Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(95): eadi4191, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728412

ABSTRACT

Conventional dendritic cells (DCs) are essential mediators of antitumor immunity. As a result, cancers have developed poorly understood mechanisms to render DCs dysfunctional within the tumor microenvironment (TME). After identification of CD63 as a specific surface marker, we demonstrate that mature regulatory DCs (mregDCs) migrate to tumor-draining lymph node tissues and suppress DC antigen cross-presentation in trans while promoting T helper 2 and regulatory T cell differentiation. Transcriptional and metabolic studies showed that mregDC functionality is dependent on the mevalonate biosynthetic pathway and its master transcription factor, SREBP2. We found that melanoma-derived lactate activates SREBP2 in tumor DCs and drives conventional DC transformation into mregDCs via homeostatic or tolerogenic maturation. DC-specific genetic silencing and pharmacologic inhibition of SREBP2 promoted antitumor CD8+ T cell activation and suppressed melanoma progression. CD63+ mregDCs were found to reside within the lymph nodes of several preclinical tumor models and in the sentinel lymph nodes of patients with melanoma. Collectively, this work suggests that a tumor lactate-stimulated SREBP2-dependent program promotes CD63+ mregDC development and function while serving as a promising therapeutic target for overcoming immune tolerance in the TME.


Subject(s)
Dendritic Cells , Lactic Acid , Mice, Inbred C57BL , Signal Transduction , Sterol Regulatory Element Binding Protein 2 , Dendritic Cells/immunology , Animals , Mice , Humans , Sterol Regulatory Element Binding Protein 2/immunology , Lactic Acid/metabolism , Signal Transduction/immunology , Melanoma/immunology , Melanoma/pathology , Disease Progression , Immune Tolerance/immunology , Female , Cell Line, Tumor , Tumor Microenvironment/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology
2.
bioRxiv ; 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38617347

ABSTRACT

Therapeutic resistance to immune checkpoint blockade has been commonly linked to the process of mesenchymal transformation (MT) and remains a prevalent obstacle across many cancer types. An improved mechanistic understanding for MT-mediated immune evasion promises to lead to more effective combination therapeutic regimens. Herein, we identify the Hedgehog transcription factor, Gli2, as a key node of tumor-mediated immune evasion and immunotherapy resistance during MT. Mechanistic studies reveal that Gli2 generates an immunotolerant tumor microenvironment through the upregulation of Wnt ligand production and increased prostaglandin synthesis. This pathway drives the recruitment, viability, and function of granulocytic myeloid-derived suppressor cells (PMN-MDSCs) while also impairing type I conventional dendritic cell, CD8 + T cell, and NK cell functionality. Pharmacologic EP2/EP4 prostaglandin receptor inhibition and Wnt ligand inhibition each reverses a subset of these effects, while preventing primary and adaptive resistance to anti-PD-1 immunotherapy, respectively. A transcriptional Gli2 signature correlates with resistance to anti-PD-1 immunotherapy in stage IV melanoma patients, providing a translational roadmap to direct combination immunotherapeutics in the clinic. SIGNIFICANCE: Gli2-induced EMT promotes immune evasion and immunotherapeutic resistance via coordinated prostaglandin and Wnt signaling.

3.
bioRxiv ; 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37162965

ABSTRACT

Dendritic cells (cDCs) are essential mediators of anti-tumor immunity. Cancers have developed mechanisms to render DCs dysfunctional within the tumor microenvironment. Utilizing CD63 as a unique surface marker, we demonstrate that mature regulatory DCs (mregDCs) suppress DC antigen cross-presentation while driving T H 2 and regulatory T cell differentiation within tumor-draining lymph node tissues. Transcriptional and metabolic studies show that mregDC functionality is dependent upon the mevalonate biosynthetic pathway and the master transcription factor, SREBP2. Melanoma-derived lactate activates DC SREBP2 in the tumor microenvironment (TME) and drives mregDC development from conventional DCs. DC-specific genetic silencing and pharmacologic inhibition of SREBP2 promotes anti-tumor CD8 + T cell activation and suppresses melanoma progression. CD63 + mregDCs reside within the sentinel lymph nodes of melanoma patients. Collectively, this work describes a tumor-driven SREBP2-dependent program that promotes CD63 + mregDC development and function while serving as a promising therapeutic target for overcoming immune tolerance in the TME. One Sentence Summary: The metabolic transcription factor, SREBF2, regulates the development and tolerogenic function of the mregDC population within the tumor microenvironment.

4.
Sci Transl Med ; 14(672): eabq7019, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36417489

ABSTRACT

The tumor-intrinsic NOD-, LRR- and pyrin domain-containing protein-3 (NLRP3) inflammasome-heat shock protein 70 (HSP70) signaling axis is triggered by CD8+ T cell cytotoxicity and contributes to the development of adaptive resistance to anti-programmed cell death protein 1 (PD-1) immunotherapy by recruiting granulocytic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) into the tumor microenvironment. Here, we demonstrate that the tumor NLRP3-HSP70 axis also drives the accumulation of PMN-MDSCs into distant lung tissues in a manner that depends on lung epithelial cell Toll-like receptor 4 (TLR4) signaling, establishing a premetastatic niche that supports disease hyperprogression in response to anti-PD-1 immunotherapy. Lung epithelial HSP70-TLR4 signaling induces the downstream Wnt5a-dependent release of granulocyte colony-stimulating factor (G-CSF) and C-X-C motif chemokine ligand 5 (CXCL5), thus promoting myeloid granulopoiesis and recruitment of PMN-MDSCs into pulmonary tissues. Treatment with anti-PD-1 immunotherapy enhanced the activation of this pathway through immunologic pressure and drove disease progression in the setting of Nlrp3 amplification. Genetic and pharmacologic inhibition of NLRP3 and HSP70 blocked PMN-MDSC accumulation in the lung in response to anti-PD-1 therapy and suppressed metastatic progression in preclinical models of melanoma and breast cancer. Elevated baseline concentrations of plasma HSP70 and evidence of NLRP3 signaling activity in tumor tissue specimens correlated with the development of disease hyperprogression and inferior survival in patients with stage IV melanoma undergoing anti-PD-1 immunotherapy. Together, this work describes a pathogenic mechanism underlying the phenomenon of disease hyperprogression in melanoma and offers candidate targets and markers capable of improving the management of patients with melanoma.


Subject(s)
Melanoma , Toll-Like Receptor 4 , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , HSP70 Heat-Shock Proteins/metabolism , Immunotherapy , Melanoma/pathology , Disease Progression , Tumor Microenvironment
5.
Cell Rep ; 35(5): 109071, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33951424

ABSTRACT

While immune checkpoint blockade is associated with prolonged responses in multiple cancers, most patients still do not benefit from this therapeutic strategy. The Wnt-ß-catenin pathway is associated with diminished T cell infiltration; however, activating mutations are rare, implicating a role for autocrine/paracrine Wnt ligand-driven signaling in immune evasion. In this study, we show that proximal mediators of the Wnt signaling pathway are associated with anti-PD-1 resistance, and pharmacologic inhibition of Wnt ligand signaling supports anti-PD-1 efficacy by reversing dendritic cell tolerization and the recruitment of granulocytic myeloid-derived suppressor cells in autochthonous tumor models. We further demonstrate that the inhibition of Wnt signaling promotes the development of a tumor microenvironment that is more conducive to favorable responses to checkpoint blockade in cancer patients. These findings support a rationale for Wnt ligand-focused treatment approaches in future immunotherapy clinical trials and suggest a strategy for selecting those tumors more responsive to Wnt inhibition.


Subject(s)
Immunotherapy/methods , Ligands , Wnt1 Protein/metabolism , Animals , Disease Models, Animal , Humans , Mice , Tumor Microenvironment
6.
Int Immunol ; 32(7): 485-491, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32449776

ABSTRACT

The dendritic cell (DC) is recognized as a vital mediator of anti-tumor immunity. More recent studies have also demonstrated the important role of DCs in the generation of effective responses to checkpoint inhibitor immunotherapy. Metabolic programming of DCs dictates their functionality and can determine which DCs become immunostimulatory versus those that develop a tolerized phenotype capable of actively suppressing effector T-cell responses to cancers. As a result, there is great interest in understanding what mechanisms have evolved in cancers to alter these metabolic pathways, thereby allowing for their continued progression and metastasis. The therapeutic strategies developed to reverse these processes of DC tolerization in the tumor microenvironment represent promising candidates for future testing in combination immunotherapy clinical trials.


Subject(s)
Dendritic Cells/metabolism , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Dendritic Cells/immunology , Humans , Immunotherapy , Neoplasms/therapy
7.
J Extracell Vesicles ; 10(2): e12042, 2020 12.
Article in English | MEDLINE | ID: mdl-33408816

ABSTRACT

Primary tumours can establish long-range communication with distant organs to transform them into fertile soil for circulating tumour cells to implant and proliferate, a process called pre-metastatic niche (PMN) formation. Tumour-derived extracellular vesicles (EV) are potent mediators of PMN formation due to their diverse complement of pro-malignant molecular cargo and their propensity to target specific cell types (Costa-Silva et al., 2015; Hoshino et al., 2015; Peinado et al., 2012; Peinado et al., 2017). While significant progress has been made to understand the mechanisms by which pro-metastatic EVs create tumour-favouring microenvironments at pre-metastatic organ sites, comparatively little attention has been paid to the factors intrinsic to recipient cells that may modify the extent to which pro-metastatic EV signalling is received and transduced. Here, we investigated the role of recipient cell cholesterol homeostasis in prostate cancer (PCa) EV-mediated signalling and metastasis. Using a bone metastatic model of enzalutamide-resistant PCa, we first characterized an axis of EV-mediated communication between PCa cells and bone marrow that is marked by in vitro and in vivo PCa EV uptake by bone marrow myeloid cells, activation of NF-κB signalling, enhanced osteoclast differentiation, and reduced myeloid thrombospondin-1 expression. We then employed a targeted, biomimetic approach to reduce myeloid cell cholesterol in vitro and in vivo prior to conditioning with PCa EVs. Reducing myeloid cell cholesterol prevented the uptake of PCa EVs by recipient myeloid cells, abolished NF-κB activity and osteoclast differentiation, stabilized thrombospondin-1 expression, and reduced metastatic burden by 77%. These results demonstrate that cholesterol homeostasis in bone marrow myeloid cells regulates pro-metastatic EV signalling and metastasis by acting as a gatekeeper for EV signal transduction.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Marrow Cells/pathology , Bone Neoplasms/secondary , Cell Communication , Cholesterol/metabolism , Extracellular Vesicles/pathology , Prostatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Marrow Cells/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Proliferation , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
8.
Front Immunol ; 10: 2876, 2019.
Article in English | MEDLINE | ID: mdl-31921140

ABSTRACT

The vast majority of cancer-related deaths are due to metastasis, a process that requires evasion of the host immune system. In addition, a significant percentage of cancer patients do not benefit from our current immunotherapy arsenal due to either primary or secondary immunotherapy resistance. Importantly, select subsets of dendritic cells (DCs) have been shown to be indispensable for generating responses to checkpoint inhibitor immunotherapy. These observations are consistent with the critical role of DCs in antigen cross-presentation and the generation of effective anti-tumor immunity. Therefore, the evolution of efficient tumor-extrinsic mechanisms to modulate DCs is expected to be a potent strategy to escape immunosurveillance and various immunotherapy strategies. Despite this critical role, little is known regarding the methods by which cancers subvert DC function. Herein, we focus on those select mechanisms utilized by developing cancers to co-opt and tolerize local DC populations. We discuss the reported mechanisms utilized by cancers to induce DC tolerization in the tumor microenvironment, describing various parallels between the evolution of these mechanisms and the process of mesenchymal transformation involved in tumorigenesis and metastasis, and we highlight strategies to reverse these mechanisms in order to enhance the efficacy of the currently available checkpoint inhibitor immunotherapies.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Neoplasms/immunology , Tumor Escape , Tumor Microenvironment/immunology , Humans , Immunotherapy , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy
9.
Neoplasia ; 20(9): 930-942, 2018 09.
Article in English | MEDLINE | ID: mdl-30118999

ABSTRACT

We have previously demonstrated that apigenin promotes the expression of antiangiogenic protein thrombospondin-1 (TSP1) via a mechanism driven by mRNA-binding protein HuR. Here, we generated a novel mouse model with whole-body THBS-1 gene knockout on SKH-1 genetic background, which allows studies of UVB-induced acute skin damage and carcinogenesis and tests TSP1 involvement in apigenin's anticancer effects. Apigenin significantly inhibited UVB-induced carcinogenesis in the wild-type (WT) animals but not in TSP1 KO (TKO) mice, suggesting that TSP1 is a critical component of apigenin's chemopreventive function in UVB-induced skin cancer. Importantly, TKO mice presented with the elevated cutaneous inflammation at baseline, which was manifested by increased inflammatory infiltrates (neutrophils and macrophages) and elevated levels of the two key inflammatory cytokines, IL-6 and IL-12. In agreement, maintaining normal TSP1 expression in the UVB-irradiated skin of WT mice using topical apigenin application caused a marked decrease of circulating inflammatory cytokines. Finally, TKO mice showed an altered population dynamics of the bone marrow myeloid progenitor cells (CD11b+), with dramatic expansion of the population of neutrophil progenitors (Ly6ClowLy6Ghigh) compared to the WT control. Our results indicate that the cutaneous tumor suppressor TSP1 is a critical mediator of the in vivo anticancer effect of apigenin in skin, specifically of its anti-inflammatory action.


Subject(s)
Apigenin/pharmacology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Sunscreening Agents/pharmacology , Ultraviolet Rays/adverse effects , Animals , Anti-Inflammatory Agents , Cell Line, Tumor , Chemoprevention , Disease Models, Animal , Genotype , Humans , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Peroxidase/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/prevention & control , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Xenograft Model Antitumor Assays
10.
Oncoscience ; 5(5-6): 164-166, 2018 May.
Article in English | MEDLINE | ID: mdl-30035178
11.
Mol Cell Endocrinol ; 468: 19-30, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29410272

ABSTRACT

A fundamental goal in biology is to understand how distinct cell types containing the same genetic information arise from a single stem cell throughout development. Sex determination is a key developmental process that requires a unidirectional commitment of an initially bipotential gonad towards either the male or female fate. This makes sex determination a unique model to study cell fate commitment and differentiation in vivo. We have focused this review on the accumulating evidence that epigenetic mechanisms contribute to the bipotential state of the fetal gonad and to the regulation of chromatin accessibility during and immediately downstream of the primary sex-determining switch that establishes the male fate.


Subject(s)
Epigenesis, Genetic , Animals , Chromatin/metabolism , DNA Methylation/genetics , Genes, sry , Histones/metabolism , Male , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism
12.
Mol Cancer Ther ; 17(3): 686-697, 2018 03.
Article in English | MEDLINE | ID: mdl-29282300

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are innate immune cells that potently inhibit T cells. In cancer, novel therapies aimed to activate T cells can be rendered ineffective due to the activity of MDSCs. Thus, targeted inhibition of MDSCs may greatly enhance T-cell-mediated antitumor immunity, but mechanisms remain obscure. Here we show, for the first time, that scavenger receptor type B-1 (SCARB1), a high-affinity receptor for spherical high-density lipoprotein (HDL), is expressed by MDSCs. Furthermore, we demonstrate that SCARB1 is specifically targeted by synthetic high-density lipoprotein-like nanoparticles (HDL NP), which reduce MDSC activity. Using in vitro T-cell proliferation assays, data show that HDL NPs specifically bind SCARB1 to inhibit MDSC activity. In murine cancer models, HDL NP treatment significantly reduces tumor growth, metastatic tumor burden, and increases survival due to enhanced adaptive immunity. Flow cytometry and IHC demonstrate that HDL NP-mediated suppression of MDSCs increased CD8+ T cells and reduced Treg cells in the metastatic tumor microenvironment. Using transgenic mice lacking SCARB1, in vivo data clearly show that the HDL NPs specifically target this receptor for suppressing MDSCs. Ultimately, our data provide a new mechanism and targeted therapy, HDL NPs, to modulate a critical innate immune cell checkpoint to enhance the immune response to cancer. Mol Cancer Ther; 17(3); 686-97. ©2017 AACR.


Subject(s)
Lipoproteins, HDL/toxicity , Myeloid-Derived Suppressor Cells/drug effects , Nanoparticles/toxicity , Neoplasms, Experimental/prevention & control , Scavenger Receptors, Class B/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Scavenger Receptors, Class B/genetics , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
13.
Nat Commun ; 8(1): 1319, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29105655

ABSTRACT

Metastatic cancers produce exosomes that condition pre-metastatic niches in remote microenvironments to favor metastasis. In contrast, here we show that exosomes from poorly metastatic melanoma cells can potently inhibit metastasis to the lung. These "non-metastatic" exosomes stimulate an innate immune response through the expansion of Ly6Clow patrolling monocytes (PMo) in the bone marrow, which then cause cancer cell clearance at the pre-metastatic niche, via the recruitment of NK cells and TRAIL-dependent killing of melanoma cells by macrophages. These events require the induction of the Nr4a1 transcription factor and are dependent on pigment epithelium-derived factor (PEDF) on the outer surface of exosomes. Importantly, exosomes isolated from patients with non-metastatic primary melanomas have a similar ability to suppress lung metastasis. This study thus demonstrates that pre-metastatic tumors produce exosomes, which elicit a broad range of PMo-reliant innate immune responses via trigger(s) of immune surveillance, causing cancer cell clearance at the pre-metastatic niche.


Subject(s)
Exosomes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Monocytes/immunology , Animals , Cell Differentiation/immunology , Eye Proteins/immunology , Female , Humans , Immunity, Innate , Immunologic Surveillance , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Macrophages/immunology , Macrophages/pathology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Monocytes/pathology , Nerve Growth Factors/immunology , Phagocytosis/immunology , Serpins/immunology , Tumor Microenvironment/immunology
14.
Sci Rep ; 6: 22915, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26964503

ABSTRACT

Exosomes are produced by cells to mediate intercellular communication, and have been shown to perpetuate diseases, including cancer. New tools are needed to understand exosome biology, detect exosomes from specific cell types in complex biological media, and to modify exosomes. Our data demonstrate a cellular pathway whereby membrane-bound scavenger receptor type B-1 (SR-B1) in parent cells becomes incorporated into exosomes. We tailored synthetic HDL-like nanoparticles (HDL NP), high-affinity ligands for SR-B1, to carry a fluorescently labeled phospholipid. Data show SR-B1-dependent transfer of the fluorescent phospholipid from HDL NPs to exosomes. Modified exosomes are stable in serum and can be directly detected using flow cytometry. As proof-of-concept, human serum exosomes were found to express SR-B1, and HDL NPs can be used to label and isolate them. Ultimately, we discovered a natural cellular pathway and nanoparticle-receptor pair that enables exosome modulation, detection, and isolation.


Subject(s)
Biosensing Techniques , Cell Communication/genetics , Exosomes/metabolism , Scavenger Receptors, Class B/isolation & purification , Exosomes/chemistry , Humans , Ligands , Lipid Metabolism/genetics , Lipoproteins, HDL/chemistry , Nanoparticles/chemistry , Phospholipids/chemistry , Phospholipids/metabolism , Protein Binding , Scavenger Receptors, Class B/blood , Scavenger Receptors, Class B/chemistry , Scavenger Receptors, Class B/genetics
15.
Adv Funct Mater ; 26(43): 7824-7835, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-28717350

ABSTRACT

Efficient systemic administration of therapeutic short interfering RNA (siRNA) is challenging. High-density lipoproteins (HDL) are natural in vivo RNA delivery vehicles. Specifically, native HDLs: 1) Load single-stranded RNA; 2) Are anionic, which requires charge reconciliation between the RNA and HDL, and 3) Actively target scavenger receptor type B-1 (SR-B1) to deliver RNA. Emphasizing these particular parameters, we employed templated lipoprotein particles (TLP), mimics of spherical HDLs, and self-assembled them with single-stranded complements of, presumably, any highly unmodified siRNA duplex pair after formulation with a cationic lipid. Resulting siRNA templated lipoprotein particles (siRNA-TLP) are anionic and tunable with regard to RNA assembly and function. Data demonstrate that the siRNA-TLPs actively target SR-B1 to potently reduce androgen receptor (AR) and enhancer of zeste homolog 2 (EZH2) proteins in multiple cancer cell lines. Systemic administration of siRNA-TLPs demonstrated no off-target toxicity and significantly reduced the growth of prostate cancer xenografts. Thus, native HDLs inspired the synthesis of a hybrid siRNA delivery vehicle that can modularly load single-stranded RNA complements after charge reconciliation with a cationic lipid, and that function due to active targeting of SR-B1.

16.
Sci Rep ; 5: 15724, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26511855

ABSTRACT

Exosomes are nanoscale vesicles that mediate intercellular communication. Cellular exosome uptake mechanisms are not well defined partly due to the lack of specific inhibitors of this complex cellular process. Exosome uptake depends on cholesterol-rich membrane microdomains called lipid rafts, and can be blocked by non-specific depletion of plasma membrane cholesterol. Scavenger receptor type B-1 (SR-B1), found in lipid rafts, is a receptor for cholesterol-rich high-density lipoproteins (HDL). We hypothesized that a synthetic nanoparticle mimic of HDL (HDL NP) that binds SR-B1 and removes cholesterol through this receptor would inhibit cellular exosome uptake. In cell models, our data show that HDL NPs bind SR-B1, activate cholesterol efflux, and attenuate the influx of esterified cholesterol. As a result, HDL NP treatment results in decreased dynamics and clustering of SR-B1 contained in lipid rafts and potently inhibits cellular exosome uptake. Thus, SR-B1 and targeted HDL NPs provide a fundamental advance in studying cholesterol-dependent cellular uptake mechanisms.


Subject(s)
Biomimetic Materials , Cholesterol/metabolism , Exosomes/metabolism , Lipoproteins, HDL , Nanoparticles/chemistry , Scavenger Receptors, Class B/metabolism , Animals , Biological Transport, Active , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line, Tumor , Humans , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/pharmacology , Mice
17.
Curr Opin Oncol ; 25(6): 646-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24097107

ABSTRACT

PURPOSE OF REVIEW: To summarize the most recent preclinical and clinical advancements in therapeutic nano-oncology. RECENT FINDINGS: First-generation nanotherapies are well tolerated in humans and evidence shows that they are efficacious, while at the same time reducing the burden of side-effects. Most of these therapies are not specifically targeted, but take advantage of enhanced passive accumulation within tumors to preferentially deliver chemotherapies that demonstrate off-target toxicities when administered as free drugs. Also, actively targeted nanotherapies are entering the clinical arena and preliminary data are encouraging. Finally, a number of exciting preclinical developments in nanotechnology provide clear evidence that nanotherapies will continue to enter the clinic and will have a significant impact in oncology. SUMMARY: A number of intriguing nanoparticle therapies are being tested in preclinical and clinical trials. Nanoparticles with increasing molecular sophistication, specific targeting properties, and unique mechanisms of action will find their way to the clinic. Certainly, nanoparticle-based therapies will be increasingly represented in drug development pipelines, and will continue to provide efficacious and well tolerated drug options for patients with cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems/trends , Molecular Targeted Therapy , Nanomedicine/trends , Nanoparticles/therapeutic use , Neoplasms/drug therapy , RNA, Small Interfering/therapeutic use , Animals , Clinical Trials as Topic , Drug Delivery Systems/methods , Drug Design , Humans , Male , Mice , Molecular Targeted Therapy/trends , Nanomedicine/methods
18.
Anal Biochem ; 397(1): 73-8, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19818327

ABSTRACT

Many cancers are characterized by changes in protein phosphorylation as a result of kinase dysregulation. Disruption of Abl kinase signaling through the Philadelphia chromosome (causing the Bcr-Abl mutation) in chronic myeloid leukemia (CML) has provided a paradigm for development of kinase inhibitor drugs such as the specific inhibitor imatinib (also known as STI571 or Gleevec). However, because patients are treated indefinitely with this drug to maintain remission, resistance is increasingly becoming an issue. Although there are many ways to detect kinase activity, most lack the ability to "multiplex" the analysis (i.e., to detect more than one substrate simultaneously). Here we report a novel biosensor for detecting Abl kinase activity and sensitivity to inhibitor in live intact cells overexpressing a CML model Abl kinase construct. This straightforward methodology could eventually provide a new tool for detecting kinase activity and inhibitor drug response in cancer cells that overexpress oncogenic kinases.


Subject(s)
Biosensing Techniques/methods , Peptides/analysis , Proto-Oncogene Proteins c-abl/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Cell Line , Fluorescent Dyes/chemistry , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Molecular Sequence Data , Peptides/metabolism , Phosphorylation , Proto-Oncogene Proteins c-abl/genetics , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...