Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2018

Open Access 01.12.2018 | Review

Cytokine release syndrome: grading, modeling, and new therapy

verfasst von: Delong Liu, Juanjuan Zhao

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2018

Abstract

Genetically modified T cells that express a chimeric antigen receptor (CAR) are opening a new frontier in cancer immunotherapy. CAR T cells currently are in clinical trials for many cancer types. Cytokine release syndrome (CRS) and neurotoxicities (CAR-related encephalopathy syndrome, CRES) are major adverse events limiting wide deployment of the CAR T cell treatment. Major efforts are ongoing to characterize the pathogenesis and etiology of CRS and CRES. Mouse models have been established to facilitate the study of pathogenesis of the major toxicities of CAR T cells. Myeloid cells including macrophages and monocytes, not the CAR T cells, were found to be the major cells mediating CRS and CRES by releasing IL-1 and IL-6 among other cytokines. Blocking IL-1 or depletion of monocytes abolished both CRS and CRES, whereas IL-6 blocker can ameliorate CRS but not CRES. Therefore, both IL-1 and IL-6 are major cytokines for CRS, though IL-1 is responsible for CRES. It was also demonstrated in the mouse models that blocking CRS does not interfere with the CAR T cell antitumor functions. We summarized new developments in the grading, modeling, and possible new therapeutic approaches for CRS and CRES in this review.
Abkürzungen
CAR
Chimeric antigen receptor
CRES
CAR-related encephalopathy syndrome
CRS
Cytokine release syndrome

Background

Genetically modified T cells that express a chimeric antigen receptor (CAR) are opening a new frontier in cancer immunotherapy [16]. Clinical trials of CAR T cells have been reported for many cancer types throughout the world [720]. Two types of CD19-directed CAR T cells, tisagenlecleucel and axicabtagene ciloleucel, have been approved for treatment of refractory/relapsed (R/R) non-Hodgkin lymphoma, whereas tisagenlecleucel was also approved for treatment of R/R acute lymphoid leukemia [15, 2123].
Currently, the structure of a CAR contains an antigen-recognition domain, a transmembrane domain, a costimulatory segment, and a signaling domain [1, 2426]. A single-chain fragment of variable region (scFv) replaces the conventional T cell antigen recognition domain. Therefore, the scFv confers the specificity of the engineered CAR T cells. More sophisticated CARs are being engineered, leading to more flexibility and controllability [2729].
CAR T cells currently are in clinical trials for both hematological malignancies and solid tumors [11, 3039]. Cytokine release syndrome (CRS) and neurotoxicities (CAR-related encephalopathy syndrome, CRES) are major adverse events limiting wide deployment of the CAR T cell treatment. Major efforts are ongoing to characterize the pathogenesis and etiology of CRS and CRES [40]. Mouse models of CRS have been established to facilitate the study of pathogenesis of the major toxicities of CAR T cells in animals. We summarized new developments in the grading, modeling, and therapeutic approaches for CRS and neurotoxicities in this review.

CRS grading

CRS has characteristic clinical presentations, manifested typically with high fever, hypotension, hypoxia, and respiratory distress [4144]. Organ dysfunctions such as liver transaminitis and renal insufficiency can occur. CRS toxicities occur as frequently as 90%, half of which is severe. Severe CRS complications are life-threatening and frequently require ICU care with vasopressors and/or ventilation support. The severities of CRS and neurotoxicities are directly related to the disease burden of ALL and the magnitude of CAR T cell expansion [22, 45, 46].
Several CRS grading systems have been proposed (Table 1) [4144]. The CTCAE grading of CRS was initially developed for CRS related to blinatumomab infusion. The Lee and Neelapu grading systems allowed low-dose vasopressors and low requirement for oxygen (< 40%) as grade 2, whereas the Penn grading scale placed those to grade 3 when IV fluid resuscitation, any vasopressors, and/or oxygen is required. Clinically, it is more practical to place those who require any vasopressors to grade 3 (severe) since ICU care is required in most hospitals in such situation, whereas those who only require low-flow oxygen (mild hypoxia) and fluid resuscitation (mild hypotension) belong to grade 2. The different CRS grading system may have accounted for the reported differences in the rate of severe grades 3–4 CRS in different clinical trials [22, 47].
Table 1
CRS grading scales: Penn grading scale, CTCAE v4.0, 2014 Lee et al. scale, and MDACC grading
 
Penn grading scale
CTCAE v4.0
2014 Lee et al.
MDACC
Grade 1
Mild reaction: treated with supportive care such as antipyretics and antiemetics
Mild reaction; infusion interruption not indicated; intervention not indicated
Symptoms are not life-threatening and require symptomatic treatment only, e.g., fever, nausea, fatigue, headache, myalgias, and malaise
Temperature ≥ 38 °C (fever) or grade 1 organ toxicity
Grade 2
Moderate reaction: some signs of organ dysfunction (e.g., grade 2 creatinine or grade 3 LFTs) related to CRS and not attributable to any other condition. Hospitalization for management of CRS-related symptoms, including fevers with associated neutropenia and need for IV therapies (not including fluid resuscitation for hypotension)
Therapy or infusion interruption indicated but responds promptly to symptomatic treatment (e.g., antihistamines, NSAIDs, narcotics, IV fluids); prophylactic medications indicated for ≤ 24 h
Symptoms require and respond to moderate intervention. Oxygen requirement < 40% or hypotension responsive to fluids or low-dose pressors or grade 2 organ toxicity
Systolic blood pressure < 90 mmHg (hypotension) but responds to IV fluids or low-dose vasopressors or needing oxygen(FiO2 < 40%) for SaO2 > 90% (hypoxia) or grade 2 organ toxicity
Grade 3
More severe reaction: hospitalization required for management of symptoms related to organ dysfunction, including grade 4 LFTs or grade 3 creatinine related to CRS and not attributable to any other conditions; this excludes management of fever or myalgias; includes hypotension treated with IV fluids (defined as multiple fluid boluses for blood pressure support) or low-dose vasopressors, coagulopathy requiring fresh frozen plasma or cryoprecipitate or fibrinogen concentrate, and hypoxia requiring supplemental oxygen (nasal cannula oxygen, high-flow oxygen, CPAP, or BiPAP). Patients admitted for management of suspected infection due to fevers and/or neutropenia may have grade 2 CRS
Prolonged reaction (e.g., not rapidly responsive to symptomatic medication and/or brief interruption of infusion); recurrence of symptoms following initial improvement; indicated for clinical sequelae (e.g., renal impairment, pulmonary infiltrates)
Symptoms require and respond to aggressive intervention. Oxygen requirement ≥ 40% or hypotension requiring high-dose or multiple pressors or grade 3 organ toxicity or grade 4 transaminitis
Systolic blood pressure < 90 mmHg (hypotension) and needs high-dose or multiple vasopressors or needing oxygen(FiO2 ≥ 40%) for SaO2 > 90% (hypoxia) or grade 3 organ toxicity or grade 4 transaminitis
Grade 4
Life-threatening complications such as hypotension requiring high-dose vasopressors and a hypoxia requiring mechanical ventilation
Life-threatening consequences; pressor or ventilator support indicated
Life-threatening symptoms. Requirements for ventilator support or grade 4 oxygen toxicity (excluding transaminitis)
Life-threatening hypotension or needing ventilator support or grade 4 oxygen toxicity (excluding transaminitis)
BiPAP bilevel positive airway pressure, CPAP continuous positive airway pressure therapy, CRS cytokine release syndrome, CTCAE Common Terminology Criteria for Adverse Events, IV intravenous, LFT liver function test, NSAID nonsteroidal anti-inflammatory drug, FiO2 fraction of inspired oxygen, SaO2 arterial oxygen saturation
aSee specific definition of high-dose vasopressors [41]
Cytokine storm appears to be a distinct entity [41], even though clinical manifestations are similar to those of CRS. Cytokine storm appears to be mainly a result of non-specific T cell activation and occurs typically early in the course, shortly after CAR T cell infusion. The major cytokines in cytokine storm are TNF and IFN gamma [41]. Steroids are usually the first choice of therapy. CRS is usually the direct consequence of CAR T cell activation upon engagement of CAR with specific antigens. Therefore, CRS onset can vary in different diseases as well as with different CARs. For CTL019 clinical trials, the CRS onset timing ranged from 1 to 71 days [41], though most cases occurred between 1 and 14 days. The CRS onset timing in these CTL019 (tisagenlecleucel) trials varies according to the severities, with grades 1–3 CRS onset between a median of 8 and 9 days, whereas grade 4 CRS onset occurred at a median of 1 day (range 1–8 days) [8, 41, 48]. The median duration of CRS was reported to be between 5 and 12 days (range 1–23 days) [41]. CRS onset appears to be earlier in lymphoma trials. The key cytokine mediator is IL-6. Tocilizumab, the IL-6 receptor antibody, is the first-line of therapy.
Compartmental CRS (C-CRS) has been reported in a patient with advanced ovarian cancer treated with mesothelin-targeted CAR T cells [49]. In this case, elevation of IL-6 and accumulation of CAR T cells were seen in the pleural fluid. Tocilizumab therapy was effective. C-CRS was also reported in the brain, where IL-6, IFN gamma, and CAR T cells were significantly higher in the CSF than in the blood [50]. High-dose methylprednisolone successfully alleviated the cerebral CRS.

CRS modeling

Establishment of CRS mouse models

CRS remains as a major life-threatening complication with complex pathogenesis. To further study and to better therapy of CRS, two mouse models have been reported [51, 52]. One model used SCID-beige mouse [51]. Raji tumor cells were injected intraperitoneally and allowed to grow 3 weeks. The tumor cells formed vascularized solid masses. Thirty million human 1928z CAR T cells were used to target the B leukemia cells and solicit CRS, which occurred 2–3 days after CAR T infusion. The mouse model recapitulated the constellation of CRS symptoms and signs. Serum cytokines of human origin, IL-2, IL-3, IL-6, IFN gamma, and GMCSF were elevated in CRS.
The other mouse model used humanized NSG mice which produce human cytokines of IL-3, stem cell factor, and GM-CSF to support and enhance hematopoiesis from human stem cells [52]. Patient-derived ALL cell line, ALL-CM, was used as targets for CD19.BBz and CD19.28z CAR T cells. After the CAR T cell challenge, a violent systemic inflammatory syndrome occurred, recapitulating severe human CRS.

Macrophages and monocytes are mediators of CRS

Using the SCID-beige mouse model, Sadelain’s group chased after the source of IL-6 released during CRS. The source was traced to macrophages. It was discovered through RNA-seq analysis that the main source of IL-6 came from the macrophages and monocytes. Production of major inflammatory cytokines was confined to the site of CAR T cell-tumor tissue colocalization in the peritoneum.
In the humanized NSG model, monocytes were found to be the main source of CRS cytokines. Monocyte depletion essentially abolished CRS and protected mice from CRS-induced lethality.
These two separate mouse model systems independently confirmed that macrophages and monocytes are the direct mediators of CRS, not CAR T cells.

IL-1 is the primary cytokine for CRS and CRES

It is well known that IL-6 is elevated in patients with CRS, and IL-6 receptor antagonist, tocilizumab, is an effective therapy for CRS. This was reproducible in the mouse models. However, it was demonstrated in both models that IL-1 is the primary cytokine responsible for both CRS and CRES. Blocking IL-1 receptor with anakinra effectively abolished CRS as well as neurotoxicities, whereas blockage of IL-6 with tocilizumab abrogated only CRS, not neurotoxicities. It is known that macrophages/monocytes produce IL-1 upon activation, and IL-1 in turn can elicit production of IL-6 by the macrophages/monocytes. Sadelain’s group further demonstrated that by engineering CAR T cells to constitutively express an IL-1 receptor antagonist, CRS was prevented [51].

Hypotension associated with CRS is induced by nitrogen oxide (NO)

It was further demonstrated in the SCID-beige mouse model that NO synthetase (NOS) was induced by IL-1 and IL-6 during CRS. As a result, NO production by macrophages was increased, leading to hypotension which is a major life-threatening complication of CAR T-induced severe CRS. Using NOS inhibitors, L-NIL or 1400W, the investigators were able to alleviate the systemic toxicities and reduce mortality secondary to severe CRS. These findings provided new evidence for CRS pathophysiology and a new means to possibly prevent and effectively treat severe CRS. Inhibition of NOS activity may significantly reduce CRS severity and possibly eliminate or reduce requirements for ICU care of CRS toxicity.
NOS was downregulated by blocking either IL-1 or IL-6, leading to reduced mortality in the mouse model. This confirms that upregulation of NOS is a unifying mechanism for both IL-1- and IL-6-induced severe CRS. However, simultaneous blockade of IL-1 and IL-6 does not further reduce the fraction of NOS+ macrophages [51]. This implies that clinically only blocking one of the two cytokines is needed to abrogate CRS.

Blocking CRS does not reduce CAR T functions

There have been concerns that treatment for CRS could reduce the activity of CAR T cells. Therefore prophylaxis for CRS by steroids and IL-6 blockage has not been encouraged currently in the clinical practice.
This question was specifically addressed in the mouse models. Using the IL-1R antagonist producing CAR T cells, it was demonstrated that blocking CRS did not reduce the 1928z CAR T cell antitumor efficacy. Therefore, preventing CRS does not affect CAR T cell functions. The preservation of antitumor efficacy was also demonstrated where similar leukemia clearance by CAR T cells in the humanized HuSGM3 mice treated with either tocilizumab or anakinra [52] occurred. These observations suggest that clinically it may be preferred to apply tocilizumab as early as prophylaxis for CRS so that morbidity and mortality from CART-related toxicities (CARTox) can be significantly reduced.

IL-1 mediates both CRS and CRES

In the HuSGM3 mouse model, occurrence of lethal neurological syndrome with paralysis and seizures was unexpectedly observed in the mice that received either vehicle or tocilizumab. Meningeal thickening together with human macrophage infiltration in these mice was demonstrated. It was further shown that the meningeal thickening was prevented by administration of anakinra, but not by tocilizumab, even though CRS was effectively abolished by both agents. It was concluded that only anakinra prophylaxis prevented both CRS and neurotoxicity and increased mice survival. This study established that both IL-1 and IL-6 are causal cytokines for CRS, though IL-1 is responsible for severe neurotoxicities (CRES).

New approach for clinical management of CRS and CRES

Currently, for early-onset CRES, such as confusion, hallucination, aphasia, and seizure, steroids are generally used first. For CRS characterized by high fever, hypotension, and hypoxia, tocilizumab is infused over an hour. The dosage is calculated according to patients’ weight. For patients’ weight less than 30 kg, the dosage is 12 mg/kg, whereas the dosage is 8 mg/kg when the weight is more than 30 kg (for official recommendations, please refer to the full prescribing information at https://​www.​gene.​com/​download/​pdf/​actemra_​prescribing.​pdf). The treatment can be repeated when clinically necessary, up to three more doses, at least 8 hours apart.
With the new discovery of IL-1 being a major cytokine for both CRS and CRES from the mouse models, it is reasonable to consider anakinra, an IL-1R antagonist, for CAR T-induced toxicities (CARTox), namely, CRS and CRES. Since blocking CARTox has no adverse effect on CAR T antitumor efficacy, it is reasonable to use anakinra or tocilizumab for CRS prophylaxis. Anakinra may be the preferred agent for prophylaxis since it is effective to prevent both CRS and CRES. Tocilizumab may be used to prevent CRS but not effective for prophylaxis of neurotoxicities. In the SCID-beige mouse model, NOS inhibitors were shown to reduce systemic toxicities of CRS. Therefore, these agents may have potential for the management of CARTox and reduce the use of vasopressors. However, these were reports from animal models. Clinical trials are needed to confirm the effect of anakinra for both CRS and CRES as well as NOS inhibitors. Once it is confirmed clinically, a significant reduction in CARTox is expected in the near future. Clinical confirmation of these significant findings from mouse models is urgently needed.
Currently, tocilizumab is being studied in a clinical trial for prophylaxis of CRS (Table 2). Since it has been observed that CRS severity is directly related to leukemia burden in ALL and severe CRS occurs earlier, timing of tocilizumab therapy is being studied in a two-cohort open-label pilot study (Table 2). Tocilizumab is scheduled to start earlier in high-leukemia-burden patients.
Table 2
Clinical trials of tocilizumab for cytokine release syndrome
NCT no.
Trials
Conditions
Interventions
Locations
03533101
Tocilizumab for Cytokine Release Syndrome Prophylaxis in Haploidentical Transplantation
•Cytokine release syndrome
•Stem cell transplant complications
•Drug: tocilizumab
•Hospital Universitario Dr. Jose E Gonzalez UANL, Monterrey, Nuevo Leon, Mexico
03275493
Humanized CD19 CAR-T Cells With CRS Suppression Technology for r/r CD19+ Acute Lymphoblastic Leukemia
•Acute lymphoblastic leukemia
•CD19 positive
•Relapse
•Refractory
•Biological: humanized CD19 CAR-T cells
•Biological: humanized CD19 CAR-T cells with CRS suppression technology
•The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
02906371
Study of the Tocilizumab Optimization Timing for CART19 Associated Cytokine Release Syndrome
•Lymphoblastic leukemia, acute, childhood
•Drug: tocilizumab
•Biological: CART 19
•Children’s Hospital of Philadelphia, Philadelphia, PA, USA

Conclusions

Currently, there are several CRS scoring models (Table 1). These were used for different clinical trials. It is ideal to have a consensus and to unify the scoring systems. There is no CRES-specific scoring system available yet. In addition to prophylaxis, new CAR design is needed to better control and adjust the CAR T cell engagement of target cancer cells. SUPRA CAR design offers a flexible, switchable, and more adjustable construct as demonstrated in the mouse model [53]. This SUPRA CAR design may bring more control in CRS. Clinical trial of the SUPRA CAR T cells is clearly needed.
Blinatumomab is a bispecific T cell engager (BiTE) approved for therapy of acute lymphoblastic leukemia [5456]. Bispecific antibodies are associated with the similar profile of CRES and CRS seen in CAR T therapy, albeit at a lower incidence and with less severity [5759]. Since the pathophysiology of the CRS and CRES incited by blinatumomab is similar to CARTox, and IL-6 blockade is equally effective for CRS, it is possible that macrophage-produced IL-1 plays a major role in the toxicities [42]. Therefore, anakinra may be a therapeutic and prophylactic option for the CRS and CRES associated with blinatumomab. Clinical trial of anakinra is indicated.
A variety of systemic inflammatory syndromes, such as acute respiratory distress syndrome (ARDS) and macrophage activation syndrome/hemophagocytic lymphohistiocytosis (HLH), have similar clinical manifestations with high fever, hypoxia, and hypotension. The findings from the CRS models appear to be provocative for implications of management for ARDS and HLH. Etanercept, a soluble TNFa receptor, has been used successfully for CRS therapy [42]. It is possible that IL-1 and/or IL-6 is also a major cytokine for the syndromes [6062]. Further studies on use of anakinra and/or tocilizumab in these syndromes would be indicated. A clinical trial of tocilizumab for HLH is underway (NCT02007239).

Funding

The study is partly supported by the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.

Availability of data and materials

The material supporting the conclusion of this review has been included within the article.
This is not applicable for this review.
This is not applicable for this review.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361–5.CrossRefPubMed June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361–5.CrossRefPubMed
4.
Zurück zum Zitat Sadelain M, Brentjens R, Riviere I, Park J. CD19 CAR therapy for acute lymphoblastic leukemia. Am Soc Clin Oncol Educ Book. 2015:e360–3.CrossRef Sadelain M, Brentjens R, Riviere I, Park J. CD19 CAR therapy for acute lymphoblastic leukemia. Am Soc Clin Oncol Educ Book. 2015:e360–3.CrossRef
7.
Zurück zum Zitat Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, Lacey SF, Melenhorst JJ, McGettigan SE, Cook DR, Zhang C, Xu J, Do P, Hulitt J, Kudchodkar SB, Cogdill AP, Gill S, Porter DL, Woyach JA, Long M, Johnson AJ, Maddocks K, Muthusamy N, Levine BL, June CH, Byrd JC, Maus MV. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016;127(9):1117–27.CrossRefPubMedCentralPubMed Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, Lacey SF, Melenhorst JJ, McGettigan SE, Cook DR, Zhang C, Xu J, Do P, Hulitt J, Kudchodkar SB, Cogdill AP, Gill S, Porter DL, Woyach JA, Long M, Johnson AJ, Maddocks K, Muthusamy N, Levine BL, June CH, Byrd JC, Maus MV. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016;127(9):1117–27.CrossRefPubMedCentralPubMed
8.
Zurück zum Zitat Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O'Connor RS, Hwang WT, Pequignot E, Ambrose DE, Zhang C, Wilcox N, Bedoya F, Dorfmeier C, Chen F, Tian L, Parakandi H, Gupta M, Young RM, Johnson FB, Kulikovskaya I, Liu L, Xu J, Kassim SH, Davis MM, Levine BL, Frey NV, Siegel DL, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018;24(5):563–71.CrossRefPubMedCentralPubMed Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O'Connor RS, Hwang WT, Pequignot E, Ambrose DE, Zhang C, Wilcox N, Bedoya F, Dorfmeier C, Chen F, Tian L, Parakandi H, Gupta M, Young RM, Johnson FB, Kulikovskaya I, Liu L, Xu J, Kassim SH, Davis MM, Levine BL, Frey NV, Siegel DL, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018;24(5):563–71.CrossRefPubMedCentralPubMed
10.
Zurück zum Zitat Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL, Grupp SA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17.CrossRefPubMedCentralPubMed Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL, Grupp SA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17.CrossRefPubMedCentralPubMed
11.
Zurück zum Zitat Neelapu SS, Locke FL, Bartlett NL, Lekakis L, Miklos D, Jacobson CA, Braunschweig I, Oluwole O, Siddiqi T, Lin Y, Timmerman J, Stiff PJ, Friedberg J, Flinn I, Goy A, Smith M, Deol A, Farooq U, McSweeney P, Munoz J, Avivi I, Castro JE, Westin JR, Chavez JC, Ghobadi A, Komanduri KV, Levy R, Jacobsen ED, Reagan P, Bot A, et al. Kte-C19 (anti-CD19 CAR T cells) induces complete remissions in patients with refractory diffuse large B-cell lymphoma (DLBCL): results from the pivotal phase 2 Zuma-1. Blood. 2016;128(22):LBA-6-LBA-6. Neelapu SS, Locke FL, Bartlett NL, Lekakis L, Miklos D, Jacobson CA, Braunschweig I, Oluwole O, Siddiqi T, Lin Y, Timmerman J, Stiff PJ, Friedberg J, Flinn I, Goy A, Smith M, Deol A, Farooq U, McSweeney P, Munoz J, Avivi I, Castro JE, Westin JR, Chavez JC, Ghobadi A, Komanduri KV, Levy R, Jacobsen ED, Reagan P, Bot A, et al. Kte-C19 (anti-CD19 CAR T cells) induces complete remissions in patients with refractory diffuse large B-cell lymphoma (DLBCL): results from the pivotal phase 2 Zuma-1. Blood. 2016;128(22):LBA-6-LBA-6.
12.
Zurück zum Zitat Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33.CrossRefPubMedCentralPubMed Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33.CrossRefPubMedCentralPubMed
13.
Zurück zum Zitat Qin L, Lai Y, Zhao R, Wei X, Weng J, Lai P, Li B, Lin S, Wang S, Wu Q, Liang Q, Li Y, Zhang X, Wu Y, Liu P, Yao Y, Pei D, Du X, Li P. Incorporation of a hinge domain improves the expansion of chimeric antigen receptor T cells. J Hematol Oncol. 2017;10(1):68.CrossRefPubMedCentralPubMed Qin L, Lai Y, Zhao R, Wei X, Weng J, Lai P, Li B, Lin S, Wang S, Wu Q, Liang Q, Li Y, Zhang X, Wu Y, Liu P, Yao Y, Pei D, Du X, Li P. Incorporation of a hinge domain improves the expansion of chimeric antigen receptor T cells. J Hematol Oncol. 2017;10(1):68.CrossRefPubMedCentralPubMed
14.
Zurück zum Zitat Qin L, Zhao R, Li P. Incorporation of functional elements enhances the antitumor capacity of CAR T cells. Experimental Hematology & Oncology. 2017;6(1):28.CrossRef Qin L, Zhao R, Li P. Incorporation of functional elements enhances the antitumor capacity of CAR T cells. Experimental Hematology & Oncology. 2017;6(1):28.CrossRef
15.
16.
Zurück zum Zitat Song D-G, Ye Q, Poussin M, Chacon JA, Figini M, Powell DJ. Effective adoptive immunotherapy of triple-negative breast cancer by folate receptor-alpha redirected CAR T cells is influenced by surface antigen expression level. J Hematol Oncol. 2016;9(1):56.CrossRefPubMedCentralPubMed Song D-G, Ye Q, Poussin M, Chacon JA, Figini M, Powell DJ. Effective adoptive immunotherapy of triple-negative breast cancer by folate receptor-alpha redirected CAR T cells is influenced by surface antigen expression level. J Hematol Oncol. 2016;9(1):56.CrossRefPubMedCentralPubMed
18.
Zurück zum Zitat Wei G, Ding L, Wang J, Hu Y, Huang H. Advances of CD19-directed chimeric antigen receptor-modified T cells in refractory/relapsed acute lymphoblastic leukemia. Experimental Hematology & Oncology. 2017;6(1):10.CrossRef Wei G, Ding L, Wang J, Hu Y, Huang H. Advances of CD19-directed chimeric antigen receptor-modified T cells in refractory/relapsed acute lymphoblastic leukemia. Experimental Hematology & Oncology. 2017;6(1):10.CrossRef
19.
Zurück zum Zitat Yu S, Li A, Liu Q, Li T, Yuan X, Han X, Wu K. Chimeric antigen receptor T cells: a novel therapy for solid tumors. J Hematol Oncol. 2017;10(1):78.CrossRefPubMedCentralPubMed Yu S, Li A, Liu Q, Li T, Yuan X, Han X, Wu K. Chimeric antigen receptor T cells: a novel therapy for solid tumors. J Hematol Oncol. 2017;10(1):78.CrossRefPubMedCentralPubMed
20.
Zurück zum Zitat Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, Ostberg JR, Blanchard MS, Kilpatrick J, Simpson J, Kurien A, Priceman SJ, Wang X, Harshbarger TL, D’Apuzzo M, Ressler JA, Jensen MC, Barish ME, Chen M, Portnow J, Forman SJ, Badie B. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375(26):2561–9.CrossRefPubMedCentralPubMed Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, Ostberg JR, Blanchard MS, Kilpatrick J, Simpson J, Kurien A, Priceman SJ, Wang X, Harshbarger TL, D’Apuzzo M, Ressler JA, Jensen MC, Barish ME, Chen M, Portnow J, Forman SJ, Badie B. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375(26):2561–9.CrossRefPubMedCentralPubMed
21.
Zurück zum Zitat Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, Ghobadi A, Budde LE, Bot A, Rossi JM, Jiang Y, Xue AX, Elias M, Aycock J, Wiezorek J, Go WY. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol Ther. 2017;25(1):285–95.CrossRefPubMedCentralPubMed Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, Ghobadi A, Budde LE, Bot A, Rossi JM, Jiang Y, Xue AX, Elias M, Aycock J, Wiezorek J, Go WY. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol Ther. 2017;25(1):285–95.CrossRefPubMedCentralPubMed
22.
Zurück zum Zitat Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, Qayed M, De Moerloose B, Hiramatsu H, Schlis K, Davis KL, Martin PL, Nemecek ER, Yanik GA, Peters C, Baruchel A, Boissel N, Mechinaud F, Balduzzi A, Krueger J, June CH, Levine BL, Wood P, Taran T, Leung M, Mueller KT, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–48.CrossRefPubMedCentralPubMed Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, Qayed M, De Moerloose B, Hiramatsu H, Schlis K, Davis KL, Martin PL, Nemecek ER, Yanik GA, Peters C, Baruchel A, Boissel N, Mechinaud F, Balduzzi A, Krueger J, June CH, Levine BL, Wood P, Taran T, Leung M, Mueller KT, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–48.CrossRefPubMedCentralPubMed
25.
26.
Zurück zum Zitat Zhang L, Tai Y-T, Ho MZG, Qiu L, Anderson KC. Interferon-alpha-based immunotherapies in the treatment of B cell-derived hematologic neoplasms in today’s treat-to-target era. Experimental Hematology & Oncology. 2017;6(1):20.CrossRef Zhang L, Tai Y-T, Ho MZG, Qiu L, Anderson KC. Interferon-alpha-based immunotherapies in the treatment of B cell-derived hematologic neoplasms in today’s treat-to-target era. Experimental Hematology & Oncology. 2017;6(1):20.CrossRef
27.
Zurück zum Zitat Casucci M, Falcone L, Camisa B, Norelli M, Porcellini S, Stornaiuolo A, Ciceri F, Traversari C, Bordignon C, Bonini C, Bondanza A. Extracellular NGFR spacers allow efficient tracking and enrichment of fully functional CAR-T cells co-expressing a suicide gene. Front Immunol. 2018;9:507.CrossRefPubMedCentralPubMed Casucci M, Falcone L, Camisa B, Norelli M, Porcellini S, Stornaiuolo A, Ciceri F, Traversari C, Bordignon C, Bonini C, Bondanza A. Extracellular NGFR spacers allow efficient tracking and enrichment of fully functional CAR-T cells co-expressing a suicide gene. Front Immunol. 2018;9:507.CrossRefPubMedCentralPubMed
28.
Zurück zum Zitat Fan M, Li M, Gao L, Geng S, Wang J, Wang Y, Yan Z, Yu L. Chimeric antigen receptors for adoptive T cell therapy in acute myeloid leukemia. J Hematol Oncol. 2017;10(1):151.CrossRefPubMedCentralPubMed Fan M, Li M, Gao L, Geng S, Wang J, Wang Y, Yan Z, Yu L. Chimeric antigen receptors for adoptive T cell therapy in acute myeloid leukemia. J Hematol Oncol. 2017;10(1):151.CrossRefPubMedCentralPubMed
29.
Zurück zum Zitat Cho JH, Collins JJ, Wong WW. Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell. 2018;173(6):1426–38.PubMedPubMedCentral Cho JH, Collins JJ, Wong WW. Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell. 2018;173(6):1426–38.PubMedPubMedCentral
30.
Zurück zum Zitat O'Rourke DM, Nasrallah M, Morrissette JJ, Melenhorst JJ, Lacey SF, Mansfield K, Martinez-Lage M, Desai AS, Brem S, Maloney E, Mohan S, Wang S, Verma G, Navenot J-M, Shen A, Zheng Z, Levine B, Okada H, June CH, Maus MV. Pilot study of T cells redirected to EGFRvIII with a chimeric antigen receptor in patients with EGFRvIII+ glioblastoma. J Clin Oncol. 2016;34(15_suppl):2067.CrossRef O'Rourke DM, Nasrallah M, Morrissette JJ, Melenhorst JJ, Lacey SF, Mansfield K, Martinez-Lage M, Desai AS, Brem S, Maloney E, Mohan S, Wang S, Verma G, Navenot J-M, Shen A, Zheng Z, Levine B, Okada H, June CH, Maus MV. Pilot study of T cells redirected to EGFRvIII with a chimeric antigen receptor in patients with EGFRvIII+ glioblastoma. J Clin Oncol. 2016;34(15_suppl):2067.CrossRef
31.
Zurück zum Zitat Shibahara I, Saito R, Zhang R, Chonan M, Shoji T, Kanamori M, Sonoda Y, Kumabe T, Kanehira M, Kikuchi T, So T, Watanabe T, Takahashi H, Iwabuchi E, Tanaka Y, Shibahara Y, Sasano H, Ishii N, Tominaga T. OX40 ligand expressed in glioblastoma modulates adaptive immunity depending on the microenvironment: a clue for successful immunotherapy. Mol Cancer. 2015;14:41.CrossRefPubMedCentralPubMed Shibahara I, Saito R, Zhang R, Chonan M, Shoji T, Kanamori M, Sonoda Y, Kumabe T, Kanehira M, Kikuchi T, So T, Watanabe T, Takahashi H, Iwabuchi E, Tanaka Y, Shibahara Y, Sasano H, Ishii N, Tominaga T. OX40 ligand expressed in glioblastoma modulates adaptive immunity depending on the microenvironment: a clue for successful immunotherapy. Mol Cancer. 2015;14:41.CrossRefPubMedCentralPubMed
32.
Zurück zum Zitat Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, Hawkins R, Chaney C, Cherian S, Chen X, Soma L, Wood B, Li D, Heimfeld S, Riddell SR, Maloney DG. Immunotherapy of non-Hodgkin's lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116.CrossRefPubMedCentralPubMed Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, Hawkins R, Chaney C, Cherian S, Chen X, Soma L, Wood B, Li D, Heimfeld S, Riddell SR, Maloney DG. Immunotherapy of non-Hodgkin's lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116.CrossRefPubMedCentralPubMed
34.
Zurück zum Zitat Wang CM, Wu ZQ, Wang Y, Guo YL, Dai HR, Wang XH, Li X, Zhang YJ, Zhang WY, Chen MX, Zhang Y, Feng KC, Liu Y, Li SX, Yang QM, Han WD. Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial. Clin Cancer Res. 2017;23(5):1156–66.CrossRefPubMed Wang CM, Wu ZQ, Wang Y, Guo YL, Dai HR, Wang XH, Li X, Zhang YJ, Zhang WY, Chen MX, Zhang Y, Feng KC, Liu Y, Li SX, Yang QM, Han WD. Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial. Clin Cancer Res. 2017;23(5):1156–66.CrossRefPubMed
35.
Zurück zum Zitat Wang QS, Wang Y, Lv HY, Han QW, Fan H, Guo B, Wang LL, Han WD. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther. 2015;23(1):184–91.CrossRefPubMed Wang QS, Wang Y, Lv HY, Han QW, Fan H, Guo B, Wang LL, Han WD. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther. 2015;23(1):184–91.CrossRefPubMed
36.
Zurück zum Zitat Wang Y, Chen M, Wu Z, Tong C, Huang J, Lv H, Dai H, Feng K, Guo Y, Liu Y, Yang Q, Han W. CD133-redirected chimeric antigen receptor engineered autologous T-cell treatment in patients with advanced and metastatic malignancies. J Clin Oncol. 2017;35(15_suppl):3042.CrossRef Wang Y, Chen M, Wu Z, Tong C, Huang J, Lv H, Dai H, Feng K, Guo Y, Liu Y, Yang Q, Han W. CD133-redirected chimeric antigen receptor engineered autologous T-cell treatment in patients with advanced and metastatic malignancies. J Clin Oncol. 2017;35(15_suppl):3042.CrossRef
37.
Zurück zum Zitat Zhai B, Shi D, Gao H, Qi X, Jiang H, Zhang Y, Chi J, Ruan H, Wang H, Ru QC, Li Z. A phase I study of anti-GPC3 chimeric antigen receptor modified T cells (GPC3 CAR-T) in Chinese patients with refractory or relapsed GPC3+ hepatocellular carcinoma (r/r GPC3+ HCC). Journal of Clinical Oncology. 2017;35(15_suppl):3049.CrossRef Zhai B, Shi D, Gao H, Qi X, Jiang H, Zhang Y, Chi J, Ruan H, Wang H, Ru QC, Li Z. A phase I study of anti-GPC3 chimeric antigen receptor modified T cells (GPC3 CAR-T) in Chinese patients with refractory or relapsed GPC3+ hepatocellular carcinoma (r/r GPC3+ HCC). Journal of Clinical Oncology. 2017;35(15_suppl):3049.CrossRef
38.
Zurück zum Zitat Hegde M, Wakefield A, Brawley VS, Grada Z, Byrd TT, Chow KK, Krebs SS, Heslop HE, Gottschalk SM, Yvon E, Ahmed N. Genetic modification of T cells with a novel bispecific chimeric antigen receptor to enhance the control of high-grade glioma (HGG). J Clin Oncol. 2014;32(15_suppl):10027. Hegde M, Wakefield A, Brawley VS, Grada Z, Byrd TT, Chow KK, Krebs SS, Heslop HE, Gottschalk SM, Yvon E, Ahmed N. Genetic modification of T cells with a novel bispecific chimeric antigen receptor to enhance the control of high-grade glioma (HGG). J Clin Oncol. 2014;32(15_suppl):10027.
39.
Zurück zum Zitat Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, Lin AA, Schlom J, June CH, Sherwin SA. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.CrossRefPubMedCentralPubMed Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, Lin AA, Schlom J, June CH, Sherwin SA. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22.CrossRefPubMedCentralPubMed
40.
41.
Zurück zum Zitat Porter D, Frey N, Wood PA, Weng Y, Grupp SA. Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel. J Hematol Oncol. 2018;11(1):35.CrossRefPubMedCentralPubMed Porter D, Frey N, Wood PA, Weng Y, Grupp SA. Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel. J Hematol Oncol. 2018;11(1):35.CrossRefPubMedCentralPubMed
42.
Zurück zum Zitat Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA, Mackall CL. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–95.CrossRefPubMedCentralPubMed Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA, Mackall CL. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–95.CrossRefPubMedCentralPubMed
43.
Zurück zum Zitat Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, Westin J, Gulbis AM, Loghin ME, de Groot JF, Adkins S, Davis SE, Rezvani K, Hwu P, Shpall EJ. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62.CrossRefPubMed Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, Westin J, Gulbis AM, Loghin ME, de Groot JF, Adkins S, Davis SE, Rezvani K, Hwu P, Shpall EJ. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62.CrossRefPubMed
44.
Zurück zum Zitat Neelapu SS, Tummala S, Kebriaei P, Wierda W, Locke FL, Lin Y, Jain N, Daver N, Gulbis AM, Adkins S, Rezvani K, Hwu P, Shpall EJ. Toxicity management after chimeric antigen receptor T cell therapy: one size does not fit 'ALL'. Nat Rev Clin Oncol. 2018;15(4):218.CrossRefPubMedCentralPubMed Neelapu SS, Tummala S, Kebriaei P, Wierda W, Locke FL, Lin Y, Jain N, Daver N, Gulbis AM, Adkins S, Rezvani K, Hwu P, Shpall EJ. Toxicity management after chimeric antigen receptor T cell therapy: one size does not fit 'ALL'. Nat Rev Clin Oncol. 2018;15(4):218.CrossRefPubMedCentralPubMed
45.
Zurück zum Zitat Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ, Sauter C, Wang Y, Santomasso B, Mead E, Roshal M, Maslak P, Davila M, Brentjens RJ, Sadelain M. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378:449–59.CrossRefPubMedCentralPubMed Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ, Sauter C, Wang Y, Santomasso B, Mead E, Roshal M, Maslak P, Davila M, Brentjens RJ, Sadelain M. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378:449–59.CrossRefPubMedCentralPubMed
46.
Zurück zum Zitat Santomasso BD, Park JH, Salloum D, Riviere I, Flynn J, Mead E, Halton E, Wang X, Senechal B, Purdon T, Cross JR, Liu H, Vachha B, Chen X, DeAngelis LM, Li D, Bernal Y, Gonen M, Wendel HG, Sadelain M, Brentjens RJ. Clinical and biologic correlates of neurotoxicity associated with CAR T cell therapy in patients with B-cell acute lymphoblastic leukemia (B-ALL). Cancer Discov. 2018;8(8):958–971.CrossRefPubMedCentralPubMed Santomasso BD, Park JH, Salloum D, Riviere I, Flynn J, Mead E, Halton E, Wang X, Senechal B, Purdon T, Cross JR, Liu H, Vachha B, Chen X, DeAngelis LM, Li D, Bernal Y, Gonen M, Wendel HG, Sadelain M, Brentjens RJ. Clinical and biologic correlates of neurotoxicity associated with CAR T cell therapy in patients with B-cell acute lymphoblastic leukemia (B-ALL). Cancer Discov. 2018;8(8):958–971.CrossRefPubMedCentralPubMed
47.
Zurück zum Zitat Zhang L-N, Song Y, Liu D. CD19 CAR-T cell therapy for relapsed/refractory acute lymphoblastic leukemia: factors affecting toxicities and long-term efficacies. Journal of Hematology & Oncology. 2018;11(1):41. Zhang L-N, Song Y, Liu D. CD19 CAR-T cell therapy for relapsed/refractory acute lymphoblastic leukemia: factors affecting toxicities and long-term efficacies. Journal of Hematology & Oncology. 2018;11(1):41.
48.
Zurück zum Zitat Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, Reich TJ, Cogdill AP, Morrissette JJD, DeNizio JE, Reddy S, Hwang Y, Gohil M, Kulikovskaya I, Nazimuddin F, Gupta M, Chen F, Everett JK, Alexander KA, Lin-Shiao E, Gee MH, Liu X, Young RM, Ambrose D, Wang Y, Xu J, Jordan MS, Marcucci KT, Levine BL, Garcia KC, Zhao Y, et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018;558(7709):307–12.CrossRefPubMedCentralPubMed Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, Reich TJ, Cogdill AP, Morrissette JJD, DeNizio JE, Reddy S, Hwang Y, Gohil M, Kulikovskaya I, Nazimuddin F, Gupta M, Chen F, Everett JK, Alexander KA, Lin-Shiao E, Gee MH, Liu X, Young RM, Ambrose D, Wang Y, Xu J, Jordan MS, Marcucci KT, Levine BL, Garcia KC, Zhao Y, et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018;558(7709):307–12.CrossRefPubMedCentralPubMed
49.
Zurück zum Zitat Tanyi JL, Stashwick C, Plesa G, Morgan MA, Porter D, Maus MV, June CH. Possible compartmental cytokine release syndrome in a patient with recurrent ovarian cancer after treatment with mesothelin-targeted CAR-T cells. J Immunother. 2017;40(3):104–7.CrossRefPubMed Tanyi JL, Stashwick C, Plesa G, Morgan MA, Porter D, Maus MV, June CH. Possible compartmental cytokine release syndrome in a patient with recurrent ovarian cancer after treatment with mesothelin-targeted CAR-T cells. J Immunother. 2017;40(3):104–7.CrossRefPubMed
50.
Zurück zum Zitat Hu Y, Sun J, Wu Z, Yu J, Cui Q, Pu C, Liang B, Luo Y, Shi J, Jin A, Xiao L, Huang H. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J Hematol Oncol. 2016;9(1):70.CrossRefPubMedCentralPubMed Hu Y, Sun J, Wu Z, Yu J, Cui Q, Pu C, Liang B, Luo Y, Shi J, Jin A, Xiao L, Huang H. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J Hematol Oncol. 2016;9(1):70.CrossRefPubMedCentralPubMed
51.
Zurück zum Zitat Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 2018;24(6):731–8.CrossRefPubMedCentralPubMed Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 2018;24(6):731–8.CrossRefPubMedCentralPubMed
52.
Zurück zum Zitat Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, Traversari C, Bordignon C, Ciceri F, Ostuni R, Bonini C, Casucci M, Bondanza A. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–48.CrossRefPubMed Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, Traversari C, Bordignon C, Ciceri F, Ostuni R, Bonini C, Casucci M, Bondanza A. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–48.CrossRefPubMed
53.
54.
Zurück zum Zitat Topp MS, Gokbuget N, Stein AS, Zugmaier G, O'Brien S, Bargou RC, Dombret H, Fielding AK, Heffner L, Larson RA, Neumann S, Foa R, Litzow M, Ribera JM, Rambaldi A, Schiller G, Bruggemann M, Horst HA, Holland C, Jia C, Maniar T, Huber B, Nagorsen D, Forman SJ, Kantarjian HM. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2014;16(1):57–66.CrossRefPubMed Topp MS, Gokbuget N, Stein AS, Zugmaier G, O'Brien S, Bargou RC, Dombret H, Fielding AK, Heffner L, Larson RA, Neumann S, Foa R, Litzow M, Ribera JM, Rambaldi A, Schiller G, Bruggemann M, Horst HA, Holland C, Jia C, Maniar T, Huber B, Nagorsen D, Forman SJ, Kantarjian HM. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2014;16(1):57–66.CrossRefPubMed
55.
Zurück zum Zitat Zugmaier G, Goekbuget N, Viardot A, Stelljes M, Neumann S, Horst HA, Reichle A, Marks R, Faul C, Brueggemann M, Holland C, Schmidt M, Mergen N, Goebeler M-E, Einsele H, Bargou RC, Topp MS. Long-term survival in adult patients with relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL) who achieved minimal residual disease (MRD) response following anti-CD19 BiTE® blinatumomab. Blood. 2014;124(21):2287. Zugmaier G, Goekbuget N, Viardot A, Stelljes M, Neumann S, Horst HA, Reichle A, Marks R, Faul C, Brueggemann M, Holland C, Schmidt M, Mergen N, Goebeler M-E, Einsele H, Bargou RC, Topp MS. Long-term survival in adult patients with relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL) who achieved minimal residual disease (MRD) response following anti-CD19 BiTE® blinatumomab. Blood. 2014;124(21):2287.
56.
Zurück zum Zitat Zugmaier G, Topp MS, Alekar S, Viardot A, Horst HA, Neumann S, Stelljes M, Bargou RC, Goebeler M, Wessiepe D, Degenhard E, Gokbuget N, Klinger M. Long-term follow-up of serum immunoglobulin levels in blinatumomab-treated patients with minimal residual disease-positive B-precursor acute lymphoblastic leukemia. Blood Cancer J. 2014;4:244.CrossRefPubMedCentralPubMed Zugmaier G, Topp MS, Alekar S, Viardot A, Horst HA, Neumann S, Stelljes M, Bargou RC, Goebeler M, Wessiepe D, Degenhard E, Gokbuget N, Klinger M. Long-term follow-up of serum immunoglobulin levels in blinatumomab-treated patients with minimal residual disease-positive B-precursor acute lymphoblastic leukemia. Blood Cancer J. 2014;4:244.CrossRefPubMedCentralPubMed
57.
Zurück zum Zitat Yu S, Li A, Liu Q, Yuan X, Xu H, Jiao D, Pestell RG, Han X, Wu K. Recent advances of bispecific antibodies in solid tumors. J Hematol Oncol. 2017;10(1):155.CrossRefPubMedCentralPubMed Yu S, Li A, Liu Q, Yuan X, Xu H, Jiao D, Pestell RG, Han X, Wu K. Recent advances of bispecific antibodies in solid tumors. J Hematol Oncol. 2017;10(1):155.CrossRefPubMedCentralPubMed
58.
Zurück zum Zitat Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, Wu K. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Experimental Hematology & Oncology. 2017;6(1):31.CrossRef Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, Wu K. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Experimental Hematology & Oncology. 2017;6(1):31.CrossRef
59.
Zurück zum Zitat Zhang X, Yang Y, Fan D, Xiong D. The development of bispecific antibodies and their applications in tumor immune escape. Experimental Hematology & Oncology. 2017;6(1):12.CrossRef Zhang X, Yang Y, Fan D, Xiong D. The development of bispecific antibodies and their applications in tumor immune escape. Experimental Hematology & Oncology. 2017;6(1):12.CrossRef
60.
Zurück zum Zitat Flammiger A, Fiedler W, Bacher U, Bokemeyer C, Schneider M, Binder M. Critical imbalance of TNF-alpha and soluble TNF receptor 1 in a patient with macrophage activation syndrome: potential implications for diagnostics and treatment. Acta Haematol. 2012;128(2):69–72.CrossRefPubMed Flammiger A, Fiedler W, Bacher U, Bokemeyer C, Schneider M, Binder M. Critical imbalance of TNF-alpha and soluble TNF receptor 1 in a patient with macrophage activation syndrome: potential implications for diagnostics and treatment. Acta Haematol. 2012;128(2):69–72.CrossRefPubMed
61.
Zurück zum Zitat Prahalad S, Bove KE, Dickens D, Lovell DJ, Grom AA. Etanercept in the treatment of macrophage activation syndrome. J Rheumatol. 2001;28(9):2120–4.PubMed Prahalad S, Bove KE, Dickens D, Lovell DJ, Grom AA. Etanercept in the treatment of macrophage activation syndrome. J Rheumatol. 2001;28(9):2120–4.PubMed
62.
Zurück zum Zitat Gabay C, Lamacchia C, Palmer G. IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol. 2010;6(4):232–41.CrossRefPubMed Gabay C, Lamacchia C, Palmer G. IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol. 2010;6(4):232–41.CrossRefPubMed
Metadaten
Titel
Cytokine release syndrome: grading, modeling, and new therapy
verfasst von
Delong Liu
Juanjuan Zhao
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2018
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-018-0653-x

Weitere Artikel der Ausgabe 1/2018

Journal of Hematology & Oncology 1/2018 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.