Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 217
Filter
2.
Mol Med ; 27(1): 49, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34022793

ABSTRACT

A SARS-like coronavirus 2 (SARS-CoV-2) has caused a pandemic Coronavirus Disease 2019 (COVID-19) that killed more than 3.3 million people worldwide. Like the SARS-CoV, SARS-CoV-2 also employs a receptor-binding motif (RBM) of its spike protein to bind a host receptor, the angiotensin-converting enzyme 2 (ACE2), to gain entry. Currently, several mRNA or adenoviral vaccines encoding for the spike protein of SARS-CoV-2 are being used to boost antibodies capable of inhibiting spike-ACE2 interaction and viral entry. However, recent evidence has also suggested an anti-inflammatory effect of spike-reactive antibodies, suggesting that some SARS-CoV-2 spike-based vaccines may elicit protective antibodies capable of inhibiting GM-CSF production and COVID-19 progression.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Neutralizing/metabolism , COVID-19 Vaccines/therapeutic use , COVID-19/prevention & control , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/metabolism , Antibodies, Neutralizing/immunology , COVID-19/metabolism , COVID-19/virology , COVID-19 Vaccines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Host-Pathogen Interactions/drug effects , Humans , Protein Binding/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/immunology , Virus Internalization/drug effects , Virus Replication/drug effects
3.
Nat Commun ; 12(1): 1032, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589587

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is a devastating lung disease caused by abnormal surfactant homeostasis, with a prevalence of 6-7 cases per million population worldwide. While mutations causing hereditary PAP have been reported, the genetic basis contributing to autoimmune PAP (aPAP) has not been thoroughly investigated. Here, we conducted a genome-wide association study of aPAP in 198 patients and 395 control participants of Japanese ancestry. The common genetic variant, rs138024423 at 6p21, in the major-histocompatibility-complex (MHC) region was significantly associated with disease risk (Odds ratio [OR] = 5.2; P = 2.4 × 10-12). HLA fine-mapping revealed that the common HLA class II allele, HLA-DRB1*08:03, strongly drove this signal (OR = 4.8; P = 4.8 × 10-12), followed by an additional independent risk allele at HLA-DPß1 amino acid position 8 (OR = 0.28; P = 3.4 × 10-7). HLA-DRB1*08:03 was also associated with an increased level of anti-GM-CSF antibody, a key driver of the disease (ß = 0.32; P = 0.035). Our study demonstrated a heritable component of aPAP, suggesting an underlying genetic predisposition toward an abnormal antibody production.


Subject(s)
Autoantibodies/genetics , Autoimmune Diseases/genetics , Genetic Predisposition to Disease , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , HLA-DRB1 Chains/genetics , Pulmonary Alveolar Proteinosis/genetics , Adult , Aged , Alleles , Asian People , Autoantibodies/biosynthesis , Autoimmune Diseases/ethnology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Case-Control Studies , Chromosomes, Human, Pair 6 , Female , Gene Expression , Gene Frequency , Genome-Wide Association Study , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HLA-DRB1 Chains/immunology , Humans , Japan , Male , Middle Aged , Odds Ratio , Protein Isoforms/genetics , Pulmonary Alveolar Proteinosis/ethnology , Pulmonary Alveolar Proteinosis/immunology , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Surfactants/immunology , Pulmonary Surfactants/metabolism , Risk
4.
Cancer Invest ; 39(4): 310-314, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33428503

ABSTRACT

Diagnosis by biopsy is difficult in the ovary since it is located deep in the abdomen. As a result, ovarian cancer is mostly found insidiously during exploratory laparotomy. Consequently, the early diagnosis of ovarian cancer is often difficult. The likelihood of peritoneal dissemination increases with the progress of ovarian cancer. With further progression, ovarian cancer metastasizes to the momentum, retroperitoneal lymph nodes, large intestine, small intestine, diaphragm, spleen, and other organs. Ovarian cancer has been considered a tumor that has a favorable response to chemotherapy, but more effective treatments are still being explored. Tumors use their own immune escape mechanism to evade host immunity. The immune checkpoint (IC) mechanism, one of the immune escape mechanisms, is established by programmed cell death-1 (PD-1)/PD-ligand-1 (PD-L1) communication. It has been shown that inhibiting PD-1/PD-L1 communication in various malignancies produces antitumor effects. However, the antitumor effect of ICI monotherapy on ovarian cancer is limited in actual clinical practice. In this review, we describe a novel cancer immunotherapeutic agent that targets myeloid-derived suppressor cells (MDSCs).


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Myeloid-Derived Suppressor Cells/drug effects , Ovarian Neoplasms/drug therapy , Tumor Escape/drug effects , Animals , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/adverse effects , Drug Resistance, Neoplasm , Female , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immune Checkpoint Inhibitors/adverse effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
5.
Sci Rep ; 10(1): 19516, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33177636

ABSTRACT

Health problems caused by airborne particulate matter with a diameter less than 2.5 (PM2.5), especially in the respiratory system, have become a worldwide problem, but the influence and mechanisms of PM2.5 on the ocular surface have not been sufficiently elucidated. We investigated in vitro the onset and pathogenesis of corneal damage induced by PM2.5. Two types of PM2.5 samples originating from Beijing (designated #28) and the Gobi Desert (designated #30) were added to the culture medium of immortalized cultured human corneal epithelial cells (HCECs) to examine the effects on survival rates, autophagy, and proinflammatory cytokine production. Both types of PM2.5 significantly reduced the HCEC survival rate in a concentration-dependent manner by triggering autophagy. In particular, compared with #30, #28 induced much more severe damage in HCECs. Physical contact between PM2.5 and HCECs was not a primary contributor to PM2.5-induced HCEC damage. Among the 38 proinflammatory cytokines examined in this study, significant increases in the granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-6 levels and a significant reduction in the interleukin-8 level were detected in culture medium of PM2.5-exposed HCECs. Simultaneous addition of a GM-CSF inhibitor, suramin, alleviated the HCEC impairment induced by PM2.5. In conclusion, PM2.5 induces HCEC death by triggering autophagy. Some cytokines that are released from HCECs, including GM-CSF, may be involved in HCEC damage caused by PM2.5 exposure.


Subject(s)
Air Pollutants/toxicity , Cornea/drug effects , Cornea/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Particulate Matter/chemistry , Particulate Matter/toxicity , Autophagy , Cell Line , Cell Survival , China , Cornea/cytology , Epithelial Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-2/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Interleukin-8/metabolism , Suramin/pharmacology , Tumor Necrosis Factor-alpha/metabolism
6.
Mayo Clin Proc ; 95(11): 2382-2394, 2020 11.
Article in English | MEDLINE | ID: mdl-33153629

ABSTRACT

OBJECTIVE: To assess the efficacy and safety of lenzilumab in patients with severe coronavirus disease 2019 (COVID-19) pneumonia. METHODS: Hospitalized patients with COVID-19 pneumonia and risk factors for poor outcomes were treated with lenzilumab 600 mg intravenously for three doses through an emergency single-use investigational new drug application. Patient characteristics, clinical and laboratory outcomes, and adverse events were recorded. We also identified a cohort of patients matched to the lenzilumab patients for age, sex, and disease severity. Study dates were March 13, 2020, to June 18, 2020. All patients were followed through hospital discharge or death. RESULTS: Twelve patients were treated with lenzilumab; 27 patients comprised the matched control cohort (untreated). Clinical improvement, defined as improvement of at least 2 points on the 8-point ordinal clinical endpoints scale, was observed in 11 of 12 (91.7%) patients treated with lenzilumab and 22 of 27 (81.5%) untreated patients. The time to clinical improvement was significantly shorter for the lenzilumab-treated group compared with the untreated cohort with a median of 5 days versus 11 days (P=.006). Similarly, the proportion of patients with acute respiratory distress syndrome (oxygen saturation/fraction of inspired oxygen<315 mm Hg) was significantly reduced over time when treated with lenzilumab compared with untreated (P<.001). Significant improvement in inflammatory markers (C-reactive protein and interleukin 6) and markers of disease severity (absolute lymphocyte count) were observed in patients who received lenzilumab, but not in untreated patients. Cytokine analysis showed a reduction in inflammatory myeloid cells 2 days after lenzilumab treatment. There were no treatment-emergent adverse events attributable to lenzilumab. CONCLUSION: In high-risk COVID-19 patients with severe pneumonia, granulocyte-macrophage colony-stimulating factor neutralization with lenzilumab was safe and associated with faster improvement in clinical outcomes, including oxygenation, and greater reductions in inflammatory markers compared with a matched control cohort of patients hospitalized with severe COVID-19 pneumonia. A randomized, placebo-controlled clinical trial to validate these findings is ongoing (NCT04351152).


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , COVID-19 Drug Treatment , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , SARS-CoV-2 , Aged , COVID-19/epidemiology , COVID-19/metabolism , Dose-Response Relationship, Drug , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Infusions, Intravenous , Male , Middle Aged , Pandemics , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 117(44): 27141-27147, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33024017

ABSTRACT

The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has highlighted the urgent need to rapidly develop therapeutic strategies for such emerging viruses without effective vaccines or drugs. Here, we report a decoy nanoparticle against COVID-19 through a powerful two-step neutralization approach: virus neutralization in the first step followed by cytokine neutralization in the second step. The nanodecoy, made by fusing cellular membrane nanovesicles derived from human monocytes and genetically engineered cells stably expressing angiotensin converting enzyme II (ACE2) receptors, possesses an antigenic exterior the same as source cells. By competing with host cells for virus binding, these nanodecoys effectively protect host cells from the infection of pseudoviruses and authentic SARS-CoV-2. Moreover, relying on abundant cytokine receptors on the surface, the nanodecoys efficiently bind and neutralize inflammatory cytokines including interleukin 6 (IL-6) and granulocyte-macrophage colony-stimulating factor (GM-CSF), and significantly suppress immune disorder and lung injury in an acute pneumonia mouse model. Our work presents a simple, safe, and robust antiviral nanotechnology for ongoing COVID-19 and future potential epidemics.


Subject(s)
Coronavirus Infections/therapy , Cytokines/antagonists & inhibitors , Nanoparticles/therapeutic use , Pneumonia, Viral/therapy , Virus Internalization/drug effects , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus , COVID-19 , Cell Membrane/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , HEK293 Cells , Humans , Interleukin-6/antagonists & inhibitors , Mice , Mice, Inbred ICR , Monocytes , Nanoparticles/chemistry , Pandemics , Peptidyl-Dipeptidase A/metabolism , Receptors, Cytokine/metabolism , SARS-CoV-2 , THP-1 Cells
8.
Front Immunol ; 11: 1625, 2020.
Article in English | MEDLINE | ID: mdl-32719685

ABSTRACT

COVID-19 is a clinical syndrome ranging from mild symptoms to severe pneumonia that often leads to respiratory failure, need for mechanical ventilation, and death. Most of the lung damage is driven by a surge in inflammatory cytokines [interleukin-6, interferon-γ, and granulocyte-monocyte stimulating factor (GM-CSF)]. Blunting this hyperinflammation with immunomodulation may lead to clinical improvement. GM-CSF is produced by many cells, including macrophages and T-cells. GM-CSF-derived signals are involved in differentiation of macrophages, including alveolar macrophages (AMs). In animal models of respiratory infections, the intranasal administration of GM-CSF increased the proliferation of AMs and improved outcomes. Increased levels of GM-CSF have been recently described in patients with COVID-19 compared to healthy controls. While GM-CSF might be beneficial in some circumstances as an appropriate response, in this case the inflammatory response is maladaptive by virtue of being later and disproportionate. The inhibition of GM-CSF signaling may be beneficial in improving the hyperinflammation-related lung damage in the most severe cases of COVID-19. This blockade can be achieved through antagonism of the GM-CSF receptor or the direct binding of circulating GM-CSF. Initial findings from patients with COVID-19 treated with a single intravenous dose of mavrilimumab, a monoclonal antibody binding GM-CSF receptor α, showed oxygenation improvement and shorter hospitalization. Prospective, randomized, placebo-controlled trials are ongoing. Anti-GM-CSF monoclonal antibodies, TJ003234 and gimsilumab, will be tested in clinical trials in patients with COVID-19, while lenzilumab received FDA approval for compassionate use. These trials will help inform whether blunting the inflammatory signaling provided by the GM-CSF axis in COVID-19 is beneficial.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Betacoronavirus/immunology , Coronavirus Infections , Drug Delivery Systems , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Pandemics , Pneumonia, Viral , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , SARS-CoV-2 , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
10.
Immunotherapy ; 12(15): 1121-1126, 2020 10.
Article in English | MEDLINE | ID: mdl-32546029

ABSTRACT

Background: Coronavirus disease 2019 (COVID-19) is a novel disease associated with a cytokine-mediated, severe, acute respiratory syndrome. Tocilizumab and lenzilumab are recombinant monoclonal antibodies against IL-6 and granulocyte macrophage colony-stimulating factor, respectively, and have been proposed as a potential treatment for acute, hypoxic respiratory failure associated with COVID-19. Results & methodology: We present the case of a 68-year-old man with COVID-19 who was initially treated with hydroxychloroquine and lenzilumab, but continued to develop hypoxemia, requiring an increase in respiratory support with an associated rise in serum inflammatory markers. He was subsequently treated with tocilizumab with marked clinical improvement and a decrease in acute phase reactants within 48 h. Discussion & conclusion: This case demonstrates the effective use of tocilizumab in the treatment of COVID-19 and suggests the superiority of tocilizumab over lenzilumab in the management of this cytokine-mediated syndrome.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Coronavirus Infections/therapy , Pneumonia, Viral/therapy , Aged , Betacoronavirus , COVID-19 , Combined Modality Therapy , Coronavirus Infections/drug therapy , Coronavirus Infections/pathology , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Male , Pandemics , Pneumonia, Viral/pathology , Receptors, Interleukin-6/antagonists & inhibitors , Respiratory Insufficiency/pathology , Respiratory Insufficiency/therapy , SARS-CoV-2 , Treatment Outcome , COVID-19 Drug Treatment
11.
Lancet Respir Med ; 8(8): 822-830, 2020 08.
Article in English | MEDLINE | ID: mdl-32559419

ABSTRACT

The COVID-19 pandemic is a global public health crisis, with considerable mortality and morbidity exerting pressure on health-care resources, including critical care. An excessive host inflammatory response in a subgroup of patients with severe COVID-19 might contribute to the development of acute respiratory distress syndrome (ARDS) and multiorgan failure. Timely therapeutic intervention with immunomodulation in patients with hyperinflammation could prevent disease progression to ARDS and obviate the need for invasive ventilation. Granulocyte macrophage colony-stimulating factor (GM-CSF) is an immunoregulatory cytokine with a pivotal role in initiation and perpetuation of inflammatory diseases. GM-CSF could link T-cell-driven acute pulmonary inflammation with an autocrine, self-amplifying cytokine loop leading to monocyte and macrophage activation. This axis has been targeted in cytokine storm syndromes and chronic inflammatory disorders. Here, we consider the scientific rationale for therapeutic targeting of GM-CSF in COVID-19-associated hyperinflammation. Since GM-CSF also has a key role in homoeostasis and host defence, we discuss potential risks associated with inhibition of GM-CSF in the context of viral infection and the challenges of doing clinical trials in this setting, highlighting in particular the need for a patient risk-stratification algorithm.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Immunologic Factors/therapeutic use , Pneumonia, Viral/drug therapy , Respiratory Distress Syndrome/prevention & control , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/virology , Disease Progression , Humans , Immunomodulation , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/virology , Respiratory Distress Syndrome/virology , SARS-CoV-2 , COVID-19 Drug Treatment
12.
J Craniofac Surg ; 31(5): 1483-1487, 2020.
Article in English | MEDLINE | ID: mdl-32502102

ABSTRACT

The present study is based on the concept of neuro-aging and how it may affect surrounding skin cells. It has been shown that many factors play a significant role in skin homeostasis by interfering with various cytokines, either through activation or inhibition. Granulocyte macrophage colony-stimulating factor (GM-CSF) is generally recognized as an inflammatory cytokine, and our previous study has shown its effects on neuronal senescence after ultraviolet (UV) irradiation of skin cells. Following our previous work, this study was performed to investigate the neuroprotective effects of a GM-CSF antagonist, and how it may play an essential role in mediating anti-senescence and anti-inflammatory effects in the keratinocyte/nerve aging model. When human blastoma cells (SH-SY5Y) were treated with 10 ng/ml of GM-CSF, the levels of regulatory RNAs associated with aging, such as matrix metalloproteinase-9 (MMP9), nuclear factor NF-kappa-B p50 subunit (NFKB), inducible nitric oxide synthase (iNOS), and interleukin 1 beta (IL-1ß) increased, whereas GM-CSF inhibition caused their expression to decrease. A decrease in the antioxidant, glutathione (GSH) was observed after SH-SY5Y cells were treated with GM-CSF. This study confirms that this GM-CSF antagonist may play an important role in neural senescence, where inhibition may be a new target in the skin/nerve aging model.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Neurons/drug effects , Skin/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-1beta/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B p50 Subunit/metabolism , Nerve Tissue/drug effects , Nerve Tissue/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , Skin/metabolism , Skin Aging/drug effects , Ultraviolet Rays
13.
J Neuroimmunol ; 345: 577281, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32534388

ABSTRACT

To identify the mechanisms relevant for the therapeutic effect of glatiramer acetate (GA), we studied T- and B- regulatory cells as well as GM-CSF expression in mice recovered from experimental autoimmune encephalomyelitis (EAE). Selective depletion of Tregs reduced but did not eliminate the ability of GA to ameliorate EAE, indicating a role for additional immune-subsets. The prevalence of Bregs in the periphery and the CNS of EAE-mice increased following GA-treatment. Furthermore, GA downregulated the pathological expression of GM-CSF, on both the protein and mRNA levels. These findings corroborate the broad immunomodulatory mechanism of action of GA in EAE/MS.


Subject(s)
B-Lymphocytes, Regulatory/metabolism , Glatiramer Acetate/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunosuppressive Agents/pharmacology , Multiple Sclerosis/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , B-Lymphocytes, Regulatory/drug effects , Disease Models, Animal , Female , Glatiramer Acetate/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/drug therapy , T-Lymphocytes, Regulatory/drug effects
15.
J Immunother Cancer ; 8(1)2020 05.
Article in English | MEDLINE | ID: mdl-32385146

ABSTRACT

The pandemic caused by the novel coronavirus SARS-CoV-2 has placed an unprecedented burden on healthcare systems around the world. In patients who experience severe disease, acute respiratory distress is often accompanied by a pathological immune reaction, sometimes referred to as 'cytokine storm'. One hallmark feature of the profound inflammatory state seen in patients with COVID-19 who succumb to pneumonia and hypoxia is marked elevation of serum cytokines, especially interferon gamma, tumor necrosis factor alpha, interleukin 17 (IL-17), interleukin 8 (IL-8) and interleukin 6 (IL-6). Initial experience from the outbreaks in Italy, China and the USA has anecdotally demonstrated improved outcomes for critically ill patients with COVID-19 with the administration of cytokine-modulatory therapies, especially anti-IL-6 agents. Although ongoing trials are investigating anti-IL-6 therapies, access to these therapies is a concern, especially as the numbers of cases worldwide continue to climb. An immunology-informed approach may help identify alternative agents to modulate the pathological inflammation seen in patients with COVID-19. Drawing on extensive experience administering these and other immune-modulating therapies, the Society for Immunotherapy of Cancer offers this perspective on potential alternatives to anti-IL-6 that may also warrant consideration for management of the systemic inflammatory response and pulmonary compromise that can be seen in patients with severe COVID-19.


Subject(s)
Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Immunotherapy , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , Pneumonia, Viral/complications , Pneumonia, Viral/drug therapy , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/drug therapy , Societies, Medical , Adoptive Transfer , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Inflammation/complications , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/antagonists & inhibitors , Interleukin-1/antagonists & inhibitors , Interleukin-17/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinases/antagonists & inhibitors , Neoplasms/immunology , Neoplasms/therapy , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , STAT Transcription Factors/antagonists & inhibitors , Severe Acute Respiratory Syndrome/pathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors
16.
Cancer Immunol Immunother ; 69(8): 1477-1492, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32285172

ABSTRACT

The interactions between tumor immune microenvironment (TIME) and pancreatic cancer cells can affect chemotherapeutic efficacy; however, the mechanisms still remain largely unknown. Thirty items in TIME were comprehensively screened by using tissue microarray from pancreatic cancer patients. Their expressions, interconnections and predictive roles for survival were analyzed. Twenty-one of 30 items could stratify the survival of the patients; however, multivariate analysis found that only 5 independent risk factors could predict worse survival (M2-polarized tumor-associated macrophages (TAMs), IgG4 positive cells, TGF-ß1, GM-CSF and lymphangiogenesis). They had a much higher expression levels in tumoral tissue, compared to peritumoral tissue. The Spearman analysis showed that M2-polarized TAM, TGF-ß1 and GM-CSF were positively correlated with pancreatic cancer stem cells (PCSC), angiogenesis and lymphangiogenesis. Both human and murine pancreatic cancer cells could induce M2-polarized TAM, which showed substantial roles to decease chemotherapeutic effects. After treated by gemcitabine, both human and murine pancreatic cancer cell lines expressed higher level of immune check points, PCSC markers and varieties of immunosuppressive factors; however, TGF-ß1 and GM-CSF had the highest increase. Based on the above results, TGF-ß1 and GM-CSF were proposed to be the optimal potential targets to improve chemotherapeutic effects. In immunocompetent murine models, we demonstrated that combined blockade of TGF-ß1 and GM-CSF improved the chemotherapeutic effects by inhibition of M2-polarized TAM and induction of CD8 positive T cells. This study presents a novel promising combined strategy to improve the chemotherapeutic effects for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/drug therapy , Transforming Growth Factor beta1/antagonists & inhibitors , Tumor Microenvironment/drug effects , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Cohort Studies , Deoxycytidine/pharmacology , Female , Gene Expression Regulation, Neoplastic , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymphangiogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Survival Rate , Transforming Growth Factor beta1/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
17.
Sci Rep ; 10(1): 1567, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005854

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a multipotent cytokine that prompts the proliferation of bone marrow-derived macrophages and granulocytes. In addition to its effects as a growth factor, GM-CSF plays an important role in chronic inflammatory autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. Reports have identified monocytes as the primary target of GM-CSF; however, its effect on monocyte activation has been under-estimated. Here, using flow cytometry and ELISA we show that GM-CSF induces an inflammatory profile in human monocytes, which includes an upregulated expression of HLA-DR and CD86 molecules and increased production of TNF-α and IL-1ß. Conversely, blockage of endogenous GM-CSF with antibody treatment not only inhibited the inflammatory profile of these cells, but also induced an immunomodulatory one, as shown by increased IL-10 production by monocytes. Further analysis with qPCR, flow cytometry and ELISA experiments revealed that GM-CSF blockage in monocytes stimulated production of the chemokine CXCL-11, which suppressed T cell proliferation. Blockade of CXCL-11 abrogated anti-GM-CSF treatment and induced inflammatory monocytes. Our findings show that anti-GM-CSF treatment induces modulatory monocytes that act in a CXCL-11-dependent manner, a mechanism that can be used in the development of novel approaches to treat chronic inflammatory autoimmune diseases.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Monocytes/drug effects , B7-2 Antigen/metabolism , Chemokine CXCL11/antagonists & inhibitors , Chemokine CXCL11/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , HLA-DR Antigens/metabolism , Humans , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Monocytes/metabolism , Monocytes/physiology , Phenotype , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
18.
Osteoarthritis Cartilage ; 28(4): 486-491, 2020 04.
Article in English | MEDLINE | ID: mdl-32028021

ABSTRACT

OBJECTIVES: Pharmacological options for treating osteoarthritis (OA) are limited and alternative treatments are required. Given the clinical data indicating that granulocyte macrophage-colony stimulating factor (GM-CSF) may be a therapeutic target in human OA, we evaluated different treatment regimens with a neutralizing anti-GM-CSF monoclonal antibody (mAb) in an experimental OA model to determine their effectiveness on amelioration of pain and disease. METHODS: The collagenase-induced osteoarthritis (CiOA) model was induced in C57BL/6 mice, followed by different treatment regimens of anti-GM-CSF mAb or isotype control. Anti-CCL17 mAb treatment was also administered continually during the late stage of CiOA. Pain-related behavior (change in weight distribution of hind limbs), and disease (cartilage damage and osteophyte size) were assessed. RESULTS: Blocking GM-CSF only during early synovitis in CiOA prevented pain and disease development. Once OA pain was established, regardless of the treatment regimen, anti-GM-CSF mAb treatment rapidly and efficiently ameliorated it; however, unless the treatment was continued, pain returned and disease progressed. Continual late stage blockade of GM-CSF was able to ameliorate pain (between-group difference: -6.567; 95% confidence interval (CI): -10.12, -3.011) and suppress cartilage damage (P = 0.0317, 95% CI: -1.75, -0.0556). Continual late stage blockade of CCL17 showed similar effects on pain and disease development. CONCLUSIONS: Early and short-term GM-CSF neutralization is effective at preventing CiOA pain and disease development but, once pain is evident, continual GM-CSF blockade is required to prevent pain from returning and to suppress disease progression in mice. These data reinforce the potential benefits of anti-GM-CSF (and anti-CCL17) mAb therapy in OA and should inform further clinical trials.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cartilage, Articular/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Osteoarthritis, Knee/pathology , Stifle/drug effects , Synovial Membrane/drug effects , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Chemokine CCL17/antagonists & inhibitors , Collagenases/toxicity , Disease Progression , Early Medical Intervention , Injections, Intra-Articular , Mice , Osteoarthritis, Knee/chemically induced , Osteophyte/pathology , Pain Measurement , Stifle/pathology , Synovial Membrane/pathology , Synovitis/pathology
19.
J Immunol ; 204(4): 923-932, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900338

ABSTRACT

The transcription factor BHLHE40 is an emerging regulator of the immune system. Recent studies suggest that BHLHE40 regulates type 2 immunity, but this has not been demonstrated in vivo. We found that BHLHE40 is required in T cells for a protective TH2 cell response in mice infected with the helminth Heligmosomoides polygyrus bakeri H. polygyrus elicited changes in gene and cytokine expression by lamina propria CD4+ T cells, many of which were BHLHE40 dependent, including production of the common ß (CSF2RB) chain family cytokines GM-CSF and IL-5. In contrast to deficiency in GM-CSF or IL-5 alone, loss of both GM-CSF and IL-5 signaling impaired protection against H. polygyrus Overall, we show that BHLHE40 regulates the TH2 cell transcriptional program during helminth infection to support normal expression of Csf2, Il5, and other genes required for protection and reveal unexpected redundancy of common ß chain-dependent cytokines previously thought to possess substantially divergent functions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Homeodomain Proteins/metabolism , Interleukin-5/metabolism , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/metabolism , Disease Models, Animal , Female , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Homeodomain Proteins/genetics , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Interleukin-5/antagonists & inhibitors , Interleukin-5/genetics , Interleukin-5/immunology , Mice , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Strongylida Infections/parasitology , Th2 Cells/drug effects , Transcription, Genetic/immunology
20.
Arterioscler Thromb Vasc Biol ; 39(7): 1275-1287, 2019 07.
Article in English | MEDLINE | ID: mdl-31092009

ABSTRACT

Growth factors, such as CSFs (colony-stimulating factors), EGFs (epidermal growth factors), and FGFs (fibroblast growth factors), are signaling proteins that control a wide range of cellular functions. Although growth factor networks are critical for intercellular communication and tissue homeostasis, their abnormal production or regulation occurs in various pathologies. Clinical strategies that target growth factors or their receptors are used to treat a variety of conditions but have yet to be adopted for cardiovascular disease. In this review, we focus on M-CSF (macrophage-CSF), GM-CSF (granulocyte-M-CSF), IL (interleukin)-3, EGFR (epidermal growth factor receptor), and FGF21 (fibroblast growth factor 21). We first discuss the efficacy of targeting these growth factors in other disease contexts (ie, inflammatory/autoimmune diseases, cancer, or metabolic disorders) and then consider arguments for or against targeting them to treat cardiovascular disease. Visual Overview- An online visual overview is available for this article.


Subject(s)
Cardiovascular Diseases/drug therapy , ErbB Receptors/antagonists & inhibitors , Fibroblast Growth Factors/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Immunotherapy , Interleukin-3/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...