Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Transl Med ; 22(1): 621, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961395

ABSTRACT

BACKGROUND: The tumor microenvironment is profoundly heterogeneous particularly when comparing sites of metastases. Establishing the extent of this heterogeneity may provide guidance on how best to design lipid-based drug delivery systems to treat metastatic disease. Building on our previous research, the current study employs a murine model of metastatic cancer to explore the distribution of ~ 100 nm liposomes. METHODS: Female NCr nude mice were inoculated with a fluorescently labeled, Her2/neu-positive, trastuzumab-resistant breast cancer cell line, JIMT-1mkate, either in the mammary fat pad to create an orthotopic tumor (OT), or via intracardiac injection (IC) to establish tumors throughout the body. Animals were dosed with fluorescent and radio-labeled liposomes. In vivo and ex vivo fluorescent imaging was used to track liposome distribution over a period of 48 h. Liposome distribution in orthotopic tumors was compared to sites of tumor growth that arose following IC injection. RESULTS: A significant amount of inter-vessel heterogeneity for DiR distribution was observed, with most tumor blood vessels showing little to no presence of the DiR-labelled liposomes. Further, there was limited extravascular distribution of DiR liposomes in the perivascular regions around DiR-positive vessels. While all OT tumors contained at least some DiR-positive vessels, many metastases had very little or none. Despite the apparent limited distribution of liposomes within metastases, two liposomal drug formulations, Irinophore C and Doxil, showed similar efficacy for both the OT and IC JIMT-1mkate models. CONCLUSION: These findings suggest that liposomal formulations achieve therapeutic benefits through mechanisms that extend beyond the enhanced permeability and retention effect.


Subject(s)
Antineoplastic Agents , Liposomes , Mice, Nude , Neoplasm Metastasis , Animals , Cell Line, Tumor , Female , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Humans , Treatment Outcome , Mice
2.
Anticancer Res ; 44(7): 2793-2803, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925821

ABSTRACT

BACKGROUND/AIM: The aim of this study was to develop an enhanced intestinal toxicity assay with three outputs assessing proliferation, villi morphology and DNA damage after irradiation. MATERIALS AND METHODS: Whole 5 cm jejunal lengths were collected from mice following total body x-ray irradiation (0-15 Gy) at 0-84 h. Tissues were wrapped into swirls for cryopreservation and immunohistochemically stained for EdU, CD31, and γH2AX. A semi-automated image analysis was developed for the proliferation, villi morphology, and DNA damage models. RESULTS: Proliferation assessed via EdU staining varied with cycles of damage repair, hyperproliferation, and homeostasis after radiation, with the time to onset of each cycle variable based on radiation dose. An analysis model evaluating the amount of proliferation per unit length of jejunum analyzed was developed, with a dose-response curve identified at 48 h post treatment. The villi length model measured the length of intact and healthy CD31-stained capillary beds between the crypts and villi tips at 3.5 days post treatment within a 0-10 Gy dose range. The DNA damage model evaluated the intensity of γH2AX staining within cellular nuclei, with a useful dose-response identified at 1 h post-radiation treatment. CONCLUSION: This assay demonstrates flexibility for assessing radiation-induced damage, with analysis of proliferation, villi length, or direct DNA damage achievable at defined time points and within useful radiation dose curves. The software-assisted image analysis allows for rapid, comprehensive, and objective data generation with an assay turnover time of days instead of weeks on samples that are representative of most of the treated jejunum.


Subject(s)
Cell Proliferation , DNA Damage , Animals , Mice , Cell Proliferation/radiation effects , DNA Damage/radiation effects , Jejunum/radiation effects , Jejunum/pathology , Radiation Tolerance , Intestinal Mucosa/radiation effects , Intestinal Mucosa/pathology , Intestines/radiation effects , Intestines/pathology , Whole-Body Irradiation/adverse effects , Dose-Response Relationship, Radiation , Histones/metabolism , Male , Mice, Inbred C57BL
3.
Radiother Oncol ; 196: 110326, 2024 07.
Article in English | MEDLINE | ID: mdl-38735536

ABSTRACT

PURPOSE: The oxygen depletion hypothesis has been proposed as a rationale to explain the observed phenomenon of FLASH-radiotherapy (FLASH-RT) sparing normal tissues while simultaneously maintaining tumor control. In this study we examined the distribution of DNA Damage Response (DDR) markers in irradiated 3D multicellular spheroids to explore the relationship between FLASH-RT protection and radiolytic-oxygen-consumption (ROC) in tissues. METHODS: Studies were performed using a Varian Truebeam linear accelerator delivering 10 MeV electrons with an average dose rate above 50 Gy/s. Irradiations were carried out on 3D spheroids maintained under a range of O2 and temperature conditions to control O2 consumption and create gradients representative of in vivo tissues. RESULTS: Staining for pDNA-PK (Ser2056) produced a linear radiation dose response whereas γH2AX (Ser139) showed saturation with increasing dose. Using the pDNA-PK staining, radiation response was then characterised for FLASH compared to standard-dose-rates as a function of depth into the spheroids. At 4 °C, chosen to minimize the development of metabolic oxygen gradients within the tissues, FLASH protection could be observed at all distances under oxygen conditions of 0.3-1 % O2. Whereas at 37 °C a FLASH-protective effect was limited to the outer cell layers of tissues, an effect only observed at 3 % O2. Modelling of changes in the pDNA-PK-based oxygen enhancement ratio (OER) yielded a tissue ROC g0-value estimate of 0.73 ± 0.25 µM/Gy with a km of 5.4 µM at FLASH dose rates. CONCLUSIONS: DNA damage response markers are sensitive to the effects of transient oxygen depletion during FLASH radiotherapy. Findings support the rationale that well-oxygenated tissues would benefit more from FLASH-dose-rate protection relative to poorly-oxygenated tissues.


Subject(s)
DNA Damage , Spheroids, Cellular , DNA Damage/radiation effects , Humans , Spheroids, Cellular/radiation effects , Histones/metabolism , Histones/analysis , Oxygen Consumption/radiation effects , Dose-Response Relationship, Radiation , Organ Sparing Treatments/methods
4.
DNA Repair (Amst) ; 139: 103689, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749239

ABSTRACT

The effectiveness of radiotherapy depends on the sensitivities of 'normal' and cancer cells to the administered radiation dose. Increasing the radiosensitivity of cancers by inhibiting DNA damage repair is a goal of much current research, however success depends on avoiding concomitant sensitization of normal tissues inevitably irradiated during therapy. In this study we investigated the mechanisms of radiosensitization for DNA-PK and PARP inhibitors by examining the impacts on proliferating vs quiescent cell populations. Experiments were performed in BRCA1/2null and wild-type parental cancer models in vitro and in vivo. Overall AZD7648 has greater radiosensitizing activity relative to Olaparib, with BRCA2-deficient models showing the greatest sensitivity. However, DNA-PK inhibitor AZD7648 also produced greater toxicity in all irradiated mice. While both DNA-PK and PARP inhibition sensitizes wild type tumor cells to radiation, in BRCA1/2 deficient cells PARP inhibition by Olaparib had limited radiosensitization capacity. Quiescent cells are more radioresistant than proliferating cells, and these were also effectively sensitized by AZD7648 while Olaparib was unable to increase radiation-induced cell kill, even in BRCA1/2null cells. These findings underscore the distinct mechanisms of radiosensitization for DNA-PK and PARP inhibitors. While DNA-PK inhibitors are able to target both proliferating and non-proliferating tumor cells for greater overall anti-cancer benefit, their application is limited by exacerbation of normal tissue toxicities. Conversely, PARP inhibitors exhibit selective activity for proliferating cells, providing a mechanism for targeting activity to cancers, but due to poor activity in non-proliferating cells they have an overall reduced impact on tumor growth control. This study highlights the importance of creating a therapeutic ratio with DNA damage repair inhibition radiation sensitizing strategies.


Subject(s)
BRCA1 Protein , BRCA2 Protein , DNA-Activated Protein Kinase , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors , Radiation-Sensitizing Agents , Phthalazines/pharmacology , Piperazines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , Humans , BRCA1 Protein/metabolism , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Mice , Cell Line, Tumor , Female , BRCA2 Protein/genetics , Cell Proliferation/drug effects , Radiation Tolerance/drug effects , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 23(9): 1230-1240, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-38781104

ABSTRACT

Inhibitors of DNA-dependent protein kinase (PRKDC; DNA-PK) sensitize cancers to radiotherapy and DNA-damaging chemotherapies, with candidates in clinical trials. However, the degree to which DNA-PK inhibitors also sensitize normal tissues remains poorly characterized. In this study, we compare tumor growth control and normal tissue sensitization following DNA-PK inhibitors in combination with radiation and etoposide. FaDu tumor xenografts implanted in mice were treated with 10 to 15 Gy irradiation ± 3 to 100 mg/kg AZD7648. A dose-dependent increase in time to tumor volume doubling following AZD7648 was proportional to an increase in toxicity scores of the overlying skin. Similar effects were seen in the intestinal jejunum, tongue, and FaDu tumor xenografts of mice assessed for proliferation rates at 3.5 days after treatment with etoposide or 5 Gy whole body irradiation ± DNA-PK inhibitors AZD7648 or peposertib (M3814). Additional organs were examined for sensitivity to DNA-PK inhibitor activity in ATM-deficient mice, where DNA-PK activity is indicated by surrogate marker γH2AX. Inhibition was observed in the heart, brain, pancreas, thymus, tongue, and salivary glands of ATM-deficient mice treated with the DNA-PK inhibitors relative to radiation alone. Similar reductions are also seen in ATM-deficient FaDu tumor xenografts where both pDNA-PK and γH2AX staining could be performed. DNA-PK inhibitor-mediated sensitization to radiation and DNA-damaging chemotherapy are not only limited to tumor tissues, but also extends to normal tissues sustaining DNA damage. These data are useful for interpretation of the sensitizing effects of DNA damage repair inhibitors, where a therapeutic index showing greater cell-killing effects on cancer cells is crucial for optimal clinical translation.


Subject(s)
DNA-Activated Protein Kinase , Xenograft Model Antitumor Assays , Animals , DNA-Activated Protein Kinase/antagonists & inhibitors , Humans , Mice , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Etoposide/pharmacology , Radiation-Sensitizing Agents/pharmacology , DNA Damage/drug effects , Cell Proliferation/drug effects , Tumor Burden/drug effects , Tumor Burden/radiation effects
6.
Sci Rep ; 13(1): 12429, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37528151

ABSTRACT

Type II topoisomerase (Top2) poisoning therapy is used to treat a broad range of cancers via induction of double strand breaks (DSBs) in cells undergoing replication and transcription. Preventing the repair of DSBs via inhibition of DNA-PK, an inhibitor of non-homologous end-joining (NHEJ), increases cell kill with Top2 poisons and has led to the initiation of several clinical trials. To elucidate the cellular mechanisms leading to synergistic activity of dual DNA-PK/Top2 inhibition we looked at their effects in cycling versus non-cycling cells, in 3D spheroids and in xenograft models. Combined DNA-PK/Top2 inhibition was found to not only increase the cell kill in proliferating cells, the cell population that is typically most vulnerable to Top2 poisoning, but also in non-proliferative but transcriptionally active cells. This effect was observed in both cancer and normal tissue models, killing more cells than high concentrations of etoposide alone. The combination treatment delayed tumor growth in mice compared to Top2 poisoning alone, but also led to increased toxicity. These findings demonstrate sensitization of Top2ß-expressing, non-cycling cells to Top2 poisoning by DNA-PK inhibition. Expansion of the target cell population of Top2 poison treatment to include non-proliferating cells via combination with DNA damage repair inhibitors has implications for efficacy and toxicity of these combinations, including for inhibitors of DNA-PK currently in clinical trial.


Subject(s)
DNA-Binding Proteins , Neoplasms , Humans , Animals , Mice , DNA-Binding Proteins/genetics , DNA Topoisomerases, Type II/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Etoposide/pharmacology , Isomerases/genetics , DNA-Activated Protein Kinase/genetics , Neoplasms/drug therapy , DNA , Topoisomerase II Inhibitors/pharmacology
7.
Radiother Oncol ; 187: 109795, 2023 10.
Article in English | MEDLINE | ID: mdl-37414252

ABSTRACT

PURPOSE: There is a significant need for a widely available, translatable, sensitive and non-invasive imaging biomarker for tumor hypoxia in radiation oncology. Treatment-induced changes in tumor tissue oxygenation can alter the sensitivity of cancer tissues to radiation, but the relative difficulty in monitoring the tumor microenvironment results in scarce clinical and research data. Oxygen-Enhanced MRI (OE-MRI) uses inhaled oxygen as a contrast agent to measure tissue oxygenation. Here we investigate the utility of dOE-MRI, a previously validated imaging approach employing a cycling gas challenge and independent component analysis (ICA), to detect VEGF-ablation treatment-induced changes in tumor oxygenation that result in radiosensitization. METHODS: Murine squamous cell carcinoma (SCCVII) tumor-bearing mice were treated with 5 mg/kg anti-VEGF murine antibody B20 (B20-4.1.1, Genentech) 2-7 days prior to radiation treatment, tissue collection or MR imaging using a 7 T scanner. dOE-MRI scans were acquired for a total of three repeated cycles of air (2 min) and 100% oxygen (2 min) with responding voxels indicating tissue oxygenation. DCE-MRI scans were acquired using a high molecular weight (MW) contrast agent (Gd-DOTA based hyperbranched polygylcerol; HPG-GdF, 500 kDa) to obtain fractional plasma volume (fPV) and apparent permeability-surface area product (aPS) parameters derived from the MR concentration-time curves. Changes to the tumor microenvironment were evaluated histologically, with cryosections stained and imaged for hypoxia, DNA damage, vasculature and perfusion. Radiosensitizing effects of B20-mediated increases in oxygenation were evaluated by clonogenic survival assays and by staining for DNA damage marker γH2AX. RESULTS: Tumors from mice treated with B20 exhibit changes to their vasculature that are consistent with a vascular normalization response, and result in a temporary period of reduced hypoxia. DCE-MRI using injectable contrast agent HPG-GDF measured decreased vessel permeability in treated tumors, while dOE-MRI using inhaled oxygen as a contrast agent showed greater tissue oxygenation. These treatment-induced changes to the tumor microenvironment result in significantly increased radiation sensitivity, illustrating the utility of dOE-MRI as a non-invasive biomarker of treatment response and tumor sensitivity during cancer interventions. CONCLUSIONS: VEGF-ablation therapy-mediated changes to tumor vascular function measurable using DCE-MRI techniques may be monitored using the less invasive approach of dOE-MRI, an effective biomarker of tissue oxygenation that can monitor treatment response and predict radiation sensitivity.


Subject(s)
Neoplasms , Oxygen , Mice , Animals , Oxygen/metabolism , Contrast Media , Magnetic Resonance Imaging/methods , Hypoxia , Biomarkers , Tumor Microenvironment
8.
Int J Mol Sci ; 22(12)2021 Jun 13.
Article in English | MEDLINE | ID: mdl-34199298

ABSTRACT

BACKGROUND: Preclinical drug development studies rarely consider the impact of a candidate drug on established metastatic disease. This may explain why agents that are successful in subcutaneous and even orthotopic preclinical models often fail to demonstrate efficacy in clinical trials. It is reasonable to anticipate that sites of metastasis will be phenotypically unique, as each tumor will have evolved heterogeneously with respect to gene expression as well as the associated phenotypic outcome of that expression. The objective for the studies described here was to gain an understanding of the tumor heterogeneity that exists in established metastatic disease and use this information to define a preclinical model that is more predictive of treatment outcome when testing novel drug candidates clinically. METHODS: Female NCr nude mice were inoculated with fluorescent (mKate), Her2/neu-positive human breast cancer cells (JIMT-mKate), either in the mammary fat pad (orthotopic; OT) to replicate a primary tumor, or directly into the left ventricle (intracardiac; IC), where cells eventually localize in multiple sites to create a model of established metastasis. Tumor development was monitored by in vivo fluorescence imaging (IVFI). Subsequently, animals were sacrificed, and tumor tissues were isolated and imaged ex vivo. Tumors within organ tissues were further analyzed via multiplex immunohistochemistry (mIHC) for Her2/neu expression, blood vessels (CD31), as well as a nuclear marker (Hoechst) and fluorescence (mKate) expressed by the tumor cells. RESULTS: Following IC injection, JIMT-1mKate cells consistently formed tumors in the lung, liver, brain, kidney, ovaries, and adrenal glands. Disseminated tumors were highly variable when assessing vessel density (CD31) and tumor marker expression (mkate, Her2/neu). Interestingly, tumors which developed within an organ did not adopt a vessel microarchitecture that mimicked the organ where growth occurred, nor did the vessel microarchitecture appear comparable to the primary tumor. Rather, metastatic lesions showed considerable variability, suggesting that each secondary tumor is a distinct disease entity from a microenvironmental perspective. CONCLUSIONS: The data indicate that more phenotypic heterogeneity in the tumor microenvironment exists in models of metastatic disease than has been previously appreciated, and this heterogeneity may better reflect the metastatic cancer in patients typically enrolled in early-stage Phase I/II clinical trials. Similar to the suggestion of others in the past, the use of models of established metastasis preclinically should be required as part of the anticancer drug candidate development process, and this may be particularly important for targeted therapeutics and/or nanotherapeutics.


Subject(s)
Biomarkers, Tumor/metabolism , Tumor Microenvironment , Animals , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Lung/pathology , Mice, Nude , Neoplasm Metastasis
9.
J Appl Clin Med Phys ; 22(6): 50-59, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34028969

ABSTRACT

PURPOSE: To assess dosimetric properties and identify required updates to commonly used protocols (including use of film and ionization chamber) pertaining to a clinical linac configured into FLASH (ultra-high dose rate) electron mode. METHODS: An 18MV photon beam of a Varian iX linac was converted to FLASH electron beam by replacing the target and the flattening filter with an electron scattering foil. The dose was prescribed by entering the MUs through the console. Fundamental beam properties, including energy, dose rate, dose reproducibility, field size, and dose rate dependence on the SAD, were examined in preparation for radiobiological experiments. Gafchromic EBT-XD film was evaluated for usability in measurements at ultra-high dose rates by comparing the measured dose to the inverse square model. Selected previously reported models of chamber efficiencies were fitted to measurements in a broad range of dose rates. RESULTS: The performance of the modified linac was found adequate for FLASH radiobiological experiments. With exception of the increase in the dose per MU on increase in the repetition rate, all fundamental beam properties proved to be in line with expectations developed with conventional linacs. The field size followed the theorem of similar triangles. The highest average dose rate (2 × 104  Gy/s) was found next to the internal monitor chamber, with the field size of FWHM = 1.5 cm. Independence of the dose readings on the dose rate (up to 2 × 104  Gy/s) was demonstrated for the EBT-XD film. A model of recombination in an ionization chamber was identified that provided good agreement with the measured chamber efficiencies for the average dose rates up to at least 2 × 103  Gy/s. CONCLUSION: Dosimetric measurements were performed to characterize a linac converted to FLASH dose rates. Gafchromic EBT-XD film and dose rate-corrected cc13 ionization chamber were demonstrated usable at FLASH dose rates.


Subject(s)
Electrons , Particle Accelerators , Film Dosimetry , Humans , Radiometry , Reproducibility of Results
10.
Cancer Immunol Res ; 7(7): 1064-1078, 2019 07.
Article in English | MEDLINE | ID: mdl-31088846

ABSTRACT

Treatment strategies involving immune-checkpoint blockade (ICB) have significantly improved survival for a subset of patients across a broad spectrum of advanced solid cancers. Despite this, considerable room for improving response rates remains. The tumor microenvironment (TME) is a hurdle to immune function, as the altered metabolism-related acidic microenvironment of solid tumors decreases immune activity. Here, we determined that expression of the hypoxia-induced, cell-surface pH regulatory enzyme carbonic anhydrase IX (CAIX) is associated with worse overall survival in a cohort of 449 patients with melanoma. We found that targeting CAIX with the small-molecule SLC-0111 reduced glycolytic metabolism of tumor cells and extracellular acidification, resulting in increased immune cell killing. SLC-0111 treatment in combination with immune-checkpoint inhibitors led to the sensitization of tumors to ICB, which led to an enhanced Th1 response, decreased tumor growth, and reduced metastasis. We identified that increased expression of CA9 is associated with a reduced Th1 response in metastatic melanoma and basal-like breast cancer TCGA cohorts. These data suggest that targeting CAIX in the TME in combination with ICB is a potential therapeutic strategy for enhancing response and survival in patients with hypoxic solid malignancies.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Carbonic Anhydrases/chemistry , Hypoxia/physiopathology , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Phenylurea Compounds/pharmacology , Sulfonamides/pharmacology , Animals , Apoptosis , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , CTLA-4 Antigen/antagonists & inhibitors , Carbonic Anhydrases/metabolism , Cell Proliferation , Drug Therapy, Combination , Enzyme Induction , Female , Gene Expression Regulation, Enzymologic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Melanoma/enzymology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Survival Rate , Tumor Cells, Cultured , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
11.
Cell Rep ; 27(6): 1769-1780.e4, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31067462

ABSTRACT

The sterile alpha motif (SAM) and SRC homology 3 (SH3) domain containing protein 1 (Sash1) acts as a scaffold in TLR4 signaling. We generated Sash1-/- mice, which die in the perinatal period due to respiratory distress. Constitutive or endothelial-restricted Sash1 loss leads to a delay in maturation of alveolar epithelial cells causing reduced surfactant-associated protein synthesis. We show that Sash1 interacts with ß-arrestin 1 downstream of the TLR4 pathway to activate Akt and endothelial nitric oxide synthase (eNOS) in microvascular endothelial cells. Generation of nitric oxide downstream of Sash1 in endothelial cells affects alveolar epithelial cells in a cGMP-dependent manner, inducing maturation of alveolar type 1 and 2 cells. Thus, we identify a critical cell nonautonomous function for Sash1 in embryonic development in which endothelial Sash1 regulates alveolar epithelial cell maturation and promotes pulmonary surfactant production through nitric oxide signaling. Lung immaturity is a major cause of respiratory distress and mortality in preterm infants, and these findings identify the endothelium as a potential target for therapy.


Subject(s)
Endothelial Cells/metabolism , Lung/growth & development , Nitric Oxide/metabolism , Signal Transduction , Animals , Animals, Newborn , Cell Line , Cyclic GMP/metabolism , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endothelium/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Lung/ultrastructure , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Alveoli/pathology , Pulmonary Surfactant-Associated Proteins/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , beta-Arrestins/metabolism
12.
Radiat Res ; 191(4): 380-381, 2019 04.
Article in English | MEDLINE | ID: mdl-30958759
13.
Magn Reson Med ; 81(4): 2514-2525, 2019 04.
Article in English | MEDLINE | ID: mdl-30368892

ABSTRACT

PURPOSE: There is a critical need for non-invasive imaging biomarkers of tumor oxygenation to assist in patient stratification and development of hypoxia targeting therapies. Using a cycling gas challenge and independent component analysis (ICA), we sought to improve the sensitivity and speed of existing oxygen enhanced MRI (OE-MRI) techniques to detect changes in oxygenation with dynamically acquired T1 W signal intensity images (dOE-MRI). METHODS: Mice were implanted with SCCVII, HCT-116, BT-474, or SKOV3 tumors in the dorsal subcutaneous region and imaged at 7T. T1 W images were acquired during a respiratory challenge with alternating 2-minute periods of air and 100% oxygen for three cycles. Data were analyzed with ICA and oxygenation maps were generated and compared to corresponding histology sections stained for hypoxia (pimonidazole) and blood vessels (CD31). RESULTS: Cycling air-oxygen-air gas challenges were well tolerated and ICA permitted extraction of the oxygen-enhancing component in all imaged tumors from four different models. Comparison with synthetic response functions showed that dOE-MRI does not require any a-priori knowledge of the physiological response. The fraction of O2 -negative dOE-MRI voxels that correlate inversely with the ICA gas-cycling component correspond well with the histological hypoxic fraction in SCCVII tumors (r = 0.91, p = 0.0016) but did not correlate in HCT-116 tumors (r = 0.13, p = 0.81). CONCLUSIONS: Using ICA and adding a cycling gas challenge extends the sensitivity of OE-MRI and allows the oxygenation status of tumors to be assessed in as little as six minutes. These findings support further development of OE-MRI as a biomarker of tumor oxygenation.


Subject(s)
Biomarkers/metabolism , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Neoplasms/diagnostic imaging , Oxygen/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Hypoxia , Cell Line, Tumor , Female , HCT116 Cells , Humans , Mice , Neoplasm Transplantation , Nitroimidazoles/therapeutic use , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Tumor Microenvironment
14.
Clin Exp Metastasis ; 35(7): 691-705, 2018 10.
Article in English | MEDLINE | ID: mdl-30196384

ABSTRACT

Most HER2-positive metastatic breast cancer patients continue to relapse. Incomplete access to all target HER2-positive cells in metastases and tumor tissues is a potential mechanism of resistance to trastuzumab. The location of locally bound trastuzumab was evaluated in HER2-positive tissues in vivo and as in vivo xenografts or metastases models in mice. Microenvironmental elements of tumors were related to bound trastuzumab using immunohistochemical staining and include tight junctions, vasculature, vascular maturity, vessel patency, hypoxia and HER2 to look for correlations. Trastuzumab was evaluated alone and in combination with bevacizumab. Dynamic contrast-enhanced magnetic resonance imaging parameters of overall vascular function, perfusion and apparent permeability were compared with matched histological images of trastuzumab distribution and vascular patency. Trastuzumab distribution is highly heterogeneous in all models examined, including avascular micrometastases of the brain and lung. Trastuzumab distributes well through the extravascular compartment even in conditions of high HER2 expression and poor convective flow in vivo. Microregional patterns of trastuzumab distribution in vivo do not consistently correlate with vascular density, patency, function or maturity; areas of poor trastuzumab access are not necessarily those with poor vascular supply. The number of vessels with perivascular trastuzumab increases with time and higher doses and dramatically decreases when pre-treated with bevacizumab. Areas of HER2-positive tissue without bound trastuzumab persist in all conditions. These data directly demonstrate tissue- and vessel-level barriers to trastuzumab distribution in vivo that can effectively limit access of the drug to target cells in brain metastases and elsewhere.


Subject(s)
Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacokinetics , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bevacizumab/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Brain Neoplasms/secondary , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Receptor, ErbB-2/biosynthesis , Trastuzumab/administration & dosage , Tumor Microenvironment , Xenograft Model Antitumor Assays
15.
Oncotarget ; 7(41): 66970-66988, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27556700

ABSTRACT

Autophagy, a lysosome-mediated degradation and recycling process, functions in advanced malignancies to promote cancer cell survival and contribute to cancer progression and drug resistance. While various autophagy inhibition strategies are under investigation for cancer treatment, corresponding patient selection criteria for these autophagy inhibitors need to be developed. Due to its central roles in the autophagy process, the cysteine protease ATG4B is one of the autophagy proteins being pursued as a potential therapeutic target. In this study, we investigated the expression of ATG4B in breast cancer, a heterogeneous disease comprised of several molecular subtypes. We examined a panel of breast cancer cell lines, xenograft tumors, and breast cancer patient specimens for the protein expression of ATG4B, and found a positive association between HER2 and ATG4B protein expression. We showed that HER2-positive cells, but not HER2-negative breast cancer cells, require ATG4B to survive under stress. In HER2-positive cells, cytoprotective autophagy was dependent on ATG4B under both starvation and HER2 inhibition conditions. Combined knockdown of ATG4B and HER2 by siRNA resulted in a significant decrease in cell viability, and the combination of ATG4B knockdown with trastuzumab resulted in a greater reduction in cell viability compared to trastuzumab treatment alone, in both trastuzumab-sensitive and -resistant HER2 overexpressing breast cancer cells. Together these results demonstrate a novel association of ATG4B positive expression with HER2 positive breast cancers and indicate that this subtype is suitable for emerging ATG4B inhibition strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy-Related Proteins/biosynthesis , Autophagy/physiology , Breast Neoplasms/metabolism , Cysteine Endopeptidases/biosynthesis , Drug Resistance, Neoplasm/drug effects , Adult , Aged , Animals , Autophagy/drug effects , Drug Resistance, Neoplasm/physiology , Female , Heterografts , Humans , Mice , Middle Aged , Receptor, ErbB-2/biosynthesis , Trastuzumab/pharmacology , Xenograft Model Antitumor Assays
16.
Clin Epigenetics ; 8: 53, 2016.
Article in English | MEDLINE | ID: mdl-27175220

ABSTRACT

BACKGROUND: The tumor-specific microregional effects of the anticancer agent RRx-001, a novel epigenetic-based radio/chemosensitizer with nitrogen oxide-donating properties in phase II clinical trials, were investigated with whole tissue section quantitative immunohistological staining in mouse SCCVII and human U87 tumors. RESULTS: SCCVII tumors exhibited regions of intermittent perfusion exemplified by co-localization of vessels with the hypoxia marker pimonidazole commonly occurring throughout the tissue. A moderate increase in perfusion (21 to 28 %) was observed after a bolus dose of the perivascular stain DiOC7(3), however, with the absence of an increase in tissue oxygenation. U87 tumors showed an absence of blood flow over large areas of treated tumors after dosing with RRx-001. However, these areas did not become necrotic and returned to near normal levels after 12 h. No significant change in tumor hypoxia was seen at 90 min or 12 h. For both tumor types, RRx-001 treatment resulted in the loss of perfusion in the large regions of the tumor; however, at the 12-h time point, both tumor types showed an increase in vessel perfusion but no significant decrease in hypoxia. CONCLUSIONS: These data suggest a redistribution of blood flow within the tumor for both tumor types akin to vascular normalization. Differences between the tumors were related to tumor architecture and distribution of alpha-smooth muscle actin (α-SMA). RRx-001 shows promise for short-term blood flow redistribution in tumors with a pericyte- and α-SMA-rich vasculature. Expression of α-SMA in tumor vasculature could therefore be useful for predicting tumor response to RRx-001.


Subject(s)
Antineoplastic Agents/administration & dosage , Azetidines/administration & dosage , Neoplasms/blood supply , Neoplasms/drug therapy , Nitro Compounds/administration & dosage , Actins/metabolism , Animals , Antineoplastic Agents/pharmacology , Azetidines/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Epigenesis, Genetic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neoplasm Transplantation , Neoplasms/metabolism , Nitro Compounds/pharmacology , Nitroimidazoles/administration & dosage , Nitroimidazoles/pharmacology , Tumor Microenvironment/drug effects
17.
Angiogenesis ; 19(2): 229-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26961182

ABSTRACT

BACKGROUND: Metronomic chemotherapy has shown promising activity against solid tumors and is believed to act in an antiangiogenic manner. The current study describes and quantifies the therapeutic efficacy, and mode of activity, of metronomic gemcitabine and a dedicated antiangiogenic agent (DC101) in patient-derived xenografts of pancreatic cancer. METHODS: Two primary human pancreatic cancer xenograft lines were dosed metronomically with gemcitabine or DC101 weekly. Changes in tumor growth, vascular function, and metabolism over time were measured with magnetic resonance imaging, positron emission tomography, and immunofluorescence microscopy to determine the anti-tumor effects of the respective treatments. RESULTS: Tumors treated with metronomic gemcitabine were 10-fold smaller than those in the control and DC101 groups. Metronomic gemcitabine, but not DC101, reduced the tumors' avidity for glucose, proliferation, and apoptosis. Metronomic gemcitabine-treated tumors had higher perfusion rates and uniformly distributed blood flow within the tumor, whereas perfusion rates in DC101-treated tumors were lower and confined to the periphery. DC101 treatment reduced the tumor's vascular density, but did not change their function. In contrast, metronomic gemcitabine increased vessel density, improved tumor perfusion transiently, and decreased hypoxia. CONCLUSION: The aggregate data suggest that metronomic gemcitabine treatment affects both tumor vasculature and tumor cells continuously, and the overall effect is to significantly slow tumor growth. The observed increase in tumor perfusion induced by metronomic gemcitabine may be used as a therapeutic window for the administration of a second drug or radiation therapy. Non-invasive imaging could be used to detect early changes in tumor physiology before reductions in tumor volume were evident.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Deoxycytidine/analogs & derivatives , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Xenograft Model Antitumor Assays , Administration, Metronomic , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Humans , Male , Mice, SCID , Microvessels/drug effects , Microvessels/pathology , Necrosis , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Perfusion , Gemcitabine
18.
Contrast Media Mol Imaging ; 11(1): 77-88, 2016.
Article in English | MEDLINE | ID: mdl-26268906

ABSTRACT

Macromolecular gadolinium (Gd)-based contrast agents are in development as blood pool markers for MRI. HPG-GdF is a 583 kDa hyperbranched polyglycerol doubly tagged with Gd and Alexa 647 nm dye, making it both MR and histologically visible. In this study we examined the location of HPG-GdF in whole-tumor xenograft sections matched to in vivo DCE-MR images of both HPG-GdF and Gadovist. Despite its large size, we have shown that HPG-GdF extravasates from some tumor vessels and accumulates over time, but does not distribute beyond a few cell diameters from vessels. Fractional plasma volume (fPV) and apparent permeability-surface area product (aPS) parameters were derived from the MR concentration-time curves of HPG-GdF. Non-viable necrotic tumor tissue was excluded from the analysis by applying a novel bolus arrival time (BAT) algorithm to all voxels. aPS derived from HPG-GdF was the only MR parameter to identify a difference in vascular function between HCT116 and HT29 colorectal tumors. This study is the first to relate low and high molecular weight contrast agents with matched whole-tumor histological sections. These detailed comparisons identified tumor regions that appear distinct from each other using the HPG-GdF biomarkers related to perfusion and vessel leakiness, while Gadovist-imaged parameter measures in the same regions were unable to detect variation in vascular function. We have established HPG-GdF as a biocompatible multi-modal high molecular weight contrast agent with application for examining vascular function in both MR and histological modalities.


Subject(s)
Contrast Media/administration & dosage , Glycerol/administration & dosage , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Polymers/administration & dosage , Animals , Cell Line, Tumor , Contrast Media/chemistry , Gadolinium DTPA/administration & dosage , Gadolinium DTPA/chemistry , Glycerol/chemistry , Humans , Mice , Neovascularization, Pathologic/diagnostic imaging , Organometallic Compounds/administration & dosage , Organometallic Compounds/chemistry , Polymers/chemistry , Radiography , Xenograft Model Antitumor Assays
19.
Microvasc Res ; 101: 38-47, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26092680

ABSTRACT

Pericytes are perivascular support cells, the origin of which in tumor tissue is not clear. Recently, we identified a Tie1(+) precursor cell that differentiates into vascular smooth muscle, in a Notch-dependent manner. To understand the involvement of Notch in the ontogeny of tumor pericytes we used a novel flow immunophenotyping strategy to define CD146(+)/CD45(-)/CD31(-/lo) pericytes in the tumor stroma. This strategy combined with ex vivo co-culture experiments identified a novel pericyte progenitor cell population defined as Sca1(hi)/CD146(-)/CD45(-)/CD31(-). The differentiation of these progenitor cells was stimulated by co-culture with endothelial cells. Overexpression of the Notch ligand Jagged1 in endothelial cells further stimulated the differentiation of Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells into pericytes, while inhibition of Notch signaling with a γ-secretase inhibitor reduced this differentiation. However, Notch inhibition specifically in Tie1-expressing cells did not change the abundance of pericytes in tumors, suggesting that the pericyte precursor is distinct from the vascular smooth muscle cell precursor. Transplant experiments showed that the bone marrow contributes minimally to tumor pericytes. Immunophenotyping revealed that Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells have greater potential to differentiate into pericytes and have increased expression of classic mesenchymal stem cell markers (CD13, CD44, Nt5e and Thy-1) compared to Sca1(-/lo)/CD146(-)/CD45(-)/CD31(-) cells. Our results suggest that a local Sca1(hi)/CD146(-)/CD45(-)/CD31(-) pericyte progenitor resides in the tumor microenvironment and requires Notch signaling for differentiation into mature pericytes.


Subject(s)
Neoplasms/metabolism , Pericytes/cytology , Receptors, Notch/metabolism , Animals , Ataxin-1/metabolism , Bone Marrow Transplantation , CD146 Antigen/metabolism , Carcinoma, Lewis Lung , Cell Differentiation , Coculture Techniques , Flow Cytometry , Human Umbilical Vein Endothelial Cells , Humans , Leukocyte Common Antigens/metabolism , Melanoma, Experimental , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, TIE-1/metabolism , Signal Transduction , Stem Cells/cytology
20.
Cancer Cell ; 27(5): 682-97, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25965573

ABSTRACT

Metastatic dissemination is the leading cause of death in cancer patients, which is particularly evident for high-risk sarcomas such as Ewing sarcoma, osteosarcoma, and rhabdomyosarcoma. Previous research identified a crucial role for YB-1 in the epithelial-to-mesenchymal transition (EMT) and metastasis of epithelial malignancies. Based on clinical data and two distinct animal models, we now report that YB-1 is also a major metastatic driver in high-risk sarcomas. Our data establish YB-1 as a critical regulator of hypoxia-inducible factor 1α (HIF1α) expression in sarcoma cells. YB-1 enhances HIF1α protein expression by directly binding to and activating translation of HIF1A messages. This leads to HIF1α-mediated sarcoma cell invasion and enhanced metastatic capacity in vivo, highlighting a translationally regulated YB-1-HIF1α axis in sarcoma metastasis.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Neoplasm Metastasis , Protein Biosynthesis , Sarcoma/pathology , Y-Box-Binding Protein 1/physiology , Humans , Neoplasm Invasiveness , Sarcoma/genetics , Von Hippel-Lindau Tumor Suppressor Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL