Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
2.
Cell Rep Med ; 4(3): 100971, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36871558

ABSTRACT

Identifying the molecular mechanisms that promote optimal immune responses to coronavirus disease 2019 (COVID-19) vaccination is critical for future rational vaccine design. Here, we longitudinally profile innate and adaptive immune responses in 102 adults after the first, second, and third doses of mRNA or adenovirus-vectored COVID-19 vaccines. Using a multi-omics approach, we identify key differences in the immune responses induced by ChAdOx1-S and BNT162b2 that correlate with antigen-specific antibody and T cell responses or vaccine reactogenicity. Unexpectedly, we observe that vaccination with ChAdOx1-S, but not BNT162b2, induces an adenoviral vector-specific memory response after the first dose, which correlates with the expression of proteins with roles in thrombosis with potential implications for thrombosis with thrombocytopenia syndrome (TTS), a rare but serious adverse event linked to adenovirus-vectored vaccines. The COVID-19 Vaccine Immune Responses Study thus represents a major resource that can be used to understand the immunogenicity and reactogenicity of these COVID-19 vaccines.


Subject(s)
COVID-19 Vaccines , COVID-19 , Vaccines , Adult , Humans , Adenoviridae/genetics , Antibodies , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , RNA, Messenger/genetics
3.
Cell Rep Med ; 2(12): 100464, 2021 12 21.
Article in English | MEDLINE | ID: mdl-35028606

ABSTRACT

Immune agonist antibodies (IAAs) are promising immunotherapies that target co-stimulatory receptors to induce potent anti-tumor immune responses, particularly when combined with checkpoint inhibitors. Unfortunately, their clinical translation is hampered by serious dose-limiting, immune-mediated toxicities, including high-grade and sometimes fatal liver damage, cytokine release syndrome (CRS), and colitis. We show that the immunotoxicity, induced by the IAAs anti-CD40 and anti-CD137, is dependent on the gut microbiota. Germ-free or antibiotic-treated mice have significantly reduced colitis, CRS, and liver damage following IAA treatment compared with conventional mice or germ-free mice recolonized via fecal microbiota transplant. MyD88 signaling is required for IAA-induced CRS and for anti-CD137-induced, but not anti-CD40-induced, liver damage. Importantly, antibiotic treatment does not impair IAA anti-tumor efficacy, alone or in combination with anti-PD1. Our results suggest that microbiota-targeted therapies could overcome the toxicity induced by IAAs without impairing their anti-tumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , CD40 Antigens/immunology , Gastrointestinal Microbiome , Immunotherapy/adverse effects , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Anti-Bacterial Agents/pharmacology , Bile Acids and Salts/metabolism , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Inflammation/pathology , Interferon Type I/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/immunology , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
4.
Br J Cancer ; 122(5): 680-691, 2020 03.
Article in English | MEDLINE | ID: mdl-31919402

ABSTRACT

BACKGROUND: Predictive preclinical models play an important role in the assessment of new treatment strategies and as avatar models for personalised medicine; however, reliable and timely model generation is challenging. We investigated the feasibility of establishing patient-derived xenograft (PDX) models of high-risk neuroblastoma from a range of tumour-bearing patient materials and assessed approaches to improve engraftment efficiency. METHODS: PDX model development was attempted in NSG mice by using tumour materials from 12 patients, including primary and metastatic solid tumour samples, bone marrow, pleural fluid and residual cells from cytogenetic analysis. Subcutaneous, intramuscular and orthotopic engraftment were directly compared for three patients. RESULTS: PDX models were established for 44% (4/9) of patients at diagnosis and 100% (5/5) at relapse. In one case, attempted engraftment from pleural fluid resulted in an EBV-associated atypical lymphoid proliferation. Xenogeneic graft versus host disease was observed with attempted engraftment from lymph node and bone marrow tumour samples but could be prevented by T-cell depletion. Orthotopic engraftment was more efficient than subcutaneous or intramuscular engraftment. CONCLUSIONS: High-risk neuroblastoma PDX models can be reliably established from diverse sample types. Orthotopic implantation allows more rapid model development, increasing the likelihood of developing an avatar model within a clinically useful timeframe.


Subject(s)
Neoplasm Transplantation/methods , Neuroblastoma/pathology , Neuroblastoma/therapy , Precision Medicine/methods , Xenograft Model Antitumor Assays/methods , Animals , Feasibility Studies , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Neuroblastoma/genetics , Random Allocation , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
Cancer Res ; 79(3): 611-624, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30545920

ABSTRACT

Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)-dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1ß and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)-dependent manner. IL1ß and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest-derived cells. Proteomic analysis revealed that enrichment of IL1ß and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited. SIGNIFICANCE: These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression.


Subject(s)
Arginine/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Neuroblastoma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Arginase/metabolism , Cell Line, Tumor , Humans , Interleukin-1beta/immunology , MAP Kinase Signaling System , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neuroblastoma/immunology , Neuroblastoma/pathology , Sarcoma, Ewing/immunology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/immunology
6.
Eur J Cancer ; 83: 132-141, 2017 09.
Article in English | MEDLINE | ID: mdl-28735070

ABSTRACT

The ATP-binding cassette transporter ABCC4 (multidrug resistance protein 4, MRP4) mRNA level is a strong predictor of poor clinical outcome in neuroblastoma which may relate to its export of endogenous signalling molecules and chemotherapeutic agents. We sought to determine whether ABCC4 contributes to development, growth and drug response in neuroblastoma in vivo. In neuroblastoma patients, high ABCC4 protein levels were associated with reduced overall survival. Inducible knockdown of ABCC4 strongly inhibited the growth of human neuroblastoma cells in vitro and impaired the growth of neuroblastoma xenografts. Loss of Abcc4 in the Th-MYCN transgenic neuroblastoma mouse model did not impact tumour formation; however, Abcc4-null neuroblastomas were strongly sensitised to the ABCC4 substrate drug irinotecan. Our findings demonstrate a role for ABCC4 in neuroblastoma cell proliferation and chemoresistance and provide rationale for a strategy where inhibition of ABCC4 should both attenuate the growth of neuroblastoma and sensitise tumours to ABCC4 chemotherapeutic substrates.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Multidrug Resistance-Associated Proteins/deficiency , Neuroblastoma/drug therapy , Animals , Blotting, Western , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Doxycycline/pharmacology , Heterografts/drug effects , Irinotecan , Mice , Mice, Knockout , Multidrug Resistance-Associated Proteins/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
World J Gastroenterol ; 22(37): 8322-8333, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27729739

ABSTRACT

AIM: To investigate the effects of orally gavaged aqueous rhubarb extract (RE) on 5-fluorouracil (5-FU)-induced intestinal mucositis in rats. METHODS: Female Dark Agouti rats (n = 8/group) were gavaged daily (1 mL) with water, high-dose RE (HDR; 200 mg/kg) or low-dose RE (LDR; 20mg/kg) for eight days. Intestinal mucositis was induced (day 5) with 5-FU (150 mg/kg) via intraperitoneal injection. Intestinal tissue samples were collected for myeloperoxidase (MPO) activity and histological examination. Xenopus oocytes expressing aquaporin 4 water channels were prepared to examine the effect of aqueous RE on cell volume, indicating a potential mechanism responsible for modulating net fluid absorption and secretion in the gastrointestinal tract. Statistical significance was assumed at P < 0.05 by one-way ANOVA. RESULTS: Bodyweight was significantly reduced in rats administered 5-FU compared to healthy controls (P < 0.01). Rats administered 5-FU significantly increased intestinal MPO levels (≥ 307%; P < 0.001), compared to healthy controls. However, LDR attenuated this effect in 5-FU treated rats, significantly decreasing ileal MPO activity (by 45%; P < 0.05), as compared to 5-FU controls. 5-FU significantly reduced intestinal mucosal thickness (by ≥ 29% P < 0.001) as compared to healthy controls. LDR significantly increased ileal mucosal thickness in 5-FU treated rats (19%; P < 0.05) relative to 5-FU controls. In xenopus oocytes expressing AQP4 water channels, RE selectively blocked water influx into the cell, induced by a decrease in external osmotic pressure. As water efflux was unaltered by the presence of extracellular RE, the directional flow of water across the epithelial barrier, in the presence of extracellular RE, indicated that RE may alleviate water loss across the epithelial barrier and promote intestinal health in chemotherapy-induced intestinal mucositis. CONCLUSION: In summary, low dose RE improves selected parameters of mucosal integrity and reduces ileal inflammation, manifesting from 5-FU-induced intestinal mucositis.


Subject(s)
Antineoplastic Agents/adverse effects , Fluorouracil/adverse effects , Intestinal Mucosa/physiopathology , Mucositis/physiopathology , Plant Extracts/chemistry , Rheum/chemistry , Animals , Aquaporin 4/metabolism , Body Weight , Feces , Female , Inflammation/pathology , Intestinal Mucosa/pathology , Oocytes/metabolism , Peroxidase/metabolism , Rats , Xenopus
8.
Nutr Cancer ; 68(7): 1171-80, 2016 10.
Article in English | MEDLINE | ID: mdl-27618153

ABSTRACT

Chemotherapy-induced mucositis is characterized by inflammation and ulcerating lesions lining the alimentary tract. Emu Oil and Lyprinol™ have independently demonstrated their therapeutic potential in intestinal inflammatory disorders, including mucositis. We investigated Emu Oil and Lyprinol™ in combination for their further potential to alleviate chemotherapy-induced mucositis in rats. Rats were gavaged with (1 ml) water, Olive Oil, Emu Oil + Olive Oil, Lyprinol™ + Olive Oil or Emu Oil + Lyprinol™ from Days 0 to 7, injected with saline (control) or 5-Fluorouracil (5-FU) on Day 5 and euthanized on Day 8. Myeloperoxidase (MPO) activity (indicative of acute inflammation), histological severity scores, and intestinal architecture were quantified. Myeloperoxidase activity was significantly increased in the jejunum and ileum following 5-FU, compared to saline controls. Both Olive Oil and Emu Oil + Lyprinol™ significantly reduced jejunal MPO levels (1.8-fold and 1.7-fold, respectively), whereas only Emu Oil + Lyprinol™ significantly decreased ileal MPO levels, relative to 5-FU controls. All oil treatments decreased histological severity scores in the jejunum and ileum, and normalized crypt depth in the mid small intestine, relative to 5-FU controls. Emu Oil combined with Lyprinol™ partially reduced acute small intestinal inflammation. Isolating bioactive constituents of these naturally sourced oils could provide a more targeted strategy to protect against intestinal mucositis.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Dietary Supplements , Enteritis/prevention & control , Fluorouracil/adverse effects , Lipids/therapeutic use , Mucositis/prevention & control , Oils/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Dasyproctidae , Dietary Supplements/analysis , Enteritis/chemically induced , Enteritis/immunology , Enteritis/metabolism , Female , Gastrointestinal Agents/chemistry , Gastrointestinal Agents/therapeutic use , Ileum/drug effects , Ileum/immunology , Ileum/metabolism , Ileum/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Jejunum/drug effects , Jejunum/immunology , Jejunum/metabolism , Jejunum/pathology , Lipids/chemistry , Mucositis/chemically induced , Mucositis/immunology , Mucositis/metabolism , Oils/chemistry , Olive Oil/chemistry , Olive Oil/therapeutic use , Organ Size/drug effects , Protective Agents/chemistry , Protective Agents/therapeutic use , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL