Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Angiogenesis ; 26(2): 279-293, 2023 05.
Article in English | MEDLINE | ID: mdl-36459240

ABSTRACT

PURPOSE: Ongoing angiogenesis renders the tumor endothelium unresponsive to inflammatory cytokines and interferes with adhesion of leukocytes, resulting in escape from immunity. This process is referred to as tumor endothelial cell anergy. We aimed to investigate whether anti-angiogenic agents can overcome endothelial cell anergy and provide pro-inflammatory conditions. EXPERIMENTAL DESIGN: Tissues of renal cell carcinoma (RCC) patients treated with VEGF pathway-targeted drugs and control tissues were subject to RNAseq and immunohistochemical profiling of the leukocyte infiltrate. Analysis of adhesion molecule regulation in cultured endothelial cells, in a preclinical model and in human tissues was performed and correlated to leukocyte infiltration. RESULTS: It is shown that treatment of RCC patients with the drugs sunitinib or bevacizumab overcomes tumor endothelial cell anergy. This treatment resulted in an augmented inflammatory state of the tumor, characterized by enhanced infiltration of all major leukocyte subsets, including T cells, regulatory T cells, macrophages of both M1- and M2-like phenotypes and activated dendritic cells. In vitro, exposure of angiogenic endothelial cells to anti-angiogenic drugs normalized ICAM-1 expression. In addition, a panel of tyrosine kinase inhibitors was shown to increase transendothelial migration of both non-adherent and monocytic leukocytes. In primary tumors of RCC patients, ICAM-1 expression was found to be significantly increased in both the sunitinib and bevacizumab-treated groups. Genomic analysis confirmed the correlation between increased immune cell infiltration and ICAM-1 expression upon VEGF-targeted treatment. CONCLUSION: The results support the emerging concept that anti-angiogenic therapy can boost immunity and show how immunotherapy approaches can benefit from combination with anti-angiogenic compounds.


Subject(s)
Angiogenesis Inhibitors , Carcinoma, Renal Cell , Endothelial Cells , Kidney Neoplasms , Neovascularization, Pathologic , Humans , Bevacizumab/immunology , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium/drug effects , Endothelium/immunology , Endothelium/pathology , Intercellular Adhesion Molecule-1/immunology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Sunitinib/immunology , Sunitinib/pharmacology , Sunitinib/therapeutic use , Vascular Endothelial Growth Factor A/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Neoplasm Invasiveness/immunology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use
2.
Angiogenesis ; 24(3): 597-611, 2021 08.
Article in English | MEDLINE | ID: mdl-33629198

ABSTRACT

Antibiotic-induced microbial imbalance, or dysbiosis, has systemic and long-lasting effects on the host and response to cancer therapies. However, the effects on tumor endothelial cells are largely unknown. Therefore, the goal of the current study was to generate matched B16-F10 melanoma associated endothelial cell lines isolated from mice with and without antibiotic-induced dysbiosis. After validating endothelial cell markers on a genomic and proteomic level, functional angiogenesis assays (i.e., migration and tube formation) also confirmed their vasculature origin. Subsequently, we found that tumor endothelial cells derived from dysbiotic mice (TEC-Dys) were more sensitive to ionizing radiotherapy in the range of clinically-relevant hypofractionated doses, as compared to tumor endothelial cells derived from orthobiotic mice (TEC-Ortho). In order to identify tumor vasculature-associated drug targets during dysbiosis, we used tandem mass tag mass spectroscopy and focused on the statistically significant cellular membrane proteins overexpressed in TEC-Dys. By these criteria c-Met was the most differentially expressed protein, which was validated histologically by comparing tumors with or without dysbiosis. Moreover, in vitro, c-Met inhibitors Foretinib, Crizotinib and Cabozantinib were significantly more effective against TEC-Dys than TEC-Ortho. In vivo, Foretinib inhibited tumor growth to a greater extent during dysbiosis as compared to orthobiotic conditions. Thus, we surmise that tumor response in dysbiotic patients may be greatly improved by targeting dysbiosis-induced pathways, such as c-Met, distinct from the many targets suppressed due to dysbiosis.


Subject(s)
Dysbiosis , Endothelial Cells/enzymology , Melanoma, Experimental , Neovascularization, Pathologic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met , Animals , Dysbiosis/enzymology , Dysbiosis/microbiology , Melanoma, Experimental/blood supply , Melanoma, Experimental/enzymology , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/microbiology , Neovascularization, Pathologic/therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Radiotherapy
3.
J Appl Toxicol ; 41(9): 1456-1466, 2021 09.
Article in English | MEDLINE | ID: mdl-33417269

ABSTRACT

The use of synthetic materials for biomedical applications is ever expanding. One of the major requirements for these materials is biocompatibility, which includes prevention of immune system responses. Due to the inherent complexity of their structural composition, the polyurethane (PU) family of polymers is being used in a variety of medical applications, from soft and hard tissue scaffolds to intricate coatings on implantable devices. Herein, we investigated whether two polymer materials, D3 and D7, induced an immune response, measured by their effects on a dendritic cell (DC) line, JAWS II. Using a lactate dehydrogenase cytotoxicity assay and Annexin V/PI staining, we found that the PU materials did not induce cytotoxicity in DC cells. Using confocal microscopy, we also showed that the materials did not induce activation or maturation, as compared to positive controls. This was confirmed by looking at various markers, CD80, CD86, MHC class I, and MHC class II, via flow cytometry. Overall, the results indicated that the investigated PU films are biocompatible in terms of immunotoxicology and immunogenicity and show great promise for use in regenerative medicine.


Subject(s)
Biocompatible Materials , Dendritic Cells/drug effects , Dendritic Cells/immunology , Materials Testing/methods , Polyurethanes/pharmacology , Animals , Bone Marrow Cells/drug effects , Cell Survival/drug effects , Ethers , Mice , Mice, Inbred C57BL , Nanostructures/toxicity , Regenerative Medicine , Tissue Engineering , Tissue Scaffolds
4.
Angiogenesis ; 21(3): 425-532, 2018 08.
Article in English | MEDLINE | ID: mdl-29766399

ABSTRACT

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Subject(s)
Biological Assay/methods , Neoplasms , Neovascularization, Pathologic , Animals , Biological Assay/instrumentation , Guidelines as Topic , Humans , Mice , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
5.
BMC Microbiol ; 18(1): 227, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30591021

ABSTRACT

BACKGROUND: Here we investigated the influence of different stabilization and storage strategies on the quality and composition of the fecal microbial community. Namely, same-day isolated murine DNA was compared to samples stored for 1 month in air at ambient temperature, with or without preservative buffers (i.e. EDTA and lysis buffer), different temperatures (i.e. 4 °C, - 20 °C, and - 80 °C), and hypoxic conditions. RESULTS: Only storage in lysis buffer significantly reduced DNA content, yet without integrity loss. Storage in EDTA affected alpha diversity the most, which was also reflected in cluster separation. Distinct changes were also seen in the phyla and bacterial species abundance per storage strategy. Metabolic function analysis showed 22 pathways not significantly affected by storage conditions, whereas the tyrosine metabolism pathway was significantly changed in all strategies except by EDTA. CONCLUSION: Each long-term storage strategy introduced a unique post-collection bias, which is important to take into account when interpreting data.


Subject(s)
Bacteria/isolation & purification , Microbiota , Preservation, Biological/methods , Animals , Bacteria/classification , Bacteria/genetics , Feces/microbiology , Mice , Specimen Handling , Temperature
6.
Int J Hyperthermia ; 34(1): 19-29, 2018 02.
Article in English | MEDLINE | ID: mdl-28540812

ABSTRACT

PURPOSE: To demonstrate delivery of Au nanocages to cells using the galectin-1 binding peptide anginex (Ax) and to demonstrate the value of this targeting for selective in vitro photothermal cell killing. MATERIALS AND METHODS: Au nanocages were synthesised, coated with polydopamine (PDA), and conjugated with Ax. Tumour and endothelial cell viability was measured with and without laser irradiation. Photoacoustic (PA) mapping and PA flow cytometry were used to confirm cell targeting in vitro and in tissue slices ex vivo. RESULTS: Cell viability was maintained at ≥50% at 100 pM suggesting low toxicity of the nanocage alone. Combining the targeted construct (25 pM) with low power 808 nm laser irradiation for 10-20 min (a duration previously shown to induce rapid and sustained heating of Au nanocages [AuNC] in solution), resulted in over 50% killing of endothelial and tumour cells. In contrast, the untargeted construct combined with laser irradiation resulted in negligible cell killing. We estimate approximately 6 × 104 peptides were conjugated to each nanocage, which also resulted in inhibition of cell migration. Binding of the targeted nanocage reached a plateau after three hours, and cell association was 20-fold higher than non-targeted nanocages both in vitro and ex vivo on tumour tissue slices. A threefold increase in tumour accumulation was observed in preliminary in vivo studies. CONCLUSIONS: These studies demonstrate Ax's potential as an effective targeting agent for Au-based theranostics to tumour and endothelial cells, enabling photothermal killing. This platform further suggests potential for multimodal in vivo therapy via next-generation drug-loaded nanocages.


Subject(s)
Galectin 1/metabolism , Gold/metabolism , Nanostructures/chemistry , Phototherapy/methods , Animals , Mice , Mice, Inbred BALB C
7.
Int J Mol Sci ; 19(3)2018 Mar 19.
Article in English | MEDLINE | ID: mdl-29562695

ABSTRACT

Galectins are a family of small, highly conserved, molecular effectors that mediate various biological processes, including chemotaxis and angiogenesis, and that function by interacting with various cell surface glycoconjugates, usually targeting ß-galactoside epitopes. Because of their significant involvement in various biological functions and pathologies, galectins have become a focus of therapeutic discovery for clinical intervention against cancer, among other pathological disorders. In this review, we focus on understanding galectin structure-function relationships, their mechanisms of action on the molecular level, and targeting them for therapeutic intervention against cancer.


Subject(s)
Galectins/metabolism , Molecular Targeted Therapy , Animals , Carbohydrates/chemistry , Drug Therapy, Combination , Galectins/chemistry , Humans
8.
Biochem Biophys Res Commun ; 492(3): 507-512, 2017 10 21.
Article in English | MEDLINE | ID: mdl-28822765

ABSTRACT

Noninvasive biological readouts of tumor metastatic risk and therapeutic efficacy are needed as healthcare costs rise. CTCs are the source of metastasis in distant organs that are responsible for the majority of cancer-related deaths. Here we demonstrate the acute and long-term effect of vascular disrupting therapies (high-dose radiotherapy and tumor necrosis factor-alpha (TNF)) on CTCs released from the primary tumor with a non-invasive real-time in vivo flow cytometry system. Using our innovative flow cytometry platform, we show here that radiation and nanodrug treatment can lead to short term release of CTC from the primary tumor. There was no increase in metastasis frequency or extent between control and TNF-treated mice; however, a significant reduction in lung metastasis was noted in the radiotherapy alone group. Mice treated with both TNF and radiotherapy had a slightly elevated metastatic profile between that of radiation alone and control (untreated) tumors. Possible mechanisms based on therapy specific vessel disruption and cell death are discussed. Overall, CTCs correlated with tumor progression and suggest CTC enumeration described herein may be useful in clinical management of solid tumor malignancies.


Subject(s)
Flow Cytometry , Gold/pharmacology , Nanoparticles/chemistry , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/radiation effects , Polyethylene Glycols/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Mice , Neoplastic Cells, Circulating/pathology , Time Factors
9.
Int J Mol Sci ; 18(12)2017 Dec 09.
Article in English | MEDLINE | ID: mdl-29232825

ABSTRACT

Galectin-1 is a hypoxia-regulated protein and a prognostic marker in head and neck squamous cell carcinomas (HNSCC). Here we assessed the ability of non-peptidic galectin-1 inhibitor OTX008 to improve tumor oxygenation levels via tumor vessel normalization as well as tumor growth inhibition in two human HNSCC tumor models, the human laryngeal squamous carcinoma SQ20B and the human epithelial type 2 HEp-2. Tumor-bearing mice were treated with OTX008, Anginex, or Avastin and oxygen levels were determined by fiber-optics and molecular marker pimonidazole binding. Immuno-fluorescence was used to determine vessel normalization status. Continued OTX008 treatment caused a transient reoxygenation in SQ20B tumors peaking on day 14, while a steady increase in tumor oxygenation was observed over 21 days in the HEp-2 model. A >50% decrease in immunohistochemical staining for tumor hypoxia verified the oxygenation data measured using a partial pressure of oxygen (pO2) probe. Additionally, OTX008 induced tumor vessel normalization as tumor pericyte coverage increased by approximately 40% without inducing any toxicity. Moreover, OTX008 inhibited tumor growth as effectively as Anginex and Avastin, except in the HEp-2 model where Avastin was found to suspend tumor growth. Galectin-1 inhibitor OTX008 transiently increased overall tumor oxygenation via vessel normalization to various degrees in both HNSCC models. These findings suggest that targeting galectin-1-e.g., by OTX008-may be an effective approach to treat cancer patients as stand-alone therapy or in combination with other standards of care.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Calixarenes/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Oxygen/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab/administration & dosage , Bevacizumab/pharmacology , Calixarenes/pharmacology , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Galectin 1/antagonists & inhibitors , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/metabolism , Humans , Mice , Peptides/administration & dosage , Peptides/pharmacology , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
10.
Biochim Biophys Acta ; 1830(6): 3454-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23403135

ABSTRACT

BACKGROUND: Dodecapeptide SC4 is a broad-spectrum bactericidal agent that functions by disintegrating bacterial membranes and neutralizing endotoxins. For insight into which SC4 amino acids are functionally important, we assessed Gram-negative bactericidal effects in structure-activity relationship experiments. Subsequently, SC4 was tested in a murine bacteremia model to combine and compare the efficacy with Zosyn, a first-line antibiotic against Pseudomonas aeruginosa (P. aeruginosa). METHODS: SC4 alanine-scanning analogs and their activities on were tested on P. aeruginosa. Survival studies in P. aeruginosa challenged mice were executed to monitor overall efficacy of SC4 and Zosyn, as a single modality and also as combination treatment. ELISAs were used to measure blood serum levels of selected inflammatory cytokines during treatment. RESULTS: Cationic residues were found to play a crucial role in terms of bactericidal activity against P. aeruginosa. In vivo, while only 9% (3/34) of control animals survived to day two and beyond, 44% (12/27) to 41% (14/34) of animals treated with SC4 or Zosyn, respectively, survived beyond one week. Combination treatment of SC4 and Zosyn demonstrated improved survival, i.e. 60% (12/20). The TNFα, IL-1, and IL-6 serum levels were attenuated in each treatment group compared to the control group. CONCLUSIONS: These data show that combination treatment of SC4 and Zosyn is most effective at killing P. aeruginosa and attenuating inflammatory cytokine levels in vivo. GENERAL SIGNIFICANCE: Combination treatment of SC4 and Zosyn may be useful in the clinic as a more effective antibiotic therapy against Gram-negative infectious diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteremia/drug therapy , Peptide Fragments/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa , Animals , Bacteremia/blood , Cytokines/blood , Disease Models, Animal , Inflammation Mediators/blood , Male , Mice , Penicillanic Acid/analogs & derivatives , Penicillanic Acid/pharmacology , Piperacillin/pharmacology , Piperacillin, Tazobactam Drug Combination , Pseudomonas Infections/blood , Time Factors
11.
Semin Radiat Oncol ; 34(3): 284-291, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38880537

ABSTRACT

The standard of care for radiation therapy is numerous, low-dose fractions that are distributed homogeneously throughout the tumor. An alternative strategy under scrutiny is to apply spatially fractionated radiotherapy (high and low doses throughout the tumor) in one or several fractions, either alone or followed by conventional radiation fractionation . Spatial fractionation allows for significant sparing of normal tissue, and the regions of tumor or normal tissue that received sublethal doses can give rise to beneficial bystander effects in both cases. Bystander effects are broadly defined as biological responses that are significantly greater than would be anticipated based on the radiation dose received. Typically these effects are initiated by diffusion of reactive oxygen species and secretion of various cytokines. As demonstrated in the literature, spatial fractionation related bystander effects can occur locally from cell to cell and in what are known as "cohort effects," which tend to take the form of restructuring of the vasculature, enhanced immune infiltration, and development of immunological memory. Other bystander effects can take place at distant sites in what are known as "abscopal effects." While these events are rare, they are mediated by the immune system and can result in the eradication of secondary and metastatic disease. Currently, due to the complexity and variability of these bystander effects, they are not thoroughly understood, but as knowledge improves they may present significant opportunities for improved clinical outcomes.


Subject(s)
Bystander Effect , Dose Fractionation, Radiation , Neoplasms , Bystander Effect/radiation effects , Humans , Neoplasms/radiotherapy , Reactive Oxygen Species/metabolism , Animals
12.
Radiat Res ; 201(2): 174-187, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38329819

ABSTRACT

Resistance to radiation remains a significant clinical challenge in non-small cell lung carcinoma (NSCLC). It is therefore important to identify the underlying molecular and cellular features that drive acquired resistance. We generated genetically matched NSCLC cell lines to investigate characteristics of acquired resistance. Murine Lewis lung carcinoma (LLC) and human A549 cells acquired an approximate 1.5-2.5-fold increase in radiation resistance as compared to their parental match, which each had unique intrinsic radio-sensitivities. The radiation resistance (RR) was reflected in higher levels of DNA damage and repair marker γH2AX and reduced apoptosis induction after radiation. Morphologically, we found that radiation resistance A549 (A549-RR) cells exhibited a greater nucleus-to-cytosol (N/C) ratio as compared to its parental counterpart. Since the N/C ratio is linked to the differentiation state, we next investigated the epithelial-to-mesenchymal transition (EMT) phenotype and cellular plasticity. We found that A549 cells had a greater radiation-induced plasticity, as measured by E-cadherin, vimentin and double-positive (DP) modulation, as compared to LLC. Additionally, migration was suppressed in A549-RR cells, as compared to A549 cells. Subsequently, we confirmed in vivo that the LLC-RR and A549-RR cells are also more resistance to radiation than their isogenic-matched counterpart. Moreover, we found that the acquired radiation resistance also induced resistance to cisplatin, but not carboplatin or oxaliplatin. This cross-resistance was attributed to induced elevation of thiol levels. Gamma-glutamylcysteine synthetase inhibitor buthionine sulfoximine (BSO) sensitized the resistant cells to cisplatin by decreasing the amount of thiols to levels prior to obtaining acquired radiation resistance. By generating radiation-resistance genetically matched NSCLC we were able to identify and overcome cisplatin cross-resistance. This is an important finding arguing for combinatorial treatment regimens including glutathione pathway disruptors in patients with the potential of improving clinical outcomes in the future.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Cisplatin/pharmacology , Cisplatin/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carboplatin , Buthionine Sulfoximine/pharmacology , Buthionine Sulfoximine/therapeutic use , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor
13.
J Pharmacol Exp Ther ; 344(3): 589-99, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23232447

ABSTRACT

Galectin-1 (gal-1), which binds ß-galactoside groups on various cell surface receptors, is crucial to cell adhesion and migration, and is found to be elevated in several cancers. Previously, we reported on 6DBF7, a dibenzofuran (DBF)-based peptidomimetic of the gal-1 antagonist anginex. In the present study, we used a structure-based approach to optimize 6DBF7. Initial NMR studies showed that 6DBF7 binds to gal-1 on one side of the ß-sandwich away from the lectin's carbohydrate binding site. Although an alanine scan of 6DBF7 showed that the two cationic groups (lysines) in the partial peptide are crucial to its angiostatic activity, it is the hydrophobic face of the amphipath that appears to interact directly with the surface of gal-1. Based on this structural information, we designed and tested additional DBF analogs. In particular, substitution of the C-terminal Asp for alanine and branched alkyl side chains (Val, Leu, Ile) for linear ones (Nle, Nva) rendered the greatest improvements in activity. Flow cytometry with gal-1(-/-) splenocytes showed that 6DBF7 and two of its more potent analogs (DB16 and DB21) can fully inhibit fluorescein isothiocyanate-gal-1 binding. Moreover, heteronuclear single-quantum coherence NMR titrations showed that the presence of DB16 decreases gal-1 affinity for lactose, indicating that the peptidomimetic targets gal-1 as a noncompetitive, allosteric inhibitor of glycan binding. Using tumor mouse models (B16F10 melanoma, LS174 lung, and MA148 ovarian), we found that DB21 inhibits tumor angiogenesis and tumor growth significantly better than 6DBF7, DB16, or anginex. DB21 is currently being developed further and holds promise for the management of human cancer in the clinic.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Benzofurans/chemistry , Benzofurans/pharmacology , Galectin 1/antagonists & inhibitors , Alanine/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Female , Galectin 1/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lactose/metabolism , Magnetic Resonance Spectroscopy/methods , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Protein Binding , Structure-Activity Relationship
14.
Invest New Drugs ; 31(5): 1142-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23392775

ABSTRACT

Previously, we reported on the anti-tumor activities of two designed calix[4]arene-based topomimetics (PTX008 and PTX009) of the amphipathic, angiostatic peptide Anginex. Here, we chemically modified the hydrophobic and hydrophilic faces of PTX008 and PTX009, and discovered new calixarene compounds that are more potent, cytotoxic anti-tumor agents. One of them, PTX013, is particularly effective at inhibiting the growth of several human cancer cell lines, as well as drug resistant cancer cells. Mechanistically, PTX013 induces cell cycle arrest in sub-G1 and G0/G1 phases of e.g. SQ20B cells, a radio-resistant human head and neck carcinoma model. In the syngeneic B16F10 melanoma tumor mouse model, PTX013 (0.5 mg/Kg) inhibits tumor growth by about 50-fold better than parent PTX008. A preliminary pharmacodynamics study strongly suggests that PTX013 exhibits good in vivo exposure and a relatively long half-life. Overall, this research contributes to the discovery of novel therapeutics as potentially useful agents against cancer in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Calixarenes/pharmacology , Cytotoxins/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Calixarenes/therapeutic use , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytotoxins/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Fibroblasts/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Tumor Burden/drug effects
15.
ACS Appl Mater Interfaces ; 15(50): 58241-58250, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38059477

ABSTRACT

Gold nanomaterials have been shown to augment radiation therapy both in vitro and in vivo. However, studies on these materials are mostly phenomenological due to nanoparticle heterogeneity and the complexity of biological systems. Even accurate quantification of the particle dose still results in bulk average biases; the effect on individual cells is not measured but rather the effect on the overall population. To perform quantitative nanobiology, we coated glass coverslips uniformly at varying densities with Au nanoparticle preparations with different morphologies (45 nm cages, 25 nm spheres, and 30 nm rods). Consequently, the effect of a specific number of particles per unit area in contact with breast cancer cells growing on the coated surfaces was ascertained. Gold nanocages showed the highest degree of radiosensitization on a per particle basis, followed by gold nanospheres and gold nanorods, respectively. All three materials showed little cytotoxic effect at 0 Gy, but clonogenic survival decreased proportionally with the radiation dose and particle coverage density. A similar trend was seen in vivo in the combined treatment antitumor response in 4T1 tumor-bearing animals. The presence of gold affected the type and quantity of reactive oxygen species generated, specifically superoxide and hydroxyl radicals, and the concentration of nanocages correlated with the development of more numerous double-stranded DNA breaks and increased protein oxidation as measured by carbonylation. This work demonstrates the dependence on morphology and concentration of radiation enhancement by gold nanomaterials and may lead to a novel method to differentiate intra- and extracellular functionalities of gold nanomedicine treatment strategies. It further provides insights that can guide the rational development of gold nanomaterial-based radiosensitizers for clinical use.


Subject(s)
Metal Nanoparticles , Nanostructures , Radiation-Sensitizing Agents , Animals , Gold/pharmacology , Gold/metabolism , Apoptosis , Radiation-Sensitizing Agents/pharmacology
16.
Int J Hyperthermia ; 27(1): 42-52, 2011.
Article in English | MEDLINE | ID: mdl-21204622

ABSTRACT

The purpose of this study was to delineate the mechanisms by which stromal components of cancer may induce tumour thermotolerance and exploit alterations in stromal and tumour physiology to enhance radiation therapy. The vascular thermoresponse was monitored by daily one-hour 41.5°C heatings in two murine solid tumour models, SCK murine mammary carcinoma and B16F10 melanoma. A transient increase was seen in overall tumour oxygenation for 2-3 days, followed by a progressive decline in tumour pO(2) upon continued daily heatings. Vascular thermotolerance was further studied by treating tumours with different heating strategies, i.e. (1) a single 60 min 41.5°C treatment; (2) two consecutive daily treatments of 41.5°C for 60 min; (3) a single 60 min 43°C treatment or (4) two days of 41.5°C for 60 min followed by treatment with 43°C for 60 min on the third day. Pre-heating tumours with mild temperature hyperthermia induced vascular thermotolerance, which was accompanied by evidence of vessel normalisation, i.e. a decrease in microvessel density and an increase in pericyte coverage. Rational scheduling of fractionated radiation during heat-induced increases in tumour oxygen levels rendered a significantly greater, synergistic, tumour growth inhibition. In vitro clonogenic survival responses of the individual cell types associated (endothelial cells, fibroblasts, pericytes and tumour cells) indicated only a direct cellular thermotolerance in endothelial cells. Overall, this suggests that tumour thermotolerance is a physiological phenomenon mediated through improvement of functional vasculature.


Subject(s)
Hyperthermia, Induced , Neoplasms/blood supply , Animals , Combined Modality Therapy , Female , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/physiopathology , Mammary Neoplasms, Experimental/radiotherapy , Mammary Neoplasms, Experimental/therapy , Mice , Neoplasms/physiopathology , Neoplasms/radiotherapy , Neoplasms/therapy , Oxygen/metabolism , Partial Pressure
17.
Biochem Biophys Rep ; 28: 101116, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34485713

ABSTRACT

BACKGROUND: Even though members of the family of adhesion/growth-regulatory galectins are increasingly detected to be co-expressed, they are still being routinely tested separately. The recent discovery of heterodimer formation among galectins-1, -3, and -7 in mixtures prompts further study of their functional activities in mixtures. METHODS: Cell agglutination, galectin binding to cells, as well as effects on cell proliferation, onset of apoptosis and migration were determined in assays using various cell types and mixtures of galectins-1, -3, and -7. RESULTS: Evidence for a more than additive increases of experimental parameters was consistently obtained. CONCLUSION: Testing galectins in mixtures simulates the situation of co-expression in situ and reveals unsuspected over-additive activities. This new insight is relevant for analyzing galectin functionality in (patho)physiological conditions.

18.
Neoplasia ; 23(1): 49-57, 2021 01.
Article in English | MEDLINE | ID: mdl-33220616

ABSTRACT

Fractionated radiation therapy is believed to reoxygenate and subsequently radiosensitize surviving hypoxic cancer cells. Measuring tumor reoxygenation between radiation fractions could conceivably provide an early biomarker of treatment response. However, the relationship between tumor reoxygenation and local control is not well understood. We used noninvasive optical fiber-based diffuse reflectance spectroscopy to monitor radiation-induced changes in hemoglobin oxygen saturation (sO2) in tumor xenografts grown from two head and neck squamous cell carcinoma cell lines - UM-SCC-22B and UM-SCC-47. Tumors were treated with 4 doses of 2 Gy over 2 consecutive weeks and diffuse reflectance spectra were acquired every day during the 2-week period. There was a statistically significant increase in sO2 in the treatment-responsive UM-SCC-22B tumors immediately following radiation. This reoxygenation trend was due to an increase in oxygenated hemoglobin (HbO2) and disappeared over the next 48 h as sO2 returned to preradiation baseline values. Conversely, sO2 in the relatively radiation-resistant UM-SCC-47 tumors increased after every dose of radiation and was driven by a significant decrease in deoxygenated hemoglobin (dHb). Immunohistochemical analysis revealed significantly elevated expression of hypoxia-inducible factor (HIF-1) in the UM-SCC-47 tumors prior to radiation and up to 48 h postradiation compared with the UM-SCC-22B tumors. Our observation of a decrease in dHb, a corresponding increase in sO2, as well as greater HIF-1α expression only in UM-SCC-47 tumors strongly suggests that the reoxygenation within these tumors is due to a decrease in oxygen consumption in the cancer cells, which could potentially play a role in promoting radiation resistance.


Subject(s)
Oxidation-Reduction/radiation effects , Oxygen Consumption/radiation effects , Oxygen/analysis , Oxygen/metabolism , Radiation Tolerance , Radiation , Spectrum Analysis , Animals , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Humans , Immunohistochemistry , Mice , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/radiotherapy , Optical Imaging , Radiotherapy , Spectrum Analysis/methods , Xenograft Model Antitumor Assays
19.
PLoS One ; 16(8): e0241882, 2021.
Article in English | MEDLINE | ID: mdl-34460818

ABSTRACT

Gold nanoparticles (AuNPs) hold great promise in nanomedicine, yet their successful clinical translation has not been realized. Some challenges include effective AuNP targeting and delivery to improve modulation of immune cells of interest while limiting potential adverse effects. In order to overcome these challenges, we must fully understand how AuNPs impact different immune cell subsets, particularly within the dendritic cell and T cell compartments. Herein, we show that polyethylene glycol coated (PEG) gold nanorods (AuNRs) and PEG AuNRs covered with a thin layer of silver (AuNR/Ag) may enhance the immune response towards immune suppression or activation. We also studied the ability to enhance CD4+ Foxp3+ Tregs in vitro using AuNRs functionalized with interleukin 2 (IL2), a cytokine that is important in Treg development and homeostasis. Our results indicate that AuNRs enhance different immune cells and that NP composition matters in immune targeting. This knowledge will help us understand how to better design AuNRs to target and enhance the immune system.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Forkhead Transcription Factors/metabolism , Gold/pharmacology , Metal Nanoparticles/administration & dosage , Nanotubes/chemistry , Animals , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Immune System/drug effects , Immune System/metabolism , Immunity/drug effects , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Polyethylene Glycols/chemistry , Silver/administration & dosage
20.
Bioconjug Chem ; 21(1): 20-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20020769

ABSTRACT

Targeted delivery of therapeutic drugs promises to become the norm to treat cancer. Here, we conjugated the cytotoxic agent 6-hydroxypropylacylfulvene (HPAF) to anginex, a peptide that targets galectin-1, which is highly expressed in endothelial cells of tumor vessels. In a human ovarian cancer model in mice, the conjugate inhibited tumor growth better than equivalent doses of either compound alone. Immunofluorescence on tumor tissue demonstrated that the conjugate, like parent anginex, selectively targeted tumor vasculature and inhibited tumor angiogenesis. Increased activity from the conjugate further suggests that HPAF retains at least some of its normal cytotoxic activity when linked to anginex. More importantly perhaps is the observation that the conjugate abrogates apparent systemic toxicity from treatment with HPAF. This work contributes to the development of tumor vascular targeting agents against cancer in the clinic.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Galectin 1/antagonists & inhibitors , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Proteins/chemistry , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Galectin 1/metabolism , Humans , Neoplasms/drug therapy , Peptides , Proteins/pharmacology , Proteins/therapeutic use , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Tumor Cells, Cultured , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL