Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.072
Filter
1.
World J Gastroenterol ; 30(25): 3147-3151, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39006388

ABSTRACT

In this editorial, we discussed the apparent discrepancy between the findings described by Colapietro et al, in their case report and data found in the literature. Colapietro et al reported a case of hepatitis B virus (HBV)-related hepatic decompensation in a patient with chronic myeloid leukemia and a previously resolved HBV infection who was receiving Bruton's tyrosine kinase (BTK) inhibitor therapy. First of all, we recapitulated the main aspects of the immune system involved in the response to HBV infection in order to underline the role of the innate and adaptive response, focusing our attention on the protective role of anti-HBs. We then carefully analyzed literature data on the risk of HBV reactivation (HBVr) in patients with previous HBV infection who were treated with either tyrosine kinase inhibitors or BTK inhibitors for their hematologic malignancies. Based on literature data, we suggested that several factors may contribute to the different risks of HBVr: The type of hematologic malignancy; the type of therapy (BTK inhibitors, especially second-generation, seem to be at a higher risk of HBVr than those with tyrosine kinase inhibitors); previous exposure to an anti-CD20 as first-line therapy; and ethnicity and HBV genotype. Therefore, the warning regarding HBVr in the specific setting of patients with hematologic malignancies requires further investigation.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Hematologic Neoplasms , Hepatitis B virus , Protein Kinase Inhibitors , Virus Activation , Humans , Virus Activation/drug effects , Virus Activation/immunology , Hepatitis B virus/drug effects , Hepatitis B virus/immunology , Hepatitis B virus/pathogenicity , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Hematologic Neoplasms/immunology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/virology , Hepatitis B/virology , Hepatitis B/diagnosis , Hepatitis B/drug therapy , Hepatitis B/immunology , Risk Factors , Antiviral Agents/therapeutic use , Hepatitis B Antibodies/blood , Hepatitis B Antibodies/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/virology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use
3.
Blood Cancer J ; 11(11): 182, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34785653

ABSTRACT

Tyrosine kinase inhibitors (TKIs) that target BCR-ABL are the frontline treatments in chronic myeloid leukemia (CML). Growing evidence has shown that TKIs also enhance immunity. Since gamma-delta T (γδT) cells possess the potent anticancer capability, here we investigated the potential involvement of γδT cells in TKI treatments for CML. We characterized γδT cells isolated from chronic-phase CML patients before and during TKI treatments. γδT expression increased significantly in CML patients who achieved major molecular response (MMR) and deep molecular response (DMR). Their Vδ2 subset of γδT also expanded, and increased expression of activating molecules, namely IFN-γ, perforin, and CD107a, as well as γδT cytotoxicity. Mechanistically, TKIs augmented the efflux of isopentenyl pyrophosphate (IPP) from CML cells, which stimulated IFN-γ production and γδT expansion. Notably, the size of the IFN-γ+ naïve γδT population in TKI-treated CML patients was strongly correlated with their rates to reach DMR and with the duration on DMR. Statistical analysis suggests that a cutoff of 7.5% IFN-γ+ naïve subpopulation of γδT in CML patients could serve as a determinant for MR4.0 sustainability. Our results highlight γδT cells as a positive regulator for TKI responses in CML patients.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Protein Kinase Inhibitors/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Female , Fusion Proteins, bcr-abl/immunology , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , T-Lymphocytes/pathology
4.
JCI Insight ; 6(23)2021 12 08.
Article in English | MEDLINE | ID: mdl-34727093

ABSTRACT

Leukemia stem cells (LSCs) promote the disease and seem resistant to therapy and immune control. Why LSCs are selectively resistant against elimination by CD8+ cytotoxic T cells (CTLs) is still unknown. In this study, we demonstrate that LSCs in chronic myeloid leukemia (CML) can be recognized and killed by CD8+ CTLs in vitro. However, Tregs, which preferentially localized close to CD8+ CTLs in CML BM, protected LSCs from MHC class I-dependent CD8+ CTL-mediated elimination in vivo. BM Tregs in CML were characterized by the selective expression of tumor necrosis factor receptor 4 (Tnfrsf4). Stimulation of Tnfrsf4 signaling did not deplete Tregs but reduced the capacity of Tregs to protect LSCs from CD8+ CTL-mediated killing. In the BM of newly diagnosed CML patients, TNFRSF4 mRNA levels were significantly increased and correlated with the expression of the Treg-restricted transcription factor FOXP3. Overall, these results identify Tregs as key regulators of immune escape of LSCs and TNFRSF4 as a potential target to reduce the function of Tregs and boost antileukemic immunity in CML.


Subject(s)
Immunotherapy/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Receptors, OX40/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology , Animals , Chronic Disease , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice
5.
BMC Cancer ; 21(1): 1002, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34493241

ABSTRACT

BACKGROUND: Leukemic cells facilitate the creation of the tumor-favorable microenvironment in the bone marrow niche using their secreted factors. There are not comprehensive details about immunosuppressive properties of chronic myelogenous leukemia-derived exosomes in the bone marrow stromal and immune compartment. We explained here that K562-derived exosomes could affect the gene expression, cytokine secretion, nitric oxide (NO) production, and redox potential of human primary cord blood-derived T cells (CB T cells). METHODS: Human primary cord blood-derived T cells were treated with K562-derived exosomes. We evaluated the expression variation of some critical genes activated in suppressor T cells. The alterations of some inflammatory and anti-inflammatory cytokines levels were assessed using ELISA assay and real-time PCR. Finally, NO production and intracellular ROS level in CB T cells were evaluated using Greiss assay and flow cytometry, respectively. RESULTS: Our results showed the over-expression of the genes involved in inhibitory T cells, including NQO1, PD1, and FoxP3. In contrast, genes involved in T cell activation such as CD3d and NFATc3 have been reduced significantly. Also, the expression of interleukin 10 (IL-10) and interleukin 6 (IL-6) mRNAs were significantly up-regulated in these cells upon exosome treatment. In addition, secretion of the interleukin 10, interleukin 6, and interleukin 17 (IL-17) proteins increased in T cells exposed to K562-derived exosomes. Finally, K562-derived exosomes induce significant changes in the NO production and intracellular ROS levels in CB T cells. CONCLUSIONS: These results demonstrate that K562-derived exosomes stimulate the immunosuppressive properties in CB-derived T cells by inducing anti-inflammatory cytokines such as IL-10, reducting ROS levels, and arising of NO synthesis in these cells. Moreover, considering the elevation of FOXP3, IL-6, and IL-17 levels in these cells, exosomes secreted by CML cells may induce the fates of T cells toward tumor favorable T cells instead of conventional activated T cells.


Subject(s)
Cytokines/metabolism , Exosomes/immunology , Fetal Blood/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Tumor Microenvironment/immunology , Cell Proliferation , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
6.
Cancer Med ; 10(18): 6310-6316, 2021 09.
Article in English | MEDLINE | ID: mdl-34464516

ABSTRACT

BACKGROUND: Patients with hematological malignancies are at an increased risk of SARS-CoV-2 disease (COVID-19) and adverse outcome. However, a low mortality rate has been reported in patients with chronic myeloid leukemia (CML). Preclinical evidence suggests that tyrosine kinase inhibitors (TKIs) may have a protective role against severe COVID-19. METHODS: We conducted a cross-sectional study of 564 consecutive patients with CML who were tested for anti-SARS-CoV-2 IgG/IgM antibodies at their first outpatient visit between May and early November 2020 in five hematologic centers representative of three Italian regions. RESULTS: The estimated serological prevalence of SARS-CoV-2 infection in patients with CML after the first pandemic wave was similar to that in the general population (about 2%), both at national and regional levels. CML patients with positive anti-SARS-CoV-2 serology were more frequently male (p = 0.027) and active workers (p = 0.012), while there was no significant association with TKI treatment type. Only 3 out of 11 IgG-positive patients had previously received a molecular diagnosis of COVID-19, while the remainders were asymptomatic or with mild symptoms. CONCLUSIONS: Our data confirm that the course of SARS-CoV-2 infection in patients with CML is generally mild and reassure about the safety of continuing TKIs during the COVID-19 pandemic. Furthermore, we suggest that patients with CML succeed to mount an antibody response after exposure to SARS-CoV-2, similar to the general population.


Subject(s)
COVID-19/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/virology , SARS-CoV-2/immunology , Adult , Aged , Aged, 80 and over , COVID-19 Serological Testing/methods , Cross-Sectional Studies , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Italy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Male , Middle Aged , Prevalence , Young Adult
7.
Immunohorizons ; 5(8): 687-702, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433624

ABSTRACT

Tyrosine kinase inhibitor (TKI)-treated chronic myeloid leukemia (CML) patients with increased NK cell number have a better prognosis, and thus, NK cells may suppress CML. However, the efficacy of TKIs varies for reasons yet to be fully elucidated. As NK cell activity is modulated by interactions between their killer cell Ig-like receptors (KIRs) and HLAs of target cells, the combination of their polymorphisms may have functional significance. We previously showed that allelic polymorphisms of KIR3DL1 and HLAs were associated with the prognosis of TKI-treated CML patients. In this study, we focus on differential NK cell activity modulation through KIR3DL1 allotypes. KIR3DL1 expression levels varied according to their alleles. The combination of KIR3DL1 expression level and HLA-Bw4 motifs defined NK cell activity in response to the CML-derived K562 cell line, and Ab-mediated KIR3DL1 blocking reversed this activity. The TKI dasatinib enhanced NK cell activation and cytotoxicity in a KIR3DL1 allotype-dependent manner but did not significantly decrease effector regulatory T cells, suggesting that it directly activated NK cells. Dasatinib also enhanced NK cell cytotoxicity against K562 bearing the BCR-ABL1 T315I TKI resistance-conferring mutation, depending on KIR3DL1/HLA-Bw4 allotypes. Transduction of KIR3DL1*01502 into the NK cell line NK-92 resulted in KIR3DL1 expression and suppression of NK-92 activity by HLA-B ligation, which was reversed by anti-KIR3DL1 Ab. Finally, KIR3DL1 expression levels also defined activation patterns in CML patient-derived NK cells. Our findings raise the possibility of a novel strategy to enhance antitumor NK cell immunity against CML in a KIR3DL1 allotype-dependent manner.


Subject(s)
Gene Expression Regulation, Leukemic/immunology , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Receptors, KIR3DL1/immunology , Alleles , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Dasatinib/pharmacology , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/immunology , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-B Antigens/metabolism , Humans , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mutation , Protein Kinase Inhibitors/pharmacology , Receptors, KIR3DL1/genetics , Receptors, KIR3DL1/metabolism
8.
Immunobiology ; 226(4): 152111, 2021 07.
Article in English | MEDLINE | ID: mdl-34237654

ABSTRACT

T cell dysfunction is a common characteristic in leukemia patients that significantly impacts clinical treatment and prognosis. However, the mechanism underlying T cell dysfunction and its reversal remains unclear. In this study, in accordance with our previous findings, we found that the expression of NFAT2 and pri-miR-17 ~ 92 are lower in peripheral blood CD3+ T cells from chronic myelogenous leukemia (CML) patients by gene expression analysis. We further demonstrate that the NFAT2-induced activation, differentiation, and expression of cytokines in human umbilical cord blood CD8+ naïve T cells are miR-20a-5p dependent. We also preliminarily explored the relationship between NFAT2 and miR-20a-5p in naive T cells. These results suggest that NFAT2 and miR-20a are crucial for regulating functional CD8+ T cells. Additionally, their alteration may be related to CD8+ T cell dysfunction in CML patients; thus, NFAT2 and miR-20a-5p may be considered potential targets for revising T cell function in leukemia immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , MicroRNAs/immunology , NFATC Transcription Factors/immunology , Cell Differentiation , Cells, Cultured , Cytokines/immunology , Fetal Blood/cytology , Fetal Blood/immunology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Signal Transduction
9.
Br J Haematol ; 194(6): 999-1006, 2021 09.
Article in English | MEDLINE | ID: mdl-34085278

ABSTRACT

Patients receiving targeted cancer treatments such as tyrosine kinase inhibitors (TKIs) have been classified in the clinically extremely vulnerable group to develop severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), including patients with chronic myeloid leukaemia (CML) taking TKIs. In addition, concerns that immunocompromised individuals with solid and haematological malignancies may not mount an adequate immune response to a single dose of SARS-CoV-2 BNT162b2 (Pfizer-BioNTech) vaccine have been raised. In the present study, we evaluated humoral and cellular immune responses after a first injection of BNT162b2 vaccine in 16 patients with CML. Seroconversion and cellular immune response before and after vaccination were assessed. By day 21 after vaccination, anti-Spike immunoglobulin G was detected in 14/16 (87·5%) of the patients with CML and all developed a neutralising antibody response [serum dilution that inhibits 50% infection (ID50 ) >50], including medium (ID50 of 200-500) or high (ID50 of 501-2000) neutralising antibodies titres in nine of the 16 (56·25%) patients. T-cell response was seen in 14/15 (93·3%) evaluable patients, with polyfunctional responses seen in 12/15 (80%) patients (polyfunctional CD4+ response nine of 15, polyfunctional CD8+ T-cell response nine of 15). These data demonstrate the immunogenicity of a single dose of SARS-CoV-2 BNT162b2 vaccine in most patients with CML, with both neutralising antibodies and polyfunctional T-cell responses seen in contrast to patients with solid tumour or lymphoid haematological malignancies.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 , Hematologic Neoplasms/immunology , Immunity, Cellular/drug effects , Immunoglobulin G/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , SARS-CoV-2/immunology , Adult , Aged , BNT162 Vaccine , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Female , Hematologic Neoplasms/drug therapy , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Spike Glycoprotein, Coronavirus/immunology
11.
Leukemia ; 35(5): 1229-1242, 2021 05.
Article in English | MEDLINE | ID: mdl-33833387

ABSTRACT

The quest for treatment-free remission (TFR) and deep molecular response (DMR) in chronic myeloid leukemia (CML) has been profoundly impacted by tyrosine kinase inhibitors (TKIs). Immunologic surveillance of residual leukemic cells is hypothesized to be one of the critical factors in successful TFR, with self-renewing leukemic stem cells implicated in relapse. Immunological characterization in CML may help to develop novel immunotherapies that specifically target residual leukemic cells upon TKI discontinuation to improve TFR rates. This review focuses on immune dysfunction in newly diagnosed CML patients, and the role that TKIs and other therapies have in restoring immune surveillance. Immune dysfunction and immunosurveillance in CML points towards several emerging areas in the key goals of DMR and TFR, including: (1) Aspects of innate immune system, in particular natural killer cells and the newly emerging target plasmacytoid dendritic cells. (2) The adaptive immune system, with promise shown in regard to leukemia-associated antigen vaccine-induced CD8 cytotoxic T-cells (CTL) responses, increased CTL expansion, and immune checkpoint inhibitors. (3) Immune suppressive myeloid-derived suppressor cells and T regulatory cells that are reduced in DMR and TFR. (4) Immunomodulator mesenchymal stromal cells that critically contribute to leukomogenesis through immunosuppressive properties and TKI- resistance. Therapeutic strategies that leverage existing immunological approaches include donor lymphocyte infusions, that continue to be used, often in combination with TKIs, in patients relapsing following allogeneic stem cell transplant. Furthermore, previous standards-of-care, including interferon-α, hold promise in attaining TFR in the post-TKI era. A deeper understanding of the immunological landscape in CML is therefore vital for both the development of novel and the repurposing of older therapies to improve TFR outcomes.


Subject(s)
Immune System/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Animals , Chronic Disease , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Protein Kinase Inhibitors/immunology , Treatment Outcome
12.
Front Immunol ; 12: 634402, 2021.
Article in English | MEDLINE | ID: mdl-33679785

ABSTRACT

Beta-glucans are naturally occurring polysaccharides present in cell walls of fungi, yeast, bacteria, cereals, seaweed, and algae. These microbe-associated molecular patterns (MAMPs) possess immunomodulatory properties. In human, it has been suggested that NK cells can be activated by ß-glucans. Here, we aimed to elucidate whether ß-glucans modulate porcine NK cell responses in vitro and if so, how these effects are mediated. We investigated the effect of two ß-glucans, Macrogard and Curdlan, which differ in solubility and structure. Direct addition of ß-glucans to purified porcine NK cells did not affect cytotoxicity of these cells against K562 target cells. However, when using PBMC instead of purified NK cells, ß-glucan addition significantly increased NK cell-mediated cytotoxicity. This effect depended on factors secreted by CD14+ monocytes upon ß-glucan priming. Further analysis showed that monocytes secrete TNF-α, IL-6, and IL-10 upon ß-glucan addition. Of these, IL-10 turned out to play a critical role in ß-glucan-triggered NK cell cytotoxicity, since depletion of IL-10 completely abrogated the ß-glucan-induced increase in cytotoxicity. Furthermore, addition of recombinant IL-10 to purified NK cells was sufficient to enhance cytotoxicity. In conclusion, we show that ß-glucans trigger IL-10 secretion by porcine monocytes, which in turn leads to increased NK cell cytotoxicity, and thereby identify IL-10 as a potent stimulus of porcine NK cell cytotoxicity.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-10/metabolism , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Monocytes/drug effects , Paracrine Communication , beta-Glucans/pharmacology , Animals , Coculture Techniques , Humans , K562 Cells , Killer Cells, Natural/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Monocytes/immunology , Monocytes/metabolism , Secretory Pathway , Sus scrofa
13.
Int J Hematol ; 113(5): 642-647, 2021 May.
Article in English | MEDLINE | ID: mdl-33651270

ABSTRACT

Chronic myeloid leukemia (CML) is caused by the reciprocal translocation t(9;22)(q34;q11), resulting in the BCR-ABL1 fusion gene. BCR-ABL1 tyrosine kinase inhibitors (TKIs) improve overall survival in patients with chronic phase CML (CML-CP). Approximately half of the patients who achieve a durable deep molecular response can achieve sustained treatment-free remission (TFR) after TKI discontinuation; thus TFR is now a therapeutic goal for most patients with CML-CP. Sensitive BCL-ABL1 transcript detection methods reveal that evidence of residual CML cells remains in patients who achieve sustained TFR, indicating that the host immune system protects against CML relapse. The human immune system is composed of innate and adaptive arms. Natural killer cells are major components of the innate immune system, while T cells are major components of the adaptive immune system. Myeloid-derived suppressor cells and regulatory T cells, both suppressors of the immune response, have important roles in the regulation of CML. Here, we review the current understanding of the immune response in CML, especially in TFR.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunity/drug effects , Immunologic Factors/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Humans , Immunologic Surveillance/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Remission Induction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
14.
Immunol Med ; 44(2): 61-68, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32715973

ABSTRACT

The development of BCR-ABL1 tyrosine kinase inhibitors (TKIs) markedly improved the prognosis of patients with chronic myeloid leukemia (CML). Approximately 50% of patients who achieve deep molecular response (DMR) remain in treatment-free remission (TFR) even after discontinuation of TKIs. Although TKIs may achieve clinical "cure" after TKI treatment for specific periods, there are no reliable biomarkers for predicting the response to TKIs and the probability of TFR in CML. An increase in natural killer (NK) cells in the peripheral blood of TKI-treated CML patients is correlated with better outcomes, suggesting that TKIs induce antitumor NK cell immunity against CML cells. Killer immunoglobulin-like receptors (KIRs) are highly polymorphic NK cell receptors that play important roles in the regulation of immune responses. The identification of allelic polymorphisms of KIRs by next-generation sequencing uncovered novel aspects of KIRs. Here we summarize the current knowledge of the genetic and immunological aspects of KIRs and discuss the association between allelic polymorphisms of KIRs and TKI-treated CML.


Subject(s)
Alleles , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Polymorphism, Genetic , Receptors, KIR/genetics , Receptors, KIR/immunology , Fusion Proteins, bcr-abl/antagonists & inhibitors , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Treatment Outcome
15.
Toxicol Appl Pharmacol ; 412: 115353, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33301752

ABSTRACT

Chronic exposure to arsenic causes cancers in various organs including the skin, liver, lung, and bladder in humans, but the mechanisms of the multi-organ carcinogenicity of arsenic remain unknown. Natural killer (NK) cells play important roles in the immune surveillance and elimination of tumor cells. Although accumulating evidence has indicated that arsenic has immunosuppressive properties, little is known about the effects of arsenic on the tumoricidal functions of NK cells. We examined the effects of arsenite on the cytotoxic activities of human and mouse NK cells toward target tumor cells. Exposure of human NK-92 cells and primary mouse NK cells to sublethal doses of arsenite reduced the IL-2-activated cytotoxic activities toward human K562 cells and murine YAC-1 cells, respectively. NK cells recognize target cells via integrated signals from both activating and inhibitory receptors and induce apoptosis of target cells via a granzyme/perforin system. We found that exposure of NK-92 cells to arsenite diminished the IL-2-activated down-regulation of the inhibitory receptors, KIR2DL2 and KIR2DL3, and the up-regulation of granzyme B and lymphotoxin-α. The IL-2-activated increases in secretion of interferon-γ and IL-10 were also slightly reduced by arsenite. Thus, arsenite suppressed the IL-2-activated cytotoxic activity of NK cells by disrupting multiple pathways required for the recognition and killing of target tumor cells. Our findings provide new insights into the roles of NK cell-mediated tumor immunity in cancer development by arsenic.


Subject(s)
Arsenites/toxicity , Cytotoxicity, Immunologic/drug effects , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Sodium Compounds/toxicity , Tumor Escape/drug effects , Animals , Coculture Techniques , Granzymes/genetics , Granzymes/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/metabolism , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Receptors, KIR2DL2/genetics , Receptors, KIR2DL2/metabolism , Receptors, KIR2DL3/genetics , Receptors, KIR2DL3/metabolism
16.
Br J Cancer ; 124(4): 817-830, 2021 02.
Article in English | MEDLINE | ID: mdl-33214684

ABSTRACT

BACKGROUND: Interferon (IFN) signalling pathways, a key element of the innate immune response, contribute to resistance to conventional chemotherapy, radiotherapy, and immunotherapy, and are often deregulated in cancer. The deubiquitylating enzyme USP18 is a major negative regulator of the IFN signalling cascade and is the predominant human protease that cleaves ISG15, a ubiquitin-like protein tightly regulated in the context of innate immunity, from its modified substrate proteins in vivo. METHODS: In this study, using advanced proteomic techniques, we have significantly expanded the USP18-dependent ISGylome and proteome in a chronic myeloid leukaemia (CML)-derived cell line. USP18-dependent effects were explored further in CML and colorectal carcinoma cellular models. RESULTS: Novel ISGylation targets were characterised that modulate the sensing of innate ligands, antigen presentation and secretion of cytokines. Consequently, CML USP18-deficient cells are more antigenic, driving increased activation of cytotoxic T lymphocytes (CTLs) and are more susceptible to irradiation. CONCLUSIONS: Our results provide strong evidence for USP18 in regulating antigenicity and radiosensitivity, highlighting its potential as a cancer target.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/immunology , Cytokines/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Ubiquitin Thiolesterase/metabolism , Ubiquitins/metabolism , Antigenic Variation , Cell Line, Tumor , Colorectal Neoplasms/radiotherapy , Gene Knockout Techniques , HCT116 Cells , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/radiotherapy , Radiation Tolerance/genetics , Radiation Tolerance/immunology , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics
17.
Transpl Infect Dis ; 23(2): e13477, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32989856

ABSTRACT

Immunosuppressed patients such as solid organ transplant and hematologic malignancy patients appear to be at increased risk for morbidity and mortality due to coronavirus disease 2019 (COVID-19) caused by SARS coronavirus 2 (SARS-CoV-2). Convalescent plasma, a method of passive immunization that has been applied to prior viral pandemics, holds promise as a potential treatment for COVID-19. Immunocompromised patients may experience more benefit from convalescent plasma given underlying deficits in B and T cell immunity as well as contraindications to antiviral and immunomodulatory therapy. We describe our institutional experience with four immunosuppressed patients (two kidney transplant recipients, one lung transplant recipient, and one chronic myelogenous leukemia patient) treated with COVID-19 convalescent plasma through the Expanded Access Program (NCT04338360). All patients clinically improved after administration (two fully recovered and two discharged to skilled nursing facilities) and none experienced a transfusion reaction. We also report the characteristics of convalescent plasma product from a local blood center including positive SARS-CoV-2 IgG and negative SARS-CoV-2 PCR in all samples tested. This preliminary evidence suggest that convalescent plasma may be safe among immunosuppressed patients with COVID-19 and emphasizes the need for further data on the efficacy of convalescent plasma as either primary or adjunctive therapy for COVID-19.


Subject(s)
COVID-19/therapy , Graft Rejection/prevention & control , Immunocompromised Host , Immunosuppressive Agents/adverse effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Adult , Aged , COVID-19/immunology , Female , Humans , Immunization, Passive/methods , Kidney Transplantation , Lung Transplantation , Male , Middle Aged , SARS-CoV-2 , Severity of Illness Index , Treatment Outcome , COVID-19 Serotherapy
18.
Mol Cancer Ther ; 20(1): 142-149, 2021 01.
Article in English | MEDLINE | ID: mdl-33082274

ABSTRACT

Treatment-free remission (TFR) is one of the therapeutic goals for patients with chronic phase chronic myeloid leukemia (CML-CP). Although previous reports indicated that antitumor immunity contributes to TFR, its determinants are still unclear. We previously reported that allelic polymorphisms of killer immunoglobulin-like receptors (KIR) and human leukocyte antigens (HLA) are associated with achievement of deep molecular response (DMR) in patients with CML-CP. Here, we examined the association between TFR and polymorphisms of KIRs and HLAs in patients who discontinued tyrosine kinase inhibitors (TKI). Seventy-six patients were enrolled, and their KIR and HLA polymorphisms and natural killer (NK) cell activation status were investigated as previously described. Overall, 33 patients discontinued TKIs, and 21 of 33 achieved TFR [63.6%; 95% confidence interval (CI), 44.9%-77.5%] at 1 year. Multivariate analysis revealed that male sex (HR, 0.157; 95% CI, 0.031-0.804; P = 0.003) and HLA-A*02:01, *11:01, or *24:02 (HR, 6.386; 95% CI, 1.701-23.980; P = 0.006) were associated with TFR. Patients who achieved DMR and discontinued TKIs exhibited higher NK cell activation status than those who did not. By contrast, there were no significant differences in NK cell activation status between the patients who achieved TFR and those who experienced molecular relapse. These results suggest NK cell activation status contributes to achievement of DMR, whereas T-cell-mediated immunity contributes to TFR in patients with CML-CP.


Subject(s)
HLA Antigens/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Polymorphism, Genetic , Protein Kinase Inhibitors/therapeutic use , Withholding Treatment , Adult , Aged , Aged, 80 and over , Female , Humans , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Multivariate Analysis , Remission Induction , Risk Factors
19.
Radiat Environ Biophys ; 60(1): 41-47, 2021 03.
Article in English | MEDLINE | ID: mdl-33125593

ABSTRACT

A model of post-diagnosis chronic myeloid leukemia (CML) dynamics across treatment cessations is applied here to pre-diagnosis scenarios of A-bomb survivors. The main result is that perturbing two parameters of a two-state simplification of this model captures the essence of two A-bomb survivor mysteries: (1) in those exposed to > 1 Sv in Hiroshima, four of six female onsets arose as a cluster in 1969-1974, well after 5-10-year latencies expected and observed in two of six female- and nine of ten male cases (about one background case was expected in this high-dose cohort); and (2) no Nagasaki adult cases exposed to > 0.2 Sv were observed though about nine were expected (~ 1.5 background + ~ 7.5 radiation-induced). Overall, it is concluded that: (1) whole-body radiation co-creates malignant and benign BCR-ABL clones; (2) benign clones are more likely to act as anti-CML vaccines in females than in males; (3) the Hong Kong flu of 1968 (and H3N2 seasonal flu thereafter) exhausted anti-CML immunity, thereby releasing radiation-induced clones latent in high-dose Hiroshima females; and (4) benign cells of 1-2 are CD4+ as human T-cell leukemia-lymphoma virus-1 endemic to Nagasaki but not Hiroshima expands numbers of such cells. The next goal is to see if these conclusions can be substantiated using banked A-bomb survivor blood samples.


Subject(s)
Atomic Bomb Survivors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Radiation-Induced/diagnosis , Models, Biological , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Influenza A Virus, H3N2 Subtype , Influenza, Human/immunology , Japan/epidemiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Radiation-Induced/immunology , Male , Neoplastic Stem Cells/immunology
20.
Cancer Control ; 27(1): 1073274820976594, 2020.
Article in English | MEDLINE | ID: mdl-33297765

ABSTRACT

Hepatitis B reactivation (HBVr) in cancer patients is a well-established complication due to chemotherapy-induced immunosuppression. Studies have reported HBVr associated with immunosuppressive medications, such as rituximab, methotrexate, and high dose steroids. There are different risks for different types of chemotherapy with rituximab carrying one of the highest risks for hepatitis B reactivation. Tyrosine kinase inhibitors (TKIs) are the standard of care in patients with chronic myeloid leukemia (CML). The risk of HBVr in chronic myeloid leukemia has been reported in many studies, but to this date, there are no clear guidelines or recommendations regarding screening and monitoring of HBV in CML patients receiving TKIs. We conducted this review to identify the risk of HBVr in patients with CML who are treated with tyrosine kinase inhibitors. We recommend testing for HBV status in patients who are to be treated with TKIs and to consider giving prophylaxis in those who are positive for HBsAg at baseline. More studies are needed to assess the risk of reactivation in patients with Hepatitis B core antibody positive receiving TKIs. Currently, monitoring such patients for reactivation may be the best strategy.


Subject(s)
Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/adverse effects , Virus Activation/immunology , Antibiotic Prophylaxis/methods , Antibiotic Prophylaxis/standards , Antiviral Agents/therapeutic use , Drug Monitoring/standards , Hepatitis B Surface Antigens/immunology , Hepatitis B Surface Antigens/isolation & purification , Hepatitis B virus/physiology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/prevention & control , Hepatitis B, Chronic/virology , Humans , Immunocompromised Host , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Mass Screening/standards , Mass Screening/statistics & numerical data , Practice Guidelines as Topic , Virus Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL