Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Eur J Cancer ; 51(16): 2321-9, 2015 Nov.
Article En | MEDLINE | ID: mdl-26283035

BACKGROUND: Treatment options after first-line chemotherapy are limited in non-small cell lung cancer (NSCLC). Belagenpumatucel-L is a therapeutic vaccine comprised of 4 transforming growth factor (TGF)-ß2-antisense gene-modified, irradiated, allogeneic NSCLC cell lines that may be useful for maintenance after initial treatment. METHODS: Stage III/IV NSCLC patients who did not progress after platinum-based chemotherapy were randomised 1:1 to receive maintenance belagenpumatucel-L or placebo. Patients were eligible for randomisation between one and four months from the end of induction chemotherapy. The primary endpoint was overall survival. RESULTS: This phase III trial enrolled 270 patients in the belagenpumatucel-L arm and 262 in the control arm. Belagenpumatucel-L was well tolerated with no serious safety concerns. There was no difference in survival between the arms (median survival 20.3 versus 17.8months with belagenpumatucel-L versus placebo, respectively; hazard ratio (HR) 0.94, p=0.594). There were also no differences in progression-free survival (4.3months versus 4.0 for belagenpumatucel-L vs placebo, respectively; HR 0.99, p=0.947). A prespecified Cox regression analysis demonstrated that the time elapsed between randomisation and the end of induction chemotherapy had a significant impact on survival (p=0.002) and that prior radiation was a positive prognostic factor (median survival 28.4months with belagenpumatucel-L versus 16.0months with placebo; HR 0.61, p=0.032). CONCLUSIONS: Although the overall trial did not meet its survival endpoint, improved survival for belagenpumatucel-L is suggested in patients who were randomised within 12weeks of completion of chemotherapy and in those who had received prior radiation. Further studies of belagenpumatucel-L in NSCLC are warranted.


Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Maintenance Chemotherapy/methods , Adult , Aged , Cancer Vaccines/adverse effects , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Disease-Free Survival , Double-Blind Method , Female , Humans , Intention to Treat Analysis , Kaplan-Meier Estimate , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Proportional Hazards Models , Time Factors , Treatment Outcome
2.
Cancer Gene Ther ; 16(8): 620-4, 2009 Aug.
Article En | MEDLINE | ID: mdl-19287371

In a previous dose escalation trial we demonstrated dose related survival correlation to Belagenpumatucel-L. In order to further evaluate safety and response at the previously defined optimal dose and schedule and to gain preliminary evidence on a hypothesis that the level of circulating tumor cells (CTCs) in blood may correlate with the overall survival of patients with stage IV NSCLC, we initiated a phase II trial. Patients received intradermal immunization of 2.5 x 10(7) transfected allogeneic tumor cells (Belagenpumatucel-L, supplied by NovaRx) 1 x every month for a total of 16 months. Circulating tumor cells (Veridex, Raritan, NJ) were measured every 4 weeks. Twenty-one advanced NSCLC patients were enrolled on this study. No significant toxic effect was observed. Overall survival was 562 days. The median survival was 660 days in patients having less than 2 CTCs at baseline compared to 150 days in patients with 2 or more CTCs (P=0.025). Phase II results of safety and response are consistent with prior experience following treatment with Belagenpumatucel-L and there is a suggestion that the number of circulating tumor cells at baseline appears to correlate with overall survival. A larger clinical trial is warranted to further explore this observation.


Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Transforming Growth Factor beta2/antagonists & inhibitors , Adult , Aged , Cancer Vaccines/adverse effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , DNA, Antisense/biosynthesis , Disease Progression , Dose-Response Relationship, Drug , Female , Genetic Therapy/methods , Humans , Injections, Intradermal , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Transforming Growth Factor beta2/genetics , Tumor Cells, Cultured
3.
Cancer Gene Ther ; 13(12): 1052-60, 2006 Dec.
Article En | MEDLINE | ID: mdl-16826191

We performed a phase I clinical trial in grade IV astrocytoma to assess the safety of a whole-cell vaccine comprising autologous tumor cells genetically modified by a transforming growth factor-beta2 (TGF-beta2) antisense vector. Blocking secretion of the immunosuppressive molecule TGF-beta in this manner should inhibit one of the major mechanisms by which tumor cells evade immune surveillance and should lead to clinically effective antitumor immunity. Six patients with progressive WHO grade IV astrocytoma were enrolled in the trial. Patients received 2-7 subcutaneous injections of 5 x 10(6)-2 x 10(7) autologous tumor cells per injection. TGF-beta2 secretion by the tumor cells used to vaccinate patients was inhibited by 53-98%. Treatment was well tolerated with only low-grade, transient treatment-related toxicities reported. Two patients had partial regressions and two had stable disease following therapy. The overall median survival was 68 weeks. Median survival of the responding patients was 78 weeks, compared to a historic value of 47 weeks for glioma patients treated conventionally. There were indications of humoral and cellular immunity induced by the vaccine. These findings support further clinical evaluation of vaccines comprised of TGF-beta antisense-modified tumor cells.


Cancer Vaccines/therapeutic use , Central Nervous System Neoplasms/drug therapy , Glioma/drug therapy , Oligonucleotides, Antisense/genetics , Transforming Growth Factor beta2/genetics , Adult , Antibody Formation , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Female , Glioma/immunology , Glioma/pathology , Humans , Injections, Intradermal , Male , Middle Aged , Transforming Growth Factor beta2/drug effects , Transforming Growth Factor beta2/metabolism , Treatment Outcome , Tumor Cells, Cultured
4.
Clin Cancer Res ; 5(9): 2359-65, 1999 Sep.
Article En | MEDLINE | ID: mdl-10499605

The purpose of this study was to determine the safety, toxicity, and antitumor immune response following S.C. immunizations with a mixture of irradiated, autologous tumor cells and autologous fibroblasts that were genetically modified to express the gene for interleukin 2 (IL-2) in patients with colorectal carcinoma. Ten patients were treated with a fixed dose of tumor cells (10(7)) and escalating doses of fibroblasts secreting IL-2 (per 24 h): 100 units (three patients), 200 units (three patients), 400 units (three patients), and 800 units (one patient). Pre- and posttreatment peripheral blood mononuclear cells were evaluated for evidence of antitumor immune responses. Fatigue and/or flu-like symptoms were experienced by seven patients and delayed-type hypersensitivity-like skin reactions were observed at the sites of the second or subsequent vaccinations in five patients. Low frequencies of tumor cytotoxic T-cell precursors (range, 1/190,000-1/1,320,000 peripheral blood mononuclear cells) were detected prior to therapy in four of seven patients. There was a 5-fold increase following treatment in the frequency of tumor cytotoxic T-cell precursors in two of six evaluable patients. Some patients with colorectal cancer have low frequencies of tumor cytotoxic T-cell precursors that may be increased by this well-tolerated form of IL-2 gene therapy, which warrants continued clinical evaluation.


Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Fibroblasts/metabolism , Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Interleukin-2/biosynthesis , Interleukin-2/genetics , Cancer Vaccines/immunology , Cell Transplantation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Combined Modality Therapy , Fibroblasts/physiology , Fibroblasts/transplantation , Genetic Engineering , Genetic Therapy/adverse effects , Humans , Hypersensitivity, Delayed/etiology , Hypersensitivity, Delayed/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/radiation effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/radiation effects , T-Lymphocytes, Cytotoxic/transplantation
5.
J Biol Chem ; 274(34): 24187-94, 1999 Aug 20.
Article En | MEDLINE | ID: mdl-10446193

Human polydeoxyribonucleotide kinase is an enzyme that has the capacity to phosphorylate DNA at 5'-hydroxyl termini and dephosphorylate 3'-phosphate termini and, therefore, can be considered a putative DNA repair enzyme. The enzyme was purified from HeLa cells. Amino acid sequence was obtained for several tryptic fragments by mass spectrometry. The sequences were matched through the dbEST data base with an incomplete human cDNA clone, which was used as a probe to retrieve the 5'-end of the cDNA sequence from a separate cDNA library. The complete cDNA, which codes for a 521-amino acid protein (57.1 kDa), was expressed in Escherichia coli, and the recombinant protein was shown to possess the kinase and phosphatase activities. Comparison with other sequenced proteins identified a P-loop motif, indicative of an ATP-binding domain, and a second motif associated with several different phosphatases. There is reasonable sequence similarity to putative open reading frames in the genomes of Caenorhabditis elegans and Schizosaccharomyces pombe, but similarity to bacteriophage T4 polynucleotide kinase is limited to the kinase and phosphatase domains noted above. Northern hybridization revealed a major transcript of approximately 2.3 kilobases and a minor transcript of approximately 7 kilobases. Pancreas, heart, and kidney appear to have higher levels of mRNA than brain, lung, or liver. Confocal microscopy of human A549 cells indicated that the kinase resides predominantly in the nucleus. The gene encoding the enzyme was mapped to chromosome band 19q13.4.


Polynucleotide 5'-Hydroxyl-Kinase/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary/isolation & purification , HeLa Cells , Humans , Molecular Sequence Data , Polynucleotide 5'-Hydroxyl-Kinase/chemistry , Polynucleotide 5'-Hydroxyl-Kinase/isolation & purification , Rabbits
6.
Neurol Res ; 20(8): 742-7, 1998 Dec.
Article En | MEDLINE | ID: mdl-9864741

Using an intracranial rat C6 glioma model, we tested the hypothesis that gene modification of glioma cells to block the expression of the immunosuppressive cytokine TGF-beta (transforming growth factor beta) may enhance anti-tumor immune responses and thereby prolong survival of tumor-bearing animals. The cDNA for simian TGF-beta 2 was ligated in antisense orientation into the episomal plasmid mammalian expression vector pCEP-4. This TGF-beta-antisense vector was transfected into C6 glioma cells by standard electroporation techniques. PCR was used to determine that the rat C6 clones were successfully transfected with the antisense-TGF beta construct. Twenty-nine adult female Wistar rats harboring 7-day-old intracranial C6 tumors were then subcutaneously injected with either saline (n = 9), unmodified C6 glioma cells (n = 10), or TGF-beta-antisense-modified C6 cells (n = 10). Animals were followed for survival, and Fisher's exact method was used to interpret the significance of difference between experimental groups. The survival of tumor-bearing rats injected with TGF-beta-antisense-modified C6 cells was significantly prolonged, relative to the survival of rats receiving injections of saline or unmodified C6 cells alone. Six of the ten (60%) TGF-beta-antisense treated animals survived for 12 weeks, whereas none of the nine (0%) animals treated with saline and none of ten (0%) of those treated with C6 cells alone survived past 5 weeks. These results indicate that the genetic inhibition of immunosuppressive cytokines (such as TGF-beta) may reverse the phenotypic immunosuppression caused by such factors, and thereby prolong the survival of C6 tumor-bearing animals. Future investigations using cytokine gene modifications in other brain tumor models are warranted.


Brain Neoplasms/therapy , Genetic Therapy , Glioma/therapy , Transforming Growth Factor beta/genetics , Animals , Antisense Elements (Genetics) , Brain/pathology , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Female , Glioma/immunology , Glioma/mortality , Necrosis , Phenotype , Polymerase Chain Reaction , Rats , Rats, Wistar , Survival Analysis , Transfection , Transforming Growth Factor beta/immunology , Treatment Outcome
7.
Gynecol Oncol ; 71(2): 204-10, 1998 Nov.
Article En | MEDLINE | ID: mdl-9826461

The immunosuppressive protein transforming growth factor beta (TGF-beta) inhibits the activation of various immune effector cells including cytotoxic T lymphocytes and may therefore inhibit the efficacy of immunostimulatory interleukin-2 (IL-2) gene therapy. In this study, we investigated the effect of TGF-beta downregulation on IL-2 gene therapy in the intraperitoneal model of murine ovarian teratoma (MOT). MOT cells, like many human ovarian carcinomas, were found to produce TGF-beta. Production of TGF-beta by MOT cells was suppressed using a TGF-beta antisense plasmid vector (pCEP4/TGF-beta antisense). Subcutaneous immunization of C3H mice with a mixture of IL-2 gene-transduced fibroblasts and TGF-beta antisense-modified MOT cells induced significantly better protection against a subsequent intraperitoneal tumor challenge compared with immunization with unmodified MOT cells alone [11/16 (69%) vs 4/21 (19%) tumor-free animals, P < 0.01]. Immunization with either a mixture of IL-2 gene modified fibroblasts and unmodified MOT cells [2/12 (17%) tumor-free animals] or TGF-beta antisense-modified MOT cells alone (0/13 tumor free animals) failed to induce significant protection compared with immunization with unmodified MOT cells. These data show that combined TGF-beta antisense and IL-2 gene therapy is required to generate effective antitumor responses in the MOT model. Our findings suggest that tumor cell expression of immunosuppressive factors may inhibit cytokine immunogene therapy and may have potential implications for the development of future clinical immunogene therapy protocols.


