Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Publication year range
1.
Glycobiology ; 28(12): 958-967, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30007349

ABSTRACT

Modification of hyaluronan (HA) accumulation has been shown to play a key role in regulating inflammatory processes linked to the progression of multiple sclerosis (MS). The aim of this study was to characterize the enzymatic activity involved in HA degradation observed within focal demyelinating lesions in the experimental autoimmune encephalomyelitis (EAE) animal model. EAE was induced in 3-month-old female C57BL/6J mice by immunization with myelin oligodendrocyte glycoprotein 33-35 (MOG33-35) peptide. The mice were monitored for 21 days. Formalin-fixed paraffin-embedded tissue from control and EAE mice were labeled with an immunoadhesin against HA, antibodies against KIAA1199 and glial fibrillary acidic protein, a marker for astrocytes. In situ hybridization was conducted using a KIAA1199 nucleic acid probe. In histologic sections of spinal cord from EAE mice, abnormal HA accumulation was observed in the close vicinity of the affected areas, whereas HA was totally degraded within the focal loci of damaged tissue. KIAA1199 immunoreactivity was exclusively associated with focal loci in damaged white columns of the spinal cord. KIAA1199 was mainly expressed by activated astrocytes that invaded damaged tissue. Similar findings were observed in tissue from an MS patient. Here, we show that KIAA1199, a protein that plays a role in a HA degradation pathway independent of the canonical hyaluronidases such as PH20, is specifically expressed in tissue lesions in which HA is degraded. KIAA1199 expression by activated astrocytes may explain the focal HA degradation observed during progression of MS and could represent a possible new therapeutic target.


Subject(s)
Hyaluronic Acid/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Proteins/metabolism , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
2.
Biophys J ; 110(9): 2106-19, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27166818

ABSTRACT

Elevated interstitial fluid pressure can present a substantial barrier to drug delivery in solid tumors. This is particularly true of pancreatic ductal adenocarcinoma, a highly lethal disease characterized by a robust fibroinflammatory response, widespread vascular collapse, and hypoperfusion that together serve as primary mechanisms of treatment resistance. Free-fluid pressures, however, are relatively low in pancreatic ductal adenocarcinoma and cannot account for the vascular collapse. Indeed, we have shown that the overexpression and deposition in the interstitium of high-molecular-weight hyaluronan (HA) is principally responsible for generating pressures that can reach 100 mmHg through the creation of a large gel-fluid phase. By interrogating a variety of tissues, tumor types, and experimental model systems, we show that an HA-dependent fluid phase contributes substantially to pressures in many solid tumors and has been largely unappreciated heretofore. We investigated the relative contributions of both freely mobile fluid and gel fluid to interstitial fluid pressure by performing simultaneous, real-time fluid-pressure measurements with both the classical wick-in-needle method (to estimate free-fluid pressure) and a piezoelectric pressure catheter transducer (which is capable of capturing pressures associated with either phase). We demonstrate further that systemic treatment with pegylated recombinant hyaluronidase (PEGPH20) depletes interstitial HA and eliminates the gel-fluid phase. This significantly reduces interstitial pressures and leaves primarily free fluid behind, relieving the barrier to drug delivery. These findings argue that quantifying the contributions of free- and gel-fluid phases to hydraulically transmitted pressures in a given cancer will be essential to designing the most appropriate and effective strategies to overcome this important and frequently underestimated resistance mechanism.


Subject(s)
Adenocarcinoma/pathology , Extracellular Fluid/metabolism , Pancreatic Neoplasms/pathology , Animals , Extracellular Fluid/drug effects , Hyaluronic Acid/pharmacology , Hydrostatic Pressure , Mice , NIH 3T3 Cells , Pancreatic Neoplasms/metabolism , Viscosity
3.
J Immunol ; 192(11): 5285-95, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24778442

ABSTRACT

Hyaluronidase (Hyal) and low m.w. hyaluronan (LMW HA) fragments have been widely reported to stimulate the innate immune response. However, most hyaluronidases used were purified from animal tissues (e.g., bovine testis Hyal [BTH]), and contain endotoxin and other unrelated proteins. We tested a highly purified recombinant human Hyal (rHuPH20) and endotoxin-free HA fragments from M(r) 5,000 to 1,500,000 in the rodent air pouch model of inflammation to determine their potential for stimulation of the innate immune response. Exogenous LMW HA fragments (average M(r) 200,000) failed to induce either cytokine/chemokine production or neutrophil infiltration into the air pouch. Challenging the air pouch with LPS or BTH stimulated production of cytokines and chemokines but rHuPH20 did not, suggesting that neither PH20 nor generation of LMW HA fragments in situ stimulates cytokine and chemokine production. LPS and BTH also induced neutrophil infiltration into the air pouch, which was not observed with rHuPH20 treatment. Endotoxin-depleted BTH had much reduced proinflammatory activity, suggesting that the difference in inflammatory responses between rHuPH20 and BTH is likely due to endotoxin contaminants in BTH. When rHuPH20 was dosed with LPS, the induction of cytokines and chemokines was the same as LPS alone, but neutrophil infiltration was inhibited, likely by interrupting HA-CD44 interaction. Our results indicate that neither rHuPH20 nor its directly generated HA catabolites have inflammatory properties in the air pouch model, and rHuPH20 can instead inhibit some aspects of inflammation, such as neutrophil infiltration into the air pouch.


Subject(s)
Antigens, Neoplasm/pharmacology , Histone Acetyltransferases/pharmacology , Hyaluronic Acid/immunology , Hyaluronoglucosaminidase/pharmacology , Lipopolysaccharides/toxicity , Neutrophil Infiltration/drug effects , Neutrophils/immunology , Acute Disease , Animals , Antigens, Neoplasm/immunology , Cattle , Cell Line , Cytokines/immunology , Histone Acetyltransferases/immunology , Humans , Hyaluronoglucosaminidase/immunology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Male , Mice , Neutrophil Infiltration/immunology , Neutrophils/pathology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology
4.
J Biol Chem ; 288(9): 6629-39, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23322779

ABSTRACT

Conditionally active proteins regulated by a physiological parameter represent a potential new class of protein therapeutics. By systematically creating point mutations in the catalytic and linker domains of human MMP-1, we generated a protein library amenable to physiological parameter-based screening. Mutants screened for temperature-sensitive activity had mutations clustered at or near amino acids critical for metal binding. One mutant, GVSK (Gly(159) to Val, Ser(208) to Lys), contains mutations in regions of the catalytic domain involved in calcium and zinc binding. The in vitro activity of GVSK at 37 °C in high Ca(2+) (10 mm) was comparable with MMP-1 (wild type), but in low Ca(2+) (1 mm), there was an over 10-fold loss in activity despite having similar kinetic parameters. Activity decreased over 50% within 15 min and correlated with the degradation of the activated protein, suggesting that GVSK was unstable in low Ca(2+). Varying the concentration of Zn(2+) had no effect on GVSK activity in vitro. As compared with MMP-1, GVSK degraded soluble collagen I at the high but not the low Ca(2+) concentration. In vivo, MMP-1 and GVSK degraded collagen I when perfused in Zucker rat ventral skin and formed higher molecular weight complexes with α2-macroglobulin, an inhibitor of MMPs. In vitro and in vivo complex formation and subsequent enzyme inactivation occurred faster with GVSK, especially at the low Ca(2+) concentration. These data suggest that the activity of the human MMP-1 mutant GVSK can be regulated by Ca(2+) both in vitro and in vivo and may represent a novel approach to engineering matrix-remodeling enzymes for therapeutic applications.


Subject(s)
Calcium/metabolism , Matrix Metalloproteinase 1/metabolism , Mutation, Missense , Amino Acid Substitution , Animals , Calcium/chemistry , Collagen Type I/genetics , Collagen Type I/metabolism , Humans , Matrix Metalloproteinase 1/chemistry , Matrix Metalloproteinase 1/genetics , Protein Binding , Protein Structure, Tertiary , Proteolysis , Rats , Rats, Zucker , Zinc/chemistry , Zinc/metabolism
5.
Gut ; 62(1): 112-20, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22466618

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA. METHODS: Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies. RESULTS: PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility. CONCLUSIONS: The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/physiology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Delivery Systems , Drug Resistance, Neoplasm/physiology , Hyaluronic Acid/physiology , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/physiopathology , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/pharmacology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/pharmacology , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/physiopathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tissue Array Analysis , Treatment Outcome , Gemcitabine
6.
FASEB J ; 22(12): 4316-26, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18772348

ABSTRACT

The metabolism of hyaluronan (HA) relies on HA synthases and hyaluronidases, among which hyaluronidase-1 (HYAL1) and -2 (HYAL2) have been proposed as key actors. Congenital HYAL1 deficiency leads to mucopolysaccharidosis IX (MPS IX), a rare lysosomal storage disorder characterized by joint abnormalities. Knowledge of HYAL2 is limited. This protein displays weak in vitro hyaluronidase activity and acts as a receptor for oncogenic ovine retroviruses. We have generated HYAL2-deficient mice through a conditional Cre-lox system. Hyal2(-/-) mice are viable and fertile. They exhibit localized congenital defects in frontonasal and vertebral bone formation and suffer from mild thrombocytopenia and chronic, possibly intravascular, hemolysis. In addition, Hyal2(-/-) mice display 10-fold increases in plasma levels of HA and 2-fold increases in plasma hyaluronidase activity. Globally, there is no HA accumulation in tissues, including bones, but liver sinusoidal cells seem overloaded with undigested HA. Taken together, these elements demonstrate for the first time that murine HYAL2 has a physiological activity in vivo that is relevant for craniovertebral bone formation, maintenance of plasma HA concentrations, and erythrocyte and platelet homeostasis. In addition, the viability of HYAL2-deficient mice raises the possibility that a similar defect, defining a new MPS disorder, exists in humans.


Subject(s)
Hyaluronoglucosaminidase/deficiency , Mucopolysaccharidoses/genetics , Musculoskeletal Abnormalities/genetics , Thrombocytopenia/genetics , Animals , Female , Genotype , Hematologic Tests , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/blood , Male , Mice , Phenotype , Polymerase Chain Reaction , Skull/abnormalities
7.
Diabetes Technol Ther ; 11(6): 345-52, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19459762

ABSTRACT

BACKGROUND: This phase 1 study investigated the pharmacokinetics (PK) and glucodynamics of insulin lispro (Humalog; Eli Lilly and Co., Indianapolis, IN) or regular human insulin (Humulin R; Eli Lilly and Co.) administered with or without (+/-) recombinant human hyaluronidase (rHuPH20). METHODS: Healthy male volunteers (n = 26), 18-55 years old with body mass index 18-28 kg/m(2), weight >70 kg, and normal fasting glucose, were randomized to a crossover sequence of two subcutaneous injections, each followed by a 6-h euglycemic clamp targeting glucose 90-110 mg/dL: Cohort 1 received 20 U of Humalog +/- 300 U of rHuPH20 (11.3 microg/mL), whereas Cohort 2 received 20 U of Humulin R +/- 240 U of rHuPH20 (10 microg/mL). Pharmacokinetic parameters included peak serum insulin concentration (C(max)), time to C(max) (t(max)), and area under the curve (AUC) of serum concentration versus time. Glucodynamic parameters included time to maximal glucose infusion rate (tGIR(max)) and area under the GIR-versus-time curve (G). RESULTS: For Humalog and Humulin R, respectively, rHuPH20 co-administration reduced t(max) by 51% (P = 0.0006) and 58% (P = 0.0002), increased C(max) by 90% (P = 0.0003) and 142% (P < 0.0001), increased early exposure (AUC(0-2h)) by 85% (P < 0.0001) and 211% (P < 0.0001), and reduced late exposure (AUC(4-6h)) by 41% (P < 0.0001) and 48% (P < 0.0001). Similarly, rHuPH20 reduced tGIR(max) by 41% (P = 0.006) and 35% (P = 0.01), increased early metabolism (G(0-2h)) by 52% (P = 0.001) and 127% (P < 0.0001), and reduced late metabolism (G(4-6h)) by 29% (P = 0.002) and 26% (P = 0.03) for Humalog and Humulin R, respectively. Injections were well tolerated. CONCLUSIONS: Co-administration of rHuPH20 accelerated the PK and glucodynamics of both insulin formulations. Additional studies are necessary to evaluate the clinical relevance of these findings in patients with diabetes.


Subject(s)
Blood Glucose/metabolism , Hyaluronoglucosaminidase/pharmacology , Insulin/analogs & derivatives , Insulin/blood , Adolescent , Adult , Blood Glucose/drug effects , Cross-Over Studies , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacokinetics , Insulin/pharmacology , Insulin Lispro , Male , Middle Aged , Recombinant Proteins/pharmacology , Reference Values , Young Adult
8.
Expert Opin Drug Deliv ; 4(4): 427-40, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17683255

ABSTRACT

The extracellular matrix is a significant barrier to the effective subcutaneous delivery of many drugs, limiting both pharmacokinetic parameters and injection volumes. The space outside adipocytes in the hypodermis is not a fluid, but rather a solid extracellular matrix of collageneous fibrils embedded within a glycosaminoglycan-rich viscoelastic gel that buffers convective forces. The extracellular matrix limits the volume of drug that can be injected at a single site, as well as the rate and amount that reach the vascular compartment. A fully human recombinant DNA-derived hyaluronidase enzyme (rHuPH20) has been developed to leverage the historical efficacy of animal testes extract-derived spreading factors to reversibly modify the hypodermis, in light of discovery of the human hyaluronidase gene family. The application of this technology to increase both injection volumes and bioavailability from subcutaneous injection may overcome some key limitations of this route of administration in multiple settings of care.


Subject(s)
Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/metabolism , Dermis/metabolism , Extracellular Matrix/metabolism , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules/adverse effects , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Humans , Hyaluronoglucosaminidase/adverse effects , Hyaluronoglucosaminidase/chemistry , Hyaluronoglucosaminidase/genetics , Injections, Subcutaneous , Molecular Sequence Data , Protein Engineering , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Solubility
9.
Ann Clin Transl Neurol ; 4(3): 191-211, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28275653

ABSTRACT

OBJECTIVE: Expression of Spam1/PH20 and its modulation of high/low molecular weight hyaluronan substrate have been proposed to play an important role in murine oligodendrocyte precursor cell (OPC) maturation in vitro and in normal and demyelinated central nervous system (CNS). We reexamined this using highly purified PH20. METHODS: Steady-state expression of mRNA in OPCs was evaluated by quantitative polymerase chain reaction; the role of PH20 in bovine testicular hyaluronidase (BTH) inhibition of OPC differentiation was explored by comparing BTH to a purified recombinant human PH20 (rHuPH20). Contaminants in commercial BTH were identified and their impact on OPC differentiation characterized. Spam1/PH20 expression in normal and demyelinated mouse CNS tissue was investigated using deep RNA sequencing and immunohistological methods with two antibodies directed against recombinant murine PH20. RESULTS: BTH, but not rHuPH20, inhibited OPC differentiation in vitro. Basic fibroblast growth factor (bFGF) was identified as a significant contaminant in BTH, and bFGF immunodepletion reversed the inhibitory effects of BTH on OPC differentiation. Spam1 mRNA was undetected in OPCs in vitro and in vivo; PH20 immunolabeling was undetected in normal and demyelinated CNS. INTERPRETATION: We were unable to detect Spam1/PH20 expression in OPCs or in normal or demyelinated CNS using the most sensitive methods currently available. Further, "BTH" effects on OPC differentiation are not due to PH20, but may be attributable to contaminating bFGF. Our data suggest that caution be exercised when using some commercially available hyaluronidases, and reports of Spam1/PH20 morphogenic activity in the CNS may be due to contaminants in reagents.

10.
Cancer Res ; 63(18): 5895-901, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522915

ABSTRACT

Immunotherapy is an attractive strategy for cancer treatment. However, self-tolerance is one of the major mechanisms that dampen immune responses against self-tumor antigens. We have demonstrated that Her-2/neu transgenic mice (neu mice) are tolerant to neu antigens and contain only a low avidity repertoire for neu. However, this repertoire has antitumor activity. Immunizations of neu mice are capable of activating the low-avidity T cells that, at best, retard the tumor growth. To increase the efficacy of the antitumor responses in neu mice, we hypothesized that immunotherapy in combination with antiangiogenic therapy would be a more efficient strategy for tumor eradication. The rationale for using this combination was that by decreasing the growth rate of the tumor with an antiangiogenic therapy, the low-avidity repertoire of neu mice stimulated by immunotherapeutic intervention would be more effective in destroying the slow growing tumor. To test this hypothesis, we stably expressed a soluble form of the Flt-1 vascular endothelial growth factor receptor (sFlt-1) on N202.1A cells, using a retrovirus vector. Expression of sFlt-1 on N202.1A (N202-Flt) cells significantly inhibited the tumor growth compared with N202.1A parental cells. In contrast to the application of immunotherapy alone or antiangiogenic therapy alone, which delayed the tumor growth, the combination of the two therapies provided complete inhibition of tumor growth in Her-2/neu mice. These results indicate that the use of tumor targeting with immunotherapy in simultaneous combination with antiangiogenic therapy provides a more efficient strategy for the treatment of solid tumors.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/therapy , Receptor, ErbB-2/physiology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Division/genetics , Combined Modality Therapy , Extracellular Matrix Proteins/genetics , Female , Genetic Vectors/genetics , Immune Tolerance , Mice , Mice, Transgenic , Myosin Heavy Chains , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Nonmuscle Myosin Type IIB , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Retroviridae/genetics , Transduction, Genetic , Vascular Endothelial Growth Factor Receptor-1
11.
Mol Cancer Ther ; 14(2): 523-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25512619

ABSTRACT

Despite tremendous progress in cancer immunotherapy for solid tumors, clinical success of monoclonal antibody (mAb) therapy is often limited by poorly understood mechanisms associated with the tumor microenvironment (TME). Accumulation of hyaluronan (HA), a major component of the TME, occurs in many solid tumor types, and is associated with poor prognosis and treatment resistance in multiple malignancies. In this study, we describe that a physical barrier associated with high levels of HA (HA(high)) in the TME restricts antibody and immune cell access to tumors, suggesting a novel mechanism of in vivo resistance to mAb therapy. We determined that approximately 60% of HER2(3+) primary breast tumors and approximately 40% of EGFR(+) head and neck squamous cell carcinomas are HA(high), and hypothesized that HA(high) tumors may be refractory to mAb therapy. We found that the pericellular matrix produced by HA(high) tumor cells inhibited both natural killer (NK) immune cell access to tumor cells and antibody-dependent cell-mediated cytotoxicity (ADCC) in vitro. Depletion of HA by PEGPH20, a pegylated recombinant human PH20 hyaluronidase, resulted in increased NK cell access to HA(high) tumor cells, and greatly enhanced trastuzumab- or cetuximab-dependent ADCC in vitro. Furthermore, PEGPH20 treatment enhanced trastuzumab and NK cell access to HA(high) tumors, resulting in enhanced trastuzumab- and NK cell-mediated tumor growth inhibition in vivo. These results suggest that HA(high) matrix in vivo may form a barrier inhibiting access of both mAb and NK cells, and that PEGPH20 treatment in combination with anticancer mAbs may be an effective adjunctive therapy for HA(high) tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hyaluronic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Extracellular Matrix/metabolism , Female , Humans , Hyaluronic Acid/pharmacology , Killer Cells, Natural/metabolism , Mice, Nude , Neoplasms/pathology , Receptor, ErbB-2/metabolism , Trastuzumab , Treatment Outcome , Xenograft Model Antitumor Assays
12.
AAPS J ; 17(5): 1144-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25967925

ABSTRACT

Recombinant human PH20 hyaluronidase (rHuPH20) is used to facilitate dispersion of subcutaneously delivered fluids and drugs. This report summarizes rHuPH20 immunogenicity findings from clinical trials where rHuPH20 was co-administered with SC human immunoglobulin, trastuzumab, rituximab, or insulin. Plasma samples were obtained from evaluable subjects participating in ten different clinical trials as well as from healthy plasma donors. A bridging immunoassay and a modified hyaluronidase activity assay were used to determine rHuPH20-reactive antibody titers and neutralizing antibodies, respectively. rHuPH20-binding antibody populations from selected subjects with positive titers were affinity-purified and subjected to further characterization such as cross-reactivity with endogenous PH20. Among individual trials, the prevalence of pre-existing rHuPH20-reactive antibodies varied between 3 and 12%, excepting the primary immunodeficiency (PID) studies. Incidence of treatment-induced rHuPH20 antibodies was 2 to 18%, with the highest titers (81,920) observed in PID. No neutralizing antibodies were observed. Within most trials, the kinetics of antibody responses were comparable between pre-existing and treatment-induced antibody responses, although responses classified as persistent were more common in subjects with pre-existing titers. There was no association between antibody positivity and either local or systemic adverse events. Pre-existing and treatment-induced antibody populations were of similar immunoglobulin isotypes and cross-reacted to endogenous PH20 to similar extents. No cross-reactivity to PH20 paralogs was detected. rHuPH20 induces only modest immunogenicity which has no association with adverse events. In addition, antibodies purified from baseline-positive individuals are qualitatively similar to those purified from individuals developing rHuPH20-reactive antibodies following exposure to the enzyme.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies/blood , Hyaluronoglucosaminidase/administration & dosage , Recombinant Proteins/administration & dosage , Clinical Trials as Topic , Humans , Hyaluronoglucosaminidase/immunology , Injections, Subcutaneous , Insulin/administration & dosage , Recombinant Proteins/immunology , Rituximab/administration & dosage , Trastuzumab/administration & dosage
13.
In Vivo ; 17(5): 377-88, 2003.
Article in English | MEDLINE | ID: mdl-14598599

ABSTRACT

BACKGROUND: Animal models are crucial to further our understanding of the mechanisms of (progressive) growth of prostatic cancer. MATERIALS AND METHODS: We have developed an intravital microscopic model based on the dorsal skinfold chamber technique in mice, allowing continuous measurements of growth and angiogenesis of small tumor spheroids. A histone H2B-GFP fusion protein has been introduced in our cell lines, allowing evaluation of mitotic and apoptotic indices. This system was used to evaluate the growth and angiogenesis of LnCAP, PC3 and DU145 tumor spheroids. RESULTS: LNCAP spheroids regressed rapidly, with complete tumor cell death within 7 days. PC3 spheroids regressed at a slower rate demonstrating a high apoptotic rate, though considerably lower than LNCAP. DU145 spheroids regressed the slowest, demonstrating considerably lower apoptotic rates and measurable mitotic rates. CONCLUSION: The inability to effectively the innate immune system by encapsulation prevents the survival of small human micro-tumor xenografts. Thus, the complex interactions between tumor cells and TAMs, which are pivotal in tumor biology, are extensively perturbed by "foreign" human tumor cells.


Subject(s)
Adenocarcinoma/pathology , Cell Communication , Epithelial Cells/pathology , Mesoderm/pathology , Prostatic Neoplasms/pathology , Adenocarcinoma/blood supply , Adenocarcinoma/immunology , Animals , Apoptosis , Cell Communication/immunology , Cell Division , Cell Line, Tumor , Chimera , Epithelial Cells/immunology , Epithelial Cells/metabolism , Green Fluorescent Proteins , Humans , Immunocompromised Host , Indicators and Reagents , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mesoderm/immunology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/immunology , Spheroids, Cellular/immunology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transduction, Genetic
14.
Int J Dermatol ; 53(6): 777-85, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24168080

ABSTRACT

BACKGROUND: Edema commonly accompanies surgical procedures and when excessive, can adversely affect surgical outcomes. The skin extracellular matrix, including one of its primary components, hyaluronan (HA), is a significant barrier to effective drainage of accumulated edematous fluid. Recombinant human hyaluronidase (rHuPH20) is a human hyaluronidase that acts transiently and locally to depolymerize HA. A non-liposomal gel formulation that provides a sustained release of rHuPH20 was tested in vivo in a preclinical murine model of acquired lymphedema. METHODS: Lymphedemic mice were injected 24 hours before surgery, and at 2 and 12 days following surgery with rHuPH20 sustained release gel (PH20 SR gel). Quantitative assessment of treatment response indicated that a single dose of PH20 SR gel resulted in accelerated resolution and reduced severity of post-surgical edema as compared to the gel vehicle (control). RESULTS: Statistically significant enzymatic degradation of HA was demonstrated up to 5 mm from the injection site, and histological analysis confirmed removal of HA up to 72 hours following PH20 SR gel administration. CONCLUSIONS: These results demonstrate sustained hyaluronidase enzymatic activity that promotes diffusion of accumulated post-surgical edematous fluid, suggesting that PH20 SR gel may be a useful adjuvant in promoting postoperative edema resolution.


Subject(s)
Delayed-Action Preparations/therapeutic use , Hyaluronoglucosaminidase/therapeutic use , Lymphedema/drug therapy , Postoperative Complications/drug therapy , Analysis of Variance , Animals , Disease Models, Animal , Injections, Intralesional , Lymphedema/etiology , Mice , Postoperative Care , Postoperative Complications/diagnosis , Preoperative Care , Random Allocation , Recombination, Genetic , Reference Values , Time Factors , Treatment Outcome , Wound Healing/drug effects , Wound Healing/physiology
15.
J Histochem Cytochem ; 62(9): 672-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24891594

ABSTRACT

Tumor necrosis factor-Stimulated Gene 6 protein (TSG-6) is a hyaluronan (HA)-binding glycoprotein containing an HA-binding Link module. Because of its well-defined structure, HA binding properties and small size, TSG-6 is an excellent candidate as an alternative to animal-derived HA-binding protein (HABP) for the detection of HA. The present work describes the generation and characterization of a novel recombinant HA-binding probe obtained by fusion of a modified TSG-6 Link module with mutationally inactivated heparin-binding sequence and the Fc portion of human IgG1 (TSG-6-ΔHep-Fc) for tissue HA detection in histological samples. Direct binding assays indicated strong binding of TSG-6-ΔHep-Fc to HA, with little residual binding to heparin. Histolocalization of HA in formalin-fixed, paraffin-embedded tissue sections using biotin-TSG-6-ΔHep-Fc resulted in hyaluronidase-sensitive staining patterns similar to those obtained with biotin-HABP, but with improved sensitivity. HA was detected in many human tissues, and was most abundant in soft connective tissues such as the skin dermis and the stroma of various glands. Digital image analysis revealed a linear correlation between biotin-HABP and biotin-TSG-6-ΔHep-Fc staining intensity in a subset of normal and malignant human tissues. These results demonstrate that TSG-6-ΔHep-Fc is a sensitive and specific probe for the detection of HA by histological methods.


Subject(s)
Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Recombinant Proteins/metabolism , Animals , Biotinylation , CHO Cells , Cricetinae , Cricetulus , Formaldehyde/pharmacology , Heparin/metabolism , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , Hyaluronan Receptors/isolation & purification , Immunoglobulin Fc Fragments/immunology , Mutation , Paraffin Embedding , Protein Engineering , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Tissue Fixation
16.
Biomed Res Int ; 2014: 817613, 2014.
Article in English | MEDLINE | ID: mdl-25147816

ABSTRACT

Extensive accumulation of the glycosaminoglycan hyaluronan is found in pancreatic cancer. The role of hyaluronan synthases 2 and 3 (HAS2, 3) was investigated in pancreatic cancer growth and the tumor microenvironment. Overexpression of HAS3 increased hyaluronan synthesis in BxPC-3 pancreatic cancer cells. In vivo, overexpression of HAS3 led to faster growing xenograft tumors with abundant extracellular hyaluronan accumulation. Treatment with pegylated human recombinant hyaluronidase (PEGPH20) removed extracellular hyaluronan and dramatically decreased the growth rate of BxPC-3 HAS3 tumors compared to parental tumors. PEGPH20 had a weaker effect on HAS2-overexpressing tumors which grew more slowly and contained both extracellular and intracellular hyaluronan. Accumulation of hyaluronan was associated with loss of plasma membrane E-cadherin and accumulation of cytoplasmic ß-catenin, suggesting disruption of adherens junctions. PEGPH20 decreased the amount of nuclear hypoxia-related proteins and induced translocation of E-cadherin and ß-catenin to the plasma membrane. Translocation of E-cadherin was also seen in tumors from a transgenic mouse model of pancreatic cancer and in a human non-small cell lung cancer sample from a patient treated with PEGPH20. In conclusion, hyaluronan accumulation by HAS3 favors pancreatic cancer growth, at least in part by decreasing epithelial cell adhesion, and PEGPH20 inhibits these changes and suppresses tumor growth.


Subject(s)
Glucuronosyltransferase/metabolism , Hyaluronic Acid/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment/physiology , Animals , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Humans , Hyaluronan Synthases , Hyaluronoglucosaminidase/metabolism , Mice , beta Catenin/metabolism
17.
Matrix Biol ; 31(2): 81-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22142621

ABSTRACT

The metabolism of hyaluronan (HA), especially its catabolism, is still far from being elucidated. Although several studies suggest that HA is degraded locally in tissues and through the lymphatic or circulatory systems, much needs to be learned about the enzymes, receptors and cell types that support this dynamic process. In the current work, the clearance of exogenously administered HA was examined in a C57BL/6 mouse model. Hyaluronidase-sensitive fluorescein-labeled 1.2MDa hyaluronan (flHA) was administered either intravenously (i.v.) or subcutaneously (s.c.) into wild type C57BL/6 mice. Plasma was sampled for pharmacokinetic analysis and tissues were harvested for histological examination of the cell types responsible for uptake using immunofluorescent localization and for size exclusion chromatography analysis. We observed that flHA could be degraded locally in the skin or be taken up by sinusoidal cells in lymph nodes, liver and spleen. I.v. administration of flHA revealed non-linear Michaelis-Menten pharmacokinetics compatible with a saturable, receptor-mediated clearance system (K(m)=11.6µg/ml±46.0%, V(max)=1.69µg/ml/min±59.7%). Through a combination of immunofluorescence microscopy, pharmacokinetic, and chromatographic analyses of labeled substrate in vivo, our results shed additional light on the mechanisms by which HA is catabolized in mammals, and serve as a basis for future studies.


Subject(s)
Hyaluronic Acid/pharmacokinetics , Liver/metabolism , Lymph Nodes/metabolism , Skin/metabolism , Spleen/metabolism , Animals , Chromatography, Gel , Fluorescein/metabolism , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/blood , Hyaluronoglucosaminidase/metabolism , Injections, Intravenous , Injections, Subcutaneous , Liver/cytology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Models, Animal , Molecular Weight , Spleen/cytology , Staining and Labeling
18.
Cancers (Basel) ; 4(3): 873-903, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-24213471

ABSTRACT

The tumor stroma, consisting of non-malignant cells and the extracellular matrix, undergoes significant quantitative and qualitative changes throughout malignant transformation and tumor progression. With increasing recognition of the role of the tumor microenvironment in disease progression, stromal components of the tumor have become attractive targets for therapeutic intervention. Stromal accumulation of the glycosaminoglycan hyaluronan occurs in many tumor types and is frequently associated with a negative disease prognosis. Hyaluronan interacts with other extracellular molecules as well as cellular receptors to form a complex interaction network influencing physicochemical properties, signal transduction, and biological behavior of cancer cells. In preclinical animal models, enzymatic removal of hyaluronan is associated with remodeling of the tumor stroma, reduction of tumor interstitial fluid pressure, expansion of tumor blood vessels and facilitated delivery of chemotherapy. This leads to inhibition of tumor growth and increased survival. Current evidence shows that abnormal accumulation of hyaluronan may be an important stromal target for cancer therapy. In this review we highlight the role of hyaluronan and hyaluronan-mediated interactions in cancer, and discuss historical and recent data on hyaluronidase-based therapies and the effect of hyaluronan removal on tumor growth.

19.
Anticancer Res ; 32(4): 1203-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22493350

ABSTRACT

BACKGROUND: The tumor microenvironment is an emerging source of novel therapeutic targets in cancer. The glycosaminoglycan hyaluronan (HA) accumulates in 20-30% of tumors and is often associated with poor prognosis. MATERIALS AND METHODS: We developed a digitized, semiquantitative scoring system for tumor-associated HA content, then grouped tumors (from animal models or patients) according to the degree of HA accumulation (HA+1,2,3). The antitumor response to HA-depletion by pegylated PH20 hyaluronidase (PEGPH20) was then characterized as a function of HA accumulation. RESULTS: Semiquantitative grouping of tumors demonstrated that HA accumulation predicts the response of tumors in animal models to PEGPH20. Prospective analysis of HA content was used to predict response to PEGPH20 of squamous cell-type explants from patients with non-small cell lung cancer in nude mice. CONCLUSION: Measurement of HA is a viable biomarker approach for predicting antitumor response in animal models to the HA-depleting agent, PEGPH20.


Subject(s)
Neoplasms, Experimental/therapy , Tumor Microenvironment , Animals , Base Sequence , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , DNA Primers , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays
20.
Mol Cancer Ther ; 9(11): 3052-64, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20978165

ABSTRACT

Hyaluronan (HA) is a glycosaminoglycan polymer that often accumulates in malignancy. Megadalton complexes of HA with proteoglycans create a hydrated connective tissue matrix, which may play an important role in tumor stroma formation. Through its colloid osmotic effects, HA complexes contribute to tumor interstitial fluid pressure, limiting the effect of therapeutic molecules on malignant cells. The therapeutic potential of enzymatic remodeling of the tumor microenvironment through HA depletion was initially investigated using a recombinant human HA-degrading enzyme, rHuPH20, which removed HA-dependent tumor cell extracellular matrices in vitro. However, rHuPH20 showed a short serum half-life (t(1/2) < 3 minutes), making depletion of tumor HA in vivo impractical. A pegylated variant of rHuPH20, PEGPH20, was therefore evaluated. Pegylation improved serum half-life (t(1/2) = 10.3 hours), making it feasible to probe the effects of sustained HA depletion on tumor physiology. In high-HA prostate PC3 tumors, i.v. administration of PEGPH20 depleted tumor HA, decreased tumor interstitial fluid pressure by 84%, decreased water content by 7%, decompressed tumor vessels, and increased tumor vascular area >3-fold. Following repeat PEGPH20 administration, tumor growth was significantly inhibited (tumor growth inhibition, 70%). Furthermore, PEGPH20 enhanced both docetaxel and liposomal doxorubicin activity in PC3 tumors (P < 0.05) but did not significantly improve the activity of docetaxel in low-HA prostate DU145 tumors. The ability of PEGPH20 to enhance chemotherapy efficacy is likely due to increased drug perfusion combined with other tumor structural changes. These results support enzymatic remodeling of the tumor stroma with PEGPH20 to treat tumors characterized by the accumulation of HA.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/pharmacology , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/pharmacology , Animals , Antineoplastic Agents/administration & dosage , CHO Cells , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/pharmacokinetics , Cricetinae , Cricetulus , Drug Synergism , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/pharmacokinetics , Male , Mice , Mice, Inbred ICR , Mice, Nude , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Tumor Cells, Cultured , Up-Regulation/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL