Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Sci Rep ; 14(1): 3285, 2024 02 08.
Article in English | MEDLINE | ID: mdl-38332021

ABSTRACT

The pharmaceutical industry is an important industry for the national economy and the people's livelihood, which is not only beneficial to the people's livelihood, but also has huge commercial value. How to promote the development of Chinese pharmaceutical industry is an urgent problem to be solved. In this study, 47 listed pharmaceutical companies are taken as cases, and Qualitative Comparative Analysis of Fuzzy Sets (fsQCA) is used to analyze the influence of five antecedent conditions on the total factor productivity of pharmaceutical enterprises from the perspective of corporate governance, and to explore the composition to Total Factor Productivity (TFP) improvement. The results are as follows. First, single corporate governance factor does not constitute the necessary condition to improve the TFP of pharmaceutical enterprises. Second, there are three configurations of high TFP of pharmaceutical enterprises, among these, two configurations belong to regulatory constraints type and one configuration belongs to the active board type. There is only one configurations to low TFP of pharmaceutical enterprises: the passive board. Based on the perspective of configuration, this paper discusses how corporate governance drives TFP improvement in pharmaceutical enterprises, which can provide systematic thinking and practical guidance for each company to promote its TFP improvement according to its own corporate structure.


Subject(s)
Medicine , Pharmacy , Humans , Asian People , Drug Industry , Pharmaceutical Preparations , China
2.
Nanoscale ; 12(42): 21519-21533, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33095224

ABSTRACT

Hypoxia, as a typical factor in a tumor microenvironment, plays a vital role in tumor treatment resistance, tumor invasion and migration. Hypoxia inducible factor (HIF), as the vital response element of hypoxia, mediates these untoward effects through a series of downstream reactions. Cancer treatments such as photodynamic therapy (PDT), radiotherapy (RT) and chemotherapy are severely hindered by hypoxia and HIF, back, however, could be intelligently manipulated through nanocomposite materials for their great potentiality to combine different functions. Herein, we reviewed the smart strategies in emerging research studies to overcome hypoxia toward the enhancement of tumor therapy.


Subject(s)
Neoplasms , Photochemotherapy , Tumor Hypoxia , Cell Hypoxia , Cell Line, Tumor , Humans , Hypoxia , Neoplasms/drug therapy , Tumor Microenvironment
4.
Biophys Rep ; 4(5): 254-264, 2018.
Article in English | MEDLINE | ID: mdl-30533490

ABSTRACT

ABSTRACT: Integrin αvß6 is expressed at an undetectable level in normal tissues, but is remarkably upregulated during many pathological processes, especially in cancer and fibrosis. Noninvasive imaging of integrin αvß6 expression using a radiotracer with favorable in vivo pharmacokinetics would facilitate disease diagnosis and therapy monitoring. Through disulfide-cyclized method, we synthesized in this study, a new integrin αvß6-targeted cyclic peptide (denoted as cHK), and radiolabeled it with 99mTc. The ability of the resulting radiotracer 99mTc-HYNIC-cHK to detect integrin αvß6 expression in pancreatic cancer xenografts and idiopathic pulmonary fibrosis was evaluated using small-animal single-photon emission computed tomography (SPECT)/computed tomography (CT). 99mTc-HYNIC-cHK showed significantly improved in vivo metabolic stability compared to the linear peptide-based radiotracer 99mTc-HYNIC-HK. 99mTc-HYNIC-cHK exhibited similar biodistribution properties to 99mTc-HYNIC-HK, but the tumor-to-muscle ratio was significantly increased (2.99 ± 0.87 vs. 1.82 ± 0.27, P < 0.05). High-contrast images of integrin αvß6-positive tumors and bleomycin-induced fibrotic lungs were obtained by SPECT/CT imaging using 99mTc-HYNIC-cHK. Overall, our studies demonstrate that 99mTc-HYNIC-cHK is a promising SPECT radiotracer for the noninvasive imaging of integrin αvß6 in living subjects.

5.
Biomaterials ; 158: 1-9, 2018 03.
Article in English | MEDLINE | ID: mdl-29272764

ABSTRACT

Increasing evidence indicates that the overexpression of galectin-1, a member of the galectin family, is related to tumor progression and invasion, as well as tumor resistance to therapies (e.g., radiotherapy). Herein, we investigated whether near-infrared fluorescence (NIRF) imaging and positron-emission tomography (PET) were sensitive approaches for detecting and quantitating galectin-1 upregulation in vivo. An anti-galectin-1 antibody was labeled with either an NIRF dye or 64Cu, and NIRF and PET imaging using the resulting probes (Dye-αGal-1 and 64Cu- 1,4,7-triazacyclononane-1,4,7-triacetic acid [NOTA]-αGal-1) were performed in 4T1 breast cancer-bearing mice treated with several rounds of sorafenib. Radiotherapy was performed in vitro and in vivo to identify the role of galectin-1 in radioresistance. NIRF and PET imaging both revealed significantly increased upregulation of galectin-1 in the hypoxic tumors after sorafenib treatment, which was verified by ex vivo biodistribution, western blotting, and enzyme-linked immunosorbent assays. Galectin-1 specific inhibition by thiodigalactoside dramatically improved the efficacy of radiotherapy, and overcame sorafenib-induced radiotherapy resistance. Taken together, galectin-1 is a key mediator of tumor resistance to radiotherapy. Targeted molecular imaging allows for real-time, noninvasive, and quantitative detection of the dynamic changes in galectin-1 levels in vivo; this introduces the possibility of early detection of tumor resistance to therapies.


Subject(s)
Drug Resistance, Multiple , Galectin 1/biosynthesis , Molecular Imaging , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Animals , Cell Line, Tumor , Female , Galectin 1/analysis , Gene Expression Regulation/radiation effects , Humans , Mice, Inbred BALB C , Optical Imaging , Positron-Emission Tomography , Sorafenib/pharmacology , Sorafenib/therapeutic use
6.
Theranostics ; 7(17): 4276-4288, 2017.
Article in English | MEDLINE | ID: mdl-29158825

ABSTRACT

Tumor relapse after initial regression post-chemotherapy is a major challenge in cancer treatment, as it usually leads to local-regional recurrence or inoperable distant metastasis. M2 macrophages diminish the tumor-inhibitory effect of chemotherapy and correlate with distant metastasis and poor prognosis. In this study, we investigated whether molecular imaging of M2 macrophages could serve as an early biomarker for tumor relapse after chemotherapy and tumor lymph node metastasis in preclinical mouse models. Methods: We developed M2 macrophage-targeted probes for near-infrared fluorescence (NIRF) imaging and single-photon emission computed tomography (SPECT) using an anti-CD206 monoclonal antibody. The specific targeting capacity and potential applications of the NIRF and SPECT probes were investigated in subcutaneous tumor and lymph node metastasis models of 4T1 murine breast cancer. Results: M2 macrophage infiltration was significantly increased in the 4T1 tumors that later underwent relapse but not in non-relapsing 4T1 tumors after cyclophosphamide treatment. Through NIRF imaging and SPECT using our synthesized probes, the infiltration of M2 macrophages in relapsing tumors and tumor lymph node metastasis could be sensitively detected. Importantly, early prediction of tumor relapse by molecular imaging of M2 macrophages resulted in an effective eradication of tumors upon combination with additional radiotherapy. Conclusion: Our findings demonstrate that M2 macrophage-targeted imaging allows for noninvasively predicting post-chemotherapy tumor relapse and sensitively detecting the metastatic lymph nodes in vivo. This imaging strategy could provide a better understanding of cancer progression, enable early prediction of tumor resistance, and have implications on the rational design of cancer therapeutics.


Subject(s)
Biomarkers/metabolism , Breast Neoplasms/metabolism , Lectins, C-Type/metabolism , Macrophages/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Line, Tumor , Female , Flow Cytometry , Lymphatic Metastasis/physiopathology , Mannose Receptor , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local/metabolism
7.
Cancer Res ; 77(21): 6021-6032, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28916656

ABSTRACT

Increased recruitment of tumor-associated macrophages (TAM) to tumors following chemotherapy promotes tumor resistance and recurrence and correlates with poor prognosis. TAM depletion suppresses tumor growth, but is not highly effective due to the effects of tumorigenic mediators from other stromal sources. Here, we report that adoptive macrophage transfer led to a dramatically enhanced photodynamic therapy (PDT) effect of 2-(1-hexyloxyethyl)-2-devinyl pyropheophor-bide-alpha (HPPH)-coated polyethylene glycosylated nanographene oxide [GO(HPPH)-PEG] by increasing its tumor accumulation. Moreover, tumor treatment with commonly used chemotherapeutic drugs induced an increase in macrophage infiltration into tumors, which also enhanced tumor uptake and the PDT effects of GO(HPPH)-PEG, resulting in tumor eradication. Macrophage recruitment to tumors after chemotherapy was visualized noninvasively by near-infrared fluorescence and single-photon emission CT imaging using F4/80-specific imaging probes. Our results demonstrate that chemotherapy combined with GO(HPPH)-PEG PDT is a promising strategy for the treatment of tumors, especially those resistant to chemotherapy. Furthermore, TAM-targeted molecular imaging could potentially be used to predict the efficacy of combination therapy and select patients who would most benefit from this treatment approach. Cancer Res; 77(21); 6021-32. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Graphite/chemistry , Macrophages/drug effects , Nanoparticles/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Animals , Cell Line , Cell Line, Tumor , Drug Therapy/methods , Fluorescent Dyes/chemistry , Macrophages/metabolism , Macrophages/pathology , Mice , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Outcome Assessment, Health Care/methods , Reproducibility of Results , Tomography, Emission-Computed, Single-Photon/methods
8.
ACS Nano ; 11(10): 10147-10158, 2017 10 24.
Article in English | MEDLINE | ID: mdl-28901740

ABSTRACT

Effective cancer therapy depends not only on destroying the primary tumor but also on conditioning the host immune system to recognize and eliminate residual tumor cells and prevent metastasis. In this study, a tumor integrin αvß6-targeting peptide (the HK peptide)-functionalized graphene oxide (GO) was coated with a photosensitizer (HPPH). The resulting GO conjugate, GO(HPPH)-PEG-HK, was investigated whether it could destroy primary tumors and boost host antitumor immunity. We found that GO(HPPH)-PEG-HK exhibited significantly higher tumor uptake than GO(HPPH)-PEG and HPPH. Photodynamic therapy (PDT) using GO(HPPH)-PEG suppressed tumor growth in both subcutaneous and lung metastatic mouse models. Necrotic tumor cells caused by GO(HPPH)-PEG-HK PDT activated dendritic cells and significantly prevented tumor growth and lung metastasis by increasing the infiltration of cytotoxic CD8+ T lymphocytes within tumors as evidenced by in vivo optical and single-photon emission computed tomography (SPECT)/CT imaging. These results demonstrate that tumor-targeted PDT using GO(HPPH)-PEG-HK could effectively ablate primary tumors and destroy residual tumor cells, thereby preventing distant metastasis by activating host antitumor immunity and suppressing tumor relapse by stimulation of immunological memory.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Graphite/pharmacology , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Photochemotherapy , Photosensitizing Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Graphite/chemistry , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Singlet Oxygen/analysis , Tumor Cells, Cultured
9.
Theranostics ; 6(5): 627-37, 2016.
Article in English | MEDLINE | ID: mdl-27022411

ABSTRACT

"Training" the host immune system to recognize and systemically eliminate residual tumor lesions and micrometastases is a promising strategy for cancer therapy. In this study, we investigated whether integrin αvß6-targeted photodynamic therapy (PDT) of tumors using a phthalocyanine dye-labeled probe (termed DSAB-HK) could trigger the host immune response, and whether PDT in combination with anti-PD-1 immune checkpoint inhibition could be used for the effective therapy of primary tumors and metastases. By near-infrared fluorescence imaging, DSAB-HK was demonstrated to specifically target either subcutaneous tumors in a 4T1 mouse breast cancer model or firefly luciferase stably transfected 4T1 (4T1-fLuc) lung metastatic tumors. Upon light irradiation, PDT by DSAB-HK significantly inhibited the growth of subcutaneous 4T1 tumors, and in addition promoted the maturation of dendritic cells and their production of cytokines, which subsequently stimulated the tumor recruitment of CD8(+) cytotoxic T lymphocytes. Furthermore, DSAB-HK PDT of the first tumor followed by PD-1 blockade markedly suppressed the growth of a second subcutaneous tumor, and also slowed the growth of 4T1-fLuc lung metastasis as demonstrated by serial bioluminescence imaging. Together, our results demonstrated the synergistic effect of tumor-targeted PDT and immune checkpoint inhibition for improving anti-tumor immunity and suppressing tumor growth/metastasis.


Subject(s)
Antigens, Neoplasm/metabolism , Breast Neoplasms/therapy , Immunologic Factors/administration & dosage , Indoles/administration & dosage , Integrins/metabolism , Lung Neoplasms/therapy , Photochemotherapy/methods , Radiation-Sensitizing Agents/administration & dosage , Animals , CD8-Positive T-Lymphocytes , Cytotoxicity, Immunologic , Disease Models, Animal , Isoindoles , Light , Lung Neoplasms/secondary , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Treatment Outcome
10.
Biomaterials ; 84: 1-12, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26803407

ABSTRACT

Tumor-associated macrophages (TAMs) play essential roles in tumor invasion and metastasis, and contribute to drug resistance. Clinical evidence suggests that TAM levels are correlated with local tumor relapse, distant metastasis, and poor prognosis in patients. In this study, we synthesized a TAM-targeted probe (IRD-αCD206) by conjugating a monoclonal anti-CD206 antibody with a near-infrared phthalocyanine dye. We then investigated the potential application of the IRD-αCD206 probe to near-infrared fluorescence (NIRF) imaging and photoimmunotherapy (PIT) of tumors resistant to treatment with the kinase inhibitor sorafenib. Sorafenib treatment had no effect on tumor growth in a 4T1 mouse model of breast cancer, but induced M2 macrophage polarization in tumors. M2 macrophage recruitment by sorafenib-treated 4T1 tumors was noninvasively visualized by in vivo NIRF imaging of IRD-αCD206. Small-animal single-photon emission computed tomography (SPECT)/CT and intratumoral microdistribution analysis indicated TAM-specific localization of the IRD-αCD206 probe in 4T1 tumors after several rounds of sorafenib treatment. Upon light irradiation, IRD-αCD206 suppressed the growth of sorafenib-resistant tumors. In vivo CT imaging and ex vivo histological analysis confirmed the inhibition of lung metastasis in mice by IRD-αCD206 PIT. These results demonstrate the utility of the IRD-αCD206 probe for TAM-targeted diagnostic imaging and treatment of tumors that are resistant to conventional therapeutics.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Immunotherapy/methods , Macrophages/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Phototherapy/methods , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , RAW 264.7 Cells , Sorafenib
11.
J Nucl Med ; 57(2): 321-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26383149

ABSTRACT

UNLABELLED: Noninvasive, real-time, quantitative measurement of key biomarkers associated with cancer therapeutic interventions could provide a better understanding of cancer biology. We investigated in this study whether incorporating multiple molecular imaging approaches could be used to guide dasatinib anti-Src therapy and aid in the rational design of a combination therapy regimen. METHODS: Bioluminescence imaging, (18)F-FDG PET, integrin αvß3-targeted SPECT/CT, and vascular endothelial growth factor-targeted near-infrared fluorescence imaging were performed before and after dasatinib treatment in a tumor mouse model. RESULTS: There was no significant difference in the bioluminescence imaging signal or (18)F-FDG tumor uptake in dasatinib-treated tumors compared with the control tumors. However, the uptake of (99m)T-3PRGD2 (integrin αvß3-specific) and DyLight755-ranibizumab (vascular endothelial growth factor-specific) in the dasatinib-treated tumors was significantly lower than that in the control tumors. In vitro studies confirmed the antiangiogenic effects of dasatinib but indicated a lack of cytotoxicity. Dasatinib plus cytotoxic docetaxel elicited marked synergistic tumor growth inhibition in vivo. CONCLUSION: Visualization of post-Src inhibition tumor signatures through multiple imaging approaches facilitates sensitive and quantitative measurement of cancer biomarkers in vivo, thus aiding in the rational design of dasatinib combination therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Dasatinib/therapeutic use , Genes, src/drug effects , Neoplasms/drug therapy , Animals , Drug Therapy, Combination , Fluorodeoxyglucose F18/pharmacokinetics , Integrin alphaVbeta3/drug effects , Luminescence , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Imaging , Radioligand Assay , Radiopharmaceuticals/pharmacokinetics , Ranibizumab/pharmacokinetics , Tomography, Emission-Computed, Single-Photon , Vascular Endothelial Growth Factor A/drug effects
12.
Radiology ; 279(2): 502-12, 2016 May.
Article in English | MEDLINE | ID: mdl-26669696

ABSTRACT

PURPOSE: To assess the potential utility of an integrin αvß3-targeting radiotracer, technetium 99m-PEG4-E[PEG4-cyclo(arginine-glycine-aspartic acid-D-phenylalanine-lysine)]2 ((99m)Tc-3PRGD2), for single photon emission computed tomography (SPECT)/computed tomography (CT) for monitoring of the progression and prognosis of liver fibrosis in a rat model. MATERIALS AND METHODS: All animal experiments were performed by following the protocol approved by the institutional animal care and use committee. (99m)Tc-3PRGD2 was prepared and longitudinal SPECT/CT was performed to monitor the progression (n = 8) and recovery (n = 5) of liver fibrosis induced in a rat model by means of thioacetamide (TAA) administration. The mean liver-to-background radioactivity per unit volume ratio was analyzed for comparisons between the TAA and control (saline) groups at different stages of liver fibrosis. Data were compared by using Student t and Mann-Whitney tests. Results:of SPECT/CT were compared with those of ex vivo biodistribution analysis (n = 5). RESULTS: Accumulation of (99m)Tc-3PRGD2 in the liver increased in proportion to the progression of fibrosis and TAA exposure time; accumulation levels were significantly different between the TAA and control groups as early as week 4 of TAA administration (liver-to-background ratio: 32.30 ± 3.39 vs 19.01 ± 3.31; P = .0002). Results of ex vivo immunofluorescence staining demonstrated the positive expression of integrin αvß3 on the activated hepatic stellate cells, and the integrin αvß3 levels in the liver corresponded to the results of SPECT/CT (R(2) = 0.75, P < .0001). (99m)Tc-3PRGD2 uptake in the fibrotic liver decreased after antifibrotic therapy with interferon α2b compared with that in the control group (relative liver-to-background ratio: 0.45 ± 0.05 vs 1.01 ± 0.05; P < .0001) or spontaneous recovery (relative liver-to-background ratio: 0.56 ± 0.06 vs 1.01 ± 0.05; P < .0001). CONCLUSION: (99m)Tc-3PRGD2 SPECT/CT was successfully used to monitor the progression and recovery of liver fibrosis and shows potential applications for noninvasive diagnosis of early stage liver fibrosis.


Subject(s)
Integrin alphaVbeta3/metabolism , Liver Cirrhosis/metabolism , Multimodal Imaging , Radiopharmaceuticals/pharmacokinetics , Sodium Pertechnetate Tc 99m/pharmacokinetics , Tomography, Emission-Computed, Single-Photon , Tomography, Spiral Computed , Animals , Disease Models, Animal , Imaging, Three-Dimensional , Liver Cirrhosis/diagnostic imaging , Male , Rats , Rats, Wistar
13.
Curr Drug Targets ; 16(6): 634-44, 2015.
Article in English | MEDLINE | ID: mdl-26028045

ABSTRACT

Cancer-targeted radionuclide therapy is a promising approach for the treatment of a wide variety of malignancies, especially those resistant to conventional therapies. However, to improve the use of targeted radionuclide therapy for the management of cancer patients, the in vivo behaviors, dosimetry, and efficacy of radiotherapeutic agents need to be well characterized and monitored. Molecular imaging, which is a powerful tool for the noninvasive characterization and quantification of biological processes in living subjects at the cellular and molecular levels, plays an important role in the guidance of cancer radionuclide therapy. In this review, we introduce the radiotherapeutics for cancer-targeted therapy and summarize the most recent evidence supporting the use of molecular imaging to guide cancer radionuclide therapy.


Subject(s)
Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Radiotherapy, Image-Guided/methods , Chelating Agents/chemistry , Drug Delivery Systems , Humans , Molecular Imaging/methods , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use
14.
Biomaterials ; 53: 229-38, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25890722

ABSTRACT

Integrin αvß6 is widely upregulated in variant malignant cancers but is undetectable in normal organs, making it a promising target for cancer diagnostic imaging and therapy. Using streptavidin-biotin chemistry, we synthesized an integrin αvß6-targeted near-infrared phthalocyanine dye-labeled agent, termed Dye-SA-B-HK, and investigated whether it could be used for cancer imaging, optical imaging-guided surgery, and phototherapy in pancreatic cancer mouse models. Dye-SA-B-HK specifically bound to integrin αvß6 in vitro and in vivo with high receptor binding affinity. Using small-animal optical imaging, we detected subcutaneous and orthotopic BxPC-3 human pancreatic cancer xenografts in vivo. Upon optical image-guidance, the orthotopically growing pancreatic cancer lesions could be successfully removed by surgery. Using light irradiation, Dye-SA-B-HK manifested remarkable antitumor effects both in vitro and in vivo. (18)F-FDG positron emission tomography (PET) imaging and ex vivo fluorescence staining validated the observed decrease in proliferation of treated tumors by Dye-DA-B-HK phototherapy. Tissue microarray results revealed overexpression of integrin αvß6 in over 95% cases of human pancreatic cancer, indicating that theranostic application of Dye-DA-B-HK has clear translational potential. Overall, the results of this study demonstrated that integrin αvß6-specific Dye-SA-B-HK is a promising theranostic agent for the management of pancreatic cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Indoles/administration & dosage , Integrins/metabolism , Pancreatic Neoplasms/therapy , Theranostic Nanomedicine , Animals , Coloring Agents/administration & dosage , Heterografts , Humans , Isoindoles , Male , Mice , Mice, Inbred BALB C , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology
15.
Theranostics ; 5(6): 597-608, 2015.
Article in English | MEDLINE | ID: mdl-25825599

ABSTRACT

Significant evidence has indicated that tumor-associated macrophages (TAMs) play a critical role in the proliferation, invasion, angiogenesis, and metastasis of a variety of human carcinomas. In this study, we investigated whether near-infrared fluorescence (NIRF) imaging using a macrophage mannose receptor (MMR; CD206)-targeting agent could be used to noninvasively visualize and quantify changes in TAMs in vivo. The CD206-targeting NIRF agent, Dye-anti-CD206, was prepared and characterized in vitro and in vivo. By using NIRF imaging, we were able to noninvasively image tumor-infiltrating macrophages in the 4T1 mouse breast cancer model. Importantly, longitudinal NIRF imaging revealed the depletion of macrophages in response to zoledronic acid (ZA) treatment. However, ZA alone did not lead to the inhibition of 4T1 tumor growth. We therefore combined anti-macrophage ZA therapy and tumor cytotoxic docetaxel (DTX) therapy in the mouse model. The results demonstrated that this combination strategy could significantly inhibit tumor growth as well as tumor metastasis to the lungs. Based on these findings, we concluded that CD206-targeted molecular imaging can sensitively detect the dynamic changes in tumor-infiltrating macrophages, and that the combination of macrophage depletion and cytotoxic therapy is a promising strategy for the effective treatment of solid tumors.


Subject(s)
Macrophages/pathology , Mammary Neoplasms, Experimental/pathology , Spectroscopy, Near-Infrared/methods , Animals , Antineoplastic Agents/therapeutic use , Cell Line , Cell Line, Tumor , Diphosphonates/therapeutic use , Docetaxel , Female , Imidazoles/therapeutic use , Lectins, C-Type/metabolism , Lung Neoplasms/secondary , Macrophages/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/metabolism , Sensitivity and Specificity , Taxoids/therapeutic use , Zoledronic Acid
16.
Mol Pharm ; 12(1): 10-7, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25397907

ABSTRACT

Cetuximab is an antiepidermal growth factor receptor (EGFR) monoclonal antibody that has received the approval of the Food and Drug Administration (FDA) for cancer treatment. However, most clinical studies indicate that cetuximab can only elicit positive effects on a subset of cancer patients. In this study, we investigated whether near-infrared fluorescence (NIRF) imaging of tumor vascular endothelial growth factor (VEGF) expression could be a biomarker for tumor early response to cetuximab therapy in preclinical wild-type and mutant tumor models of the KRAS gene. The treatment efficacy of cetuximab was determined in both HT-29 (wild-type KRAS) and HTC-116 (mutant KRAS) human colon cancer models. A VEGF-specific optical imaging probe (Dye755-Ran) was synthesized by conjugating ranibizumab (an anti-VEGF antibody Fab fragment) with a NIRF dye. Serial optical scans with Dye755-Ran were performed in HT-29 and HTC-116 xenograft models. By using longitudinal NIRF imaging, we were able to detect early tumor response on day 3 and day 5 after initiation of cetuximab treatment in the cetuximab-responsive HT-29 tumor model. Enzyme-linked immunosorbent assay (ELISA) confirmed that cetuximab treatment inhibited human VEGF expression in the KRAS wild-type HT-29 tumor but not in the KRAS mutant HCT-116 tumor. We have demonstrated that the antitumor effect of cetuximab can be noninvasively monitored by serial fluorescence imaging using Dye755-Ran. VEGF expression detected by optical imaging could serve as a sensitive biomarker for tumor early response to drugs that directly or indirectly act on VEGF.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemistry , Fluorescent Dyes/chemistry , Immunotherapy/methods , Neoplasms/drug therapy , Animals , Biomarkers/metabolism , Cell Line, Tumor , Cetuximab , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/pathology , Neovascularization, Pathologic , Ranibizumab , Vascular Endothelial Growth Factor A/metabolism
17.
Cancer Res ; 74(20): 5711-22, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25261236

ABSTRACT

Immune escape is a fundamental trait of cancer. Dendritic cells (DC) that interact with T cells represent a crucial site for the development of tolerance to tumor antigens, but there remains incomplete knowledge about how DC-tolerizing signals evolve during tumorigenesis. In this study, we show that DCs isolated from patients with metastatic or locally advanced breast cancer express high levels of the adiponectin receptors AdipoR1 and AdipoR2, which are sufficient to blunt antitumor immunity. Mechanistic investigations of ligand-receptor interactions on DCs revealed novel signaling pathways for each receptor. AdipoR1 stimulated IL10 production by activating the AMPK and MAPKp38 pathways, whereas AdipoR2 modified inflammatory processes by activating the COX-2 and PPARγ pathways. Stimulation of these pathways was sufficient to block activation of NF-κB in DC, thereby attenuating their ability to stimulate antigen-specific T-cell responses. Together, our findings reveal novel insights into how DC-tolerizing signals evolve in cancer to promote immune escape. Furthermore, by defining a critical role for adiponectin signaling in this process, our work suggests new and broadly applicable strategies for immunometabolic therapy in patients with cancer.


Subject(s)
Breast Neoplasms/immunology , Dendritic Cells/metabolism , Receptors, Adiponectin/metabolism , Tumor Escape , Adiponectin/physiology , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Clonal Anergy , Cyclooxygenase 2/metabolism , Cytotoxicity, Immunologic , Disease Progression , Enzyme Activation , Female , Humans , Interleukin-10/metabolism , MAP Kinase Signaling System , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasm Transplantation , PPAR gamma/metabolism , T-Lymphocytes, Cytotoxic/immunology
18.
J Nucl Med ; 55(6): 1002-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24732154

ABSTRACT

UNLABELLED: Previous in vitro studies demonstrated that treating tumors expressing both epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 with trastuzumab resulted in increased EGFR homodimerization and subsequent rapid downregulation of EGFR. We investigated whether molecular imaging using near-infrared fluorescence (NIRF) imaging and PET probes could sensitively detect trastuzumab-induced EGFR downregulation in vivo. METHODS: The F(ab')2 antibody fragment PaniF(ab')2 was generated by digesting the anti-EGFR monoclonal antibody panitumumab. PaniF(ab')2 was labeled with either a NIRF dye or (68)Ga, and optical imaging and small-animal PET imaging of Dye-PaniF(ab')2 and (68)Ga-PaniF(ab')2, respectively, were performed in HT-29 tumor-bearing nude mice treated with trastuzumab or untreated control. RESULTS: Longitudinal NIRF imaging studies revealed significantly reduced tumor uptake of Dye-PaniF(ab')2 on days 5 and 7 in trastuzumab-treated HT-29 tumors, compared with control. Western blotting confirmed the downregulation of EGFR after treatment with trastuzumab. Small-animal PET on day 5 after trastuzumab treatment also demonstrated decreased (68)Ga-PaniF(ab')2 uptake in trastuzumab-treated HT-29 tumors. The tumor uptake value of (68)Ga-PaniF(ab')2 obtained from PET imaging had an excellent linear correlation with the uptake value measured using biodistribution. CONCLUSION: The downregulation of EGFR induced by trastuzumab treatment could be detected noninvasively using optical and PET imaging. This molecular imaging strategy could provide a dynamic readout of changes in the tumor signaling and may facilitate the noninvasive monitoring of the early tumor response to drug treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Down-Regulation/drug effects , ErbB Receptors/metabolism , Optical Imaging , Positron-Emission Tomography , Animals , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Coloring Agents/chemistry , Gallium Radioisotopes , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Mice , Mice, Nude , Panitumumab , Receptor, ErbB-2/metabolism , Trastuzumab
19.
Oncoimmunology ; 2(1): e22590, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23483821

ABSTRACT

In this study, we generated human MHC Class I-restricted CD4+ T cells specific for Epstein-Barr virus (EBV) and cytomegalovirus (CMV), two herpesviridae associated with lymphoma, nasopharyngeal carcinoma and medulloblastoma, respectively. Retroviral transfer of virus-specific, HLA-A2-restricted TCR-coding genes generated CD4+ T cells that recognized HLA-A2/peptide multimers and produced cytokines when stimulated with MHC Class II-deficient cells presenting the relevant viral peptides in the context of HLA-A2. Peptide titration revealed that CD4+ T cells had a 10-fold lower avidity than CD8+ T cells expressing the same TCR. The impaired avidity of CD4+ T cells was corrected by simultaneously transferring TCR- and CD8-coding genes. The CD8 co-receptor did not alter the cytokine signature of CD4+ T cells, which remained distinct from that of CD8+ T cells. Using the xenogeneic NOD/SCID mouse model, we demonstrated that human CD4+ T cells expressing a specific TCR and CD8 can confer efficient protection against the growth of tumors expressing the EBV or CMV antigens recognized by the TCR. In summary, we describe a robust approach for generating therapeutic CD4+ T cells capable of providing MHC Class I-restricted immunity against MHC Class II-negative tumors in vivo.

20.
Haematologica ; 95(1): 126-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19679884

ABSTRACT

BACKGROUND: The Wilms' tumor antigen (WT1) is an attractive target for immunotherapy of leukemia. In the past, we isolated and characterized the specificity and function of a WT1-specific T-cell receptor. The goal of this translational study was to develop a safe and efficient WT1-T-cell receptor retroviral vector for an adoptive immunotherapy trial with engineered T cells. DESIGN AND METHODS: We generated a panel of retroviral constructs containing unmodified or codon-optimized WT1-T-cell receptor alpha and beta genes, linked via internal ribosome entry sites or 2A sequences, with or without an additional inter-chain disulfide bond in the T-cell receptor constant domains. These constructs were functionally analyzed in vitro, and the best one was tested in an autologous primary leukemia model in vivo. RESULTS: We identified a WT1-T-cell receptor construct that showed optimal tetramer staining, antigen-specific cytokine production and killing activity when introduced into primary human T cells. Fresh CD34(+) cells purified from a patient with leukemia were engrafted into NOD/SCID mice, followed by adoptive immunotherapy with patient's autologous T cells transduced with the WT1-T-cell receptor. This therapeutic treatment evidently decreased leukemia engraftment in mice and resulted in a substantial improvement of leukemia-free survival. CONCLUSIONS: This is the first report that patient's T cells, engineered to express the WT1-T-cell receptor, can eliminate autologous leukemia progenitor cells in an in vivo model. This study provides a firm basis for the planned WT1-T-cell receptor gene therapy trial in leukemia patients.


Subject(s)
Antigens, Neoplasm/immunology , Blast Crisis/immunology , Genetic Engineering/methods , Leukemia/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/pathology , Wilms Tumor/pathology , Adult , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blast Crisis/genetics , Blast Crisis/therapy , Genetic Therapy/methods , Genetic Vectors/biosynthesis , Genetic Vectors/chemistry , Hepatitis B Virus, Woodchuck/genetics , Humans , Jurkat Cells , Leukemia/genetics , Leukemia/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Autologous/immunology , Wilms Tumor/immunology , Wilms Tumor/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...