Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Sci Rep ; 11(1): 1399, 2021 01 14.
Article En | MEDLINE | ID: mdl-33446805

SHP2 is a ubiquitous tyrosine phosphatase involved in regulating both tumor and immune cell signaling. In this study, we discovered a novel immune modulatory function of SHP2. Targeting this protein with allosteric SHP2 inhibitors promoted anti-tumor immunity, including enhancing T cell cytotoxic function and immune-mediated tumor regression. Knockout of SHP2 using CRISPR/Cas9 gene editing showed that targeting SHP2 in cancer cells contributes to this immune response. Inhibition of SHP2 activity augmented tumor intrinsic IFNγ signaling resulting in enhanced chemoattractant cytokine release and cytotoxic T cell recruitment, as well as increased expression of MHC Class I and PD-L1 on the cancer cell surface. Furthermore, SHP2 inhibition diminished the differentiation and inhibitory function of immune suppressive myeloid cells in the tumor microenvironment. SHP2 inhibition enhanced responses to anti-PD-1 blockade in syngeneic mouse models. Overall, our study reveals novel functions of SHP2 in tumor immunity and proposes that targeting SHP2 is a promising strategy for cancer immunotherapy.


Immunity, Cellular , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction/genetics
2.
Nat Commun ; 11(1): 6315, 2020 12 09.
Article En | MEDLINE | ID: mdl-33298926

Despite the increasing interest in targeting stromal elements of the tumor microenvironment, we still face tremendous challenges in developing adequate therapeutics to modify the tumor stromal landscape. A major obstacle to this is our poor understanding of the phenotypic and functional heterogeneity of stromal cells in tumors. Herein, we perform an unbiased interrogation of tumor mesenchymal cells, delineating the co-existence of distinct subsets of cancer-associated fibroblasts (CAFs) in the microenvironment of murine carcinomas, each endowed with unique phenotypic features and functions. Furthermore, our study shows that neutralization of TGFß in vivo leads to remodeling of CAF dynamics, greatly reducing the frequency and activity of the myofibroblast subset, while promoting the formation of a fibroblast population characterized by strong response to interferon and heightened immunomodulatory properties. These changes correlate with the development of productive anti-tumor immunity and greater efficacy of PD1 immunotherapy. Along with providing the scientific rationale for the evaluation of TGFß and PD1 co-blockade in the clinical setting, this study also supports the concept of plasticity of the stromal cell landscape in tumors, laying the foundation for future investigations aimed at defining pathways and molecules to program CAF composition for cancer therapy.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cancer-Associated Fibroblasts/immunology , Carcinoma/drug therapy , Interferon-beta/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer-Associated Fibroblasts/drug effects , Carcinoma/immunology , Carcinoma/pathology , Cell Line, Tumor/transplantation , Cell Plasticity/drug effects , Cell Plasticity/immunology , Disease Models, Animal , Drug Synergism , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Stromal Cells/drug effects , Stromal Cells/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
Proc Natl Acad Sci U S A ; 116(7): 2662-2671, 2019 02 12.
Article En | MEDLINE | ID: mdl-30700544

Cooption of the host vasculature is a strategy that some cancers use to sustain tumor progression without-or before-angiogenesis or in response to antiangiogenic therapy. Facilitated by certain growth factors, cooption can mediate tumor infiltration and confer resistance to antiangiogenic drugs. Unfortunately, this mode of tumor progression is difficult to target because the underlying mechanisms are not fully understood. Here, we analyzed the dynamics of vessel cooption during tumor progression and in response to antiangiogenic treatment in gliomas and brain metastases. We followed tumor evolution during escape from antiangiogenic treatment as cancer cells coopted, and apparently mechanically compressed, host vessels. To gain deeper understanding, we developed a mathematical model, which incorporated compression of coopted vessels, resulting in hypoxia and formation of new vessels by angiogenesis. Even if antiangiogenic therapy can block such secondary angiogenesis, the tumor can sustain itself by coopting existing vessels. Hence, tumor progression can only be stopped by combination therapies that judiciously block both angiogenesis and cooption. Furthermore, the model suggests that sequential blockade is likely to be more beneficial than simultaneous blockade.


Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/pathology , Angiogenesis Inhibitors/therapeutic use , Angiopoietin-2/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Microscopy/methods , Neoplasm Invasiveness , Neovascularization, Pathologic/prevention & control , Oxygen/metabolism , Rats , Reproducibility of Results , Vascular Endothelial Growth Factor A/metabolism
4.
J Cell Biol ; 217(6): 1941-1955, 2018 06 04.
Article En | MEDLINE | ID: mdl-29588376

The multiciliated cell (MCC) is an evolutionarily conserved cell type, which in vertebrates functions to promote directional fluid flow across epithelial tissues. In the conducting airway, MCCs are generated by basal stem/progenitor cells and act in concert with secretory cells to perform mucociliary clearance to expel pathogens from the lung. Studies in multiple systems, including Xenopus laevis epidermis, murine trachea, and zebrafish kidney, have uncovered a transcriptional network that regulates multiple steps of multiciliogenesis, ultimately leading to an MCC with hundreds of motile cilia extended from their apical surface, which beat in a coordinated fashion. Here, we used a pool-based short hairpin RNA screening approach and identified TRRAP, an essential component of multiple histone acetyltransferase complexes, as a central regulator of MCC formation. Using a combination of immunofluorescence, signaling pathway modulation, and genomic approaches, we show that (a) TRRAP acts downstream of the Notch2-mediated basal progenitor cell fate decision and upstream of Multicilin to control MCC differentiation; and (b) TRRAP binds to the promoters and regulates the expression of a network of genes involved in MCC differentiation and function, including several genes associated with human ciliopathies.


Adaptor Proteins, Signal Transducing/metabolism , Cilia/metabolism , Nuclear Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Lineage , Epigenesis, Genetic , Epithelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Lung/cytology , RNA, Small Interfering/metabolism , Receptor, Notch2 , Signal Transduction , Transcription Factors
5.
Proc Natl Acad Sci U S A ; 113(16): 4470-5, 2016 Apr 19.
Article En | MEDLINE | ID: mdl-27044097

Glioblastomas (GBMs) rapidly become refractory to anti-VEGF therapies. We previously demonstrated that ectopic overexpression of angiopoietin-2 (Ang-2) compromises the benefits of anti-VEGF receptor (VEGFR) treatment in murine GBM models and that circulating Ang-2 levels in GBM patients rebound after an initial decrease following cediranib (a pan-VEGFR tyrosine kinase inhibitor) administration. Here we tested whether dual inhibition of VEGFR/Ang-2 could improve survival in two orthotopic models of GBM, Gl261 and U87. Dual therapy using cediranib and MEDI3617 (an anti-Ang-2-neutralizing antibody) improved survival over each therapy alone by delaying Gl261 growth and increasing U87 necrosis, effectively reducing viable tumor burden. Consistent with their vascular-modulating function, the dual therapies enhanced morphological normalization of vessels. Dual therapy also led to changes in tumor-associated macrophages (TAMs). Inhibition of TAM recruitment using an anti-colony-stimulating factor-1 antibody compromised the survival benefit of dual therapy. Thus, dual inhibition of VEGFR/Ang-2 prolongs survival in preclinical GBM models by reducing tumor burden, improving normalization, and altering TAMs. This approach may represent a potential therapeutic strategy to overcome the limitations of anti-VEGFR monotherapy in GBM patients by integrating the complementary effects of anti-Ang2 treatment on vessels and immune cells.


Antibodies, Neoplasm/pharmacology , Glioblastoma , Macrophages , Neoplasm Proteins , Neoplasms, Experimental , Neovascularization, Pathologic , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor , Ribonuclease, Pancreatic , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Ribonuclease, Pancreatic/antagonists & inhibitors , Ribonuclease, Pancreatic/metabolism
6.
Proc Natl Acad Sci U S A ; 113(16): 4476-81, 2016 Apr 19.
Article En | MEDLINE | ID: mdl-27044098

Inhibition of the vascular endothelial growth factor (VEGF) pathway has failed to improve overall survival of patients with glioblastoma (GBM). We previously showed that angiopoietin-2 (Ang-2) overexpression compromised the benefit from anti-VEGF therapy in a preclinical GBM model. Here we investigated whether dual Ang-2/VEGF inhibition could overcome resistance to anti-VEGF treatment. We treated mice bearing orthotopic syngeneic (Gl261) GBMs or human (MGG8) GBM xenografts with antibodies inhibiting VEGF (B20), or Ang-2/VEGF (CrossMab, A2V). We examined the effects of treatment on the tumor vasculature, immune cell populations, tumor growth, and survival in both the Gl261 and MGG8 tumor models. We found that in the Gl261 model, which displays a highly abnormal tumor vasculature, A2V decreased vessel density, delayed tumor growth, and prolonged survival compared with B20. In the MGG8 model, which displays a low degree of vessel abnormality, A2V induced no significant changes in the tumor vasculature but still prolonged survival. In both the Gl261 and MGG8 models A2V reprogrammed protumor M2 macrophages toward the antitumor M1 phenotype. Our findings indicate that A2V may prolong survival in mice with GBM by reprogramming the tumor immune microenvironment and delaying tumor growth.


Antibodies, Bispecific/pharmacology , Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Macrophages/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Ribonuclease, Pancreatic/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vesicular Transport Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Macrophages/pathology , Mice , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Ribonuclease, Pancreatic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vesicular Transport Proteins/metabolism , Xenograft Model Antitumor Assays
7.
J Natl Cancer Inst ; 107(4)2015 Apr.
Article En | MEDLINE | ID: mdl-25710962

BACKGROUND: Matrix metalloproteinase (MMP) 14 may mediate tumor progression through vascular and immune-modulatory effects. METHODS: Orthotopic murine breast tumors (4T1 and E0771 with high and low MMP14 expression, respectively; n = 5-10 per group) were treated with an anti-MMP14 inhibitory antibody (DX-2400), IgG control, fractionated radiation therapy, or their combination. We assessed primary tumor growth, transforming growth factor ß (TGFß) and inducible nitric oxide synthase (iNOS) expression, macrophage phenotype, and vascular parameters. A linear mixed model with repeated observations, with Mann-Whitney or analysis of variance with Bonferroni post hoc adjustment, was used to determine statistical significance. All statistical tests were two-sided. RESULTS: DX-2400 inhibited tumor growth compared with IgG control treatment, increased macrophage numbers, and shifted the macrophage phenotype towards antitumor M1-like. These effects were associated with a reduction in active TGFß and SMAD2/3 signaling. DX-2400 also transiently increased iNOS expression and tumor perfusion, reduced tissue hypoxia (median % area: control, 20.2%, interquartile range (IQR) = 6.4%-38.9%; DX-2400: 1.2%, IQR = 0.2%-3.2%, P = .044), and synergistically enhanced radiation therapy (days to grow to 800mm(3): control, 12 days, IQR = 9-13 days; DX-2400 plus radiation, 29 days, IQR = 26-30 days, P < .001) in the 4T1 model. The selective iNOS inhibitor, 1400W, abolished the effects of DX-2400 on vessel perfusion and radiotherapy. On the other hand, DX-2400 was not capable of inducing iNOS expression or synergizing with radiation in E0771 tumors. CONCLUSION: MMP14 blockade decreased immunosuppressive TGFß, polarized macrophages to an antitumor phenotype, increased iNOS, and improved tumor perfusion, resulting in reduced primary tumor growth and enhanced response to radiation therapy, especially in high MMP14-expressing tumors.


Amidines/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Benzylamines/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Macrophages/drug effects , Matrix Metalloproteinase 14/drug effects , Matrix Metalloproteinase 14/metabolism , Nitric Oxide Synthase Type II/drug effects , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Line, Tumor , Dose Fractionation, Radiation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin G/blood , Macrophages/enzymology , Mammary Neoplasms, Experimental , Mice , Neovascularization, Pathologic , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phenotype , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
8.
J Natl Cancer Inst ; 105(16): 1188-201, 2013 Aug 21.
Article En | MEDLINE | ID: mdl-23899555

BACKGROUND: The solid tumor microvasculature is characterized by structural and functional abnormality and mediates several deleterious aspects of tumor behavior. Here we determine the role of vascular endothelial protein tyrosine phosphatase (VE-PTP), which deactivates endothelial cell (EC) Tie-2 receptor tyrosine kinase, thereby impairing maturation of tumor vessels. METHODS: AKB-9778 is a first-in-class VE-PTP inhibitor. We examined its effects on ECs in vitro and on embryonic angiogenesis in vivo using zebrafish assays. We studied the impact of AKB-9778 therapy on the tumor vasculature, tumor growth, and metastatic progression using orthotopic models of murine mammary carcinoma as well as spontaneous and experimental metastasis models. Finally, we used endothelial nitric oxide synthase (eNOS)-deficient mice to establish the role of eNOS in mediating the effects of VE-PTP inhibition. All statistical tests were two-sided. RESULTS: AKB-9778 induced ligand-independent Tie-2 activation in ECs and impaired embryonic zebrafish angiogenesis. AKB-9778 delayed the early phase of mammary tumor growth by maintaining vascular maturity (P < .01, t test); slowed growth of micrometastases (P < .01, χ(2) test) by preventing extravasation of tumor cells (P < 0.01, Fisher exact test), resulting in a trend toward prolonged survival (27.0 vs 36.5 days; hazard ratio of death = 0.33, 95% confidence interval = 0.11 to 1.03; P = .05, Mantel-Cox test); and stabilized established primary tumor blood vessels, enhancing tumor perfusion (P = .03 for 4T1 tumor model and 0.05 for E0771 tumor model, by two-sided t tests) and, hence, radiation response (P < .01, analysis of variance; n = 7 mice per group). The effects of AKB-9778 on tumor vessels were mediated in part by endothelial nitric oxide synthase activation. CONCLUSIONS: Our results demonstrate that pharmacological VE-PTP inhibition can normalize the structure and function of tumor vessels through Tie-2 activation, which delays tumor growth, slows metastatic progression, and enhances response to concomitant cytotoxic treatments.


Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Lung Neoplasms/prevention & control , Neovascularization, Pathologic/drug therapy , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Zebrafish Proteins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Breast Neoplasms/blood supply , Disease Progression , Drug Synergism , Enzyme Activation/drug effects , Female , Human Umbilical Vein Endothelial Cells , Lung Neoplasms/secondary , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Receptor, TIE-2/metabolism , Xenograft Model Antitumor Assays , Zebrafish
9.
Neuro Oncol ; 15(8): 1079-87, 2013 Aug.
Article En | MEDLINE | ID: mdl-23828240

Antiangiogenic therapy is associated with increased radiographic responses in glioblastomas, but tumors invariably recur. Because tumor-associated macrophages have been shown to mediate escape from antiangiogenic therapy in preclinical models, we examined the role of macrophages in patients with recurrent glioblastoma. We compared autopsy brain specimens from 20 patients with recurrent glioblastoma who received antiangiogenic treatment and chemoradiation with 8 patients who received chemotherapy and/or radiotherapy without antiangiogenic therapy or no treatment. Tumor-associated macrophages were morphologically and phenotypically analyzed using flow cytometry and immunohistochemistry for CD68, CD14, CD163, and CD11b expression. Flow cytometry showed an increase in macrophages in the antiangiogenic-treated patients. Immunohistochemical analysis demonstrated an increase in CD68+ macrophages in the tumor bulk (P < .01) and infiltrative areas (P = .02) in antiangiogenic-treated patients. We also observed an increase in CD11b+ cells in the tumor bulk (P < .01) and an increase in CD163+ macrophages in infiltrative tumor (P = .02). Of note, an increased number of CD11b+ cells in bulk and infiltrative tumors (P = .05 and P = .05, respectively) correlated with poor overall survival among patients who first received antiangiogenic therapy at recurrence. In summary, recurrent glioblastomas showed an increased infiltration in myeloid populations in the tumor bulk and in the infiltrative regions after antiangiogenic therapy. Higher numbers of CD11b+ cells correlated with poor survival among these patients. These data suggest that tumor-associated macrophages may participate in escape from antiangiogenic therapy and may represent a potential biomarker of resistance and a potential therapeutic target in recurrent glioblastoma.


Angiogenesis Inhibitors/therapeutic use , Biomarkers, Tumor/metabolism , Brain Neoplasms/mortality , Glioblastoma/mortality , Macrophages/pathology , Neoplasm Recurrence, Local/mortality , Tumor Microenvironment/drug effects , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Autopsy , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Female , Flow Cytometry , Follow-Up Studies , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Macrophages/drug effects , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Prognosis , Survival Rate , Young Adult
10.
Cell ; 152(5): 1065-76, 2013 Feb 28.
Article En | MEDLINE | ID: mdl-23452854

Medulloblastoma is the most common pediatric malignant brain tumor. Although current therapies improve survival, these regimens are highly toxic and are associated with significant morbidity. Here, we report that placental growth factor (PlGF) is expressed in the majority of medulloblastomas, independent of their subtype. Moreover, high expression of PlGF receptor neuropilin 1 (Nrp1) correlates with poor overall survival in patients. We demonstrate that PlGF and Nrp1 are required for the growth and spread of medulloblastoma: PlGF/Nrp1 blockade results in direct antitumor effects in vivo, resulting in medulloblastoma regression, decreased metastasis, and increased mouse survival. We reveal that PlGF is produced in the cerebellar stroma via tumor-derived Sonic hedgehog (Shh) and show that PlGF acts through Nrp1-and not vascular endothelial growth factor receptor 1-to promote tumor cell survival. This critical tumor-stroma interaction-mediated by Shh, PlGF, and Nrp1 across medulloblastoma subtypes-supports the development of therapies targeting PlGF/Nrp1 pathway.


Cerebellar Neoplasms/pathology , Cerebellum/metabolism , Medulloblastoma/pathology , Neuropilin-1/metabolism , Pregnancy Proteins/metabolism , Signal Transduction , Animals , Cells, Cultured , Cerebellar Neoplasms/metabolism , Humans , Medulloblastoma/metabolism , Mice , Mice, Knockout , Neoplasm Transplantation , Paracrine Communication , Placenta Growth Factor , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-1/metabolism
11.
Neoplasia ; 14(2): 84-94, 2012 Feb.
Article En | MEDLINE | ID: mdl-22431917

Neurofibromatosis type 2 (NF2) is an autosomal-dominant multiple neoplasia syndrome that results from mutations in the NF2 tumor suppressor gene. Patients with NF2 develop hallmark schwannomas that require surgery or radiation, both of which have significant adverse effects. Recent studies have indicated that the tumor microenvironment-in particular, tumor blood vessels-of schwannomas may be an important therapeutic target. Furthermore, although much has been done to understand how merlin, the NF2 gene product, functions as a tumor suppressor gene in schwannoma cells, the functional role of merlin in the tumor microenvironment and the mechanism(s) by which merlin regulates angiogenesis to support schwannoma growth is largely unexplored. Here we report that the expression of semaphorin 3F (SEMA3F) was specifically downregulated in schwannoma cells lacking merlin/NF2. When we reintroduced SEMA3F in schwannoma cells, we observed normalized tumor blood vessels, reduced tumor burden, and extended survival in nude mice bearing merlin-deficient brain tumors. Next, using chemical inhibitors and gene knockdown with RNA interference, we found that merlin regulated expression of SEMA3F through Rho GTPase family member Rac1. This study shows that, in addition to the tumor-suppressing activity of merlin, it also functions to maintain physiological angiogenesis in the nervous system by regulating antiangiogenic factors such as SEMA3F. Restoring the relative balance of proangiogenic and antiangiogenic factors, such as increases in SEMA3F, in schwannoma microenvironment may represent a novel strategy to alleviate the clinical symptoms of NF2-related schwannomas.


Brain Neoplasms/blood supply , Membrane Proteins/metabolism , Neovascularization, Pathologic , Nerve Tissue Proteins/metabolism , Neurilemmoma/blood supply , Neurofibromin 2/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Blood Vessels/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Kaplan-Meier Estimate , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Nerve Tissue Proteins/genetics , Neurilemmoma/metabolism , Neurilemmoma/pathology , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2/genetics , Permeability , Signal Transduction , Thrombospondins/genetics , Thrombospondins/metabolism
12.
Intravital ; 1(1)2012.
Article En | MEDLINE | ID: mdl-24353926

The abnormal tumor microenvironment fuels tumor progression, metastasis, immune suppression, and treatment resistance. Over last several decades, developments in and applications of intravital microscopy have provided unprecedented insights into the dynamics of the tumor microenvironment. In particular, intravital multiphoton microscopy has revealed the abnormal structure and function of tumor-associated blood and lymphatic vessels, the role of aberrant tumor matrix in drug delivery, invasion and metastasis of tumor cells, the dynamics of immune cell trafficking to and within tumors, and gene expression in tumors. However, traditional multiphoton microscopy suffers from inherently slow imaging rates-only a few frames per second, thus unable to capture more rapid events such as blood flow, lymphatic flow, and cell movement within vessels. Here, we report the development and implementation of a video-rate multiphoton microscope (VR-MPLSM) based on resonant galvanometer mirror scanning that is capable of recording at 30 frames per second and acquiring intravital multispectral images. We show that the design of the system can be readily implemented and is adaptable to various experimental models. As examples, we demonstrate the utility of the system to directly measure flow within tumors, capture metastatic cancer cells moving within the brain vasculature and cells in lymphatic vessels, and image acute responses to changes in a vascular network. VR-MPLSM thus has the potential to further advance intravital imaging and provide new insight into the biology of the tumor microenvironment.

13.
Cancer Res ; 71(13): 4527-38, 2011 Jul 01.
Article En | MEDLINE | ID: mdl-21571860

Functional roles for the cancer cell-associated membrane type I matrix metalloproteinase (MT1-MMP) during early steps of the metastatic cascade in primary tumors remain unresolved. In an effort to determine its significance, we determined the in vivo effects of RNAi-mediated downregulation in mammary cancer cells on the migration, blood and lymphatic vessel invasion (LVI), and lymph node and lung metastasis. We also correlated the expression of cancer cell MT1-MMP with blood vessel invasion (BVI) in 102 breast cancer biopsies. MT1-MMP downregulation in cancer cells decreased lung metastasis without affecting primary tumor growth. The inhibition of lung metastasis correlated with reduced cancer cell migration and BVI. Furthermore, cancer cell-expressed MT1-MMP upregulated the expression of MT1-MMP in vascular endothelial cells, but did not affect MT1-MMP expression in lymphatic endothelial cells, LVI, or lymph node metastasis. Of clinical importance, we observed that elevated MT1-MMP expression correlated with BVI in biopsies from triple-negative breast cancers (TNBC), which have a poor prognosis and high incidence of distant metastasis, relative to other breast cancer subtypes. Together, our findings established that MT1-MMP activity in breast tumors is essential for BVI, but not LVI, and that MT1-MMP should be further explored as a predictor and therapeutic target of hematogenous metastasis in TNBC patients.


Breast Neoplasms/blood supply , Matrix Metalloproteinase 14/biosynthesis , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Down-Regulation , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Matrix Metalloproteinase 14/metabolism , Mice , Mice, SCID , Neoplasm Metastasis , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rats , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/deficiency , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/deficiency , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/deficiency
14.
Clin Cancer Res ; 17(8): 2074-80, 2011 Apr 15.
Article En | MEDLINE | ID: mdl-21349998

Addition of multiple molecularly targeted agents to the existing armamentarium of chemotherapeutics and radiotherapies represents a significant advance in the management of several advanced cancers. In certain tumor types with no efficacious therapy options, these agents have become the first line of therapy, for example, sorafenib in advanced hepatocellular carcinoma or bevacizumab in recurrent glioblastoma. Unfortunately, in many cases, the survival benefits are modest, lasting only weeks to a few months. Moreover, they may not show benefit in patients with localized disease (i.e., in the adjuvant setting). Recent studies have provided increasing evidence that activation of the chemokine CXCL12 (SDF1α) pathway is a potential mechanism of tumor resistance to both conventional therapies and biological agents via multiple complementary actions: (i) by directly promoting cancer cell survival, invasion, and the cancer stem and/or tumor-initiating cell phenotype; (ii) by recruiting "distal stroma" (i.e., myeloid bone marrow-derived cells) to indirectly facilitate tumor recurrence and metastasis; and (iii) by promoting angiogenesis directly or in a paracrine manner. Here, we discuss recent preclinical and clinical data that support the potential use of anti-CXCL12 agents (e.g., AMD3100, NOX-A12, or CCX2066) as sensitizers to currently available therapies by targeting the CXCL12/CXCR4 and CXCL12/CXCR7 pathways.


Chemokine CXCL12/metabolism , Neoplasms/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Animals , Benzylamines , Cyclams , Heterocyclic Compounds/therapeutic use , Humans , Models, Biological , Neoplasms/drug therapy , Neoplasms/pathology , Peptides/therapeutic use , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction/drug effects
15.
Clin Cancer Res ; 16(14): 3618-27, 2010 Jul 15.
Article En | MEDLINE | ID: mdl-20501615

PURPOSE: In brain tumors, cerebral edema is a significant source of morbidity and mortality. Recent studies have shown that inhibition of vascular endothelial growth factor (VEGF) signaling induces transient vascular normalization and reduces cerebral edema, resulting in a modest survival benefit in glioblastoma patients. During anti-VEGF treatment, circulating levels of angiopoietin (Ang)-2 remained high after an initial minor reduction. It is not known, however, whether Ang-2 can modulate anti-VEGF treatment of glioblastoma. Here, we used an orthotopic glioma model to test the hypothesis that Ang-2 is an additional target for improving the efficacy of current anti-VEGF therapies in glioma patients. EXPERIMENTAL DESIGN: To recapitulate high levels of Ang-2 in glioblastoma patients during anti-VEGF treatment, Ang-2 was ectopically expressed in U87 glioma cells. Animal survival and tumor growth were assessed to determine the effects of Ang-2 and anti-VEGF receptor 2 (VEGFR2) treatment. We also monitored morphologic and functional vascular changes using multiphoton laser scanning microscopy and immunohistochemistry. RESULTS: Ectopic expression of Ang-2 had no effect on vascular permeability, tumor growth, or survival, although it resulted in higher vascular density, with dilated vessels and reduced mural cell coverage. On the other hand, when combined with anti-VEGFR2 treatment, Ang-2 destabilized vessels without affecting vessel regression and compromised the survival benefit of VEGFR2 inhibition by increasing vascular permeability. VEGFR2 inhibition normalized tumor vasculature whereas ectopic expression of Ang-2 diminished the beneficial effects of VEGFR2 blockade by inhibiting vessel normalization. CONCLUSION: Cancer treatment regimens combining anti-VEGF and anti-Ang-2 agents may be an effective strategy to improve the efficacy of current anti-VEGF therapies.


Angiopoietin-2/metabolism , Glioma/therapy , Neovascularization, Pathologic/therapy , Vascular Endothelial Growth Factor Receptor-2/metabolism , Angiopoietin-2/genetics , Animals , Cell Survival , Disease Models, Animal , Glioma/blood supply , Humans , Immunohistochemistry , Mice , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
16.
Cancer Res ; 69(6): 2260-8, 2009 Mar 15.
Article En | MEDLINE | ID: mdl-19276390

The external pH of solid tumors is acidic as a consequence of increased metabolism of glucose and poor perfusion. Acid pH has been shown to stimulate tumor cell invasion and metastasis in vitro and in cells before tail vein injection in vivo. The present study investigates whether inhibition of this tumor acidity will reduce the incidence of in vivo metastases. Here, we show that oral NaHCO(3) selectively increased the pH of tumors and reduced the formation of spontaneous metastases in mouse models of metastatic breast cancer. This treatment regimen was shown to significantly increase the extracellular pH, but not the intracellular pH, of tumors by (31)P magnetic resonance spectroscopy and the export of acid from growing tumors by fluorescence microscopy of tumors grown in window chambers. NaHCO(3) therapy also reduced the rate of lymph node involvement, yet did not affect the levels of circulating tumor cells, suggesting that reduced organ metastases were not due to increased intravasation. In contrast, NaHCO(3) therapy significantly reduced the formation of hepatic metastases following intrasplenic injection, suggesting that it did inhibit extravasation and colonization. In tail vein injections of alternative cancer models, bicarbonate had mixed results, inhibiting the formation of metastases from PC3M prostate cancer cells, but not those of B16 melanoma. Although the mechanism of this therapy is not known with certainty, low pH was shown to increase the release of active cathepsin B, an important matrix remodeling protease.


Breast Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Prostatic Neoplasms/drug therapy , Sodium Bicarbonate/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
17.
Appl Opt ; 47(25): 4560-8, 2008 Sep 01.
Article En | MEDLINE | ID: mdl-18758526

The emission and transmission properties of three commercially produced coherent fiber optic imaging bundles were evaluated. Full fluorescence excitation versus emission data were collected from 250 to 650 nm excitation for high-resolution Sumitomo, Fujikura, and Schott fiber bundles. The results generated show regions of autofluorescence and inelastic Raman scattering in the imaging bundles that represent a wavelength-dependent background signal when these fibers are used for imaging applications. The high-resolution fiber bundles also exhibit significant variation in transmission with the angle of illumination, which affects the overall coupling and transmission efficiency. Knowledge of these properties allows users of high-resolution imaging bundles to optimally design systems that utilize such bundles.


Fiber Optic Technology/instrumentation , Microscopy, Fluorescence/instrumentation , Equipment Design , Equipment Failure Analysis , Optical Fibers , Scattering, Radiation
18.
Mol Imaging ; 7(1): 50-7, 2008.
Article En | MEDLINE | ID: mdl-18384724

Molecularly targeted gold nanorods were investigated for applications in both diagnostic imaging and disease treatment with cellular resolution. The nanorods were tested in two genetically engineered cell lines derived from the human colon carcinoma HCT-116, a model for studying ligand-receptor interactions. One of these lines was modified to express delta opioid receptor (deltaOR) and green fluorescent protein, whereas the other was receptor free and expressed a red fluorescent protein, to serve as the control. Deltorphin, a high-affinity ligand for deltaOR, was stably attached to the gold nanorods through a thiol-terminated linker. In a mixed population of cells, we demonstrated selective imaging and destruction of receptor-expressing cells while sparing those cells that did not express the receptor. The molecularly targeted nanorods can be used as an in vitro ligand-binding and cytotoxic treatment assay platform and could potentially be applied in vivo for diagnostic and therapeutic purposes with endoscopic technology.


Contrast Media/metabolism , Gold/metabolism , Nanotubes , Receptors, Opioid, delta/metabolism , Amino Acid Sequence , Coculture Techniques , Green Fluorescent Proteins/metabolism , HCT116 Cells , Humans , Luminescent Measurements , Molecular Sequence Data , Oligopeptides/chemistry , Phantoms, Imaging
19.
Cancer Epidemiol Biomarkers Prev ; 16(10): 2048-57, 2007 Oct.
Article En | MEDLINE | ID: mdl-17932352

PURPOSE: Among gynecologic cancers, ovarian cancer is the second most common and has the highest mortality. Currently, there is no accurate early diagnostic technique for ovarian cancer. Furthermore, little is understood regarding the early progression of this disease. We have imaged multiphoton interactions of endogenous tissue constituents from normal and abnormal ovarian biopsies that were kept viable during transport from the operating room and microscopy. EXPERIMENTAL DESIGN: The ovarian surface and underlying stroma were assessed with two-photon excited fluorescence (2PEF) and second harmonic generation (SHG). High-resolution, optically sectioned images were analyzed for epithelial morphology based on 2PEF and collagen density and structural integrity based on SHG. Additionally, multiwavelength 2PEF provided an estimation of the cellular redox ratio of epithelial cells. RESULTS: Normal tissue exhibited a uniform epithelial layer with highly structured collagen in the stroma, whereas abnormal tissue exhibited varied epithelium with large cells and substantial quantitative changes to the collagen structure. Samples from patients at high risk for developing ovarian cancer (based on their personal/family history of cancer) exhibited highly variable cellular redox ratios and changes in collagen structure that trended toward cancer samples. CONCLUSION: This study highlights differences in endogenous signals in viable ovarian biopsies based on quantitative collagen structural changes and redox ratio estimates that may lead to improved detection and further insights in ovarian cancer, particularly in the early stages of the disease.


Biomarkers, Tumor/analysis , Microscopy, Fluorescence, Multiphoton/instrumentation , Ovarian Neoplasms/pathology , Biopsy , Collagen/analysis , Epithelium/pathology , Female , Genetic Predisposition to Disease/genetics , Humans , Image Processing, Computer-Assisted , NAD/analysis , NADP/analysis , Ovarian Neoplasms/genetics , Ovary/pathology , Oxidation-Reduction , Reference Values , Risk Factors , Stromal Cells/pathology
20.
Am J Physiol Heart Circ Physiol ; 292(6): H3198-206, 2007 Jun.
Article En | MEDLINE | ID: mdl-17307995

To better understand interstitial matrix remodeling during angiogenesis, we probed endogenous optical signatures of collagen fibrils and cells with multiphoton microscopy to noninvasively visualize, in real-time, changes to fibril organization around angiogenic sprouts and growing neovessels. From analyses of the second-harmonic generation signal from fibrillar collagen and two-photon excited fluorescence, as well as coherent transmitted light from vascular cells, we found that microvessel fragments interacting with the collagen matrix exhibited two key features: a strong association of fibrillar collagen around the parent vessel fragment during vessel construct reconstitution and a substantial collagen fibril reorganization by sprout and neovessel tips. Results indicate that angiogenic sprouts and growing neovessels actively and differentially remodel existing collagen fibrils. This imaging approach to assess local changes in matrix organization may have a broader impact on tissue biology and mechanics during angiogenesis and allow for new insights in cardiovascular, diabetes, and cancer research.


Adipose Tissue/blood supply , Fibrillar Collagens/metabolism , Microscopy, Fluorescence, Multiphoton , Neovascularization, Physiologic , Animals , Epididymis , Male , Microcirculation/metabolism , Microcirculation/physiology , Rats , Rats, Sprague-Dawley , Signal Processing, Computer-Assisted , Time Factors , Tissue Culture Techniques
...