Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Mol Cell Biol ; 12(9): 3689-98, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1380646

RESUMO

We have studied several aspects of DNA damage formation and repair in human ovarian cancer cell lines which have become resistant to cisplatin through continued exposure to the anticancer drug. The resistant cell lines A2780/cp70 and 2008/c13*5.25 were compared with their respective parental cell lines, A2780 and 2008. Cells in culture were treated with cisplatin, and the two main DNA lesions formed, intrastrand adducts and interstrand cross-links, were quantitated before and after repair incubation. This quantitation was done for total genomic lesions and at the level of individual genes. In the overall genome, the initial frequency of both cisplatin lesions assayed was higher in the parental than in the derivative resistant cell lines. Nonetheless, the total genomic repair of each of these lesions was not increased in the resistant cells. These differences in initial lesion frequency between parental and resistant cell lines were not observed at the gene level. Resistant and parental cells had similar initial frequencies of intrastrand adducts and interstrand cross-links in the dihydrofolate reductase (DHFR) gene and in several other genes after cisplatin treatment of the cells. There was no increase in the repair efficiency of intrastrand adducts in the DHFR gene in resistant cell lines compared with the parental partners. However, a marked and consistent repair difference between parental and resistant cells was observed for the gene-specific repair of cisplatin interstrand cross-links. DNA interstrand cross-links were removed from three genes, the DHFR, multidrug resistance (MDR1), and delta-globin genes, much more efficiently in the resistant cell lines than in the parental cell lines. Our findings suggest that acquired cellular resistance to cisplatin may be associated with increased gene-specific DNA repair efficiency of a specific lesion, the interstrand cross-link.


Assuntos
Cisplatino/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Reparo do DNA , Southern Blotting , Dano ao DNA , Resistência a Medicamentos/genética , Feminino , Genoma Humano , Humanos , Neoplasias Ovarianas , Tetra-Hidrofolato Desidrogenase/genética , Células Tumorais Cultivadas , gama-Globulinas/genética
2.
Nat Biotechnol ; 14(5): 610-4, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-9630952

RESUMO

Over two-thirds of the current gene therapy protocols use retroviral gene transfer systems. We have developed an efficient retroviral-based method that allows rapid identification of gene transfer in living mammalian cells. Cells were generated containing a gene for an improved (humanized, red-shifted) version of the Aequorea victoria green fluorescent protein (hRGFP) from a retroviral vector. The hRGFP gene was used to produce an amphotropic vector producer cell line that demonstrated vibrant green fluorescence after excitation with blue light. A375 melanoma cells transduced with the retroviral vector demonstrated stable green fluorescence. Both PA317 murine fibroblasts and A375 human cell lines containing the vector were easily detected by FACS analysis. These vectors represent a substantial improvement over currently available gene transfer marking systems. Bright, long-term expression of the hRGFP gene in living eukaryotic cells will advance the study of gene transfer, gene expression, and gene product function in vitro and in vivo particularly for human gene therapy applications.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Animais , Sequência de Bases , Biotecnologia , Separação Celular , Primers do DNA/genética , Citometria de Fluxo , Fluorescência , Terapia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Camundongos , Retroviridae/genética , Células Tumorais Cultivadas
3.
Cancer Res ; 59(3): 661-8, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9973215

RESUMO

We demonstrate a novel method of concentrating radiation for tumor imaging or killing. The rat sodium/iodide symporter gene (rNIS) was cloned into a retroviral vector for transfer into cancer cells to mimic the iodide uptake of thyroid follicular cells. In vitro iodide transport shows that the symporter functions similarly in rNIS-transduced tumor cells and rat thyroid follicular cells. rNIS-transduced and control nontransduced (NV) human A375 melanoma xenografts established in vivo in athymic nude mice were imaged using a gamma camera after i.p. injections of 123I. The rNIS-transduced human A375 melanoma tumors are visually distinguishable from and accumulate significantly more radionuclides than NV tumors. In vitro clonogenic assays confirm efficacy and clearly show that rNIS-transduced A375 human melanoma, BNL.1 ME murine transformed liver, CT26 murine colon carcinoma, and IGROV human ovarian carcinoma can be selectively killed by the induced accumulation of 131I. Thus, NIS-based gene therapy may have both diagnostic and therapeutic applications for cancer.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Terapia Genética/métodos , Radioisótopos do Iodo/farmacocinética , Radioisótopos do Iodo/uso terapêutico , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Neoplasias/radioterapia , Simportadores , Tecnécio/farmacocinética , Tecnécio/uso terapêutico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/radioterapia , Animais , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/radioterapia , Feminino , Humanos , Iodetos/farmacocinética , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/radioterapia , Percloratos/farmacologia , Transplante Heterólogo , Células Tumorais Cultivadas
4.
Cancer Res ; 56(3): 569-73, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8564973

RESUMO

Carboxyamido-triazole (CAI) is a synthetic inhibitor of non-excitable calcium channels that reversibly inhibits angiogenesis, tumor cell proliferation, and metastatic potential. Inhibition of calcium influx and calcium-dependent events is a potential common mechanism underlying these effects of CAI. The cytostatic and antiangiogenic properties of CAI led to its development for clinical investigation. In a Phase I clinical trial open to patients with refractory solid tumors, 49 patients received p.o. administered CAI daily or every other day. Two oral formulations, PEG-400 CAI solution and a gelatin capsule containing CAI in PEG-400, were tested. All administered dosages of CAI yielded plasma concentration at or above the range demonstrated to be effective in inhibiting signaling and cancer progression in vitro and in preclinical models (1 microgram/ml, 2.3 microM). Toxicity of p.o. administered CAI most commonly consisted of dose-related grade 1-2 nausea, vomiting, and occasional anorexia. CAI administration at bedtime ameliorated gastrointestinal complaints in many patients; others required addition of simple antiemetic regimens, usually consisting of metoclopropamide or prochlorperazine. Gastrointestinal complaints were the cause for compliance-limiting toxicity at 175 mg/m2/day of the liquid formulation and 125 mg/m2/day of the gelatin capsule formation. Reversible and rare sensory axonal neuropathy (grade 3, 1 patient) and neutropenia (grade 4, 1 patient) were dose-limiting toxicities observed at the 330 mg/m2 every-other-day liquid CAI dose level. No evidence of cumulative end organ damage or central nervous system injury was observed. Disease stabilization and improvement in performance status was observed. Disease stabilization and improvement in performance status was observed in 49% of evaluable patients who had disease progression before CAI. Disease stabilization and associated improvement in performance status was seen in patients with renal cell carcinoma (7 months), pancreaticobiliary carcinomas (3, 5, and 5 months), melanoma (7 months), ovarian cancer (7 months), and non-small cell lung cancer (3 months). The recommended Phase II doses from this trial are 150 mg/m2/day in the liquid formation and 100 mg/m2/day in the gelatin capsule formation.


Assuntos
Antineoplásicos/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Neoplasias/tratamento farmacológico , Triazóis/uso terapêutico , Administração Oral , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Bloqueadores dos Canais de Cálcio/efeitos adversos , Bloqueadores dos Canais de Cálcio/farmacocinética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Triazóis/efeitos adversos , Triazóis/farmacocinética
5.
Oncogene ; 10(8): 1639-45, 1995 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-7731719

RESUMO

We have studied the repair of u.v.-induced cyclobutane pyrimidine dimers (CPDs) in amplified c-myc oncogene loci in human colon cancer cells to better understand the relationship between chromatin structure, transcription and DNA repair. To assess the variation in DNA repair in the same gene whether located in a chromosomal site or in a extra-chromosomal site, we have quantitated the efficiency of excision repair after u.v. exposure in the endogenous and episomal c-myc genes isolated from COLO320HSR and DM cells. In the HSR cells, c-myc is localized in a homogeneously staining region (HSR), and in the DM cells, the gene is localized in double minute chromosomes (DM). Our results indicate that the repair is less efficient in c-myc amplicons organized as double minute chromosomes than in the endogenous c-myc amplicons. The episomal gene is not repaired with the same efficiency as when it is intrachromosomal. This may reflect differences in chromatin structure. An advantage of this biological system is that the cells possess two different alleles of the c-myc gene, one that is active and another which is inactive. We have studied the relationship between DNA repair and transcriptional activity in the c-myc locus by measuring the efficiency of excision repair after u.v. exposure in the normal and rearranged alleles of the c-myc gene. Surprisingly, the c-myc gene is repaired with similar efficiency in the highly transcribed allele as in the poorly expressed allele. However, u.v. damage is selectively removed from the transcribed strand of the active c-myc allele, but DNA repair is not strand specific in the non-expressed c-myc allele.


Assuntos
Reparo do DNA , Amplificação de Genes , Genes myc , Neoplasias/genética , Mapeamento Cromossômico , Humanos , Plasmídeos , Dímeros de Pirimidina/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Transcrição Gênica , Células Tumorais Cultivadas
6.
Clin Cancer Res ; 6(1): 185-92, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10656449

RESUMO

Adenoviral vector-mediated p53 expression induced apoptosis is a well established gene therapy approach that has been evaluated extensively in epithelial tumors but only recently in lymphoid malignancies mainly due to the known resistance of the lymphoid lineage to adenovirus infection. Recently, it was shown that this resistance is not absolute and that cell lines derived from anaplastic large cell lymphoma (ALCL) and some other lymphoid malignancies are efficiently transduced by adenoviral vectors. Normal circulating T lymphocytes do not express coxsackie-adenovirus receptor (CAR) and alpha(nu)beta integrins and are relatively resistant to infection by adenovirus. These molecules serve as receptors for adenovirus entry into the cells. ALCL-derived SUDHL-1 cells were evaluated for transduction efficiency and expression of p53 after infection with an adenoviral vector containing wild-type p53 (AdWTp53). Cells derived from ALCL and circulating mononucleated cells (MNCs) were also evaluated for expression of CAR and alpha(nu)beta integrins. AdWTp53-mediated expression of p53 resulted in p21/WAF1 induction, G1 arrest, and apoptosis in SUDHL-1 cells. The expression of CAR and alpha(nu)beta5 integrin was high in SUDHL-1 cells and comparable to levels observed with epithelial tumor cells, but it was absent in MNCs. The susceptibility to adenoviral vector transduction of the tumor-derived cells implies an important biological difference between them and circulating MNCs, possibly underlying the malignant transformation that ALCL cells undergo. Further studies will be required to evaluate this initial observation in more cell lines and tissue derived from ALCL.


Assuntos
Adenoviridae , Genes p53 , Integrinas/fisiologia , Linfoma Difuso de Grandes Células B/patologia , Receptores Virais/fisiologia , Receptores de Vitronectina , Adenoviridae/fisiologia , Ciclo Celular , Sobrevivência Celular , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/fisiologia , Citometria de Fluxo , Vetores Genéticos , Humanos , Marcação In Situ das Extremidades Cortadas , Integrinas/genética , Linfoma Difuso de Grandes Células B/imunologia , Receptores Virais/genética , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
7.
Hum Gene Ther ; 10(13): 2209-19, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10498252

RESUMO

Genetic diseases are often caused by nonsense mutations. The resulting defect in protein translation can be restored by expressing suppressor tRNA in the mutant cells. Our goal was to demonstrate both protein restoration and phenotypic correction using these small transgenes. Functional activity of an arginine opal suppressor tRNA in cells expressing a nonsense mutated GFP gene was demonstrated by restored fluorescence. This suppressor tRNA was expressed in xeroderma pigmentosum group A cells, containing a homozygous nonsense mutation at Arg-207 in the XPA complementing gene. The transfected XPA cell population showed a twofold increase in cell survival after UV irradiation as determined by colony-forming assays compared with cell populations without the suppressor tRNA gene. The UV doses required for 37% survival of XP cells and XP cells expressing the suppressor tRNA were 0.6 and 1.2 J/m2. A similar twofold increase in the reactivation of UV-irradiated plasmid DNA was observed in XP cells expressing the suppressor tRNA. However, there was no detectable increase in XPA protein levels. Several potential limitations of this approach exist, including the availability of mutant RNA transcripts, the efficiency of suppression by the suppressor tRNA, and the abundance and availability and continued expression of the suppressor tRNA. The unique feature of this study is the relatively small size (88 bp) of the suppressor tRNA. Small-sized suppressor tRNAs can be synthetically constructed and subcloned into different viral vectors for delivery into the target cells. This approach may be useful for other genetic diseases caused by nonsense mutations.


