Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 73(4): 788-802.e7, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30704899

ABSTRACT

mTORC1 and GSK3 play critical roles in early stages of (macro)autophagy, but how they regulate late steps of autophagy remains poorly understood. Here we show that mTORC1 and GSK3-TIP60 signaling converge to modulate autophagosome maturation through Pacer, an autophagy regulator that was identified in our recent study. Hepatocyte-specific Pacer knockout in mice results in impaired autophagy flux, glycogen and lipid accumulation, and liver fibrosis. Under nutrient-rich conditions, mTORC1 phosphorylates Pacer at serine157 to disrupt the association of Pacer with Stx17 and the HOPS complex and thus abolishes Pacer-mediated autophagosome maturation. Importantly, dephosphorylation of Pacer under nutrient-deprived conditions promotes TIP60-mediated Pacer acetylation, which facilitates HOPS complex recruitment and is required for autophagosome maturation and lipid droplet clearance. This work not only identifies Pacer as a regulator in hepatic autophagy and liver homeostasis in vivo but also reveals a signal integration mechanism involved in late stages of autophagy and lipid metabolism.


Subject(s)
Autophagosomes/enzymology , Autophagy-Related Proteins/metabolism , Autophagy , Glycogen Synthase Kinase 3/metabolism , Lipid Metabolism , Liver/enzymology , Lysine Acetyltransferase 5/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphate-Binding Proteins/metabolism , Trans-Activators/metabolism , Acetylation , Animals , Autophagosomes/pathology , Autophagy-Related Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Glycogen Synthase Kinase 3/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipid Droplets/metabolism , Liver/pathology , Lysine Acetyltransferase 5/genetics , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Membrane Proteins , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphate-Binding Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Signal Transduction , Trans-Activators/genetics , Tumor Suppressor Proteins
2.
Cell ; 143(5): 711-24, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21074248

ABSTRACT

PI3K and PTEN lipid phosphatase control the level of cellular phosphatidylinositol (3,4,5)-trisphosphate, an activator of AKT kinases that promotes cell growth and survival. Mutations activating AKT are commonly observed in human cancers. We report here that ENTPD5, an endoplasmic reticulum (ER) enzyme, is upregulated in cell lines and primary human tumor samples with active AKT. ENTPD5 hydrolyzes UDP to UMP to promote protein N-glycosylation and folding in ER. Knockdown of ENTPD5 in PTEN null cells causes ER stress and loss of growth factor receptors. ENTPD5, together with cytidine monophosphate kinase-1 and adenylate kinase-1, constitute an ATP hydrolysis cycle that converts ATP to AMP, resulting in a compensatory increase in aerobic glycolysis known as the Warburg effect. The growth of PTEN null cells is inhibited both in vitro and in mouse xenograft tumor models. ENTPD5 is therefore an integral part of the PI3K/PTEN regulatory loop and a potential target for anticancer therapy.


Subject(s)
Cell Proliferation , Endoplasmic Reticulum/metabolism , Glycosylation , Oncogene Proteins/metabolism , Adenosine Triphosphate/metabolism , Aerobiosis , Animals , Cell Line, Tumor , Glycolysis , Guanosine Monophosphate/metabolism , Humans , Mice , Neoplasm Transplantation , Oncogene Protein v-akt/metabolism , Oncogene Proteins/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pyrophosphatases , Transplantation, Heterologous , Uridine Monophosphate/metabolism
3.
Mol Cell ; 65(6): 1029-1043.e5, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28306502

ABSTRACT

Class III PI3-kinase (PI3KC3) is essential for autophagy initiation, but whether PI3KC3 participates in other steps of autophagy remains unknown. The HOPS complex mediates the fusion of intracellular vesicles to lysosome, but how HOPS specifically tethers autophagosome to lysosome remains elusive. Here, we report Pacer (protein associated with UVRAG as autophagy enhancer) as a regulator of autophagy. Pacer localizes to autophagic structures and positively regulates autophagosome maturation. Mechanistically, Pacer antagonizes Rubicon to stimulate Vps34 kinase activity. Next, Pacer recruits PI3KC3 and HOPS complexes to the autophagosome for their site-specific activation by anchoring to the autophagosomal SNARE Stx17. Furthermore, Pacer is crucial for the degradation of hepatic lipid droplets, the suppression of Salmonella infection, and the clearance of protein aggregates. These results not only identify Pacer as a crucial multifunctional enhancer in autophagy but also uncover both the involvement of PI3KC3 and the mediators of HOPS's specific tethering activity in autophagosome maturation.


Subject(s)
Autophagosomes/enzymology , Autophagy-Related Proteins/metabolism , Autophagy , Class III Phosphatidylinositol 3-Kinases/metabolism , Qa-SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism , Autophagy-Related Proteins/genetics , Endosomes/enzymology , Enzyme Activation , HEK293 Cells , HeLa Cells , Hep G2 Cells , Hepatocytes/enzymology , Host-Pathogen Interactions , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Droplets/metabolism , Lysosomes/enzymology , Membrane Fusion , Protein Aggregates , Protein Binding , Protein Interaction Domains and Motifs , Qa-SNARE Proteins/genetics , RNA Interference , Salmonella typhimurium/growth & development , Signal Transduction , Time Factors , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vesicular Transport Proteins/genetics
4.
Gastric Cancer ; 25(5): 943-955, 2022 09.
Article in English | MEDLINE | ID: mdl-35778636

ABSTRACT

BACKGROUND: In solid tumor Phase 1/2 trials (NCT02407990; NCT04068519), tislelizumab demonstrated clinical benefit, including in advanced gastroesophageal adenocarcinoma (GEA). However, the majority of patients with GEA did not respond, highlighting the need to understand mechanisms of resistance and identify predictive biomarkers for response. METHODS: All tislelizumab-treated patients with GEA from the Phase 1/2 trials were included (N = 105). Programmed death-ligand 1 (PD-L1) expression (Tumor Area Positivity [TAP] ≥ 5%), interferon gamma (IFNγ)-related gene signature, gene expression profile, tumor mutational burden (TMB), and gene hyperamplification (HA) were analyzed for correlation with tislelizumab. RESULTS: A moderate association was observed between PD-L1 TAP ≥ 5%, IFNγ gene signature, TMB-high and efficacy. A potential correlation between hyperamplification (HA +) and worse outcomes with programmed cell death protein 1 (PD-1) inhibition was identified. Hyperamplified genes were mainly enriched in cancer progression pathways, including cell cycle and RTK-RAS-PI3K pathways. Joint PD-L1 TAP ≥ 5% and lack of hyperamplification showed the most favorable benefit with an objective response rate of 29.4%, and median progression-free survival and overall survival of 4.1 and 14.7 months, respectively. Tumors with TAP ≥ 5% and HA - had inflamed immune signatures with increased immune cell infiltration, enhanced anti-tumor cytotoxic activity and antigen presentation signatures. Findings were validated in two independent gastric and gastrointestinal cancer cohorts treated with immune checkpoint inhibitors. CONCLUSIONS: In GEA, PD-L1 positivity, IFNγ-related gene signature and TMB-high status were positively associated with tislelizumab clinical benefit, whereas HA was associated with worse clinical outcomes. Combining PD-L1 positivity and HA - may help identify patients more likely to benefit from PD-1 blockade.


Subject(s)
Adenocarcinoma , Lung Neoplasms , Stomach Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Antibodies, Monoclonal, Humanized , B7-H1 Antigen , Biomarkers, Tumor/genetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Esophageal Neoplasms , Esophagogastric Junction/pathology , Humans , Lung Neoplasms/drug therapy , Mutation , Phosphatidylinositol 3-Kinases/genetics , Programmed Cell Death 1 Receptor/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
5.
Future Oncol ; 17(22): 2893-2905, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34189951

ABSTRACT

Aims: To explore the prognostic value of high PD-L1 expression on tumor cells (TC) and tumor-infiltrating immune cells (TIIC) in urothelial carcinoma (UC). Patients & methods: 162 UC specimens were evaluated for PD-L1 expression on TIIC and TC with the SP263 assay. High PD-L1 expression was defined as ≥25% staining. Results: High PD-L1 expression on TC in UC patients with stage T1-4 disease was associated with poor overall survival. However, high PD-L1 expression on TIIC in UC patients with stage T1-4 disease revealed favorable disease-free and overall survival; more significant differences were observed in patients with stages T2-4. Multivariate analysis revealed that high PD-L1 expression on TIIC was an independent prognostic predictor for better disease-free and overall survival. Conclusion: High PD-L1 expression on TIIC, but not on TC, is a favorable prognostic factor in UC.


Lay abstract Bladder cancer is the tenth most common form of cancer worldwide, and urothelial carcinoma is the most common type of bladder cancer. PD-L1 is a protein that can be expressed on the surface of many tissue types, including tumor cells (TC) and tumor-infiltrating immune cells (TIIC). PD-L1 can help the tumor evade the body's natural immune defense system. The expression of PD-L1 not only related to the response of immunotherapy but is also associated with the prognosis in bladder cancer. However, the prognostic significance of PD-L1 expression on TC and TIIC remains controversial. This study drew a conclusion that high PD-L1 expression on TIIC, but not on TC, is a favorable prognostic factor in urothelial carcinoma.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/mortality , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasm Recurrence, Local/epidemiology , Urinary Bladder Neoplasms/mortality , Adult , Aged , B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Biopsy , Carcinoma, Transitional Cell/diagnosis , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/surgery , Cystectomy , Disease-Free Survival , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Staging , Prognosis , Retrospective Studies , Risk Assessment/methods , Tumor Microenvironment/immunology , Urinary Bladder/immunology , Urinary Bladder/pathology , Urinary Bladder/surgery , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/surgery
6.
Nature ; 506(7489): E4-6, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24572428

ABSTRACT

Sirtuins can promote deacetylation of a wide range of substrates in diverse cellular compartments and regulate many cellular processes¹,². Recently Narayan et al., reported that SIRT2 was required for necroptosis based on their findings that SIRT2 inhibition, knock-down or knock-out prevented necroptosis. We sought to confirm and explore the role of SIRT2 in necroptosis and tested four different sources of the SIRT2 inhibitor AGK2, three independent siRNAs against SIRT2, and cells from two independently generated Sirt2−/− mouse strains, however we were unable to show that inhibiting or depleting SIRT2 protected cells from necroptosis. Furthermore, Sirt2−/− mice succumbed to TNF induced Systemic Inflammatory Response Syndrome (SIRS) more rapidly than wild type mice while Ripk3−/− mice were resistant. Our results therefore question the importance of SIRT2 in the necroptosis cell death pathway.


Subject(s)
Necrosis/enzymology , Sirtuin 2/genetics , Sirtuin 2/metabolism , Animals , Female , Humans , Male
7.
Nucleic Acids Res ; 45(22): e180, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29036617

ABSTRACT

Forward genetic screens using mammalian embryonic stem (ES) cells have identified genes required for numerous cellular processes. However, loss-of-function screens are more difficult to conduct in diploid cells because, in most cases, both alleles of a gene must be mutated to exhibit a phenotype. Recently, mammalian haploid ES cell lines were successfully established and applied to several recessive genetic screens. However, all these screens were performed in mixed pools of mutant cells and were mainly based on positive selection. In general, negative screening is not easy to apply to these mixed pools, although quantitative deep sequencing of mutagen insertions can help to identify some 'missing' mutants. Moreover, the interplay between different mutant cells in the mixed pools would interfere with the readout of the screens. Here, we developed a method for rapidly generating arrayed haploid mutant libraries in which the proportion of homozygous mutant clones can reach 85%. After screening thousands of individual mutant clones, we identified a number of novel factors required for the onset of differentiation in ES cells. A negative screen was also conducted to discover mutations conferring cells with increased sensitivity to DNA double-strand breaks induced by the drug doxorubicin. Both of these screens illustrate the value of this system.


Subject(s)
Genetic Testing/methods , Haploidy , Mouse Embryonic Stem Cells/metabolism , Mutation , Animals , Cell Line , Cells, Cultured , Clone Cells/cytology , Clone Cells/metabolism , DNA Transposable Elements/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Mutagenesis, Insertional/methods , Phenotype
8.
Proc Natl Acad Sci U S A ; 112(16): 5017-22, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25852146

ABSTRACT

Receptor-interacting protein kinase 3, RIP3, and a pseudokinase mixed lineage kinase-domain like protein, MLKL, constitute the core components of the necroptosis pathway, which causes programmed necrotic death in mammalian cells. Latent RIP3 in the cytosol is activated by several upstream signals including the related kinase RIP1, which transduces signals from the tumor necrosis factor (TNF) family of cytokines. We report here that RIP3 activation following the induction of necroptosis requires the activity of an HSP90 and CDC37 cochaperone complex. This complex physically associates with RIP3. Chemical inhibitors of HSP90 efficiently block necroptosis by preventing RIP3 activation. Cells with knocked down CDC37 were unable to respond to necroptosis stimuli. Moreover, an HSP90 inhibitor that is currently under clinical development as a cancer therapy was able to prevent systemic inflammatory response syndrome in rats treated with TNF-α. HSP90 and CDC37 cochaperone complex-mediated protein folding is thus an important part of the RIP3 activation process during necroptosis.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Cytosol/metabolism , HSP90 Heat-Shock Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Macrophages/metabolism , Mice , Necrosis , Protein Binding , Protein Kinases/metabolism , Rats
9.
Proc Natl Acad Sci U S A ; 112(32): 9990-5, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26216950

ABSTRACT

The empirical criteria for defining a clinical subtype of lung cancer are gradually transiting from histopathology to genetic variations in driver genes. Targeting these driver mutations, such as sensitizing epidermal growth factor receptor (EGFR) mutations, has dramatically improved the prognosis of advanced non-small cell lung cancer (NSCLC). However, the clinical benefit of molecularly targeted therapy on NSCLC appears to be different between lung adenocarcinomas and squamous cell carcinomas (SqCCs). We report here that the resistance of lung SqCC harboring EGFR mutations to EGFR tyrosine kinase inhibitors (EGFR-TKIs) was due to the activation of BMP-BMPR-Smad1/5-p70S6K. The combined treatment of these tumor cells with EGFR-TKI, together with inhibitors specific to BMPR or downstream mTOR, effectively reversed the resistance to EGFR-TKI. Moreover, blocking the whole PI3K-AKT-mTOR pathway with the PI3K/mTOR dual inhibitor BEZ235 also showed efficacy in treating this subtype of lung SqCC. This study details the empirical basis for a feasible clinical solution for squamous cell carcinomas with EGFR mutations.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Carcinoma, Squamous Cell/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/drug therapy , Female , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Male , Mice, Nude , Middle Aged , Multivariate Analysis , Reproducibility of Results , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Smad Proteins/metabolism
10.
Cell Mol Life Sci ; 73(11-12): 2177-81, 2016 06.
Article in English | MEDLINE | ID: mdl-27066893

ABSTRACT

Necroptosis has been extensively studied recently, and the receptor-interacting kinase 3 (RIP3 or RIPK3) and its substrate, the pseudokinase mixed lineage kinase domain-like protein, have been discovered to be core components of this process. Classical necroptosis requires RIP1 (or RIPK1) for the activation of RIP3 through the induction of RIP1/RIP3 necrosomes. Increasing evidence from genetic and pharmacological studies has been expanding the view that necroptosis plays important roles in the etiology and/or progression of many human diseases, such as pancreatitis, ischemic injury, and neurodegenerative diseases, among others. Ongoing progress in translational research about necroptosis has highlighted the increasingly important need for the identification of biomarkers for use in disease diagnosis, monitoring, and drug development. This review presents a discussion of the current status of biomarkers that can be used to detect necroptosis both in vitro and in vivo.


Subject(s)
Apoptosis/physiology , Necrosis/pathology , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Biomarkers/metabolism , Cytokines/metabolism , Humans , Mice , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 111(41): 14782-7, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25275009

ABSTRACT

Intrinsic apoptotic stimuli initiate mammalian cells' apoptotic program by first activating the proteins that have only Bcl-2 homology domain 3 (BH3), such as Bcl-2 interacting mediator of cell death (Bim) and truncated BH3 interacting death domain agonist (tBid), which in turn trigger conformational changes in BCL2-associated X (Bax) and BCL2-antagonist/killer (Bak) proteins that enable oligomer formation on the mitochondria, causing cytochrome c and other apoptogenic proteins in the intermembrane space to leak out. Leaked cytochrome c then initiates apoptotic caspase activation through a well-defined biochemical pathway. However, how oligomerized Bax and Bak cause cytochrome c release from mitochondria remains unknown. We report here the establishment of cell lines in which Bim or tBid can be inducibly expressed to initiate apoptosis in a controlled, quantitative manner. We used these cell lines to examine apoptotic events after Bax and Bak oligomerization but before cytochrome c release. The mitochondrial metalloprotease OMA1 was activated in this system in a Bax- and Bak-dependent fashion. Activated OMA1 cleaved the dynamin-like GTPase, optical nerve atrophy 1, an event that is critical for remodeling of mitochondrial cristae. Knockdown or knockout of OMA1 in these cells attenuated cytochrome c release. Thus it is clear that oligomerized Bax and Bak trigger apoptosis by causing both the permeabilization of the mitochondrial outer membrane and activation OMA1.


Subject(s)
Apoptosis , Cytochromes c/metabolism , Metalloendopeptidases/metabolism , Mitochondria/enzymology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Line, Tumor , Enzyme Activation , GTP Phosphohydrolases/metabolism , Gene Knockdown Techniques , Humans , Protein Stability
12.
J Med Genet ; 52(8): 523-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986071

ABSTRACT

BACKGROUND: Auditory neuropathy spectrum disorder (ANSD) is a form of hearing loss in which auditory signal transmission from the inner ear to the auditory nerve and brain stem is distorted, giving rise to speech perception difficulties beyond that expected for the observed degree of hearing loss. For many cases of ANSD, the underlying molecular pathology and the site of lesion remain unclear. The X-linked form of the condition, AUNX1, has been mapped to Xq23-q27.3, although the causative gene has yet to be identified. METHODS: We performed whole-exome sequencing on DNA samples from the AUNX1 family and another small phenotypically similar but unrelated ANSD family. RESULTS: We identified two missense mutations in AIFM1 in these families: c.1352G>A (p.R451Q) in the AUNX1 family and c.1030C>T (p.L344F) in the second ANSD family. Mutation screening in a large cohort of 3 additional unrelated families and 93 sporadic cases with ANSD identified 9 more missense mutations in AIFM1. Bioinformatics analysis and expression studies support this gene as being causative of ANSD. CONCLUSIONS: Variants in AIFM1 gene are a common cause of familial and sporadic ANSD and provide insight into the expanded spectrum of AIFM1-associated diseases. The finding of cochlear nerve hypoplasia in some patients was AIFM1-related ANSD implies that MRI may be of value in localising the site of lesion and suggests that cochlea implantation in these patients may have limited success.


Subject(s)
Hearing Loss, Central/genetics , Animals , Apoptosis Inducing Factor/chemistry , Apoptosis Inducing Factor/genetics , Chromosome Mapping , Cohort Studies , DNA Mutational Analysis , Exome/genetics , Female , Genes, X-Linked , Hearing Loss, Central/pathology , Humans , Male , Mice , Mutation, Missense , Pedigree , Protein Structure, Tertiary
13.
Structure ; 32(5): 550-561.e5, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38460520

ABSTRACT

TIGIT is mainly expressed on T cells and is an inhibitory checkpoint receptor that binds to its ligand PVR in the tumor microenvironment. Anti-TIGIT monoclonal antibodies (mAbs) such as Ociperlimab and Tiragolumab block the TIGIT-PVR interaction and are in clinical development. However, the molecular blockade mechanism of these mAbs remains elusive. Here, we report the crystal structures of TIGIT in complex with Ociperlimab_Fab and Tiragolumab_Fab revealing that both mAbs bind TIGIT with a large steric clash with PVR. Furthermore, several critical epitopic residues are identified. Interestingly, the binding affinity of Ociperlimab toward TIGIT increases approximately 17-fold when lowering the pH from 7.4 to 6.0. Our structure shows a strong electrostatic interaction between ASP103HCDR3 and HIS76TIGIT explaining the pH-responsive mechanism of Ociperlimab. In contrast, Tiragolumab does not show an acidic pH-dependent binding enhancement. Our results provide valuable information that could help to improve the efficacy of therapeutic antibodies for cancer treatment.


Subject(s)
Models, Molecular , Protein Binding , Receptors, Immunologic , Hydrogen-Ion Concentration , Humans , Receptors, Immunologic/metabolism , Receptors, Immunologic/chemistry , Crystallography, X-Ray , Antibodies, Monoclonal/chemistry , Binding Sites , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/immunology
14.
Leuk Lymphoma ; 65(8): 1079-1089, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38775302

ABSTRACT

To evaluate the effects of gene mutations on Bruton tyrosine kinase inhibitor, zanubrutinib's effectiveness in patients with diffuse large B-cell lymphoma (DLBCL), we examined pooled data from four single-arm studies (BGB-3111-AU-003 [NCT02343120], BGB-3111-207 [NCT03145064], BGB-3111_GA101_Study_001 [NCT02569476], BGB-3111-213 [NCT03520920]; n = 121). Objective response rate (ORR) was higher, though not statistically significant, in patients with activated B-cell-like (ABC)- and unclassified DLBCL (42.9% [21/49]) versus those with germinal-center B-cell-like DLBCL (14.3% [1/7]; p = 0.15). Patients with CD79B mutations had better ORR (60%) versus patients with wild-type alleles (25.9%, p < 0.01). Higher TCL1A expression correlated with better zanubrutinib response (p = 0.03), longer progression-free survival (p = 0.01), and longer overall survival (p = 0.12). TCL1A expression was higher in ABC-DLBCL (p < 0.001) and MYD88/CD79B-mutated subtypes (p < 0.0001). Eighteen patients with high MYC/BCL-2 expression responded better to zanubrutinib (ORR = 61 vs. 29%, p = 0.02). Our results support assessing CD79B mutations, co-expressor DLBCL, and TCL1A expression status to identify patients with DLBCL who will benefit from zanubrutinib.


Subject(s)
CD79 Antigens , Lymphoma, Large B-Cell, Diffuse , Mutation , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins c-myc , Proto-Oncogene Proteins , Pyrimidines , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Middle Aged , Female , Male , Aged , Pyrimidines/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , CD79 Antigens/genetics , Proto-Oncogene Proteins/genetics , Adult , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Aged, 80 and over , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Treatment Outcome , Germinal Center/pathology , Germinal Center/metabolism , Germinal Center/drug effects
15.
Blood Adv ; 8(7): 1639-1650, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38315878

ABSTRACT

ABSTRACT: The phase 3 ASPEN trial (NCT03053440) compared Bruton tyrosine kinase inhibitors (BTKis), zanubrutinib and ibrutinib, in patients with Waldenström macroglobulinemia (WM). Post-hoc biomarker analysis was performed using next-generation sequencing on pretreatment bone marrow samples from 98 patients treated with zanubrutinib and 92 patients treated with ibrutinib with mutated (MUT) MYD88 and 20 patients with wild-type (WT) MYD88 treated with zanubrutinib. Of 329 mutations in 52 genes, mutations in CXCR4 (25.7%), TP53 (24.8%), ARID1A (15.7%), and TERT (9.0%) were most common. TP53MUT, ARID1AMUT, and TERTMUT were associated with higher rates of CXCR4MUT (P < .05). Patients with CXCR4MUT (frameshift or nonsense [NS] mutations) had lower very good partial response (VGPR) and complete response rates (CR; 17.0% vs 37.2%, P = .020) and longer time to response (11.1 vs 8.4 months) than patients with CXCR4WT treated with BTKis. CXCR4NS was associated with inferior progression-free survival (PFS; hazard ratio [HR], 3.39; P = .017) in patients treated with ibrutinib but not in those treated with zanubrutinib (HR, 0.67; P = .598), but VGPR + CR rates were similar between treatment groups (14.3% vs 15.4%). Compared with ibrutinib, patients with CXCR4NS treated with zanubrutinib had a favorable major response rate (MRR; 85.7% vs 53.8%; P = .09) and PFS (HR, 0.30; P = .093). In patients with TP53MUT, significantly lower MRRs were observed for patients treated with ibrutinib (63.6% vs 85.7%; P = .04) but not for those treated with zanubrutinib (80.8% vs 81.9%; P = .978). In TP53MUT, compared with ibrutinib, patients treated with zanubrutinib had higher VGPR and CR (34.6% vs 13.6%; P < .05), numerically improved MRR (80.8% vs 63.6%; P = .11), and longer PFS (not reached vs 44.2 months; HR, 0.66; P = .37). Collectively, patients with WM with CXCR4MUT or TP53MUT had worse prognosis compared with patients with WT alleles, and zanubrutinib led to better clinical outcomes.


Subject(s)
Adenine/analogs & derivatives , Piperidines , Pyrazoles , Pyrimidines , Waldenstrom Macroglobulinemia , Humans , Waldenstrom Macroglobulinemia/drug therapy , Waldenstrom Macroglobulinemia/genetics , Myeloid Differentiation Factor 88/genetics , Biomarkers
16.
Biomark Res ; 11(1): 25, 2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36879284

ABSTRACT

BACKGROUND: Activated immune cells (IC) in the tumor microenvironment (TME) are critical for anti-tumor efficacy. Greater understanding of the dynamic diversity and crosstalk between IC is needed to clarify their association with immune checkpoint inhibitor efficacy. METHODS: Patients from three tislelizumab monotherapy trials in solid tumors (NCT02407990, NCT04068519, NCT04004221) were retrospectively divided into subgroups by CD8+ T-cell and macrophage (Mφ) levels, assessed via multiplex immunohistochemistry (mIHC; n = 67) or gene expression profiling (GEP; n = 629). RESULTS: A trend of longer survival was observed in patients with both high CD8+ T-cell and Mφ levels versus other subgroups in the mIHC analysis (P = 0.11), which was confirmed with greater statistical significance in the GEP analysis (P = 0.0001). Co-existence of CD8+ T cells and Mφ was coupled with elevated CD8+ T-cell cytotoxicity, T-cell trafficking, MHC class I antigen presentation signatures/genes, and enrichment of the pro-inflammatory Mφ polarization pathway. Additionally, a high level of pro-inflammatory CD64+ Mφ density was associated with an immune-activated TME and survival benefit with tislelizumab (15.2 vs. 5.9 months for low density; P = 0.042). Spatial proximity analysis revealed that closer proximity between CD8+ T cells and CD64+ Mφ was associated with a survival benefit with tislelizumab (15.2 vs. 5.3 months for low proximity; P = 0.024). CONCLUSIONS: These findings support the potential role of crosstalk between pro-inflammatory Mφ and cytotoxic T cells in the clinical benefit of tislelizumab. TRIAL REGISTRATION: NCT02407990, NCT04068519, NCT04004221.

17.
Cell Rep Med ; 4(12): 101302, 2023 12 19.
Article in English | MEDLINE | ID: mdl-38052215

ABSTRACT

The RATIONALE-307 study (ClinicalTrials.gov: NCT03594747) demonstrates prolonged progression-free survival (PFS) with first-line tislelizumab plus chemotherapy versus chemotherapy in advanced lung squamous cell carcinoma (LUSC; N = 360). Here we describe an immune-related gene expression signature (GES), composed of genes involved in both innate and adaptive immunity, that appears to differentiate tislelizumab plus chemotherapy PFS benefit versus chemotherapy. In contrast, a tislelizumab plus chemotherapy PFS benefit is observed regardless of programmed death ligand 1 (PD-L1) expression or tumor mutational burden (TMB). Genetic analysis reveals that NRF2 pathway activation is enriched in PD-L1positive and TMBhigh patients. NRF2 pathway activation is negatively associated with PFS, which affects efficacy outcomes associated with PD-L1 and TMB status, impairing their predictive potential. Mechanistic studies demonstrate that NRF2 directly mediates PD-L1 constitutive expression independent of adaptive PD-L1 regulation in LUSC. In summary, the GES is an immune signature that might identify LUSC patients likely to benefit from first-line tislelizumab plus chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Humans , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , NF-E2-Related Factor 2/genetics , Programmed Cell Death 1 Receptor , Treatment Outcome , Tumor Microenvironment/genetics
18.
Front Med ; 17(6): 1170-1185, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37747585

ABSTRACT

OX40 is a costimulatory receptor that is expressed primarily on activated CD4+, CD8+, and regulatory T cells. The ligation of OX40 to its sole ligand OX40L potentiates T cell expansion, differentiation, and activation and also promotes dendritic cells to mature to enhance their cytokine production. Therefore, the use of agonistic anti-OX40 antibodies for cancer immunotherapy has gained great interest. However, most of the agonistic anti-OX40 antibodies in the clinic are OX40L-competitive and show limited efficacy. Here, we discovered that BGB-A445, a non-ligand-competitive agonistic anti-OX40 antibody currently under clinical investigation, induced optimal T cell activation without impairing dendritic cell function. In addition, BGB-A445 dose-dependently and significantly depleted regulatory T cells in vitro and in vivo via antibody-dependent cellular cytotoxicity. In the MC38 syngeneic model established in humanized OX40 knock-in mice, BGB-A445 demonstrated robust and dose-dependent antitumor efficacy, whereas the ligand-competitive anti-OX40 antibody showed antitumor efficacy characterized by a hook effect. Furthermore, BGB-A445 demonstrated a strong combination antitumor effect with an anti-PD-1 antibody. Taken together, our findings show that BGB-A445, which does not block OX40-OX40L interaction in contrast to clinical-stage anti-OX40 antibodies, shows superior immune-stimulating effects and antitumor efficacy and thus warrants further clinical investigation.


Subject(s)
Antineoplastic Agents , Receptors, Tumor Necrosis Factor , Mice , Animals , Receptors, Tumor Necrosis Factor/physiology , Receptors, OX40 , Membrane Glycoproteins , Ligands , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology
19.
MAbs ; 15(1): 2292305, 2023.
Article in English | MEDLINE | ID: mdl-38095560

ABSTRACT

Pharmaceutical companies have recently focused on accelerating the timeline for initiating first-in-human (FIH) trials to allow quick assessment of biologic drugs. For example, a stable cell pool can be used to produce materials for the toxicology (Tox) study, reducing time to the clinic by 4-5 months. During the coronavirus disease 2019 (COVID-19) pandemic, the anti-COVID drugs timeline from DNA transfection to the clinical stage was decreased to 6 months using a stable pool to generate a clinical drug substrate (DS) with limited stability, virus clearance, and Tox study package. However, a lean chemistry, manufacturing, and controls (CMC) package raises safety and comparability risks and may leave extra work in the late-stage development and commercialization phase. In addition, whether these accelerated COVID-19 drug development strategies can be applied to non-COVID projects and established as a standard practice in biologics development is uncertain. Here, we present a case study of a novel anti-tumor drug in which application of "fast-to-FIH" approaches in combination with BeiGene's de-risk strategy achieved successful delivery of a complete CMC package within 10 months. A comprehensive comparability study demonstrated that the DS generated from a stable pool and a single-cell-derived master cell bank were highly comparable with regards to process performance, product quality, and potency. This accomplishment can be a blueprint for non-COVID drug programs that approach the pace of drug development during the pandemic, with no adverse impact on the safety, quality, and late-stage development of biologics.


Subject(s)
Antineoplastic Agents , Biological Products , COVID-19 , Humans , Antibodies, Monoclonal , Pharmaceutical Preparations , Antineoplastic Agents/therapeutic use
20.
J Environ Public Health ; 2022: 8506676, 2022.
Article in English | MEDLINE | ID: mdl-35958389

ABSTRACT

EMC (economic management cost) of the government has become a hot topic of concern from all walks of life. Controlling and reducing government EMC is the requirement for building a conservation-oriented society and deepening the reform of public budget. Dynamic cost accounting calculates the cost of cost objects through the confirmation, measurement, and distribution of production costs. In this article, it is innovatively proposed to integrate BSC (balanced score card) into EMC control, and analyze the logical relationship between system objectives and functions by using the mathematical model. Based on GA (genetic algorithm) cost optimization control concept and specific control ideas, individuals who meet the evolutionary characteristics are stored in the crossover database and participate in crossover operation as parents when crossing. In order to improve the local optimization ability of the algorithm, parallel mutation operation mechanism is introduced, which can execute multiple mutation rules at the same time. The results show that the average convergence time of this algorithm is 0.186s and the variance of population fitness is 288.19. The conclusion shows that the algorithm proposed in this article can overcome the problems of slow convergence speed, low accuracy, and local convergence of GA, and effectively improve the overall performance of the algorithm.


Subject(s)
Algorithms , Conservation of Natural Resources , Government , Humans , Models, Theoretical
SELECTION OF CITATIONS
SEARCH DETAIL