Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
iScience ; 27(4): 109593, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38632987

ABSTRACT

Precise regulation of Type I interferon signaling is crucial for combating infection and cancer while avoiding autoimmunity. Type I interferon signaling is negatively regulated by USP18. USP18 cleaves ISG15, an interferon-induced ubiquitin-like modification, via its canonical catalytic function, and inhibits Type I interferon receptor activity through its scaffold role. USP18 loss-of-function dramatically impacts immune regulation, pathogen susceptibility, and tumor growth. However, prior studies have reached conflicting conclusions regarding the relative importance of catalytic versus scaffold function. Here, we develop biochemical and cellular methods to systematically define the physiological role of USP18. By comparing a patient-derived mutation impairing scaffold function (I60N) to a mutation disrupting catalytic activity (C64S), we demonstrate that scaffold function is critical for cancer cell vulnerability to Type I interferon. Surprisingly, we discovered that human USP18 exhibits minimal catalytic activity, in stark contrast to mouse USP18. These findings resolve human USP18's mechanism-of-action and enable USP18-targeted therapeutics.

2.
Transl Oncol ; 40: 101833, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38128467

ABSTRACT

Lung cancer remains a leading cause of cancer-related death, but scientists have made great strides in developing new treatments recently, partly owing to the use of genetically engineered mouse models (GEMMs). GEMM tumors represent a translational model that recapitulates human disease better than implanted models because tumors develop spontaneously in the lungs. However, detection of these tumors relies on in vivo imaging tools, specifically micro-Computed Tomography (micro-CT or µCT), and image analysis can be laborious with high inter-user variability. Here we present a deep learning model trained to perform fully automated segmentation of lung tumors without the interference of other soft tissues. Trained and tested on 100 3D µCT images (18,662 slices) that were manually segmented, the model demonstrated a high correlation to manual segmentations on the testing data (r2=0.99, DSC=0.78) and on an independent dataset (n = 12 3D scans or 2328 2D slices, r2=0.97, DSC=0.73). In a comparison against manual segmentation performed by multiple analysts, the model (r2=0.98, DSC=0.78) performed within inter-reader variability (r2=0.79, DSC=0.69) and close to intra-reader variability (r2=0.99, DSC=0.82), all while completing 5+ hours of manual segmentations in 1 minute. Finally, when applied to a real-world longitudinal study (n = 55 mice), the model successfully detected tumor progression over time and the differences in tumor burden between groups induced with different virus titers, aligning well with more traditional analysis methods. In conclusion, we have developed a deep learning model which can perform fast, accurate, and fully automated segmentation of µCT scans of murine lung tumors.

3.
Mol Cancer Ther ; 22(7): 891-900, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37186518

ABSTRACT

KRAS is one of the most commonly mutated oncogenes in lung, colorectal, and pancreatic cancers. Recent clinical trials directly targeting KRAS G12C presented encouraging results for a large population of non-small cell lung cancer (NSCLC), but resistance to treatment is a concern. Continued exploration of new inhibitors and preclinical models is needed to address resistance mechanisms and improve duration of patient responses. To further enable the development of KRAS G12C inhibitors, we present a preclinical framework involving translational, non-invasive imaging modalities (CT and PET) and histopathology in a conventional xenograft model and a novel KRAS G12C knock-in mouse model of NSCLC. We utilized an in-house developed KRAS G12C inhibitor (Compound A) as a tool to demonstrate the value of this framework in studying in vivo pharmacokinetic/pharmacodynamic (PK/PD) relationship and anti-tumor efficacy. We characterized the Kras G12C-driven genetically engineered mouse model (GEMM) and identify tumor growth and signaling differences compared to its Kras G12D-driven counterpart. We also find that Compound A has comparable efficacy to sotorasib in the Kras G12C-driven lung tumors arising in the GEMM, but like observations in the clinic, some tumors inevitably progress on treatment. These findings establish a foundation for evaluating future KRAS G12C inhibitors that is not limited to xenograft studies and can be applied in a translationally relevant mouse model that mirrors human disease progression and resistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Mice , Humans , Heterografts , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Transplantation, Heterologous , Disease Models, Animal , Mutation
4.
Med Image Anal ; 86: 102786, 2023 05.
Article in English | MEDLINE | ID: mdl-36878160

ABSTRACT

Spine registration for volumetric magnetic resonance (MR) and computed tomography (CT) images plays a significant role in surgical planning and surgical navigation system for the radiofrequency ablation of spine intervertebral discs. The affine transformation of each vertebra and elastic deformation of the intervertebral disc exist at the same time. This situation is a major challenge in spine registration. Existing spinal image registration methods failed to solve the optimal affine-elastic deformation field (AEDF) simultaneously, only consider the overall rigid or elastic alignment with the help of a manual spine mask, and encounter difficulty in meeting the accuracy requirements of clinical registration application. In this study, we propose a novel affine-elastic registration framework named SpineRegNet. The SpineRegNet consists of a Multiple Affine Matrices Estimation (MAME) Module for multiple vertebrae alignment, an Affine-Elastic Fusion (AEF) Module for joint estimation of the overall AEDF, and a Local Rigidity Constraint (LRC) Module for preserving the rigidity of each vertebra. Experiments on T2-weighted volumetric MR and CT images show that the proposed approach achieves impressive performance with mean Dice similarity coefficients of 91.36%, 81.60%, and 83.08% for the mask of the vertebrae on Datasets A-C, respectively. The proposed technique does not require a mask or manual participation during the tests and provides a useful tool for clinical spinal disease surgical planning and surgical navigation systems.


Subject(s)
Algorithms , Intervertebral Disc , Humans , Magnetic Resonance Imaging/methods , Tomography, X-Ray Computed/methods , Magnetic Resonance Spectroscopy , Image Processing, Computer-Assisted/methods
5.
Polymers (Basel) ; 15(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38232011

ABSTRACT

Composite polymolecular separation membranes were prepared by combining multi-branched ZIF-L with high-porosity electrospinning nanofibers PI. Meanwhile, PDA and PEI were introduced into the membrane in order to improve its adhesion. The new membrane is called the "PI@PDA@PEI/ZIF-L-4" composite membrane. Compared with the PI@PDA@PEI/ZIF-8 composite membrane, the new membrane's filtration rates for heavy metal ions such as Cd2+, Cr3+, and Pb2+ were increased by 7.0%, 6.6%, and 9.3%, respectively. Furthermore, the new membrane has a permeability of up to 1140.0 L·m-2·h-1·bar-1, and displayed a very stable performance after four repeated uses. The separation mechanism of the PI@PDA@PEI/ZIF-L composite membrane was analyzed further in order to provide a basis to support the production of separation membranes with a high barrier rate and high flux.

6.
Cytotherapy ; 24(8): 827-834, 2022 08.
Article in English | MEDLINE | ID: mdl-35400595

ABSTRACT

BACKGROUND AIMS: Chimeric antigen receptor (CAR) T-cell therapy can be associated with significant toxicities. CAR-engineered natural killer (NK) cells provide a safer alternative while maintaining anti-tumor effects. Activated NK (aNK) cells are a clinical-grade cellular product obtained from the NK-92 cell line that have demonstrated both safety and potent cytotoxicity toward a wide range of cancers in phase 1 trials. Genetically engineered variants of aNK cells expressing a high-affinity Fc receptor (haNK) or co-expressing a CAR (t-haNK) are currently in phase 1/2 clinical trials. A key factor in the efficacy of cellular immunotherapies is biodistribution and tumor infiltration, which affect the local effector:target ratio. The chemokines CCL19 and CCL21 can drive recruitment of CCR7 receptor-expressing immune cells to secondary lymphoid organs. METHODS: Since NK-92 cells do not spontaneously express CCR7, clinical-grade aNK cells were transfected with a non-viral vector containing the CCR7 receptor, an anti-CD19 CAR and a high-affinity CD16 Fc receptor. RESULTS: CCR7-engineered CD19 t-haNK showed significant migration in vitro toward K562 cells engineered to secrete CCL19. This observation was confirmed in a NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mouse model in which subcutaneous tumors of CCL19-expressing K562 cells displayed a higher number of infiltrating CCR7_CD19 t-haNK cells than CCR7-negative CD19 t-haNK cells. In NSG mice inoculated either intravenously or subcutaneously with CCL19-secreting Raji cells, treatment with CCR7_CD19 t-haNK improved survival and tumor control compared with CD19 t-haNK or vehicle. CONCLUSIONS: Expression of CCR7 receptor by off-the-shelf t-haNK cells improves their homing toward lymph node chemokines both in vitro and in vivo, resulting in superior tumor control.


Subject(s)
Immunotherapy, Adoptive , Lymphoma , Receptors, CCR7 , Animals , Antigens, CD19 , Cell Line, Tumor , Chemokine CCL19/genetics , Chemokine CCL19/metabolism , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural , Lymphoma/therapy , Mice , Mice, Inbred NOD , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, Chimeric Antigen/metabolism , Receptors, Fc/metabolism , Tissue Distribution
7.
PLoS One ; 16(9): e0257694, 2021.
Article in English | MEDLINE | ID: mdl-34543354

ABSTRACT

In oncology research, while xenograft tumor models are easily visualized and humane endpoints can be clearly defined, metastatic tumor models are often based on more subjective clinical observations as endpoints. This study aimed at identifying objective non-invasive criteria for predicting imminent distress and mortality in metastatic lung tumor-bearing mice. BALB/c and C57BL/6 mice were inoculated with CT26 or B16F10 cells, respectively. The mice were housed in Vium smart cages to continuously monitor and stream respiratory rate and locomotion for up to 28 days until scheduled euthanasia or humane endpoint criteria were met. Body weight and body temperature were measured during the study. On days 11, 14, 17 and 28, lungs of subsets of animals were microCT imaged in vivo to assess lung metastasis progression and then euthanized for lung microscopic evaluations. Beginning at day 21, most tumor-bearing animals developed increased respiratory rates followed by decreased locomotion 1-2 days later, compared with the baseline values. Increases in respiratory rate did not correlate to surface tumor nodule counts or lung weight. Body weight measurement did not show significant changes from days 14-28 in either tumor-bearing or control animals. We propose that increases in respiratory rate (1.3-1.5 X) can be used to provide an objective benchmark to signal the need for increased clinical observations or euthanasia. Adoption of this novel humane endpoint criterion would allow investigators time to collect tissue samples prior to spontaneous morbidity or death and significantly reduce the distress of mice in the terminal stages of these metastatic lung tumor models.


Subject(s)
Lung Neoplasms , Respiratory Rate , Animals , Body Temperature , Disease Models, Animal , Mice
8.
MAbs ; 13(1): 1958662, 2021.
Article in English | MEDLINE | ID: mdl-34347577

ABSTRACT

IL13Rα2 is a cell surface tumor antigen that is overexpressed in multiple tumor types. Here, we studied biodistribution and targeting potential of an anti-IL13Rα2 antibody (Ab) and anti-tumor activity of anti-IL13Rα2-antibody-drug conjugate (ADC). The anti-IL13Rα2 Ab was labeled with fluorophore AF680 or radioisotope 89Zr for in vivo tracking using fluorescence molecular tomography (FMT) or positron emission tomography (PET) imaging, respectively. Both imaging modalities showed that the tumor was the major uptake site for anti-IL13Rα2-Ab, with peak uptake of 5-8% ID and 10% ID/g as quantified from FMT and PET, respectively. Pharmacological in vivo competition with excess of unlabeled anti-IL13Rα2-Ab significantly reduced the tumor uptake, indicative of antigen-specific tumor accumulation. Further, FMT imaging demonstrated similar biodistribution and pharmacokinetic profiles of an auristatin-conjugated anti-IL13Rα2-ADC as compared to the parental Ab. Finally, the anti-IL13Rα2-ADC exhibited a dose-dependent anti-tumor effect on A375 xenografts, with 90% complete responders at a dose of 3 mg/kg. Taken together, both FMT and PET showed a favorable biodistribution profile for anti-IL13Rα2-Ab/ADC, along with antigen-specific tumor targeting and excellent therapeutic efficacy in the A375 xenograft model. This work shows the great potential of this anti-IL13Rα2-ADC as a targeted anti-cancer agent.


Subject(s)
Aminobenzoates , Antineoplastic Agents, Immunological , Immunoconjugates , Interleukin-13 Receptor alpha2 Subunit , Melanoma, Experimental , Neoplasm Proteins , Oligopeptides , Aminobenzoates/immunology , Aminobenzoates/pharmacokinetics , Aminobenzoates/pharmacology , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Mice , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Oligopeptides/immunology , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Xenograft Model Antitumor Assays
9.
PLoS One ; 16(6): e0252950, 2021.
Article in English | MEDLINE | ID: mdl-34138905

ABSTRACT

Unlike the majority of cancers, survival for lung cancer has not shown much improvement since the early 1970s and survival rates remain low. Genetically engineered mice tumor models are of high translational relevance as we can generate tissue specific mutations which are observed in lung cancer patients. Since these tumors cannot be detected and quantified by traditional methods, we use micro-computed tomography imaging for longitudinal evaluation and to measure response to therapy. Conventionally, we analyze microCT images of lung cancer via a manual segmentation. Manual segmentation is time-consuming and sensitive to intra- and inter-analyst variation. To overcome the limitations of manual segmentation, we set out to develop a fully-automated alternative, the Mouse Lung Automated Segmentation Tool (MLAST). MLAST locates the thoracic region of interest, thresholds and categorizes the lung field into three tissue categories: soft tissue, intermediate, and lung. An increase in the tumor burden was measured by a decrease in lung volume with a simultaneous increase in soft and intermediate tissue quantities. MLAST segmentation was validated against three methods: manual scoring, manual segmentation, and histology. MLAST was applied in an efficacy trial using a Kras/Lkb1 non-small cell lung cancer model and demonstrated adequate precision and sensitivity in quantifying tumor growth inhibition after drug treatment. Implementation of MLAST has considerably accelerated the microCT data analysis, allowing for larger study sizes and mid-study readouts. This study illustrates how automated image analysis tools for large datasets can be used in preclinical imaging to deliver high throughput and quantitative results.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Radiographic Image Interpretation, Computer-Assisted/methods , AMP-Activated Protein Kinases , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Mice , Neoplasms, Experimental , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Burden , X-Ray Microtomography
10.
Cancer Res ; 80(13): 2903-2913, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32409308

ABSTRACT

CD8-expressing T cells are the main effector cells in cancer immunotherapy. Treatment-induced changes in intratumoral CD8+ T cells may represent a biomarker to identify patients responding to cancer immunotherapy. Here, we have used a 89Zr-radiolabeled human CD8-specific minibody (89Zr-Df-IAB22M2C) to monitor CD8+ T-cell tumor infiltrates by PET. The ability of this tracer to quantify CD8+ T-cell tumor infiltrates was evaluated in preclinical studies following single-agent treatment with FOLR1-T-cell bispecific (TCB) antibody and combination therapy of CEA-TCB (RG7802) and CEA-targeted 4-1BB agonist CEA-4-1BBL. In vitro cytotoxicity assays with peripheral blood mononuclear cells and CEA-expressing MKN-45 gastric or FOLR1-expressing HeLa cervical cancer cells confirmed noninterference of the anti-CD8-PET-tracer with the mode of action of CEA-TCB/CEA-4-1BBL and FOLR1-TCB at relevant doses. In vivo, the extent of tumor regression induced by combination treatment with CEA-TCB/CEA-4-1BBL in MKN-45 tumor-bearing humanized mice correlated with intratumoral CD8+ T-cell infiltration. This was detectable by 89Zr-IAB22M2C-PET and γ-counting. Similarly, single-agent treatment with FOLR1-TCB induced strong CD8+ T-cell infiltration in HeLa tumors, where 89Zr-Df-IAB22M2C again was able to detect CD8 tumor infiltrates. CD8-IHC confirmed the PET imaging results. Taken together, the anti-CD8-minibody 89Zr-Df-IAB22M2C revealed a high sensitivity for the detection of intratumoral CD8+ T-cell infiltrates upon either single or combination treatment with TCB antibody-based fusion proteins. These results provide further evidence that the anti-CD8 tracer, which is currently in clinical phase II, is a promising monitoring tool for intratumoral CD8+ T cells in patients treated with cancer immunotherapy. SIGNIFICANCE: Monitoring the pharmacodynamic activity of cancer immunotherapy with novel molecular imaging tools such as 89Zr-Df-IAB22M2C for PET imaging is of prime importance to identify patients responding early to cancer immunotherapy.


Subject(s)
Antibodies, Bispecific/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Molecular Imaging/methods , Positron-Emission Tomography/methods , Uterine Cervical Neoplasms/immunology , Zirconium/metabolism , Animals , Antibodies, Bispecific/immunology , Carcinoembryonic Antigen , Female , Folate Receptor 1/immunology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Radiopharmaceuticals/metabolism , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/therapy
11.
J Orthop Res ; 37(7): 1638-1648, 2019 07.
Article in English | MEDLINE | ID: mdl-30737824

ABSTRACT

The correlation between BMP-2 and osteosarcoma growth has gained increased interest in the recent years, however, there is still no consensus. In this study, we tested the effects of BMP-2 on osteosarcoma cells through both in vitro and in vivo experiments. The effect of BMP-2 on the proliferation, migration and invasion of osteosarcoma cells was tested in vitro. Subcutaneous and intratibial tumor models were used for the in vivo experiments in nude mice. The effects of BMP-2 on EMT of osteosarcoma cells and the Wnt/ß-catenin signaling pathway were also tested using a variety of biochemical methods. In vitro tests did not show a significant effect of BMP-2 on tumor cell proliferation. However, BMP-2 increased the mobility of tumor cells and the invasion assay demonstrated that BMP-2 promoted invasion of osteosarcoma cells in vitro. In vivo animal study showed that BMP-2 dramatically enhanced tumor growth. We also found that BMP-2 induced EMT of osteosarcoma cells. The expression levels of Axin2 and Dkk-1 were both down regulated by BMP-2 treatment, while ß-catenin, c-myc and Cyclin-D1 were all upregulated. The expression of Wnt3α and p-GSK-3ß were also significantly upregulated indicating that the Wnt/ß-catenin signaling pathway was activated during the EMT of osteosarcoma driven by BMP-2. From this study, we can conclude that BMP-2 significantly promotes growth of osteosarcoma cells (143B, MG63), and enhances mobility and invasiveness of tumor cells as demonstrated in vitro. The underlying mechanism might be that BMP-2 promotes EMT of osteosarcoma through the Wnt/ß-catenin signaling pathway. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1638-1648, 2019.


Subject(s)
Bone Morphogenetic Protein 2/adverse effects , Bone Neoplasms , Epithelial-Mesenchymal Transition/drug effects , Osteosarcoma , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, SCID , Wnt Signaling Pathway
12.
Clin Cancer Res ; 25(1): 188-200, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30301826

ABSTRACT

PURPOSE: The inability to intraoperatively distinguish primary tumor, as well as lymphatic spread, increases the probability of positive surgical margins, tumor recurrence, and surgical toxicity. The goal of this study was to develop a tumor-specific optical probe for real-time fluorescence-guided surgery. EXPERIMENTAL DESIGN: A humanized antibody fragment against PSCA (A11 minibody, A11 Mb) was conjugated with a near-infrared fluorophore, IRDye800CW. The integrity and binding of the probe to PSCA were confirmed by gel electrophoresis, size-exclusion chromatography, and flow cytometry, respectively. The ability of the probe to detect tumor-infiltrated lymph nodes and metastatic lesions was evaluated in 2 xenograft models, as well as in transgenic mice expressing human PSCA (hPSCA). An invasive intramuscular model was utilized to evaluate the efficacy of the A11 Mb-IRDye800CW-guided surgery. RESULTS: A11 Mb was successfully conjugated with IRDye800CW and retained specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 48 hours. The A11 Mb-IRDye800CW specifically detected PSCA-positive primary tumors, tumor-infiltrated lymph nodes, and distant metastases with high contrast. Fluorescence guidance facilitated more complete tumor resection, reduced tumor recurrence, and improved overall survival, compared with conventional white light surgery. The probe successfully identified primary orthotopic tumors and metastatic lesions in hPSCA transgenic mice. CONCLUSIONS: Real-time fluorescence image-guided surgery with A11 Mb-IRDye800CW enabled detection of lymph node metastases and positive surgical margins, facilitated more complete tumor removal, and improved survival, compared with white light surgery. These results may be translatable into clinical practice to improve surgical and patient outcomes.


Subject(s)
Antigens, Surface/genetics , Glutamate Carboxypeptidase II/genetics , Indoles/pharmacology , Prostatic Neoplasms/diagnostic imaging , Surgery, Computer-Assisted , Animals , Antigens, Surface/isolation & purification , Cell Line, Tumor , Disease Models, Animal , Fluorescence , Gene Expression Regulation, Neoplastic/genetics , Glutamate Carboxypeptidase II/isolation & purification , Heterografts , Humans , Infrared Rays , Male , Margins of Excision , Mice , Optical Imaging , Prostate/surgery , Prostatectomy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Spectroscopy, Near-Infrared
13.
ACS Nano ; 10(1): 1417-24, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26731174

ABSTRACT

A pretargeted oncologic positron emission tomography (PET) imaging that leverages the power of supramolecular nanoparticles with in vivo bioorthogonal chemistry was demonstrated for the clinically relevant problem of tumor imaging. The advantages of this approach are that (i) the pharmacokinetics (PKs) of tumor-targeting and imaging agents can be independently altered via chemical alteration to achieve the desired in vivo performance and (ii) the interplay between the two PKs and other controllable variables confers a second layer of control toward improved PET imaging. In brief, we utilized supramolecular chemistry to synthesize tumor-targeting nanoparticles containing transcyclooctene (TCO, a bioorthogonal reactive motif), called TCO⊂SNPs. After the intravenous injection and subsequent concentration of the TCO⊂SNPs in the tumors of living mice, a small molecule containing both the complementary bioorthogonal motif (tetrazine, Tz) and a positron-emitting radioisotope ((64)Cu) was injected to react selectively and irreversibly to TCO. High-contrast PET imaging of the tumor mass was accomplished after the rapid clearance of the unreacted (64)Cu-Tz probe. Our nanoparticle approach encompasses a wider gamut of tumor types due to the use of EPR effects, which is a universal phenomenon for most solid tumors.


Subject(s)
Cyclooctanes/chemistry , Glioblastoma/diagnostic imaging , Glioblastoma/diagnosis , Heterocyclic Compounds, 1-Ring/chemistry , Nanoparticles/chemistry , Positron-Emission Tomography/methods , Animals , Copper Radioisotopes/administration & dosage , Copper Radioisotopes/chemistry , Dendrimers/chemistry , Glioblastoma/pathology , Heterocyclic Compounds, 1-Ring/administration & dosage , Humans , Injections, Subcutaneous , Mice , Mice, Nude , Nanoparticles/ultrastructure , Neoplasm Transplantation , Permeability , Polyethylenes/chemistry , Transplantation, Heterologous
14.
Clin Cancer Res ; 22(6): 1403-12, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26490315

ABSTRACT

PURPOSE: The inability to visualize cancer during prostatectomy contributes to positive margins, cancer recurrence, and surgical side effects. A molecularly targeted fluorescent probe offers the potential for real-time intraoperative imaging. The goal of this study was to develop a probe for image-guided prostate cancer surgery. EXPERIMENTAL DESIGN: An antibody fragment (cys-diabody, cDb) against prostate stem cell antigen (PSCA) was conjugated to a far-red fluorophore, Cy5. The integrity and binding of the probe to PSCA was confirmed by gel electrophoresis, size exclusion, and flow cytometry, respectively. Subcutaneous models of PSCA-expressing xenografts were used to assess the biodistribution and in vivo kinetics, whereas an invasive intramuscular model was utilized to explore the performance of Cy5-cDb-mediated fluorescence guidance in representative surgical scenarios. Finally, a prospective, randomized study comparing surgical resection with and without fluorescent guidance was performed to determine whether this probe could reduce the incidence of positive margins. RESULTS: Cy5-cDb demonstrated excellent purity, stability, and specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 6 hours. In mice carrying PSCA(+) and negative (-) dual xenografts, the mean fluorescence ratio of PSCA(+/-) tumors was 4.4:1. In surgical resection experiments, residual tumors <1 mm that were missed on white light surgery were identified and resected using fluorescence guidance, which reduced the incidence of positive surgical margins (0/8) compared with white light surgery alone (7/7). CONCLUSIONS: Fluorescently labeled cDb enables real-time in vivo imaging of prostate cancer xenografts in mice, and facilitates more complete tumor removal than conventional white light surgery alone.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoglobulin Fragments/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Surgery, Computer-Assisted , Animals , Antigens, Neoplasm/metabolism , Disease Models, Animal , Fluorescent Dyes , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Male , Mice , Neoplasm Proteins/metabolism , Optical Imaging/methods , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Surgery, Computer-Assisted/methods , Xenograft Model Antitumor Assays
15.
Cancer Res ; 75(22): 4742-52, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26471360

ABSTRACT

Malignant ascites is a common complication in the late stages of epithelial ovarian cancer (EOC) that greatly diminishes the quality of life of patients. Malignant ascites is a known consequence of vascular dysfunction, but current approved treatments are not effective in preventing fluid accumulation. In this study, we investigated an alternative strategy of targeting macrophage functions to reverse the vascular pathology of malignant ascites using fluid from human patients and an immunocompetent murine model (ID8) of EOC that mirrors human disease by developing progressive vascular disorganization and leakiness culminating in massive ascites. We demonstrate that the macrophage content in ascites fluid from human patients and the ID8 model directly correlates with vascular permeability. To further substantiate macrophages' role in the pathogenesis of malignant ascites, we blocked macrophage function in ID8 mice using a colony-stimulating factor 1 receptor kinase inhibitor (GW2580). Administration of GW2580 in the late stages of disease resulted in reduced infiltration of protumorigenic (M2) macrophages and dramatically decreased ascites volume. Moreover, the disorganized peritoneal vasculature became normalized and sera from GW2580-treated ascites protected against endothelial permeability. Therefore, our findings suggest that macrophage-targeted treatment may be a promising strategy toward a safe and effective means to control malignant ascites of EOC.


Subject(s)
Anisoles/pharmacology , Ascites/prevention & control , Capillary Permeability/drug effects , Macrophages/drug effects , Neoplasms, Glandular and Epithelial/complications , Ovarian Neoplasms/complications , Pyrimidines/pharmacology , Animals , Ascites/etiology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
16.
Cancer Res ; 75(6): 950-62, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25736687

ABSTRACT

Growing evidence suggests that tumor-associated macrophages (TAM) promote cancer progression and therapeutic resistance by enhancing angiogenesis, matrix-remodeling, and immunosuppression. In this study, prostate cancer under androgen blockade therapy (ABT) was investigated, demonstrating that TAMs contribute to prostate cancer disease recurrence through paracrine signaling processes. ABT induced the tumor cells to express macrophage colony-stimulating factor 1 (M-CSF1 or CSF1) and other cytokines that recruit and modulate macrophages, causing a significant increase in TAM infiltration. Inhibitors of CSF1 signaling through its receptor, CSF1R, were tested in combination with ABT, demonstrating that blockade of TAM influx in this setting disrupts tumor promotion and sustains a more durable therapeutic response compared with ABT alone.


Subject(s)
Androgen Antagonists/therapeutic use , Macrophages/physiology , Prostatic Neoplasms/drug therapy , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Carcinogenesis , Cells, Cultured , Drug Resistance, Neoplasm , Humans , Male , Mice , Prostatic Neoplasms/pathology
17.
Clin Cancer Res ; 20(24): 6367-78, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25326233

ABSTRACT

PURPOSE: Prostate stem cell antigen (PSCA) is highly expressed in local prostate cancers and prostate cancer bone metastases and its expression correlates with androgen receptor activation and a poor prognosis. In this study, we investigate the potential clinical applications of immunoPET with the anti-PSCA A11 minibody, an antibody fragment optimized for use as an imaging agent. We compare A11 minibody immunoPET to (18)F-Fluoride PET bone scans for detecting prostate cancer bone tumors and evaluate the ability of the A11 minibody to image tumor response to androgen deprivation. EXPERIMENTAL DESIGN: Osteoblastic, PSCA-expressing, LAPC-9 intratibial xenografts were imaged with serial (124)I-anti-PSCA A11 minibody immunoPET and (18)F-Fluoride bone scans. Mice bearing LAPC-9 subcutaneous xenografts were treated with either vehicle or MDV-3100 and imaged with A11 minibody immunoPET/CT scans pre- and posttreatment. Ex vivo flow cytometry measured the change in PSCA expression in response to androgen deprivation. RESULTS: A11 minibody demonstrated improved sensitivity and specificity over (18)F-Fluoride bone scans for detecting LAPC-9 intratibial xenografts at all time points. LAPC-9 subcutaneous xenografts showed downregulation of PSCA when treated with MDV-3100 which A11 minibody immunoPET was able to detect in vivo. CONCLUSIONS: A11 minibody immunoPET has the potential to improve the sensitivity and specificity of clinical prostate cancer metastasis detection over bone scans, which are the current clinical standard-of-care. A11 minibody immunoPET additionally has the potential to image the activity of the androgen signaling axis in vivo which may help evaluate the clinical response to androgen deprivation and the development of castration resistance.


Subject(s)
Antigens, Neoplasm , Immunoglobulin Fragments , Iodine Radioisotopes , Neoplasm Proteins , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnosis , Androgen Antagonists/administration & dosage , Androgen Antagonists/pharmacology , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Benzamides , Disease Models, Animal , Disease Progression , GPI-Linked Proteins/immunology , Heterografts , Humans , Immunoglobulin Fragments/immunology , Male , Mice , Neoplasm Proteins/immunology , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Tissue Distribution , Treatment Outcome , Tumor Burden/drug effects , X-Ray Microtomography
18.
PLoS One ; 8(9): e73650, 2013.
Article in English | MEDLINE | ID: mdl-24023896

ABSTRACT

Tumor-specific adenoviral vectors comprise a fruitful gene-based diagnostic imaging and therapy research area for advanced stage of cancer, including metastatic disease. However, clinical translation of viral vectors has encountered considerable obstacles, largely due to host immune responses against the virus. Here, we explored the utilization of an immunosuppressant, rapamycin, to circumvent the anti-adenovirus immunity in immunocompetent murine prostate cancer models. Rapamycin diminished adenoviral-induced acute immune response by inhibiting NF-κB activation; it also reduced the scale and delayed the onset of inflammatory cytokine secretion. Further, we found that rapamycin abrogated anti-adenovirus antibody production and retarded the function of myeloid cells and lymphocytes that were activated upon viral administration in pre-immunized hosts. Thus, the co-administration of rapamycin prolonged and enhanced adenovirus-delivered transgene expression in vivo, and thereby augmented the imaging capability of adenoviral vectors in both bioluminescent and positron emission tomography modalities. Furthermore, we showed that despite an excellent response of cancer cells to a cytotoxic gene therapeutic vector in vitro, only minimal therapeutic effects were observed in vivo in pre-immunized mice. However, when we combined gene therapy with transient immunosuppression, complete tumor growth arrest was achieved. Overall, transient immunosuppression by rapamycin was able to boost the diagnostic utility and therapeutic potentials of adenoviral vectors.


Subject(s)
Adenoviridae/genetics , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Molecular Imaging , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Sirolimus/pharmacology , Adaptive Immunity/drug effects , Animals , Cell Line, Tumor , Ganciclovir/pharmacology , Genetic Therapy/adverse effects , Genetic Vectors/genetics , Immunity, Innate/drug effects , Immunization , Male , Mice , Molecular Imaging/adverse effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Safety , Thymidine Kinase/genetics , Transgenes/genetics
19.
J Control Release ; 166(1): 75-85, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23247040

ABSTRACT

We sought to modify adenoviral (Ad) particles by incorporating the advantageous characteristics of non-viral gene delivery vehicles to complement the viral vectors. α-Amino acid-N-carboxyanhydride chemistry was used to synthesize homopolypeptides and diblock copolypeptides that possess well-defined secondary structures. Using cryo-electron and fluorescence microscopy, we showed that these polypeptides can coat the surfaces of Ad particles in a non-covalent manner to modify their transduction properties. The coated Ad particles were found to bind to and be internalized by cells. In contrast to reports using covalently PEGylated Ad particles, we found that our physically coated Ad hybrid complexes facilitate gene transfer both in vitro and in vivo. We showed that our polypeptide coating was able to shield the Ad particles from the neutralizing effect of antibodies and mitigate the binding of blood coagulation factor (Factor X) in vitro. The coating also reduced the antigenicity of Ad in immunocompetent mice. The biodistribution of the systemically administered hybrid complexes mirrored the behavior of both viral and non-viral vectors, exhibiting liver tropism as well as enhanced lung transduction. These data demonstrated that our non-covalent modification was able to alter Ad's interactions with cells and organs with retention of transduction efficiency. Advantages such as facile coating of the Ad vector, design flexibility and ease of attaching ligands to the polypeptides make this system potentially useful as a platform for adding functionalities to Ad to target cancer metastasis.


Subject(s)
Adenoviridae/genetics , Drug Carriers/chemistry , Gene Transfer Techniques , Genetic Vectors , Peptides/chemistry , Transduction, Genetic , Animals , Antibodies, Viral/blood , Cell Line , Cryoelectron Microscopy , Drug Stability , Green Fluorescent Proteins/genetics , Humans , Luciferases, Firefly/genetics , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Microscopy, Fluorescence , Particle Size , Scattering, Radiation , Surface Properties
20.
Mol Ther ; 21(3): 554-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23247102

ABSTRACT

An imaging modality that can accurately discern prostate cancer (PCa) foci would be useful to detect PCa early or guide treatment. We have engineered numerous adenoviral vectors (Ads) to carry out reporter gene-based imaging using specific promoters to express a potent transcriptional activator, which in turn activates the reporter gene in PCa. This two-step transcriptional amplification (TSTA) method can boost promoters' activity, while maintaining cell specificity. Here, we examined a dual TSTA (DTSTA) approach, which utilizes TSTA not only to express the imaging reporter, but also to direct viral genome replication of a conditionally replicating Ad (CRAd) to further augment the expression levels of the reporter gene by genomic amplification supported in trans by coadministered CRAd. In vitro studies showed up to 50-fold increase of the reporter genome by DTSTA. Compared with TSTA reporter alone, DTSTA application exhibited a 25-fold increase in imaging signal in PCa xenografts. DTSTA approach is also beneficial for a combination of two TSTA Ads with distinct promoters, although amplification is observed only when TSTA-CRAd can replicate. Consequently, the DTSTA approach is a hybrid method of transcriptional and genomic augmentation that can provide higher level reporter gene expression potentially with a lower dose of viral administration.


Subject(s)
Genes, Reporter , Molecular Imaging/methods , Prostatic Neoplasms/diagnosis , Adenoviridae/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Male , Mice , Mice, SCID , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...