Assuntos
Proteínas de Ligação a DNA/genética , Técnicas de Transferência de Genes , RNA de Transferência de Arginina/biossíntese , Xeroderma Pigmentoso/patologia , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Herpesvirus Humano 1/genética , Humanos , Mutação Puntual , RNA de Transferência de Arginina/genética , Supressão Genética , Transfecção , Raios Ultravioleta , Xeroderma Pigmentoso/genética , Xeroderma Pigmentoso/metabolismo , Proteína de Xeroderma Pigmentoso Grupo A
8.
Hum Gene Ther ; 5(3): 343-79, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8018748

RESUMO

Murine retroviral vectors can infect a wide variety of proliferating mammalian cell types (e.g. lymphocytes). Non-proliferating tissues (e.g. neurons) are not transduced by murine retroviral vectors. These findings suggest that this type of vector may be useful for the selective introduction of genes into growing tumors in the brain, since the tumor is essentially the only tissue that will integrate and express the vector genes.


Assuntos
Neoplasias Encefálicas/terapia , Ganciclovir/uso terapêutico , Terapia Genética , Proteínas Recombinantes de Fusão/genética , Simplexvirus/enzimologia , Timidina Quinase/genética , Proteínas Virais/genética , Adulto , Animais , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/líquido cefalorraquidiano , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/cirurgia , Sobrevivência Celular , Células Cultivadas/microbiologia , Células Cultivadas/transplante , Protocolos Clínicos , Terapia Combinada , Feminino , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Consentimento Livre e Esclarecido , Injeções Espinhais/instrumentação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Leucemia Murina de Moloney/genética , Ratos , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Terapia de Salvação , Sarcoma Experimental/terapia , Simplexvirus/genética , Timidina Quinase/antagonistas & inibidores , Células Tumorais Cultivadas , Proteínas Virais/antagonistas & inibidores
9.
Hum Gene Ther ; 9(1): 115-34, 1998 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9458247

RESUMO

This study will evaluate the safety and efficacy of allogenic donor lymphocyte infusions in patients who have relapsed hematologic malignancies after allogeneic bone marrow transplantation (BMT). Donor lymphocyte transfusions have resulted in the cure of some patients with relapsed leukemia or lymphoproliferative disorder after allogeneic BMT, but has been complicated by the development of graft versus host disease (GvHD). We hypothesize that a retroviral vector containing the Herpes simplex thymidine kinase (HStk) gene will allow for retention of the anti-leukemia response of transfused donor lymphocytes while allowing for the adverse effects of GVHD to be mitigated. Patients with relapsed hematologic malignancies after allogeneic BMT will be infused with ex vivo gene modified donor lymphocytes. The Herpes Simplex thymidine kinase (HStk) gene will be transduced into the cells ex vivo using LTKOSN. 1 vector supernate. Insertion of the HStk gene into lymphocytes confers a sensitivity to the anti-herpes drug ganciclovir (GCV). This selective destruction of donor lymphocytes in situ will be used to abrogate the effect of graft versus host disease, if it develops.


Assuntos
Protocolos Clínicos , Imunoterapia Adotiva/métodos , Leucemia Linfoide/terapia , Timidina Quinase/genética , Estudos de Avaliação como Assunto , Vetores Genéticos , Humanos , Imunoterapia Adotiva/efeitos adversos , Linfócitos/citologia , Linfócitos/metabolismo , Seleção de Pacientes , Indução de Remissão/métodos , Simplexvirus/enzimologia , Timidina Quinase/metabolismo
10.
Semin Hematol ; 36(1 Suppl 3): 38-42, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9989489

RESUMO

A variety of approaches to antitumor therapy are currently being explored that use both antigen-encoding DNA and noncoding nucleotides as a component of gene vaccination. Among the specific strategies reviewed are a construct that fuses a single-chain variable fragment (scFv) that incorporates both the variable-region genes necessary to encode the idiotypic determinants with fragment C (FrC) of tetanus toxin; a novel vector system using herpes simplex virus 1 (HSV-1) for in vivo gene delivery; the possibility of eliciting hyperacute xenograft response to treat human cancer; and the use of gene gun-mediated granulocyte-macrophage colony-stimulating factor (GM-CSF) cDNA-based tumor cell vaccines. The protection provided by DNA vaccination against viral diseases such as influenza suggested a role for such vaccines against cancer. However, unlike vaccines against infectious diseases, cancer vaccines are therapeutic, rather than prophylactic. With multiple myeloma, for example, it is possible that the optimal timing of administration of such a vaccine is during a remission that has been induced by traditional therapies, to eliminate residual disease. DNA cancer vaccines are designed to activate immune responses to tumor antigens to which the immune system has already been exposed. To do so, the vaccines must first overcome immune tolerance that may have already developed to the tumor. There is increasing evidence that tumor antigens, unlike viral or bacterial antigens, do not consistently activate an immune response. One major factor in determining whether a reaction occurs appears to be whether antigen presentation is accompanied by danger signals. With viral or bacterial infection, the accompanying tissue destruction and inflammation produce costimulatory signals that promote T-cell activation. However, inflammatory and tissue-destructive processes are absent during initial tumor transformation. The typical outcome may be immunologic tolerance.


Assuntos
Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Vacinas de DNA/imunologia , Terapia Genética , Humanos , Mieloma Múltiplo/genética
11.
Am J Med ; 101(2): 217-25, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8757363

RESUMO

Borderline tumor of the ovary (BOT) is an epithelial tumor with a low rate of growth and a low potential to invade or metastasize. This tumor often is associated with a significantly better prognosis than epithelial ovarian cancer. Most tumors are either serous or mucinous in histology and present as early stage lesions. However, stage III lesions with peritoneal implants are not uncommon. Patients with early stage lesions have an excellent prognosis. Patients with higher stage lesions have a worse prognosis. Long-term follow-up of patients with BOT is required since the tumor can recur up to 20 years after the initial diagnosis. Recently, investigators have begun to identify subsets of patients with a worse prognosis, such as patients with aneuploid tumors. Treatment for early stage lesions is surgical and conservative surgery can be accomplished successfully in younger patients who desire to maintain fertility. Treatment for later stage lesions has been approached in a variety of ways. All approaches initially begin with maximal cytoreductive surgery. Studies suggest that early stage disease should be managed with surgery alone. Conflicting results on the usefulness of adjuvant therapy for patients with later stage disease have been obtained. At this time, the usefulness of adjuvant therapy for advanced disease remains undetermined. Further understanding of the basis of the disease and analysis of specific higher risk subsets might identify patients in whom adjuvant therapy could be tested in the setting of controlled clinical trials.


Assuntos
Neoplasias Ovarianas , Biomarcadores Tumorais/sangue , Feminino , Humanos , Inoculação de Neoplasia , Estadiamento de Neoplasias , Neoplasias Epiteliais e Glandulares , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Neoplasias Peritoneais/secundário , Prognóstico , Análise de Sobrevida
12.
Biotechniques ; 22(1): 162-7, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8994663

RESUMO

We have developed murine retroviral vectors (RVs) containing an optimized green fluorescent protein (GFP) gene to study retroviral gene transfer and expression in living cells. We used the codon "humanized", "red-shifted" GFP gene, hGFP-S65T, a gain of function variant of the wild-type GFP from the jellyfish Aequorea victoria. We cloned the hGFP-S65T gene into the RV plasmid pLNCX (pLNChG65T). A stable amphotropic RV-producer cell line (VPC), designated LNChG65T VPC, was generated that exhibited bright fluorescence in greater than 95% of the cells. Human A375 melanoma cells and IGROV ovarian carcinoma cells transduced from LNCh-G65T VPC demonstrated high levels of fluorescence. The expression of a single integrated hGFP-S65T gene in eukaryotic cells provides a powerful tool to study gene transfer, expression and functional studies in vitro and in vivo.


Assuntos
Expressão Gênica/genética , Técnicas de Transferência de Genes , Vetores Genéticos/isolamento & purificação , Proteínas Luminescentes/genética , Retroviridae/genética , Animais , Clonagem Molecular , Citometria de Fluxo , Marcadores Genéticos , Humanos , Proteínas Luminescentes/análise , Camundongos , Cifozoários/genética , Cifozoários/metabolismo , Transfecção , Células Tumorais Cultivadas
13.
Bone Marrow Transplant ; 32 Suppl 1: S33-5, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12931238

RESUMO

Transduction of stem cells with a marking gene holds promise to determine if tissue repair or regeneration is derived from the adult hematopoietic stem cell and if relapse of an autoimmune disease should occur whether relapse arises from the stem cell compartment or from lymphocytes surviving the conditioning regimen. New safety concerns about gene-modified stem cell would entail new safety testing such as documentation of the insertional site prior to release.


Assuntos
Genes Reporter/genética , Células-Tronco Hematopoéticas/metabolismo , Canamicina Quinase/genética , Transdução Genética/métodos , Antígenos CD/análise , Antígenos CD34/análise , Técnicas de Cultura de Células , Diferenciação Celular , Vetores Genéticos , Humanos , Retroviridae/genética , Fatores de Tempo
14.
Bone Marrow Transplant ; 24(10): 1043-51, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10578153

RESUMO

Patients with recurrent leukemia after an allogeneic hematopoietic stem cell transplant may be treated with donor lymphocyte infusions (DLI). The transfusion of lymphocytes from the original hematopoietic stem cell donor induces remission in approximately one third of relapsed AML cases and 80% of relapsed CML. DLI may be complicated by delayed and sometimes lethal graft-versus-host disease (GVHD). In an attempt to avoid this complication, several centers have initiated DLI trials in which the infused lymphocytes carry a suicide gene, herpes simplex thymidine kinase (HStk), which confers sensitivity to ganciclovir (GCV). In the event of severe GVHD, administration of GCV should terminate or ameliorate GVHD.


Assuntos
Ganciclovir/uso terapêutico , Leucemia Mieloide Aguda/terapia , Transfusão de Linfócitos , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Humanos , Timidina Quinase/metabolismo , Transfecção
15.
Cancer Treat Res ; 101: 369-75, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10800657

RESUMO

The overall goal of adoptive immunotherapy with genetically modified lymphocytes is to decrease the morbidity and mortality associated with allogeneic bone marrow transplantation. The initial data reviewed here suggest that the behavior of the allogeneic HStk transgenic cells can be modified after administration to patients. Further study is needed to identify the response rates and risks associated with this procedure. In particular, larger studies will be needed with appropriate randomization to determine if the response rate to genetically modified cells is equivalent to the response rates with unmodified cells. Wider application of these techniques in the initial setting of allogeneic transplantation will undoubtedly occur and such trials have been initiated at several institutions. Careful attention to vector, suicide gene, selectable marker, efficiency of transduction, and cell dose will be necessary when comparing different trials since these variables will probably affect transgenic cell survival and response rates. [figure: see text]


Assuntos
Transfusão de Sangue Autóloga , Ganciclovir/uso terapêutico , Terapia Genética , Leucemia/terapia , Transfusão de Linfócitos , Simplexvirus/genética , Timidina Quinase/genética , Transplante de Medula Óssea/efeitos adversos , Efeito Enxerto vs Leucemia , Humanos , Imunoterapia Adotiva , Transplante Homólogo
16.
Anticancer Res ; 18(4A): 2301-8, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9703870

RESUMO

Xenograft hyperacute rejection in humans occurs as a secondary response to a cellular glycosylation incompatibility with most non-human mammalian species. A key component of hyperacute rejection, alpha(1,3)galactosyl (agal) epitopes present on the surface of most non-human mammal cells, is bound by host anti-agal IgG antibodies leading to the activation of complement and, cellular lysis (1). The enzyme causing specific glycosylation patterns, alpha(1,3)galactosyltransferase [alpha(1,3)GT], directs the addition of agal to N-acetyl glucosamine residues in the trans Golgi apparatus in most mammalian species including Mus musculus, but not old world primates, apes or humans. In this report, we cloned both a truncated and full length murine alpha(1,3)GT gene into a retroviral vector backbone in order to transfer alpha(1,3)galactosyl epitopes into human A375 melanoma cells. Expression of agal epitopes on A375 cells after alpha(1,3)GT gene transfer was demonstrated using FITC-labeled ligand and FACS analysis. These cells were exposed to human serum for 30 minutes and > 90% of the agal expressing cells were killed by this treatment. These pretreated cells failed to establish tumors after implantation into athymic nude mice. This is the first report of retroviral vector transfer of the alpha(1,3)GT gene into human tumor cells in an attempt to elicit hyperacute rejection as a novel anti-cancer gene therapy strategy.


Assuntos
Neoplasias da Mama/terapia , Galactosiltransferases/genética , Melanoma/terapia , Neoplasias Ovarianas/terapia , Células 3T3 , Animais , Sangue , Neoplasias da Mama/patologia , Linhagem Celular , Sobrevivência Celular , Chlorocebus aethiops , Clonagem Molecular , Feminino , Citometria de Fluxo , Galactosiltransferases/biossíntese , Humanos , Melanoma/patologia , Camundongos , Camundongos Nus , Neoplasias Ovarianas/patologia , Proteínas Recombinantes/biossíntese , Transplante Heterólogo , Células Tumorais Cultivadas
17.
Mutat Res ; 336(2): 115-21, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7885382

RESUMO

We have measured gene specific DNA repair in a normal human fibroblast cell line, and in fibroblast lines from two patients with familial Alzheimer disease (AD). Cells were treated with either ultraviolet radiation (UV) or the chemotherapeutic alkylating agent, nitrogen mustard (HN2). DNA damage formation and repair were studied in the active dihydrofolate reductase (DHFR) gene for the main lesions introduced by each of these two types of DNA damaging agents. The gene specific repair of UV induced cyclobutane pyrimidine dimers in the human DHFR gene was 86% complete in the AD cells after 24 h of repair incubation. This repair efficiency was similar to what we and others have found in normal human fibroblasts. After treatment of the AD cells with HN2, we found the frequency of HN2 induced lesions in the DHFR gene to be similar to the frequency in the transcriptionally inactive delta-globin gene. The gene specific repair of HN2 induced lesions in the DHFR gene was completed within 8-24 h in the normal fibroblast line and in the familial AD line, and the repair kinetics were similar for both cell lines. These results indicate that familial AD fibroblasts have normal gene specific repair of both UV induced and HN2 induced DNA damage in active genes.


Assuntos
Doença de Alzheimer/metabolismo , Reparo do DNA/fisiologia , Mecloretamina/efeitos adversos , Tetra-Hidrofolato Desidrogenase/genética , Raios Ultravioleta/efeitos adversos , Células Cultivadas , Dano ao DNA , Fibroblastos , Globinas/genética , Humanos , Dímeros de Pirimidina
18.
Am Surg ; 63(7): 617-20, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9202536

RESUMO

The herpes simplex thymidine kinase (HStk) gene induces regression of epithelial tumors after ganciclovir (GCV) administration. This observation has been attributed to both gene transfer and metabolic cooperation between cells (the bystander effect). This study evaluates the relative roles of the bystander effect and gene delivery of the HStk gene by the LTKOSN.2 vector. MC38 colon adenocarcinoma cells, syngeneic for C57/B16 mice, were used. Whereas in vitro proliferation assays demonstrated a bystander effect, significantly greater inhibition of proliferation occurred with HStk gene transfer. In mixtures containing 75 per cent MC38 cells with no vector (MC38 NV) and 25 per cent MC38 pretransduced with LTKOSN.2 (MC38 TK), proliferation was inhibited by 62 +/- 5 per cent. In mixtures containing 75 per cent MC38 NV with 25 per cent HStk vector-producing cells (LTKOSN.2 VPC), proliferation was inhibited by 97 +/- 1 per cent. In vivo subcutaneous mixture experiments utilized MC38 NV cells inoculated at a 1:1 ratio with various treatment cell groups followed by administration of GCV. Tumor volumes (mean +/- standard error) at 30 days were: 264 +/- 66 mm3 for MC38 TK, 0 for LTKOSN.2 VPC, 1009 +/- 335 mm3 for lacZ VPC (beta-galactosidase VPC), and 1012 +/- 212 mm3 for NIH3T3 (nontransduced cells). These data suggest that in vivo, the bystander effect alone causes tumor inhibition, but gene transfer is necessary for complete tumor elimination in immunocompetent mice.


Assuntos
Adenocarcinoma/terapia , Neoplasias do Colo/terapia , Técnicas de Transferência de Genes , Genes Virais , Terapia Genética , Herpes Simples/enzimologia , Timidina Quinase/genética , Adenocarcinoma/patologia , Animais , Divisão Celular , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Herpes Simples/genética , Camundongos , Camundongos Endogâmicos , Células Tumorais Cultivadas
19.
In Vivo ; 12(1): 89-93, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9575431

RESUMO

Neoplasms are derived from normal tissues of the body by cellular transformation. Tumors often represent a less differentiated or an undifferentiated version of the histology of the neoplasms original tissue. Primary tumor(s) may spread by direct extension or by metastasis. For the purpose of this review, second primary tumors will be defined to exclude metastatic lesions or recurrences from an original primary tumor. Second primary tumors arise in several different clinical situations that basically are the result of either inherited or acquired genetic mutations. Second primary tumors may develop soon or very late after treatment of the first primary tumor and may reflect an underlying genetic or immunologic defect in the patient, treatment related genetic damage, or environmental exposure to carcinogens. With the greater success of modern chemotherapy and radiotherapy in achieving long-term remissions in many patients, second primary tumors are a rapidly developing disease-category. This paper will review the clinical circumstances associated with a significantly higher incidence of second primary tumors in patients with an initial primary tumor.


Assuntos
Segunda Neoplasia Primária/etiologia , Humanos
20.
In Vivo ; 14(5): 635-41, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11212841

RESUMO

Retroviral vector producer cells (VPC) can effectively transfer genes in vivo. To develop a safe method to target gene delivery into intraperitoneal tumors, we have examined the toxicity of intraperitoneal (i.p.) infusion of retroviral VPC in a xenogeneic canine model. Mongrel dogs were injected intraperitoneally (i.p.) with 2 x 10(9) murine LTKOSN.2 VPC. The animals did not demonstrate acute toxicity and tolerated the i.p. infusion of the cells without difficulty. Starting 7 days after i.p. injection, the dogs received intravenous injections of ganciclovir (GCV) twice daily (5 mg/kg) for 7 days. The treatment dogs underwent peritoneal washings on days 3, 7 and 14 after their initial infusion of cells to study the persistence of the VPC. GCV treatment did not cause significant toxicities. Dogs underwent serial blood tests to evaluate bone marrow, renal, liver and immunological function. Complete blood counts, electrolytes and renal function remained normal throughout the study. Although, transient mild elevations occurred of serum alkaline phosphate, the remaining hepatic enzymes remained normal. Histologic examination of tissues from animals sacrificed after the i.p. administration of the VPC revealed no tissue destruction of the normal peritoneal lining. The dogs mounted an antibody response to the murine VPC that was first observed 7 days post injection. PCR analysis of selected tissues after GCV administration did not reveal persistent vector sequences. These results demonstrated that the injection of xenogeneic VPC is not accompanied by significant adverse effects over a 1 month period following administration into the canine peritoneal cavity. These data support the potential clinical application of the VPC in Phase I clinical trials in humans.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Cavidade Peritoneal/citologia , Fosfatase Alcalina/sangue , Animais , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Cães , Ganciclovir/farmacologia , Testes Hematológicos , Herpesvirus Humano 1/enzimologia , Humanos , Testes de Função Renal , Testes de Função Hepática , Camundongos , Modelos Animais , Lavagem Peritoneal , Reação em Cadeia da Polimerase , Timidina Quinase/genética , Transfecção , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/transplante
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa