Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Sci Rep ; 14(1): 8350, 2024 04 09.
Article in English | MEDLINE | ID: mdl-38594383

ABSTRACT

This study aimed to evaluate the sensitivity of AI in screening acute leukemia and its capability to classify either physiological or pathological cells. Utilizing an acute leukemia orientation tube (ALOT), one of the protocols of Euroflow, flow cytometry efficiently identifies various forms of acute leukemia. However, the analysis of flow cytometry can be time-consuming work. This retrospective study included 241 patients who underwent flow cytometry examination using ALOT between 2017 and 2022. The collected flow cytometry data were used to train an artificial intelligence using deep learning. The trained AI demonstrated a 94.6% sensitivity in detecting acute myeloid leukemia (AML) patients and a 98.2% sensitivity for B-lymphoblastic leukemia (B-ALL) patients. The sensitivities of physiological cells were at least 80%, with variable performance for pathological cells. In conclusion, the AI, trained with ResNet-50 and EverFlow, shows promising results in identifying patients with AML and B-ALL, as well as classifying physiological cells.


Subject(s)
Deep Learning , Leukemia, Myeloid, Acute , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Retrospective Studies , Flow Cytometry/methods , Artificial Intelligence , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/pathology , Acute Disease , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Immunophenotyping
2.
Cancers (Basel) ; 16(6)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38539418

ABSTRACT

BACKGROUND: Allogeneic hematopoietic stem cell transplantation (HSCT) is rarely recommended for unfit patients with newly diagnosed acute myeloid leukemia (AML). Patient survival can improve with venetoclax plus azacitidine (VEN plus AZA). However, the long-term outcome of this treatment strategy is still unsatisfactory. The high response and low treatment toxicity rates of patients receiving VEN plus AZA can provide an opportunity for HSCT among unfit patients. Nevertheless, the outcomes and complications of VEN plus AZA, followed by HSCT, remain unclear. METHODS: This single-center retrospective study aimed to compare patients with newly diagnosed AML receiving VEN plus AZA as induction therapy (n = 27) to those receiving the conventional I3A7 regimen as induction therapy (n = 34). RESULT: The 1-year overall survival, relapse, and non-relapse mortality rates in the two groups were similar. The cytogenetic risks and the hematopoietic cell transplantation-specific comorbidity index are the most significant predictive factors of overall survival. CONCLUSION: In older patients unfit for intensive chemotherapy, a low-intensity regimen with VEN plus AZA is a suitable bridge therapy. Furthermore, allo-HSCT is feasible and can be a curative option.

3.
Hematol Rep ; 15(1): 50-56, 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36648883

ABSTRACT

Myelodysplastic syndrome (MDS) immunity plays an important role in the proliferation and apoptosis of aberrant cells. Immune dysregulation has been studied in various prognostic subgroups. This study analyzed 60 patients with MDS via multidimensional flow cytometry to evaluate the expression of aberrant markers, such as CD7 and cytoplasmic CD3 on lymphocytes. The Revised International Prognostic Scoring System (IPSS-R) scores were used to classify the patients into risk groups. The results showed a significant downregulation of CyCD3- T cells in low-intermediate versus high-risk patients (p = 0.013). This study is the first to show that a significant decrease in cyCD3- T cells in patients with a lower IPSS-R score may indicate microenvironmental changes conducive to transformation in MDS.

4.
BMC Cancer ; 22(1): 169, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35168547

ABSTRACT

BACKGROUND: The treatment of recurrent or metastatic head and neck squamous-cell carcinoma (R/M HNSCC) remains challenging. Preclinical studies revealed that B cell depletion could modulate the microenvironment and overcome chemoresistance. We conducted a phase I study to evaluate the feasibility and safety of B cell depletion using the anti-CD20 antibody rituximab to treat HNSCC. METHODS: Ten patients were enrolled in two protocols. The first four patients treated using protocol 1 received rituximab 1000 mg on days -14 and -7, followed by gemcitabine/cisplatin every 3 weeks, and rituximab was administered every 6 months thereafter. Because of disease hyperprogression, protocol 1 was amended to protocol 2, which consisted of the concomitant administration of rituximab 375 mg/m2 and gemcitabine/cisplatin every 3 weeks. Another six patients were enrolled and treated using protocol 2. RESULTS: Three patients treated using protocol 1 exhibited rapid disease progression, and the remaining patient could not undergo evaluation after rituximab treatment. Conversely, no unpredicted harm was observed in the six patients treated using protocol 2. Among these patients, one achieved complete response, and two had partial responses. The disease-free durations in these patients were 7.0, 6.2, and 7.1 months, respectively. Immune cell analysis revealed a higher ratio of cytotoxic T cells to regulatory T cells in responders than in non-responders. CONCLUSIONS: B cell depletion using rituximab alone in patients with HNSCC can cause hyperprogressive disease. Contrarily, the co-administration of rituximab and cisplatin/gemcitabine was feasible and safe. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04361409 , 24 April 2020, retrospectively registered.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cisplatin/administration & dosage , Deoxycytidine/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Rituximab/administration & dosage , Squamous Cell Carcinoma of Head and Neck/drug therapy , Adult , Deoxycytidine/administration & dosage , Disease-Free Survival , Drug Administration Schedule , Feasibility Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Pilot Projects , Treatment Outcome , Gemcitabine
5.
Int J Hematol ; 115(4): 525-533, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35226308

ABSTRACT

BACKGROUND: Post-transplant cyclophosphamide (PT-Cy) and calcineurin inhibitors used in peripheral blood haplo-identical stem cell transplantation (haploSCT) increase the risk of acute/chronic graft-versus-host disease (GVHD). Whether ATG/PT-Cy is feasible for Asian patients undergoing haploSCT is unclear, and an optimal strategy for GVHD prophylaxis is needed. MATERIALS AND METHODS: We retrospectively analyzed 61 hematologic malignancy patients who underwent peripheral blood haploSCT using ATG/PT-Cy from January 2013 to December 2018. We also compared the effects of ATG/PT-Cy (ATG group; n = 61) with historical data from patients who underwent haploSCT using sirolimus/PT-Cy (non-ATG group; n = 22). RESULTS: Cumulative incidences of grades II-IV acute GVHD and moderate to severe chronic GVHD did not differ significantly. The ATG group had higher incidence of Epstein-Barr virus (EBV) reactivation, but neither group had post-transplant lymphoproliferative disorders. The ATG group also had a higher OS rate (2-year OS in ATG group vs. non-ATG group: 43.4% vs. 27.3%, respectively; P = 0.071). CONCLUSION: ATG/PT-Cy is an acceptable strategy for GVHD prophylaxis in Asian patients undergoing haploSCT.


Subject(s)
Epstein-Barr Virus Infections , Graft vs Host Disease , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Peripheral Blood Stem Cell Transplantation , Antilymphocyte Serum/therapeutic use , Cyclophosphamide/therapeutic use , Epstein-Barr Virus Infections/drug therapy , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Herpesvirus 4, Human , Humans , Peripheral Blood Stem Cell Transplantation/adverse effects , Retrospective Studies , Transplantation Conditioning
6.
Ann Hematol ; 97(4): 697-707, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29349655

ABSTRACT

Total body irradiation (TBI) is frequently used in hematopoietic stem cell transplantation (HSCT) and is associated with many complications due to radiation injury to the normal cells, including normal stem cells. Nevertheless, the effects of TBI on the mesenchymal stromal stem cell (MSC) are not fully understood. Bone marrow-derived MSCs (BM-MSCs) isolated from normal adults were irradiated with 200 cGy twice daily for consecutive 3 days, a regimen identical to that used in TBI-conditioning HSCT. The characteristics, differentiation potential, cytogenetics, hematopoiesis-supporting function, and carcinogenicity of the irradiated BM-MSCs were then compared to the non-irradiated control. The irradiated and non-irradiated MSCs shared similar morphology, phenotype, and hematopoiesis-supporting function. However, irradiated MSCs showed much lower proliferative and differentiative potential. Irradiation also induced clonal cytogenetic abnormalities of MSCs. Nevertheless, the carcinogenicity of irradiated MSCs is low in vitro and in vivo. In parallel with the ex vivo irradiation experiments, decreased proliferative and differentiative abilities and clonal cytogenetic abnormalities can also be found in MSCs isolated from transplant recipients who had received TBI-based conditioning previously. Thus, TBI used in HSCT drastically injury MSCs and may contribute to the development of some long-term complications associated with clonal cytogenetic abnormality and poor adipogenesis and osteogenesis after TBI.


Subject(s)
Apoptosis/radiation effects , Bone Marrow Cells/radiation effects , Chromosome Aberrations/radiation effects , Hematopoietic Stem Cells/radiation effects , Mesenchymal Stem Cells/radiation effects , Radiation Injuries/pathology , Whole-Body Irradiation/adverse effects , Adult , Adult Stem Cells/radiation effects , Bone Marrow Cells/cytology , Bone Marrow Cells/pathology , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , China , Chromosome Disorders/etiology , Chromosome Disorders/pathology , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Hospitals, University , Humans , Leukemia/pathology , Leukemia/therapy , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/pathology , Necrosis , Radiation Injuries/etiology , Transplantation Conditioning/adverse effects , Tumor Cells, Cultured , Young Adult
7.
Leuk Lymphoma ; 58(9): 1-12, 2017 09.
Article in English | MEDLINE | ID: mdl-28140713

ABSTRACT

Deferasirox (DFX), in addition to its iron-chelation property, has marked anti-proliferative effects on cancer cells. However, the activity and mechanism by which DFX inhibits acute myeloid leukemia (AML) cells remain to be elucidated. Furthermore, the anti-leukemia effect of combining DFX with currently recommended agents doxorubicin (DOX) and cytosine arabinoside (Ara-C) has not been studied. In this study, we show that DFX significantly reduces the viability of three AML cell lines, HL60, THP1, and WEHI3 and two primary leukemic cells harvested from AML patients. DFX induces cell cycle arrest at G1 phase and apoptosis and inhibits phosphorylation of ERK. We also showed that DFX antagonizes the anti-leukemic effect of DOX. On the contrary, combining DFX with Ara-C created a synergistic effect. Our study confirms the anti-leukemia activity of DFX and provides important information on how to select a partner drug for DFX for the treatment of AML in future clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Doxorubicin/pharmacology , Triazoles/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deferasirox , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Iron/metabolism , Leukemia/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism
8.
In Vivo ; 30(3): 265-9, 2016.
Article in English | MEDLINE | ID: mdl-27107085

ABSTRACT

Mounting evidence supports the notion that inflammatory processes play a role in carcinogenesis, and interleukin-10 (IL10) is an important inflammatory cytokine. This study aimed to evaluate the contribution of IL10 A-1082G (rs1800896), T-819C (rs3021097) and A-592C (rs1800872) genotypes to the risk of childhood acute lymphoblastic leukemia (ALL) in Taiwan. Associations of these IL10 polymorphic genotypes with ALL risk were analyzed in 266 patients with childhood ALL patients and 266 non-cancer healthy controls by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) methodology. The results showed that CC genotype carriers at IL10 T-819C were at lower risk for childhood ALL (odds ratio=0.33, 95% confidence interval=0.16-0.68). On the contrary, AC and CC genotype carriers at IL10 A-592C were at higher risk for childhood ALL (odds ratio=1.73 and 6.34, 95% confidence interval=1.19-2.51 and 3.16-12.72, respectively). There was no difference in the distribution of A-1082G genotypes between childhood ALL and control groups. The genotypes at IL10 T-819C and A-592C may serve as predictive biomarkers for childhood ALL in Taiwan.


Subject(s)
Genetic Predisposition to Disease/genetics , Interleukin-10/genetics , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Asian People/genetics , Case-Control Studies , Child , Child, Preschool , Female , Gene Frequency , Genetic Predisposition to Disease/ethnology , Genotype , Humans , Male , Odds Ratio , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Precursor Cell Lymphoblastic Leukemia-Lymphoma/ethnology , Risk Factors , Taiwan
10.
Blood ; 121(8): 1357-66, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23243276

ABSTRACT

Adult patients with disseminated nontuberculous mycobacterial (dNTM) infections usually have severe immune system defects. Recently, several studies have shown that anti-IFN-γ autoantibodies may play an important role in the pathogenicity of dNTM infections. A considerable proportion of reported cases of anti-IFN-γ autoantibodies show either clinical or laboratory evidence of autoimmune disease. In the present study, we identified 19 formerly healthy adults who later developed dNTM infections, of whom 17 were further investigated immunologically. High-titer anti-IFN-γ autoantibodies capable of inhibiting IL-12 production in vitro were found in the plasma of all of these patients. In addition to dNTM infection, 35% and 71% of our patients also suffered from salmonellosis and herpes zoster, respectively. This observation suggests that IFN-γ may be crucial in controlling salmonella infection and reactivating latent varicella-zoster virus infection in humans. 2 HLA alleles, DRB1*16:02 DQB1*05:02 (odds ratio 8.68; 95% confidence interval, 3.47-21.90; P = 1.1 × 10(-6); Pc = 3.08 × 10(-5) and odds ratio 7.16; 95% confidence interval, 3.02-17.05; P = 1 × 10(-7); Pc = 1.4 × 10(-6), respectively), were found in 82% (14 of 17) of our patients. In conclusion, our data suggest that anti-IFN-γ autoantibodies may play a critical role in the pathogenesis of dNTM infections and reactivation of latent varicella-zoster virus infection and are associated with HLA-DRB1*16:02 and HLA-DQB1*05:02.


Subject(s)
Autoantibodies/immunology , HLA-DQ beta-Chains/immunology , HLA-DRB1 Chains/immunology , Herpes Zoster/immunology , Interferon-gamma/immunology , Mycobacterium Infections, Nontuberculous/immunology , Aged , Aged, 80 and over , Autoantibodies/blood , Autoantibodies/genetics , Coinfection/genetics , Coinfection/immunology , Coinfection/mortality , Female , Gene Frequency , HLA-DQ beta-Chains/genetics , HLA-DRB1 Chains/genetics , Herpes Zoster/genetics , Herpes Zoster/mortality , Herpesvirus 3, Human/immunology , Histocompatibility Testing , Humans , Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interferon-gamma/blood , Interleukin-12 Subunit p40/blood , Interleukin-12 Subunit p40/immunology , Male , Middle Aged , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/mortality , Seroepidemiologic Studies , Virus Latency/immunology
11.
PLoS One ; 7(9): e44416, 2012.
Article in English | MEDLINE | ID: mdl-22957070

ABSTRACT

BACKGROUND: CD4(+)interferon (IFN)-γ(+) T cell (Th1) and CD4(+)interleukin (IL)-4(+) T cell (Th2) polarizations are crucial in the pathogenesis of graft-versus-host disease (GVHD). However, this hypothesis is largely based on animal experiments of Parent-into-F1 GVHD model. The causal relationship between kinetics of Th1, Th2 and associated cytokines and the clinical activity of GVHD in a real world situation remains unknown. METHODOLOGY: Peripheral blood was collected every week prospectively from Day 0 to Day 210 (patients without GVHD) or Day 300 (patients with chronic GVHD) after allogeneic peripheral blood stem cell transplantation in consecutive 27 patients. The frequencies of Th1 and Th2 within CD4(+) T cells were determined by flow cytometry and pplasma IFN-γ, IL-12, IL-4, and IL-10 were determined by ELISA. PRINCIPAL FINDINGS: Kinetics of Th1, Th2 frequency, and the plasma IL-10 and IFN-γ more commonly coincided with, rather than predicted, the activity of GVHD. These markers are significantly higher when acute or chronic GVHD developed. The kinetics of IL-10 is especially correlated well with the activity of GVHD during clinical course of immunosuppressive treatment. For patients with hepatic GVHD, there is a positive correlation between plasma IL-10 levels and the severity of hepatic injury. The frequency of Th2 is also significant higher in acute GVHD and tends to be higher in chronic GVHD. Interestingly, there is a very good positive correlation between the frequency of Th1 and Th2 (r = 0.951, p<0.001). The plasma level of IL-4 and IL-12 are not associated with the activity of GVHD. CONCLUSIONS: The frequency of Th1, Th2 within CD4(+) T cells and plasma IL-10 and IFN-γ are good biomarkers of GVHD. Plasma IL-10 can also be used to monitor the therapeutic responsiveness. Furthermore, both Th1 and Th2 likely contribute to the pathogenesis of GVHD.


Subject(s)
Cytokines/metabolism , Graft vs Host Disease/metabolism , Interferons/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Animals , Biomarkers/metabolism , Flow Cytometry/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-4/metabolism , Kinetics , Th1 Cells/cytology , Th2 Cells/cytology , Time Factors
12.
Ann Hematol ; 91(2): 163-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21573981

ABSTRACT

Both bone marrow hematopoietic cells (BM-HCs) and mesenchymal stem cells (BM-MSCs) may have cytogenetic aberrations in leukemic patients, and anti-leukemic therapy may induce cytogenetic remission of BM-HCs. The impact of anti-leukemic therapy on BM-MSCs remains unknown. Cytogenetic studies of BM-MSCs from 15 leukemic patients with documented cytogenetic abnormalities of BM-HCs were investigated. To see the influence of anti-leukemic therapy on BM-MSCs, cytogenetic studies were carried out in seven of them after the completion of anti-leukemic therapy, including anthracycline/Ara-C-based chemotherapy in two patients, high-dose busulfan/cyclophosphamide-based allogeneic transplantation in two patients, and total body irradiation (TBI)-based allogeneic transplantation in three patients. To simulate the effect of TBI in vitro, three BM-MSCs from one leukemic patient and two normal adults were irradiated using the same dosage and dosing schedule of TBI and cytogenetics were re-examined after irradiation. At the diagnosis of leukemia, two BM-MSCs had cytogenetic aberration, which were completely different to their BM-HCs counterpart. After the completion of anti-leukemic therapy, cytogenetic aberration was no longer detectable in one patient. Unexpectedly, BM-MSCs from three patients receiving TBI-based allogeneic transplantation acquired new, clonal cytogenetic abnormalities after transplantation. Similarly, complex cytogenetic abnormalities were found in all the three BM-MSCs exposed to in vitro irradiation. In conclusion, anti-leukemic treatments induce not only "cytogenetic remission" but also new cytogenetic abnormalities of BM-MSCs. TBI especially exerts detrimental effect on the chromosomal integrity of BM-MSCs and highlights the equal importance of investigating long-term adverse effect of anti-leukemic therapy on BM-MSCs as opposed to beneficial effect on BM-HCs.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Marrow Cells , Chromosome Aberrations , Leukemia/therapy , Mesenchymal Stem Cells , Transplantation, Homologous/adverse effects , Whole-Body Irradiation/adverse effects , Adolescent , Adult , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , Cytogenetics/methods , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Humans , Karyotype , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/radiation effects , Middle Aged , Remission Induction , Young Adult
13.
Leuk Lymphoma ; 53(6): 1196-202, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22080758

ABSTRACT

There is little evidence to demonstrate the importance of the Sonic hedgehog homolog (Shh) pathway to differentiation therapy in the treatment of hematological neoplasms. Here we characterize the changes in acute myelogenous leukemia (HL-60) cells after blocking the Shh pathway by an antagonist of Smoothened, cyclopamine. Cyclopamine induces apoptosis of HL-60 cells in a dose- and time-dependent manner with increased G0/G1 cycle fraction. Treatment with cyclopamine increases the expression of monocytic cell markers CD11b and CD14, but the expression of CD13, CD33 and CD38 is unchanged. The monocytic differentiation of HL-60 cells induced by cyclopamine is also evidenced by an increase in Egr-1 expression. Importantly, cyclopamine down-regulates the phosphorylation of Akt and ERK, but activates AMP-activated protein kinase (AMPK) signaling. Further investigations should determine the clinical application of modulating the Shh pathway in the treatment of hematological malignancies.


Subject(s)
Cell Differentiation/drug effects , Hedgehog Proteins/antagonists & inhibitors , Monocytes/drug effects , Veratrum Alkaloids/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/genetics , Drug Evaluation, Preclinical , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Monocytes/metabolism , Monocytes/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smoothened Receptor , Transcription Factors/genetics , Transcription Factors/metabolism , Validation Studies as Topic , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
SELECTION OF CITATIONS
SEARCH DETAIL