Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Ther ; 22(5): 1048-55, 2014 May.
Article in English | MEDLINE | ID: mdl-24572293

ABSTRACT

G207, a mutant herpes simplex virus (HSV) type 1, is safe when inoculated into recurrent malignant glioma. We conducted a phase 1 trial of G207 to demonstrate the safety of stereotactic intratumoral administration when given 24 hours prior to a single 5 Gy radiation dose in patients with recurrent malignant glioma. Nine patients with progressive, recurrent malignant glioma despite standard therapy were included. Patients received one dose of G207 stereotactically inoculated into the multiple sites of the enhancing tumor margin and were then treated focally with 5 Gy radiation. Treatment was well tolerated, and no patient developed HSV encephalitis. The median interval between initial diagnosis and G207 inoculation was 18 months (mean: 23 months; range: 11-51 months). Six of the nine patients had stable disease or partial response for at least one time point. Three instances of marked radiographic response to treatment occurred. The median survival time from G207 inoculation until death was 7.5 months (95% confidence interval: 3.0-12.7). In conclusion, this study showed the safety and the potential for clinical response of single-dose oncolytic HSV therapy augmented with radiation in the treatment of malignant glioma patients. Additional studies with oncolytic HSV such as G207 in the treatment of human glioma are recommended.


Subject(s)
Genetic Therapy , Glioma/genetics , Glioma/radiotherapy , Herpesvirus 1, Human/genetics , Adult , Female , Glioma/diagnostic imaging , Glioma/virology , Herpesvirus 1, Human/pathogenicity , Humans , Male , Middle Aged , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/pathogenicity , Radiography , Virus Replication/genetics
2.
PLoS One ; 8(11): e81768, 2013.
Article in English | MEDLINE | ID: mdl-24312353

ABSTRACT

Oncolytic type-1 herpes simplex viruses (oHSVs) lacking the γ134.5 neurovirulence gene are being evaluated for treatment of a variety of malignancies. oHSVs replicate within and directly kill permissive cancer cells. To augment their anti-tumor activity, oHSVs have been engineered to express immunostimulatory molecules, including cytokines, to elicit tumor-specific immune responses. Interleukin-15 (IL-15) holds potential as an immunotherapeutic cytokine because it has been demonstrated to promote both natural killer (NK) cell-mediated and CD8(+) T cell-mediated cytotoxicity against cancer cells. The purpose of these studies was to engineer an oHSV producing bioactive IL-15. Two oHSVs were constructed encoding murine (m)IL-15 alone (J100) or with the mIL-15 receptor α (mIL-15Rα, J100D) to determine whether co-expression of these proteins is required for production of bioactive mIL-15 from oHSV. The following were demonstrated: i) both oHSVs retain replication competence and cytotoxicity in permissive tumor cell lines. ii) Enhanced production of mIL-15 was detected in cell lysates of neuro-2a cells following J100D infection as compared to J100 infection, suggesting that mIL-15Rα improved mIL-15 production. iii) Soluble mIL-15 in complex with mIL-15Rα was detected in supernates from J100D-infected, but not J100-infected, neuro-2a, GL261, and CT-2A cells. These cell lines vary in permissiveness to oHSV replication and cytotoxicity, demonstrating soluble mIL-15/IL-15Rα complex production from J100D was independent of direct oHSV effects. iv) The soluble mIL-15/IL-15Rα complex produced by J100D was bioactive, stimulating NK cells to proliferate and reduce the viability of syngeneic GL261 and CT-2A cells. v) J100 and J100D were aneurovirulent inasmuch as no neuropathologic effects were documented following direct inoculation into brains of CBA/J mice at up to 1x10(7) plaque forming units. The production of mIL-15/mIL-15Rα from multiple tumor lines, as well as the lack of neurovirulence, renders J100D suitable for investigating the combined effects of oHSV and mIL-15/IL-15Rα in various cancer models.


Subject(s)
Genetic Engineering/methods , Herpesvirus 1, Human/genetics , Interleukin-15 Receptor alpha Subunit/metabolism , Interleukin-15/biosynthesis , Interleukin-15/metabolism , Oncolytic Viruses/genetics , Virus Replication , Animals , Cell Line, Tumor , Genes, Viral/genetics , Herpesvirus 1, Human/physiology , Humans , Injections , Interleukin-15/chemistry , Interleukin-15/genetics , Interleukin-15 Receptor alpha Subunit/genetics , Mice , Oncolytic Viruses/physiology , Protein Binding , Solubility
3.
Pediatr Blood Cancer ; 60(1): 45-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22408058

ABSTRACT

BACKGROUND: Rhabdomyosarcoma (RMS) is characterized by features of skeletal muscle and is comprised of two major histological subtypes, embryonal (E-RMS), and alveolar (A-RMS). Subsets of each RMS subtype demonstrate resistance to multimodal therapy leading to treatment failure. Cancer stem cells or cancer-initiating cells (CIC) represent a theorized population of cells that give rise to tumors and are responsible for treatment resistance. PROCEDURE: We investigated the ability of CD133, a putative CIC marker, to distinguish a chemoresistant, myogenically primitive population in alveolar (RH30), and embryonal (RD) RMS cell lines. We tested CD133+/- cells for sensitivity to engineered herpes simplex virus (oHSV). RESULTS: Relative to CD133- cells, CD133+ A-RMS, and E-RMS cells demonstrate an enhanced colony-forming ability, are less differentiated myogenically, and are more resistant to cytotoxic chemotherapy but equally sensitive to oHSV oncolysis. Compared to CD133- RD cells, CD133+ cells express relatively high levels of genes typically expressed in skeletal muscle progenitor satellite cells including PAX7, c-MET, and the GLI effectors of the hedgehog signaling pathway. In contrast, CD133+ RH30 cells were not associated with enhanced expression of satellite cell markers or Hh targets. CONCLUSIONS: Our findings demonstrate that CD133+ cells from A-RMS and E-RMS cell lines are characterized by a myogenically primitive phenotype. These cells have the capacity to form colonies in vitro and are more resistant to chemotherapy than CD133- cells. CD133 expression may denote a subset of RMS cells with an important role in tumorigenesis and treatment failure. These resistant cells may be effectively targeted by oHSV therapy.


Subject(s)
Antigens, CD/analysis , Drug Resistance, Neoplasm , Glycoproteins/analysis , Oncolytic Virotherapy , Peptides/analysis , Rhabdomyosarcoma/therapy , Simplexvirus/genetics , Simplexvirus/physiology , AC133 Antigen , Antigens, CD/physiology , Cell Line, Tumor , Genetic Engineering , Glycoproteins/physiology , Humans , Peptides/physiology , Receptor, Fibroblast Growth Factor, Type 3/analysis , Rhabdomyosarcoma/chemistry , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Signal Transduction
4.
J Virol ; 86(9): 5304-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22379082

ABSTRACT

Herpes simplex virus 1 (HSV-1) mutants that lack the γ(1)34.5 gene are unable to replicate in the central nervous system but maintain replication competence in dividing cell populations, such as those found in brain tumors. We have previously demonstrated that a γ(1)34.5-deleted HSV-1 expressing murine interleukin-12 (IL-12; M002) prolonged survival of immunocompetent mice in intracranial models of brain tumors. We hypothesized that M002 would be suitable for use in clinical trials for patients with malignant glioma. To test this hypothesis, we (i) compared the efficacy of M002 to three other HSV-1 mutants, R3659, R8306, and G207, in murine models of brain tumors, (ii) examined the safety and biodistribution of M002 in the HSV-1-sensitive primate Aotus nancymae following intracerebral inoculation, and (iii) determined whether murine IL-12 produced by M002 was capable of activating primate lymphocytes. Results are summarized as follows: (i) M002 demonstrated superior antitumor activity in two different murine brain tumor models compared to three other genetically engineered HSV-1 mutants; (ii) no significant clinical or magnetic resonance imaging evidence of toxicity was observed following direct inoculation of M002 into the right frontal lobes of A. nancymae; (iii) there was no histopathologic evidence of disease in A. nancymae 1 month or 5.5 years following direct inoculation; and (iv) murine IL-12 produced by M002 activates A. nancymae lymphocytes in vitro. We conclude that the safety and preclinical efficacy of M002 warrants the advancement of a Δγ(1)34.5 virus expressing IL-12 to phase I clinical trials for patients with recurrent malignant glioma.


Subject(s)
Genetic Vectors/genetics , Interleukin-12/genetics , Simplexvirus/genetics , Acyclovir/pharmacology , Animals , Antiviral Agents/pharmacology , Aotidae , Brain/pathology , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical , Female , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Glioma/genetics , Glioma/mortality , Glioma/therapy , Humans , Interleukin-12/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, SCID , Simplexvirus/drug effects , Survival Analysis , Virus Replication/drug effects , Virus Replication/genetics , Xenograft Model Antitumor Assays
5.
Int J Breast Cancer ; 2012: 628697, 2012.
Article in English | MEDLINE | ID: mdl-23346408

ABSTRACT

The metastasis of breast cancer to the brain and central nervous system (CNS) is a problem of increasing importance. As improving treatments continue to extend patient survival, the incidence of CNS metastases from breast cancer is on the rise. New treatments are needed, as current treatments are limited by deleterious side effects and are generally palliative. We have previously described an oncolytic herpes simplex virus (HSV), designated M002, which lacks both copies of the γ(1)34.5 neurovirulence gene and carries a murine interleukin 12 (IL-12) expression cassette, and have validated its antitumor efficacy in a variety of preclinical models of primary brain tumors. However, M002 has not been yet evaluated for use against metastatic brain tumors. Here, we demonstrate the following: both human breast cancer and murine mammary carcinoma cells support viral replication and IL-12 expression from M002; M002 replicates in and destroys breast cancer cells from a variety of histological subtypes, including "triple-negative" and HER2 overexpressing; M002 improves survival in an immunocompetent model more effectively than does a non-cytokine control virus. Thus, we conclude from this proof-of-principle study that a γ(1)34.5-deleted IL-12 expressing oncolytic HSV may be a potential new therapy for breast cancer brain metastases.

6.
Mol Cell Biol ; 31(16): 3196-207, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21690298

ABSTRACT

Inherently unstable mRNAs contain AU-rich elements (AREs) in the 3' untranslated regions. Expression of ARE-containing type I interferon transcripts is robustly induced upon viral infection and rapidly shut off thereafter. Their transient accumulation is partly mediated through posttranscriptional regulation. Here we show that mouse embryonic fibroblasts derived from knockout mice deficient in KH-type splicing regulatory protein (KSRP), an RNA-binding protein required for ARE-mediated mRNA decay, produce higher levels of Ifna and Ifnb mRNAs in response to viral infection as a result of decreased mRNA decay. Functional analysis showed that KSRP is required for the decay of Ifna4 and Ifnb mRNAs by interaction with AREs. The increased IFN expression renders Ksrp(-)(/)(-) cells refractory to herpes simplex virus type 1 and vesicular stomatitis virus infection. These findings support a role of a posttranscriptional mechanism in the control of type I IFN expression and highlight the function of KSRP in innate immunity by negatively regulating IFN production.


Subject(s)
Immunity, Innate , Interferon Type I/genetics , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , RNA Interference/immunology , RNA-Binding Proteins/immunology , Trans-Activators/immunology , Virus Diseases/immunology , Animals , Cells, Cultured , Herpes Simplex/immunology , Mice , Mice, Knockout , RNA Stability/immunology , Rhabdoviridae Infections/immunology
7.
Mol Pharm ; 8(1): 44-9, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21142023

ABSTRACT

Conditionally replication-competent Herpes Simplex Virus Type 1 (HSV-1) vectors expressing foreign genes have been developed as experimental therapeutic agents. Traditional methods of virus construction, including growth selection based on thymidine kinase gene expression, and color selection based on a reporter gene expression are often time-consuming and relatively inefficient. This review summarizes the various strategies developed in recent years for the rapid and efficient construction of novel conditionally replication-competent mutant HSV expressing multiple foreign genes. Additionally, two new modifications of existing strategies, which have not been previously reported, are discussed.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Herpesvirus 1, Human/physiology , Neoplasms/therapy , Virus Replication/genetics , Herpesvirus 1, Human/genetics , Humans , Neoplasms/genetics
8.
J Neurooncol ; 95(2): 199-209, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19521665

ABSTRACT

Oncolytic herpes simplex viruses (HSV) hold promise for therapy of glioblastoma multiforme (GBM) resistant to traditional therapies. We examined the ability of genetically engineered HSV to infect and kill cells that express CD133, a putative marker of glioma progenitor cells (GPC), to determine if GPC have an inherent therapeutic resistance to HSV. Expression of CD133 and CD111 (nectin-1), the major entry molecule for HSV, was variable in six human glioma xenografts, at initial disaggregation and after tissue culture. Importantly, both CD133+ and CD133- populations of glioma cells expressed CD111 in similar relative proportions in five xenografts, and CD133+ and CD133- glioma cell subpopulations were equally sensitive to killing in vitro by graded dilutions of wild-type HSV-1(F) or several different gamma(1)34.5-deleted viruses. GPC did not display an inherent resistance to HSV. While CD111 expression was an important factor for determining sensitivity of glioma cells to HSV oncolysis, it was not the only factor. Our findings support the notion that HSV will not be able to effectively enter, infect, and kill cells in tumors that have low CD111 expression (<20%). However, virotherapy with HSV may be very effective against CD111+ GPC resistant to traditional therapies.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/therapy , Cell Adhesion Molecules/metabolism , Glioblastoma/therapy , Glycoproteins/metabolism , Herpesvirus 1, Human/physiology , Oncolytic Virotherapy , Peptides/metabolism , AC133 Antigen , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Cytopathogenic Effect, Viral , Genetic Engineering/methods , Genetic Vectors , Glioblastoma/metabolism , Glioblastoma/virology , Humans , Mice , Mice, Nude , Nectins , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
9.
Mol Ther ; 17(1): 199-207, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18957964

ABSTRACT

We have previously demonstrated safety of G207, a doubly mutated (deletion of both gamma(1)34.5 loci, insertional inactivation of U(L)39) herpes simplex virus (HSV) for patients stereotactically inoculated in enhancing portions of recurrent malignant gliomas. We have now determined safety of two inoculations of G207, before and after tumor resection. Inclusion criteria were histologically proven recurrent malignant glioma, Karnofsky score >or=70, and ability to resect the tumor without ventricular system breach. Patients received two doses of G207 totaling 1.15 x 10(9) plaque-forming units with 13% of this total injected via a catheter placed stereotactically in the tumor. Two or five days later, tumor was resected en bloc with catheter in place. The balance of G207 dose was injected into brain surrounding the resection cavity. Six patients with recurrent glioblastoma multiforme were enrolled. Two days after the second G207 inoculation, one patient experienced transient fever, delirium, and hemiparesis, which entirely resolved on high-dose dexamethasone. No patient developed HSV encephalitis or required treatment with acyclovir. Radiographic and neuropathologic evidence suggestive of antitumor activity is reported. Evidence of viral replication was demonstrated. G207 appears safe for multiple dose delivery, including direct inoculation into the brain surrounding tumor resection cavity.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Simplexvirus/physiology , Acyclovir/therapeutic use , Adult , Aged , Antibodies, Viral/blood , Brain Neoplasms/drug therapy , Female , Glioblastoma/drug therapy , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Simplexvirus/genetics , Simplexvirus/immunology , Treatment Outcome , Virus Replication
10.
Viruses ; 1(3): 510-22, 2009 12.
Article in English | MEDLINE | ID: mdl-21994558

ABSTRACT

A virulent recombinant HSV lacking the diploid γ(1)34.5 gene (Δγ(1)34.5) have been investigated over the last two decades both for anti-tumor therapy and as vaccine vectors. The first generation vectors, while safe, are incapable of sustained replication in the majority of treated patients. An interferon inducible host antiviral kinase, protein kinase R (PKR), limits late viral protein synthesis and replication of Δγ(1)34.5 viruses. This review describes the development of new Δγ(1)34.5 vectors, through serial passage selection and direct viral genome engineering, which demonstrate selective PKR evasion in targeted cells and improved viral replication without restoring neurovirulence.

11.
Vaccine ; 25(37-38): 6764-73, 2007 Sep 17.
Article in English | MEDLINE | ID: mdl-17706843

ABSTRACT

We have constructed a replication competent, gamma(1)34.5-deleted herpes simplex virus type-1 (HSV-1) vector (J200) that expresses the gag gene from human immunodeficiency virus type-1, primary isolate 89.6 (HIV-1(89.6)), as a candidate vaccine for HIV-1. J200 replicates in vitro, resulting in abundant Gag protein production and accumulation in the extracellular media. Immunization of Balb/c mice with a single intraperitoneal injection of J200 elicited strong Gag-specific CD8 responses, as measured by intracellular IFN-gamma staining and flow cytometry analysis. Responses were highest between 6 weeks and 4 months, but persisted at 9 months post-immunization, the last time-point evaluated. These data highlight the potential utility of neuroattenuated, replication competent HSV-1 vectors for delivery of HIV-1 immunogens.


Subject(s)
Gene Expression Regulation, Viral , Gene Products, gag/immunology , Gene Products, gag/metabolism , Genetic Vectors/genetics , HIV-1/physiology , Virus Replication , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Chlorocebus aethiops , Female , Gene Products, gag/genetics , Immunization , Mice , Mice, Inbred BALB C
12.
Herpes ; 13(3): 66-71, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17147910

ABSTRACT

The prognosis for patients diagnosed with malignant glioma, the most common primary tumour of the central nervous system, remains poor despite decades of research and advances in surgery, radiation and chemotherapy. The development of new approaches for the treatment of these tumours has led to the emergence of oncolytic viral therapy, with the use of conditionally replicating viruses, as a potential new intervention. Herpes simplex virus type 1 has emerged as the leading candidate oncolytic virus, with six different trials either completed or underway for patients with malignant glioma. In this review, the background of this approach will be discussed, followed by a discussion of the clinical trials, as well as potential directions for future trials.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy/methods , Clinical Trials as Topic , Genetic Therapy , Genetic Vectors , Humans
13.
J Virol ; 80(15): 7308-15, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16840311

ABSTRACT

Previous studies have described in vitro serial passage of a Deltagamma(1)34.5 herpes simplex virus type 1 (HSV-1) strain in SK-N-SH neuroblastoma cells and selection of mutants that have acquired the ability to infect and replicate in this previously nonpermissive cell line. Here we describe the selection of a mutant HSV-1 strain by in vivo serial passage, which prolongs survival in two separate experimental murine brain tumor models. Two conditionally replication-competent Deltagamma(1)34.5 viruses, M002, which expresses murine interleukin-12, and its parent virus, R3659, were serially passaged within human malignant glioma D54-MG cell lines in vitro or flank tumor xenografts in vivo. The major findings are (i) viruses passaged in vivo demonstrate decreased neurovirulence, whereas those passaged in vitro demonstrate a partial recovery of the neurovirulence associated with HSV-1; and (ii) vvD54-M002, the virus selected after in vivo serial passage of M002 in D54-MG tumors, improves survival in two independent murine brain tumor models compared to the parent (unpassaged) M002. Additionally, in vitro-passaged, but not in vivo-passaged, M002 displayed changes in the protein synthesis profile in previously nonpermissive cell lines, as well as early U(S)11 transcription. Thus, a mutant HSV-1 strain expressing a foreign gene can be selected for enhanced antitumor efficacy via in vivo serial passage within flank D54-MG tumor xenografts. The enhanced antitumor efficacy of vvD54-M002 is not due to restoration of protein synthesis or early U(S)11 expression. This finding emphasizes the contribution of the in vivo tumor environment for selecting novel oncolytic HSV specifically adapted for tumor cell destruction in vivo.


Subject(s)
Brain Neoplasms/virology , Glioma/virology , Herpesvirus 1, Human/pathogenicity , Neuroblastoma/therapy , Virulence/physiology , Animals , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Herpesvirus 1, Human/physiology , Humans , Interleukin-12/metabolism , Mice , Mice, Inbred A , Mice, Inbred CBA , Mice, SCID , Mutation/genetics , Neoplasms, Experimental/virology , Neuroblastoma/metabolism , Neuroblastoma/virology , Transplantation, Heterologous , Tumor Cells, Cultured , Vero Cells , Virus Replication
14.
Vaccine ; 24(10): 1644-52, 2006 Mar 06.
Article in English | MEDLINE | ID: mdl-16243413

ABSTRACT

We are using genetically modified, conditionally replicating herpes simplex virus (HSV) that express either interleukin (IL)-12 or granulocyte macrophage-colony stimulating factor (GM-CSF) as live, attenuated vaccine candidates for protection against HSV infection and/or disease. We report the following: (1) animals previously vaccinated with these candidate vaccines exhibited dose-dependent protection after intranasal, intraperitoneal or intracranial challenge with the highly virulent E377-MB wild-type HSV-1; (2) the IL-12 expressing virus (M002) consistently conferred protection at lower immunization doses than GM-CSF expressing virus (M004); (3) between 80 and 100% protection from E377-MB challenge was conferred after intramuscular immunization of mice with any of the three Deltagamma1 34.5 HSV, as opposed to 50% protection elicited after immunization with wild-type HSV-1 (F); and (4) latent virus was not detected at a higher rate in animals immunized and subsequently challenged with E377-MB than in immunized animals alone. These data suggest that conditionally replicating, cytokine-expressing HSV are able to elicit protective immune responses while retaining safety in an experimental murine model.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Herpes Simplex Virus Vaccines/immunology , Interleukin-12/genetics , Simplexvirus/genetics , Vaccines, Synthetic/immunology , Animals , Female , Genetic Engineering , Herpes Simplex Virus Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Simplexvirus/pathogenicity , Simplexvirus/physiology , Trigeminal Ganglion/virology , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Virulence , Virus Activation , Virus Replication
15.
Neuro Oncol ; 7(3): 213-24, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16053696

ABSTRACT

Long-term survivors of glioblastoma multiforme, the most common form of primary intracranial malignancy in adults, are extremely rare. Experimental animal models that more closely resemble human disease are essential for the identification of effective novel therapies. We report here an extensive analysis of the 4C8 glioma model to assess its suitability for evaluating novel type 1 herpes simplex virus (HSV-1) therapies of malignant glioma. We first determined that expression of major histocompatibility complex I and II and of alphavbeta3 in the 4C8 model was comparable to that seen in human glioma cells. Next, using a panel of Delta(gamma1)34.5 HSVs, we demonstrated that, in vitro, 4C8 cells were as sensitive as human glioma cells to both infection and lysis and that the 4C8 cells supported the production of foreign gene products. Replication competence of HSV was demonstrated in vitro. Finally, 4C8 intracranial gliomas were established in immunologically competent syngeneic B6D2F1 mice, treated by intratumoral injection of selected engineered HSVs, including the interleukin-12-expressing virus, M002. Survival data from these studies demonstrated that 4C8 cells in vivo are sensitive to both direct oncolysis and HSV-mediated interleukin-12 expression. Fluorescence-activated cell sorting analyses of immune-related infiltrating cells supported the concept that survival was prolonged in part because of antitumor actions of these cells. We conclude that the 4C8/B6D2F1 syngeneic glioma model is suitable for preclinical evaluation of HSV-based therapies and that M002 is a superior virus for the treatment of murine glioma in this model.


Subject(s)
Brain Neoplasms/virology , Disease Models, Animal , Glioma/virology , Interleukin-12/metabolism , Simplexvirus/physiology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Cell Line, Tumor , Flow Cytometry , Genetic Therapy , Glioma/metabolism , Glioma/therapy , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class II/biosynthesis , Humans , Mice , Transplantation, Isogeneic
16.
J Infect Dis ; 192(1): 156-61, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15942905

ABSTRACT

Herpes simplex virus type 2 (HSV-2) resistance to antiviral drugs has been described primarily in immunocompromised patients. We report an apparently immunocompetent, human immunodeficiency virus-negative male patient who has experienced repeated HSV-2 genital outbreaks despite receiving antiviral prophylaxis with several different drugs. Several of the HSV-2 genital isolates from this patient have been confirmed as resistant to acyclovir and penciclovir. Antiviral resistance occurred in the setting of long-term prednisone treatment and intermittent acyclovir prophylaxis at suboptimal doses and persisted despite the cessation of oral steroid treatment. The patient's genital herpes outbreaks were not controlled by high-dose prophylaxis with acyclovir, valacyclovir, and famciclovir. Cessation of antiviral prophylaxis resulted in reversion of this patient's HSV-2 isolates to acyclovir and penciclovir sensitivity, although resistant virus reappeared when antiviral prophylaxis was resumed. Transmission of a sensitive HSV-2 strain from this patient to a female sex partner was observed. These observations confirm previous reports that resistance to acyclovir may develop during prophylactic therapy in an otherwise well, immunocompetent patient. These findings support the conclusion that both drug-sensitive and drug-resistant HSV-2 strains established latency in this patient and that both strains are capable of frequent reactivation.


Subject(s)
Acyclovir/analogs & derivatives , Acyclovir/pharmacology , Antiviral Agents/pharmacology , Drug Resistance, Multiple, Viral , Herpes Genitalis/virology , Herpesvirus 2, Human/drug effects , Adult , Female , Guanine , Herpes Genitalis/drug therapy , Herpes Genitalis/transmission , Humans , Immunocompetence , Male , Recurrence
17.
Cancer Gene Ther ; 12(4): 359-68, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15678154

ABSTRACT

Herpes simplex viruses type 1 (HSV-1) that lack the gamma(1)34.5 gene are unable to replicate in the central nervous system (CNS), but maintain replication competence in actively dividing tumors. To determine if antitumor therapy by M002, a gamma(1)34.5(-) HSV that expresses interleukin-12 (IL-12), could be augmented by combinatorial therapy with another gamma(1)34.5-deleted HSV-1 engineered to express the chemokine CCL2, Neuro-2a tumors were established subcutaneously in the syngeneic A/J mouse strain. Tumors received multiple injections intratumorally either of saline, the parent, non-cytokine-expressing virus R3659, M002, M010 (gamma(1)34.5(-) HSV expressing CCL2), or a combination of M002 and M010. Efficacies were evaluated by monitoring inhibition of tumor growth over time. Results demonstrated the following: (1) inhibition of tumor growth was most pronounced in tumors treated with a combination of M002 and M010; (2) enhanced tumor growth inhibition for the combinatorial treatment group was statistically significant compared to either M002 or M010 alone; and (3) the variability between slopes of the tumor growth rates within an individual treatment group appeared to be virus-dependent, and was reproducible between experiments. Our results demonstrate that combinatorial cytokine/chemokine gamma(1)34.5(-) HSV therapies can provide superior antitumor effects in experimental tumors as a model for malignancies arising in the brain.


Subject(s)
Cancer Vaccines , Chemokine CCL2/genetics , Genetic Therapy , Herpesvirus 1, Human/genetics , Immunotherapy/methods , Interleukin-12/genetics , Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Central Nervous System/metabolism , Chlorocebus aethiops , Combined Modality Therapy , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Genetic Engineering , Immunohistochemistry , Mice , Models, Genetic , Neoplasm Transplantation , Neoplasms/therapy , Plasmids/metabolism , Time Factors , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...