Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28331086

ABSTRACT

In addition to humans, only certain nonhuman primates are naturally susceptible to measles virus (MeV) infection. Disease severity is species dependent, ranging from mild to moderate for macaques to severe and even lethal for certain New World monkey species. To investigate if squirrel monkeys (Saimiri sciureus), which are reported to develop a course of disease similar to humans, may be better suited than macaques for the identification of virulence determinants or the evaluation of therapeutics, we infected them with a green fluorescent protein-expressing MeV. Compared to cynomolgus macaques (Macaca fascicularis) infected with the same virus, the squirrel monkeys developed more-severe immunosuppression, higher viral load, and a broader range of clinical signs typical for measles. In contrast, infection with an MeV unable to interact with the epithelial receptor nectin-4, while causing immunosuppression, resulted in only a mild and transient rash and a short-lived elevation of the body temperature. Similar titers of the wild-type and nectin-4-blind MeV were detected in peripheral blood mononuclear cells and lymph node homogenates, but only the wild-type virus was found in tracheal lavage fluids and urine. Thus, our study demonstrates the importance of MeV interactions with nectin-4 for clinical disease in the new and better-performing S. sciureus model of measles pathogenesis.IMPORTANCE The characterization of mechanisms underlying measles virus clinical disease has been hampered by the lack of an animal model that reproduces the course of disease seen in human patients. Here, we report that infection of squirrel monkeys (Saimiri sciureus) fulfills these requirements. Comparative infection with wild-type and epithelial cell receptor-blind viruses demonstrated the importance of epithelial cell infection for clinical disease, highlighting the spread to epithelia as an attractive target for therapeutic strategies.


Subject(s)
Cell Adhesion Molecules/metabolism , Measles virus/pathogenicity , Measles/virology , Models, Animal , Saimiri , Animals , Epithelial Cells/virology , Green Fluorescent Proteins , Humans , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/virology , Macaca fascicularis , Measles virus/physiology , Nectins , Viral Load , Virulence
2.
Mol Ther ; 21(11): 2019-31, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23913184

ABSTRACT

We developed here a vaccine-identical measles virus (MV) as an oncolytic agent against mantle cell lymphoma (MCL), an aggressive B-cell non-Hodgkin's lymphoma that is difficult to cure but radiosensitive. We armed the virus with the sodium-iodide symporter, which concentrates iodide within infected cells enabling noninvasive imaging and combination radiovirotherapy. Through high-resolution in vivo and ex vivo imaging, we visualized the spread of infections in primary and metastatic tumors for over 2 weeks after therapy, documenting homogeneous virus seeding and spread restricted to perfused tissue. Infection of metastases was more rapid and intense than primary tumors, achieving isotope uptake within about threefold the efficiency of the thyroid. Virotherapy combined with systemic (131)I resulted in more rapid disease regression than either therapy alone. In addition to ubiquitous CD46, vaccine MV retains cell entry through its immune cell-specific receptor signaling lymphocytic activation molecule (SLAM). We asked whether both receptors could sustain effective oncolysis of MCL. Strikingly, only SLAM-dependent entry sustained efficient viral spread, tumor regression, and prolonged survival. These observations shift the focus of future clinical trials to SLAM-expressing hematologic malignancies and suggest that oncolytic vectors may depend on tissue-specific receptors for both cell entry and activation of responses assisting their replication.


Subject(s)
Antigens, CD/immunology , Lymphoma, Mantle-Cell/therapy , Measles virus/physiology , Oncolytic Virotherapy/methods , Receptors, Cell Surface/immunology , Symporters/genetics , Animals , CHO Cells , Cell Line, Tumor , Chlorocebus aethiops , Combined Modality Therapy , Cricetulus , Female , Humans , Iodine Radioisotopes/therapeutic use , Lymphoma, Mantle-Cell/radiotherapy , Lymphoma, Mantle-Cell/virology , Measles Vaccine , Measles virus/genetics , Measles virus/immunology , Mice , Mice, Nude , Neoplasms, Experimental , Radiotherapy/methods , Signal Transduction , Signaling Lymphocytic Activation Molecule Family Member 1 , Symporters/metabolism , Vero Cells , Virus Internalization , Virus Replication , Xenograft Model Antitumor Assays
3.
J Virol ; 87(5): 2526-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23255790

ABSTRACT

After the contagion measles virus (MV) crosses the respiratory epithelium within myeloid cells that express the primary receptor signaling lymphocytic activation molecule (SLAM), it replicates briskly in SLAM-expressing cells in lymphatic organs. Later, the infection spreads to epithelia expressing nectin-4, an adherens junction protein expressed preferentially in the trachea, but how it gets there is not understood. To characterize the mechanisms of spread, we infected groups of 5 or 6 cynomolgus monkeys (Macaca fascicularis) with either a wild-type MV or its "N4-blind" derivative, which is unable to enter nectin-4-expressing cells because of the targeted mutation of two hemagglutinin residues. As expected, both viruses caused similar levels of immunosuppression, as monitored by reductions in white blood cell counts and lymphocyte proliferation activity. However, monkeys infected with the N4-blind MV cleared infection more rapidly. Wild-type virus-infected monkeys secreted virus, while marginal virus titers were detected in tracheal lavage fluid cells of N4-blind MV-infected hosts. Analyses of tracheal rings obtained at necropsy (day 12) documented widespread infection of individual cells or small cell clusters in the subepithelial lamina propria of monkeys infected with either virus. However, only wild-type MV spread to the epithelium, forming numerous infectious centers comprised of many contiguous columnar cells. Infected CD11c(+) myeloid (macrophage or dendritic) cells were frequently observed in the lamina propria below epithelial infectious centers. Thus, MV may use myeloid cells as vehicles not only immediately after contagion but also to infect epithelia of tissues expressing nectin-4, including the trachea.


Subject(s)
Cell Adhesion Molecules/metabolism , Macaca fascicularis/virology , Measles virus/physiology , Mucous Membrane/immunology , Respiratory Mucosa/virology , Trachea/immunology , Trachea/virology , Animals , Antigens, CD/biosynthesis , Cell Adhesion Molecules/biosynthesis , Chlorocebus aethiops/immunology , Chlorocebus aethiops/virology , Dendritic Cells/immunology , Dendritic Cells/virology , Epithelial Cells/virology , Female , Green Fluorescent Proteins/metabolism , Immunosuppression Therapy , Macrophages/immunology , Macrophages/virology , Measles/metabolism , Measles/virology , Measles virus/genetics , Measles virus/metabolism , Mucous Membrane/virology , Receptors, Cell Surface/biosynthesis , Receptors, Virus/metabolism , Respiratory Mucosa/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells
4.
J Virol ; 86(21): 11558-66, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22896607

ABSTRACT

Hepatitis C virus (HCV) infection remains a serious public health problem worldwide. Treatments are limited, and no preventive vaccine is available. Toward developing an HCV vaccine, we engineered two recombinant measles viruses (MVs) expressing structural proteins from the prototypic HCV subtype 1a strain H77. One virus directs the synthesis of the HCV capsid (C) protein and envelope glycoproteins (E1 and E2), which fold properly and form a heterodimer. The other virus expresses the E1 and E2 glycoproteins separately, with each one fused to the cytoplasmic tail of the MV fusion protein. Although these hybrid glycoproteins were transported to the plasma membrane, they were not incorporated into MV particles. Immunization of MV-susceptible, genetically modified mice with either vector induced neutralizing antibodies to MV and HCV. A boost with soluble E2 protein enhanced titers of neutralizing antibody against the homologous HCV envelope. In animals primed with MV expressing properly folded HCV C-E1-E2, boosting also induced cross-neutralizating antibodies against two heterologous HCV strains. These results show that recombinant MVs retain the ability to induce MV-specific humoral immunity while also eliciting HCV neutralizing antibodies, and that anti-HCV immunity can be boosted with a single dose of purified E2 protein. The use of MV vectors could have advantages for pediatric HCV vaccination.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Hepacivirus/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Cross Reactions , Drug Carriers/administration & dosage , Genetic Vectors , Hepacivirus/genetics , Measles virus/genetics , Mice , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage
5.
Nature ; 480(7378): 530-3, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22048310

ABSTRACT

Measles virus is an aerosol-transmitted virus that affects more than 10 million children each year and accounts for approximately 120,000 deaths. Although it was long believed to replicate in the respiratory epithelium before disseminating, it was recently shown to infect initially macrophages and dendritic cells of the airways using signalling lymphocytic activation molecule family member 1 (SLAMF1; also called CD150) as a receptor. These cells then cross the respiratory epithelium and transport the infection to lymphatic organs where measles virus replicates vigorously. How and where the virus crosses back into the airways has remained unknown. On the basis of functional analyses of surface proteins preferentially expressed on virus-permissive human epithelial cell lines, here we identify nectin-4 (ref. 8; also called poliovirus-receptor-like-4 (PVRL4)) as a candidate host exit receptor. This adherens junction protein of the immunoglobulin superfamily interacts with the viral attachment protein with high affinity through its membrane-distal domain. Nectin-4 sustains measles virus entry and non-cytopathic lateral spread in well-differentiated primary human airway epithelial sheets infected basolaterally. It is downregulated in infected epithelial cells, including those of macaque tracheae. Although other viruses use receptors to enter hosts or transit through their epithelial barriers, we suggest that measles virus targets nectin-4 to emerge in the airways. Nectin-4 is a cellular marker of several types of cancer, which has implications for ongoing measles-virus-based clinical trials of oncolysis.


Subject(s)
Cell Adhesion Molecules/metabolism , Measles virus/metabolism , Measles/metabolism , Receptors, Virus/metabolism , Animals , CHO Cells , Cell Adhesion Molecules/genetics , Cell Line , Cricetinae , Gene Expression Profiling , Humans , Receptors, Virus/genetics
6.
Mol Ther ; 19(10): 1813-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21610701

ABSTRACT

Measles virus (MV) is a promising vector for cancer therapy and multivalent vaccination, but high prevalence of pre-existing neutralizing antibodies may reduce therapeutic efficacy, particularly following systemic administration. MV has only one serotype, but here we show that its envelope glycoproteins can be exchanged with those of the closely related canine distemper virus (CDV), generating a chimeric virus capable of escaping neutralization. To target its entry, we displayed on the CDV attachment protein a single-chain antibody specific for a designated receptor. To enhance oncolytic efficacy we armed the virus with a prodrug convertase gene capable of locally activating chemotherapeutic prodrugs. The new virus achieved high titers, was genetically stable, and was resistant to neutralization by sera from both MV-immunized mice and MV-immune humans. The new virus targeted syngeneic murine tumor cells expressing the designated receptor implanted in immunocompetent mice, and synergized with a chemotherapeutic prodrug in a model of oncolysis. Importantly, the chimeric MV remained oncolytic when administered systemically even in the presence of anti-MV antibodies capable of abrogating the therapeutic efficacy of the parental, nonshielded MV. This work shows that targeting, arming, and shielding can be combined to generate a tumor-specific, neutralization-resistant virus that can synergize with chemotherapeutics.


Subject(s)
Measles virus/genetics , Oncolytic Virotherapy , Tumor Escape , Animals , Base Sequence , DNA Primers , Genetic Vectors , Humans , Mice , Mice, Inbred C57BL , Neutralization Tests , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics
7.
Mol Ther ; 19(6): 1097-106, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21468006

ABSTRACT

Oncolytic measles viruses (MV) derived from the live attenuated vaccine strain have been engineered for increased tumor-cell specificity, and are currently under investigation in clinical trials including a phase I study for glioblastoma multiforme (GBM). Recent preclinical studies have shown that the cellular tropism of several viruses can be controlled by inserting microRNA-target sequences into their genomes, thereby inhibiting spread in tissues expressing cognate microRNAs. Since neuron-specific microRNA-7 is downregulated in gliomas but highly expressed in normal brain tissue, we engineered a microRNA-sensitive virus containing target sites for microRNA-7 in the 3'-untranslated region of the viral fusion gene. In presence of microRNA-7 this modification inhibits translation of envelope proteins, restricts viral spread, and progeny production. Even though highly attenuated in presence of microRNA-7, this virus retained full efficacy against glioblastoma xenografts. Furthermore, microRNA-mediated inhibition protected genetically modified mice susceptible to MV infection from a potentially lethal intracerebral challenge. Importantly, endogenous microRNA-7 expression in primary human brain resections tightly restricted replication and spread of microRNA-sensitive virus. This is proof-of-concept that tropism restriction by tissue-specific microRNAs can be adapted to oncolytic MV to regulate viral replication and gene expression to maximize tumor specificity without compromising oncolytic efficacy.


Subject(s)
Cell Survival/physiology , Measles virus/genetics , MicroRNAs/genetics , Oncolytic Viruses/physiology , Animals , Brain Neoplasms/therapy , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Chlorocebus aethiops , Female , Genetic Vectors/genetics , Glioblastoma/therapy , Glioma/therapy , Humans , Immunoblotting , In Vitro Techniques , Measles virus/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Xenograft Model Antitumor Assays
8.
J Virol ; 84(6): 3033-42, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042501

ABSTRACT

Recent studies of primate models suggest that wild-type measles virus (MV) infects immune cells located in the airways before spreading systemically, but the identity of these cells is unknown. To identify cells supporting primary MV infection, we took advantage of mice expressing the MV receptor human signaling lymphocyte activation molecule (SLAM, CD150) with human-like tissue specificity. We infected these mice intranasally (IN) with a wild-type MV expressing green fluorescent protein. One, two, or three days after inoculation, nasal-associated lymphoid tissue (NALT), the lungs, several lymph nodes (LNs), the spleen, and the thymus were collected and analyzed by microscopy and flow cytometry, and virus isolation was attempted. One day after inoculation, MV replication was documented only in the airways, in about 2.5% of alveolar macrophages (AM) and 0.5% of dendritic cells (DC). These cells expressed human SLAM, and it was observed that MV infection temporarily enhanced SLAM expression. Later, MV infected other immune cell types, including B and T lymphocytes. Virus was isolated from lymphatic tissue as early as 2 days post-IN inoculation; the mediastinal lymph node was an early site of replication and supported high levels of infection. Three days after intraperitoneal inoculation, 1 to 8% of the mediastinal LN cells were infected. Thus, MV infection of alveolar macrophages and subepithelial dendritic cells in the airways precedes infection of lymphocytes in lymphatic organs of mice expressing human SLAM with human-like tissue specificity.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/virology , Lymphoid Tissue/virology , Macrophages, Alveolar/virology , Measles virus/metabolism , Measles/immunology , Receptors, Cell Surface/immunology , Animals , Antigens, CD/genetics , Dendritic Cells/metabolism , Humans , Lung/cytology , Lung/immunology , Lung/virology , Lymphoid Tissue/metabolism , Macrophages, Alveolar/metabolism , Measles/pathology , Measles virus/pathogenicity , Mice , Mice, Transgenic , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1
9.
Cancer Res ; 67(22): 10939-47, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006839

ABSTRACT

Combination chemotherapy regimen incorporating CD20 antibodies are commonly used in the treatment of CD20-positive non-Hodgkin's lymphoma (NHL). Fludarabine phosphate (F-araAMP), cyclophosphamide, and CD20 antibodies (Rituximab) constitute the FCR regimen for treating selected NHL, including aggressive mantle cell lymphoma (MCL). As an alternative to the CD20 antibody, we generated a CD20-targeted measles virus (MV)-based vector. This vector was also armed with the prodrug convertase purine nucleoside phosphorylase (PNP) that locally converts the active metabolite of F-araAMP to a highly diffusible substance capable of efficiently killing bystander cells. We showed in infected cells that early prodrug administration controls vector spread, whereas late administration enhances cell killing. Control of spread by early prodrug administration was also shown in an animal model: F-araAMP protected genetically modified mice susceptible to MV infection from a potentially lethal intracerebral challenge. Enhanced oncolytic potency after extensive infection was shown in a Burkitt's lymphoma xenograft model (Raji cells): After systemic vector inoculation, prodrug administration enhanced the therapeutic effect synergistically. In a MCL xenograft model (Granta 519 cells), intratumoral (i.t.) vector administration alone had high oncolytic efficacy: All mice experienced complete but temporary tumor regression, and survival was two to four times longer than that of untreated mice. Cells from MCL patients were shown to be sensitive to infection. Thus, synergy of F-araAMP with a PNP-armed and CD20-targeted MV was shown in one lymphoma therapy model after systemic vector inoculation.


Subject(s)
Antigens, CD20/biosynthesis , Lymphoma/drug therapy , Lymphoma/metabolism , Measles virus/metabolism , Vidarabine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Chlorocebus aethiops , Drug Therapy/methods , Humans , Membrane Cofactor Protein/biosynthesis , Mice , Mice, Transgenic , Prodrugs/pharmacology , Purine-Nucleoside Phosphorylase/metabolism , Vero Cells , Vidarabine/pharmacology
10.
Mol Ther ; 15(11): 1991-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17712331

ABSTRACT

An immunocompetent model is required to test therapeutic regimens for clinical trials with the oncolytic measles virus (MV). Toward developing this model, a retargeted MV that enters murine colon adenocarcinoma cells forming tumors in syngeneic C57BL/6 mice was generated. Since MV infection tends to be less efficient in murine than in human cells, the targeted virus was also armed with the prodrug convertase, purine nucleoside phosphorylase (PNP), and named MV-PNP-antiCEA. We have shown before that in cultured cells, infection with this virus activated the prodrug, 6-methylpurine-2'-deoxyriboside (MeP-dR), causing extensive cytotoxicity. When injected intratumorally (IT), MV-PNP-antiCEA inhibited subcutaneous tumor growth marginally, but subsequent administration of the prodrug enhanced the oncolytic effect. Systemic delivery of MV-PNP-antiCEA alone had no substantial oncolytic effects, but in combination with the prodrug it was therapeutic, revealing synergistic effects between virus and prodrug. Immunosuppression with cyclophosphamide (CPA) retarded the appearance of MV neutralizing antibodies and enhanced oncolytic efficacy: survival was 100%, with 9 out of 10 animals going into complete remission. This immunocompetent murine model facilitates the testing of therapeutic regimens for clinical trials.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Disease Models, Animal , Immunocompetence/immunology , Measles virus/immunology , Measles virus/metabolism , Animals , Bystander Effect , Cell Line , Chlorocebus aethiops , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclophosphamide/pharmacology , Gene Targeting , Genetic Therapy/adverse effects , Immunosuppression Therapy , Measles/genetics , Measles/immunology , Measles/metabolism , Measles/pathology , Measles virus/drug effects , Measles virus/genetics , Mice , Mice, Inbred C57BL , Prodrugs/therapeutic use , Prodrugs/toxicity , Survival Rate
11.
Cancer Res ; 66(15): 7694-700, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885371

ABSTRACT

Cancer cells secrete matrix metalloproteinases (MMP) that degrade the extracellular matrix and are responsible for some hallmarks of malignant cancer. Many viruses, including a few currently used in oncolytic virotherapy clinical trials, depend on intracellular proteases to process their proteins and activate their particles. We show here for measles virus (MV) that particle activation can be made dependent of proteases secreted by cancer cells. The MV depends on the intracellular protease furin to process and activate its envelope fusion (F) protein. To make F protein activation cancer cell specific, we introduced hexameric sequences recognized by an MMP and identified the mutant proteins most effective in fusing MMP-expressing human fibrosarcoma cells (HT1080). We showed that an MMP inhibitor interferes with syncytia formation elicited by mutant F proteins and confirmed MMP-dependent cleavage by Edman degradation sequence analysis. We generated recombinant MVs expressing the modified F proteins in place of furin-activated F. These viruses spread only in cells secreting MMP. In nude mice, an MMP-activated MV retarded HT1080 xenograft growth as efficiently as the furin-activated MV vaccine strain. In MV-susceptible mice, the furin-activated virus caused lethal encephalitis upon intracerebral inoculation, whereas the MMP-activated did not. Thus, MV particle activation can be made dependent of proteases secreted by cancer cells, enhancing safety. This study opens the perspective of combining targeting at the particle activation, receptor recognition, and selective replication levels to improve the therapeutic index of MV and other viruses in ongoing clinical trials of oncolysis.


Subject(s)
Matrix Metalloproteinases/metabolism , Measles virus/physiology , Oncolytic Virotherapy/methods , Amino Acid Sequence , Animals , Cell Line, Tumor , Chlorocebus aethiops , Fibrosarcoma/enzymology , Fibrosarcoma/therapy , Fibrosarcoma/virology , Furin/metabolism , Humans , Matrix Metalloproteinase Inhibitors , Measles virus/metabolism , Mice , Mice, Nude , Molecular Sequence Data , Oncolytic Virotherapy/adverse effects , Vero Cells , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Activation
12.
Cancer Gene Ther ; 12(7): 593-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15746945

ABSTRACT

Oncolytic viruses are promising cytoreductive agents for cancer treatment but extensive human testing will be required before they are made commercially available. Here, we investigated the oncolytic potential of two commercially available live attenuated vaccines, Moraten measles and Jeryl-Lynn mumps, in a murine model of intraperitoneal human ovarian cancer and compared their efficacies against a recombinant oncolytic measles virus (MV-CEA) that is being tested in a phase I clinical trial. The common feature of these viruses is that they express hemagglutinin and fusion therapeutic proteins that can induce extensive fusion of the infected cell with its neighbors, resulting in death of the cell monolayer. In vitro, the three viruses caused intercellular fusion in human ovarian cancer cells but with marked differences in fusion kinetics. MV-CEA was the fastest followed by Jeryl-Lynn mumps virus while Moraten measles virus was the slowest, although all viruses eventually caused comparable cell death 6 days postinfection. Tumor-bearing mice treated with 10(6) or 10(7) pfu (one thousand times the vaccine dose) of each of the three viruses responded favorably to therapy with significant prolongations in survival. All three viruses demonstrated equivalent antitumor potency. Commercially available Moraten measles and Jeryl-Lynn mumps vaccines warrant further investigation as potential anticancer agents.


Subject(s)
Measles Vaccine/therapeutic use , Mumps Vaccine/therapeutic use , Ovarian Neoplasms/therapy , Animals , Carcinoembryonic Antigen/blood , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/metabolism , Cytopathogenic Effect, Viral , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
13.
Hum Gene Ther ; 14(16): 1565-77, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14577918

ABSTRACT

In support of a proposed phase I clinical trial, we studied the biodistribution of virus-infected cells after intraperitoneal administration of oncolytic measles viruses to alpha/beta interferon-defective mice expressing human CD46 with human-like tissue specificity. Various marker genes were employed, and green fluorescent protein proved to be most informative. Mesothelium and ovarian surface epithelium were remarkably resistant to infection, but infected peritoneal macrophages were present in abundance both in peritoneal lavage fluid and in the greater omentum, where they were heavily concentrated in "milky spots". Infected macrophages were also identified outside the peritoneal cavity, along the peritoneal fluid drainage pathway and in the spleen. Thus, diaphragmatic stomata, thoracic lymphatic vessels, and parathymic lymph nodes contained numerous measles-infected cells, as did the marginal zones of the white pulp of the spleen. Splenic marginal zone macrophages were the predominant targets of infection after intravenous administration of oncolytic measles viruses. When measles-infected peritoneal macrophages were adoptively transferred, they did not migrate beyond the confines of the peritoneal cavity, suggesting that, after intraperitoneal virus administration, the positive cells in thoracic lymphatics, parathymic lymph nodes, and spleen are nonmigratory cells transduced in situ by viral particles that have exited from the peritoneal cavity.


Subject(s)
Antigens, CD/physiology , Injections, Intraperitoneal , Measles virus/physiology , Membrane Glycoproteins/physiology , Peritoneal Cavity/cytology , Recombinant Proteins/administration & dosage , Animals , Green Fluorescent Proteins , Interferon Type I/metabolism , Luminescent Proteins/metabolism , Macrophages/virology , Membrane Cofactor Protein , Mice , Mice, Transgenic , Organ Specificity , Peritoneal Cavity/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...