Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Res ; 12(5): 575-591, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38588410

ABSTRACT

Poliovirus receptor-related 2 (PVRL2, also known as nectin-2 or CD112) is believed to act as an immune checkpoint protein in cancer; however, most insight into its role is inferred from studies on its known receptor, poliovirus receptor (PVR)-related immunoglobulin domain protein (PVRIG, also known as CD112R). Here, we study PVRL2 itself. PVRL2 levels were found to be high in tumor cells and tumor-derived exosomes. Deletion of PVRL2 in multiple syngeneic mouse models of cancer showed a dramatic reduction in tumor growth that was immune dependent. This effect was even greater than that seen with deletion of PD-L1. PVRL2 was shown to function by suppressing CD8+ T and natural killer cells in the tumor microenvironment. The loss of PVRL2 suppressed tumor growth even in the absence of PVRIG. In contrast, PVRIG loss showed no additive effect in the absence of PVRL2. T-cell immunoreceptor with Ig and ITIM domains (TIGIT) blockade combined with PVRL2 deletion resulted in a near complete block in tumor growth. This effect was not recapitulated by the combined deletion of PVRL2 with its paralog, PVR, which is the ligand for TIGIT. These data uncover PVRL2 as a distinct inhibitor of the antitumor immune response with functions beyond that of its known receptor PVRIG. Moreover, the data provide a strong rationale for combinatorial targeting of PVRL2 and TIGIT for cancer immunotherapy.


Subject(s)
Nectins , Receptors, Cell Surface , Receptors, Immunologic , Tumor Microenvironment , Animals , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Nectins/metabolism , Mice , Humans , Tumor Microenvironment/immunology , Cell Line, Tumor , Signal Transduction , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism
2.
Elife ; 112022 03 08.
Article in English | MEDLINE | ID: mdl-35257663

ABSTRACT

Characterization of cell surface proteome differences between cancer and healthy cells is a valuable approach for the identification of novel diagnostic and therapeutic targets. However, selective sampling of surface proteins for proteomics requires large samples (>10e6 cells) and long labeling times. These limitations preclude analysis of material-limited biological samples or the capture of rapid surface proteomic changes. Here, we present two labeling approaches to tether exogenous peroxidases (APEX2 and HRP) directly to cells, enabling rapid, small-scale cell surface biotinylation without the need to engineer cells. We used a novel lipidated DNA-tethered APEX2 (DNA-APEX2), which upon addition to cells promoted cell agnostic membrane-proximal labeling. Alternatively, we employed horseradish peroxidase (HRP) fused to the glycan-binding domain of wheat germ agglutinin (WGA-HRP). This approach yielded a rapid and commercially inexpensive means to directly label cells containing common N-Acetylglucosamine (GlcNAc) and sialic acid glycans on their surface. The facile WGA-HRP method permitted high surface coverage of cellular samples and enabled the first comparative surface proteome characterization of cells and cell-derived small extracellular vesicles (EVs), leading to the robust quantification of 953 cell and EV surface annotated proteins. We identified a newly recognized subset of EV-enriched markers, as well as proteins that are uniquely upregulated on Myc oncogene-transformed prostate cancer EVs. These two cell-tethered enzyme surface biotinylation approaches are highly advantageous for rapidly and directly labeling surface proteins across a range of material-limited sample types.


Subject(s)
Extracellular Vesicles , Proteomics , Horseradish Peroxidase , Humans , Male , Proteome/analysis , Wheat Germ Agglutinins
3.
J Med Chem ; 62(21): 9471-9487, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31560543

ABSTRACT

Inducing protein degradation by proteolysis targeting chimeras (PROTACs) has gained tremendous momentum for its promise to discover and develop new therapies. Based upon our previously reported PROTAC MDM2 degraders, we have designed and synthesized additional analogues. Surprisingly, we found that simple structural modifications of MD-222, a bona fide MDM2 PROTAC degrader, converts it into a "molecular glue", as exemplified by MG-277. MG-277 induces only moderate MDM2 degradation and fails to activate wild-type p53 but is highly potent in inhibition of tumor cell growth in a p53-independent manner. Our mechanistic investigation established that MG-277 is not a PROTAC MDM2 degrader but instead works as a molecular glue, inducing degradation of a translation termination factor, GSPT1 to achieve its potent anticancer activity. Our study provides the first example that simple structural modifications can convert a bona fide PROTAC degrader into a molecular glue compound, which has a completely different mechanism of action.


Subject(s)
Drug Design , Proteolysis/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Models, Molecular , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Protein Conformation
4.
Sci Adv ; 5(1): eaat7911, 2019 01.
Article in English | MEDLINE | ID: mdl-30662943

ABSTRACT

A functional HIV cure requires immune reconstitution for lasting viremia control. A major immune dysfunction persisting in HIV infection is the impairment of T helper cell migration and homing to lymphoid tissues such as GALTs (gut-associated lymphoid tissues). ART (antiretroviral therapy) does not fully restore T cell motility for tissue repopulation. The molecular mechanism dictating this persistent T cell dysfunction is not understood. Cofilin is an actin-depolymerizing factor that regulates actin dynamics for T cell migration. Here, we demonstrate that blood CD4 T cells from HIV-infected patients (n = 193), with or without ART, exhibit significantly lower levels of cofilin phosphorylation (hyperactivation) than those from healthy controls (n = 100; ratio, 1.1:2.3; P < 0.001); cofilin hyperactivation is also associated with poor CD4 T cell recovery following ART. These results suggest an HIV-mediated systemic dysregulation of T cell motility that cannot be repaired solely by ART. We further demonstrate that stimulating blood CD4 T cells with an anti-human α4ß7 integrin antibody can trigger signal transduction and modulate the cofilin pathway, partially restoring T cell motility in vitro. However, we also observed that severe T cell motility defect caused by high degrees of cofilin hyperactivation was not repairable by the anti-integrin antibody, demonstrating a mechanistic hindrance to restore immune functions in vivo. Our study suggests that cofilin is a key molecule that may need to be therapeutically targeted early for T cell tissue repopulation, immune reconstitution, and immune control of viremia.


Subject(s)
Actin Depolymerizing Factors/metabolism , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/metabolism , HIV-1/metabolism , Integrins/immunology , Antirheumatic Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Cell Movement/drug effects , Cell Polarity/drug effects , Cohort Studies , HEK293 Cells , HIV Infections/drug therapy , Humans , Lim Kinases/metabolism , Okadaic Acid/pharmacology , Okadaic Acid/toxicity , Phosphorylation/drug effects , Receptors, CCR5/metabolism , Signal Transduction/drug effects , Transfection
5.
J Med Chem ; 62(2): 448-466, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30525597

ABSTRACT

Human murine double minute 2 (MDM2) protein is a primary endogenous cellular inhibitor of the tumor suppressor p53 and has been pursued as an attractive cancer therapeutic target. Several potent, nonpeptide, small-molecule inhibitors of MDM2 are currently in clinical development. In this paper, we report our design, synthesis, and evaluation of small-molecule MDM2 degraders based on the proteolysis targeting chimera (PROTAC) concept. The most promising compound (MD-224) effectively induces rapid degradation of MDM2 at concentrations <1 nM in human leukemia cells. It achieves an IC50 value of 1.5 nM in inhibition of growth of RS4;11 cells and also low nanomolar IC50 values in a panel of leukemia cell lines. MD-224 achieves complete and durable tumor regression in vivo in the RS4;11 xenograft tumor model in mice at well-tolerated dose schedules. MD-224 is thus a highly potent and efficacious MDM2 degrader and warrants extensive evaluations as a new class of anticancer agent.


Subject(s)
Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Humans , Indoles/pharmacology , Indoles/therapeutic use , Leukemia/drug therapy , Leukemia/pathology , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
6.
J Med Chem ; 61(15): 6685-6704, 2018 Aug 09.
Article in English | MEDLINE | ID: mdl-30019901

ABSTRACT

Proteins of the bromodomain and extra-terminal (BET) family are epigenetics "readers" and promising therapeutic targets for cancer and other human diseases. We describe herein a structure-guided design of [1,4]oxazepines as a new class of BET inhibitors and our subsequent design, synthesis, and evaluation of proteolysis-targeting chimeric (PROTAC) small-molecule BET degraders. Our efforts have led to the discovery of extremely potent BET degraders, exemplified by QCA570, which effectively induces degradation of BET proteins and inhibits cell growth in human acute leukemia cell lines even at low picomolar concentrations. QCA570 achieves complete and durable tumor regression in leukemia xenograft models in mice at well-tolerated dose-schedules. QCA570 is the most potent and efficacious BET degrader reported to date.


Subject(s)
Drug Design , Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Mice , Models, Molecular , Protein Conformation , Proteins/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics
7.
J Med Chem ; 60(12): 4818-4839, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28603984

ABSTRACT

We report herein the design, synthesis, and evaluation of macrocyclic peptidomimetics that bind to WD repeat domain 5 (WDR5) and block the WDR5-mixed lineage leukemia (MLL) protein-protein interaction. Compound 18 (MM-589) binds to WDR5 with an IC50 value of 0.90 nM (Ki value <1 nM) and inhibits the MLL H3K4 methyltransferase (HMT) activity with an IC50 value of 12.7 nM. Compound 18 potently and selectively inhibits cell growth in human leukemia cell lines harboring MLL translocations and is >40 times better than the previously reported compound MM-401. Cocrystal structures of 16 and 18 complexed with WDR5 provide structural basis for their high affinity binding to WDR5. Additionally, we have developed and optimized a new AlphaLISA-based MLL HMT functional assay to facilitate the functional evaluation of these designed compounds. Compound 18 represents the most potent inhibitor of the WDR5-MLL interaction reported to date, and further optimization of 18 may yield a new therapy for acute leukemia.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Peptides, Cyclic/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Animals , Binding, Competitive , Cell Line, Tumor , Cell Survival/drug effects , Chemistry Techniques, Synthetic , Drug Discovery , Drug Stability , High-Throughput Screening Assays/methods , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Intracellular Signaling Peptides and Proteins , Leukemia/drug therapy , Leukemia/pathology , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Magnetic Resonance Spectroscopy , Mice , Microsomes/drug effects , Molecular Docking Simulation , Myeloid-Lymphoid Leukemia Protein/genetics , Peptides, Cyclic/chemistry , Peptidomimetics/metabolism , Protein Interaction Domains and Motifs , Rats
8.
PLoS One ; 7(9): e45290, 2012.
Article in English | MEDLINE | ID: mdl-22984631

ABSTRACT

BACKGROUND: Lysosomal protein transmembrane 4 beta (LAPTM4B) is a novel cancer-related gene which has two alleles designated LAPTM4B*1 and LAPTM4B*2. In this study we investigated the correlation of LAPTM4B genotype with prognosis and clinicopathologic features in patients who had undergone curative resection for gallbladder carcinoma (GBC). METHODOLOGY/PRINCIPAL FINDINGS: PCR assay was performed to determine the LAPTM4B genotype in 85 patients. The correlation of LAPTM4B genotype with clinicopathologic parameters was assessed with the Chi-squared test. Differences in patient survival were determined by the Kaplan-Meier method. Multivariate analysis of prognostic factors was carried out with Cox regression analysis. Patients with LAPTM4B *2 had both significantly shorter overall survival (OS) and shorter disease-free survival (DFS) (both P<0.001). Multivariate analysis showed that LAPTM4B genotype is a prognostic factor for OS and DFS (both P<0.001). CONCLUSIONS/SIGNIFICANCE: LAPTM4B allele *2 is a risk factor associated with poor prognosis in patients with resected GBC, and LAPTM4B status may be therefore be useful preoperatively as an adjunct in evaluation of the operability of GBC.


Subject(s)
Biomarkers, Tumor/genetics , Gallbladder Neoplasms/genetics , Membrane Proteins/genetics , Oncogene Proteins/genetics , Adult , Aged , Alleles , Chi-Square Distribution , Female , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/surgery , Genotype , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Risk Assessment/statistics & numerical data , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...