Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Hematol ; 99(4): 662-678, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38197307

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy, an innovative immunotherapeutic against relapsed/refractory B-cell lymphoma, faces challenges due to frequent viral infections. Despite this, a comprehensive review addressing risk assessment, surveillance, and treatment management is notably absent. This review elucidates immune response compromises during viral infections in CAR-T recipients, collates susceptibility risk factors, and deliberates on preventive strategies. In the post-pandemic era, marked by the Omicron variant, new and severe threats to CAR-T therapy emerge, necessitating exploration of preventive and treatment measures for COVID-19. Overall, the review provides recommendations for viral infection prophylaxis and management, enhancing CAR-T product safety and recipient survival.


Subject(s)
Lymphoma, B-Cell , Receptors, Chimeric Antigen , Virus Diseases , Humans , Receptors, Chimeric Antigen/therapeutic use , Immunotherapy, Adoptive/adverse effects , Lymphoma, B-Cell/therapy , Virus Diseases/etiology , Antigens, CD19 , Cell- and Tissue-Based Therapy
2.
Front Biosci (Landmark Ed) ; 28(11): 299, 2023 11 24.
Article in English | MEDLINE | ID: mdl-38062808

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T-cell therapy carries the risk of inducing severe and life-threatening toxicities such as cytokine release syndrome (CRS), neurotoxicity, and infection. Although CRS and infections have similar symptoms, their treatment strategies differ, and early diagnosis is very important. For CRS and infections, the fastest detection time currently takes more than 24 h, so a quick and simple method to identify a fever after CAR T-cell infusion is urgently needed. METHODS: We enrolled 27 patients with recurrent fever treated with different types of CAR T-cells, including cluster of differentiation (CD) 7, CD19, CD22, and CD19-CD22 bicistronic CAR T-cells, and evaluated the infection events occurring in these patients. We detailed the morphology of CAR T-cells in peripheral blood smears (PBS) and reported the infection events, CAR transgene copy number, and inflammatory indicators within the first month after treatment. RESULTS: Similar morphological characteristics were observed in the PBS of different CAR T-cells, namely, enlarged cell bodies, deep outside and shallow inside basophilic blue cytoplasm, and natural killer (NK) cell-like purplish red granules. There were ten infections in nine of the twenty-seven patients (33%). The percentage of atypical lymphocytes in PBS was significantly associated with CAR transgene copy number and absolute lymphocyte count in all patients. The atypical lymphocyte percentage was significantly higher in the non-infection group. CONCLUSIONS: In conclusion, the unique morphology of CAR T-cells in PBS can be used to evaluate CAR T-cell kinetics and provide reliable evidence for the rapid early identification of fever after CAR T-cell infusion. CLINICAL TRIAL REGISTRATIONS: ChiCTR-OPN-16008526; ChiCTR-OPN-16009847; ChiCTR2000038641; NCT05618041; NCT05388695.


Subject(s)
Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cytokine Release Syndrome , Killer Cells, Natural , Antigens, CD19
3.
Crit Rev Oncol Hematol ; 192: 104186, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37863402

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are a specific dendritic cell type stemming from the myeloid lineage. Clinically and pathologically, neoplasms associated with pDCs are classified as blastic plasmacytoid dendritic cell neoplasm (BPDCN), mature plasmacytoid dendritic myeloid neoplasm (MPDMN) and pDC expansion in myeloid neoplasms (MNs). BPDCN was considered a rare and aggressive neoplasm in the 2016 World Health Organization (WHO) classification. MPDMN, known as mature pDC-derived neoplasm, is closely related to MNs and was first recognized in the latest 2022 WHO classification, proposing a new concept that acute myeloid leukemia cases could show clonally expanded pDCs (pDC-AML). With the advances in detection techniques, an increasing number of pDC expansion in MNs have been reported, but whether the pathogenesis is similar to that of MPDMN remains unclear. This review focuses on patient characteristics, diagnosis and treatment of pDC expansion in MNs to gain further insight into this novel and unique provisional subtype.


Subject(s)
Hematologic Neoplasms , Leukemia, Myeloid, Acute , Skin Neoplasms , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/metabolism , Immunophenotyping , Dendritic Cells/pathology , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/therapy , Hematologic Neoplasms/pathology , Skin Neoplasms/pathology
4.
Ther Adv Hematol ; 14: 20406207231183330, 2023.
Article in English | MEDLINE | ID: mdl-37547364

ABSTRACT

Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematological diseases and a high risk for transformation to acute myeloid leukemia (AML). The identification of key genetic alterations in MDS has enhanced our understanding of the pathogenesis and evolution. In recent years, it has been found that both innate and adaptive immune signaling are activated in the hematopoietic niche of MDS with aberrant cytokine secretion in the bone marrow microenvironment. It is also clear that immune dysregulation plays an important role in the occurrence and progression of MDS, especially the destruction of the bone marrow microenvironment, including hematopoiesis and stromal components. The purpose of this review is to explore the role of immune cells, the immune microenvironment, and cytokines in the pathogenesis of MDS. Insights into the mechanisms of these variants may facilitate the development of novel effective treatments to prevent disease progression.

6.
Blood Sci ; 4(4): 205-208, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36518236

ABSTRACT

Plasmablastic lymphoma (PBL) is an aggressive lymphoma with limited treatment strategies. Tuberculosis (TB) infection poses a high risk for patients with hematologic malignancies, especially those treated with immune agents but were never reported post-daratumumab treatment. Herein, we reported a TB infection in a 57-year-old male diagnosed with HIV-negative PBL receiving daratumumab-based treatment, who showed atypical lung infection and yielded Mycobacterium tuberculosis and cytomegalovirus (CMV) in the bronchoalveolar lavage fluid. Anti-TB therapy was administered, and the following daratumumab treatment was complete with good tolerance. In this case, we demonstrated that TB infection might occur after daratumumab therapy, and adequate attention should be paid to atypical symptoms.

7.
Hematol Oncol ; 40(5): 821-834, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35635796

ABSTRACT

Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy has rapidly changed current treatment pattern, providing a better option for individuals with primary refractory or relapsed B-cell non-Hodgkin lymphoma (r/r B-NHL) and B-cell acute lymphoblastic leukemia (r/r B-ALL). However, despite the outstanding efficacy, a high relapse rate is still found in some B-cell malignancies after anti-CD19 CAR T-cell therapy, which emerges as a main barrier for improving the overall response and long-term outcomes. Understanding the resistance mechanism is crucial to improve current CAR T products, better incorporate them into the current therapy system and develop novel CAR approaches. Herein, we discuss the latest advances in understanding the mechanisms limiting efficacy of CAR T-cell therapy, resulting in CD19 negative (CD19- ) and CD19 positive (CD19+ ) relapses. We also provide a whole scenario of current potential strategies to overcome these barriers.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Humans
8.
Signal Transduct Target Ther ; 7(1): 101, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35399106

ABSTRACT

TP53 gene alteration confers inferior prognosis in refractory/relapse aggressive B-cell non-Hodgkin lymphoma (r/r B-NHL). From September 2016 to September 2020, 257 r/r B-NHL patients were assessed for eligibility for two trials in our center, assessing anti-CD19 and anti-CD22 chimeric antigen receptor (CAR19/22) T-cell cocktail treatment alone or in combination with autologous stem cell transplantation (ASCT). TP53 alterations were screened in 123 enrolled patients and confirmed in 60. CAR19/22 T-cell administration resulted in best objective (ORR) and complete (CRR) response rate of 87.1% and 45.2% in patients with TP53 alterations, respectively. Following a median follow-up of 16.7 months, median progression-free survival (PFS) was 14.8 months, and 24-month overall survival (OS) was estimated at 56.3%. Comparable ORR, PFS, and OS were determined in individuals with or without TP53 alterations, and in individuals at different risk levels based on functional stratification of TP53 alterations. CAR19/22 T-cell treatment in combination with ASCT resulted in higher ORR, CRR, PFS, and OS, but reduced occurrence of severe CRS in this patient population, even in individuals showing stable or progressive disease before transplantation. The best ORR and CRR in patients with TP53 alterations were 92.9% and 82.1%, respectively. Following a median follow-up of 21.2 months, 24-month PFS and OS rates in patients with TP53 alterations were estimated at 77.5% and 89.3%, respectively. In multivariable analysis, this combination strategy predicted improved OS. In conclusion, CAR19/22 T-cell therapy is efficacious in r/r aggressive B-NHL with TP53 alterations. Combining CAR-T cell administration with ASCT further improves long-term outcome of these patients.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Lymphoma, B-Cell , Receptors, Chimeric Antigen , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Transplantation, Autologous , Tumor Suppressor Protein p53/genetics
9.
Exp Hematol Oncol ; 11(1): 11, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236415

ABSTRACT

Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell diseases arising from the bone marrow (BM), and approximately 30% of MDS eventually progress to AML, associated with increasingly aggressive neoplastic hematopoietic clones and poor survival. Dysregulated immune microenvironment has been recognized as a key pathogenic driver of MDS and AML, causing high rate of intramedullary apoptosis in lower-risk MDS to immunosuppression in higher-risk MDS and AML. Immune checkpoint molecules, including programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1), play important roles in oncogenesis by maintaining an immunosuppressive tumor microenvironment. Recently, both molecules have been examined in MDS and AML. Abnormal inflammatory signaling, genetic and/or epigenetic alterations, interactions between cells, and treatment of patients all have been involved in dysregulating PD-1/PD-L1 signaling in these two diseases. Furthermore, with the PD-1/PD-L1 pathway activated in immune microenvironment, the milieu of BM shift to immunosuppressive, contributing to a clonal evolution of blasts. Nevertheless, numerous preclinical studies have suggested a potential response of patients to PD-1/PD-L1 blocker. Current clinical trials employing these drugs in MDS and AML have reported mixed clinical responses. In this paper, we focus on the recent preclinical advances of the PD-1/PD-L1 signaling in MDS and AML, and available and ongoing outcomes of PD-1/PD-L1 inhibitor in patients. We also discuss the novel PD-1/PD-L1 blocker-based immunotherapeutic strategies and challenges, including identifying reliable biomarkers, determining settings, and exploring optimal combination therapies.

11.
Neurosci Lett ; 731: 135024, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32380142

ABSTRACT

Prepulse inhibition (PPI), a measure of sensorimotor gating, has been shown to be disrupted in several animal models of neuropsychiatric disorders, such as schizophrenia. The neural circuits involving the hippocampus and nucleus accumbens (NAC) have been studied in rats to uncover the neurochemical and neuroanatomical substrates that regulate PPI. Majority of the studies of the hippocampus on PPI to date have been focused on CA1, CA2, and dentate gyrus (DG) area. Little is known about the role of the subiculum, which maintains the hippocampal formation intact, on the sensorimotor gating. In this study, the PPI disruption was induced by intraperitoneal injection of MK-801 in rats, and the neuronal activity in the dorsal and ventral subiculum by c-Fos immunostaining was examined. The projections from the subiculum to the nucleus accumbens (NAC) were detected by retrograde tracing of cholera toxin B subunit, in the PPI dysfunctional animals. The results showed an increase in neuronal activity in the ventral subiculum (vSub) while remaining constant in the dorsal subiculum during PPI disruption. The excitatory projections from the vSub to the NAC shell were significantly enhanced when PPI was disrupted. Muscimol Inhibition of vSub could significantly ameliorate the MK801-induced PPI deficit. This data suggests that the enhancement of neuronal activity in the vSub was associated with the PPI impairment, possibly due to the enhanced excitatory output from vSub the NAC shell.


Subject(s)
Neural Pathways/physiology , Neurons/physiology , Nucleus Accumbens/physiology , Prepulse Inhibition/physiology , Animals , Dizocilpine Maleate/pharmacology , Male , Neural Pathways/drug effects , Neurons/drug effects , Prepulse Inhibition/drug effects , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Reflex, Startle/physiology
12.
PLoS One ; 7(12): e50902, 2012.
Article in English | MEDLINE | ID: mdl-23236402

ABSTRACT

Two ruthenium(II) complexes, Λ-[Ru(phen)(2)(p-HPIP)](2+) and Δ-[Ru(phen)(2)(p-HPIP)](2+), were synthesized and characterized via proton nuclear magnetic resonance spectroscopy, electrospray ionization-mass spectrometry, and circular dichroism spectroscopy. This study aims to clarify the anticancer effect of metal complexes as novel and potent telomerase inhibitors and cellular nucleus target drug. First, the chiral selectivity of the compounds and their ability to stabilize quadruplex DNA were studied via absorption and emission analyses, circular dichroism spectroscopy, fluorescence-resonance energy transfer melting assay, electrophoretic mobility shift assay, and polymerase chain reaction stop assay. The two chiral compounds selectively induced and stabilized the G-quadruplex of telomeric DNA with or without metal cations. These results provide new insights into the development of chiral anticancer agents for G-quadruplex DNA targeting. Telomerase repeat amplification protocol reveals the higher inhibitory activity of Λ-[Ru(phen)(2)(p-HPIP)](2+) against telomerase, suggesting that Λ-[Ru(phen)(2)(p-HPIP)](2+) may be a potential telomerase inhibitor for cancer chemotherapy. MTT assay results show that these chiral complexes have significant antitumor activities in HepG2 cells. More interestingly, cellular uptake and laser-scanning confocal microscopic studies reveal the efficient uptake of Λ-[Ru(phen)(2)(p-HPIP)](2+) by HepG2 cells. This complex then enters the cytoplasm and tends to accumulate in the nucleus. This nuclear penetration of the ruthenium complexes and their subsequent accumulation are associated with the chirality of the isomers as well as with the subtle environment of the ruthenium complexes. Therefore, the nucleus can be the cellular target of chiral ruthenium complexes for anticancer therapy.


Subject(s)
Enzyme Inhibitors/chemistry , G-Quadruplexes , Ruthenium Compounds/chemistry , Telomerase/antagonists & inhibitors , Cell Line, Tumor , Circular Dichroism , Enzyme Inhibitors/chemical synthesis , Humans , Magnetic Resonance Spectroscopy , Ruthenium Compounds/chemical synthesis , Stereoisomerism , Telomerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...