebenwirkungen.Review der Literatur zu CAR-T-Zell-Therapie, Toxizitäten und Nebenwirkungsmanagement ERGEBNIS: Das „cytokine release syndrome“ (CRS) und das „immune effector cell-associated neurotoxicity syndrome“ (ICANS) treten regelhaft im Rahmen einer Therapie mit CAR-T-Zellen auf. Das CRS kann von einer milden grippeähnlichen Symptomatik bis hin zu einem Multiorganversagen führen. Bei ICANS kann sich letztendlich ein lebensbedrohliches Hirnödem entwickeln. Bei der Pathophysiologie des CRS spielt insbesondere das Interleukin‑6 eine entscheidende Rolle. Die Pathophysiologie des ICANS ist nicht vollständig verstanden. Das Management richtet sich nach dem Schweregrad gemäß Grading der American Society for Transplantation and Cellular Therapy (ASTCT). Beim CRS werden Tocilizumab und Kortikosteroide, für das ICANS Kortikosteroide empfohlen.Nach einer CAR-T-Zell-Therapie kommt es regelhaft zu potenziell lebensbedrohlichen Komplikationen. Weitergehende Therapieansätze sind bislang nicht gut untersucht. Eine interdisziplinäre Zusammenarbeit zwischen Intensivmediziner*innen, Hämatolog*innen, Neurolog*innen und Ärzt*innen anderer Fachabteilungen ist von entscheidender Bedeutung. Aufgrund der steigenden Patientenzahlen ist von einer Zunahme an Patienten mit Notwendigkeit einer intensivmedizinischen Behandlung im Rahmen einer CAR-T-Zelltherapie auszugehen.
- Published
- 2020
17. [Genetically modified regulatory T cells: therapeutic concepts and regulatory aspects]
- Author
-
Attila, Sebe, Brigitte, Anliker, Juliane, Rau, and Matthias, Renner
- Subjects
Immune tolerance ,Adoptive T‑cell therapy ,Regulatory T cells ,Immuntoleranz ,Genetisch modifizierte Zellen ,Immunotherapy, Adoptive ,T-Lymphocytes, Regulatory ,Regulatorische T‑Zellen ,Regulatorische Rahmenbedingungen ,Regulatory framework ,Germany ,Leitthema ,Humans ,Adoptive T‑Zelltherapie ,Genetically modified cells - Abstract
Adoptive T‑Zelltherapien sind neuartige Konzepte zur Behandlung verschiedener Krankheiten. CAR-T-Zellen sind dabei als Letztlinientherapie für fortgeschrittene B‑Zelllymphome und die B‑Zellleukämie etabliert und zugelassen. TCR-basierte T‑Zellen als Behandlungsoption verschiedener hämatologischer und solider Tumoren befinden sich in der klinischen Entwicklung. Genetisch modifizierte regulatorische T‑Zellen stehen dagegen noch am Anfang ihrer klinischen Entwicklung zur Induktion von Immuntoleranz in einer Vielzahl von Anwendungsgebieten. In diesem Artikel wird zunächst ein Überblick über die Funktion der regulatorischen T‑Zellen für die Induktion der Immuntoleranz sowie über ihre Rolle im Pathomechanismus bestimmter Immunerkrankungen gegeben und der aktuelle Stand der klinischen Entwicklungen von therapeutischen Ansätzen auf Basis genetisch modifizierter regulatorischer T‑Zellen zusammengefasst. Im Weiteren werden die regulatorisch-wissenschaftlichen Anforderungen und Herausforderungen hinsichtlich Herstellung und Qualitätskontrolle sowie nichtklinischer und klinischer Testung genetisch modifizierter regulatorischer T‑Zellen als Arzneimittel für neuartige Therapien diskutiert.
- Published
- 2020
18. [Batten down the hatches: CAR T-cells - immuno-oncology meets intensive care medicine]
- Author
-
Jorge Garcia, Borrega, Michael, von Bergwelt-Baildon, and Boris, Böll
- Subjects
Critical Care ,Lymphoma ,Humans ,Neurotoxicity Syndromes ,Cytokine Release Syndrome ,Immunotherapy, Adoptive - Abstract
CAR T-cell treatment has brought a significant benefit for patients with relapsed and refractory lymphoma and even long lasting remissions seem to be possible. Despite the good results CAR T-cell treatment is associated with severe and potentially life-threatening adverse effects. Patients regularly develop Cytokine Release Syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) after the transfusion of CAR T-cells. In addition to these immunological reactions patients can develop severe and life-threatening infections and other related adverse effects. Due to the increasing number of patients receiving CAR T-cells and the upcoming clinical trials more and more patients will potentially require intensive care treatment for the adverse effects after CAR T-cell treatment. The management of these patients requires an extensive diagnostic workup and complex treatment. An interdisciplinary team of hematologist, intensivist and others like neurologist and palliative medicine consultants is crucial for the best treatment of these patients. Together with the increasing number of patients this can lead to logistical issues for the intensive care departments of centers offering CAR T-cell therapies.Das Cytokine-Release-Syndrome (CRS) ist die häufigste Nebenwirkung einer CAR-T-Zell-Therapie und kann von leichtem Fieber bis zu einem Multiorganversagen führen. Pathophysiologisch kommt es beim CRS zu einem Zytokinsturm und trotz einer Therapie mit Tocilizumab sind refraktäre und tödliche Verläufe beschrieben. Die Symptome des Immune-Effector-Cell-associated-Neurotoxicity-Syndrome (ICANS) variieren von leichter Desorientiertheit bis zum lebensbedrohlichen Hirnödem. Die Pathophysiologie und Therapie des ICANS sind noch nicht ausreichend erforscht. Die Differenzialdiagnosen von CRS und ICANS sind komplex und umfassen neben Infektionen und Sepsis unter anderem auch eine Toxizität der vorhergehenden Therapie, ein Tumorlysesyndrom und nicht zuletzt einen Progress der Grunderkrankung. Ein klinischer oder laborchemischer Parameter zum sicheren Beweis oder Ausschluss eines CRS oder ICANS gibt es zum heutigen Zeitpunkt nicht. INTENSIVMEDIZINISCHE RELEVANZ UND POTENZIELLE ENTWICKLUNGEN DER CAR-T-ZELL-THERAPIE: Erste Auswertungen von Real-world-Daten deuten auf eine höhere Rate an schweren Nebenwirkungen im Rahmen der CAR-T-Zell-Therapie als in den Zulassungsstudien hin. Für die Indikation r/r-DLBCL könnten schätzungsweise bis zu maximal 300 Patienten pro Jahr in Deutschland eine intensivmedizinische Betreuung im Rahmen der CAR-T-Zell-Therapie benötigen. Studien mit wesentlich häufigeren soliden Tumoren könnten die Patientenzahl drastisch erhöhen. THERAPIEZIEL BEI CAR-T-ZELL-PATIENTEN UND ENTSCHEIDUNGEN BEI THERAPIEZIELäNDERUNG: Aufgrund des neuen Therapiekonzepts kann ein Konflikt zwischen bislang palliativem Patientenkollektiv und nun möglicherweise langfristigen Remissionen entstehen. Eine frühzeitige Aufklärung über potenziell lebensbedrohliche Nebenwirkungen im Rahmen der Therapie und eine interdisziplinäre Besprechung der Therapieziele mit den Patienten ist entscheidend.
- Published
- 2019
19. [Oncological Intensive Care Medicine: From CPR to CAR T cells]
- Author
-
Michael, von Bergwelt-Baildon
- Subjects
Critical Care ,Neoplasms ,Humans ,Medical Oncology ,Immunotherapy, Adoptive ,Cardiopulmonary Resuscitation - Published
- 2019
20. [Immuno-Oncology Meets Intensive Care Medicine: CAR-T cells]
- Author
-
Boris, Böll, Jorge Garcia, Borrega, Peter, Schellongowski, Marion, Subklewe, and Michael, von Bergwelt-Baildon
- Subjects
Disease Models, Animal ,Mice ,Critical Care ,Neoplasms ,Animals ,Humans ,Cytokine Release Syndrome ,Immunotherapy, Adoptive - Abstract
CAR-T cells, genetically modified tumor-targeted t-cells, revolutionized the treatment of refractory B-cell malignancies. CAR-T cell treatment however, is invariably associated with severe immune-mediated toxicities, namely Cytokine Release Syndrome (CRS) and neurological toxicity (CRES/ICANS). Although knowledge on the pathomechanism of these toxicities is still very limited, recent findings including mouse models might allow prediction, earlier recognition and targeted treatment of patients suffering from these potentially life-threatening side effects. This article summarizes current knowledge and recent findings on the management of severe CAR-T associated toxicities and gaps of knowledge stressing the importance of an interdisciplinary approach including hematology-oncology and Intensive Care Medicine.
- Published
- 2019
21. [Bridging before CAR T-cell therapy-a new opportunity for radiotherapy?]
- Author
-
Oertel M and Eich HT
- Subjects
- Humans, T-Lymphocytes, Immunotherapy, Adoptive, Radiation Oncology
- Published
- 2021
- Full Text
- View/download PDF
22. [What is established in cell therapies? : Possibilities and limits in immuno-oncology]
- Author
-
A, Quaiser and U, Köhl
- Subjects
Killer Cells, Natural ,Neoplasms ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Humans ,Immunotherapy, Adoptive - Abstract
Cell and gene therapy as part of immuno-oncology has reached an important milestone in medicine. After decades of experience stem cell transplantation is well established with worldwide1 million transplantations to date. Due to the improved success of the last years using chimeric antigen receptor (CAR) T cells for CD19 positive leukemia and lymphomas, the interest in cellular therapies is continuously increasing. The current review also gives a short overview about donor lymphocytes, antigen-specific T cells, regulatory T cells, natural killer (NK) cells, mesenchymal stromal cells and induced pluripotent stem (iPS) cells in immuno-oncology.
- Published
- 2018
23. [CARs, CRS and neurotoxicity: severe complications after administration of immunotherapy : Essentials for intensivists]
- Author
-
J, Prinz, Y, d'Hargues, P, Gödel, A, Shimabukuro-Vornhagen, M, Kochanek, and B, Böll
- Subjects
Receptors, Chimeric Antigen ,Cytokines ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Cytokine Release Syndrome ,Immunotherapy, Adoptive - Abstract
The development of chimeric antigen receptor (CAR) T‑cells has shown promising results in relapsed/refractory B‑cell acute lymphoblastic leukemia/lymphoma (B-ALL) and diffuse large cell B‑cell lymphoma. Complications, especially cytokine release syndrome (CRS) and CAR T‑cell related encephalopathy syndrome (CRES), can be life threatening. The management of both plays a key role in CAR T‑cell therapy.Diagnosis, clinical presentation and development of complications in the treatment with CAR T‑cells.Summary of incidence, mortality and treatment of severe complications after administration of CAR T‑cells referring to current studies and therapy recommendations.Complications after administration of CAR T‑cells, especially CRS and CRES, can be life threatening. The timely identification of side effects and their appropriate treatment usually leads to complete recovery.Using a therapy algorithm in the treatment with CAR T‑cells allows safe management of toxicities and can be helpful in recognizing them in time.
- Published
- 2018
24. [CAR T-cell therapy for children and adolescents with acute lymphatic leukemia].
- Author
-
Bader P and Klingebiel T
- Subjects
- Acute Disease, Adolescent, Child, Humans, Immunotherapy, Adoptive, Hematopoietic Stem Cell Transplantation, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Background: Acute lymphatic leukemia is the most common malignant disease in children. Despite a good prognosis, new therapeutic concepts are needed for patients with refractory and relapsed disease., Question: The importance of chimeric antigen receptor (CAR) T‑cell therapy for patients with refractory and post-stem cell transplantation., Material and Methods: Overview of studies in childhood; presentation of extraction, use, side effects and prognostic factors., Results: In 30 patients with a leukemia burden of <5% at the time of lymphocytic chemotherapy, the probability of event-free survival after 12 months was 0.70; in patients with a leukemia burden of >5%, the probability of event-free survival was only 0.23., Conclusions: Further improvements in vector design will help to improve the effectiveness of this treatment. It will become important to isolate factors that will make it possible to identify patients who will respond to this therapy in the long term.
- Published
- 2021
- Full Text
- View/download PDF
25. [Chimeric antigen receptors (CARs): universal tools in adoptive cell therapy].
- Author
-
Holzinger A and Abken H
- Subjects
- Cell- and Tissue-Based Therapy, Humans, Immunotherapy, Adoptive, Receptors, Antigen, T-Cell genetics, T-Lymphocytes, Receptors, Chimeric Antigen genetics
- Abstract
Background: The observation that tumor-infiltrating lymphocytes (TIL) after ex vivo amplification can control tumors in the long term led to the concept of redirecting patients' cytolytic T‑cells by a receptor with defined specificity against the tumor., Objectives: Development of a recombinant receptor-signal molecule (chimeric antigen receptor, CAR) to increase selectivity and enhance anti-tumor immunity., Methods: Description of a prototype CAR, overview of the modular composition and further development of CAR technology for use in adoptive immune cell therapy., Results: Intensive research over the last two decades has shown how CAR-mediated T‑cell activation is influenced by factors such as binding affinity, the epitope of the target antigen, its expression density and accessibility on the tumor cells, as well as by the signaling domains and their combination to induce T‑cell activation. The quality and duration of the T‑cell response can be specifically modulated by modifying the modular composition of the CAR; CAR T‑cells can act as "biopharmaceutical factories" (T-cells redirected for unrestricted cytokine-mediated killing, TRUCK) in the tissue by CAR-mediated release of transgenic therapeutic proteins., Conclusion: Adoptive CAR T‑cell therapy has shown clinical efficacy in the treatment of hematological malignancies; the treatment of solid tumors, however, is more challenging. Allogeneic CAR T‑cell technology is aimed at generating "off-the-shelf" CAR T‑cells that are accessible for a large number of patients. A further promising approach is the use of CAR T‑cells for other therapeutic applications such as the treatment of autoimmune diseases.
- Published
- 2021
- Full Text
- View/download PDF
26. [CAR T-cell therapy for multiple myeloma].
- Author
-
Zhou X, Einsele H, and Danhof S
- Subjects
- B-Cell Maturation Antigen, Humans, Immunotherapy, Adoptive, Neoplasm Recurrence, Local, Multiple Myeloma therapy, Receptors, Chimeric Antigen
- Abstract
Chimeric antigen receptor (CAR) T-cell treatment is a novel immunotherapy utilizing the patient's own immune system as a "weapon against tumor cells". In patients with multiple myeloma (MM), CAR T-cell therapy has been investigated in clinical trials. The current data on B‑cell maturation antigen (BCMA)-targeted CAR T-cells have shown impressive efficacy, and official approval is expected shortly. However, the majority of patients relapse after CAR T-cell therapy. Moreover, the treatment can cause severe adverse events such as cytokine release syndrome and neurotoxicity with lethal outcome. The cost-benefit ratio of this treatment also needs to be optimized. Despite these limitations, CAR T-cell therapy represents an attractive option for patients with MM and has the potential to be incorporated into the standard of care.
- Published
- 2021
- Full Text
- View/download PDF
27. [CAR T-cell therapy for malignant B-cell lymphoma : A new treatment paradigm].
- Author
-
Balke-Want H and Borchmann P
- Subjects
- Child, Humans, Immunotherapy, Adoptive, T-Lymphocytes, Leukemia, Lymphocytic, Chronic, B-Cell, Lymphoma, B-Cell therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Abstract
Following the first demonstration of efficacy of anti-CD19-directed chimeric antigen receptor (CAR) T cells in a patient with relapsed chronic lymphocytic leukemia (CLL) in 2011, pivotal studies for this innovative therapy were initially conducted in multiple relapsed or refractory (r/r) childhood and young adult acute B‑cell leukemia and in aggressive adult B‑cell lymphoma. The studies demonstrated efficacy even in chemotherapy-refractory disease, resulting in the first approval of autologous and genetically engineered T cells for the treatment of r/r B‑cell acute lymphoblastic leukemia (B-ALL) in the US for the product tisagenlecleucel (Kymriah®, Novartis) back in 2018. Approval for the treatment of r/r aggressive B‑cell lymphoma followed shortly thereafter for tisagenlecleucel and axicabtagene ciloleucel (Yescarta, Kite/Gilead). This review focuses on the treatment of aggressive B‑cell lymphoma and other CD19 positive B‑cell lymphomas by summarizing the study results of clinically tested CAR T cells, discussing possible resistance mechanisms, and providing an outlook on ongoing studies with new target antigens for the treatment of B‑cell lymphomas.
- Published
- 2021
- Full Text
- View/download PDF
28. [Multiple myeloma-soon curable?]
- Author
-
Goldschmidt H
- Subjects
- Antibodies, Monoclonal, Germany, Humans, Proteasome Inhibitors, T-Lymphocytes, Multiple Myeloma diagnosis, Multiple Myeloma therapy
- Abstract
Multiple myeloma (MM) is one of the most frequent cancerous diseases of the hemopoietic system. Over the past 60 years the systemic treatment has undergone multiple changes, from alkylating agents to high-dose therapy followed by autologous peripheral blood stem cell transplantation up to immunomodulating substances and proteasome inhibitors. The treatment of MM is currently undergoing a renewed transition. In recent years monoclonal antibodies have decisively extended the treatment options. Long-term remission is achieved more often. Due to progress in immuno-oncological treatment the prognosis of intensively treated patients with a very short life-expectancy can be improved in the future. It is to be expected that MM will be curable in the medium term. The concentration of free light chains in serum, lesions in magnetic resonance imaging (MRI) and bone marrow infiltration are parameters that are incorporated into the treatment indications. In clinical studies patients with smoldering myeloma are already being treated to delay progression, to increase the remission rates or to achieve long-term remission with negative minimal residual disease. Taking the chromosomal alterations and serological parameters into consideration, the prognosis of patients with MM can nowadays be very well discriminated. In currently running studies high-risk patients are being separately and mostly aggressively treated. Imaging is of great importance in MM. Using MRI focal lesions can be detected even before bone destruction. In this year chimeric antigen receptor (CAR) T cell treatment of MM will be approved for the first time in Germany. Novel antibody constructs, such as belantamab mafodotin, are or will be introduced for a late recurrence.
- Published
- 2021
- Full Text
- View/download PDF
29. [CAR T cells as drugs for novel therapies (advanced therapy medicinal products)].
- Author
-
Köhl U and Abken H
- Subjects
- Humans, T-Lymphocytes, Neoplasms, Pharmaceutical Preparations, Receptors, Chimeric Antigen genetics
- Abstract
Background: Two commercial chimeric antigen receptor (CAR) T cell products, axicabtagene-ciloleucel (Yescarta®) and tisagenlecleucel (Kymriah®), are registered for the treatment of B cell neoplasia, for which an increased supply of CAR T cell products is required., Problem: The production of patient-specific CAR T cells as advanced therapy medicinal products (ATMPs) poses considerable challenges with respect to logistics, regulation, and manufacturing., Method: Review of the CAR T cell manufacturing process and the regulatory network, the current challenges, and future development capabilities of CAR T cells for adoptive immunotherapy., Results: CAR T cells are manufactured under individualized, laborious, good manufacturing practice-conforming processes in decentralized or in specialized centers. Starting from the patient's leukapheresis product, T cells are genetically engineered ex vivo with a CAR, amplified, and after extensive quality control re-applied to the patient. Most CAR T cell products are manufactured in a manual or semi-automated process; fully automated, supervised, and closed systems are increasingly applied to meet the need for a growing number of CAR T cell products. In this setting, research aims at providing allogeneic CAR T cell products or non-T cells such as natural killer cells for broad applications., Conclusion: The significance of CAR T cells in adoptive immunotherapy is continuously growing. As individualized cell products, manufacturing requires highly efficient processes under the control of harmonized protocols and regulations so as to ensure the quality of the ATMP in view of increasing demand and to develop new fields in therapy.
- Published
- 2021
- Full Text
- View/download PDF
30. [The critically ill CAR T-cell patient : Relevant toxicities, their management and challenges in critical care].
- Author
-
Garcia Borrega J, Heindel K, Kochanek M, Warnke C, Stemmler J, von Bergwelt-Baildon M, Liebregts T, and Böll B
- Subjects
- Critical Care, Cytokine Release Syndrome, Humans, T-Lymphocytes, Critical Illness therapy, Immunotherapy, Adoptive
- Abstract
Background: CAR‑T cell therapy has been implemented as clinical routine treatment option during the last decade. Despite beneficial outcomes in many patients severe side effects and toxicities are seen regularly that can compromise the treatment success., Methods: Literature review: CAR T‑cell therapy, toxicities and their management RESULTS: The cytokine release syndrome (CRS) and the immune effector cell-associated neurotoxicity syndrome (ICANS) are seen regularly after CAR T‑cell treatment. CRS symptoms can range from mild flu-like symptoms to severe organ dysfunction requiring vasopressor therapy, mechanical ventilation and other intensive care support. ICANS symptoms usually develop later and can range from disorientation and aphasia to potentially life-threatening brain edema. IL‑6 is a key factor in the pathophysiology of CRS. The pathophysiology of ICANS is not fully understood. The ASTCT consensus grading is recommended to stratify patients for different management options. An interdisciplinary team including hematologist, intensivist, neurologists and other specialties is needed to optimize the treatment., Discussion: Severe and potentially life-threatening toxicities occur regularly after CAR T‑cell therapy. Treatment strategies for CRS and ICANS still need to be evaluated prospectively. Due to the increasing number of patients treated with CAR T‑cells the number of patients requiring temporary intensive care management due to CRS and ICANS is expected to increase during the next years.
- Published
- 2021
- Full Text
- View/download PDF
31. [Perspectives on CAR T-cell treatment].
- Author
-
Viardot A
- Subjects
- Antigens, CD19, Humans, Receptors, Antigen, T-Cell, Receptors, Chimeric Antigen, Immunotherapy, Adoptive, T-Lymphocytes
- Abstract
CARTs (T-cells with a chimeric antigen receptor) are one of the most innovative and exciting developments in cancer medicine. In August 2018, two novel drugs were approved in the European Union, both for the treatment of relapsed and refractory aggressive lymphoma, one of them also for the treatment of acute lymphoblastic leukemia in children and young adults up to 25 years. In the foreseeable future, further approvals are expected, e.g., in mantle cell lymphoma and multiple myeloma. Clinical trials with new constructs are ongoing in almost all cancer entities. The introduction of this completely new principle implicates unusual challenges: (1) The use of genetically modified "living drugs" is challenging from a regulatory point of view and might require a lifelong surveillance of the patient, (2) companies and authorities make high demands on the quality management at the sites, (3) the price of about 280,000 € for the approved treatments rises new socioeconomic and ethical questions. However, CARTs will change the therapeutic landscape in many cancers in upcoming years.
- Published
- 2020
- Full Text
- View/download PDF
32. [Chimeric antigen receptors in oncology: clinical applications and new developments].
- Author
-
Michels A, Hartmann J, and Buchholz CJ
- Subjects
- Animals, Germany, Humans, Immunotherapy, Adoptive, Medical Oncology, Receptors, Antigen, T-Cell genetics, T-Lymphocytes, Receptors, Chimeric Antigen
- Abstract
In 2018, two novel cancer therapies based on chimeric antigen receptors (CARs) were granted marketing authorization in the European Union. Authorized for use against advanced lymphoma and/or leukemia, the products were at the center of international attention, not only due to their novel mode of action and their encouraging efficacy but also because of their sometimes severe side effects and the economic and logistic challenges posed by their manufacture. Now, almost two years later, hundreds of active clinical trials emphasize the global drive to harness the full potential of CAR technology.In this article, we describe the mode of action of CAR T and CAR NK cells and review the clinical testing situation as well as early real-world data. In recent years, preclinical studies using advanced animal models have provided first insights into the mechanisms underlying the severe side effects of CAR T therapy. We summarize their results and describe the available models. Additionally, we discuss potential solutions to the hurdles currently limiting CAR technology. So far used as last-line treatment for patients with aggressive disease, CAR technology has the potential to become a new, broadly effective standard for tumor therapy.
- Published
- 2020
- Full Text
- View/download PDF
33. [Genetically modified regulatory T cells: therapeutic concepts and regulatory aspects].
- Author
-
Sebe A, Anliker B, Rau J, and Renner M
- Subjects
- Germany, Humans, Immunotherapy, Adoptive, T-Lymphocytes, Regulatory
- Abstract
Adoptive T‑cell therapies are emerging tools to combat various human diseases. CAR‑T cells are approved and marketed as last line therapeutics in advanced B‑cell lymphomas and leukemias. TCR-engineered T cells are being evaluated in clinical trials for a variety of hematological and solid tumors. Genetically modified regulatory T cells, however, are still in the initial stages of clinical development for the induction of immune tolerance in various indications.Here we outline the general role of regulatory T cells in establishing self-tolerance and the mechanisms by which these suppress the effector immune cells. Further, the role of regulatory T cells in the pathomechanism of certain immune diseases is presented, and the current status of clinical developments of genetically modified Treg cells is discussed. We also present the regulatory framework for genetically modified regulatory T cells as advanced therapy medicinal products, including aspects of manufacture and quality control, as well as nonclinical and clinical development requirements.
- Published
- 2020
- Full Text
- View/download PDF
34. [CARs, CRS and neurotoxicity: severe complications after administration of immunotherapy : Essentials for intensivists].
- Author
-
Prinz J, d'Hargues Y, Gödel P, Shimabukuro-Vornhagen A, Kochanek M, and Böll B
- Subjects
- Cytokine Release Syndrome, Cytokines, Humans, Immunotherapy, Adoptive, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Chimeric Antigen
- Abstract
Background: The development of chimeric antigen receptor (CAR) T‑cells has shown promising results in relapsed/refractory B‑cell acute lymphoblastic leukemia/lymphoma (B-ALL) and diffuse large cell B‑cell lymphoma. Complications, especially cytokine release syndrome (CRS) and CAR T‑cell related encephalopathy syndrome (CRES), can be life threatening. The management of both plays a key role in CAR T‑cell therapy., Objectives: Diagnosis, clinical presentation and development of complications in the treatment with CAR T‑cells., Materials and Methods: Summary of incidence, mortality and treatment of severe complications after administration of CAR T‑cells referring to current studies and therapy recommendations., Results: Complications after administration of CAR T‑cells, especially CRS and CRES, can be life threatening. The timely identification of side effects and their appropriate treatment usually leads to complete recovery., Conclusions: Using a therapy algorithm in the treatment with CAR T‑cells allows safe management of toxicities and can be helpful in recognizing them in time.
- Published
- 2020
- Full Text
- View/download PDF
35. Exploiting natural killer cells for therapy of melanoma
- Author
-
Oliver Hölsken, Adelheid Cerwenka, and Matthias Miller
- Subjects
Lymphokine-activated killer cell ,Skin Neoplasms ,medicine.medical_treatment ,Melanoma ,Models, Immunological ,Dermatology ,Immunotherapy ,Human leukocyte antigen ,Biology ,Natural killer T cell ,medicine.disease ,Immunotherapy, Adoptive ,Killer Cells, Natural ,Interleukin 21 ,NK-92 ,Immunology ,Interleukin 12 ,medicine ,Humans - Abstract
During the recent years, immunotherapy has obtained substantial impact on the clinical treatment of melanoma. Besides promising approaches based on T lymphocytes, natural killer (NK) cells have gained more and more attention as anti-melanoma effector cells. NK cell activation is inhibited by HLA class I molecules expressed by target cells, so they preferentially attack tumor cells that express low levels of HLA class I. Partial or complete loss of HLA class I expression is a frequent event during the development of melanoma. In parallel, ligands for activating NK cell receptors become induced upon malignant transformation. Thus, melanoma cells are often efficiently recognized and lysed by NK cells at least in vitro. In vivo, however, melanomas have developed multiple sophisticated strategies to escape from NK cell mediated attack. Several novel approaches aim at harnessing NK cells to treat melanoma patients and to counteract existing tumor escape mechanisms. This review summarizes the most recent advances in the field.
- Published
- 2015
36. [Oncological Intensive Care Medicine: From CPR to CAR T cells].
- Author
-
von Bergwelt-Baildon M
- Subjects
- Cardiopulmonary Resuscitation, Humans, Immunotherapy, Adoptive, Critical Care, Medical Oncology, Neoplasms therapy
- Abstract
Competing Interests: Disclosure The authors report no conflicts of interest in this work.
- Published
- 2019
- Full Text
- View/download PDF
37. [What is established in cell therapies? : Possibilities and limits in immuno-oncology].
- Author
-
Quaiser A and Köhl U
- Subjects
- Humans, Killer Cells, Natural, T-Lymphocytes, Cell- and Tissue-Based Therapy, Immunotherapy, Adoptive, Neoplasms therapy
- Abstract
Cell and gene therapy as part of immuno-oncology has reached an important milestone in medicine. After decades of experience stem cell transplantation is well established with worldwide >1 million transplantations to date. Due to the improved success of the last years using chimeric antigen receptor (CAR) T cells for CD19 positive leukemia and lymphomas, the interest in cellular therapies is continuously increasing. The current review also gives a short overview about donor lymphocytes, antigen-specific T cells, regulatory T cells, natural killer (NK) cells, mesenchymal stromal cells and induced pluripotent stem (iPS) cells in immuno-oncology.
- Published
- 2018
- Full Text
- View/download PDF
38. [Perspectives on immunotherapy for hepatocellular carcinoma]
- Author
-
N, Schmidt, N, Büttner, and R, Thimme
- Subjects
CD4-Positive T-Lymphocytes ,Carcinoma, Hepatocellular ,Lymphocytes, Tumor-Infiltrating ,Liver Neoplasms ,Humans ,Immunotherapy ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive - Abstract
Hepatocellular carcinoma (HCC) is the fifth most common tumor worldwide with a high mortality. Available therapeutical options are limited, thus the development of new, innovative therapeutic strategies is crucial. Based on the immune system's antitumoral effect, immunotherapy is a promising new treatment option. Specific antitumoral T-cell responses can be detected in patients with HCC, however, their impact on tumor control seems to be rather weak. Various different immunosuppressive mechanisms seem to contribute to the failure of tumor-specific T-cell responses. Thus, the aim of immunotherapeutic strategies is to address these mechanisms of T-cell failure and to induce new or to boost existing antitumoral immune responses.
- Published
- 2013
39. [The role of radiotherapy in the induction of antitumor immune responses]
- Author
-
G, Multhoff, U S, Gaipl, and G, Niedermann
- Subjects
CD4-Positive T-Lymphocytes ,Immunity, Cellular ,Apoptosis ,Chemoradiotherapy ,Dendritic Cells ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,Combined Modality Therapy ,Immunotherapy, Adoptive ,Killer Cells, Natural ,Antibody Specificity ,HLA Antigens ,Neoplasms ,Immune Tolerance ,Humans - Published
- 2012
40. [Treatment of a questionable prostate carcinoma recurrence with oncolytic viruses, dendritic cells and heat shock proteins in established naturopathy practice]
- Author
-
Rainer, Hakimi
- Subjects
Male ,Oncolytic Virotherapy ,Treatment Outcome ,Naturopathy ,Artesunate ,Humans ,Prostatic Neoplasms ,Dendritic Cells ,Neoplasm Recurrence, Local ,Immunotherapy, Adoptive ,Artemisinins ,Heat-Shock Proteins - Published
- 2012
41. [Adoptive T-cell therapy of rhabdomyosarcoma]
- Author
-
K, Simon-Keller, A, Paschen, S, Eichmüller, S, Gattenlöhner, S, Barth, E, Koscielniak, I, Leuschner, P, Stöbel, A, Hombach, H, Abken, and A, Marx
- Subjects
Cell Line, Tumor ,T-Lymphocytes ,Rhabdomyosarcoma ,Receptors, Antigen, T-Cell ,Humans ,Receptors, Cholinergic ,Cytotoxicity Tests, Immunologic ,Chimerism ,Immunoglobulin Fragments ,Immunotherapy, Adoptive ,Immunoglobulin Fc Fragments - Abstract
To improve survival of patients with advanced rhabdomyosarcomas (RMS), we aimed to adoptively transfer T-cells with redirected specificity for the fetal acetylcholine receptor (AChR), an RMS-specific cell surface antigen.A "second generation" chimeric antigen receptor (CAR) with a combined CD28-CD3ζ signaling domain was derived from our previously described chimeric antigen receptor composed of an extracellular human anti-fAChR antibody fragment, an Fc hinge region, and the intracellular T-cell receptor zeta chain. Lymphocytes from the peripheral blood were modified by retroviral transduction and monitored by FACS analysis. Cytotoxicity of modified T-cells towards RMS cells was recorded by MTT-based viability tests; expression of co-stimulatory molecules and anti-apoptotic genes was studied by FACS and qRT-PCR analysis.Co-stimulatory molecules were expressed in low levels on RMS cells giving the rationale to generate a CD28-CD3ζ signalling CAR (chimeric antigen receptor) for redirecting T-cells. T-cells were successfully engineered with the "second generation" AChR-specific chimeric antigen receptor. Despite of high CAR expression engineered T-cells showed low killing efficiency towards RMS compared to redirected killing of CD20+ lymphoma or CEA-expressing adenocarcinoma cell lines when redirected by CD20- and/or CEA-specific CAR.Data suggest that RMS cells exhibit resistance to a T-cell attack redirected by a fAChR-specific CAR. Inhibition of anti-apoptotic pathways in those cells may improve sensitivity to conventional as well as T-cell-based therapeutics.
- Published
- 2010
42. [Renal cell carcinoma associated proteins. Isolation, cloning and immunogenicity evaluation]
- Author
-
A, Haferkamp, M, Hohenfellner, R, Hautmann, and M, Zöller
- Subjects
B-Lymphocytes ,Mice, SCID ,Cancer Vaccines ,Immunotherapy, Adoptive ,Kidney Neoplasms ,Mice ,Antigens, Neoplasm ,Biomarkers, Tumor ,Disease Progression ,Vaccines, DNA ,Animals ,Humans ,Cloning, Molecular ,Mitogen-Activated Protein Kinases ,Carcinoma, Renal Cell ,Neoplasm Staging ,T-Lymphocytes, Cytotoxic - Published
- 2007
43. Dendritic cell-based immunotherapy of malignant melanoma: success and limitations
- Author
-
Edgar Schmitt, Jürgen Knop, Andrea Tuettenberg, and Helmut Jonuleit
- Subjects
Skin Neoplasms ,Effector ,T cell ,medicine.medical_treatment ,Melanoma ,Models, Immunological ,Cancer ,Dermatology ,Immunotherapy ,Dendritic cell ,Dendritic Cells ,Biology ,medicine.disease ,Immunotherapy, Adoptive ,Melanoma Vaccine ,medicine.anatomical_structure ,Immune system ,Treatment Outcome ,Immunology ,Practice Guidelines as Topic ,medicine ,Humans ,Practice Patterns, Physicians' - Abstract
Dendritic cells (DC) are professional antigen-presenting cells in the immune system which are able to induce primary T-cell responses. Because of their central role in the initiation of immune responses, DC are an important tool for tumor-antigen-specific immunotherapy of cancer. DC vaccination using tumor-antigen-loaded DC has led to tumor regression in individual advanced-stage cancer patients. However, there is a discrepancy between strong and antigen-specific T cell responses in vaccinated cancer patients detectable ex vivo and only weak clinical responses. In most cases the immune system of advanced stage IV cancer patients allows only a temporary anti-tumor response and increasing evidence exists that active suppressive mechanisms of the immune system as well as of the tumor itself ultimately prevent "autoaggressive" immune reactions against the tumor. Active counter-regulation of effector T cells by tumor-antigen-specific regulatory T-cell (Treg) populations play a central role in limiting the efficacy of the vaccines. Nevertheless, recent studies have shown that DC,additionally activated byToll-Like-receptor ligands (TLRL) can neutralize these suppressive effects of Treg and facilitate the induction of long-lasting effector T cell responses even in the presence of activated Treg. These studies open a new way for "conditioning" of DC by TLRL and might significantly enhance the efficiency of DC-based melanoma vaccines in the future.
- Published
- 2007
44. [Squamous cell carcinoma of the head and neck. Principles and current concepts of immunotherapy]
- Author
-
T K, Hoffmann, T L, Whiteside, and H, Bier
- Subjects
Head and Neck Neoplasms ,Practice Guidelines as Topic ,Vaccination ,Humans ,Genetic Therapy ,Immunotherapy ,Practice Patterns, Physicians' ,Cancer Vaccines ,Immunotherapy, Adoptive - Abstract
Biologic therapies able to induce or up-regulate anti-tumor immune responses could represent a complementary approach to improve the conventional treatment of squamous cell carcinomas of the head and neck (SCCHN). Patients with SCCHN are frequently immunocompromised due to the elimination and dysfunction of critical immune effector cells. Therefore, it might be necessary to restore these immune functions to allow for the generation of effective anti-tumor host responses. Simultaneously, to prevent tumor escape from immunological recognition and destruction, it might also be necessary to alter antigenic and immunogenic attributes of the malignant cells. The present overview summarizes general aspects, historical data, and recent advances in the field of immunotherapy of SCCHN, including non-specific immune stimulation, transfer of immunocompetent cells, gene therapy, use of monoclonal antibodies, and anti-cancer vaccines.
- Published
- 2005
45. [Significance of dendritic cells for the immunotherapy of tumors]
- Author
-
J B, Weise, S, Maune, D, Kabelitz, and A, Heiser
- Subjects
Neoplasms ,Immune Tolerance ,Animals ,Humans ,Dendritic Cells ,Immunotherapy, Adoptive - Published
- 2005
46. [Specific cellular immunotherapy of renal cell carcinoma. Current status and prospects]
- Author
-
M, Ringhoffer and J E, Gschwend
- Subjects
Survival Rate ,T-Lymphocytes ,Humans ,Dendritic Cells ,Cancer Vaccines ,Carcinoma, Renal Cell ,Immunotherapy, Adoptive ,Kidney Neoplasms - Abstract
Renal cell carcinoma (RCC) is susceptible to immunomodulating therapies. This is proven by clinical responses to unspecific immunotherapy with cytokines. Understanding the mechanisms of antigen presentation and recognition by T cells enables us to expand T-cell clones which are capable of recognizing specific tumor-associated antigens (TAA). The use of dendritic cells (DC) in specific cellular immunotherapy could be beneficial because of their outstanding properties in antigen presentation and T-cell costimulation. In order to circumvent the escape of some tumor cells under T-cell pressure, polyvalent vaccination strategies should be developed. This goal can be achieved by either pulsing respective transfecting DC with tumor cell lysates, RNA or DNA libraries, or a pool of peptide antigens. Careful monitoring of the elicited T-cell response and quality assurance (GMP and GCP) are mandatory to establish a rationale for specific immunotherapy against RCC and to bring it from the bench to the bedside.
- Published
- 2002
47. [T-cell therapy in oncology. Therapeutic vaccination, allogeneic blood stem cell transplantation and adoptive T-cell transfer]
- Author
-
J, Kuball, H-G, Derigs, and T, Wölfel
- Subjects
Neoplasms ,T-Lymphocytes ,Hematopoietic Stem Cell Transplantation ,Animals ,Humans ,Transplantation, Homologous ,Cancer Vaccines ,Immunotherapy, Adoptive - Published
- 2002
48. [Cell therapy of hematological neoplasms]
- Author
-
H J, Kolb and E, Mischak-Weissinger
- Subjects
Leukemia ,Transplantation Conditioning ,Lymphoma ,HLA Antigens ,Hematologic Neoplasms ,Hematopoietic Stem Cell Transplantation ,Humans ,Genetic Therapy ,Immunotherapy, Adoptive ,Bone Marrow Transplantation - Published
- 2001
49. [Antibody and immunotherapy in oncology. Which patients benefit?]
- Author
-
G, Schlimok
- Subjects
Treatment Outcome ,Antibody Specificity ,Neoplasms ,Antibodies, Monoclonal ,Humans ,Immunotherapy, Active ,Immunotherapy ,Immunotherapy, Adoptive - Abstract
Among the various immunotherapeutic approaches, monoclonal antibodies in particular have acquired clinical relevance. In patients with non-Hodgkin's lymphoma and in breast cancer it has been shown that antibody therapy and chemotherapy are not rival forms of treatment, but that the best clinical response is obtained with a combination of the two. Cellular immunotherapy with lymphokine-activated killer cells, tumor-infiltrating lymphocytes, allogeneic T-cells and dendritic cells is still in the experimental stage. This is also true of therapeutic vaccination with tumor antigens. The best clinical results of treatment with antibodies or immunotherapy may be expected in patients with minimal residual disease.
- Published
- 2001
50. [HCC: Adoptive immunotherapy promises success]
- Author
-
S, Krome
- Subjects
Postoperative Care ,Carcinoma, Hepatocellular ,Time Factors ,Liver Neoplasms ,Preoperative Care ,Humans ,Neoplasm Recurrence, Local ,Combined Modality Therapy ,Immunotherapy, Adoptive ,Follow-Up Studies - Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.