Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Exp Cell Res ; 371(2): 364-371, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30149000

ABSTRACT

Tumor-derived exosomes (TEX) play an important role in the escape of tumor cells from immune surveillance. However, the details of the mechanism are not fully understood. In this study, the apoptosis of CD4+ T cells increased during treatment with B16-derived exosomes in vitro and in vivo, resulting in accelerated growth of melanoma cells in mice. While the release of exosomes was blocked by disrupting the expression of Rab27a, tumor growth was clearly inhibited, and the percentage of T cells in the tumor environment increased. At the same time, Western blot showed that TEX could increase the activation of caspase-3, caspase-7 and caspase-9 but not caspase-8, down-regulating the anti-apoptotic proteins, including BCL-2, MCL-1 and BCL-xL in CD4+ T cells, and indicating that the TEX activates the mitochondrial apoptotic pathway of CD4+ T cells. These reductions were probably associated with the release of microRNAs, such as miR-690, from TEX to T cells. Our present study reveals for the first time that melanoma-released exosomes may directly activate the mitochondrial apoptotic pathway of CD4+ T cells through their microRNA cargo.


Subject(s)
Apoptosis/genetics , CD4-Positive T-Lymphocytes/metabolism , Exosomes/metabolism , Melanoma, Experimental/chemistry , MicroRNAs/genetics , Mitochondria/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation , Exosomes/chemistry , Exosomes/pathology , Female , Gene Expression Regulation , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Mitochondria/pathology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , bcl-X Protein/genetics , bcl-X Protein/metabolism
2.
Biochem Biophys Res Commun ; 496(4): 1357-1361, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29421654

ABSTRACT

In solid tumors, cancer cells survive and proliferate under conditions of microenvironment stress such as poor nutrients and hypoxia due to inadequate vascularization. These stress conditions in turn activate autophagy, which is important for cancer cell survival. However, autophagy has a contrary effect of inducing cell death in cancer cells cultured in vitro under conditions of glucose deprivation. In this study, we hypothesized that supplementation of lactic acid serves as a means of cell survival under glucose-deprived conditions. At neutral pH, cell death of B16 murine melanoma cells by autophagy under glucose-deprived conditions was observed. However, supplementation of lactic acid suppressed cell death and autophagy in B16 melanoma cells when cultured in glucose-deprived conditions. Sodium lactate, which does not change extracellular pH, did not inhibit cell death, while HCl-adjusted acidic pH suppressed cell death under glucose-deprived conditions. These results suggested that an acidic pH is crucial for cell survival under glucose-deprived conditions.


Subject(s)
Apoptosis , Autophagy , Glucose/metabolism , Lactic Acid/metabolism , Melanoma, Experimental/metabolism , Melanoma/chemistry , Melanoma/metabolism , Animals , Cell Line, Tumor , Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Hydrogen-Ion Concentration , Lactic Acid/chemistry , Melanoma/pathology , Melanoma, Experimental/chemistry , Melanoma, Experimental/pathology , Mice , Tumor Microenvironment
3.
J Surg Res ; 194(2): 688-695, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466519

ABSTRACT

BACKGROUND: Some malignant cancers show high levels of local invasiveness by the secretion of soluble factors that can degrade adjacent tissues and suppress surrounding cell growth. We investigated the possibility of treating fibroproliferative scars based on these properties of malignant melanoma. MATERIAL AND METHODS: B16 melanoma-conditioned medium (B16 M-CM) was added to keloid fibroblasts (KFs), and proliferation, migration, and type I collagen production were measured. The cell cycle and signaling pathways were also analyzed. Proteins associated with cell proliferation were measured with Western blot analysis. Animal experiments using a rabbit ear model was performed to confirm the effect of B16 M-CM in vivo. RESULTS: B16 M-CM reduced proliferation, migration, and type I collagen production of KFs. This treatment also increased the number of cells in the subG1 phase and decreased phosphorylation levels of AKT, extracellular signal-regulated kinase1/2, cyclin D1, and c-Myc of KFs. Additionally, B16 M-CM reduced the thickness of rabbit ear scars in the rabbit ear model in vivo. CONCLUSIONS: B16 M-CM can suppress proliferation, migration, and type I collagen production of KFs. In addition, concentrated B16 M-CM reduced scar thickness in the rabbit ear model. The specific proteins involved should be identified in a future study.


Subject(s)
Fibroblasts/drug effects , Keloid/prevention & control , Melanoma, Experimental/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/biosynthesis , Culture Media, Conditioned/pharmacology , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rabbits
4.
Nanotechnology ; 26(28): 285101, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26111959

ABSTRACT

This study examines the effects of polyethylene glycol (PEG) and peptide conjugation on the biodistribution of ultrasmall (2.7 nm) gold nanoparticles in mice bearing B16 melanoma allografts. Nanoparticles were delivered intravenously, and biodistribution was measured at specific timepoints by organ digestion and inductively coupled plasma mass spectrometry. All major organs were examined. Two peptides were tested: the cyclic RGD peptide (cRGD, which targets integrins); and a recently described peptide derived from the myxoma virus. We found the greatest specific tumor delivery using the myxoma peptide, with or without PEGylation. Un-PEGylated cRGD performed poorly, but PEGylated RGD showed a significant transient collection in the tumor. Liver and kidney were the primary targets of all constructs. None of the particles were able to cross the blood-brain barrier. Although it was able to deliver Au to B16 cells, the myxoma peptide did not show any cytotoxic activity against these cells, in contrast to previous reports. These results indicate that the effect of passive targeting by PEGylation and active targeting by peptides can be independent or combined, and that they should be evaluated on a case-by-case basis when designing new nanosystems for targeted therapies. Both myxoma peptide and cRGD should be considered for specific targeting to melanoma, but a thorough investigation of the cytotoxicity of the myxoma peptide to different cell lines remains to be performed.


Subject(s)
Gold/chemistry , Melanoma, Experimental/chemistry , Metal Nanoparticles/chemistry , Peptides/pharmacokinetics , Allografts , Animals , Body Fluids/chemistry , Cell Line, Tumor , Drug Delivery Systems , Gold/pharmacokinetics , Mice , Myxoma virus/chemistry , Peptides/chemistry , Peptides, Cyclic/pharmacokinetics , Polyethylene Glycols/chemistry , Tissue Distribution
5.
J Surg Res ; 190(2): 528-34, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24923630

ABSTRACT

BACKGROUND: Dendritic cells (DC) are localized in close proximity to cancer cells in many well-known tumors, and thus maybe a useful target for tumor margin assessment. MATERIALS AND METHODS: [(99m)Tc]- cyanine 7 (Cy7)-tilmanocept was synthesized and in vitro binding assays to bone marrow-derived DC were performed. Fifteen mice, implanted with either 4T1 mouse mammary or K1735 mouse melanoma tumors, were administered 1.0 nmol of [(99m)Tc]-Cy7-tilmanocept via tail vein injection. After fluorescence imaging 1 or 2 h after injection, the tumor, muscle, and blood were assayed for radioactivity to calculate percent-injected dose. Digital images of the tumors after immunohistochemical staining for DC were analyzed to determine DC density. RESULTS: In vitro binding demonstrated subnanomolar affinity of [(99m)Tc]-Cy7-tilmanocept to DC (KA = 0.31 ± 0.11 nM). After administration of [(99m)Tc]-Cy7-tilmanocept, fluorescence imaging showed a 5.5-fold increase in tumor signal as compared with preinjection images and a 3.3-fold difference in fluorescence activity when comparing the tumor with the surgical bed after tumor excision. Immunohistochemical staining analysis demonstrated that DC density positively correlated with tumor percent of injected dose per gram (r = 0.672, P = 0.03), and higher DC density was observed at the periphery versus center of the tumor (186 ± 54 K versus 64 ± 16 K arbitrary units, P = 0.001). CONCLUSIONS: [(99m)Tc]-Cy7-tilmanocept exhibits in vitro and in vivo tumor-specific binding to DC and maybe useful as a tumor margin targeting agent.


Subject(s)
Benzothiazoles , Carbocyanines , Dendritic Cells/pathology , Dextrans , Mammary Neoplasms, Experimental/pathology , Mannans , Melanoma, Experimental/pathology , Technetium Tc 99m Pentetate/analogs & derivatives , Animals , Benzothiazoles/chemistry , CD11c Antigen/analysis , CD11c Antigen/chemistry , Carbocyanines/chemistry , Cell Line , Cell Line, Tumor , Dendritic Cells/chemistry , Dextrans/chemistry , Female , Mammary Neoplasms, Experimental/chemistry , Mannans/chemistry , Melanoma, Experimental/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Microscopy, Fluorescence , Technetium Tc 99m Pentetate/chemistry , Ultraviolet Rays
6.
Naturwissenschaften ; 101(2): 87-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24424785

ABSTRACT

During the first 24 h after removal from incubation, melanoma cells in culture displayed reliable increases in emissions of photons of specific wavelengths during discrete portions of this interval. Applications of specific filters revealed marked and protracted increases in infrared (950 nm) photons about 7 h after removal followed 3 h later by marked and protracted increases in near ultraviolet (370 nm) photon emissions. Specific wavelengths within the visible (400 to 800 nm) peaked 12 to 24 h later. Specific activators or inhibitors for specific wavelengths based upon Cosic's resonant recognition model elicited either enhancement or diminishment of photons at the specific wavelength as predicted. Inhibitors or activators predicted for other wavelengths, even within 10 nm, were less or not effective. There is now evidence for quantitative coupling between the wavelength of photon emissions and intrinsic cellular chemistry. The results are consistent with initial activation of signaling molecules associated with infrared followed about 3 h later by growth and protein-structural factors associated with ultraviolet. The greater-than-expected photon counts compared with raw measures through the various filters, which also function as reflective material to other photons, suggest that photons of different wavelengths might be self-stimulatory and could play a significant role in cell-to-cell communication.


Subject(s)
Macromolecular Substances/chemistry , Melanoma, Experimental/physiopathology , Models, Theoretical , Photons , Light , Melanoma, Experimental/chemistry , Tumor Cells, Cultured , Ultraviolet Rays
7.
Phytother Res ; 28(2): 200-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23519930

ABSTRACT

It is well-documented that macrophages have the functions to regulate antitumor immune response. Antitumor response can be launched by a series of events, starting with inflammation mediated by monocyte/macrophages, which stimulates natural killer and dendritic cells and finally activates the cytotoxic lymphoid system. Monocytes/macrophages may be the first line of defense in tumors. However, specific and nonspecific immunotherapy for human cancer has shown no success or limited success in clinical trials. Part of the reasons attribute to tumor-derived soluble factors that suppress functions of immune cells or induce apoptosis of these cells, including macrophages. Therefore, antagonism of the suppression on the macrophages is an important goal for tumor immunotherapy. To achieve this purpose, Ganoderma lucidum polysaccharides (Gl-PS) with multiple bioactivities were used on mouse peritoneal macrophages incubating with culture supernatants of B16F10 melanoma cells (B16F10-CS). It was shown that the viability, phagocytic activity, NO production, TNF-α production and activity in peritoneal macrophages after activation by lipopolysaccharide were suppressed by B16F10-CS, while the suppressions were fully or partially antagonized by Gl-PS. In conclusion, B16F10-CS is suppressive to the viability, phagocytic activity, NO production, TNF-α production and activity in peritoneal macrophages while Gl-PS had the antagonistic effects against this suppression, suggesting this potential of Gl-PS to facilitate cancer immunotherapy.


Subject(s)
Culture Media/chemistry , Macrophages, Peritoneal/drug effects , Melanoma, Experimental/chemistry , Polysaccharides/pharmacology , Reishi/chemistry , Animals , Cell Survival , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Phagocytosis/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
Magn Reson Med ; 70(6): 1748-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23401099

ABSTRACT

PURPOSE: Combined therapeutic and diagnostic agents, "theranostics" are emerging valuable tools for noninvasive imaging and drug delivery. Here, we report on a solid biodegradable multifunctional nanoparticle that combines both features. METHODS: Poly(lactide-co-glycolide) nanoparticles were engineered to confine superparamagnetic iron oxide contrast for magnetic resonance imaging while enabling controlled drug delivery and targeting to specific cells. To achieve this dual modality, fatty acids were used as anchors for surface ligands and for encapsulated iron oxide in the polymer matrix. RESULTS: We demonstrate that fatty acid modified iron oxide prolonged retention of the contrast agent in the polymer matrix during degradative release of drug. Antibody-fatty acid surface modification facilitated cellular targeting and subsequent internalization in cells while inducing clustering of encapsulated fatty-acid modified superparamagnetic iron oxide during particle formulation. This induced clustered confinement led to an aggregation within the nanoparticle and, hence, higher transverse relaxivity, r2 , (294 mM(-1) s(-1) ) compared with nanoparticles without fatty-acid ligands (160 mM(-1) s(-1) ) and higher than commercially available superparamagnetic iron oxide nanoparticles (89 mM(-1) s(-1) ). CONCLUSION: Clustering of superparamagnetic iron oxide in poly(lactide-co-glycolide) did not affect the controlled release of encapsulated drugs such as methotrexate or clodronate and their subsequent pharmacological activity, thus highlighting the full theranostic capability of our system.


Subject(s)
Absorbable Implants , Dextrans/chemistry , Macrophages/chemistry , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Melanoma, Experimental/chemistry , Nanocapsules/chemistry , Animals , Cells, Cultured , Dextrans/therapeutic use , Diffusion , Drug Compounding/methods , Magnetite Nanoparticles/therapeutic use , Materials Testing , Melanoma, Experimental/diagnosis , Melanoma, Experimental/therapy , Mice , Nanocapsules/therapeutic use
9.
Int J Hyperthermia ; 29(2): 121-32, 2013.
Article in English | MEDLINE | ID: mdl-23418916

ABSTRACT

PURPOSE: Evaluation of the effectiveness of highly focalised thermotherapy (HFT) in a melanoma mouse model, using a ferrimagnetic cement (FC) and repeated low hyperthermia treatments. MATERIALS AND METHODS: A melanoma mouse model was induced with B16F10 cells in C57BL6 mice. The FC, injected into the tumour, was used as the magnetic vehicle for HFT. FC location within the tumour was assessed by radiography and its capability to generate heat, when exposed to an external high frequency magnetic field (HFMF), monitored by thermal camera. The HFT treatment consisted of three HFMF exposures, with 48-h intervals, each one lasting 30 min, with a 5-6°C tumour temperature increase. At the end of the experiment, FC samples were characterised by scanning electron microscopy (SEM) and energy dispersion spectroscopy (EDS). The presence of iron contents was analysed in the tumour, lungs, liver and spleen. Histological evaluation and immunohistochemical staining for caspase-3 were performed. Tumour growth was monitored during the experiment. RESULTS: Surface analysis showed FC stabilisation within the tumour, and iron was absent. The thermal camera confirmed the localised temperature increase in the tumour. HFT treatments inhibited the tumour growth by ∼70% compared to controls. This was due to cell destruction by necrosis and apoptosis. CONCLUSIONS: The HFT, using the FC, proved to be a minimally invasive technique that statistically inhibited tumour growth. Results suggested that this methodology seems to be a promising technique for the treatment of solid tumours, allowing repeated low hyperthermia treatments, which can be easier and less traumatic than other hyperthermia techniques.


Subject(s)
Bone Cements/therapeutic use , Ferric Compounds/therapeutic use , Glass Ionomer Cements/therapeutic use , Hyperthermia, Induced/methods , Melanoma, Experimental/therapy , Animals , Caspase 3/analysis , Cell Line, Tumor , Female , Melanoma, Experimental/chemistry , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL
10.
Article in English | MEDLINE | ID: mdl-22038357

ABSTRACT

This study explored the possibility that the components in melanoma cytoplasm induce murine BMSCs transformation and expression of Melan-A by morphologically observing the changes of BMSCs and immunocytochemically detecting Melan-A in the cells after culturing BMSCs in medium containing melanoma cytoplasm components (MCC). MCC of B16 melanoma cells was prepared and BMSCs were cultured and induced by adding the MCC into culture medium. The cells were morphologically observed and Melan-A was immunohistochemically detected to confirm BMSCs transformation. MCC-induced BMSCs underwent morphological changes. A number of melanin granules appeared in the cytoplasm of the cells and some were released into surrounding areas. Several cells that might come from one cell formed a cluster, and their granules, together with those secreted by other induced BMSCs, formed a so-called "sphere-formed structure". The induced BMSCs expressed Melan-A. We are led to conclude that there might be some factors in the cytoplasm of melanoma cells that might induce BMSCs transformation toward melanogenic cell, or even melanoma.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cytoplasm/chemistry , MART-1 Antigen/metabolism , Melanoma, Experimental/pathology , Skin Neoplasms/pathology , Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Coculture Techniques , Humans , Male , Melanoma, Experimental/chemistry , Mice , Skin Neoplasms/chemistry
11.
Clin Cancer Res ; 27(7): 1932-1940, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33408249

ABSTRACT

PURPOSE: Therapeutic checkpoint inhibitors on tumor-infiltrating lymphocytes (TIL) are being increasingly utilized in the clinic. The T-cell immunoreceptor with Ig and ITIM domains (TIGIT) is an inhibitory receptor expressed on T and natural killer cells. The TIGIT signaling pathway is an alternative target for checkpoint blockade to current PD-1/CTLA-4 strategies. Elevated TIGIT expression in the tumor microenvironment correlates with better therapeutic responses to anti-TIGIT therapies in preclinical models. Therefore, quantifying TIGIT expression in tumors is necessary for determining whether a patient may respond to anti-TIGIT therapy. PET imaging of TIGIT expression on TILs can therefore aid diagnosis and in monitoring therapeutic responses. EXPERIMENTAL DESIGN: Antibody-based TIGIT imaging radiotracers were developed with the PET radionuclides copper-64 (64Cu) and zirconium-89 (89Zr). In vitro characterization of the imaging probes was followed by in vivo evaluation in both xenografts and syngeneic tumor models in mouse. RESULTS: Two anti-TIGIT probes were developed and exhibited immunoreactivity of >72%, serum stability of >95%, and specificity for TIGIT with both mouse TIGIT-expressing HeLa cells and ex vivo-activated primary splenocytes. In vivo, the 89Zr-labeled probe demonstrated superior contrast than the 64Cu probe due to 89Zr's longer half-life matching the TIGIT antibody's pharmacokinetics. The 89Zr probe was used to quantify TIGIT expression on TILs in B16 melanoma in immunocompetent mice and confirmed by ex vivo flow cytometry. CONCLUSIONS: This study develops and validates novel TIGIT-specific 64Cu and 89Zr PET probes for quantifying TIGIT expression on TILs for diagnosis of patient selection for anti-TIGIT therapies.


Subject(s)
Lymphocytes, Tumor-Infiltrating/chemistry , Positron-Emission Tomography/methods , Receptors, Immunologic/analysis , Animals , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Female , Humans , Isotope Labeling , Melanoma, Experimental/chemistry , Mice , Mice, Inbred C57BL , Receptors, Immunologic/antagonists & inhibitors , Tumor Microenvironment
12.
J Org Chem ; 75(22): 7920-3, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20954692

ABSTRACT

A straightforward access to pyrrolidine-based analogues of jaspine B was developed. Five stereoisomers were prepared including the all-cis derivatives presenting the configuration of the natural anhydrophytosphingosine. The synthesis of the latter relied on an original Staudinger-type cyclization process. The compounds were evaluated regarding their ability to alter tumor cells' viability and to interfere with the metabolism of sphingolipids.


Subject(s)
Melanoma, Experimental/chemistry , Pyrrolidines/chemistry , Sphingosine/analogs & derivatives , Animals , Cell Line, Tumor , Humans , Melanoma, Experimental/metabolism , Mice , Sphingosine/chemical synthesis , Sphingosine/chemistry , Sphingosine/metabolism , Sphingosine/pharmacology , Stereoisomerism
13.
EBioMedicine ; 58: 102902, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32707448

ABSTRACT

BACKGROUND: The developing zebrafish is an emerging tool in nanomedicine, allowing non-invasive live imaging of the whole animal at higher resolution than is possible in the more commonly used mouse models. In addition, several transgenic fish lines are available endowed with selected cell types expressing fluorescent proteins; this allows nanoparticles to be visualized together with host cells. METHODS: Here, we introduce the zebrafish neural tube as a robust injection site for cancer cells, excellently suited for high resolution imaging. We use light and electron microscopy to evaluate cancer growth and to follow the fate of intravenously injected nanoparticles. FINDINGS: Fluorescently labelled mouse melanoma B16 cells, when injected into this structure proliferated rapidly and stimulated angiogenesis of new vessels. In addition, macrophages, but not neutrophils, selectively accumulated in the tumour region. When injected intravenously, nanoparticles made of Cy5-labelled poly(ethylene glycol)-block-poly(2-(diisopropyl amino) ethyl methacrylate) (PEG-PDPA) selectively accumulated in the neural tube cancer region and were seen in individual cancer cells and tumour associated macrophages. Moreover, when doxorubicin was released from PEG-PDPA, in a pH dependant manner, these nanoparticles could strongly reduce toxicity and improve the treatment outcome compared to the free drug in zebrafish xenotransplanted with mouse melanoma B16 or human derived melanoma cells. INTERPRETATION: The zebrafish has the potential of becoming an important intermediate step, before the mouse model, for testing nanomedicines against patient-derived cancer cells. FUNDING: We received funding from the Norwegian research council and the Norwegian cancer society.


Subject(s)
Doxorubicin/administration & dosage , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/drug therapy , Polymethacrylic Acids/administration & dosage , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/drug therapy , Administration, Intravenous , Animals , Carbocyanines/chemistry , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Humans , Macrophages/chemistry , Melanoma, Experimental/chemistry , Melanoma, Experimental/pathology , Mice , Microscopy, Electron , Nanoparticles , Neoplasm Transplantation , Neural Tube/chemistry , Neutrophils/chemistry , Polyethylene Glycols/chemistry , Polymethacrylic Acids/chemistry , Skin Neoplasms/chemistry , Skin Neoplasms/pathology , Treatment Outcome , Zebrafish
14.
Biomater Sci ; 8(11): 3052-3062, 2020 Jun 07.
Article in English | MEDLINE | ID: mdl-32347852

ABSTRACT

Inefficient nanoparticle accumulation in solid tumors hinders the clinical translation of cancer nanomedicines. Herein, we proposed that sildenafil, a vasodilator ampholyte, could be used to promote nanoparticle accumulation by inducing vasodilation after its tumor acidity-triggered release from the nanocarriers. To confirm this, sildenafil was first encapsulated in a cisplatin-incorporated polymeric micelle. The dense PEG shell of the micelle reduced its endocytosis by cancer cells, which in return resulted in accumulative extracellular release of protonated sildenafil in the acidic tumor microenvironment. The released sildenafil was found to be more effective in enlarging the tumor blood vessels than could be achieved without sildenafil. As a result, we demonstrated considerable improvement in the intratumoral accumulation of the sildenafil-cisplatin co-loaded nanoparticle and its enhanced cancer therapeutic efficacy over the control group. Given the generality of a dense PEG shell and a hydrophobic part in most clinically developed nanomedicines, this work implies the great potential of sildenafil as a simple and universal adjuvant to selectively promote the intratumoral accumulation of nanomedicines, thus improving their clinical translation.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Melanoma, Experimental/metabolism , Nanoparticles/administration & dosage , Sildenafil Citrate/administration & dosage , Vasodilator Agents/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Transport , Blood Vessels/physiology , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Drug Liberation , Female , Hydrogen-Ion Concentration , Melanoma, Experimental/blood supply , Melanoma, Experimental/chemistry , Melanoma, Experimental/drug therapy , Mice, Inbred C57BL , Micelles , Nanoparticles/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/chemistry , Polyglutamic Acid/pharmacokinetics , Rats, Sprague-Dawley , Sildenafil Citrate/chemistry , Sildenafil Citrate/pharmacokinetics , Tissue Distribution , Vasodilation , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacokinetics
15.
Cancer Immunol Immunother ; 58(5): 769-75, 2009 May.
Article in English | MEDLINE | ID: mdl-18633610

ABSTRACT

Intravenously-applied bacteria tend to accumulate in tumors and can sporadically lead to tumor regression. Systemic administration of attenuated Salmonella typhimurium is safe and has shown no significant adverse effects in humans. The purpose of this study was to test the hypothesis that engineering S. typhimurium to express a chemokine, CCL21, would increase anti-tumor activity. We engineered an attenuated strain of S. typhimurium to produce the chemokine CCL21. Attenuated S. typhimurium expressing CCL21 significantly inhibited the growth of primary tumors and pulmonary metastases in preclinical models of multi-drug-resistant murine carcinomas, while control bacteria did not. Histological analysis of tumors showed marked inflammatory cell infiltrates in mice treated with CCL21-expressing but not control bacteria. Levels of cytokines and chemokines known to be induced by CCL21 [e.g., interferon-gamma (INFgamma), CXCL9, and CXCL10] were significantly elevated in tumors of mice treated with CCL21-expressing but not control S. typhimurium. The anti-tumor activity was found to be dependent on CD4- and CD8-expressing cells, based on antibody-mediated in vivo immuno-depletion experiments. Anti-tumor activity was achieved without evidence of toxicity. In summary, chemokine-expressing, attenuated bacteria may provide a novel approach to cancer immunotherapy for effective and well-tolerated in vivo delivery of immunomodulatory proteins.


Subject(s)
Adenocarcinoma/therapy , Bacterial Vaccines/therapeutic use , Cancer Vaccines/therapeutic use , Chemokine CCL21/physiology , Immunotherapy, Active , Melanoma, Experimental/therapy , Salmonella typhimurium/genetics , Adenocarcinoma/chemistry , Adenocarcinoma/immunology , Adenocarcinoma/prevention & control , Adenocarcinoma/secondary , Animals , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor/transplantation , Chemokine CCL21/genetics , Chemokines/analysis , Colonic Neoplasms/pathology , Cytokines/analysis , Female , Lung Neoplasms/chemistry , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/chemistry , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/physiology , Salmonella typhimurium/metabolism , Vaccines, Attenuated/therapeutic use , Xenograft Model Antitumor Assays
16.
Science ; 257(5073): 1121-4, 1992 Aug 21.
Article in English | MEDLINE | ID: mdl-1509264

ABSTRACT

Complementary DNA clones from the pink-eyed dilution (p) locus of mouse chromosome 7 were isolated from murine melanoma and melanocyte libraries. The transcript from this gene is missing or altered in six independent mutant alleles of the p locus, suggesting that disruption of this gene results in the hypopigmentation phenotype that defines mutant p alleles. Characterization of the human homolog revealed that it is localized to human chromosome 15 at q11.2-q12, a region associated with Prader-Willi and Angelman syndromes, suggesting that altered expression of this gene may be responsible for the hypopigmentation phenotype exhibited by certain individuals with these disorders.


Subject(s)
Carrier Proteins , Membrane Proteins , Membrane Transport Proteins , Pigmentation Disorders/genetics , Prader-Willi Syndrome/genetics , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 15 , Cloning, Molecular , DNA/genetics , Humans , Melanocytes/chemistry , Melanoma, Experimental/chemistry , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutation , Nucleic Acid Hybridization , Phenotype , Proteins/chemistry , Sequence Homology, Nucleic Acid
17.
Nutr Cancer ; 61(4): 518-29, 2009.
Article in English | MEDLINE | ID: mdl-19838924

ABSTRACT

Methionine (Met) deprivation stress (MDS) is proposed in association with chemotherapy in the treatment of some cancers. A synergistic effect of this combination is generally acknowledged. However, little is known on the mechanism of the response to this therapeutic strategy. A model of B16 melanoma tumor in vivo was treated by MDS alone and in combination with chloroethylnitrosourea (CENU). It was applied recent developments in proton-NMR spectroscopy-based metabolomics for providing information on the metabolic response of tumors to MDS and combination with chemotherapy. MDS inhibited tumor growth during the deprivation period and growth resumption thereafter. The combination of MDS with CENU induced an effective time-dependent synergy on growth inhibition. Metabolite profiling during MDS showed a decreased Met content (P < 0.01) despite the preservation of the protein content, disorders in sulfur-containing amino acids, glutamine/proline, and phospholipid metabolism [increase of glycerophosphorylcholine (P < 0.01), decrease in phosphocholine (P < 0.05)]. The metabolic profile of MDS combined with CENU and ANOVA analysis revealed the implication of Met and phospholipid metabolism in the observed synergy, which may be interpreted as a Met-sparing metabolic reprogramming of tumors. It follows that combination therapy of MDS with CENU seems to intensify adaptive processes, which may set limitations to this therapeutic strategy.


Subject(s)
Adaptation, Physiological/drug effects , Antineoplastic Agents/therapeutic use , Melanoma, Experimental/therapy , Methionine/deficiency , Methionine/metabolism , Nitrosourea Compounds/therapeutic use , Phospholipids/metabolism , Analysis of Variance , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Combined Modality Therapy , Confidence Intervals , Growth Inhibitors/therapeutic use , Liver/drug effects , Male , Melanoma, Experimental/chemistry , Melanoma, Experimental/diet therapy , Melanoma, Experimental/metabolism , Metabolome/drug effects , Methionine/analysis , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Organ Size/drug effects , Phosphatidylethanolamine N-Methyltransferase/metabolism , Phospholipids/analysis , Random Allocation , Stress, Physiological/drug effects , Time Factors , Tumor Burden/drug effects
18.
Oncol Res ; 17(11-12): 601-12, 2009.
Article in English | MEDLINE | ID: mdl-19806791

ABSTRACT

Metastasis is a complex, multistep process by which a cancer cell leaves the primary tumor, travels to a distant site via the circulatory system, and establishes a secondary cancer. A deeper understanding of the molecular events underlying metastasis will provide information that will be useful for the development of new diagnostic and therapeutic strategies. The B16 and B16F10 mouse melanoma cell lines are widely used as model system for studying many aspects of cancer biology including metastasis. Compared with B16, which has a low metastatic potential, the highly metastatic cell line B16F10 displayed a higher metastatic ability along with higher expression levels of the metastasis-associated phosphatase of regenerating liver-3 (PRL-3). B16 cells transfected with PRL-3 cDNA (B16-PRL3) had metastatic abilities comparable to those of Bl16F10 cells. To study the molecular mechanisms that underlie metastasis, the proteomes of the B16, B16F10, and B16-PRL3 cell lines were compared using two-dimensional differential in-gel electrophoresis. Proteins that varied significantly in levels between these cell lines were selected and identified using mass spectrometry. Interestingly, many proteins, especially those present in membrane fractions, were similarly up- or downregulated in both the Bl16F10 and B16-PRL3 cells lines compared to B16 cell lines. The list of similarly regulated proteins included heat shock protein 70, fascin-1, septin-6, ATP synthase beta subunit, and bone morphogenic protein receptor type IB. These proteins may play a causal role in PRL-3-mediated metastasis. These investigations open an avenue for the further characterization of the molecular mechanisms that underlie metastasis.


Subject(s)
Immediate-Early Proteins/analysis , Melanoma, Experimental/chemistry , Protein Tyrosine Phosphatases/analysis , Proteomics , Animals , Bone Morphogenetic Protein Receptors, Type I/analysis , Cell Line, Tumor , Cell Movement , Electrophoresis, Gel, Two-Dimensional , HSP70 Heat-Shock Proteins/analysis , Immediate-Early Proteins/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Protein Tyrosine Phosphatases/genetics , Proton-Translocating ATPases/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Clin Cancer Res ; 14(21): 6877-85, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18980982

ABSTRACT

PURPOSE: The National Cancer Institute has completed a first-in-human clinical pharmacodynamic trial of the targeted agent ABT-888, a poly (ADP-ribose) polymerase (PARP) inhibitor, under the auspices of the U.S. Food and Drug Administration's Exploratory Investigational New Drug Application. Performance of the study design, needle biopsy procedure, and validated pharmacodynamic assay were evaluated in human tumor xenograft models. EXPERIMENTAL DESIGN: A validated ELISA was used to quantify PAR, a product of the PARP 1/2 enzyme activity. Sampling variability from tumor heterogeneity was determined by comparing PAR content in multiple tumors, and in different areas of the same tumor in a particular animal, collected under anesthesia by needle biopsy or resection before and after administration of nontoxic doses of ABT-888. The degree of PARP inhibition following single-dose treatment was evaluated in the time frame anticipated for biopsy in humans. RESULTS: Sampling variability around the mean (approximately 50%) for untreated and vehicle-treated animals was random and due to specimen heterogeneity. PAR levels in initial and repeat tumor biopsies, separated by 1 week, were not altered by the stress induced by daily handling of the animals. A single ABT-888 dose (3 or 12.5 mg/kg) reduced intratumor PAR levels by >95%. ABT-888 (1.56-25 mg/kg) significantly decreased PAR levels at 2 h post-dosing. CONCLUSION: The detailed methodologies developed for this study facilitated the design of a phase 0, first-in-human clinical trial of ABT-888 and could serve as a model for developing proof-of-principle clinical trials of molecularly targeted anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Clinical Trials as Topic , Melanoma, Experimental/chemistry , Models, Biological , Poly(ADP-ribose) Polymerases/analysis , Animals , Benzimidazoles/therapeutic use , Biopsy, Needle , Drug Delivery Systems , Humans , Melanoma, Experimental/drug therapy , Mice , Mice, Nude , Poly(ADP-ribose) Polymerase Inhibitors , Transplantation, Heterologous , Xenograft Model Antitumor Assays
20.
Int J Radiat Oncol Biol Phys ; 105(5): 1126-1136, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31461675

ABSTRACT

PURPOSE: Synchrotron microbeam radiation therapy (MRT) is a method that spatially distributes the x-ray beam into several microbeams of very high dose (peak dose), regularly separated by low-dose intervals (valley dose). MRT selectively spares normal tissues, relative to conventional (uniform broad beam [BB]) radiation therapy. METHODS AND MATERIALS: To evaluate the effect of MRT on radioresistant melanoma, B16-F10 murine melanomas were implanted into mice ears. Tumors were either treated with MRT (407.6 Gy peak; 6.2 Gy valley dose) or uniform BB irradiation (6.2 Gy). RESULTS: MRT induced significantly longer tumor regrowth delay than did BB irradiation. A significant 24% reduction in blood vessel perfusion was observed 5 days after MRT, and the cell proliferation index was significantly lower in melanomas treated by MRT compared with BB. MRT provoked a greater induction of senescence in melanoma cells. Bio-Plex analyses revealed enhanced concentration of monocyte-attracting chemokines in the MRT group: MCP-1 at D5, MIP-1α, MIP-1ß, IL12p40, and RANTES at D9. This was associated with leukocytic infiltration at D9 after MRT, attributed mainly to CD8 T cells, natural killer cells, and macrophages. CONCLUSIONS: In light of its potential to disrupt blood vessels that promote infiltration of the tumor by immune cells and its induction of senescence, MRT could be a new therapeutic approach for radioresistant melanoma.


Subject(s)
Ear Neoplasms/radiotherapy , Melanoma, Experimental/radiotherapy , Radiation Tolerance , Synchrotrons , Animals , Cell Proliferation/radiation effects , Cellular Senescence , Ear Neoplasms/blood supply , Ear Neoplasms/metabolism , Ear Neoplasms/pathology , Female , Melanoma, Experimental/blood supply , Melanoma, Experimental/chemistry , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/metabolism , Staining and Labeling , Tumor Burden , Tumor Microenvironment , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL