Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Ann Med ; 56(1): 2349796, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38738799

ABSTRACT

BACKGROUND: Relapse/refractory B-cell acute lymphoblastic leukaemia (r/r B-ALL) represents paediatric cancer with a challenging prognosis. CAR T-cell treatment, considered an advanced treatment, remains controversial due to high relapse rates and adverse events. This study assessed the efficacy and safety of CAR T-cell therapy for r/r B-ALL. METHODS: The literature search was performed on four databases. Efficacy parameters included minimal residual disease negative complete remission (MRD-CR) and relapse rate (RR). Safety parameters constituted cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). RESULTS: Anti-CD22 showed superior efficacy with the highest MRD-CR event rate and lowest RR, compared to anti-CD19. Combining CAR T-cell therapy with haploidentical stem cell transplantation improved RR. Safety-wise, bispecific anti-CD19/22 had the lowest CRS rate, and anti-CD22 showed the fewest ICANS. Analysis of the costimulatory receptors showed that adding CD28ζ to anti-CD19 CAR T-cell demonstrated superior efficacy in reducing relapses with favorable safety profiles. CONCLUSION: Choosing a more efficacious and safer CAR T-cell treatment is crucial for improving overall survival in acute leukaemia. Beyond the promising anti-CD22 CAR T-cell, exploring costimulatory domains and new CD targets could enhance treatment effectiveness for r/r B-ALL.


Subject(s)
Antigens, CD19 , Immunotherapy, Adoptive , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Sialic Acid Binding Ig-like Lectin 2 , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Antigens, CD19/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Receptors, Chimeric Antigen/immunology , Child , Treatment Outcome , Neoplasm, Residual , Cytokine Release Syndrome/etiology , Recurrence , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology
2.
J Hazard Mater ; 473: 134607, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38761765

ABSTRACT

Paraquat (PQ) exposure is strongly associated with neurotoxicity. However, research on the neurotoxicity mechanisms of PQ varies in terms of endpoints of toxic assessment, resulting in a great challenge to understand the early neurotoxic effects of PQ. In this study, we developed an adverse outcome pathway (AOP) to investigate PQ-induced neuro-immunotoxicity from an immunological perspective, combining of traditional toxicology methods and computer simulations. In vivo, PQ can microstructurally lead to an early synaptic loss in the brain mice, which is a large degree regarded as a main reason for cognitive impairment to mice behavior. Both in vitro and in vivo demonstrated synapse loss is caused by excessive activation of the complement C1q/C3-CD11b pathway, which mediates microglial phagocytosis dysfunction. Additionally, the interaction between PQ and C1q was validated by molecular simulation docking. Our findings extend the AOP framework related to PQ neurotoxicity from a neuro-immunotoxic perspective, highlighting C1q activation as the initiating event for PQ-induced neuro-immunotoxicity. In addition, downstream complement cascades induce abnormal microglial phagocytosis, resulting in reduced synaptic density and subsequent non-motor dysfunction. These findings deepen our understanding of neurotoxicity and provide a theoretical basis for ecological risk assessment of PQ.


Subject(s)
Complement C1q , Computer Simulation , Microglia , Paraquat , Phagocytosis , Paraquat/toxicity , Animals , Complement C1q/immunology , Complement C1q/metabolism , Phagocytosis/drug effects , Microglia/drug effects , Adverse Outcome Pathways , Male , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/etiology , Mice , Brain/drug effects , Herbicides/toxicity , CD11b Antigen/metabolism , Complement C3/metabolism , Molecular Docking Simulation , Synapses/drug effects , Mice, Inbred C57BL
3.
Int Immunopharmacol ; 135: 112336, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38801809