Antisense Elements (Genetics)/therapeutic use , Cancer Vaccines/therapeutic use , Genetic Therapy , Interleukin-2/genetics , Ovarian Neoplasms/therapy , Teratoma/therapy , Transforming Growth Factor beta/antagonists & inhibitors , 3T3 Cells , Animals , Down-Regulation , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
8.
In Vitro Cell Dev Biol Anim ; 34(6): 455-62, 1998 Jun.
Article En | MEDLINE | ID: mdl-9661048

Six human glioma cell lines were established from tissues obtained from five patients diagnosed with Kernohan grade IV glioblastoma multiforme and one from a patient with a grade II astrocytoma. One line was from a recurrent patient who had received prior therapy; the other lines were derived from patients at initial diagnosis and/or before cytoreductive therapies other than surgery were given. Considerable variability in phenotypic, karyotypic, and cell surface marker expression was displayed between the six human glioma cell lines. The karyotypes ranged from apparently normal (grade II astrocytoma) to those with complex rearrangements. Trisomy of chromosome 7 was the most common abnormality. The extensive cytogenetic and molecular characterization of these lines may facilitate their utilization in cellular and molecular biologic studies.


Brain Neoplasms , Glioblastoma , Adult , Aged , Animals , Astrocytoma/classification , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Biomarkers, Tumor/metabolism , Brain Neoplasms/classification , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , DNA, Neoplasm/analysis , Female , Glioblastoma/classification , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Karyotyping , Male , Middle Aged , Polymerase Chain Reaction , Receptors, Platelet-Derived Growth Factor/metabolism , Transforming Growth Factor beta/analysis , Tumor Cells, Cultured
9.
J Immunother ; 20(6): 437-48, 1997 Nov.
Article En | MEDLINE | ID: mdl-9409449

Several investigators have employed interleukin-2 (IL-2) gene transfer to enhance the immunogenicity of tumor cell vaccines. We describe in this report the construction and characterization of retroviral vectors for IL-2 gene therapy. Human IL-2 cDNA with a chimeric rat preproinsulin/IL-2 DNA leader sequence was subcloned into the pLXSN (long terminal repeat promoter) and pLNCX (cytomegalovirus [CMV] promoter) vectors to generate the plasmids pLXSN-iIL2 and pLNCX-iIL2, respectively. Human IL-2 cDNA with a chimeric human tissue factor/IL-2 DNA leader sequence was utilized to construct the vector pLXSN-tIL2. The levels of IL-2 secreted by transduced tumor cells and fibroblasts were evaluated by enzyme-linked immunosorbent assay (ELISA) of culture supernatants and compared with those of normal peripheral blood mononuclear cells (PBMC) activated in vitro with calcium ionophore and phorbol 12-myristate 13-acetate. The highest levels of IL-2 secreted by transduced tumor cells (760 units/10(6) cells/24 h), adult fibroblasts (625 units/10(6) cells/24 h), and embryonic fibroblasts (3,975 units/10(6) cells/24 h) were 150- to 1,000-fold higher than than secreted by the activated PBMC (4 units/10(6) cells/24 h). Similar levels of IL-2 were expressed by human fibroblasts transduced with pLXSN vectors employing the preproinsulin (pLXSN-iIL2) or tissue factor (pLXSN-tIL2) leader sequences (range in IL-2 units/10(6) cells/24 h pLXSN-iIL2 = 375-625 vs. pLXSN-tIL2 = 90-440). Because IL-2-transduced cells for clinical applications are generally irradiated to prevent cellular proliferation, we evaluated the effects of radiation on IL-2 production. Radiation doses between 1,500 and 10,000 cGy resulted in gradual decreases in IL-2 secretion by transduced cells. The range of the decrease in IL-2 secretion was 7-11% by day 7, 0-29% by day 14, and 25-50% by day 35. For clinical applications, stable production of the vector in high concentrations is an important consideration. The retroviral vector pLXSN-tIL2 produced the highest viral titer and was chosen for further characterization. Southern blot analysis of SacI-digested genomic DNA from the LXSN-tIL2 producer cell line and SacI-digested pLXSN-tIL2 plasmid DNA revealed the expected 3.2-kbp fragment, suggesting the absence of transgene rearrangement and the suitability of this vector as a candidate for clinical applications.


Genetic Therapy , Genetic Vectors , Interleukin-2/genetics , Retroviridae/genetics , 3T3 Cells , Animals , Base Sequence , Cell Line , Gene Expression , Gene Transfer Techniques , Humans , Insulin , Lymphocytes/metabolism , Mice , Plasmids , Polymerase Chain Reaction , Proinsulin/genetics , Protein Precursors/genetics , RNA, Messenger/analysis , Rats , Thromboplastin/genetics
11.
Oncol Rep ; 4(1): 135-8, 1997.
Article En | MEDLINE | ID: mdl-21590028

We previously demonstrated the efficacy of a genetically engineered vaccine composed of syngeneic tumor cells mixed with syngeneic, IL-2 gene transduced fibroblasts in the CT-26 model of murine colorectal carcinoma. In this report, we describe a more practical approach to fibroblast mediated IL-2 gene therapy that employs syngeneic tumor cells mixed with allogeneic, IL-2 gene transduced fibroblasts. BALB/c mice were challenged with an injection of CT-26, 14 days following immunization with IL-2 modified syngeneic BALB/c 3T3 (H-2(d)) or allogeneic C3H 3T3 (H-2(k)) fibroblasts mixed with irradiated CT-26. Both syngeneic and allogeneic IL-2 modified fibroblasts provided significantly better protection compared to animals treated with control fibroblasts (syngeneic IL-2 fibroblasts 32/40-80% vs. control 15/45-33%, p<0.01; allogeneic IL-2 fibroblasts 25/37-68% vs. control 15/45-3345, p<0.01). There was no statistically significant difference between the groups immunized with syngeneic or allogeneic IL-2 modified fibroblasts. These findings support the evaluation of allogeneic IL-2 modified fibroblasts as a practical form of cytokine gene therapy for cancer.

12.
Int J Cancer ; 66(5): 669-77, 1996 May 29.
Article En | MEDLINE | ID: mdl-8647631

Numerous established human tumor lines co-express platelet-derived growth factor (PDGF) and cognate receptors, suggesting that an autocrine and/or paracrine growth mechanism may be a causal or contributing mechanism to their transformed phenotype. Indeed, it is known that a PDGF-autocrine system is functional in several established tumor lines, especially in human gliomas, and a model for a functional paracrine mechanism has been established in a human melanoma line. However, at least 168 human cell lines representing 26 different human tumor types have been reported to continuously express PDGF-A and/or -B chains, and 55 of these also express PDGF receptors. For the majority of these cases, the significance of co-expression and the relative roles of autocrine and paracrine mechanisms in transformation remains unclear. Here, we show that human glioblastoma T98G cells co-express PDGF-B/c-sis and moderate levels of the cognate beta-type PDGF receptor (PR-beta) but are not tumorigenic in athymic mice. In contrast, human breast carcinoma MCF-7 cells do not express PR-beta and are tumorigenic. Clonal lines of each cell type with greatly increased secretion of p16w(T98Gsis and MCF-7sis cells) were characterized. T98Gsis cells are 85% tumorigenic and occasionally develop pulmonary metastases, showing that endogenous PR-beta can mediate complete transformation upon sufficient stimulation. In contrast, MCF-7sis cells exhibit some growth slowing in vitro and an exactly proportional decrease in tumor growth rate. We conclude that a PDGF-autocrine, and not a paracrine, mechanism best accounts for the acquired tumorigenicity of T98Gsis cells, thereby emphasizing the potential significance of expression of even moderate levels of PR-beta by human tumor cells.


Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Cell Transformation, Neoplastic/pathology , Glioblastoma/pathology , Glioblastoma/ultrastructure , Platelet-Derived Growth Factor/physiology , Proto-Oncogene Proteins/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Animals , Antibodies, Neoplasm/pharmacology , Antibody Specificity , Breast Neoplasms/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Female , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phenotype , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Precipitin Tests , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-sis , Receptors, Platelet-Derived Growth Factor/biosynthesis , Transfection , Tumor Cells, Cultured
13.
Proc Natl Acad Sci U S A ; 93(7): 2909-14, 1996 Apr 02.
Article En | MEDLINE | ID: mdl-8610141

Like human gliomas, the rat 9L gliosarcoma secretes the immunosuppressive transforming growth factor beta (TGF-beta). Using the 9L model, we tested our hypothesis that genetic modification of glioma cells to block TGF-beta expression may enhance their immunogenicity and make them more suitable for active tumor immunotherapy. Subcutaneous immunizations of tumor-bearing animals with 9L cells genetically modified to inhibit TGF-beta expression with an antisense plasmid vector resulted in a significantly higher number of animals surviving for 12 weeks (11/11, 100%) compared to immunizations with control vector-modified 9L cells (2/15, 13%) or 9L cells transduced with an interleukin 2 retroviral vector (3/10, 30%) (P < 0.001 for both comparisons). Histologic evaluation of implantation sites 12 weeks after treatment revealed no evidence of residual tumor. In vitro tumor cytotoxicity assays with lymph node effector cells revealed a 3- to 4-fold increase in lytic activity for the animals immunized with TGF-beta antisense-modified tumor cells compared to immunizations with control vector or interleukin 2 gene-modified tumor cells. These results indicate that inhibition of TGF-beta expression significantly enhances tumor-cell immunogenicity and supports future clinical evaluation of TGF-beta antisense gene therapy for TGF-beta-expressing tumors.


Brain Neoplasms/therapy , DNA, Antisense/therapeutic use , Genetic Therapy , Gliosarcoma/therapy , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Animals , Brain Neoplasms/pathology , Cytotoxicity, Immunologic , DNA, Antisense/administration & dosage , Gene Expression/drug effects , Genetic Therapy/methods , Genetic Vectors , Gliosarcoma/pathology , Humans , Immunotherapy/methods , Interleukin-2/biosynthesis , Lymphocytes/immunology , Plasmids , Rats , Rats, Inbred F344 , Retroviridae , Time Factors
14.
Cancer Res ; 56(2): 280-6, 1996 Jan 15.
Article En | MEDLINE | ID: mdl-8542581

Autocrine stimulation by platelet-derived growth factor-B (PDGF-B)-like factors has been widely implicated as an important mechanism in the cause and/or maintenance of a variety of human tumors. However, normal human cells appear to be resistant to transformation by PDGF-B-like molecules, and a direct demonstration of the tumor-promoting or tumor-maintaining property of a PDGF-B autocrine system is lacking. T98G human glioblastoma cells are nontumorigenic in athymic mice. We show that these cells express predominantly PDGF-beta type receptors and continuously secrete small amount of PDGF-B/c-sis. Addition of suramin or specific anti-PDGF-B/v-sis antibody inhibits proliferation in culture. Conversely, multiple clonal lines that stably overexpress PDGF-B/v-sis (T98Gsis cells) exhibit a striking 200-250% increased proliferation rate and an enhanced colony-forming frequency in soft agar. Clonal lines with stable expression of PDGF-B/v-sis (T98Gsis cells) reliably (80%) develop tumors in 4-6 weeks, whereas the empty-vector control cells are nontumorigenic. Moreover, in some cases, T98Gsis cells disseminate to form bilateral and multifocal pulmonary metastases. The results show that T98G cells contain functional PDGF receptors that, upon sufficient stimulation, can cause greatly increased mitogenic response, which may account for the development of the malignant phenotype. Metastatic tumor formation in athymic mice by PDGF stimulation has not been reported previously. The mechanism may depend on preexisting changes such as the lost p53 function of these cells. T98Gsis cells provide a model of growth factor-dependent tumorigenesis and metastases, which may be helpful in elucidating these relationships.


Glioblastoma/pathology , Platelet-Derived Growth Factor/physiology , Proto-Oncogene Proteins/physiology , Retroviridae Proteins, Oncogenic/physiology , Animals , Antibodies, Neoplasm/pharmacology , Antibody Specificity , Cell Division/physiology , Cell Transformation, Neoplastic , Female , Glioblastoma/metabolism , Glioblastoma/secondary , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Middle Aged , Oncogene Proteins v-sis , Phenotype , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-sis , Retroviridae Proteins, Oncogenic/biosynthesis , Retroviridae Proteins, Oncogenic/genetics , Transfection , Tumor Cells, Cultured
15.
Hum Gene Ther ; 6(5): 591-601, 1995 May.
Article En | MEDLINE | ID: mdl-7578396

We evaluated the effects of different doses of interleukin-2 (IL-2)-transduced fibroblasts in the treatment of colorectal carcinoma in the CT-26 murine tumor model. Immunization with a mixture of irradiated tumor cells and IL-2-transduced fibroblasts (100 units of IL-2/24 hr) induced significantly greater protection against a live tumor challenge compared to irradiated tumor cells alone (22/35, 65% vs. 10/30, 33%, p < 0.02). Protective effects were observed with doses of IL-2-transduced fibroblasts secreting from 5 to 100 units of IL-2/24 hr. Parallel experiments in nude mice produced no protection, indicating that the effects of immunization were mediated by a T-cell-dependent mechanism. In animals with established tumors, complete tumor remissions were observed following immunization with a mixture of irradiated tumor cells and IL-2-transduced fibroblasts secreting 100 units of IL-2/24 hr, but not after immunization with irradiated tumor cells alone (7/16 vs. 0/11 complete remissions, p < 0.02). Fibroblasts secreting higher doses of IL-2 were ineffective in generating systemic immunity, but were required to prevent tumor implantation. A statistically significant difference in the prevention of tumor implantation was observed between groups inoculated with a mixture of live tumor cells and IL-2-transduced fibroblasts (1,750 units of IL-2/24 hr) compared to control fibroblasts (6/8 vs. 0/12, p < 0.001). Similar results were observed in nude mice, suggesting that the implantation rejection response is mediated in part by cells other than thymus-derived T cells. Our data support the utility of IL-2-transduced fibroblasts and indicate that the level of IL-2 expression is an important variable in activating different effector components of antitumor immune responses in IL-2 gene therapy.


Colorectal Neoplasms/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Interleukin-2/genetics , T-Lymphocytes/immunology , Adenocarcinoma/pathology , Animals , Cell Line , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Cytotoxicity, Immunologic , Fibroblasts , Gene Expression , Genetic Vectors , Humans , Immunity, Cellular , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Retroviridae/genetics
16.
J Immunother Emphasis Tumor Immunol ; 17(4): 201-8, 1995 May.
Article En | MEDLINE | ID: mdl-7582256

We compared the efficacy of gene therapy mediated by interleukin-2 (IL-2) gene-modified tumor cells to gene therapy mediated by IL-2 transduced fibroblasts in the CT-26 model of murine colorectal carcinoma. We transduced CT-26 tumor cells and BALB/c 3T3 fibroblasts with three different retroviral vectors using three different promoters for the human IL-2 gene: DC/TKIL-2 (thymidine kinase promoter), LXSN-iIL2 (long terminal repeat promoter), and LNCX-iIL2 (cytomegalovirus promoter). These transductions resulted in CT-26 and 3T3 subclones that secreted different amounts of IL-2. Immunization of animals with either CT-26/IL-2 cells or with fibroblast/IL-2 cells mixed with CT-26 induced similar levels of immunity that protected 62-82% of animals against a subsequent tumor challenge with parental CT-26. However, mice developed tumors at the site of inoculation in 46% of the animals immunized with CT-26/IL-2 cells. In a separate experiment, CT-26/IL-2 cells were exposed to 6,000 cGy of gamma irradiation to prevent tumor growth at the site of inoculation. Although the CT-26/IL-2 cells continued to secrete IL-2 after irradiation, they were no longer effective at inducing antitumor immunity. In contrast, both irradiated and nonirradiated fibroblast/IL-2 cells, mixed with irradiated CT-26, were equally effective at inducing antitumor immunity. These data suggest that in the CT-26 model, fibroblast-mediated IL-2 gene therapy has advantages for the induction of antitumor immunity and abrogation of tumorigenic potential at the site of inoculation compared with tumor cell-mediated IL-2 gene therapy.


3T3 Cells/drug effects , Colorectal Neoplasms/therapy , Genetic Therapy , Interleukin-2/genetics , Transfection , 3T3 Cells/radiation effects , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Disease Models, Animal , Humans , Interleukin-2/pharmacology , Interleukin-2/radiation effects , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured
17.
In Vitro Cell Dev Biol Anim ; 31(3): 207-14, 1995 Mar.
Article En | MEDLINE | ID: mdl-7757303

We have established and characterized a new glioblastoma cell line, termed GT9, from a biopsy sample of a female adult patient with glioblastoma multiforme. The line has now undergone over 60 passages and has been successfully cultured after cryopreservation. Immunofluorescence analyses with a panel of monoclonal antibodies were positive for glial fibrillary acidic protein and vimentin, and negative for neurofilament, galactocerebroside, and fibronectin, a pattern typical of glial cells. Based on a tetraploid, the composite karyotype of GT9 cells included the loss of chromosome 10, gain of chromosome 7, and the presence of double minute chromosomes, three of the most common karyotypic abnormalities in glioblastoma. Sequence analysis of p53 cDNA revealed a homozygous double mutation at codon 249 (commonly mutated in aflatoxin-associated hepatocellular carcinoma) and codon 250. Moreover, there was a complete absence of wild-type p53. However, unlike the majority of human glioblastomas previously described, the expression of platelet-derived growth factor-B (PDGF-B), a potent mitogenic autocrine factor, was low in GT9 cells. The expression and phosphorylation of c-Jun and Jun-B, downstream mediators of the PDGF pathway, were also low. Thus, deregulation of the PDGF pathway does not appear to be involved in the pathogenesis of the GT9 glioblastoma. Conversely, Jun-D, a negative regulator of cell growth, was also low. In addition, Phosphorylated Egr-1, a recently reported suppressor of PDGF-B/v-sis-transformed cells, was also low, suggesting that the lack of activation of the PDGF pathway was not due to these suppressive mechanisms.(ABSTRACT TRUNCATED AT 250 WORDS)


Genes, p53 , Glioblastoma/genetics , Immediate-Early Proteins , Mutation , Platelet-Derived Growth Factor/metabolism , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Antibodies, Monoclonal , Chromosome Aberrations , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 7 , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Female , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , Glioblastoma/pathology , Humans , Middle Aged , Molecular Sequence Data , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Vimentin/analysis
18.
Gene Ther ; 2(2): 164-7, 1995 Mar.
Article En | MEDLINE | ID: mdl-7719933

A patient with glioblastoma multiforme (GBM) who had failed conventional therapy was treated with IL-2 gene therapy. The patient received 10 subcutaneous immunizations with autologous tumor cells and fibroblasts genetically modified to secrete IL-2 by retroviral gene transfer. An antitumor immune response mediated in part by CD8+ cytotoxic T cells was demonstrated with the patient's peripheral blood mononuclear cells. A magnetic resonance imaging (MRI) scan performed 4 weeks after the highest treatment dose revealed marked tumor necrosis. These results support the evaluation of this form of IL-2 gene therapy in additional patients with glioblastoma.


Brain Neoplasms/therapy , Fibroblasts/metabolism , Fibroblasts/transplantation , Genetic Therapy , Glioblastoma/therapy , Interleukin-2/therapeutic use , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Recombinant Fusion Proteins/therapeutic use , Salvage Therapy , Temporal Lobe , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cells, Cultured/transplantation , Combined Modality Therapy , Fatal Outcome , Female , Genetic Vectors , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Injections, Subcutaneous , Interleukin-2/genetics , Interleukin-2/metabolism , Isotretinoin/therapeutic use , Lomustine/administration & dosage , Magnetic Resonance Imaging , Middle Aged , Necrosis , Neoplasm Recurrence, Local , Procarbazine/administration & dosage , Radioimmunotherapy , Radiosurgery , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , T-Lymphocytes, Cytotoxic/immunology , Tamoxifen/therapeutic use , Transfection , Tumor Cells, Cultured/transplantation , Vincristine/administration & dosage
20.
J Biol Chem ; 267(6): 4023-9, 1992 Feb 25.
Article En | MEDLINE | ID: mdl-1371282

Biliverdin reductase is a unique dual cofactor- and pH-dependent enzyme that converts biliverdin to bilirubin and displays extensive inter-organ pI and molecular weight microheterogeneity. Presently we have explored the molecular basis for these properties. The amino acid composition and the sequences of NH2 termini plus five tryptic fragments of purified rat liver and kidney enzymes were obtained. A 62-nucleotide DNA probe was designed and in combination with antibody was used to screen a rat kidney cDNA library. A cDNA sequence of 1108 base pairs (bp) containing an 885-bp open reading frame was generated. The cloned cDNA probe detected a single mRNA of approximately 1500 bp in liver and kidney. The open reading frame encodes a 295 amino acid protein. Methionine and aspartic acid residues at positions 1 and 2 of the deduced protein are removed during processing. The deduced amino acid composition of the mature protein closely matched that of the purified rat liver and kidney enzymes. All liver peptides were found in the deduced amino acid sequence of kidney enzyme and the NH2 termini of both enzymes were identical. The expressed protein co-migrated with purified reductase and was recognized by antiserum to the enzyme. The expressed reductase displayed two distinct pH optima using a different cofactor at each pH: NADH at the lower pH 6.7-6.9 range and NADPH at pH 8.5-8.7. The findings suggest that the liver and kidney enzymes are the products of the same transcript(s) and that their microheterogeneity may reflect tissue-specific post-translational modifications.


Kidney/enzymology , Liver/enzymology , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/genetics , Amino Acid Sequence , Amino Acids/analysis , Animals , Base Sequence , Blotting, Northern , Chromatography, High Pressure Liquid , DNA , Electrophoresis, Polyacrylamide Gel , Gene Expression , Hydrogen-Ion Concentration , Male , Molecular Sequence Data , NAD/metabolism , NADP/metabolism , Open Reading Frames , Oxidoreductases/metabolism , Poly A/genetics , RNA/genetics , RNA, Messenger , Rats , Rats, Inbred Strains , Restriction Mapping , Transcription, Genetic
...