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










Publication year range
1.
Oncoimmunology ; 11(1): 2077898, 2022.
Article in English | MEDLINE | ID: mdl-35655707

ABSTRACT

The past decades witnessed the clinical employment of targeted therapies including but not limited to tyrosine kinase inhibitors (TKIs) that restrain a broad variety of pro-tumorigenic signals. TKIs can be categorized into (i) agents that directly target cancer cells, (ii) normalize angiogenesis or (iii) affect cells of the hematologic lineage. However, a clear distinction of TKIs based on this definition is limited by the fact that many TKIs designed to inhibit cancer cells have also effects on immune cells that are being discovered. Additionally, TKIs originally designed to target hematological cancers exhibit bioactivities on healthy cells of the same hematological lineage. TKIs have been described to improve immune recognition and cancer immunosurveillance, providing the scientific basis to combine TKIs with immunotherapy. Indeed, combination of TKIs with immunotherapy showed synergistic effects in preclinical models and clinical trials and some combinations of TKIs normalizing angiogenesis with immune checkpoint blocking antibodies have already been approved by the FDA for cancer therapy. However, the identification of appropriate drug combinations as well as optimal dosing and scheduling needs to be improved in order to obtain tangible progress in cancer care. This Trial Watch summarizes active clinical trials combining TKIs with various immunotherapeutic strategies to treat cancer patients.


Subject(s)
Immunotherapy , Neoplasms , Clinical Trials as Topic , Humans , Immunologic Factors/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Oncoimmunology ; 10(1): 1973197, 2021.
Article in English | MEDLINE | ID: mdl-34712511

ABSTRACT

Immunogenic cell death (ICD) has initially been discovered in the context of chemotherapy. High-dose crizotinib also stimulates ICD, as we described for non-small cell lung cancer lacking activating chromosomal aberrations of ALK or ROS1, the usual targets of crizotinib, indicating that crizotinib may act through off-target effects. However, we found that low-dose of ALK inhibitors, crizotinib and ceritinib, may stimulate ICD in anaplastic large cell lymphoma, in which ALK is activated due to a chromosomal translocation, suggesting on target ICD-promoting effects.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Crizotinib/therapeutic use , Humans , Immunogenic Cell Death , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Pyrimidines , Sulfones
3.
Autophagy ; 13(3): 567-578, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28059587

ABSTRACT

Starvation is a strong physiological stimulus of macroautophagy/autophagy. In this study, we addressed the question as to whether it would be possible to measure autophagy in blood cells after nutrient deprivation. Fasting of mice for 48 h (which causes ∼20% weight loss) or starvation of human volunteers for up to 4 d (which causes <2% weight loss) provokes major changes in the plasma metabolome, yet induces only relatively minor alterations in the intracellular metabolome of circulating leukocytes. White blood cells from mice and human volunteers responded to fasting with a marked reduction in protein lysine acetylation, affecting both nuclear and cytoplasmic compartments. In circulating leukocytes from mice that underwent 48-h fasting, an increase in LC3B lipidation (as assessed by immunoblotting and immunofluorescence) only became detectable if the protease inhibitor leupeptin was injected 2 h before drawing blood. Consistently, measurement of an enhanced autophagic flux was only possible if white blood cells from starved human volunteers were cultured in the presence or absence of leupeptin. Whereas all murine leukocyte subpopulations significantly increased the number of LC3B+ puncta per cell in response to nutrient deprivation, only neutrophils from starved volunteers showed signs of activated autophagy (as determined by a combination of multi-color immunofluorescence, cytofluorometry and image analysis). Altogether, these results suggest that white blood cells are suitable for monitoring autophagic flux. In addition, we propose that the evaluation of protein acetylation in circulating leukocytes can be adopted as a biochemical marker of organismal energetic status.


Subject(s)
Fasting/blood , Fasting/metabolism , Acetylation , Adult , Animals , Autophagy , Cells, Cultured , Female , Humans , Lysine/metabolism , Male , Metabolome , Metabolomics , Mice, Inbred C57BL , Middle Aged , Neutrophils/metabolism , Starvation/blood , Starvation/metabolism , Young Adult
4.
Methods Mol Biol ; 1419: 1-16, 2016.
Article in English | MEDLINE | ID: mdl-27108427

ABSTRACT

Different modes of regulated cell death (RCD) can be initiated by distinct molecular machineries and their morphological manifestations can be difficult to discriminate. Moreover, cells responding to stress often activate an adaptive response centered around autophagy, and whether such a response is cytoprotective or cytotoxic cannot be predicted based on morphological parameters only. Molecular definitions are therefore important to understand various RCD subroutines from a mechanistic perspective. In vitro, various forms of RCD including apoptosis and autophagic cell death can be easily discriminated from each other with assays that involve chemical or pharmacological interventions targeting key components of either pathway. Here, we detail a straightforward method to discriminate apoptosis from autophagic cell death by flow cytometry, based on the broad-spectrum caspase inhibitor Z-VAD-fmk and the genetic inhibition of ATG5.


Subject(s)
Apoptosis , Autophagy , Flow Cytometry/methods , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Humans
5.
6.
Nat Cell Biol ; 10(6): 676-87, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18454141

ABSTRACT

Multiple cellular stressors, including activation of the tumour suppressor p53, can stimulate autophagy. Here we show that deletion, depletion or inhibition of p53 can induce autophagy in human, mouse and nematode cells subjected to knockout, knockdown or pharmacological inhibition of p53. Enhanced autophagy improved the survival of p53-deficient cancer cells under conditions of hypoxia and nutrient depletion, allowing them to maintain high ATP levels. Inhibition of p53 led to autophagy in enucleated cells, and cytoplasmic, not nuclear, p53 was able to repress the enhanced autophagy of p53(-/-) cells. Many different inducers of autophagy (for example, starvation, rapamycin and toxins affecting the endoplasmic reticulum) stimulated proteasome-mediated degradation of p53 through a pathway relying on the E3 ubiquitin ligase HDM2. Inhibition of p53 degradation prevented the activation of autophagy in several cell lines, in response to several distinct stimuli. These results provide evidence of a key signalling pathway that links autophagy to the cancer-associated dysregulation of p53.


Subject(s)
Autophagy , Cytoplasm/metabolism , Gene Expression Regulation , Genes, p53 , Tumor Suppressor Protein p53/physiology , Animals , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Humans , Hypoxia , Lysosomes/metabolism , Mice , Mice, Transgenic , Models, Biological , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Methods Mol Biol ; 445: 29-76, 2008.
Article in English | MEDLINE | ID: mdl-18425442

ABSTRACT

Autophagic (or type 2) cell death is characterized by the massive accumulation of autophagic vacuoles (autophagosomes) in the cytoplasm of cells that lack signs of apoptosis (type 1 cell death). Here we detail and critically assess a series of methods to promote and inhibit autophagy via pharmacological and genetic manipulations. We also review the techniques currently available to detect autophagy, including transmission electron microscopy, half-life assessments of long-lived proteins, detection of LC3 maturation/aggregation, fluorescence microscopy, and colocalization of mitochondrion- or endoplasmic reticulum-specific markers with lysosomal proteins. Massive autophagic vacuolization may cause cellular stress and represent a frustrated attempt of adaptation. In this case, cell death occurs with (or in spite of) autophagy. When cell death occurs through autophagy, on the contrary, the inhibition of the autophagic process should prevent cellular demise. Accordingly, we describe a strategy for discriminating cell death with autophagy from cell death through autophagy.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Animals , Cell Death/physiology , Endoplasmic Reticulum/ultrastructure , HeLa Cells , Humans , Lysosomes/ultrastructure , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mitochondria/ultrastructure , Phagosomes/ultrastructure
8.
Cell Cycle ; 6(18): 2263-7, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17890908

ABSTRACT

When added to cells, a variety of autophagy inducers that operate through distinct mechanisms and target different organelles for autophagic destruction (mitochondria in mitophagy, endoplasmic reticulum in reticulophagy) rarely induce autophagic vacuolization in more than 50% or the cells. Here we show that this heterogeneity may be explained by cell cycle-specific effects. The BH3 mimetic ABT737, lithium, rapamycin, tunicamycin or nutrient depletion stereotypically induce autophagy preferentially in the G(1) and S phases of the cell cycle, as determined by simultaneous monitoring of cell cycle markers and the cytoplasmic aggregation of GFP-LC3 in autophagic vacuoles. These results point to a hitherto neglected crosstalk between autophagic vacuolization and cell cycle regulation.


Subject(s)
Autophagy/physiology , Cell Cycle/physiology , Endoplasmic Reticulum/physiology , Mitochondria/physiology , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Endoplasmic Reticulum/pathology , HCT116 Cells , Humans , Mitochondria/pathology , Rats
9.
Nat Rev Mol Cell Biol ; 8(9): 741-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17717517

ABSTRACT

The functional relationship between apoptosis ('self-killing') and autophagy ('self-eating') is complex in the sense that, under certain circumstances, autophagy constitutes a stress adaptation that avoids cell death (and suppresses apoptosis), whereas in other cellular settings, it constitutes an alternative cell-death pathway. Autophagy and apoptosis may be triggered by common upstream signals, and sometimes this results in combined autophagy and apoptosis; in other instances, the cell switches between the two responses in a mutually exclusive manner. On a molecular level, this means that the apoptotic and autophagic response machineries share common pathways that either link or polarize the cellular responses.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Signal Transduction , Animals , Cell Cycle , Humans
10.
Nat Med ; 13(9): 1050-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704786

ABSTRACT

Conventional cancer treatments rely on radiotherapy and chemotherapy. Such treatments supposedly mediate their effects via the direct elimination of tumor cells. Here we show that the success of some protocols for anticancer therapy depends on innate and adaptive antitumor immune responses. We describe in both mice and humans a previously unrecognized pathway for the activation of tumor antigen-specific T-cell immunity that involves secretion of the high-mobility-group box 1 (HMGB1) alarmin protein by dying tumor cells and the action of HMGB1 on Toll-like receptor 4 (TLR4) expressed by dendritic cells (DCs). During chemotherapy or radiotherapy, DCs require signaling through TLR4 and its adaptor MyD88 for efficient processing and cross-presentation of antigen from dying tumor cells. Patients with breast cancer who carry a TLR4 loss-of-function allele relapse more quickly after radiotherapy and chemotherapy than those carrying the normal TLR4 allele. These results delineate a clinically relevant immunoadjuvant pathway triggered by tumor cell death.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , Toll-Like Receptor 4/immunology , Animals , Bone Neoplasms/drug therapy , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Organoplatinum Compounds/therapeutic use , Osteosarcoma/drug therapy , Pyridines/therapeutic use
11.
Autophagy ; 3(4): 350-3, 2007.
Article in English | MEDLINE | ID: mdl-17404493

ABSTRACT

The second messenger myo-inositol-1,4,5-trisphosphate (IP(3)) acts on the IP(3) receptor (IP(3)R), an IP(3)-activated Ca(2+) channel of the endoplasmic reticulum (ER). The IP(3)R agonist IP(3) inhibits starvation-induced autophagy. The IP(3)R antagonist xestospongin B induces autophagy in human cells through a pathway that requires the obligate contribution of Beclin-1, Atg5, Atg10, Atg12 and hVps34, yet is inhibited by ER-targeted Bcl-2 or Bcl-XL, two proteins that physically interact with IP(3)R. Autophagy can also be induced by depletion of the IP(3)R by small interfering RNAs. Autophagy induction by IP(3)R blockade cannot be explained by changes in steady state levels of Ca(2+) in the endoplasmic reticulum (ER) and the cytosol. Autophagy induction by IP(3)R blockade is effective in cells lacking the obligate mediator of ER stress IRE1. In contrast, IRE1 is required for autophagy induced by ER stress-inducing agents such a tunicamycin or thapsigargin. These findings suggest that there are several distinct pathways through which autophagy can be initiated at the level of the ER.


Subject(s)
Autophagy/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Anti-Bacterial Agents/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Food Deprivation , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Models, Biological , Oxazoles/pharmacology , RNA, Small Interfering/metabolism , Thapsigargin/pharmacology , Tunicamycin/pharmacology
12.
EMBO J ; 26(10): 2527-39, 2007 May 16.
Article in English | MEDLINE | ID: mdl-17446862

ABSTRACT

The anti-apoptotic proteins Bcl-2 and Bcl-X(L) bind and inhibit Beclin-1, an essential mediator of autophagy. Here, we demonstrate that this interaction involves a BH3 domain within Beclin-1 (residues 114-123). The physical interaction between Beclin-1 and Bcl-X(L) is lost when the BH3 domain of Beclin-1 or the BH3 receptor domain of Bcl-X(L) is mutated. Mutation of the BH3 domain of Beclin-1 or of the BH3 receptor domain of Bcl-X(L) abolishes the Bcl-X(L)-mediated inhibition of autophagy triggered by Beclin-1. The pharmacological BH3 mimetic ABT737 competitively inhibits the interaction between Beclin-1 and Bcl-2/Bcl-X(L), antagonizes autophagy inhibition by Bcl-2/Bcl-X(L) and hence stimulates autophagy. Knockout or knockdown of the BH3-only protein Bad reduces starvation-induced autophagy, whereas Bad overexpression induces autophagy in human cells. Gain-of-function mutation of the sole BH3-only protein from Caenorhabditis elegans, EGL-1, induces autophagy, while deletion of EGL-1 compromises starvation-induced autophagy. These results reveal a novel autophagy-stimulatory function of BH3-only proteins beyond their established role as apoptosis inducers. BH3-only proteins and pharmacological BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin-1 and Bcl-2 or Bcl-X(L).


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Membrane Proteins/chemistry , Proteins/chemistry , bcl-X Protein/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/ultrastructure , Autophagy , Beclin-1 , Blotting, Western , Cells, Cultured , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , HeLa Cells , Humans , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Mice , Mutation , Precipitin Tests , Protein Structure, Tertiary/genetics , Proteins/metabolism , Proteins/ultrastructure , RNA Interference , Two-Hybrid System Techniques , bcl-X Protein/ultrastructure
13.
Apoptosis ; 12(1): 3-18, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17080328

ABSTRACT

HeLa and HCT116 cells respond differentially to sorbitol, an osmolyte able to induce hypertonic stress. In these models, sorbitol promoted the phenotypic manifestations of early apoptosis followed by complete loss of viability in a time-, dose-, and cell type-specific fashion, by eliciting distinct yet partially overlapping molecular pathways. In HCT116 but not in HeLa cells, sorbitol caused the mitochondrial release of the caspase-independent death effector AIF, whereas in both cell lines cytochrome c was retained in mitochondria. Despite cytochrome c retention, HeLa cells exhibited the progressive activation of caspase-3, presumably due to the prior activation of caspase-8. Accordingly, caspase inhibition prevented sorbitol-induced killing in HeLa, but only partially in HCT116 cells. Both the knock-out of Bax in HCT116 cells and the knock-down of Bax in A549 cells by RNA interference reduced the AIF release and/or the mitochondrial alterations. While the knock-down of Bcl-2/Bcl-X(L) sensitized to sorbitol-induced killing, overexpression of a Bcl-2 variant that specifically localizes to mitochondria (but not of the wild-type nor of a endoplasmic reticulum-targeted form) strongly inhibited sorbitol effects. Thus, hyperosmotic stress kills cells by triggering different molecular pathways, which converge at mitochondria where pro- and anti-apoptotic members of the Bcl-2 family exert their control.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Apoptosis Inducing Factor/biosynthesis , Base Sequence , Cell Line , Cytochromes c/metabolism , HeLa Cells , Humans , Osmotic Pressure , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/genetics , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...