ABSTRACT

IIrbesartan (IRB), an angiotensin II type 1 receptor (AT1R) antagonist, has been widely employed in the medical field for its effectiveness in managing hypertension. However, there have been no documented investigations regarding the immunostimulatory properties of IRB. To address this gap, this study has been performed to assess the neuroprotective impact of IRB as an immunostimulatory agent in mitigating acute neurotoxicity induced by cyclophosphamide (CYP) in rats. mRNA levels of nuclear factor erythroid 2 (Nrf-2), interleukin (IL)-18, IL-1ß, and MMP-1 have been assessed using quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the levels of malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD) has been evaluated to assess the oxidative stress. Additionally, macrophage inflammatory protein 2 (MIP2) has been evaluated using enzyme-linked immunosorbent assay (ELISA). Western blotting has been used to investigate the protein expression of nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) and caspase-1 (CASP-1), along with an assessment of histopathological changes. Administration of IRB protected against oxidative stress by augmenting the levels of GSH and SOD as well as reducing MDA level. Also, administration of IRB led to a diminishment in the brain levels of MIP2 and MMP1. Furthermore, it led to a suppression of IL-1ß and IL-18 levels, which are correlated with a reduction in the abundance of NLRP3 and subsequently CASP-1. This study provides new insights into the immunomodulatory effects of IRB in the context of CYP-induced acute neurotoxicity. Specifically, IRB exerts its effects by reducing oxidative stress, neuroinflammation, inhibiting chemokine recruitment, and mitigating neuronal degeneration through the modulation of immune markers. Therefore, it can be inferred that the use of IRB as an immunomodulator has the potential to effectively mitigate immune disorders associated with inflammation.


Subject(s)
Cyclophosphamide , Inflammasomes , Irbesartan , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Animals , Cyclophosphamide/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Irbesartan/pharmacology , Irbesartan/therapeutic use , Male , Rats , Oxidative Stress/drug effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/immunology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Immunomodulation/drug effects , Rats, Wistar
4.
Nat Rev Clin Oncol ; 21(7): 501-521, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769449

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of several haematological malignancies and is being investigated in patients with various solid tumours. Characteristic CAR T cell-associated toxicities such as cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are now well-recognized, and improved supportive care and management with immunosuppressive agents has made CAR T cell therapy safer and more feasible than it was when the first regulatory approvals of such treatments were granted in 2017. The increasing clinical experience with these therapies has also improved recognition of previously less well-defined toxicities, including movement disorders, immune effector cell-associated haematotoxicity (ICAHT) and immune effector cell-associated haemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), as well as the substantial risk of infection in patients with persistent CAR T cell-induced B cell aplasia and hypogammaglobulinaemia. A more diverse selection of immunosuppressive and supportive-care pharmacotherapies is now being utilized for toxicity management, yet no universal algorithm for their application exists. As CAR T cell products targeting new antigens are developed, additional toxicities involving damage to non-malignant tissues expressing the target antigen are a potential hurdle. Continued prospective evaluation of toxicity management strategies and the design of less-toxic CAR T cell products are both crucial for ongoing success in this field. In this Review, we discuss the evolving understanding and clinical management of CAR T cell-associated toxicities.


Subject(s)
Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/adverse effects , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology
5.
Expert Opin Pharmacother ; 25(3): 263-279, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38588525

ABSTRACT

INTRODUCTION: Chimeric antigen receptor (CAR) T cells have revolutionized the treatment of multiple hematologic malignancies. Engineered cellular therapies now offer similar hope to transform the management of solid tumors and autoimmune diseases. However, toxicities can be serious and often require hospitalization. AREAS COVERED: We review the two chief toxicities of CAR T therapy, cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and the rarer immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. We discuss treatment paradigms and promising future pharmacologic strategies. Literature and therapies reviewed were identified by PubMed search, cited references therein, and review of registered trials. EXPERT OPINION: Management of CRS and ICANS has improved, aided by consensus definitions and guidelines that facilitate recognition and timely intervention. Further data will define optimal timing of tocilizumab and corticosteroids, current foundations of management. Pathophysiologic understanding has inspired off-label use of IL-1 receptor antagonism, IFNγ and IL-6 neutralizing antibodies, and janus kinase inhibitors, with data emerging from ongoing clinical trials. Further strategies to reduce toxicities include novel pharmacologic targets and safety features engineered into CAR T cells themselves. As these potentially curative therapies are used earlier in oncologic therapy and even in non-oncologic indications, effective accessible strategies to manage toxicities are critical.


Subject(s)
Cytokine Release Syndrome , Immunotherapy, Adoptive , Lymphohistiocytosis, Hemophagocytic , Neurotoxicity Syndromes , Receptors, Chimeric Antigen , Humans , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/therapy , Lymphohistiocytosis, Hemophagocytic/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Animals
6.
Radiology ; 302(2): 438-445, 2022 02.
Article in English | MEDLINE | ID: mdl-34751616

ABSTRACT

Background Chimeric antigen receptor (CAR) T-cell immunotherapy is increasingly used for refractory lymphoma but may lead to cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Imaging may assist in clinical management. Associations between CRS or ICANS grade and imaging findings remain not fully established. Purpose To determine associations between imaging findings and clinical grade of CRS or ICANS, evaluate response patterns, and assess imaging use following CAR T-cell treatment. Materials and Methods Patients with refractory B-cell lymphoma who received CAR T-cell infusion between 2018 and 2020 at a single center were analyzed retrospectively. Clinical CRS or ICANS toxicity grade was assessed using American Society for Transplantation and Cellular Therapy, or ASTCT, consensus grading. Thoracic and head images (radiographs, CT scans, MRI scans) were evaluated. Associations between imaging findings and clinical CRS or ICANS grade were analyzed. Wilcoxon signed-rank and χ2 tests were used to assess associations between thoracic imaging findings, clinical CRS toxicity grade, and imaging-based response. Response to therapy was evaluated according to Deauville five-point scale criteria. Results A total of 38 patients (mean age ± standard deviation, 59 years ± 10; 23 men) who received CAR T-cell infusion were included. Of these, 24 (63% [95% CI: 48, 79]) and 11 (29% [95% CI: 14, 44]) experienced clinical grade 1 or higher CRS and ICANS, respectively. Patients with grade 2 or higher CRS were more likely to have thoracic images with abnormal findings (10 of 14 patients [71%; 95% CI: 47, 96] vs five of 24 patients [21%; 95% CI: 4, 37]; P = .002) and more likely to have imaging evidence of pleural effusions (five of 14 [36%; 95% CI: 10, 62] vs two of 24 [8.3%; 95% CI: 0, 20]; P = .04) and atelectasis (eight of 14 [57%; 95% CI: 30, 84] vs six of 24 [25%; 95% CI: 7, 43]; P = .048). Positive imaging findings were identified in three of seven patients (43%) with grade 2 or higher ICANS who underwent neuroimaging. The best treatment response included 20 of 36 patients (56% [95% CI: 39, 72]) with complete response, seven of 36 (19% [95% CI: 6, 33]) with partial response, one of 36 (2.8% [95% CI: 0, 8]) with stable disease, and eight of 36 (22% [95% CI: 8, 36]) with progressive disease. Conclusion Thoracic imaging findings, including pleural effusions and atelectasis, correlated with cytokine release syndrome grade following chimeric antigen receptor (CAR) T-cell infusion. CAR T-cell therapy yielded high response rates. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Langer in this issue.


Subject(s)
Cell- and Tissue-Based Therapy/adverse effects , Cytokine Release Syndrome/etiology , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/therapy , Neurotoxicity Syndromes/etiology , Receptors, Chimeric Antigen/immunology , Cytokine Release Syndrome/diagnostic imaging , Cytokine Release Syndrome/immunology , Female , Humans , Male , Middle Aged , Neurotoxicity Syndromes/diagnostic imaging , Neurotoxicity Syndromes/immunology , Retrospective Studies , Tomography, X-Ray Computed
7.
CA Cancer J Clin ; 72(1): 78-93, 2022 01.
Article in English | MEDLINE | ID: mdl-34613616

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a promising immunotherapeutic treatment concept that is changing the treatment approach to hematologic malignancies. The development of CAR T-cell therapy represents a prime example for the successful bench-to-bedside translation of advances in immunology and cellular therapy into clinical practice. The currently available CAR T-cell products have shown high response rates and long-term remissions in patients with relapsed/refractory acute lymphoblastic leukemia and relapsed/refractory lymphoma. However, CAR T-cell therapy can induce severe life-threatening toxicities such as cytokine release syndrome, neurotoxicity, or infection, which require rapid and aggressive medical treatment in the intensive care unit setting. In this review, the authors provide an overview of the state-of-the-art in the clinical management of severe life-threatening events in CAR T-cell recipients. Furthermore, key challenges that have to be overcome to maximize the safety of CAR T cells are discussed.


Subject(s)
Critical Care/methods , Cytokine Release Syndrome/therapy , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/adverse effects , Neurotoxicity Syndromes/therapy , Cytokine Release Syndrome/immunology , Hematologic Neoplasms/immunology , Humans , Immunotherapy, Adoptive/methods , Neurotoxicity Syndromes/immunology , Receptors, Chimeric Antigen/immunology , Treatment Outcome
8.
Bull Cancer ; 108(10S): S117-S127, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34920794

ABSTRACT

CAR-T cells are modified T cells expressing a chimeric antigen receptor targeting a specific antigen. They have revolutionized the treatment of B cell malignancies (aggressive lymphomas, B-ALL), and this has raised hopes for application in many other pathologies (myeloma, AML, solid tumors, etc.). However, these therapies are associated with novel and specific toxicities (cytokine release syndrome and neurotoxicity). These complications, although mostly managed in a conventional hospitalization unit, can sometimes be life threatening, leading to admission of patients to the intensive care unit. Management relies mainly on anti-IL6R (tocilizumab) and corticosteroids. However, the optimal treatment regimen is still a matter of debate, and the management of the most severe forms is even less well codified. In addition to CRS and ICANS, infections, cytopenia and hypogammaglobulinemia are other frequent complications. This article reviews the mechanisms, risk factors, clinical presentation, and management of these toxicities.


Subject(s)
Cytokine Release Syndrome/drug therapy , Immunotherapy, Adoptive/adverse effects , Neurotoxicity Syndromes/drug therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Adrenal Cortex Hormones/therapeutic use , Agammaglobulinemia/etiology , Agammaglobulinemia/therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/analysis , Cytokine Release Syndrome/diagnosis , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Humans , Infections/etiology , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Risk Factors , T-Lymphocytes/immunology
9.
Ann Clin Lab Sci ; 51(6): 790-794, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34921032

ABSTRACT

OBJECTIVE: To investigate the cytokine release syndrome (CRS) condition for central nervous system B-cell acute lymphocytic leukemia (CNS B-ALL) patients after CAR-Ts targeting CD19 with short hairpin RNA (shRNA)-IL-6 gene silencing technology (ssCART-19s) infusion. METHODS: This prospective observational research included a total of 12 cases of patients with CNS B-ALL from March 2017 to February 2020. ssCART-19 infusions (5×106 cells/kg) were given to patients for 3 consecutive days. After infusion, the temperature of all patients was detected constantly and the CRS was carefully monitored within 1 month after treatment. The serum levels of IL-2, IL-4, IL-6, IL-10, IFN-γ, TNF-α, CRP and IL-17A were tested by enzyme-linked immunosorbent assay (ELISA) within 10 days after infusion. RESULTS: All 12 CNS B-ALL patients showed CRS with 100% incidence rate, with 3 cases (25.00%) of CRS stage I and 9 cases (75.00%) with CRS stage II. No CRS stage III~V was observed. The overall response rate was 91.67% (11/12), with 10 patients (83.33%) showed CR and 1 case (8.33%) of PR. In 9 patients with CRS stage II, the temperature increased persistently, ranging from 4 days to 14 days after infusion, and decreased gradually after 14 days of nursing treatment. The hyperthermia condition started from 1 day after infusion and returned to baseline at the following 2-10 days of nursing treatment. The levels of the inflammatory factors increased markedly after ssCAR-T19s infusion for 2-3 days compared to the baseline, and gradually returned to the baseline after treatment. After 10 days of infusion, all inflammatory factors returned to normal levels. CONCLUSION: ssCART-19s infusion induced short-term slight CRS with increased temperature and inflammatory factors, and no severe CRS was observed.


Subject(s)
Central Nervous System Neoplasms/therapy , Cytokine Release Syndrome , Gene Knockdown Techniques/methods , Immunotherapy, Adoptive , Interleukin-6 , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen , Adult , Antigens, CD19/immunology , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/physiopathology , Cytokine Release Syndrome/therapy , Female , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Interleukin-6/genetics , Interleukin-6/immunology , Male , Monitoring, Physiologic/methods , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Small Interfering/therapeutic use , Receptors, Chimeric Antigen/administration & dosage , Receptors, Chimeric Antigen/immunology , Treatment Outcome
10.
Front Immunol ; 12: 738456, 2021.
Article in English | MEDLINE | ID: mdl-34721401

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has shown unprecedented success in treating advanced hematological malignancies. Its effectiveness in solid tumors has been limited due to heterogeneous antigen expression, a suppressive tumor microenvironment, suboptimal trafficking to the tumor site and poor CAR T cell persistence. Several approaches have been developed to overcome these obstacles through various strategies including the genetic engineering of CAR T cells to blunt the signaling of immune inhibitory receptors as well as to modulate signaling of cytokine/chemokine molecules and their receptors. In this review we offer our perspective on how genetically modifying cytokine/chemokine molecules and their receptors can improve CAR T cell qualities such as functionality, persistence (e.g. resistance to pro-apoptotic signals) and infiltration into tumor sites. Understanding how such modifications can overcome barriers to CAR T cell effectiveness will undoubtedly enhance the potential of CAR T cells against solid tumors.


Subject(s)
Cytokines/genetics , Genetic Therapy , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/transplantation , Animals , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/prevention & control , Cytokines/immunology , Cytokines/metabolism , Genetic Therapy/adverse effects , Humans , Immunotherapy, Adoptive/adverse effects , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Phenotype , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Risk Factors , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape , Tumor Microenvironment
11.
Sci Rep ; 11(1): 22884, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819589

ABSTRACT

Immune cell infiltration has been implicated in neurotoxic chemotherapy for cancer treatment. However, our understanding of immune processes is still incomplete and current methods of observing immune cells are time consuming or invasive. Corneal dendritic cells are potent antigen-presenting cells and can be imaged with in-vivo corneal confocal microscopy. Corneal dendritic cell densities and nerve parameters in patients treated with neurotoxic chemotherapy were investigated. Patients treated for cancer with oxaliplatin (n = 39) or paclitaxel (n = 48), 3 to 24 months prior to assessment were recruited along with 40 healthy controls. Immature (ImDC), mature (MDC) and total dendritic cell densities (TotalDC), and corneal nerve parameters were analyzed from in-vivo corneal confocal microscopy images. ImDC was increased in the oxaliplatin group (Median, Md = 22.7 cells/mm2) compared to healthy controls (Md = 10.1 cells/mm2, p = 0.001), but not in the paclitaxel group (Md = 10.6 cells/mm2). ImDC was also associated with higher oxaliplatin cumulative dose (r = 0.33, p = 0.04) and treatment cycles (r = 0.40, p = 0.01). There was no significant difference in MDC between the three groups (p > 0.05). Corneal nerve parameters were reduced in both oxaliplatin and paclitaxel groups compared to healthy controls (p < 0.05). There is evidence of elevation of corneal ImDC in oxaliplatin-treated patients. Further investigation is required to explore this potential link through longitudinal studies and animal or laboratory-based immunohistochemical research.


Subject(s)
Antineoplastic Agents/adverse effects , Cornea/drug effects , Dendritic Cells/drug effects , Nerve Fibers/drug effects , Neurotoxicity Syndromes/etiology , Oxaliplatin/adverse effects , Paclitaxel/adverse effects , Aged , Case-Control Studies , Cornea/immunology , Cornea/innervation , Cornea/pathology , Cross-Sectional Studies , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Humans , Male , Microscopy, Confocal , Middle Aged , Nerve Fibers/pathology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Time Factors , Treatment Outcome
12.
Inflamm Res ; 70(10-12): 1151-1164, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34661679

ABSTRACT

OBJECTIVE: Gisenoside Rg1 is a potent neuroprotectant in ginseng. The aim of this study was to investigate the elimination effect of Rg1 on cadmium (Cd)-induced neurotoxicity. MATERIALS AND METHODS: A cumulative Cd exposure mouse model was established. Also, the toxicity of Cd and the protective effect of Rg1 were examined in vitro using cultured neurons and microglia. RESULTS: We found that Cd-intoxicated mice exhibited significant injury in the liver, kidney, small intestine, and testis, along with cognitive impairment. Antioxidant enzymes such as SOD, GSH-Px and CAT were reduced in the blood and brain, and correspondingly, the lipid peroxidation product MDA was elevated. In the brain, astrocytes and microglia were activated, characterized by an increase in inflammatory factors such as TNF-α, IL-1ß and IL-6, as well as their protein markers GFAP and IBA1. However, Rg1 eliminated Cd-induced toxicity and restored oxidative stress and inflammatory responses, correspondingly restoring the behavioral performance of the animals. Meanwhile, the BDNF-TrkB/Akt and Notch/HES-1 signaling axes were involved in the Rg1-mediated elimination of Cd-induced toxicity. CONCLUSION: Rg1 is a promising agent for the elimination of Cd-induced toxicity.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cadmium , Ginsenosides/therapeutic use , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Brain/drug effects , Brain/immunology , Brain/pathology , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Ginsenosides/pharmacology , Intestine, Small/drug effects , Intestine, Small/pathology , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Oxidative Stress/drug effects , Oxidoreductases/immunology , Testis/drug effects , Testis/pathology
13.
Immunotherapy ; 13(15): 1261-1269, 2021 10.
Article in English | MEDLINE | ID: mdl-34558978

ABSTRACT

Aims: Neurotoxicity (NT) is a common complication of chimeric antigen receptor (CAR) T-cell therapy. Data on early clinical identifiers for impending severe NT are lacking. Methods: The authors performed a retrospective study on 26 adult relapsed/refractory diffuse large B cell lymphoma patients treated with commercial CAR T-cell therapy (December 2017 - September 2018). Results: NT of any grade and severe NT occurred in 88 and 31% of patients, respectively. Dysgraphia (p < 0.01), disorientation (p = 0.01) and inattention (p = 0.018) were associated with severe NT, with positive predictive values of 100, 87.5 and 87.5%, respectively. Dysnomia was not associated with severe NT. Conclusion: In the authors' limited cohort, the dysgraphia, disorientation and inattention components of the CAR T-cell therapy-associated toxicity 10 scoring system were significantly associated with and predictive of impending severe NT.


Lay abstract Neurotoxicity (NT) is a common complication of chimeric antigen receptor (CAR) T-cell therapy. Information on early signs and symptoms of impending severe NT is lacking. The authors studied 26 adult patients with relapsed/refractory diffuse large B cell lymphoma who received commercial CAR T-cell therapy for the development of NT. The authors found that NT was common, with 31% of patients experiencing severe NT. In this relatively small patient population, the authors found that impaired writing, disorientation and poor attention, which are components of the CAR T-cell therapy-associated toxicity 10 scoring system, were significantly associated with and predictive of impending severe NT.


Subject(s)
Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, Large B-Cell, Diffuse/drug therapy , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/physiopathology , Receptors, Chimeric Antigen/immunology , Aged , Antigens, CD19/immunology , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Middle Aged , Retrospective Studies
14.
Article in English | MEDLINE | ID: mdl-34497101

ABSTRACT

BACKGROUND AND OBJECTIVES: To describe the marked clinical and biological responses of a targeted treatment with anti-interleukin-6 (IL-6)-receptor antibody and Janus kinase (JAK) inhibitors in a patient with a severe, corticoresistant CNS toxicity of immune-checkpoint inhibitor (ICI) therapy. METHODS: A 58-year-old man was admitted for subacute paraparesis, urinary retention, and ascending paresthesia. He was under treatment with ipilimumab and nivolumab for metastatic melanoma. Spine MRI disclosed multiple T2-hyperintense, contrast-enhancing longitudinally extensive lesions. A diagnosis of ICI-related acute transverse myelitis was made. RESULTS: ICIs were immediately discontinued, and the patient received high-dose glucocorticoids plus 1 session of plasma exchange, but he did not improve. Based on the marked elevation of CSF IL-6 (505 pg/mL), a second-line targeted therapy with anti-IL-6-receptor tocilizumab (8 mg/kg/mo for 3 infusions) plus JAK inhibitor ruxolitinib (50 mg/d) was administered. Patient neurologic status started to improve shortly after, with corresponding radiologic resolution. At 9 months, the patient was able to walk independently, presenting only slight residual disability while remaining in oncologic partial response. DISCUSSION: Our case suggests that some patients with severe, corticoresistant CNS immune-related toxicities of ICIs may benefit from cytokine blockade. Cytokine measurement in serum and CSF might help in selecting patients for personalized treatment strategies.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Immune Checkpoint Inhibitors/toxicity , Janus Kinase Inhibitors/pharmacology , Melanoma/drug therapy , Myelitis, Transverse , Neurotoxicity Syndromes , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Interleukin-6/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Humans , Ipilimumab/toxicity , Janus Kinase Inhibitors/administration & dosage , Male , Middle Aged , Myelitis, Transverse/chemically induced , Myelitis, Transverse/drug therapy , Myelitis, Transverse/immunology , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Nitriles/administration & dosage , Nivolumab/toxicity , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage
15.
N Z Med J ; 134(1542): 96-108, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34531588

ABSTRACT

Chimeric antigen receptor (CAR) T-cells are a personalised cell and gene therapy for cancer that are becoming an international standard of care for some refractory B-cell leukaemias, non-Hodgkin lymphomas and myeloma. A single CAR T-cell administration can result in durable complete response for some recipients. Domestic CAR T-cell manufacturing capability was established for Aotearoa New Zealand's first CAR T-cell trial (ENABLE, ClinicalTrials.gov NCT04049513). This article outlines CAR T-cell manufacturing and logistical considerations, with a focus on New Zealand's environment for this personalised cell and gene therapy. We discuss Maori engagement in CAR T-cell trial and clinical service design, and propose enhancing Maori guardianship (kaitiakitanga) over cells and genetic material through on-shore manufacture. Strategies to safely deliver CAR T-cells within New Zealand's healthcare system are outlined. Finally, we discuss challenges to, and opportunities for, widening CAR T-cell availability and assuring equity of access. Based on our experience, we consider Aotearoa New Zealand to be in an excellent position to develop and implement investigational and commercial CAR T-cell therapies in the future.


Subject(s)
Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Lymphoma, Non-Hodgkin/therapy , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes/transplantation , Adolescent , Adult , Aged , Female , Health Equity , Humans , Male , Middle Aged , Native Hawaiian or Other Pacific Islander , Neoplasm Recurrence, Local , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , New Zealand , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Remission Induction , T-Lymphocytes/immunology , Young Adult
16.
Dtsch Med Wochenschr ; 146(17): 1129-1137, 2021 09.
Article in German | MEDLINE | ID: mdl-34448189

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).


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Cytokine Release Syndrome , Immunotherapy, Adoptive/adverse effects , Neoplasms/immunology , Neoplasms/therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/therapy , Antineoplastic Agents, Immunological/therapeutic use , Biological Products , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Humans , Leukapheresis , Neoplasms/pathology , Neurotoxicity Syndromes/immunology , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , T-Lymphocytes/immunology , Treatment Outcome
17.
Toxicol Appl Pharmacol ; 426: 115649, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34273407

ABSTRACT

The use of the anthelmintic levamisole as a cocaine adulterant has been increasing worldwide. Complications caused by this association include systemic vasculitis, agranulocytosis, neutropenia, tissue necrosis, pulmonary hemorrhage, and renal injury. Data about toxicity of levamisole are scarce, therefore the aim of this study was to evaluate the acute and subchronic toxic effects of levamisole in rats. Male Wistar rats received saline or levamisole by intraperitoneal route at the doses of 12, 24 and 36 mg/kg in the acute toxicity test; and at 3, 6 and 12 mg/kg in the subchronic toxicity test. Toxicity was evaluated using behavioral, cognitive, renal, hematological, biochemical and histopathological parameters. Acute administration of levamisole caused behavioral and histopathological alterations. Subchronic administration caused behavioral, cognitive and hematological alterations (p < 0.0001 and p < 0.05, respectively), impairment of liver and kidney functions (p < 0.05), and changes of antioxidant defenses (p ≤ 0.0001). Both administrations produced toxic effects of clinical relevance, which make levamisole a dangerous cutting agent. Furthermore, the knowledge of these effects can contribute to the correct diagnosis and treatment of cocaine dependents with unusual systemic alterations.


Subject(s)
Antinematodal Agents/toxicity , Levamisole/toxicity , Neurotoxicity Syndromes/etiology , Animals , Behavior, Animal/drug effects , Cocaine , Leukocyte Count , Liver/drug effects , Liver/pathology , Male , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Organ Size/drug effects , Oxidative Stress/drug effects , Rats, Wistar , Toxicity Tests, Acute
18.
Bull Cancer ; 108(12S): S53-S64, 2021 Dec.
Article in French | MEDLINE | ID: mdl-34253335

ABSTRACT

CAR-T cells represent a new anti-tumor immunotherapy which has shown its clinical efficacy in B-cell malignancies. The results of clinical trials carried out in this context have shown that certain immunological characteristics of patients before (at the time of apheresis) and after the administration of the treatment, or of the CAR-T cells themselves, are correlated with the response to the treatment or to its toxicity. However, to date, there are no recommendations on the immunological monitoring of patients treated in real life. The objectives of this workshop were to determine, based on data from the literature and the experience of the centers, the immunological analyses to be carried out in patients treated with CAR-T cells. The recommendations relate to the characterization of the patient's immune cells at the time of apheresis, the characterization of the injected CAR-T cells, as well as the monitoring of the CAR-T cells and other parameters of immune reconstitution in the patient after administration of the treatment. Harmonization of practices will allow clinical-biological correlation studies to be carried out in patients treated in real life with the aim of identifying factors predictive of response and toxicity. Such data could have a major medico-economic impact by making it possible to identify the patients who will optimally benefit from these expensive treatments.


Subject(s)
Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Immune Reconstitution , Immunotherapy, Adoptive , Monitoring, Immunologic/standards , Bacterial Infections/etiology , Blood Component Removal , Cytokine Release Syndrome/immunology , Flow Cytometry , Humans , Immunity, Cellular , Immunotherapy, Adoptive/adverse effects , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Lymphocyte Depletion , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , Monitoring, Immunologic/methods , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Mycoses/etiology , Neurotoxicity Syndromes/immunology , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Recurrence , Societies, Medical , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation , Virus Diseases/etiology
19.
Front Immunol ; 12: 623610, 2021.
Article in English | MEDLINE | ID: mdl-34093519

ABSTRACT

Severe cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) strongly hampered the broad clinical applicability of chimeric antigen receptor T cell (CAR-T) therapy. Vascular endothelial activation has been suggested to contribute to the development of CRS and ICANS after CAR-T therapy. However, therapeutic strategies targeting endothelial dysfunction during CAR-T therapy have not been well studied yet. Here, we found that tumor necrosis factor α (TNFα) produced by CAR-T cells upon tumor recognition and interleukin 1ß (IL1ß) secreted by activated myeloid cells were the main cytokines in inducing endothelial activation. Therefore, we investigated the potential effectiveness of TNFα and IL1ß signaling blockade on endothelial activation in CAR-T therapy. The blockade of TNFα and IL1ß with adalimumab and anti-IL1ß antibody respectively, as well as the application of focal adhesion kinase (FAK) inhibitor, effectively ameliorated endothelial activation induced by CAR-T, tumor cells, and myeloid cells. Moreover, adalimumab and anti-IL1ß antibody exerted synergistic effect on the prevention of endothelial activation induced by CAR-T, tumor cells, and myeloid cells. Our results indicate that TNFα and IL1ß blockade might have therapeutic potential for the treatment of CAR-T therapy-associated CRS and neurotoxicity.


Subject(s)
Adalimumab/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Immunotherapy, Adoptive , Interleukin-1beta/antagonists & inhibitors , Myeloid Cells/drug effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/drug effects , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/metabolism , Cell Line, Tumor , Coculture Techniques , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/prevention & control , HEK293 Cells , Human Umbilical Vein Endothelial Cells/immunology , Humans , Immunotherapy, Adoptive/adverse effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...