Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 16(3)2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36986488

ABSTRACT

Cancer-associated fibroblasts (CAFs) execute diverse and complex functions in cancer progression. While reprogramming the crosstalk between CAFs and cancer epithelial cells is a promising avenue to evade the adverse effects of stromal depletion, drugs are limited by their suboptimal pharmacokinetics and off-target effects. Thus, there is a need to elucidate CAF-selective cell surface markers that can improve drug delivery and efficacy. Here, functional proteomic pulldown with mass spectrometry was used to identify taste receptor type 2 member 9 (TAS2R9) as a CAF target. TAS2R9 target characterization included binding assays, immunofluorescence, flow cytometry, and database mining. Liposomes conjugated to a TAS2R9-specific peptide were generated, characterized, and compared to naked liposomes in a murine pancreatic xenograft model. Proof-of-concept drug delivery experiments demonstrate that TAS2R9-targeted liposomes bind with high specificity to TAS2R9 recombinant protein and exhibit stromal colocalization in a pancreatic cancer xenograft model. Furthermore, the delivery of a CXCR2 inhibitor by TAS2R9-targeted liposomes significantly reduced cancer cell proliferation and constrained tumor growth through the inhibition of the CXCL-CXCR2 axis. Taken together, TAS2R9 is a novel cell-surface CAF-selective target that can be leveraged to facilitate small-molecule drug delivery to CAFs, paving the way for new stromal therapies.

2.
Nat Commun ; 14(1): 97, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36609611

ABSTRACT

Richter's Transformation (RT) is a poorly understood and fatal progression of chronic lymphocytic leukemia (CLL) manifesting histologically as diffuse large B-cell lymphoma. Protein arginine methyltransferase 5 (PRMT5) is implicated in lymphomagenesis, but its role in CLL or RT progression is unknown. We demonstrate herein that tumors uniformly overexpress PRMT5 in patients with progression to RT. Furthermore, mice with B-specific overexpression of hPRMT5 develop a B-lymphoid expansion with increased risk of death, and Eµ-PRMT5/TCL1 double transgenic mice develop a highly aggressive disease with transformation that histologically resembles RT; where large-scale transcriptional profiling identifies oncogenic pathways mediating PRMT5-driven disease progression. Lastly, we report the development of a SAM-competitive PRMT5 inhibitor, PRT382, with exclusive selectivity and optimal in vitro and in vivo activity compared to available PRMT5 inhibitors. Taken together, the discovery that PRMT5 drives oncogenic pathways promoting RT provides a compelling rationale for clinical investigation of PRMT5 inhibitors such as PRT382 in aggressive CLL/RT cases.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, Large B-Cell, Diffuse , Animals , Mice , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/pathology
3.
Sci Adv ; 8(37): eabp9005, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36112677

ABSTRACT

Using a genome-wide CRISPR screen, we identified CDK9, DHODH, and PRMT5 as synthetic lethal partners with gilteritinib treatment in fms-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD) acute myeloid leukemia (AML) and genetically and pharmacologically validated their roles in gilteritinib sensitivity. The presence of FLT3-ITD is associated with an increase in anaerobic glycolysis, rendering leukemia cells highly sensitive to inhibition of glycolysis. Supportive of this, our data show the enrichment of single guide RNAs targeting 28 glycolysis-related genes upon gilteritinib treatment, suggesting that switching from glycolysis to oxidative phosphorylation (OXPHOS) may represent a metabolic adaption of AML in gilteritinib resistance. CDK9i/FLT3i, DHODHi/FLT3i, and PRMT5i/FLT3i pairs mechanistically converge on OXPHOS and purine biosynthesis blockade, implying that targeting the metabolic functions of these three genes and/or proteins may represent attractive strategies to sensitize AML to gilteritinib treatment. Our findings provide the basis for maximizing therapeutic impact of FLT3-ITD inhibitors and a rationale for a clinical trial of these novel combinations.

4.
Cells ; 10(9)2021 08 30.
Article in English | MEDLINE | ID: mdl-34571895

ABSTRACT

The cytolinker and scaffolding protein, plectin, has emerged as a potent driver of malignant hallmarks in many human cancers due to its involvement in various cellular activities contributing to tumorigenesis, including cancer cell proliferation, adhesion, migration, invasion, and signal transduction. Evidence shows that beyond plectin's diverse protein interactome, its cancer-specific mislocalization to the cell surface enables its function as a potent oncoprotein. As such, therapeutic targeting of plectin, its protein interactors, and, in particular, cancer-specific plectin (CSP) presents an attractive opportunity to impede carcinogenesis directly. Here, we report on plectin's differential gene and protein expression in cancer, explore its mutational profile, and discuss the current understanding of plectin's and CSP's biological function in cancer. Moreover, we review the landscape of plectin as a prognostic marker, diagnostic biomarker, and target for imaging and therapeutic modalities. We highlight how, beyond their respective biological importance, plectin's common overexpression in cancer and CSP's cancer-specific bioavailability underscore their potential as high-value druggable targets. We discuss how recent evidence of the potent anti-cancer effects of CSP therapeutic targeting opens the door for cell-surface mislocalized proteins as novel therapeutic targets.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/drug therapy , Plectin/antagonists & inhibitors , Animals , Humans , Neoplasms/metabolism , Plectin/metabolism
5.
Mol Cancer Ther ; 20(11): 2207-2217, 2021 11.
Article in English | MEDLINE | ID: mdl-34518298

ABSTRACT

Acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITD) mutation is an aggressive hematologic malignancy associated with frequent relapse and poor overall survival. The tyrosine kinase inhibitor gilteritinib is approved for the treatment of relapse/refractory AML with FLT3 mutations, yet its mechanism of action is not completely understood. Here, we sought to identify additional therapeutic targets that can be exploited to enhance gilteritinib's antileukemic effect. Based on unbiased transcriptomic analyses, we identified the glutamine transporter SNAT1 (SLC38A1) as a novel target of gilteritinib that leads to impaired glutamine uptake and utilization within leukemic cells. Using metabolomics and metabolic flux analyses, we found that gilteritinib decreased glutamine metabolism through the TCA cycle and cellular levels of the oncometabolite 2-hydroxyglutarate. In addition, gilteritinib treatment was associated with decreased ATP production and glutathione synthesis and increased reactive oxygen species, resulting in cellular senescence. Finally, we found that the glutaminase inhibitor CB-839 enhanced antileukemic effect of gilteritinib in ex vivo studies using human primary FLT3-ITD-positive AML cells harboring mutations in the enzyme isocitrate dehydrogenase, which catalyzes the oxidative decarboxylation of isocitrate, producing α-ketoglutarate. Collectively, this work has identified a previously unrecognized, gilteritinib-sensitive metabolic pathway downstream of SLC38A1 that causes decreased glutaminolysis and disruption of redox homeostasis. These findings provide a rationale for the development and therapeutic exploration of targeted combinatorial treatment strategies for this subset of relapse/refractory AML.


Subject(s)
Aniline Compounds/therapeutic use , Glutamine/drug effects , Leukemia, Myeloid, Acute/drug therapy , Pyrazines/therapeutic use , fms-Like Tyrosine Kinase 3/metabolism , Aniline Compounds/pharmacology , Animals , Female , Humans , Mice , Pyrazines/pharmacology
6.
Clin Cancer Res ; 27(8): 2352-2366, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33542077

ABSTRACT

PURPOSE: Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors (NAMPTi) are currently in development, but may be limited as single-agent therapy due to compound-specific toxicity and cancer metabolic plasticity allowing resistance development. To potentially lower the doses of NAMPTis required for therapeutic benefit against acute myeloid leukemia (AML), we performed a genome-wide CRISPRi screen to identify rational disease-specific partners for a novel NAMPTi, KPT-9274. EXPERIMENTAL DESIGN: Cell lines and primary cells were analyzed for cell viability, self-renewal, and responses at RNA and protein levels with loss-of-function approaches and pharmacologic treatments. In vivo efficacy of combination therapy was evaluated with a xenograft model. RESULTS: We identified two histone deacetylases (HDAC), HDAC8 and SIRT6, whose knockout conferred synthetic lethality with KPT-9274 in AML. Furthermore, HDAC8-specific inhibitor, PCI-34051, or clinical class I HDAC inhibitor, AR-42, in combination with KPT-9274, synergistically decreased the survival of AML cells in a dose-dependent manner. AR-42/KPT-9274 cotreatment attenuated colony-forming potentials of patient cells while sparing healthy hematopoietic cells. Importantly, combined therapy demonstrated promising in vivo efficacy compared with KPT-9274 or AR-42 monotherapy. Mechanistically, genetic inhibition of SIRT6 potentiated the effect of KPT-9274 on PARP-1 suppression by abolishing mono-ADP ribosylation. AR-42/KPT-9274 cotreatment resulted in synergistic attenuation of homologous recombination and nonhomologous end joining pathways in cell lines and leukemia-initiating cells. CONCLUSIONS: Our findings provide evidence that HDAC8 inhibition- or shSIRT6-induced DNA repair deficiencies are potently synergistic with NAMPT targeting, with minimal toxicity toward normal cells, providing a rationale for a novel-novel combination-based treatment for AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytokines/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Sirtuins/antagonists & inhibitors , Acrylamides/pharmacology , Acrylamides/therapeutic use , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , DNA Damage , DNA End-Joining Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Knockout Techniques , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Leukemia, Myeloid, Acute/pathology , Male , Mice , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Recombinational DNA Repair/drug effects , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Xenograft Model Antitumor Assays
7.
JCI Insight ; 5(23)2020 12 03.
Article in English | MEDLINE | ID: mdl-33268594

ABSTRACT

Effective treatment for AML is challenging due to the presence of clonal heterogeneity and the evolution of polyclonal drug resistance. Here, we report that TP-0903 has potent activity against protein kinases related to STAT, AKT, and ERK signaling, as well as cell cycle regulators in biochemical and cellular assays. In vitro and in vivo, TP-0903 was active in multiple models of drug-resistant FLT3 mutant AML, including those involving the F691L gatekeeper mutation and bone marrow microenvironment-mediated factors. Furthermore, TP-0903 demonstrated preclinical activity in AML models with FLT3-ITD and common co-occurring mutations in IDH2 and NRAS genes. We also showed that TP-0903 had ex vivo activity in primary AML cells with recurrent mutations including MLL-PTD, ASXL1, SRSF2, and WT1, which are associated with poor prognosis or promote clinical resistance to AML-directed therapies. Our preclinical studies demonstrate that TP-0903 is a multikinase inhibitor with potent activity against multiple drug-resistant models of AML that will have an immediate clinical impact in a heterogeneous disease like AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Gene Duplication/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mice , Mice, Nude , Mutation/drug effects , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrimidines/metabolism , Sulfonamides/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
8.
J Hematol Oncol ; 13(1): 139, 2020 10 19.
Article in English | MEDLINE | ID: mdl-33076970

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous and complex disease, and treatments for this disease have not been curative for the majority of patients. In younger patients, internal tandem duplication of FLT3 (FLT3-ITD) is a common mutation for which two inhibitors (midostaurin and gilteritinib) with varied potency and specificity for FLT3 are clinically approved. However, the high rate of relapse or failed initial response of AML patients suggests that the addition of a second targeted therapy may be necessary to improve efficacy. Using an unbiased large-scale CRISPR screen, we genetically identified BCL2 knockout as having synergistic effects with an approved FLT3 inhibitor. Here, we provide supportive studies that validate the therapeutic potential of the combination of FLT3 inhibitors with venetoclax in vitro and in vivo against multiple models of FLT3-ITD-driven AML. Our unbiased approach provides genetic validation for co-targeting FLT3 and BCL2 and repurposes CRISPR screening data, utilizing the genome-wide scope toward mechanistic understanding.


Subject(s)
Leukemia, Myeloid, Acute/therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , CRISPR-Cas Systems , Cell Line, Tumor , Female , Gene Knockout Techniques , Genetic Therapy , Humans , Leukemia, Myeloid, Acute/genetics , Mice, SCID , Pyrazines/therapeutic use , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , Sulfonamides/therapeutic use
9.
J Clin Med ; 9(7)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708954

ABSTRACT

Sickle-cell disease (SCD) is a debilitating hematological disorder with very few approved treatment options. Therapeutic reactivation of fetal hemoglobin (HbF) is one of the most pursued methods for ameliorating the systemic manifestations of SCD. Despite this, very few pharmacological agents have advanced to clinical trials or marketing for use. In this study, we report the development of an HbF in situ intracellular immunoblot assay coupled to a high-throughput drug screen to identify Food and Drug Administration (FDA) approved drugs that can be repurposed clinically for treatment of SCD. Using this assay we evaluated the National Institute of Health (NIH) Clinical Collection (NCC), a publicly available library of 725 small molecules, and found nine candidates that can significantly re-express HbF in erythroid cell lines as well as primary erythroblasts derived from SCD patients. Furthermore, we show the strong effects on HbF expression of these candidates to occur with minimal cytotoxicity in 7 of the 9 drugs. Given these data and their proven history of use for other indications, we hypothesize that several of these candidate drugs warrant further investigation for use in SCD.

10.
Cancers (Basel) ; 12(6)2020 Jun 14.
Article in English | MEDLINE | ID: mdl-32545904

ABSTRACT

Acute myeloid leukemia (AML) is a hematopoietic stem-cell-derived leukemia with often successive derived driver mutations. Late onset acquisition of internal tandem duplication in FLT3 (FLT3-ITD) at a high variant allele frequency often contributes to full transformation to a highly proliferative, rapidly progressive disease with poor outcome. The FLT3-ITD mutation is targetable with approved FLT3 small molecule inhibitors, including midostaurin and gilteritinib. However, outside of patients receiving allogeneic transplant, most patients fail to respond or relapse, suggesting alternative approaches of therapy will be required. We employed genome-wide pooled CRISPR knockout screening as a method for large-scale identification of targets whose knockout produces a phenotypic effect that enhances the antitumor properties of FLT3 inhibitors. Among the candidate targets we identified the effect of XPO1 knockout to be synergistic with midostaurin treatment. Next, we validated the genetic finding with pharmacologic combination of the slowly reversible XPO1 inhibitor selinexor with midostaurin and gilteritinib in FLT3-ITD AML cell lines and primary patient samples. Lastly, we demonstrated improved survival with either combination therapy compared to its monotherapy components in an aggressive AML murine model, supporting further evaluation and rapid clinical translation of this combination strategy.

11.
Cancer Discov ; 8(4): 458-477, 2018 04.
Article in English | MEDLINE | ID: mdl-29386193

ABSTRACT

Bromodomain and extra-terminal (BET) family proteins are key regulators of gene expression in cancer. Herein, we utilize BRD4 profiling to identify critical pathways involved in pathogenesis of chronic lymphocytic leukemia (CLL). BRD4 is overexpressed in CLL and is enriched proximal to genes upregulated or de novo expressed in CLL with known functions in disease pathogenesis and progression. These genes, including key members of the B-cell receptor (BCR) signaling pathway, provide a rationale for this therapeutic approach to identify new targets in alternative types of cancer. Additionally, we describe PLX51107, a structurally distinct BET inhibitor with novel in vitro and in vivo pharmacologic properties that emulates or exceeds the efficacy of BCR signaling agents in preclinical models of CLL. Herein, the discovery of the involvement of BRD4 in the core CLL transcriptional program provides a compelling rationale for clinical investigation of PLX51107 as epigenetic therapy in CLL and application of BRD4 profiling in other cancers.Significance: To date, functional studies of BRD4 in CLL are lacking. Through integrated genomic, functional, and pharmacologic analyses, we uncover the existence of BRD4-regulated core CLL transcriptional programs and present preclinical proof-of-concept studies validating BET inhibition as an epigenetic approach to target BCR signaling in CLL. Cancer Discov; 8(4); 458-77. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 371.


Subject(s)
Gene Expression Regulation, Leukemic , Isoxazoles/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Nuclear Proteins/genetics , Pyridines/therapeutic use , Pyrroles/therapeutic use , Signal Transduction , Transcription Factors/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Humans , Isoxazoles/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology , Mice , Mice, SCID , Nuclear Proteins/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
12.
Blood Adv ; 1(15): 995-1000, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-29296742

ABSTRACT

We identify and characterize novel SF3B1 in-frame deletions in chronic lymphocytic leukemia.These deletions are functionally similar to well-known SF3B1 hotspot mutations and are sensitive to splicing modulation.

13.
PLoS One ; 11(5): e0155244, 2016.
Article in English | MEDLINE | ID: mdl-27186887

ABSTRACT

Next-generation sequencing has enhanced the phage display process, allowing for the quantification of millions of sequences resulting from the biopanning process. In response, many valuable analysis programs focused on specificity and finding targeted motifs or consensus sequences were developed. For targeted drug delivery and molecular imaging, it is also necessary to find peptides that are selective-targeting only the cell type or tissue of interest. We present a new analysis strategy and accompanying software, PHage Analysis for Selective Targeted PEPtides (PHASTpep), which identifies highly specific and selective peptides. Using this process, we discovered and validated, both in vitro and in vivo in mice, two sequences (HTTIPKV and APPIMSV) targeted to pancreatic cancer-associated fibroblasts that escaped identification using previously existing software. Our selectivity analysis makes it possible to discover peptides that target a specific cell type and avoid other cell types, enhancing clinical translatability by circumventing complications with systemic use.


Subject(s)
Cell Surface Display Techniques , Computational Biology/methods , High-Throughput Nucleotide Sequencing , Peptides/chemistry , Peptides/genetics , Software , Amino Acid Sequence , Peptide Library , Reproducibility of Results
14.
Elife ; 42015 Aug 19.
Article in English | MEDLINE | ID: mdl-26287525

ABSTRACT

The Reproducibility Project: Cancer Biology seeks to address growing concerns about reproducibility in scientific research by replicating selected results from a substantial number of high-profile papers in the field of cancer biology. The papers, which were published between 2010 and 2012, were selected on the basis of citations and Altmetric scores (Errington et al., 2014). This Registered report describes the proposed replication plan of key experiments from 'Wnt activity defines colon cancer stem cells and is regulated by the microenvironment' by Vermeulen and colleagues, published in Nature Cell Biology in 2010 (Vermeulen et al., 2010). The key experiments that will be replicated are those reported in Figures 2F, 6D, and 7E. In these experiments, Vermeulen and colleagues utilize a reporter for Wnt activity and show that colon cancer cells with high levels of Wnt activity also express cancer stem cell markers (Figure 2F; Vermeulen et al., 2010). Additionally, treatment either with conditioned medium derived from myofibroblasts or with hepatocyte growth factor restored clonogenic potential in low Wnt activity colon cancer cells in vitro (Figure 6D; Vermeulen et al., 2010) and in vivo (Figure 7E; Vermeulen et al., 2010). The Reproducibility Project: Cancer Biology is a collaboration between the Center for Open Science and Science Exchange and the results of the replications will be published in eLife.


Subject(s)
Neoplastic Stem Cells/physiology , Tumor Microenvironment , Wnt Signaling Pathway , Colonic Neoplasms/pathology , Humans
15.
J Control Release ; 220(Pt A): 556-567, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26122651

ABSTRACT

Although reperfusion is essential in restoring circulation to ischemic myocardium, it also leads to irreversible events including reperfusion injury, decreased cardiac function and ultimately scar formation. Various cell types are involved in the multi-phase repair process including inflammatory cells, vascular cells and cardiac fibroblasts. Therapies targeting these cell types in the infarct border zone can improve cardiac function but are limited by systemic side effects. The aim of this work was to develop liposomes with surface modifications to include peptides with affinity for cell types present in the post-infarct myocardium. To identify peptides specific for the infarct/border zone, we used in vivo phage display methods and an optical imaging approach: fluorescence molecular tomography (FMT). We identified peptides specific for cardiomyocytes, endothelial cells, myofibroblasts, and c-Kit + cells present in the border zone of the remodeling infarct. These peptides were then conjugated to liposomes and in vivo specificity and pharmacokinetics were determined. As a proof of concept, cardiomyocyte specific (I-1) liposomes were used to deliver a PARP-1 (poly [ADP-ribose] polymerase 1) inhibitor: AZ7379. Using a targeted liposomal approach, we were able to increase AZ7379 availability in the infarct/border zone at 24h post-injection as compared with free AZ7379. We observed ~3-fold higher efficiency of PARP-1 inhibition when all cell types were assessed using I-1 liposomes as compared with negative control peptide liposomes (NCP). When analyzed further, I-1 liposomes had 9-fold and 1.5-fold higher efficiencies in cardiomyocytes and macrophages, respectively, as compared with NCP liposomes. In conclusion, we have developed a modular drug delivery system that can be targeted to cell types of therapeutic interest in the infarct border zone.


Subject(s)
Cardiovascular Agents/administration & dosage , Lipids/chemistry , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Myocardium/metabolism , Peptides/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Animals , Biological Availability , Cardiovascular Agents/chemistry , Cardiovascular Agents/metabolism , Cardiovascular Agents/pharmacokinetics , Cell Surface Display Techniques , Disease Models, Animal , Drug Compounding , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Liposomes , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Peptide Library , Peptides/chemistry , Peptides/pharmacokinetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Solubility
16.
Cell Mol Life Sci ; 72(4): 659-71, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25336151

ABSTRACT

Exosomes offer new insight into cancer biology with both diagnostic and therapeutic implications. Because of their cell-to-cell communication, exosomes influence tumor progression, metastasis, and therapeutic efficacy. They can be isolated from blood and other bodily fluids to reveal disease processes occurring within the body, including cancerous growth. In addition to being a reservoir of cancer biomarkers, they can be re-engineered to reinstate tumor immunity. Tumor exosomes interact with various cells of the microenvironment to confer tumor-advantageous changes that are responsible for stromal activation, induction of the angiogenic switch, increased vascular permeability, and immune escape. Exosomes also contribute to metastasis by aiding in the epithelial-to-mesenchymal transition and formation of the pre-metastatic niche. Furthermore, exosomes protect tumor cells from the cytotoxic effects of chemotherapy drugs and transfer chemoresistance properties to nearby cells. Thus, exosomes are essential to many lethal elements of cancer and it is important to understand their biogenesis and role in cancer.


Subject(s)
Exosomes/metabolism , Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Cancer Vaccines/immunology , Endosomal Sorting Complexes Required for Transport/metabolism , Endothelium, Vascular/metabolism , Fibroblasts/metabolism , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Neoplasm Metastasis , Neoplasms/diagnosis , Neoplasms/therapy , Tumor Microenvironment
17.
Cancer Genomics Proteomics ; 9(6): 345-55, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23162074

ABSTRACT

Tumor heterogeneity has been a stumbling block in the development of effective cancer treatments. Personalized medicine has evolved with the theory that matching therapies with the unique misregulated pathways often present in tumors will increase patient prognosis. Of particular interest is prediction or determination of the metastatic potential of a tumor. Thus, biomarkers that can predict metastases represent an enormous advance to our understanding over the clinical treatment of cancer. Considerable effort has been expended to characterize the cancer proteome for early detection, however, fewer efforts have been made to develop biomarkers to distinguish the potential for and the nature of metastasis. In this review, we discuss proteomic technologies as well as existing potential metastatic biomarkers for various cancers. In the conclusion, we discuss forward thinking as to what the field needs to enable translation to the clinic.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasm Metastasis/diagnosis , Neoplasms/diagnosis , Neoplasms/metabolism , Blood Proteins/analysis , Chemokines/analysis , Cytokines/analysis , Humans , Precision Medicine , Prognosis , Proteome
SELECTION OF CITATIONS
SEARCH DETAIL
...