Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1385079, 2024.
Article in English | MEDLINE | ID: mdl-38948517

ABSTRACT

Background: 177Lu-oxodotreotide peptide receptor therapy (LuPRRT) is an efficient treatment for midgut neuroendocrine tumors (NETs) of variable radiological response. Several clinical, biological, and imaging parameters may be used to establish a relative disease prognosis but none is able to predict early efficacy or toxicities. We investigated expression levels for mRNA and miRNA involved in radiosensitivity and tumor progression searching for correlations related to patient outcome during LuPRRT therapy. Methods: Thirty-five patients received LuPRRT for G1/G2 midgut NETs between May 2019 and September 2021. Peripheral blood samples were collected prior to irradiation, before and 48 h after the second and the fourth LuPRRT, and at 6-month follow-up. Multiple regression analyses and Pearson correlations were performed to identify the miRNA/mRNA signature that will best predict response to LuPRRT. Results: Focusing on four mRNAs and three miRNAs, we identified a miRNA/mRNA signature enabling the early identification of responders to LuPRRT with significant reduced miRNA/mRNA expression after the first LuPRRT administration for patients with progressive disease at 1 year (p < 0.001). The relevance of this signature was reinforced by studying its evolution up to 6 months post-LuPRRT. Moreover, nadir absolute lymphocyte count within the first 2 months after the first LuPRRT administration was significantly related to low miRNA/mRNA expression level (p < 0.05) for patients with progressive disease. Conclusion: We present a pilot study exploring a miRNA/mRNA signature that correlates with early hematologic toxicity and therapeutic response 12 months following LuPRRT. This signature will be tested prospectively in a larger series of patients.


Subject(s)
Intestinal Neoplasms , MicroRNAs , Neuroendocrine Tumors , RNA, Messenger , Humans , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/radiotherapy , Neuroendocrine Tumors/pathology , Male , Female , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , Intestinal Neoplasms/blood , Intestinal Neoplasms/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/drug therapy , RNA, Messenger/genetics , RNA, Messenger/blood , Aged , Follow-Up Studies , Adult , Prognosis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Receptors, Peptide/genetics , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/administration & dosage , Lutetium , Radioisotopes
2.
Technol Cancer Res Treat ; 23: 15330338241252622, 2024.
Article in English | MEDLINE | ID: mdl-38845139

ABSTRACT

Purpose: The aim of this matched-pair cohort study was to evaluate the potential of intensity-modulated proton therapy (IMPT) for sparring of the pelvic bone marrow and thus reduction of hematotoxicity compared to intensity-modulated photon radiotherapy (IMRT) in the setting of postoperative irradiation of gynaecological malignancies. Secondary endpoint was the assessment of predictive parameters for the occurrence of sacral insufficiency fractures (SIF) when applying IMPT. Materials and Methods: Two cohorts were analyzed consisting of 25 patients each. Patients were treated with IMPT compared with IMRT and had uterine cervical (n = 8) or endometrial cancer (n = 17). Dose prescription, patient age, and diagnosis were matched. Dosimetric parameters delivered to the whole pelvic skeleton and subsites (ilium, lumbosacral, sacral, and lower pelvis) and hematological toxicity were evaluated. MRI follow-up for evaluation of SIF was only available for the IMPT group. Results: In the IMPT group, integral dose to the pelvic skeleton was significantly lower (23.4GyRBE vs 34.3Gy; p < 0.001), the average V5Gy, V10Gy, and V20Gy were reduced by 40%, 41%, and 28%, respectively, compared to the IMRT group (p < 0.001). In particular, for subsites ilium and lower pelvis, the low dose volume was significantly lower. Hematotoxicity was significantly more common in the IMRT group (80% vs 32%; p = 0009), especially hematotoxicity ≥ CTCAE II (36% vs 8%; p = 0.037). No patient in the IMPT group experienced hematotoxicity > CTCAE II. In the IMPT cohort, 32% of patients experienced SIF. Overall SIF occurred more frequently with a total dose of 50.4 GyRBE (37.5%) compared to 45 GyRBE (22%). No significant predictive dose parameters regarding SIF could be detected aside from a trend regarding V50Gy to the lumbosacral subsite. Conclusion: Low-dose exposure to the pelvic skeleton and thus hematotoxicity can be significantly reduced by using IMPT compared to a matched photon cohort. Sacral insufficiency fracture rates appear similar to reported rates for IMRT in the literature.


Subject(s)
Bone Marrow , Genital Neoplasms, Female , Proton Therapy , Radiotherapy Dosage , Radiotherapy, Intensity-Modulated , Humans , Female , Radiotherapy, Intensity-Modulated/adverse effects , Radiotherapy, Intensity-Modulated/methods , Proton Therapy/adverse effects , Proton Therapy/methods , Bone Marrow/radiation effects , Bone Marrow/pathology , Middle Aged , Aged , Genital Neoplasms, Female/radiotherapy , Adult , Radiotherapy Planning, Computer-Assisted , Organs at Risk/radiation effects , Organ Sparing Treatments/methods
3.
In Vivo ; 38(4): 2041-2048, 2024.
Article in English | MEDLINE | ID: mdl-38936894

ABSTRACT

BACKGROUND/AIM: Hematotoxicity is a life-threatening condition that has become the major cause of drug discontinuation in patients with acute lymphoblastic leukemia (ALL). The nudix hydrolase 15 (NUDT15) gene polymorphism (c.415C>T) is reported to have an association with the hematotoxicity of 6-mercaptopurine (6-MP) as maintenance therapy in patients with ALL. However, the prevalence of this genetic polymorphism in the Indonesian population is unknown. This study aimed to assess the frequency of NUDT15 polymorphism among Indonesian pediatric patients with ALL and its association with the hematotoxicity of 6-MP. PATIENTS AND METHODS: A total of 101 stored DNA samples from pediatric patients with ALL receiving 6-MP treatment were used for genetic testing. Direct sequencing was conducted to determine the NUDT15 c.415C>T genotype. Chi-square or Fisher's exact test were employed to examine the association between the NUDT15 c.415C>T genotype and hematotoxicity. RESULTS: All (100%) of the DNA samples from patients with ALL treated with 6-MP exhibited a homozygous variant of the NUDT15 c.415C>T genotype, 70.3% of which showed hematotoxicity to some extent. We found no significant differences in NUDT15 gene polymorphism among patients with ALL with different states of hematotoxicity. CONCLUSION: The observed high frequency of NUDT15 c.415C>T in our study population might explain the elevated prevalence of 6-MP-associated hematotoxicity in pediatric patients with ALL within the Indonesian population. Our study provides new insight regarding the NUDT15 gene polymorphism and its relation to hematotoxicity. Further studies are required to determine the necessity of adjusting the initial dose of 6-MP for Indonesian pediatric patients with ALL.


Subject(s)
Mercaptopurine , Nudix Hydrolases , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Pyrophosphatases , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Alleles , Antimetabolites, Antineoplastic/adverse effects , Gene Frequency , Genetic Predisposition to Disease , Genotype , Indonesia/epidemiology , Mercaptopurine/adverse effects , Nudix Hydrolases/genetics , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrophosphatases/genetics
4.
Chem Biol Interact ; 398: 111107, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38866309

ABSTRACT

Benzene is the main environmental pollutant and risk factor of childhood leukemia and chronic benzene poisoning. Benzene exposure leads to hematopoietic stem and progenitor cell (HSPC) dysfunction and abnormal blood cell counts. However, the key regulatory targets and mechanisms of benzene hematotoxicity are unclear. In this study, we constructed a benzene-induced hematopoietic damage mouse model to explore the underlying mechanisms. We identified that Insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) was significantly reduced in benzene-exposed mice. Moreover, targeting IGF2BP1 effectively mitigated damages to hematopoietic function and hematopoietic molecule expression caused by benzene in mice. On the mechanics, by metabolomics and transcriptomics, we discovered that branched-chain amino acid (BCAA) metabolism and fatty acid oxidation were key metabolic pathways, and Branched-chain amino acid transaminase 1 (BCAT1) and Carnitine palmitoyltransferase 1a (CPT1A) were critical metabolic enzymes involved in IGF2BP1-mediated hematopoietic injury process. The expression of the above molecules in the benzene exposure population was also examined and consistent with animal experiments. In conclusion, targeting IGF2BP1 alleviated hematopoietic injury caused by benzene exposure, possibly due to the reprogramming of BCAA metabolism and fatty acid oxidation via BCAT1 and CPT1A metabolic enzymes. IGF2BP1 is a potential regulatory and therapeutic target for benzene hematotoxicity.


Subject(s)
Amino Acids, Branched-Chain , Benzene , Fatty Acids , Oxidation-Reduction , Animals , Benzene/toxicity , Amino Acids, Branched-Chain/metabolism , Fatty Acids/metabolism , Oxidation-Reduction/drug effects , Mice , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Male , Mice, Inbred C57BL , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/drug effects
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 617-626, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708493

ABSTRACT

OBJECTIVE: To investigate immunogenic and toxic effects of graphene oxide (GO) nanoparticles in mouse skeletal muscles and in human blood in vitro. METHODS: GO nanoparticles prepared using a probe sonicator were supended in deionized H2O or PBS, and particle size and surface charge of the nanoparticles were measured with dynamic light scattering (DLS). Different concentrations (0.5, 1.0 and 2.0 mg/mL) of GO suspension or PBS were injected at multiple sites in the gastrocnemius muscle (GN) of C57BL/6 mice, and inflammatory response and immune cell infiltrations were detected with HE and immunofluorescence staining. We also examined the effects of GO nanoparticles on human red blood cell (RBC) morphology, hemolysis and blood coagulation using scanning electron microscope (SEM), spectrophotometry, and thromboelastography (TEG). RESULTS: GO nanoparticles suspended in PBS exhibited better colloidal dispersity, stability and surface charge effects than those in deionized H2O. In mouse GNs, injection of GO suspensions dose- and time-dependently resulted in sustained muscular inflammation and myofiber degeneration at the injection sites, which lasted till 8 weeks after the injection; immunofluorescence staining revealed obvious infiltration of monocytes, macrophages, dendritic cells and CD4+ T cells around the injection sites in mouse GNs. In human RBCs, incubation with GO suspensions at 0.2, 2.0 and 20 mg/mL, but not at 0.002 or 0.02 mg/mL, caused significant alterations of cell morphology and hemolysis. TEG analysis showed significant abnormalities of blood coagulation parameters following treatment with high concentrations of GO. CONCLUSION: GO nanoparticles can induce sustained inflammatory and immunological responses in mouse GNs and cause RBC hemolysis and blood coagulation impairment, suggesting its muscular toxicity and hematotoxicity at high concentrations.


Subject(s)
Erythrocytes , Graphite , Hemolysis , Mice, Inbred C57BL , Muscle, Skeletal , Nanoparticles , Animals , Graphite/toxicity , Graphite/chemistry , Mice , Erythrocytes/drug effects , Humans , Muscle, Skeletal/drug effects , Hemolysis/drug effects , Particle Size , Blood Coagulation/drug effects
6.
Biomed Environ Sci ; 37(4): 341-353, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38727157

ABSTRACT

Objective: Hydroquinone (HQ), one of the phenolic metabolites of benzene, is widely recognized as an important participant in benzene-induced hematotoxicity. However, there are few relevant proteomics in HQ-induced hematotoxicity and the mechanism hasn't been fully understood yet. Methods: In this study, we treated K562 cells with 40 µmol/L HQ for 72 h, examined and validated protein expression changes by Label-free proteomic analysis and Parallel reaction monitoring (PRM), and performed bioinformatics analysis to identify interaction networks. Results: One hundred and eighty-seven upregulated differentially expressed proteins (DEPs) and 279 downregulated DEPs were identified in HQ-exposed K562 cells, which were involved in neutrophil-mediated immunity, blood microparticle, and other GO terms, as well as the lysosome, metabolic, cell cycle, and cellular senescence-related pathways. Focusing on the 23 DEGs and 5 DEPs in erythroid differentiation-related pathways, we constructed the network of protein interactions and determined 6 DEPs (STAT1, STAT3, CASP3, KIT, STAT5B, and VEGFA) as main hub proteins with the most interactions, among which STATs made a central impact and may be potential biomarkers of HQ-induced hematotoxicity. Conclusion: Our work reinforced the use of proteomics and bioinformatic approaches to advance knowledge on molecular mechanisms of HQ-induced hematotoxicity at the protein level and provide a valuable basis for further clarification.


Subject(s)
Benzene , Hemolytic Agents , Proteome , Proteome/metabolism , Proteomics , Benzene/toxicity , K562 Cells , Humans , Toxicity Tests/methods , Hemolytic Agents/toxicity
7.
Sci Total Environ ; 930: 172608, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38653421

ABSTRACT

The effect of the lead (Pb), cadmium (Cd), mercury (Hg) and arsenic (As) mixture (MIX) on hematotoxicity development was investigated trough combined approach. In vivo subacute study (28 days) was performed on rats (5 per group): a control group and five groups orally exposed to increasing metal(loid) mixture doses, MIX 1- MIX 5 (mg/kg bw./day) (Pb: 0.003, 0.01, 0.1, 0.3, 1; Cd: 0.01, 0.03, 0.3, 0.9, 3; Hg: 0.0002, 0.0006, 0.006, 0.018, 0.06; As: 0.002, 0.006, 0.06, 0.18, 0.6). Blood was taken for analysis of hematological parameters and serum iron (Fe) analysis. MIX treatment increased thrombocyte/platelet count and MCHC and decreased Hb, HCT, MCV and MCH values compared to control, indicating the development of anemia and thrombocytosis. BMDIs with the narrowest width were identified for MCH [pg] (6.030E-03 - 1.287E-01 mg Pb/kg bw./day; 2.010E-02 - 4.290E-01 mg Cd/kg bw./day; 4.020E-04 - 8.580E-03 mg Hg/kg bw./day; 4.020E-03 - 8.580E-02 mg As/kg bw./day). In silico analysis showed target genes connected with MIX and the development of: anemia - ACHE, GSR, PARP1, TNF; thrombocytosis - JAK2, CALR, MPL, THPO; hematological diseases - FAS and ALAD. The main extracted pathways for anemia were related to apoptosis and oxidative stress; for thrombocytosis were signaling pathways of Jak-STAT and TPO. Changes in miRNAs and transcription factors enabled the mode of action (MoA) development based on the obtained results, contributing to mechanistic understanding and hematological risk related to MIX exposure.


Subject(s)
Arsenic , Cadmium , Lead , Mercury , Animals , Rats , Lead/toxicity , Cadmium/toxicity , Mercury/toxicity , Arsenic/toxicity , Computer Simulation , Male , Environmental Pollutants/toxicity
8.
Gut Microbes ; 16(1): 2323227, 2024.
Article in English | MEDLINE | ID: mdl-38436067

ABSTRACT

Due to the annual increase in its production and consumption in occupational environments, the adverse blood outcomes caused by benzene are of concern. However, the mechanism of benzene-induced hematopoietic damage remains elusive. Here, we report that benzene exposure causes hematopoietic damage in a dose-dependent manner and is associated with disturbances in gut microbiota-long chain fatty acids (LCFAs)-inflammation axis. C57BL/6J mice exposed to benzene for 45 days were found to have a significant reduction in whole blood cells and the suppression of hematopoiesis, an increase in Bacteroides acidifaciens and a decrease in Lactobacillus murinus. Recipient mice transplanted with fecal microbiota from benzene-exposed mice showed potential for hematopoietic disruption, LCFAs, and interleukin-5 (IL-5) elevation. Abnormally elevated plasma LCFAs, especially palmitoleic acid (POA) exacerbated benzene-induced immune-inflammation and hematopoietic damage via carnitine palmitoyltransferase 2 (CPT2)-mediated disorder of fatty acid oxidation. Notably, oral administration of probiotics protects the mice against benzene-induced hematopoietic toxicity. In summary, our data reveal that the gut microbiota-POA-IL-5 axis is engaged in benzene-induced hematopoietic damage. Probiotics might be a promising candidate to prevent hematopoietic abnormalities from benzene exposure.


Subject(s)
Fatty Acids, Monounsaturated , Gastrointestinal Microbiome , Interleukin-5 , Animals , Mice , Mice, Inbred C57BL , Benzene/toxicity , Fatty Acids , Inflammation
9.
Toxicology ; 503: 153758, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38367942

ABSTRACT

Benzene exposure leads to hematotoxicity, and epigenetic modification is considered to be a potential mechanism of benzene pathogenesis. As a newly discovered post-transcriptional modification, the roles of N6-methyladenosine (m6A) in benzene hematotoxicity are still unclear. m6A can only exert its gene regulatory function after being recognized by m6A reading proteins. In this study, we found that the expression of m6A reader IGF2BP1 decreased in benzene poisoning workers and in 20 µM benzene metabolite 1,4-BQ-treated AHH-1 cells. Further overexpression of IGF2BP1 in mice alleviated 50 ppm benzene-induced hematopoietic damage, suggesting that IGF2BP1 plays a critical role in benzene hematotoxicity. Next, we examined transcriptome-wide m6A methylation in vitro to search for target genes of IGF2BP1. We found that benzene metabolite 1,4-BQ treatment altered the m6A methylation levels of various genes. The comprehensive analysis of mRNA expression and m6A methylation uncovered that the hypomethylated Ribosomal Protein L36 (RPL36) and its consequent reduced expression impaired cell proliferation. Mechanically, m6A modification reduced RNA stability to down-regulate RPL36 expression. Moreover, overexpression of IGF2BP1 relieved RPL36 reduction and cell proliferation inhibition caused by benzene in vitro and in vivo by directly binding with RPL36 mRNA. In conclusion, the m6A reader IGF2BP1 attenuates the stability of RPL36 and cell proliferation to mediate benzene hematotoxicity by recognizing m6A modification. IGF2BP1 and RPL36 may be key molecules and potential therapeutic targets for benzene hematotoxicity.


Subject(s)
Adenine/analogs & derivatives , Benzene , Mice , Animals , Benzene/toxicity , Methylation , RNA, Messenger/metabolism , Biomarkers/metabolism , Cell Proliferation
10.
Drug Chem Toxicol ; : 1-14, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38326995

ABSTRACT

This study investigated the curative effect of black cumin oil (Nigella sativa, NS), which is a phytotherapeutic agent against to cypermethrin (CYP), which is known to have adverse effects on rainbow trout (Oncorhynchus mykiss)'s behavioral changes, oxidative stress-mediated neurotoxicity, hematotoxicity and hepatotoxicity parameters.At the end of the trial period; (i) evaluation of critical swimming speed (Ucrit) (ii) hematology indices [white blood cell (WBC), red blood cell (RBC), hemoglobin (Hgb), hematocrit (Hct), mean cell volume (MCV), mean cell hemoglobin) (MCH), mean cell hemoglobin concentration (MCHC)] (iii) Elucidation of the mechanism of functional damage in brain tissue of O. mykiss by neurological parameter [acetylcholinesterase (AChE)] (iv) Evaluation of oxidative damage in oxidative stress-mediated neurotoxicity and hepatotoxicity in liver, gill and brain tissue of O. mykiss with antioxidant enzymes [(Superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), Glutathione (GSH)] and [(detection by means of malondialdehyde (MDA)] (v) Obtaining applicable data in the toxicological field using a multi-biomarker approach to investigate the modulation of NS administration via target markers in the physiological pathway of O. mykiss were aimed.As a result of CYP application, it was determined that the Ucrit value of O. mykiss decreased significantly. It was determined that the changes in the values of RBC, Hgb and Hct, which are among the hematology parameters examined in the blood tissue, were statistically significant (p < 0.05). It was determined that WBC value was inhibited by CYP application and NS tried to make a positive contribution to WBC. It was determined that the AChE activity of O. mykiss in the brain tissue had a statistically significant inhibition in the CYP-treated group (p < 0.05). SOD, CAT, GPx, enzyme activities were found to be inhibited by CYP application and were statistically significant (p < 0.05). Acute toxicity of CYP was determined by antioxidant enzyme biomarkers in gill tissue. In the results obtained; While inhibitions were determined in SOD, CAT, GPx activities compared to the control group, an induction occurred in MDA value.NS administration was noted to be an important modulator of the SOD-CAT system against CYP exposure at both concentrations. Thus, it can be said that it indirectly functions as an effective antioxidant through the NS receptor protein and structurally stimulates the synthesis and activity of antioxidative enzymes under oxidative stress.

11.
Transplant Cell Ther ; 30(4): 404-414, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38281589

ABSTRACT

Prolonged hematotoxicity is the most common long-term adverse event in chimeric antigen receptor T cell therapy (CAR-T). To evaluate the impact on prolonged cytopenia of inflammatory status after CAR T infusion, we performed a single-center retrospective study and analyzed patients with B cell lymphomas after CAR-T. Among 90 patients analyzed at 90 days after infusion, the cumulative incidence was 57.5% for prolonged neutropenia, 36.7% for anemia, and 49.8% for thrombocytopenia. Patients who experienced cytokine release syndrome (CRS) had significantly higher incidence and longer duration of prolonged cytopenia. In addition, we found that among patients with grade 1 CRS, those with a longer duration of CRS-related symptoms (>5 days; grade 1b in modified CRS grading [m-CRS]) had a significantly higher incidence and longer duration of prolonged cytopenia than those whose CRS-related symptoms resolved within 5 days (grade 1a m-CRS). Multivariate analysis revealed that a higher m-CRS grade (grade 1b or 2; hazard ratio [HR], 2.42), higher peak CRP (≥10 mg/dL; HR, 1.66), longer duration of elevated CRP (≥10 days; HR, 1.83), and a decrease in serum inorganic phosphorus concentration (≥30% from baseline; HR, 1.95) were associated with significantly higher cumulative incidence of prolonged neutropenia, as well as anemia and thrombocytopenia. Using these factors, we developed a new predictive scoring model for prolonged hematotoxicity, the KyoTox a-score, which can successfully stratify the incidence and duration of cytopenia independent of the existing model, CAR-HEMATOTOX, which is based on laboratory data at lymphodepletion. Thus, this newly developed post-CAR-T inflammation-dependent score is accurate and useful for predicting prolonged hematotoxicity.


Subject(s)
Anemia , Cytopenia , Neutropenia , Receptors, Chimeric Antigen , Thrombocytopenia , Humans , Cytokine Release Syndrome/etiology , Retrospective Studies , Cell- and Tissue-Based Therapy
12.
Transplant Cell Ther ; 30(1): 38-55, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37821079

ABSTRACT

Chimeric antigen receptor (CAR) T cell (CAR-T) therapy has emerged as a revolutionary cancer treatment modality, particularly in children and young adults with B cell malignancies. Through clinical trials and real-world experience, much has been learned about the unique toxicity profile of CAR-T therapy. The past decade brought advances in identifying risk factors for severe inflammatory toxicities, investigating preventive measures to mitigate these toxicities, and exploring novel strategies to manage refractory and newly described toxicities, infectious risks, and delayed effects, such as cytopenias. Although much progress has been made, areas needing further improvements remain. Limited guidance exists regarding initial administration of tocilizumab with or without steroids and the management of inflammatory toxicities refractory to these treatments. There has not been widespread adoption of preventive strategies to mitigate inflammation in patients at high risk of severe toxicities, particularly children. Additionally, the majority of research related to CAR-T toxicity prevention and management has focused on adult populations, with only a few pediatric-specific studies published to date. Given that children and young adults undergoing CAR-T therapy represent a unique population with different underlying disease processes, physiology, and tolerance of toxicities than adults, it is important that studies be conducted to evaluate acute, delayed, and long-term toxicities following CAR-T therapy in this younger age group. In this pediatric-focused review, we summarize key findings on CAR-T therapy-related toxicities over the past decade, highlight emergent CAR-T toxicities, and identify areas of greatest need for ongoing research.


Subject(s)
Receptors, Chimeric Antigen , Humans , Child , Receptors, Chimeric Antigen/therapeutic use , Receptors, Antigen, T-Cell , T-Lymphocytes , Immunotherapy, Adoptive/adverse effects , Risk Factors
13.
Mar Environ Res ; 193: 106294, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38096712

ABSTRACT

Contamination of the aquatic environment with different insecticides is a major concern in the aquatic ecosystem today. For this reason, in the designed study, Thiamethoxam (TMX) for which there is limited information on its negative effects on Oncorhynchus mykiss was investigated, its effects on hematotoxicity, oxidative status, cytotoxicity, DNA damage and apoptotic status indicators in blood/liver tissue. However, the antitoxic potential of ulexite (UX) supplementation in the elimination of TMX-mediated toxicity has been determined. LC50-96h value determined for TMX 0.73 mg/L has been determined. As a result of hematology profile, TMX application, RBC, Hgb and Hct values showed a temporal decrease compared to the control group, while increases were determined in MCV, MCH and MCHC values. It was determined that the inhibition/induction of hematological parameters was slowed down by adding UX to the medium. During the trial (48th and 96th hours), it was noted that TMX induced cortisol level, while UX supplementation slowed this induction at 48th hour. Antioxidant enzyme activities were significantly inhibited by TMX application, and MDA and MPO values increased as a result of the stimulation of ROS. It was determined that UX added to the medium showed activity in favor of antioxidants and tried to inhibit MDA and MPO levels. When Nrf-2, one of the inflammation parameters, was compared with the administration and control groups, it was determined that it inhibited depending on time, TNF-α, IL-6, DNA damage and apoptosis were induced, and UX suppressed this situation. The results obtained were evaluated as statistically meaningful. Briefly, it was determined that TMX induced oxidative damage in all tissues at 48th - 96th hours, whereas UX mitigated this situation. The results provide possible in vivo evidence that UX supplements can reduce TMX-mediated oxidative stress and tissues damage in O. mykiss blood and liver tissues.


Subject(s)
Chemical and Drug Induced Liver Injury , Insecticides , Humans , Thiamethoxam/toxicity , Ecosystem , Oxidative Stress , Antioxidants , Insecticides/toxicity
14.
Vet World ; 16(11): 2186-2191, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38152275

ABSTRACT

Background and Aim: Cisplatin (CP) is a preferred drug for cancer treatment but it has dose-dependent side effects. Vitex agnus-castus (VAC) berry extract has antioxidant, free-radical scavenging, and anti-inflammatory activities. This study explored the mitigating effects of VAC extract (VACE) on acute hematotoxicity induced by CP in female Wistar rats. Materials and Methods: Female Wistar rats (n = 30) were randomly divided into five groups (n = 6/group). The normal control (NC) group received no treatment. The CP control group received CP (7 mg/kg.b.w. ip, single dose) and the drug control group (VACE-650) received VACE (650 mg/kg b.w. oral, daily) for 7 days. Both groups received a single dose of CP (7 mg/kg b.w. ip), followed by 350 and 650 mg/kg.b.w. of VACE daily orally (CPVACE-350 and CPVACE-650 groups, respectively) for 7 days. Results: After a single dose of CP (7 mg/kg b.w.), the red blood cells (RBC), hematocrit (HCT), white blood cells (WBC), and platelets significantly decreased. In the VAC-350 group, the reduction in total WBC count was less than that in the VAC-650 group on the 3rd day. The RBC and HCT values of the VACE groups were better than that of the CP control, but the VACE-350 treatment group showed significant improvement only on the 3rd day. Conclusion: Our findings showed that VACE can mitigate CP-induced damage to peripheral blood cells at lower doses.

15.
Cureus ; 15(10): e46577, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37936991

ABSTRACT

Background In line with the growing industrial applications of copper oxide nanoparticles (CuONPs) in various fields, concerns about their potentially harmful consequences on the environment, human, and animal health are increasing. Giloy is considered an alternative medicine to treat various ailments. Giloy's potential in helping manage diabetes, alleviating arthritis and joint pain, and addressing skin disorders such as eczema and acne underscores its multifaceted role in traditional medicine. Moreover, it is deemed beneficial for reducing stress and anxiety levels, promoting liver health, and potentially impacting heart health by regulating cholesterol levels. Emerging research also explores its potential in cancer prevention. This study aimed to evaluate the hematotoxicity of CuONPs and the alleviating effect of giloy in adult rats. Materials and methods In this experiment, 28 laboratory rats were used, set to four groups (7/group), as follows: control group without any dose; CuONPs group administered copper oxide nanoparticles at 300 mg/kg/day; CuONPs + giloy group dosed with CuONPs at 300 mg/kg/day plus giloy at 100 mg/kg/day; giloy group treated only with giloy at 100 mg/kg/day. All treatments were given by gastric gavage and continued for 28 uninterrupted days. Results Dosing animals with CuONPs led to significant adverse changes in the examined blood profile. In contrast, when the animals were coadministered with giloy, restoring the disturbed blood levels was observed. Conclusion Copper oxide nanoparticles at a high dose had notable hematotoxicity in laboratory rats and, supplemented with giloy, could reduce this hematological toxicity.

16.
Cancer Cell ; 41(11): 1871-1891.e6, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37802054

ABSTRACT

Acute myeloid leukemia (AML) poses a singular challenge for chimeric antigen receptor (CAR) therapy owing to its phenotypic heterogeneity and similarity to normal hematopoietic stem/progenitor cells (HSPCs). Here we expound a CAR strategy intended to efficiently target AML while minimizing HSPC toxicity. Quantification of target expression in relapsed/refractory patient samples and normal HSPCs reveals a therapeutic window for gated co-targeting of ADGRE2 and CLEC12A: We combine an attenuated ADGRE2-CAR with a CLEC12A-chimeric costimulatory receptor (ADCLEC.syn1) to preferentially engage ADGRE2posCLEC12Apos leukemic stem cells over ADGRE2lowCLEC12Aneg normal HSPCs. ADCLEC.syn1 prevents antigen escape in AML xenograft models, outperforms the ADGRE2-CAR alone and eradicates AML despite proximate myelopoiesis in humanized mice. Off-target HSPC toxicity is similar to that of a CD19-CAR and can be mitigated by reducing CAR T cell-derived interferon-γ. Overall, we demonstrate the ability of target density-adapted cooperative CAR targeting to selectively eliminate AML and potentially obviate the need for hematopoietic rescue.


Subject(s)
Leukemia, Myeloid, Acute , T-Lymphocytes , Humans , Animals , Mice , Cell Line, Tumor , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/metabolism , Immunotherapy, Adoptive , Hematopoietic Stem Cells , Receptors, Mitogen/metabolism , Lectins, C-Type
17.
Food Chem Toxicol ; 180: 114030, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37689099

ABSTRACT

Long-term benzene exposure is harmful and causes hematopoietic dysfunction. However, the mechanism of benzene hematopoietic toxicity is still unclear. Acyl-CoA Synthetase Long-Chain Family Member 1 (ACSL1) has been found to participate in the progress of a variety of benign and malignant diseases, but there is no research about its effect on benzene-induced hematopoietic toxicity. Herein, We exposed C57BL/6J mice to benzene to construct an in vivo model. Human peripheral blood mononuclear cells (THP-1 cells) were treated with benzene metabolite 1, 4-BQ to construct an in vitro model. We observed that the ACSL1 expression was upregulated both in vivo and in vitro. Moreover, inhibition of ACSL1 relieved inflammation and senescence development in vitro, suggesting that ACSL1 mediates inflammation and senescence. As for the regulation mechanism of ACSL1 expression, it is closely related to hydroxymethylation modification. This was proved by hydroxymethylated DNA immunoprecipitation (hMeDIP) experiments. Furthermore, oxidative stress influenced the hydroxymethylation process. These results showed that benzene hematopoietic toxicity occurs through the induction of oxidative stress and thus the regulation of ACSL1 hydroxymethylation, which in turn mediates inflammation and senescence. Thus, this study might be of great significance in identifying and preventing benzene exposure in the early stage.


Subject(s)
Benzene , Leukocytes, Mononuclear , Mice , Animals , Humans , Benzene/toxicity , Leukocytes, Mononuclear/metabolism , Mice, Inbred C57BL , Inflammation/chemically induced , Oxidative Stress , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism
18.
Curr Oncol ; 30(7): 6330-6352, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37504327

ABSTRACT

Multiple myeloma is the second-most common hematologic malignancy in adults worldwide. Despite ongoing advancement in therapeutic modalities, it remains an incurable disease with a 5-year survival rate of approximately 50%. The recent development and introduction of anti-BCMA immunotherapies into clinical practice, including chimeric antigen receptor T-cell (CAR-T) therapies and bispecific antibodies, has radically shifted the treatment paradigm. However, despite the promising potential of these therapies for broader application, frequent and significant adverse effects have been reported, both in short- and in long-term settings, requiring increasing awareness and vigilance in the treating team, close monitoring, and prompt interventions with a multidisciplinary approach. In this review, we will discuss the toxicities associated with CAR-T cell and bispecific antibody therapies, focusing on results from major clinical studies and real-world observations. In addition, we will emphasize on effective strategies for prevention, monitoring and management, and provide expert recommendations.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes , Multiple Myeloma/drug therapy , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods
19.
Environ Int ; 178: 108113, 2023 08.
Article in English | MEDLINE | ID: mdl-37506515

ABSTRACT

Recent population and animal studies have revealed a correlation between fat content and the severity of benzene-induced hematologic toxicity. However, the precise impact of lipid deposition on benzene-induced hematotoxicity and the underlying mechanisms remain unclear. In this study, we established a mouse model with moderate lipid accumulation by subjecting the mice to an 8-week high-fat diet (45% kcal from fat, HFD), followed by 28-day inhalation of benzene at doses of 0, 1, 10, and 100 ppm. The results showed that benzene exposure caused a dose-dependent reduction of peripheral white blood cell (WBC) counts in both diet groups. Notably, this reduction was less pronounced in the HFD-fed mice, suggesting that moderate lipid accumulation mitigates benzene-related hematotoxicity. To investigate the molecular basis for this effect, we performed bioinformatics analysis of high-throughput transcriptome sequencing data, which revealed that moderate lipid deposition alters mouse metabolism and stress tolerance towards xenobiotics. Consistently, the expression of key metabolic enzymes, such as Cyp2e1 and Gsta1, were upregulated in the HFD-fed mice upon benzene exposure. Furthermore, we utilized a real-time exhaled breath detection technique to monitor exhaled benzene metabolites, and the results indicated that moderate lipid deposition enhanced metabolic activation and increased the elimination of benzene metabolites. Collectively, these findings demonstrate that moderate lipid deposition confers reduced susceptibility to benzene-induced hematotoxicity in mice, at least in part, by accelerating benzene metabolism and clearance.


Subject(s)
Benzene , Leukocytes , Mice , Animals , Benzene/toxicity , Acceleration , Lipids , Lipid Metabolism
20.
Environ Int ; 177: 108007, 2023 07.
Article in English | MEDLINE | ID: mdl-37290291

ABSTRACT

BACKGROUND: Impairment of the hematopoietic system is one of the primary adverse health effects from exposure to benzene. We previously have shown that exposure to benzene at low levels (<1 ppm) affects the blood forming system and that these effects were proportionally stronger at lower versus higher levels of benzene exposure. This observation is potentially explained by saturation of enzymatic systems. METHODS: Here we extend these analyses by detailed modeling of the exposure response association of benzene and its major metabolites (i.e. catechol, muconic acid, phenol, and hydroquinone) on peripheral white blood cell (WBC) counts and its major cell-subtypes (i.e. granulocytes, lymphocytes, and monocytes) using two previously published cross-sectional studies among occupationally exposed Chinese workers. RESULTS: Supra-linear exposure response associations were observed between air benzene concentrations (range âˆ¼ 0.1 - 100 ppm) and WBC counts and its cell-subtypes, with a larger than proportional decrease in cell counts at lower than at higher levels of benzene exposure. The hematotoxicity associations were largely similar in shape when the analyses were repeated with benzene urinary metabolites suggesting that enzymatic saturation is not a full explanation of the observed non-linearity with WBC endpoints. DISCUSSION: We hypothesize that the flattening of the exposure response curve especially at higher benzene exposure levels may reflect a response by the bone marrow to maintain hematopoietic homeostasis. Toxicity to the bone marrow and an induced hyper-proliferative response could both contribute to risk of subsequently developing a hematopoietic malignancy. Additional work is needed to explore this hypothesis.


Subject(s)
Benzene , Occupational Exposure , Humans , Benzene/toxicity , Benzene/analysis , Occupational Exposure/adverse effects , Occupational Exposure/analysis , Cross-Sectional Studies , East Asian People , Phenols/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...