Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Proc Natl Acad Sci U S A ; 121(14): e2321336121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38530888

ABSTRACT

Host-directed therapies (HDTs) represent an emerging approach for bacterial clearance during tuberculosis (TB) infection. While most HDTs are designed and implemented for immuno-modulation, other host targets-such as nonimmune stromal components found in pulmonary granulomas-may prove equally viable. Building on our previous work characterizing and normalizing the aberrant granuloma-associated vasculature, here we demonstrate that FDA-approved therapies (bevacizumab and losartan, respectively) can be repurposed as HDTs to normalize blood vessels and extracellular matrix (ECM), improve drug delivery, and reduce bacterial loads in TB granulomas. Granulomas feature an overabundance of ECM and compressed blood vessels, both of which are effectively reduced by losartan treatment in the rabbit model of TB. Combining both HDTs promotes secretion of proinflammatory cytokines and improves anti-TB drug delivery. Finally, alone and in combination with second-line antitubercular agents (moxifloxacin or bedaquiline), these HDTs significantly reduce bacterial burden. RNA sequencing analysis of HDT-treated lung and granuloma tissues implicates up-regulated antimicrobial peptide and proinflammatory gene expression by ciliated epithelial airway cells as a putative mechanism of the observed antitubercular benefits in the absence of chemotherapy. These findings demonstrate that bevacizumab and losartan are well-tolerated stroma-targeting HDTs, normalize the granuloma microenvironment, and improve TB outcomes, providing the rationale to clinically test this combination in TB patients.


Subject(s)
Latent Tuberculosis , Mycobacterium tuberculosis , Tuberculosis , Humans , Animals , Rabbits , Bevacizumab/pharmacology , Losartan/pharmacology , Tuberculosis/microbiology , Antitubercular Agents/pharmacology , Granuloma , Latent Tuberculosis/microbiology
2.
Oncologist ; 24(8): 1095-1102, 2019 08.
Article in English | MEDLINE | ID: mdl-30975923

ABSTRACT

BACKGROUND: Seribantumab (MM-121) is a fully human IgG2 monoclonal antibody that binds to human epidermal growth factor receptor 3 (HER3/ErbB3) to block heregulin (HRG/NRG)-mediated ErbB3 signaling and induce receptor downregulation. This open-label, randomized phase 1/2 study evaluated safety and efficacy of seribantumab plus erlotinib in advanced non-small cell lung cancer (NSCLC). Here, we report the activity of seribantumab plus erlotinib, versus erlotinib alone, in patients with EGFR wild-type tumors and describe the potential predictive power of HRG. MATERIALS AND METHODS: Patients with EGFR wild-type NSCLC were assigned randomly to receive seribantumab + erlotinib or erlotinib alone. Patients underwent pretreatment core needle biopsy and archived tumor samples were collected to support prespecified biomarker analyses. RESULTS: One hundred twenty-nine patients received seribantumab + erlotinib (n = 85) or erlotinib alone (n = 44). Median estimated progression-free survival (PFS) in the unselected intent-to-treat (ITT) population was 8.1 and 7.7 weeks in the experimental and control arm, respectively (hazard ratio [HR], 0.822; 95% confidence interval [CI], 0.37-1.828; p = 0.63), and median estimated overall survival was 27.3 and 40.3 weeks in the experimental and control arm, respectively (HR, 1.395; 95% CI, 0.846 to 2.301; p = .1898) In patients whose tumors had detectable HRG mRNA expression, treatment benefit was observed in the seribantumab + erlotinib combination (HR, 0.35; 95% CI, 0.16-0.76; p = .008). In contrast, in patients whose tumors were HRG negative, the HR was 2.15 (95% CI, 0.97-4.76; p = .059, HRG-by-treatment interaction, p value = .0016). CONCLUSION: The addition of seribantumab to erlotinib did not result in improved PFS in unselected patients. However, predefined retrospective exploratory analyses suggest that detectable HRG mRNA levels identified patients who might benefit from seribantumab. An ongoing clinical trial of seribantumab, in combination with docetaxel, is underway in patients with advanced NSCLC and high HRG mRNA expression (NCT02387216). IMPLICATIONS FOR PRACTICE: The poor prognosis of patients with non-small cell lung cancer (NSCLC) underscores the need for more effective treatment options, highlighting the unmet medical need in this patient population. The results of this study show that a novel biomarker, heregulin, may help to identify patients with advanced NSCLC who could benefit from treatment with seribantumab. On the basis of the observed safety profile and promising clinical efficacy, a prospective, randomized, open-label, international, multicenter phase II trial (SHERLOC, NCT02387216) is under way to investigate the efficacy and safety of seribantumab in combination with docetaxel in patients with heregulin-positive advanced adenocarcinoma.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , Erlotinib Hydrochloride/therapeutic use , Lung Neoplasms/drug therapy , Neuregulin-1/analysis , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride/pharmacology , Female , Follow-Up Studies , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neuregulin-1/antagonists & inhibitors , Patient Selection , Progression-Free Survival , Receptor, ErbB-3/analysis , Receptor, ErbB-3/antagonists & inhibitors , Retrospective Studies
3.
Nanomedicine ; 17: 71-81, 2019 04.
Article in English | MEDLINE | ID: mdl-30654182

ABSTRACT

MM-302 is an anti-HER2 antibody-targeted pegylated liposomal doxorubicin designed to deliver doxorubicin specifically to HER2-expressing solid tumors. The delivery and activity of MM-302 were evaluated in orthotopic, transgenic, and intravenous breast cancer models expressing varying levels of HER2 that metastasize to some of the most common sites of dissemination for breast cancer, namely, lung, liver, and brain. Metastatic burden was quantified by gross evaluation, immunohistochemistry (IHC), and bioluminescent imaging. Liposome delivery was quantified by IHC and ex vivo fluorescent imaging. Unlike its non-targeted counterpart, pegylated liposomal doxorubicin (PLD), MM-302 showed activity at controlling both primary and metastatic tumor burden in all models tested. The effect of HER2-targeting was greatest in the lung where lymphatic vessel density and MM-302 delivery were highest. Our data indicate that the therapeutic advantage of actively targeting a nanoliposome with an antibody is influenced by both target expression and the tumor microenvironment.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Immunoconjugates/chemistry , Liposomes/chemistry , Single-Chain Antibodies/chemistry , Animals , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Drug Delivery Systems , Female , Mice , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/therapeutic use , Receptor, ErbB-2/metabolism , Tumor Microenvironment/drug effects
4.
Proc Natl Acad Sci U S A ; 113(16): 4470-5, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044097

ABSTRACT

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.


Subject(s)
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
5.
Proc Natl Acad Sci U S A ; 112(5): 1350-5, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605916

ABSTRACT

Multiplexed, phenotypic, intravital cytometric imaging requires novel fluorophore conjugates that have an appropriate size for long circulation and diffusion and show virtually no nonspecific binding to cells/serum while binding to cells of interest with high specificity. In addition, these conjugates must be stable and maintain a high quantum yield in the in vivo environments. Here, we show that this can be achieved using compact (∼15 nm in hydrodynamic diameter) and biocompatible quantum dot (QD) -Ab conjugates. We developed these conjugates by coupling whole mAbs to QDs coated with norbornene-displaying polyimidazole ligands using tetrazine-norbornene cycloaddition. Our QD immunoconstructs were used for in vivo single-cell labeling in bone marrow. The intravital imaging studies using a chronic calvarial bone window showed that our QD-Ab conjugates diffuse into the entire bone marrow and efficiently label single cells belonging to rare populations of hematopoietic stem and progenitor cells (Sca1(+)c-Kit(+) cells). This in vivo cytometric technique may be useful in a wide range of structural and functional imaging to study the interactions between cells and between a cell and its environment in intact and diseased tissues.


Subject(s)
Antibodies/immunology , Quantum Dots , Animals , Biocompatible Materials , Mice , Mice, Transgenic
6.
Proc Natl Acad Sci U S A ; 112(6): 1827-32, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25624495

ABSTRACT

Tuberculosis (TB) causes almost 2 million deaths annually, and an increasing number of patients are resistant to existing therapies. Patients who have TB require lengthy chemotherapy, possibly because of poor penetration of antibiotics into granulomas where the bacilli reside. Granulomas are morphologically similar to solid cancerous tumors in that they contain hypoxic microenvironments and can be highly fibrotic. Here, we show that TB-infected rabbits have impaired small molecule distribution into these disease sites due to a functionally abnormal vasculature, with a low-molecular-weight tracer accumulating only in peripheral regions of granulomatous lesions. Granuloma-associated vessels are morphologically and spatially heterogeneous, with poor vessel pericyte coverage in both human and experimental rabbit TB granulomas. Moreover, we found enhanced VEGF expression in both species. In tumors, antiangiogenic, specifically anti-VEGF, treatments can "normalize" their vasculature, reducing hypoxia and creating a window of opportunity for concurrent chemotherapy; thus, we investigated vessel normalization in rabbit TB granulomas. Treatment of TB-infected rabbits with the anti-VEGF antibody bevacizumab significantly decreased the total number of vessels while normalizing those vessels that remained. As a result, hypoxic fractions of these granulomas were reduced and small molecule tracer delivery was increased. These findings demonstrate that bevacizumab treatment promotes vascular normalization, improves small molecule delivery, and decreases hypoxia in TB granulomas, thereby providing a potential avenue to improve delivery and efficacy of current treatment regimens.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Blood Vessels/drug effects , Granuloma, Respiratory Tract/drug therapy , Granuloma, Respiratory Tract/metabolism , Tuberculosis/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Bevacizumab , Blood Vessels/pathology , Coloring Agents/pharmacokinetics , Granuloma, Respiratory Tract/etiology , Humans , Pericytes/pathology , Positron-Emission Tomography , Rabbits , Tomography, X-Ray Computed , Tuberculosis/complications
7.
Proc Natl Acad Sci U S A ; 110(31): 12774-9, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23861493

ABSTRACT

Efficient generation of competent vasculogenic cells is a critical challenge of human induced pluripotent stem (hiPS) cell-based regenerative medicine. Biologically relevant systems to assess functionality of the engineered vessels in vivo are equally important for such development. Here, we report a unique approach for the derivation of endothelial precursor cells from hiPS cells using a triple combination of selection markers--CD34, neuropilin 1, and human kinase insert domain-containing receptor--and an efficient 2D culture system for hiPS cell-derived endothelial precursor cell expansion. With these methods, we successfully generated endothelial cells (ECs) from hiPS cells obtained from healthy donors and formed stable functional blood vessels in vivo, lasting for 280 d in mice. In addition, we developed an approach to generate mesenchymal precursor cells (MPCs) from hiPS cells in parallel. Moreover, we successfully generated functional blood vessels in vivo using these ECs and MPCs derived from the same hiPS cell line. These data provide proof of the principle that autologous hiPS cell-derived vascular precursors can be used for in vivo applications, once safety and immunological issues of hiPS-based cellular therapy have been resolved. Additionally, the durability of hiPS-derived blood vessels in vivo demonstrates a potential translation of this approach in long-term vascularization for tissue engineering and treatment of vascular diseases. Of note, we have also successfully generated ECs and MPCs from type 1 diabetic patient-derived hiPS cell lines and use them to generate blood vessels in vivo, which is an important milestone toward clinical translation of this approach.


Subject(s)
Blood Vessel Prosthesis , Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Neovascularization, Physiologic , Tissue Engineering , Animals , Endothelial Cells/transplantation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Mice , Mice, SCID , Transplantation, Heterologous , Vascular Diseases/therapy
8.
Proc Natl Acad Sci U S A ; 109(43): 17561-6, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23045683

ABSTRACT

The recent approval of a prostate cancer vaccine has renewed hope for anticancer immunotherapies. However, the immunosuppressive tumor microenvironment may limit the effectiveness of current immunotherapies. Antiangiogenic agents have the potential to modulate the tumor microenvironment and improve immunotherapy, but they often are used at high doses in the clinic to prune tumor vessels and paradoxically may compromise various therapies. Here, we demonstrate that targeting tumor vasculature with lower vascular-normalizing doses, but not high antivascular/antiangiogenic doses, of an anti-VEGF receptor 2 (VEGFR2) antibody results in a more homogeneous distribution of functional tumor vessels. Furthermore, lower doses are superior to the high doses in polarizing tumor-associated macrophages from an immune inhibitory M2-like phenotype toward an immune stimulatory M1-like phenotype and in facilitating CD4(+) and CD8(+) T-cell tumor infiltration. Based on this mechanism, scheduling lower-dose anti-VEGFR2 therapy with T-cell activation induced by a whole cancer cell vaccine therapy enhanced anticancer efficacy in a CD8(+) T-cell-dependent manner in both immune-tolerant and immunogenic murine breast cancer models. These findings indicate that vascular-normalizing lower doses of anti-VEGFR2 antibody can reprogram the tumor microenvironment away from immunosuppression toward potentiation of cancer vaccine therapies. Given that the combinations of high doses of bevacizumab with chemotherapy have not improved overall survival of breast cancer patients, our study suggests a strategy to use antiangiogenic agents in breast cancer more effectively with active immunotherapy and potentially other anticancer therapies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/blood supply , Immunotherapy , Tumor Microenvironment , Animals , Breast Neoplasms/immunology , Female , Humans , Mice , Vascular Endothelial Growth Factor Receptor-2/immunology
9.
Proc Natl Acad Sci U S A ; 108(9): 3725-30, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21321210

ABSTRACT

Primary tumors secrete factors that alter the microenvironment of distant organs, rendering those organs as fertile soil for subsequent metastatic cancer cell colonization. Although the lungs are exposed to these factors ubiquitously, lung metastases usually develop as a series of discrete lesions. The underlining molecular mechanisms of the formation of these discrete lesions are not understood. Here we show that primary tumors induce formation of discrete foci of vascular hyperpermeability in premetastatic lungs. This is mediated by endothelial cell-focal adhesion kinase (FAK), which up-regulates E-selectin, leading to preferential homing of metastatic cancer cells to these foci. Suppression of endothelial-FAK or E-selectin activity attenuates the number of cancer cells homing to these foci. Thus, localized activation of endothelial FAK and E-selectin in the lung vasculature mediates the initial homing of metastatic cancer cells to specific foci in the lungs.


Subject(s)
Cell Movement , E-Selectin/metabolism , Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Lung/metabolism , Neoplasms/pathology , Up-Regulation , Animals , Cell Adhesion , Cell Line, Tumor , Endothelial Cells/pathology , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Humans , Lung/blood supply , Lung/pathology , Mice , Neoplasm Metastasis , Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Permeability
10.
Proc Natl Acad Sci U S A ; 108(5): 1799-803, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21224417

ABSTRACT

Delivery of blood-borne molecules and nanoparticles from the vasculature to cells in the tissue differs dramatically between tumor and normal tissues due to differences in their vascular architectures. Here we show that two simple measures of vascular geometry--δ(max) and λ--readily obtained from vascular images, capture these differences and link vascular structure to delivery in both tissue types. The longest time needed to bring materials to their destination scales with the square of δ(max), the maximum distance in the tissue from the nearest blood vessel, whereas λ, a measure of the shape of the spaces between vessels, determines the rate of delivery for shorter times. Our results are useful for evaluating how new therapeutic agents that inhibit or stimulate vascular growth alter the functional efficiency of the vasculature and more broadly for analysis of diffusion in irregularly shaped domains.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Diffusion , Humans , Mice , Neoplasms/blood supply
11.
Nat Methods ; 7(8): 655-60, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20581828

ABSTRACT

Not all tumor vessels are equal. Tumor-associated vasculature includes immature vessels, regressing vessels, transport vessels undergoing arteriogenesis and peritumor vessels influenced by tumor growth factors. Current techniques for analyzing tumor blood flow do not discriminate between vessel subtypes and only measure average changes from a population of dissimilar vessels. We developed methodologies for simultaneously quantifying blood flow (velocity, flux, hematocrit and shear rate) in extended networks at single-capillary resolution in vivo. Our approach relies on deconvolution of signals produced by labeled red blood cells as they move relative to the scanning laser of a confocal or multiphoton microscope and provides fully resolved three-dimensional flow profiles within vessel networks. Using this methodology, we show that blood velocity profiles are asymmetric near intussusceptive tissue structures in tumors in mice. Furthermore, we show that subpopulations of vessels, classified by functional parameters, exist in and around a tumor and in normal brain tissue.


Subject(s)
Erythrocytes/cytology , Microcirculation , Neoplasms/blood supply , Animals , Blood Flow Velocity , Hematocrit , Hemorheology , Mice
12.
Nat Nanotechnol ; 18(11): 1351-1363, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37443252

ABSTRACT

Intravenously administered cyclic dinucleotides and other STING agonists are hampered by low cellular uptake and poor circulatory half-life. Here we report the covalent conjugation of cyclic dinucleotides to poly(ß-amino ester) nanoparticles through a cathepsin-sensitive linker. This is shown to increase stability and loading, thereby expanding the therapeutic window in multiple syngeneic tumour models, enabling the study of how the long-term fate of the nanoparticles affects the immune response. In a melanoma mouse model, primary tumour clearance depends on the STING signalling by host cells-rather than cancer cells-and immune memory depends on the spleen. The cancer cells act as a depot for the nanoparticles, releasing them over time to activate nearby immune cells to control tumour growth. Collectively, this work highlights the importance of nanoparticle structure and nano-biointeractions in controlling immunotherapy efficacy.


Subject(s)
Melanoma , Nanoparticles , Neoplasms , Animals , Mice , Polymers/pharmacology , Neoplasms/drug therapy , Signal Transduction , Nanoparticles/therapeutic use , Nanoparticles/chemistry
13.
Clin Cancer Res ; 28(14): 3076-3090, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35584239

ABSTRACT

PURPOSE: The abnormal function of tumor blood vessels causes tissue hypoxia, promoting disease progression and treatment resistance. Although tumor microenvironment normalization strategies can alleviate hypoxia globally, how local oxygen levels change is not known because of the inability to longitudinally assess vascular and interstitial oxygen in tumors with sufficient resolution. Understanding the spatial and temporal heterogeneity should help improve the outcome of various normalization strategies. EXPERIMENTAL DESIGN: We developed a multiphoton phosphorescence quenching microscopy system using a low-molecular-weight palladium porphyrin probe to measure perfused vessels, oxygen tension, and their spatial correlations in vivo in mouse skin, bone marrow, and four different tumor models. Further, we measured the temporal and spatial changes in oxygen and vessel perfusion in tumors in response to an anti-VEGFR2 antibody (DC101) and an angiotensin-receptor blocker (losartan). RESULTS: We found that vessel function was highly dependent on tumor type. Although some tumors had vessels with greater oxygen-carrying ability than those of normal skin, most tumors had inefficient vessels. Further, intervessel heterogeneity in tumors is associated with heterogeneous response to DC101 and losartan. Using both vascular and stromal normalizing agents, we show that spatial heterogeneity in oxygen levels persists, even with reductions in mean extravascular hypoxia. CONCLUSIONS: High-resolution spatial and temporal responses of tumor vessels to two agents known to improve vascular perfusion globally reveal spatially heterogeneous changes in vessel structure and function. These dynamic vascular changes should be considered in optimizing the dose and schedule of vascular and stromal normalizing strategies to improve the therapeutic outcome.


Subject(s)
Microscopy , Neoplasms , Angiotensins , Animals , Hypoxia , Losartan , Mice , Neoplasms/therapy , Oxygen , Receptors, Angiotensin , Tumor Microenvironment
14.
Mol Cancer Ther ; 19(1): 270-281, 2020 01.
Article in English | MEDLINE | ID: mdl-31597714

ABSTRACT

Combinations of chemotherapy with immunotherapy have seen recent clinical success, including two approvals of anti-PD-1/L1 agents in combination with taxane-based chemotherapy in non-small cell lung cancer and triple-negative breast cancer. Here, we present a study on the combination activity and mechanistic rationale of a novel EphA2-targeted liposomal taxane (EphA2-ILs-DTXp) and anti-PD-1. This combination was highly active in mouse syngeneic tumor models, with complete responses observed in 3 of 5 models. In the EMT-6 tumor model, combination of EphA2-ILs-DTXp with anti-PD-1 resulted in a 60% complete response rate, with durable responses that were resistant to rechallenge. These responses were not observed in the absence of CD8+ T cells. Characterization of the immune infiltrates in EMT-6 tumors reveals increased CD8+ T cells, increased CD8+ IFNγ+ CTLs, and an increased CD8/regulatory T-cell (Treg) ratio. These immunomodulatory effects were not observed in mice treated with a combination of docetaxel and anti-PD-1. Pharmacokinetic analysis revealed that the AUC of docetaxel was increased 15 times, from 52.1 to 785 ng/mL/hour, when delivered by EphA2-ILs-DTXp. A dose reduction study of EphA2-ILs-DTXp showed a dose-response relationship for both tumor growth inhibition and the CD8/Treg ratio. Our data indicate that synergism between docetaxel and anti-PD-1 is achievable with nanoliposomal delivery.


Subject(s)
Bridged-Ring Compounds/therapeutic use , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptor, EphA2/metabolism , Taxoids/therapeutic use , Animals , Bridged-Ring Compounds/pharmacology , Disease Models, Animal , Female , Humans , Mice , Neoplasms/pathology , Taxoids/pharmacology
15.
J Control Release ; 310: 47-57, 2019 09 28.
Article in English | MEDLINE | ID: mdl-31400383

ABSTRACT

Ephrin A2 targeted immunoliposomes incorporating pH-sensitive taxane prodrugs were developed for sustained delivery of active drug to solid tumors. Here we describe the systematic formulation development and characterization of these immunoliposomes. We synthesized both paclitaxel and docetaxel prodrugs to formulate as ephrin A2-targeted liposomes stabilized in the aqueous core with sucroseoctasulfate (SOS). The optimized lipid formulation was comprised of egg-sphingomyelin, cholesterol, and polyethylene glycol distearoyl glycerol (PEG-DSG). The formulations examined had a high efficiency of prodrug encapsulation (as high as 114 mol% taxane per mole phospholipid) and subsequent stability (>3 years at 2-8 °C). The taxane prodrug was stabilized with extraliposomal citric acid and subsequently loaded into liposomes containing a gradient of SOS, resulting in highly stable SOS-drug complexes being formed inside the liposome. The internal prodrug and SOS concentrations were optimized for their impact on in vivo drug release and drug degradation. Cryo-electron microscope images revealed dense prodrug-SOS complex in the aqueous core of the immunoliposomes. Ephrin A2-targeted taxane liposomes exhibited sub-nanomolar (0.69 nM) apparent equilibrium dissociation constant toward the extracellular domain of the ephrin A2 receptor, long circulation half-life (8-12 h) in mouse plasma, a release rate dependent on intraliposomal drug concentration and stable long-term storage. At an equitoxic dose of 50 mg taxane/kg, ephrin A2-targeted liposomal prodrug showed greater antitumor activity than 10 mg/kg of docetaxel in A549 non-small cell lung, as well as MDA-MB-436 and SUM149 triple negative breast cancer xenograft models. The lead molecule entered a Phase I clinical trial in patients with solid tumors (NCT03076372).


Subject(s)
Antineoplastic Agents/administration & dosage , Bridged-Ring Compounds/administration & dosage , Drug Carriers/chemistry , Ephrin-A2/metabolism , Nanoparticles/chemistry , Prodrugs/administration & dosage , Taxoids/administration & dosage , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/pharmacokinetics , Bridged-Ring Compounds/pharmacology , Cell Line, Tumor , Drug Compounding , Drug Liberation , Female , Humans , Liposomes , Mice, Nude , Particle Size , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Protein Binding , Taxoids/chemistry , Taxoids/pharmacokinetics , Taxoids/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Nat Biomed Eng ; 3(4): 264-280, 2019 04.
Article in English | MEDLINE | ID: mdl-30952988

ABSTRACT

Antibody-mediated tumour targeting and nanoparticle-mediated encapsulation can reduce the toxicity of antitumour drugs and improve their efficacy. Here, we describe the performance of a nanotherapeutic encapsulating a hydrolytically sensitive docetaxel prodrug and conjugated to an antibody specific for EphA2-a receptor overexpressed in many tumours. Administration of the nanotherapeutic in mice led to slow and sustained release of the prodrug, reduced exposure of active docetaxel in the circulation (compared with administration of the free drug) and maintenance of optimal exposure of the drug in tumour tissue. We also show that administration of the nanotherapeutic in rats and dogs resulted in minimal haematological toxicity, as well as the absence of neutropenia and improved overall tolerability in multiple rodent models. Targeting of the nanotherapeutic to EphA2 improved tumour penetration and resulted in markedly enhanced antitumour activity (compared with administration of free docetaxel and non-targeted nanotherapeutic controls) in multiple tumour-xenografted mice. This nanomedicine could become a potent and safe therapeutic alternative for cancer patients undergoing chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Nanoparticles/therapeutic use , Receptor, EphA2/metabolism , Animals , Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Bridged-Ring Compounds/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Docetaxel/blood , Docetaxel/chemistry , Docetaxel/pharmacokinetics , Docetaxel/therapeutic use , Humans , Liposomes , Mice, Inbred NOD , Mice, SCID , Taxoids/pharmacology , Taxoids/therapeutic use , Tissue Distribution/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
IEEE Trans Biomed Eng ; 55(1): 162-70, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18232358

ABSTRACT

Intravital microscopy has been used to visualize the microcirculation by imaging fluorescent labeled red blood cells (RBCs). Traditionally, microcirculation has been modeled by computing the mean velocity of a few, randomly selected, manually tracked RBCs. However, this protocol is tedious, time consuming, and subjective with technician related bias. We present a new method for analyzing the microcirculation by modeling the RBC motion through automatic tracking. The tracking of RBCs is challenging as in each image, as many as 200 cells move through a complex network of vessels at a wide range of speeds while deforming in shape. To reliably detect RBCs traveling at a wide range of speeds, a window of temporal template matching is applied. Then, cells appearing in successive frames are corresponded based on the motion behavior constraints in terms of the direction, magnitude, and path. The performance evaluation against a ground truth indicates the detection accuracy up to 84% TP at 6% FP and a correspondence accuracy of 89%. We include an in-depth discussion on comparison of the microcirculation based on motion modeling from the proposed automated method against a mean velocity from manual analysis protocol in terms of precision, objectivity, and sensitivity.


Subject(s)
Erythrocytes/cytology , Erythrocytes/physiology , Image Interpretation, Computer-Assisted/methods , Liver Circulation/physiology , Microcirculation/cytology , Microcirculation/physiology , Microscopy, Fluorescence/methods , Animals , Cell Movement/physiology , Flow Cytometry/methods , Rats
18.
Free Radic Biol Med ; 39(10): 1320-31, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16257641

ABSTRACT

Oxidative stress may mediate vascular disruption associated with a loss of endothelial nitric oxide synthase (eNOS) activity and a hypersensitivity to the constrictor effects of endothelin-1 (ET-1). We hypothesize that this is due, in part, to uncoupling of ET(B) receptors from eNOS activation. Thus, we tested whether oxidative stress (OS) affects liver vascular relaxation by reducing basal and ET-1-induced NO production. Primary sinusoidal endothelial cell cultures were pretreated with H(2)O(2) (25 microM) for 1 or 6 h before a 10-min ET-1 stimulation. OS resulted in a significant basal and ET-1-induced decrease in NO production. Acute OS increased the monomeric form of the inhibitory protein caveolin-1 (1.2 +/- 0.05 vs 0.9 +/- 0.02, p < 0.01) and increased the eNOS-caveolin association as determined by coimmunoprecipitation (1.24 +/- 0.04 vs 0.97 +/- 0.04, p < 0.05). ET-1 stimulation further exacerbated these effects. Subacute OS inhibited ET-1-induced eNOS phosphorylation of serine 1177 (activation residue) (1 +/- 0.07 vs 1.6 +/- 0.04, p < 0.05) and dephosphorylation of the inhibitory residue threonine 495 (1.5 +/- 0.08 vs 0.7 +/- 0.02, p < 0.01). Additionally subacute OS resulted in dissociation of eNOS from ET(B) (0.8 +/- 0.09 vs 1.2 +/- 0.06, p < 0.05). Our findings indicate that acute and subacute oxidative stress result in the inhibition of induced nitric oxide synthase activity through distinct mechanisms dependent on caveolin-1 inhibition, ET(B) dissociation, and eNOS phosphorylation.


Subject(s)
Endothelium, Vascular/cytology , Liver/cytology , Nitric Oxide Synthase/metabolism , Oxidative Stress , Animals , Blotting, Western , Caveolins/metabolism , Cell Survival , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelin-1/metabolism , Enzyme Activation , Free Radicals , Hydrogen Peroxide/pharmacology , Immunoprecipitation , Liver/metabolism , Male , Models, Biological , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , RNA/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Threonine/chemistry , Time Factors
19.
Shock ; 24(5): 447-54, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16247331

ABSTRACT

In vivo studies have shown that chronic alcohol consumption sensitizes the liver to endotoxemic shock, leading to liver microcirculation disruption. In the present study, we investigated the molecular mechanisms involved, focusing on endothelial nitric oxide synthase (eNOS) activity and regulation, which represents one of the major vasodilatory pathways. Male Sprague-Dawley rats were fed an alcohol liquid diet or a control isocaloric diet for 5 weeks. Priming effects of ethanol were studied in a model with or without a 24-h LPS treatment (1 mg/kg body weight). At the end of the diet, liver tissue was harvested for western blot, reverse transcriptase-PCR, histological analysis, and immunostaining and blood for serum alanine aminotransferase analysis. Chronic ethanol and LPS alone induced a mild hepatitis and infiltration, respectively. Combined, LPS and chronic ethanol feeding showed a synergistic effect on the liver, leading to extensive steatohepatitis with extensive focal necrosis associated with significantly higher levels of serum ALT. Chronic ethanol and LPS significantly inhibited eNOS activity, but exerted their effects through different mechanisms. Caveolin-1, an eNOS inhibitory protein, was upregulated after LPS and chronic alcohol consumption. Additionally, chronic alcohol consumption down-regulated endothelin B receptor, eNOS protein levels, and eNOS phosphorylation. In conclusion, chronic ethanol consumption and LPS share a similar pathophysiology and both lead to the impairment of eNOS activity, but through distinct molecular mechanisms. The presence of focal necrosis in a mild stress model could provide a good animal study to investigate the advanced stages of alcoholic liver diseases.


Subject(s)
Endotoxins/metabolism , Ethanol/administration & dosage , Gene Expression Regulation, Enzymologic , Liver/drug effects , Nitric Oxide Synthase Type III/metabolism , Alanine Transaminase/blood , Alcohol Drinking , Animals , Blotting, Western , Body Weight , Down-Regulation , Drug Synergism , Endothelin-1/metabolism , Ethanol/pharmacology , Hepatitis , Immunohistochemistry , Lipopolysaccharides/metabolism , Liver/pathology , Male , Necrosis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , RNA/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Endothelin B/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
Shock ; 24(4): 324-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16205316

ABSTRACT

Trauma and subsequent sepsis lead to hepatic microcirculation disruption through various molecular mechanisms in which endothelin-1 (ET-1) plays a pivotal role. These stresses are thought to alter hepatic perfusion, heterogeneously leading to a mismatch of oxygen supply and demand. We hypothesize that mild remote stresses prime the liver to sequential sepsis through direct effects on the hepatic lobular flow distribution. We also propose to investigate the extent and the localization of the stress-induced microcirculation disruption. Sprague-Dawley rats were randomly divided into four experimental groups: sham, femur fracture (FFX), cecal ligation and puncture (CLP), and sequential stress (SS). Hepatic intravital microscopy was performed for in vivo assessment of the liver microcirculation flow distribution under baseline and after ET-1 infusion. Red blood cell motion distribution was used to quantify intralobular and interlobular heterogeneity of perfusion (HoP). Intralobular HoP, which reflects lobular regulation sites, was significantly increased in the FFX and CLP groups, but was not changed or decreased in the SS group compared with control. ET-1 infusion exerted opposite effects depending on the pathological condition, further increasing the difference between groups. SS induced decrease in intralobular HoP, contrasted with a significant increase in interlobular HoP, suggesting multiple disruption sites. Our data suggest that increased intralobular HoP may be indicative of a compensatory response to moderate stress; its decrease under sequential stress conditions corresponds with a total breakdown of hepatic lobular flow regulation. This may be another instance of the rich variability characteristic of normal physiology that "decomplexifies" under critical decompensated conditions.


Subject(s)
Liver/physiology , Animals , Automation , Blood Flow Velocity , Cell Survival , Endothelin-1/metabolism , Erythrocytes/cytology , Femur/pathology , Fracture Healing , Image Processing, Computer-Assisted , Liver/blood supply , Liver/metabolism , Liver/pathology , Liver Circulation , Male , Microcirculation , Perfusion , Rats , Rats, Sprague-Dawley , Sepsis , Time Factors , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL