Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Br J Cancer ; 130(12): 1990-2002, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38600326

ABSTRACT

BACKGROUND: Previous studies have suggested that patients with HER2-low breast cancers do not benefit from trastuzumab treatment although the reasons remain unclear. METHODS: We investigated the effect of trastuzumab monotherapy and its combination with different HER2 targeting treatments in a panel of breast cancer cell lines and patient-derived organoids (PDOs) using biochemical methods and cell viability assays. RESULTS: Compared to sensitive HER2 over-expressing (IHC3 + ) breast cancer cells, increasing doses of trastuzumab could not achieve IC50 in MDA-MB-361 (IHC 2 + FISH + ) and MDA-MB-453 (IHC 2 + FISH-) cells which showed an intermediate response to trastuzumab. Trastuzumab treatment induced upregulation of HER ligand release, resulting in the activation of HER receptors in these cells, which could account for their trastuzumab insensitivity. Adding a dual ADAM10/17 inhibitor to inhibit the shedding of HER ligands in combination with trastuzumab only showed a modest decrease in the cell viability of HER2-low breast cancer cells and PDOs. However, the panHER inhibitor neratinib was an effective monotherapy in HER2-low breast cancer cells and PDOs, and showed additive effects when combined with trastuzumab. CONCLUSION: This study demonstrates that neratinib in combination with trastuzumab may be effective in a subset of HER2-low breast cancers although further validation is required in a larger panel of PDOs and in future clinical studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Organoids , Quinolines , Receptor, ErbB-2 , Trastuzumab , Humans , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Female , Organoids/drug effects , Quinolines/pharmacology , Quinolines/administration & dosage , Cell Line, Tumor , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Survival/drug effects
2.
Br J Haematol ; 200(5): 573-578, 2023 03.
Article in English | MEDLINE | ID: mdl-36413792

ABSTRACT

The survival of acute myeloid leukaemia (AML) patients aged over 60 has been suboptimal historically, whether they are treated using hypomethylating agents, low-dose cytarabine (LDAC) or venetoclax-based regimens. Progress is being made, however, for subgroups with favourable molecular or cytogenetic findings. Arginine metabolism plays a key role in AML pathophysiology. We report the only randomised study of LDAC with recombinant arginase BCT-100 versus LDAC alone in older AML patients unsuitable for intensive therapy. Eighty-three patients were randomised to the study. An overall response rate was seen in 19.5% (all complete remission [CR]) and 15% (7.5% each in CR and CR without evidence of adequate count recovery [CRi]) of patients in the LDAC+BCT-100 and LDAC arms respectively (odds ratio 0.73, confidence interval 0.23-2.33; p = 0.592). No significant difference in overall or median survival between treatment arms was seen. The addition of BCT-100 to LDAC was well tolerated.


Subject(s)
Cytarabine , Leukemia, Myeloid, Acute , Humans , Middle Aged , Aged , Arginase , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Polyethylene Glycols/therapeutic use
3.
Cancer Immunol Immunother ; 72(3): 543-560, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35962843

ABSTRACT

Acute myeloid leukaemia (AML) creates an immunosuppressive environment to conventional T cells through Arginase 2 (ARG2)-induced arginine depletion. We identify that AML blasts release the acute phase protein serum amyloid A (SAA), which acts in an autocrine manner to upregulate ARG2 expression and activity, and promote AML blast viability. Following in vitro cross-talk invariant natural killer T (iNKT) cells become activated, upregulate mitochondrial capacity, and release IFN-γ. iNKT retain their ability to proliferate and be activated despite the low arginine AML environment, due to the upregulation of Large Neutral Amino Acid Transporter-1 (LAT-1) and Argininosuccinate Synthetase 1 (ASS)-dependent amino acid pathways, resulting in AML cell death. T cell proliferation is restored in vitro and in vivo. The capacity of iNKT cells to restore antigen-specific T cell immunity was similarly demonstrated against myeloid-derived suppressor cells (MDSCs) in wild-type and Jα18-/- syngeneic lymphoma-bearing models in vivo. Thus, stimulation of iNKT cell activity has the potential as an immunotherapy against AML or as an adjunct to boost antigen-specific T cell immunotherapies in haematological or solid cancers.


Subject(s)
Leukemia, Myeloid, Acute , Myeloid-Derived Suppressor Cells , Natural Killer T-Cells , Humans , Cell Proliferation , Arginine
4.
Blood ; 137(2): 203-215, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33443553

ABSTRACT

Chronic active Epstein-Barr virus (CAEBV) typically presents as persistent infectious mononucleosis-like disease and/or hemophagocytic lymphohistocytosis (HLH), reflecting ectopic Epstein-Barr virus (EBV) infection and lymphoproliferation of T and/or NK cells. Clinical behavior ranges from indolent, stable disease through to rapidly progressive, life-threatening disease. Although it is thought the chronicity and/or progression reflect an escape from immune control, very little is known about the phenotype and function of the infected cells vs coresident noninfected population, nor about the mechanisms that could underpin their evasion of host immune surveillance. To investigate these questions, we developed a multicolor flow cytometry technique combining phenotypic and functional marker staining with in situ hybridization for the EBV-encoded RNAs (EBERs) expressed in every infected cell. This allows the identification, phenotyping, and functional comparison of infected (EBERPOS) and noninfected (EBERNEG) lymphocyte subset(s) in patients' blood samples ex vivo. We have characterized CAEBV and HLH cases with monoclonal populations of discrete EBV-activated T-cell subsets, in some cases accompanied by EBV-activated NK-cell subsets, with longitudinal data on the infected cells' progression despite standard steroid-based therapy. Given that cytotoxic CD8+ T cells with relevant EBV antigen specificity were detectable in the blood of the best studied patient, we searched for means whereby host surveillance might be impaired. This revealed a unique feature in almost every patient with CAEBV studied: the presence of large numbers of myeloid-derived suppressor cells that exhibited robust inhibition of T-cell growth. We suggest that their influence is likely to explain the host's failure to contain EBV-positive T/NK-cell proliferation.


Subject(s)
Epstein-Barr Virus Infections/immunology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/virology , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocyte Subsets/virology , Adult , Flow Cytometry/methods , Herpesvirus 4, Human/immunology , Humans , Male , Middle Aged , Young Adult
5.
Cancer Immunol Immunother ; 71(7): 1583-1596, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34727230

ABSTRACT

BACKGROUND: Monocytic myeloid-derived suppressor cells (M-MDSCs) are significantly expanded in the blood of colorectal cancer (CRC) patients. However, their presence and underlying mechanisms in the tumour microenvironment of CRC have not been examined in detail. METHODS: Tumour tissues and peripheral blood from CRC patients were analysed for the presence of M-MDSCs. The mechanisms of suppression were analysed by blocking pathways by which MDSCs abrogate T cell proliferation. Co-culture of CRC cells with monocytes were performed with and without cytokine blocking antibodies to determine the mechanism by which CRC cells polarise monocytes. Multi-spectral IHC was used to demonstrate the intra-tumoral location of M-MDSCs. RESULTS: Tumour tissues and blood of CRC patients contain M-MDSCs which inhibit T cell proliferation. Whilst inhibition of arginase and nitric oxide synthase 2 fail to rescue T cell proliferation, blockade of IL-10 released by these HLA-DR- cells abrogates the suppresivity of M-MDSCs. Tumour conditioned media (TCM) significantly reduces HLA-DR expression, increases IL-10 release from monocytes and causes them to become suppressive. TGF-ß is highly expressed in the TCM and accumulates in the plasma. TGF-ß reduces HLA-DR expression and drives monocyte immunosuppressivity. The invasive margin of CRC is enriched in CD14+ HLA-DR- cells in close proximity to T cells. CONCLUSIONS: Our study demonstrates the cross-talk between CRC cells, M-MDSCs and T cells. Characterisation of CRC M-MDSCs point to therapeutic avenues to target these cells in addition to TGF-ß blockade.


Subject(s)
Colorectal Neoplasms , Myeloid-Derived Suppressor Cells , Transforming Growth Factor beta , Colorectal Neoplasms/metabolism , HLA-DR Antigens , Humans , Interleukin-10/metabolism , Monocytes , Phenotype , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
6.
Nat Immunol ; 11(11): 1039-46, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20890286

ABSTRACT

Neutrophils are the main effector cells during inflammation, but they can also control excessive inflammatory responses by secreting anti-inflammatory cytokines. However, the mechanisms that modulate their plasticity remain unclear. We now show that systemic serum amyloid A 1 (SAA-1) controls the plasticity of neutrophil differentiation. SAA-1 not only induced anti-inflammatory interleukin 10 (IL-10)-secreting neutrophils but also promoted the interaction of invariant natural killer T cells (iNKT cells) with those neutrophils, a process that limited their suppressive activity by diminishing the production of IL-10 and enhancing the production of IL-12. Because SAA-1-producing melanomas promoted differentiation of IL-10-secreting neutrophils, harnessing iNKT cells could be useful therapeutically by decreasing the frequency of immunosuppressive neutrophils and restoring tumor-specific immune responses.


Subject(s)
Cell Differentiation/immunology , Interleukin-10/immunology , Melanoma/immunology , Natural Killer T-Cells/immunology , Neutrophils/immunology , Serum Amyloid A Protein/immunology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Neutrophils/cytology
7.
Blood ; 136(10): 1155-1160, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32573723

ABSTRACT

Hematological and solid cancers catabolize the semiessential amino acid arginine to drive cell proliferation. However, the resulting low arginine microenvironment also impairs chimeric antigen receptor T cells (CAR-T) cell proliferation, limiting their efficacy in clinical trials against hematological and solid malignancies. T cells are susceptible to the low arginine microenvironment because of the low expression of the arginine resynthesis enzymes argininosuccinate synthase (ASS) and ornithine transcarbamylase (OTC). We demonstrate that T cells can be reengineered to express functional ASS or OTC enzymes, in concert with different chimeric antigen receptors. Enzyme modifications increase CAR-T cell proliferation, with no loss of CAR cytotoxicity or increased exhaustion. In vivo, enzyme-modified CAR-T cells lead to enhanced clearance of leukemia or solid tumor burden, providing the first metabolic modification to enhance CAR-T cell therapies.


Subject(s)
Arginine/metabolism , Argininosuccinate Synthase/metabolism , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Neuroblastoma/therapy , Ornithine Carbamoyltransferase/metabolism , T-Lymphocytes/transplantation , Animals , Apoptosis , Argininosuccinate Synthase/genetics , Cell Proliferation , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Metabolic Engineering/methods , Mice , Mice, Nude , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Ornithine Carbamoyltransferase/genetics , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Int J Cancer ; 145(8): 2201-2208, 2019 10 15.
Article in English | MEDLINE | ID: mdl-30485425

ABSTRACT

Therapeutic approaches which aim to target Acute Myeloid Leukaemia through enhancement of patients' immune responses have demonstrated limited efficacy to date, despite encouraging preclinical data. Examination of AML patients treated with azacitidine (AZA) and vorinostat (VOR) in a Phase II trial, demonstrated an increase in the expression of Cancer-Testis Antigens (MAGE, RAGE, LAGE, SSX2 and TRAG3) on blasts and that these can be recognised by circulating antigen-specific T cells. Although the T cells have the potential to be activated by these unmasked antigens, the low arginine microenvironment created by AML blast Arginase II activity acts a metabolic brake leading to T cell exhaustion. T cells exhibit impaired proliferation, reduced IFN-γ release and PD-1 up-regulation in response to antigen stimulation under low arginine conditions. Inhibition of arginine metabolism enhanced the proliferation and cytotoxicity of anti-NY-ESO T cells against AZA/VOR treated AML blasts, and can boost anti-CD33 Chimeric Antigen Receptor-T cell cytotoxicity. Therefore, measurement of plasma arginine concentrations in combination with therapeutic targeting of arginase activity in AML blasts could be a key adjunct to immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arginase/antagonists & inhibitors , Arginine/blood , Leukemia, Myeloid/therapy , Acute Disease , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Arginase/blood , Arginase/metabolism , Arginine/metabolism , Azacitidine/administration & dosage , Humans , Immunotherapy/methods , K562 Cells , Leukemia, Myeloid/immunology , Leukemia, Myeloid/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Sialic Acid Binding Ig-like Lectin 3/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Vorinostat/administration & dosage
9.
Int J Cancer ; 142(7): 1490-1502, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29168171

ABSTRACT

Arginine is a semi-essential amino acid that plays a key role in cell survival and proliferation in normal and malignant cells. BCT-100, a pegylated (PEG) recombinant human arginase, can deplete arginine and starve malignant cells of the amino acid. Acute lymphoblastic leukemia (ALL) is the most common cancer of childhood, yet for patients with high risk or relapsed disease prognosis remains poor. We show that BCT-100 is cytotoxic to ALL blasts from patients in vitro by necrosis, and is synergistic in combination with dexamethasone. Against ALL xenografts, BCT-100 leads to a reduction in ALL engraftment and a prolongation of survival. ALL blasts express the arginine transporter CAT-1, yet the majority of blasts are arginine auxotrophic due to deficiency in either argininosuccinate synthase (ASS) or ornithine transcarbamylase (OTC). Although endogenous upregulation or retroviral transduced increases in ASS or OTC may promote ALL survival under moderately low arginine conditions, expression of these enzymes cannot prevent BCT-100 cytotoxicity at arginine depleting doses. RNA-sequencing of ALL blasts and supporting stromal cells treated with BCT-100 identifies a number of candidate pathways which are altered in the presence of arginine depletion. Therefore, BCT-100 provides a new clinically relevant therapeutic approach to target arginine metabolism in ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Arginase/pharmacology , Arginine/metabolism , Metabolome/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recombinant Proteins/pharmacology , Animals , Cell Survival/drug effects , Dexamethasone/pharmacology , Drug Synergism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
10.
Blood ; 125(15): 2386-96, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25710880

ABSTRACT

Acute myeloid leukemia (AML) is one of the most common acute leukemias in adults and children, yet significant numbers of patients relapse and die of disease. In this study, we identify the dependence of AML blasts on arginine for proliferation. We show that AML blasts constitutively express the arginine transporters CAT-1 and CAT-2B, and that the majority of newly diagnosed patients' blasts have deficiencies in the arginine-recycling pathway enzymes argininosuccinate synthase and ornithine transcarbamylase, making them arginine auxotrophic. BCT-100, a pegylated human recombinant arginase, leads to a rapid depletion in extracellular and intracellular arginine concentrations, resulting in arrest of AML blast proliferation and a reduction in AML engraftment in vivo. BCT-100 as a single agent causes significant death of AML blasts from adults and children, and acts synergistically in combination with cytarabine. Using RNA sequencing, 20 further candidate genes which correlated with resistance have been identified. Thus, AML blasts are dependent on arginine for survival and proliferation, as well as depletion of arginine with BCT-100 of clinical value in the treatment of AML.


Subject(s)
Arginase/therapeutic use , Arginine/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Adolescent , Aged , Animals , Antimetabolites, Antineoplastic/therapeutic use , Child , Child, Preschool , Cytarabine/therapeutic use , Enzyme Therapy , Female , Humans , Infant , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/metabolism , Male , Mice, SCID , Middle Aged , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured , Young Adult
11.
Int J Cancer ; 139(3): 501-9, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26913960

ABSTRACT

Renewed interest in the use of therapeutic enzymes combined with an improved knowledge of cancer cell metabolism, has led to the translation of several arginine depletion strategies into early phase clinical trials. Arginine auxotrophic tumors are reliant on extracellular arginine, due to the downregulation of arginosuccinate synthetase or ornithine transcarbamylase-key enzymes for intracellular arginine recycling. Engineered arginine catabolic enzymes such as recombinant human arginase (rh-Arg1-PEG) and arginine deiminase (ADI-PEG) have demonstrated cytotoxicity against arginine auxotrophic tumors. In this review, we discuss the molecular events triggered by extracellular arginine depletion that contribute to tumor cell death.


Subject(s)
Antineoplastic Agents/therapeutic use , Arginine/metabolism , Enzyme Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Arginase/therapeutic use , Cell Proliferation , Cell Survival , Clinical Trials as Topic , Drug Evaluation, Preclinical , Enzyme Therapy/methods , Humans , Hydrolases/therapeutic use , Metabolic Networks and Pathways/drug effects , Neoplasms/enzymology , Signal Transduction/drug effects , Stress, Physiological/drug effects
12.
Blood ; 122(5): 749-58, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23733335

ABSTRACT

Acute myeloid leukemia (AML) is the most common acute leukemia in adults and the second most common frequent leukemia of childhood. Patients may present with lymphopenia or pancytopenia at diagnosis. We investigated the mechanisms by which AML causes pancytopenia and suppresses patients' immune response. This study identified for the first time that AML blasts alter the immune microenvironment through enhanced arginine metabolism. Arginase II is expressed and released from AML blasts and is present at high concentrations in the plasma of patients with AML, resulting in suppression of T-cell proliferation. We extended these results by demonstrating an arginase-dependent ability of AML blasts to polarize surrounding monocytes into a suppressive M2-like phenotype in vitro and in engrafted nonobese diabetic-severe combined immunodeficiency mice. In addition, AML blasts can suppress the proliferation and differentiation of murine granulocyte-monocyte progenitors and human CD34(+) progenitors. Finally, the study showed that the immunosuppressive activity of AML blasts can be modulated through small-molecule inhibitors of arginase and inducible nitric oxide synthase, suggesting a novel therapeutic target in AML. The results strongly support the hypothesis that AML creates an immunosuppressive microenvironment that contributes to the pancytopenia observed at diagnosis.


Subject(s)
Arginase/physiology , Immune Tolerance , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Tumor Microenvironment/immunology , Animals , Arginase/metabolism , Cell Proliferation , Cells, Cultured , Humans , Immune Tolerance/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Transplantation, Heterologous , Tumor Escape/physiology , Tumor Microenvironment/physiology
13.
Front Oncol ; 14: 1296576, 2024.
Article in English | MEDLINE | ID: mdl-38357205

ABSTRACT

Background: The survival for many children with relapsed/refractory cancers remains poor despite advances in therapies. Arginine metabolism plays a key role in the pathophysiology of a number of pediatric cancers. We report the first in child study of a recombinant human arginase, BCT-100, in children with relapsed/refractory hematological, solid or CNS cancers. Procedure: PARC was a single arm, Phase I/II, international, open label study. BCT-100 was given intravenously over one hour at weekly intervals. The Phase I section utilized a modified 3 + 3 design where escalation/de-escalation was based on both the safety profile and the complete depletion of arginine (defined as adequate arginine depletion; AAD <8µM arginine in the blood after 4 doses of BCT-100). The Phase II section was designed to further evaluate the clinical activity of BCT-100 at the pediatric RP2D determined in the Phase I section, by recruitment of patients with pediatric cancers into 4 individual groups. A primary evaluation of response was conducted at eight weeks with patients continuing to receive treatment until disease progression or unacceptable toxicity. Results: 49 children were recruited globally. The Phase I cohort of the trial established the Recommended Phase II Dose of 1600U/kg iv weekly in children, matching that of adults. BCT-100 was very well tolerated. No responses defined as a CR, CRi or PR were seen in any cohort within the defined 8 week primary evaluation period. However a number of these relapsed/refractory patients experienced prolonged radiological SD. Conclusion: Arginine depletion is a clinically safe and achievable strategy in children with cancer. The RP2D of BCT-100 in children with relapsed/refractory cancers is established at 1600U/kg intravenously weekly and can lead to sustained disease stability in this hard to treat population. Clinical trial registration: EudraCT, 2017-002762-44; ISRCTN, 21727048; and ClinicalTrials.gov, NCT03455140.

14.
Blood Adv ; 7(9): 1754-1761, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36521029

ABSTRACT

Cancer cells take up amino acids from the extracellular space to drive cell proliferation and viability. Similar mechanisms are applied by immune cells, resulting in the competition between conventional T cells, or indeed chimeric antigen receptor (CAR) T cells and tumor cells, for the limited availability of amino acids within the environment. We demonstrate that T cells can be re-engineered to express SLC7A5 or SLC7A11 transmembrane amino acid transporters alongside CARs. Transporter modifications increase CAR T-cell proliferation under low tryptophan or cystine conditions with no loss of CAR cytotoxicity or increased exhaustion. Transcriptomic and phenotypic analysis reveals that downstream, SLC7A5/SLC7A11-modified CAR T cells upregulate intracellular arginase expression and activity. In turn, we engineer and phenotype a further generation of CAR T cells that express functional arginase 1/arginase 2 enzymes and have enhanced CAR T-cell proliferation and antitumor activity. Thus, CAR T cells can be adapted to the amino acid metabolic microenvironment of cancer, a hitherto recognized but unaddressed barrier for successful CAR T-cell therapy.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Receptors, Chimeric Antigen/metabolism , Receptors, Antigen, T-Cell/genetics , Arginase/genetics , Arginase/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Neoplasms/metabolism , Amino Acids/metabolism , Tumor Microenvironment
15.
Clin Transl Immunology ; 11(5): e1395, 2022.
Article in English | MEDLINE | ID: mdl-35602884

ABSTRACT

Objectives: Recombinant granulocyte colony-stimulating factor (G-CSF) is frequently administered to patients with cancer to enhance granulocyte recovery post-chemotherapy. Clinical trials have also used G-CSF to modulate myeloid cell function in pregnancy and inflammatory diseases. Although the contribution of G-CSF to expanding normal granulocytes is well known, the effect of this cytokine on the phenotype and function of immunosuppressive granulocytic cells remains unclear. Here, we investigate the impact of physiological and iatrogenic G-CSF on an as yet undescribed granulocyte phenotype and ensuing outcome on T cells in the settings of cancer and pregnancy. Methods: Granulocytes from patients treated with recombinant G-CSF, patients with late-stage cancer and women enrolled on a trial of recombinant G-CSF were phenotyped by flow cytometry. The ability and mechanism of polarised granulocytes to suppress T-cell proliferation were assessed by cell proliferation assays, flow cytometry and ELISA. Results: We observed that G-CSF leads to a significant upregulation of CD14 expression on CD15+ granulocytes. These CD15+CD14+ cells are identified in the blood of patients with patients undergoing neutrophil mobilisation with recombinant G-CSF, and physiologically in women early in pregnancy or in those treated as a part of a clinical trial. Immunohistochemistry of tumor tissue or placental tissue identified the expression of G-CSF. The G-CSF upregulates the release of reactive oxygen species (ROS) in CD15+CD14+ cells leading to the suppression of T-cell proliferation. Conclusions: G-CSF induces a population of ROS+ immunosuppressive CD15+CD14+ granulocytes. Strategies for how recombinant G-CSF can be scheduled to reduce effects on T-cell therapies should be developed in future clinical studies.

16.
J Clin Invest ; 118(12): 4036-48, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19033672

ABSTRACT

Infection with influenza A virus (IAV) presents a substantial threat to public health worldwide, with young, elderly, and immunodeficient individuals being particularly susceptible. Inflammatory responses play an important role in the fatal outcome of IAV infection, but the mechanism remains unclear. We demonstrate here that the absence of invariant NKT (iNKT) cells in mice during IAV infection resulted in the expansion of myeloid-derived suppressor cells (MDSCs), which suppressed IAV-specific immune responses through the expression of both arginase and NOS, resulting in high IAV titer and increased mortality. Adoptive transfer of iNKT cells abolished the suppressive activity of MDSCs, restored IAV-specific immune responses, reduced IAV titer, and increased survival rate. The crosstalk between iNKT and MDSCs was CD1d- and CD40-dependent. Furthermore, IAV infection and exposure to TLR agonists relieved the suppressive activity of MDSCs. Finally, we extended these results to humans by demonstrating the presence of myeloid cells with suppressive activity in the PBLs of individuals infected with IAV and showed that their suppressive activity is substantially reduced by iNKT cell activation. These findings identify what we believe to be a novel immunomodulatory role of iNKT cells, which we suggest could be harnessed to abolish the immunosuppressive activity of MDSCs during IAV infection.


Subject(s)
Arginase/immunology , Immune Tolerance , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Myeloid Cells/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Arginase/genetics , Arginase/metabolism , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cells, Cultured , Humans , Immune Tolerance/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza, Human/enzymology , Influenza, Human/epidemiology , Influenza, Human/genetics , Influenza, Human/pathology , Mice , Mice, Knockout , Myeloid Cells/enzymology , Myeloid Cells/pathology , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/pathology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
17.
Proc Natl Acad Sci U S A ; 105(24): 8345-50, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18550831

ABSTRACT

Highly regulated activation of B cells is required for the production of specific antibodies necessary to provide protection from pathogen infection. This process is initiated by specific recognition of antigen through the B cell receptor (BCR), leading to early intracellular signaling followed by the late recruitment of T cell help. In this study we demonstrate that specific BCR uptake of CD1d-restricted antigens represents an effective means of enhancing invariant natural killer T (iNKT)-dependent B cell responses in vivo. This mechanism is effective over a wide range of antigen affinities but depends on exceeding a tightly regulated avidity threshold necessary for BCR-mediated internalization and CD1d-dependent presentation of particulate antigenic lipid. Subsequently, iNKT cells provide the help required for stimulating B cell proliferation and differentiation. iNKT-stimulated B cells develop within extrafollicular foci and mediate the production of high titers of specific IgM and early class-switched antibodies. Thus, we have demonstrated that in response to particulate antigenic lipids iNKT cells are recruited for the assistance of B cell activation, resulting in the enhancement of specific antibody responses. We propose that such a mechanism may operate to potentiate adaptive immune responses against pathogens in vivo.


Subject(s)
Antibody Formation , Antigens, CD1/metabolism , B-Lymphocytes/immunology , Killer Cells, Natural/immunology , Receptors, Antigen, B-Cell/metabolism , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation , Antigens, CD1d , Cell Line , Galactosylceramides/immunology , Immunization , Lymphocyte Activation , Mice , Mice, Inbred Strains
18.
Front Oncol ; 11: 674720, 2021.
Article in English | MEDLINE | ID: mdl-34094976

ABSTRACT

Tumor cells require a higher supply of nutrients for growth and proliferation than normal cells. It is well established that metabolic reprograming in cancers for increased nutrient supply exposes a host of targetable vulnerabilities. In this article we review the documented changes in expression patterns of amino acid metabolic enzymes and transporters in myeloid malignancies and the growing list of small molecules and therapeutic strategies used to disrupt amino acid metabolic circuits within the cell. Pharmacological inhibition of amino acid metabolism is effective in inducing cell death in leukemic stem cells and primary blasts, as well as in reducing tumor burden in in vivo murine models of human disease. Thus targeting amino acid metabolism provides a host of potential translational opportunities for exploitation to improve the outcomes for patients with myeloid malignancies.

19.
J Clin Invest ; 116(10): 2777-90, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016559

ABSTRACT

Active suppression of tumor-specific T lymphocytes can limit the efficacy of immune surveillance and immunotherapy. While tumor-recruited CD11b+ myeloid cells are known mediators of tumor-associated immune dysfunction, the true nature of these suppressive cells and the fine biochemical pathways governing their immunosuppressive activity remain elusive. Here we describe a population of circulating CD11b+IL-4 receptor alpha+ (CD11b+IL-4Ralpha+), inflammatory-type monocytes that is elicited by growing tumors and activated by IFN-gamma released from T lymphocytes. CD11b+IL-4Ralpha+ cells produced IL-13 and IFN-gamma and integrated the downstream signals of these cytokines to trigger the molecular pathways suppressing antigen-activated CD8+ T lymphocytes. Analogous immunosuppressive circuits were active in CD11b+ cells present within the tumor microenvironment. These suppressor cells challenge the current idea that tumor-conditioned immunosuppressive monocytes/macrophages are alternatively activated. Moreover, our data show how the inflammatory response elicited by tumors had detrimental effects on the adaptive immune system and suggest novel approaches for the treatment of tumor-induced immune dysfunctions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Monocytes/immunology , Neoplasms/immunology , Animals , Arginase/genetics , Arginase/metabolism , CD11b Antigen/analysis , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Gene Expression/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Cellular/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-13/metabolism , Interleukin-13/pharmacology , Interleukin-4/genetics , Interleukin-4/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Monocytes/drug effects , Monocytes/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptors, Cell Surface/analysis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
20.
EBioMedicine ; 47: 235-246, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31462392

ABSTRACT

BACKGROUND: Targeting of MDSCs is a major clinical challenge in the era of immunotherapy. Antibodies which deplete MDSCs in murine models can reactivate T cell responses. In humans such approaches have not developed due to difficulties in identifying targets amenable to clinical translation. METHODS: RNA-sequencing of M-MDSCs and G-MDSCs from cancer patients was undertaken. Flow cytometry and immunohistochemistry of blood and tumours determined MDSC CD33 expression. MDSCs were treated with Gemtuzumab ozogamicin and internalisation kinetics, and cell death mechanisms determined by flow cytometry, confocal microscopy and electron microscopy. Effects on T cell proliferation and CAR-T cell anti-tumour cytotoxicity were identified in the presence of Gemtuzumab ozogamicin. FINDINGS: RNA-sequencing of human M-MDSCs and G-MDSCs identified transcriptomic differences, but that CD33 is a common surface marker. Flow cytometry indicated CD33 expression is higher on M-MDSCs, and CD33+ MDSCs are found in the blood and tumours regardless of cancer subtype. Treatment of human MDSCs leads to Gemtuzumab ozogamicin internalisation, increased p-ATM, and cell death; restoring T cell proliferation. Anti-GD2-/mesothelin-/EGFRvIII-CAR-T cell activity is enhanced in combination with the anti-MDSC effects of Gemtuzumab ozogamicin. INTERPRETATION: The study identifies that M-MDSCs and G-MDSCs are transcriptomically different but CD33 is a therapeutic target on peripheral and infiltrating MDSCs across cancer subtypes. The immunotoxin Gemtuzumab ozogamicin can deplete MDSCs providing a translational approach to reactivate T cell and CAR-T cell responses against multiple cancers. In the rare conditions of HLH/MAS gemtuzumab ozogamicin provides a novel anti-myeloid strategy. FUND: This work was supported by Cancer Research UK, CCLG, Treating Children with Cancer, and the alumni and donors to the University of Birmingham.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Gemtuzumab/pharmacology , Myeloid-Derived Suppressor Cells/drug effects , Neoplasms/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers , Gemtuzumab/therapeutic use , Gene Expression Profiling , Humans , Immunohistochemistry , Immunophenotyping , Immunotherapy , Models, Biological , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , T-Lymphocytes/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL