Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Proc Natl Acad Sci U S A ; 120(21): e2209639120, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37186844

ABSTRACT

Renal medullary carcinoma (RMC) is an aggressive kidney cancer that almost exclusively develops in individuals with sickle cell trait (SCT) and is always characterized by loss of the tumor suppressor SMARCB1. Because renal ischemia induced by red blood cell sickling exacerbates chronic renal medullary hypoxia in vivo, we investigated whether the loss of SMARCB1 confers a survival advantage under the setting of SCT. Hypoxic stress, which naturally occurs within the renal medulla, is elevated under the setting of SCT. Our findings showed that hypoxia-induced SMARCB1 degradation protected renal cells from hypoxic stress. SMARCB1 wild-type renal tumors exhibited lower levels of SMARCB1 and more aggressive growth in mice harboring the SCT mutation in human hemoglobin A (HbA) than in control mice harboring wild-type human HbA. Consistent with established clinical observations, SMARCB1-null renal tumors were refractory to hypoxia-inducing therapeutic inhibition of angiogenesis. Further, reconstitution of SMARCB1 restored renal tumor sensitivity to hypoxic stress in vitro and in vivo. Together, our results demonstrate a physiological role for SMARCB1 degradation in response to hypoxic stress, connect the renal medullary hypoxia induced by SCT with an increased risk of SMARCB1-negative RMC, and shed light into the mechanisms mediating the resistance of SMARCB1-null renal tumors against angiogenesis inhibition therapies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Sickle Cell Trait , Animals , Humans , Mice , Carcinoma, Renal Cell/pathology , Hypoxia/genetics , Hypoxia/metabolism , Kidney/metabolism , Kidney Neoplasms/pathology , Sickle Cell Trait/genetics , Sickle Cell Trait/metabolism , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism
2.
Nanomedicine ; 10(6): 1121-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24650884

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal and chemo-refractory cancers, clearly, alternative treatment strategies are needed. We utilized 10nm gold nanoparticles as a scaffold to synthesize nanoconjugates bearing a targeting antibody (cetuximab, C225) and gemcitabine. Loading efficiency of gemcitabine on the gold nanoconjugates was 30%. Targeted gold nanoconjugates in combination with RF were selectively cytotoxic to EGFR expressing Hep3B and SNU449 cells when compared to isotype particles with/without RF (P<0.05). In animal experiments, targeted gold nanoconjugates halted the growth of subcutaneous Hep3B xenografts in combination with RF exposure (P<0.05). These xenografts also demonstrated increased apoptosis, necrosis and decreased proliferation compared to controls. Normal tissues were unharmed. We have demonstrated that non-invasive RF-induced hyperthermia when combined with targeted delivery of gemcitabine is more effective and safe at dosages ~275-fold lower than the current clinically-delivered systemic dose of gemcitabine. FROM THE CLINICAL EDITOR: In a model of hepatocellular carcinoma, the authors demonstrate that non-invasive RF-induced hyperthermia applied with cetuximab targeted delivery of Au NP-gemcitabine conjugate is more effective and safe at dosages ~ 275-fold lower than the current clinically-used systemic dose of gemcitabine.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Hepatocellular/therapy , Deoxycytidine/analogs & derivatives , Gold/therapeutic use , Liver Neoplasms/therapy , Nanoconjugates/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/chemistry , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cetuximab , Deoxycytidine/chemistry , Deoxycytidine/therapeutic use , Drug Delivery Systems , Gold/chemistry , Humans , Hyperthermia, Induced , Liver/drug effects , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice, Inbred BALB C , Nanoconjugates/chemistry , Gemcitabine
3.
bioRxiv ; 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37786705

ABSTRACT

Mesenchymal plasticity has been extensively described in advanced and metastatic epithelial cancers; however, its functional role in malignant progression, metastatic dissemination and therapy response is controversial. More importantly, the role of epithelial mesenchymal transition (EMT) and cell plasticity in tumor heterogeneity, clonal selection and clonal evolution is poorly understood. Functionally, our work clarifies the contribution of EMT to malignant progression and metastasis in pancreatic cancer. We leveraged ad hoc somatic mosaic genome engineering, lineage tracing and ablation technologies and dynamic genetic reporters to trace and ablate tumor-specific lineages along the phenotypic spectrum of epithelial to mesenchymal plasticity. The experimental evidences clarify the essential contribution of mesenchymal lineages to pancreatic cancer evolution and metastatic dissemination. Spatial genomic analysis combined with single cell transcriptomic and epigenomic profiling of epithelial and mesenchymal lineages reveals that EMT promotes with the emergence of chromosomal instability (CIN). Specifically tumor lineages with mesenchymal features display highly conserved patterns of genomic evolution including complex structural genomic rearrangements and chromotriptic events. Genetic ablation of mesenchymal lineages robustly abolished these mutational processes and evolutionary patterns, as confirmed by cross species analysis of pancreatic and other human epithelial cancers. Mechanistically, we discovered that malignant cells with mesenchymal features display increased chromatin accessibility, particularly in the pericentromeric and centromeric regions, which in turn results in delayed mitosis and catastrophic cell division. Therefore, EMT favors the emergence of high-fitness tumor cells, strongly supporting the concept of a cell-state, lineage-restricted patterns of evolution, where cancer cell sub-clonal speciation is propagated to progenies only through restricted functional compartments. Restraining those evolutionary routes through genetic ablation of clones capable of mesenchymal plasticity and extinction of the derived lineages completely abrogates the malignant potential of one of the most aggressive form of human cancer.

4.
Nat Cancer ; 4(7): 984-1000, 2023 07.
Article in English | MEDLINE | ID: mdl-37365326

ABSTRACT

Molecular routes to metastatic dissemination are critical determinants of aggressive cancers. Through in vivo CRISPR-Cas9 genome editing, we generated somatic mosaic genetically engineered models that faithfully recapitulate metastatic renal tumors. Disruption of 9p21 locus is an evolutionary driver to systemic disease through the rapid acquisition of complex karyotypes in cancer cells. Cross-species analysis revealed that recurrent patterns of copy number variations, including 21q loss and dysregulation of the interferon pathway, are major drivers of metastatic potential. In vitro and in vivo genomic engineering, leveraging loss-of-function studies, along with a model of partial trisomy of chromosome 21q, demonstrated a dosage-dependent effect of the interferon receptor genes cluster as an adaptive mechanism to deleterious chromosomal instability in metastatic progression. This work provides critical knowledge on drivers of renal cell carcinoma progression and defines the primary role of interferon signaling in constraining the propagation of aneuploid clones in cancer evolution.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , DNA Copy Number Variations/genetics , Chromosomal Instability/genetics , Aneuploidy , Kidney Neoplasms/genetics
5.
Int J Hyperthermia ; 28(3): 202-9, 2012.
Article in English | MEDLINE | ID: mdl-22515341

ABSTRACT

BACKGROUND: Several studies have reported targeted hyperthermia at the cellular level using remote activation of nanoparticles by radiofrequency waves. To date, methods to quantify intracellular thermal dose have not been reported. In this report we study the relationship between radio wave exposure and luciferase denaturation with and without intracellular nanoparticles. The findings are used to devise a strategy to quantify targeted thermal dose in a primary human liver cancer cell line. METHODS: Water bath or non-invasive external Kanzius RF generator (600 W, 13.56 MHz) was used for hyperthermia exposures. Luciferase activity was measured using a bioluminescence assay and viability was assessed using Annexin V-FITC and propidium iodide staining. Heat shock proteins were analysed using western blot analysis. RESULTS: Duration-dependent luciferase denaturation was observed in SNU449 cells exposed to RF field that preceded measurable loss in viability. Loss of luciferase activity was higher in cetuximab-conjugated gold nanoparticle (C225-AuNP) treated cells. Using a standard curve from water bath experiments, the intracellular thermal dose was calculated. Cells treated with C225-AuNP accumulated 6.07 times higher intracellular thermal dose than the untreated controls over initial 4 min of RF exposure. CONCLUSION: Cancer cells when exposed to an external RF field exhibit dose-dependent protein denaturation. Luciferase denaturation assay can be used to quantify thermal dose delivered after RF exposures to cancer cells with and without nanoparticles.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Hyperthermia, Induced/methods , Luciferases/metabolism , Protein Denaturation , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Gold/therapeutic use , Humans , Luminescent Measurements , Metal Nanoparticles/therapeutic use , Radiofrequency Therapy , Thermometers
6.
Nanomedicine ; 8(7): 1096-105, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22349096

ABSTRACT

The use of noninvasive radiofrequency (RF) electric fields as an energy source for thermal activation of nanoparticles within cancer cells could be a valuable addition to the emerging field of nano-mediated cancer therapies. Based on investigations of cell death through hyperthermia, and offering the ability for total-body penetration by RF fields, this technique is thought to complement and possibly outperform existing nano-heat treatments that utilize alternative heat production via optical or magnetic stimuli. However, it remains a challenge to understand fully the complex RF-nanoparticle-intracellular interactions before full system optimization can be engineered. Herein we have shown that liver cancer cells can selectively internalize antibody-conjugated gold nanoparticles (AuNPs) through receptor-mediated endocytosis, with the nanoparticles predominantly accumulating and aggregating within cytoplasmic endolysosomes. After exposure to an external RF field, nonaggregated AuNPs absorbed and dissipated energy as heat, causing thermal damage to the targeted cancer cells. We also observed that RF absorption and heat dissipation is dependent on solubility of AuNPs in the colloid, which is pH dependent. Furthermore, by modulating endolysosomal pH it is possible to prevent intracellular AuNP aggregation and enhance thermal cytotoxicity in hepatocellular cancer cells. FROM THE CLINICAL EDITOR: Gold nanoparticles absorb energy from RF fields and can exert hyperthermic effects leading to cell death. Combining this known effect with antibody-based targeting of the nanoparticles, selective cancer specific hyperthermia induced cell death therapies can be designed, as demonstrated in this article.


Subject(s)
Gold/therapeutic use , Hyperthermia, Induced/methods , Immunoconjugates/therapeutic use , Liver Neoplasms/therapy , Nanoparticles/therapeutic use , Radiofrequency Therapy , Antibodies/chemistry , Antibodies/therapeutic use , Cell Line, Tumor , Gold/chemistry , Humans , Hydrogen-Ion Concentration , Immunoconjugates/chemistry , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lysosomes/metabolism , Lysosomes/pathology , Nanoparticles/chemistry , Solubility
7.
Cancer Res ; 81(15): 4054-4065, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34117030

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is almost universally lethal. A critical unmet need exists to explore essential susceptibilities in PDAC and to identify druggable targets to improve PDAC treatment. KRAS mutations dominate the genetic landscape of PDAC and lead to activation of multiple downstream pathways and cellular processes. Here, we investigated the requirement of these pathways for tumor maintenance using an inducible KrasG12D -driven PDAC mouse model (iKras model), identifying that RAF-MEK-MAPK signaling is the major effector for oncogenic KRAS-mediated tumor maintenance. However, consistent with previous studies, MEK inhibition had minimal therapeutic effect as a single agent for PDAC in vitro and in vivo. Although MEK inhibition partially downregulated transcription of glycolysis genes, it failed to suppress glycolytic flux in PDAC cells, which is a major metabolic effector of oncogenic KRAS. Accordingly, an in vivo genetic screen identified multiple glycolysis genes as potential targets that may sensitize tumor cells to MEK inhibition. Inhibition of glucose metabolism with low-dose 2-deoxyglucose in combination with a MEK inhibitor induced apoptosis in KrasG12D -driven PDAC cells in vitro. The combination also inhibited xenograft PDAC tumor growth and prolonged overall survival in a genetically engineered PDAC mouse model. Molecular and metabolic analyses indicated that co-targeting glycolysis and MAPK signaling results in apoptosis via induction of lethal endoplasmic reticulum stress. Together, our work suggests that combined inhibition of glycolysis and the MAPK pathway may serve as an effective approach to target KRAS-driven PDAC. SIGNIFICANCE: This study demonstrates the critical role of glucose metabolism in resistance to MAPK inhibition in KRAS-driven pancreatic cancer, uncovering a potential therapeutic approach for treating this aggressive disease.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Glucose/metabolism , Protein Kinase Inhibitors/therapeutic use , Animals , Humans , Mice , Protein Kinase Inhibitors/pharmacology
9.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31557131

ABSTRACT

Transcriptomic profiling classifies pancreatic ductal adenocarcinoma (PDAC) into several molecular subtypes with distinctive histological and clinical characteristics. However, little is known about the molecular mechanisms that define each subtype and their correlation with clinical outcome. Mutant KRAS is the most prominent driver in PDAC, present in over 90% of tumors, but the dependence of tumors on oncogenic KRAS signaling varies between subtypes. In particular, the squamous subtype is relatively independent of oncogenic KRAS signaling and typically displays much more aggressive clinical behavior versus the progenitor subtype. Here, we identified that yes-associated protein 1 (YAP1) activation is enriched in the squamous subtype and associated with poor prognosis. Activation of YAP1 in progenitor subtype cancer cells profoundly enhanced malignant phenotypes and transformed progenitor subtype cells into squamous subtype. Conversely, depletion of YAP1 specifically suppressed tumorigenicity of squamous subtype PDAC cells. Mechanistically, we uncovered a significant positive correlation between WNT5A expression and YAP1 activity in human PDAC and demonstrated that WNT5A overexpression led to YAP1 activation and recapitulated a YAP1-dependent but Kras-independent phenotype of tumor progression and maintenance. Thus, our study identifies YAP1 oncogene as a major driver of squamous subtype PDAC and uncovers the role of WNT5A in driving PDAC malignancy through activation of the YAP pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Pancreatic Ductal/genetics , Oncogenes , Pancreatic Neoplasms/genetics , Transcription Factors/genetics , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Neoplasms/pathology , Wnt-5a Protein/genetics , YAP-Signaling Proteins
10.
Nat Commun ; 10(1): 5114, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704972

ABSTRACT

Triple-negative breast cancer (TNBC), which lacks estrogen receptor α (ERα), progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expression, is closely related to basal-like breast cancer. Previously, we and others report that cyclin E/cyclin-dependent kinase 2 (CDK2) phosphorylates enhancer of zeste homolog 2 (EZH2) at T416 (pT416-EZH2). Here, we show that transgenic expression of phospho-mimicking EZH2 mutant EZH2T416D in mammary glands leads to tumors with TNBC phenotype. Coexpression of EZH2T416D in mammary epithelia of HER2/Neu transgenic mice reprograms HER2-driven luminal tumors into basal-like tumors. Pharmacological inhibition of CDK2 or EZH2 allows re-expression of ERα and converts TNBC to luminal ERα-positive, rendering TNBC cells targetable by tamoxifen. Furthermore, the combination of either CDK2 or EZH2 inhibitor with tamoxifen effectively suppresses tumor growth and markedly improves the survival of the mice bearing TNBC tumors, suggesting that the mechanism-based combination therapy may be an alternative approach to treat TNBC.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Estrogen Receptor alpha/drug effects , Mammary Neoplasms, Experimental/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Benzamides/pharmacology , Biphenyl Compounds , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclic N-Oxides , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Indolizines , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Morpholines , Phosphorylation , Pyridinium Compounds/pharmacology , Pyridones/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Triple Negative Breast Neoplasms/metabolism
11.
Cancer Cell ; 33(4): 752-769.e8, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29606349

ABSTRACT

Pancreatic ribonuclease (RNase) is a secreted enzyme critical for host defense. We discover an intrinsic RNase function, serving as a ligand for epidermal growth factor receptor (EGFR), a member of receptor tyrosine kinase (RTK), in pancreatic ductal adenocarcinoma (PDAC). The closely related bovine RNase A and human RNase 5 (angiogenin [ANG]) can trigger oncogenic transformation independently of their catalytic activities via direct association with EGFR. Notably, high plasma ANG level in PDAC patients is positively associated with response to EGFR inhibitor erlotinib treatment. These results identify a role of ANG as a serum biomarker that may be used to stratify patients for EGFR-targeted therapies, and offer insights into the ligand-receptor relationship between RNase and RTK families.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Erlotinib Hydrochloride/pharmacology , Pancreatic Neoplasms/pathology , Ribonuclease, Pancreatic/blood , Ribonuclease, Pancreatic/metabolism , Animals , Binding Sites , Biomarkers/blood , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/drug therapy , Cattle , Cell Line, Tumor , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Erlotinib Hydrochloride/therapeutic use , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Mice , Neoplasm Transplantation , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/drug therapy , Ribonuclease, Pancreatic/chemistry , Signal Transduction
12.
J Natl Cancer Inst ; 106(8)2014 Aug.
Article in English | MEDLINE | ID: mdl-25128695

ABSTRACT

BACKGROUND: Gemcitabine is a potent nucleoside analogue against solid tumors, but development of drug resistance is a substantial problem. Removal of gemcitabine incorporated into DNA by repair mechanisms may contribute to resistance in chemo-refractory solid tumors. Human hepatocellular carcinoma (HCC) is usually very chemoresistant to gemcitabine. METHODS: We treated HCC in vitro and in vivo (orthotopic murine model with human Hep3B or HepG2 xenografts, 7-10 CB17SCID mice per group) with gemcitabine. The role of homologous recombination repair proteins in repairing stalled replication forks was evaluated with hyperthermia exposure and cell-cycle analysis. The Student t-test was used for two-sample comparisons. Multiple group data were analyzed using one-way analysis of variance. All statistical tests were two-sided. RESULTS: We demonstrated that Mre11-mediated homologous recombination repair of gemcitabine-stalled replication forks is crucial to survival of HCC cells. Furthermore, we demonstrated inhibition of Mre11 by an exonuclease inhibitor or concomitant hyperthermia. In orthotopic murine models of chemoresistant HCC, the Hep3B tumor mass with radiofrequency plus gemcitabine treatment (mean ± SD, 180±91mg) was statistically significantly smaller compared with gemcitabine alone (661±419mg, P = .0063). CONCLUSIONS: This study provides mechanistic understanding of homologous recombination inhibiting-strategies, such as noninvasive radiofrequency field-induced hyperthermia, to overcome resistance to gemcitabine in refractory human solid tumors.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/therapy , DNA Replication/drug effects , DNA, Neoplasm/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Hyperthermia, Induced/methods , Liver Neoplasms/therapy , Radio Waves , Recombinational DNA Repair/drug effects , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival/drug effects , Cell Survival/genetics , DNA Replication/genetics , DNA, Neoplasm/genetics , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm , Exonucleases/antagonists & inhibitors , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , MRE11 Homologue Protein , Mice , Mice, SCID , Neoplasms, Experimental/therapy , Radiofrequency Therapy , Gemcitabine
13.
Hepatol Res ; 42(7): 706-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22414001

ABSTRACT

AIM: Although thioredoxin 1 (TXN) has pleiotropic cellular functions as a redox-sensitive protein, very little is known about its role in tumor survival and growth under hypoxia. MHCC97H hepatocellular carcinoma cells have a high metastatic potential and high thioredoxin expression levels compared with their parent cell line, MHCC97. Thus, we used this cell line to explore the functional connections between TXN and hypoxia. METHODS: MHCC97H cells were cultured under normoxia and hypoxia for specific periods after nucleofection with TXN siRNA or control siRNA. We assessed the ß-phenylethyl isothiocyanate (PEITC) sensitivity of the cells, cell proliferation, cell cycle and senescence, and DNA damage response by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, colony formation assays, flow cytometry, ß-galactosidase staining, western blotting, and immunohistochemistry. RESULTS: ß-phenylethyl isothiocyanate treatment shifted reduced TXN to oxidized TXN in MHCC97H cells. Although silencing of TXN via siRNA had no effect on the PEITC sensitivity of the cells, it suppressed cell proliferation and colony formation under both normoxia and hypoxia. Under hypoxia, silencing TXN did not induce apoptosis but induced DNA damage response and cellular senescence. CONCLUSIONS: High TXN levels in MHCC97H cells protect them from DNA damage and cellular senescence under hypoxia. Targeting TXN might enhance the chemotherapeutic effects of some DNA-damaging agents against hepatocellular carcinoma.

14.
Nanotoxicology ; 5(4): 459-68, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20854190

ABSTRACT

There is a paucity of data regarding the safety of administering solid gold nanoparticles (AuNPs) in large animal tumor models. We assessed the acute toxicity and biodistribution of 5 nm and 25 nm solid AuNPs in New Zealand White rabbits (n = 6 in each) with implanted liver Vx2 tumors 24 h after intravenous injection. Gold concentration was determined by inductively coupled plasma atomic emission spectrometry (ICP) and imaged with transmission electron microscopy (TEM). There was no clinico-pathologic evidence of renal, hepatic, pulmonary, or other organ dysfunction. After 25 nm AuNP administration, the concentration of white blood cells increased after treatment (p = 0.001). Most other blood studies were unchanged. AuNPs were distributed to the spleen, liver, and Vx2 tumors, but not to other tissues. The urinary excretion of AuNPs was bimodal as measured by ICP. 25 nm AuNPs were more evenly distributed throughout tissues and may be better tools for medical therapy.


Subject(s)
Gold/pharmacokinetics , Gold/toxicity , Liver Neoplasms, Experimental/metabolism , Metal Nanoparticles/toxicity , Animals , Blood Cell Count , Gold/administration & dosage , Gold/urine , Histocytochemistry , Injections, Intravenous , Liver Neoplasms, Experimental/chemistry , Liver Neoplasms, Experimental/pathology , Mass Spectrometry , Metal Nanoparticles/administration & dosage , Microscopy, Electron, Transmission , Particle Size , Rabbits , Tissue Distribution
15.
Clin Cancer Res ; 17(17): 5822-32, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21737511

ABSTRACT

PURPOSE: The resistance of tumors to antiangiogenic therapies is becoming increasingly relevant. There are currently no validated predictive biomarkers for selecting which cancer patients will benefit from antiangiogenic therapy. Also lacking are resistance biomarkers that can identify which escape pathways should be targeted after tumors develop resistance to VEGF treatment. Recent studies showed that anti-VEGF treatment can make tumor cells more aggressive and metastatic. However, the mechanisms and mediators of this are unidentified. Therefore, we aimed this study at directly identifying the tumor cell-initiated mechanisms responsible for the resistance of pancreatic cancer to anti-VEGF treatment. EXPERIMENTAL DESIGN: We established and validated two murine models of human pancreatic cancer resistant to the VEGF-specific antibody bevacizumab in vivo. We used a genome-wide analysis to directly identify which tumor-secreted factors were overexpressed by pancreatic cancer cells that were resistant to anti-VEGF treatment. RESULTS: Rather than direct proangiogenic factors, we identified several proinflammatory factors that were expressed at higher levels in cells resistant to anti-VEGF treatment than in treatment-sensitive control cells. These proinflammatory factors acted in a paracrine manner to stimulate the recruitment of CD11b(+) proangiogenic myeloid cells. Also, we found that secreted factors overexpressed by anti-VEGF treatment-resistant pancreatic cancer cells acted in an autocrine manner to induce epithelial-to-mesenchymal transition (EMT) and were thus responsible for increased aggressiveness of bevacizumab-resistant pancreatic tumors. CONCLUSIONS: Our results identified proinflammatory factors and EMT markers as potential biomarkers for selecting patients with pancreatic cancer for antiangiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/physiopathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Bevacizumab , Biomarkers, Tumor/analysis , CD11b Antigen/biosynthesis , Cell Line, Tumor , Chemokines/biosynthesis , Cytokines/biosynthesis , Disease Progression , Drug Resistance, Neoplasm , Female , Genome-Wide Association Study , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transplantation, Heterologous , Tumor Escape/genetics
16.
Transl Oncol ; 4(3): 138-46, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21633669

ABSTRACT

Hepatocellular carcinoma (HCC) and pancreatic carcinoma (PC) cells often have inherent urea cycle defects rendering them auxotrophic for the amino acid l-arginine (l-arg). Most HCC and PC require extracellular sources of l-arg and undergo cell cycle arrest and apoptosis when l-arg is restricted. Systemic, enzyme-mediated depletion of l-arg has been investigated in mouse models and human trials. Non-human enzymes elicit neutralizing antibodies, whereas human arginases display poor pharmacological properties in serum. Co(2+) substitution of the Mn(2+) metal cofactor in human arginase I (Co-hArgI) was shown to confer more than 10-fold higher catalytic activity (k(cat)/K(m)) and 5-fold greater stability. We hypothesized that the Co-hArgI enzyme would decrease tumor burden by systemic elimination of l-arg in a murine model. Co-hArgI was conjugated to 5-kDa PEG (Co-hArgI-PEG) to enhance circulation persistence. It was used as monotherapy for HCC and PC in vitro and in vivo murine xenografts. The mechanism of cell death was also investigated. Weekly treatment of 8 mg/kg Co-hArgI-PEG effectively controlled human HepG2 (HCC) and Panc-1 (PC) tumor xenografts (P = .001 and P = .03, respectively). Both cell lines underwent apoptosis in vitro with significant increased expression of activated caspase-3 (P < .001). Furthermore, there was evidence of autophagy in vitro and in vivo. We have demonstrated that Co-hArgI-PEG is effective at controlling two types of l-arg-dependent carcinomas. Being a nonessential amino acid, arginine deprivation therapy through Co-hArgI-PEG holds promise as a new therapy in the treatment of HCC and PC.

17.
Surgery ; 148(2): 319-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20541785

ABSTRACT

BACKGROUND: Gold and carbon nanoparticles absorb nonionizing radio frequency (RF) energy and release heat. Solid gold nanoparticles are delivered to cancer cells via conjugation with targeting antibodies. Here, 20-nm gold particles were conjugated to cetuximab, which is an epidermal growth factor receptor-1 (EGFR-1) antibody. METHODS: A pancreatic carcinoma cell line that highly expresses EGFR-1, Panc-1, and Cama-1, which is a breast carcinoma cell line that minimally expresses EGFR-1, were treated with 100-nmol/L cetuximab-conjugated gold nanoparticles for 3 h (n = 4). Thirty-six hours later, the dishes were placed in an RF field with a generator power of 200 W for 5 min. After another 36 h, cell injury and death were evaluated with flow cytometry. RESULTS: The targeted cell line Panc-1 had a viability of 46% +/- 12%, whereas the Cama-1 cell had a viability of 92% +/- 2% after RF field exposure (P < .008). Transmission electron microscopy showed gold nanoparticle uptake in Panc-1 cells but negligible uptake by Cama-1 cells. Nontargeted cells do not internalize a sufficient amount of antibody-conjugated gold nanoparticles to induce injury in a noninvasive RF field. CONCLUSION: This technique could be useful in cancer treatment if a cancer-specific antibody is used to localize gold nanoparticles to malignant cells.


Subject(s)
Metal Nanoparticles/therapeutic use , Pancreatic Neoplasms/therapy , Radiofrequency Therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Survival , Cetuximab , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gold/administration & dosage , Gold/therapeutic use , Humans , Hyperthermia, Induced , Metal Nanoparticles/administration & dosage , Microscopy, Electron, Transmission , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
18.
Clin Cancer Res ; 16(23): 5712-21, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21138869

ABSTRACT

PURPOSE: Pancreatic carcinoma is one of the deadliest cancers with few effective treatments. Gold nanoparticles (AuNP) are potentially therapeutic because of the safety demonstrated thus far and their physiochemical characteristics. We used the astounding heating rates of AuNPs in nonionizing radiofrequency (RF) radiation to investigate human pancreatic xenograft destruction in a murine model. EXPERIMENTAL DESIGN: Weekly, Panc-1 and Capan-1 human pancreatic carcinoma xenografts in immunocompromised mice were exposed to an RF field 36 hours after treatment (intraperitoneal) with cetuximab- or PAM4 antibody-conjugated AuNPs, respectively. Tumor sizes were measured weekly, whereas necrosis and cleaved caspase-3 were investigated with hematoxylin-eosin staining and immunofluorescence, respectively. In addition, AuNP internalization and cytotoxicity were investigated in vitro with confocal microscopy and flow cytometry, respectively. RESULTS: Panc-1 cells demonstrated increased apoptosis with decreased viability after treatment with cetuximab-conjugated AuNPs and RF field exposure (P = 0.00005). Differences in xenograft volumes were observed within 2 weeks of initiating therapy. Cetuximab- and PAM4-conjugated AuNPs demonstrated RF field-induced destruction of Panc-1 and Capan-1 pancreatic carcinoma xenografts after 6 weeks of weekly treatment (P = 0.004 and P = 0.035, respectively). There was no evidence of injury to murine organs. Cleaved caspase-3 and necrosis were both increased in treated tumors. CONCLUSIONS: This study demonstrates a potentially novel cancer therapy by noninvasively inducing intracellular hyperthermia with targeted AuNPs in an RF field. While the therapy is dependent on the specificity of the targeting antibody, normal tissues were without toxicity despite systemic therapy and whole-body RF field exposure.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antibodies, Monoclonal/administration & dosage , Gold/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Radiofrequency Therapy , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cetuximab , Combined Modality Therapy , Drug Delivery Systems/methods , Gold/administration & dosage , Gold/adverse effects , Humans , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/adverse effects , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/pathology , Radio Waves/adverse effects , Xenograft Model Antitumor Assays
19.
Surgery ; 148(2): 310-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20466402

ABSTRACT

BACKGROUND: Hepatocellular and pancreatic carcinomas are often auxotrophic for L-arginine, a semi-essential amino acid. The purpose of this study was to investigate cancer cell death using a significantly more active, cobalt-substituted bioengineered arginase. METHODS: Panc-1, a human pancreatic carcinoma cell line, and Hep 3B, a human hepatocellular carcinoma cell line, were exposed to L-arginase. Flow cytometry was used to measure expression of Ki-67, caspase-3, and argininosuccinate synthetase-1 (ASS-1) 4 days after treatment. An MTT assay measured proliferation. The Student t test determined statistical significance. RESULTS: Viability decreased by 31% +/- 2% for Panc-1 cells (P < .0001) and 34% +/- 1% (P < .0001) for Hep 3B cells after treatment. Both cell lines demonstrated a 4-fold increase activated caspase-3 expression after high dose treatment (P < .01), and 5-fold increase in ASS-1 expression (P < .002). Ki-67 expression did not vary in Hep 3B cells but decreased for Panc-1 cells (P < .015). The 50% inhibitory concentration was 8-fold higher for Panc-1 cells than for Hep 3B cells (P < .03). CONCLUSION: Increased ASS-1 expression by these cells, in order to increase L-arginine concentration, is inadequate, suggesting a mechanism by which arginine depletion can be used in multimodality therapy for arginine-dependent cancers.


Subject(s)
Arginase/pharmacology , Argininosuccinate Synthase/biosynthesis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Caspase 3/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Apoptosis/drug effects , Apoptosis/physiology , Arginase/administration & dosage , Arginine/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Induction/drug effects , Humans , Ki-67 Antigen/metabolism , Liver Neoplasms/pathology , Models, Biological , Pancreatic Neoplasms/pathology , Protein Engineering , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
20.
Cancer Res ; 69(19): 7851-9, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19773448

ABSTRACT

Many genetic and molecular alterations, such as K-ras mutation and NF-kappaB activation, have been identified in pancreatic cancer. However, the mechanisms by which pancreatic cancer metastasizes still remain to be determined. Although we previously showed that the tropomyosin-related kinase B (TrkB) was significantly correlated with the development of liver metastasis, its function in pancreatic cancer metastasis remained unresolved. In the present study, we showed that overexpressed TrkB is an alternatively spliced transcript variant of TrkB (TrkBT1) with a unique COOH-terminal 12-amino acid sequence and is mainly localized in the cytoplasm. Our results showed that overexpression of Flag-tagged TrkBT1 but not a Flag-tagged TrkBT1 COOH-terminal deletion mutant (Flag-TrkBT1DeltaC) in nonmetastatic pancreatic cancer cells enhanced cell proliferation, promoted formation of colonies in soft agar, stimulated tumor cell invasion, and induced liver metastasis in an orthotopic xenograft mouse model of pancreatic cancer. TrkBT1 interacted with Rho GDP dissociation inhibitor (GDI) in vivo, but Flag-TrkBT1DeltaC did not. Furthermore, overexpression of Flag-TrkBT1 and knockdown of RhoGDI expression by RhoGDI short hairpin RNAs promoted RhoA activation, but Flag-TrkBT1DeltaC overexpression did not. Therefore, our results showed that TrkBT1 overexpression induces liver metastasis of pancreatic cancer and uncovered a unique signaling mechanism by which TrkBT1 sequesters GDI and activates RhoA signaling.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Liver Neoplasms/secondary , Pancreatic Neoplasms/pathology , Receptor, trkB/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cadherins/biosynthesis , Cadherins/genetics , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Enzyme Activation , Female , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Protein Subunits , Receptor, trkB/biosynthesis , Receptor, trkB/genetics , Transplantation, Heterologous , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL