Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cell Prolif ; 41(6): 843-858, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19040565

ABSTRACT

OBJECTIVES: Islet-like clusters (ILCs), differentiated from human embryonic stem cells (hESCs), were characterized both before and after transplantation under the kidney capsule of streptozotocin-induced diabetic immuno-incompetent mice. MATERIALS AND METHODS: Multiple independent ILC preparations (n = 8) were characterized by immunohistochemistry, flow cytometry and cell insulin content, with six preparations transplanted into diabetic mice (n = 42), compared to controls, which were transplanted with either a human fibroblast cell line or undifferentiated hESCs (n = 28). RESULTS: Prior to transplantation, ILCs were immunoreactive for the islet hormones insulin, C-peptide and glucagon, and for the ductal epithelial marker cytokeratin-19. ILCs also had cellular insulin contents similar to or higher than human foetal islets. Expression of islet and pancreas-specific cell markers was maintained for 70 days post-transplantation. The mean survival of recipients was increased by transplanted ILCs as compared to transplanted human fibroblast cells (P < 0.0001), or undifferentiated hESCs (P < 0.042). Graft function was confirmed by secretion of human C-peptide in response to an oral bolus of glucose. CONCLUSIONS: hESC-derived ILC grafts continued to contain cells that were positive for islet endocrine hormones and were shown to be functional by their ability to secrete human C-peptide. Further enrichment and maturation of ILCs could lead to generation of a sufficient source of insulin-producing cells for transplantation into patients with type 1 diabetes.


Subject(s)
Embryonic Stem Cells/cytology , Endocrine Cells/cytology , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Animals , Cell Differentiation , Cell Line , DNA/metabolism , Embryonic Stem Cells/ultrastructure , Endocrine Cells/ultrastructure , Flow Cytometry , Humans , Insulin/metabolism , Islets of Langerhans/ultrastructure , Kaplan-Meier Estimate , Mice , Mice, Inbred NOD
2.
Gene Ther ; 8(7): 568-78, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11319624

ABSTRACT

In human cells, telomerase activity is regulated by transcriptional control of the telomerase reverse transcriptase gene (hTERT) whose product is the catalytic subunit of the enzyme. The hTERT promoter is active in virtually all types of tumors and immortal cells, but is silent in most adult somatic tissues. In this study, we placed the herpes simplex virus thymidine kinase gene under the control of the hTERT promoter with the aim of restricting its expression to tumor cells. In transfection experiments, the hTERT promoter driven thymidine kinase gene (hTERTp/TK) conferred ganciclovir sensitivity to all tumor and immortal cell lines tested, whereas normal somatic cells remained largely unaffected. Human hTERTp/TK-positive cancer cells implanted in nude mice developed into tumors that could be eradicated by ganciclovir treatment. The hTERTp/TK cassette was inserted into an adenovirus vector and its efficacy in reducing tumor growth was compared with that of an adenovirus carrying the thymidine kinase gene under the control of the cytomegalovirus immediate-early promoter (CMVp/TK). In a xenograft model using the human 143B osteosarcoma cell line, a single injection of either virus resulted in equivalent tumor regression and survival upon ganciclovir treatment. In animals injected intratumorally with the CMVp/TK adenovirus, expression of the thymidine kinase gene was detected in tumors, as well as in liver samples. Expression of the suicide gene in combination with ganciclovir resulted in severe liver histopathology and in an elevation of hepatic enzymes. In sharp contrast, when the hTERT promoter controlled the thymidine kinase gene, transgene expression was observed in tumors, but not in liver samples. Normal liver function in these animals was confirmed by serum levels of hepatic enzymes that were indistinguishable from those of control healthy mice. These results indicate that by restricting thymidine kinase expression to tumor cells, the hTERT promoter allows the tumoricidal effect of the suicidal gene to be exerted without detrimental consequences on healthy tissues and vital organs. The tight specificity of expression imparted by the hTERT promoter will assist the development of novel approaches to the treatment of a broad array of cancer types.


Subject(s)
Genetic Therapy/methods , Liver Diseases/prevention & control , Osteosarcoma/therapy , Promoter Regions, Genetic , RNA , Telomerase/genetics , Adenoviridae/genetics , Animals , DNA-Binding Proteins , Ganciclovir/therapeutic use , Gene Expression Regulation, Neoplastic , Genetic Therapy/adverse effects , Genetic Vectors/therapeutic use , Humans , Liver Diseases/etiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Messenger/genetics , Survival Rate , Thymidine Kinase/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
3.
Cancer J ; 7 Suppl 2: S83-93, 2001.
Article in English | MEDLINE | ID: mdl-11777269

ABSTRACT

Human embryonic stem (hES) cells can proliferate extensively in culture and can differentiate into representatives of all three embryonic germ layers in vitro and in vivo. The undifferentiated hES cells have now been cultured for more than 50 passages in vitro, yet maintain a normal karyotype. The hES cells express a series of specific surface antigens, as well as OCT-4 and human telomerase, proteins associated with a pluripotent and immortal phenotype. On differentiation, OCT-4 and human telomerase expression decreases with the emergence of a maturing population of cells. During hES cell differentiation, modulation of the expression of many genes has been evaluated using microarray analysis. To improve the ease, reproducibility, and scalability of hES culture, methods have been developed to propagate the cells in the absence of mouse embryonic cell feeders. hES cells maintained in culture using extracellular matrix factors together with mouse embryonic cell conditioned medium proliferate indefinitely while maintaining a normal karyotype, proliferation rate, and complement of undifferentiated cell markers. hES cells cultured without feeder layers retain their capacity to differentiate into cells of all three germ layers in vitro and in teratomas. The hES cells can also be genetically modified transiently or stably using both plasmid and viral gene transfer agents. These analyses and technological developments will aid in the realization of the full potential of hES cells for both research and therapeutic applications.


Subject(s)
Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Stem Cell Transplantation , Transcription Factors , Animals , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Humans , Mice , Octamer Transcription Factor-3 , Oligonucleotide Array Sequence Analysis/methods , Phenotype , Polymerase Chain Reaction/methods , Stem Cells/cytology
4.
Nat Med ; 6(9): 1011-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973321

ABSTRACT

The polypeptide component of telomerase (TERT) is an attractive candidate for a broadly expressed tumor rejection antigen because telomerase is silent in normal tissues but is reactivated in more than 85% of cancers. Here we show that immunization against TERT induces immunity against tumors of unrelated origin. Immunization of mice with TERT RNA-transfected dendritic cells (DC) stimulated cytotoxic T lymphocytes (CTL), which lysed melanoma and thymoma tumor cells and inhibited the growth of three unrelated tumors in mice of distinct genetic backgrounds. TERT RNA-transfected human DC stimulated TERT-specific CTL in vitro that lysed human tumor cells, including Epstein Barr virus (EBV)-transformed B cells as well as autologous tumor targets from patients with renal and prostate cancer. Tumor RNA-transfected DC were used as surrogate targets in the CTL assays, obviating the difficulties in obtaining tumor cells from cancer patients. In one instance, where a tumor cell line was successfully established in culture from a patient with renal cancer, the patient's tumor cells were efficiently lysed by the CTL. Immunization with tumor RNA was generally more effective than immunization with TERT RNA, suggesting that an optimal immunization protocol may have to include TERT as well as additional tumor antigens.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Neoplasms, Experimental/prevention & control , T-Lymphocytes, Cytotoxic , Telomerase/therapeutic use , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , B-Lymphocytes , Cell Transformation, Viral , H-2 Antigens , Herpesvirus 4, Human , Humans , Immunotherapy , Kidney Neoplasms/immunology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Prostatic Neoplasms/immunology , RNA/genetics , RNA/immunology , RNA/therapeutic use , Telomerase/genetics , Telomerase/immunology , Vaccines, Synthetic/therapeutic use
5.
Cancer Gene Ther ; 7(1): 128-34, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10678365

ABSTRACT

New specificities may be engrafted onto lymphocytes by the transfer of genes for chimeric receptors that combine antigen recognition and signal-transducing elements. We have engineered and evaluated a new class of chimeric receptors that use the natural ligands of receptors found to be frequently overexpressed by cancer cells. The heregulin molecule, a ligand for Her3 and Her4 receptors when fused with the CD3 zeta-chain, was capable of redirecting T lymphocytes to recognize and respond to cancer cell lines that overexpress these receptors. Thus, CD8+ T lymphocytes were isolated from a healthy individual and transduced to express the chimeric heregulin-zeta receptor. These modified effector cells acquired the ability to specifically lyse a breast cancer cell line that overexpresses Her3 and Her4. A new class of chimeric receptors, such as heregulin-zeta, endowing anti-cancer effector cells with the potential to recognize and eliminate tumor targets, are likely to increase the effectiveness of adoptive immunotherapy for the treatment of cancer.


Subject(s)
Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Cytotoxicity, Immunologic , Flow Cytometry , Humans , Jurkat Cells , Ligands , Neuregulin-1/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-3/immunology , Recombinant Fusion Proteins , Signal Transduction
6.
J Immunother ; 23(1): 2-10, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10687132

ABSTRACT

MUC1 mucin peptides stimulated cytotoxic T lymphocytes (CTL) from humans with adenocarcinomas. Peripheral blood mononuclear cells, tumor-draining lymph node cells, or tumor-infiltrating lymphocytes were stimulated using mono-nuclear cells from humans with adenocarcinomas of breast or ovary, respectively, using (a) a native MUC1 mucin tandem repeat peptide of 20 amino acids (MUC1-mtr1) plus recombinant human interleukin-2 (IL-2), (b) the mutated (T3N) MUC1-mtr1 plus IL-2, or (c) immobilized anti-CD3 plus IL-2, or (d) IL-2 alone. The CTL stimulated by each of these four conditions were predominately CD4+. However, the CTL stimulated by either the native MUC1-mtr1 or (T3N) MUC1-mtr1 showed 5-10 times greater cytotoxicity of a breast cancer cell line that expresses MUC1 compared to CTL stimulated by either anti-CD3 + IL-2 or IL-2 alone. Each incubation condition generated CTL with different variable beta gene families of T-cell receptors, implying an oligoclonal expansion of a limited CTL repertoire for each. Thus, peptide-stimulated T cells showed expression of cytotoxic cells, which was not induced by nonspecific (anti-CD3 or IL-2) stimulation.


Subject(s)
Adenocarcinoma/immunology , Breast Neoplasms/immunology , Mucin-1/immunology , Ovarian Neoplasms/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenocarcinoma/blood , Amino Acid Sequence , Binding Sites , Breast Neoplasms/blood , Female , Glycosylation , Humans , K562 Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Molecular Sequence Data , Mucin-1/genetics , Mucin-1/pharmacology , Mutagenesis , Ovarian Neoplasms/blood , Peptide Fragments/genetics , Peptide Fragments/pharmacology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Cells, Cultured
7.
J Immunother ; 23(1): 168-76, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10687150

ABSTRACT

Immunization with tumor-specific-associated antigen--pulsed dendritic cells has proved to be efficacious in various animal models and is being evaluated for the treatment of cancer in humans. Use of dendritic cells pulsed with specific peptides or transfected with tumor-associated antigen genes has been a focused area of investigation for inducing potent tumor and viral immune responses. In this study, the authors demonstrate transgene expression, including the lacZ and MART-1 genes, in dendritic cells infected with adenoviral constructs. These transiently transduced dendritic cells, derived from melanoma patients' monocytes cultured with granulocyte-macrophage colony-stimulating factor and interleukin-4, express the transgene and can stimulate patients' CD8+ T cells to elicit an antitumor immune response comparable to dendritic cells loaded with a defined peptide. These cytotoxic T lymphocytes were able to recognize both known and unknown tumor-associated antigen epitopes and exhibited cytolytic activity against HLA-matched tumor cells expressing the antigen. The ability to induce tumor-specific cytotoxic T lymphocytes in vitro using gene-modified dendritic cells that transiently express tumor-associated antigens demonstrates the potential use of these antigen-presenting cells for developing in vivo cancer vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Melanoma/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenoviruses, Human , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Cells, Cultured , Dendritic Cells/cytology , Gene Expression , Genetic Vectors , Humans , Immunotherapy, Adoptive/methods , MART-1 Antigen , Melanoma/blood , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Peptides/genetics , Peptides/immunology , Transgenes , Tumor Cells, Cultured
8.
J Immunother ; 22(3): 229-36, 1999 May.
Article in English | MEDLINE | ID: mdl-10335482

ABSTRACT

Immunization with tumor-associated antigen pulsed dendritic cells (DC) has been shown to elicit both protective and therapeutic antitumor immunity in a variety of animal models and is currently being investigated for the treatment of cancer patients in clinical trials. In this study we show that DC can be generated from peripheral blood mononuclear cells of healthy donors as well as breast and melanoma cancer patients using granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-13 (IL-13) and that these DC have many of the same characteristics as DC differentiated using GM-CSF and IL-4. The DC generated in GM-CSF and IL-13 are CD14- and express high levels of the cell surface markers CD86, HLA-DR, and CD58, as do DC generated in GM-CSF and IL-4. The purity and yield of both DC populations are not significantly different. Furthermore, both populations of DC are effective at presentation of alloantigen as determined in a mixed lymphocyte response, and both are able to process and present soluble tetanus toxoid antigen to CD4+ T cells. Because we are interested in the generation of DC for antigen-specific cytotoxic T lymphocyte (CTL) generation, we compared the ability of peptide-pulsed DC differentiated in GM-CSF and IL-4 versus GM-CSF and IL-13 for the generation of influenza and MART-1 specific CTL. Both populations of DC induced CD3+ CD8+ CD4- and CD56- CTL, which could lyse the appropriate targets in an antigen-specific manner. Finally, both GM-CSF and IL-4 DC and GM-CSF and IL-13 DC yielded similar beta galactosidase expression levels after transduction with recombinant adenovirus containing the LacZ gene. These results suggest that DC generated in GM-CSF and IL-13 may be useful for immunotherapy and gene therapy protocols.


Subject(s)
Dendritic Cells/immunology , Genetic Therapy , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm , Breast Neoplasms/blood , Breast Neoplasms/immunology , Cell Differentiation , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Immunotherapy , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphocyte Culture Test, Mixed , MART-1 Antigen , Melanoma/blood , Melanoma/immunology , Neoplasm Proteins/immunology
9.
Ann N Y Acad Sci ; 886: 1-11, 1999.
Article in English | MEDLINE | ID: mdl-10667198

ABSTRACT

Telomerase is absent in most normal tissues, but is abnormally reactivated in all major cancer types. Telomerase enables tumor cells to maintain telomere length, allowing indefinite replicative capacity. Albeit not sufficient in itself to induce neoplasia, telomerase is believed to be necessary for cancer cells to grow without limit. The presence of telomerase has been detected in virtually all cancer types including the most prevalent cancers of the prostate, breast, lung, colon, bladder, uterus, ovary, and pancreas as well as in lymphomas, leukemias, and melanomas. In addition, data from cancer patients indicate that telomerase levels correlate with clinical outcome in neuroblastomas, leukemias, and prostate, gastric, and breast cancers. Studies using an antisense to the human telomerase RNA component demonstrate that telomerase in human tumor lines can be blocked ex vivo. In these experiments, telomerase inhibition led to telomere shortening and cancer cell death, validating telomerase as a target for anticancer therapy. Telomerase is a uniquely appealing target for drug discovery because its dichotomic expression in normal versus cancer cells suggests that no serious side effects would result from a treatment abrogating telomerase activity. A variety of approaches to telomerase inhibition are being investigated and are discussed.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Telomerase/antagonists & inhibitors , Humans
10.
Cytotherapy ; 1(5): 401-7, 1999.
Article in English | MEDLINE | ID: mdl-20426540

ABSTRACT

BACKGROUND: Mismatched family donor and unrelated donor BM transplants are associated with a high risk of acute GvHD. White T-cell depletion is the best method to reduce risk of acute GvHD, there was a reluctance to use T-cell depletion in the mismatched setting because of increased risk of rejection and relapse. Partial T-cell depletion, by the panning of CDS and CD8 positive T cells may reduce complications related to GvHD without compromising outcomes. METHOD: In a long-term follow-up of a Phase II study of partial T-cell depletion by panning for BM transplant, 32 recipients received transplants from a single-Ag (HLA A, B, or DR) mismatched family donor; or an HLA serologically-matched unrelated donor. Patients were studied for engraftment, GHD, relapse and survival. RESULTS: 30 (94%) of the patients marrow engrafted. The cumulative risk of Grade 2-4 acute GvHD was 62 - 9%; of Grade 3-4 GvHD, 11 - 6%. The 4-year cumulative risk of relapse was 18 - 8% and actuarial survival was 44 - 9%. DISCUSSION: Partial T-cell depletion had a low rate of severe acute GvHD without compromising engrafment or relapse risk.


Subject(s)
Bone Marrow Transplantation/methods , CD5 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Adolescent , Adult , Bone Marrow Cells/cytology , Child , Female , Follow-Up Studies , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Histocompatibility , Humans , Leukemia/therapy , Lymphoma/therapy , Male , Recurrence , Treatment Outcome
11.
Cancer Gene Ther ; 5(4): 236-46, 1998.
Article in English | MEDLINE | ID: mdl-9694075

ABSTRACT

Immunization with specific tumor-associated antigen (Ag) (TAA)-pulsed dendritic cells (DC) has proven to be efficacious in a variety of animal models and is being investigated for the treatment of cancer patients. Use of DC pulsed with specific peptides or transfected with TAA genes has been a focused area of investigation for the induction of potent tumor and viral immune responses. In this study we demonstrate transgene expression, including expression of the MART-1 gene, in DC transfected with plasmid DNA and cationic liposome complexes. These transiently transfected DC, derived from healthy donor monocytes cultured with granulocyte macrophage colony-stimulating factor and interleukin-4, express the transgene and can stimulate naive CD8+ T cells to elicit an antitumor immune response. These cytotoxic T lymphocytes (CTL) were capable of recognizing both known and unknown TAA epitopes and were able to exhibit cytolytic activity against human histocompatibility leukocyte Ag-matched tumor cells expressing the Ag. In addition to their cytolytic function, the CTL displayed an oligoclonal T-cell receptor repertoire, indicating that the presented Ag induced alterations in the T-cell population. The ability to induce tumor-specific CTL in vitro using gene-modified DC transiently expressing TAAs demonstrates the potential use of these Ag-presenting cells to generate future in vivo cancer vaccine strategies.


Subject(s)
Carcinoembryonic Antigen/genetics , Dendritic Cells/physiology , Neoplasm Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoembryonic Antigen/metabolism , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dependovirus/genetics , Genes, MHC Class I , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-4/pharmacology , Liposomes , MART-1 Antigen , Major Histocompatibility Complex , Monocytes , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Transgenes
12.
J Cell Sci ; 111 ( Pt 13): 1867-75, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9625749

ABSTRACT

Human adult hematopoietic stem cells are mostly quiescent or slow cycling. We have previously demonstrated that blocking of transforming growth factor-beta1 (TGF-beta1) is able to activate, in the presence of cytokines, primitive quiescent hematopoietic multipotent progenitors which could not grow in a two week semi-solid culture assay (short term culture). We have also shown that anti-TGF-beta1 can up-modulate c-KIT, the receptor of the stem cell factor (steel factor). To elucidate whether TGF-beta1 plays a central role in controlling the quiescence of hematopoietic primitive cells, it was necessary to demonstrate, as detailed in this study, that: (1) whatever the cytokine combination tested, addition of anti-TGF-beta1 releases from quiescence multipotent progenitors with a significantly higher hematopoietic potential than those activated by cytokines alone. (2) Other important cytokine receptors controlling the most primitive hematopoietic cells such as FLT3 and the IL6 receptor (IL6-R) are down-modulated by TGF-beta1 but rapidly up-modulated by anti-TGF-beta1. (3) Anti-TGF-beta1-sensitive multipotent and high proliferative potential progenitors express these cytokine receptors at a low level (FLT3(low) and IL6-Rlow). According to these results, we propose the working model of 'High Proliferative Potential-Quiescent cells' to refer to these primitive hematopoietic multipotent progenitors that are highly sensitive to the growth inhibitory effect of TGF-beta1. This model could be valid not only to study the human hematopoietic quiescent progenitors but also for other somatic stem cell systems.


Subject(s)
Hematopoietic Stem Cells/metabolism , Models, Biological , Antibodies/pharmacology , Antigens, CD34/analysis , Antigens, CD34/biosynthesis , Cell Division/drug effects , Cells, Cultured , Fetal Blood , Fibroblast Growth Factor 2/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Interphase/drug effects , Membrane Proteins/biosynthesis , Membrane Proteins/pharmacology , Receptors, Interleukin-6/analysis , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology
13.
J Hematother ; 6(4): 335-49, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9377072

ABSTRACT

The isolation and culture of human CD34+ cells could have broad clinical application for hematologic support following high-dose chemotherapy or bone marrow transplantation. The need for reproducible, animal product-free conditions for the culture of progenitors is crucial to the widespread clinical implementation of ex vivo cell therapies. In these studies, we explored the use of animal serum-free (ASF) medium for the culture of isolated human bone marrow and peripheral blood CD34+ cells. In this ASF system, isolated CD34+ cells were cultured using a variety of different growth factor combinations. Such ASF culture conditions yielded equivalent to superior cell and progenitor growth when directly compared with culture containing 10% fetal calf serum (FCS). In cultures containing IL-1, IL-3, and stem cell factor, total cell numbers increased, on average, 33-fold over the first 2 weeks. On phenotypic analysis, the ASF cultures demonstrated sustained proliferation of CD33+ myeloid cells throughout the culture period. CD34+ cell numbers increased during the first 7-10 days of culture, with a mean 3.4-fold expansion. Concomitant with the CD34+ cell expansion was an average 8.2-fold expansion of colony-forming unit-granulocyte-macrophage (CFU-GM) and a 102.0-fold increase in burst-forming units-erythrocytes (BFU-E). Likewise, a mean 4929-fold expansion of CD41a+ megakaryocyte progenitors was observed in these CD34+ cultures. Different combinations of growth factors affected the fold increase in cell and progenitor number. When CD34+ cell cultures from normal healthy volunteers mobilized with either G-CSF or GM-CSF were compared, similar expansions of total cell and progenitor cells resulted. However, CD41+ cells expansions were greater in those samples from G-CSF-mobilized volunteers in every case tested. These studies established the feasibility of this ASF CD34+ cell culture system to generate a population of maturing progenitors for potential use in transfusion support during cytopenic periods following high-dose chemotherapy or bone marrow transplantation.


Subject(s)
Antigens, CD34/blood , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Animals , Cell Lineage , Cell Separation , Cells, Cultured , Culture Media, Serum-Free , Erythroid Precursor Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Megakaryocytes/drug effects , Reference Values , Reproducibility of Results
14.
Biol Blood Marrow Transplant ; 3(1): 34-44, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9209739

ABSTRACT

Autologous transplantation can induce extended remission in some patients with advanced breast cancer and lymphoma yet nearly 80% and 50%, respectively, will ultimately relapse. In vitro studies suggest that activated natural killer cells (NK) mediate lytic activity against breast cancer and lymphoma cell lines. Therefore, immunotherapy with interleukin-2 (IL-2, Amgen) to activate NK may improve long-term disease-free survival when administered in a post-transplant minimal residual disease setting. To determine the feasibility of administering IL-2 and activation of NK post-transplant, twelve patients (6 breast cancer, 6 lymphoma) were enrolled on a phase I dose escalation study after autologous transplantation (median day + 94, range 50-166). IL-2 was self administered at 0.25 x 10(6) (n = 6) or 0.5 x 10(6) (n = 6) U/m2/day subcutaneously for 84 consecutive days. The best tolerated dose was 0.25 x 10(6) U/m2/day (75% of planned doses given vs. 48% at the higher dose). Dose limiting toxicity occurred in 6 patients (n = 2 at 0.25 x 10(6) U/m2/day, n = 4 at 0.5 x 10(6) U/m2/day) consisting of decreased performance status (n = 2), thrombocytopenia (n = 3, 1 at the lower dose), and mild neutropenia (n = 1 at the lower dose). However, all symptoms resolved within a week following discontinuation of IL-2 and no patient required hospitalization. Circulating soluble IL-2 receptor levels were significantly increased in all patients receiving IL-2. Patients receiving at least 28 days of IL-2 exhibited a greater than 10-fold increment in circulating CD56+bright/CD3- NK. Furthermore, lytic function was increased against NK resistant targets, MCF-7 (breast cancer), and Raji (lymphoma). In vivo IL-2 primed NK cells obtained by lymphapheresis were activated in large-scale ex vivo incubation in high dose IL-2 (1,000 U/mL) at high cell density (10 x 10(6)/mL), in gas permeable bags, and using serum-free media. NK lytic function against MCF-7 and Raji targets was further enhanced. We conclude that low dose subcutaneous IL-2 based immunotherapy is feasible, relatively safe, can be administered in an outpatient setting and hypothesize that additional ex vivo incubation in IL-2 may be used to generate NK cells with potent antitumor effects in vivo.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Bone Marrow Transplantation/immunology , Hematopoietic Stem Cell Transplantation , Interleukin-2/therapeutic use , Killer Cells, Natural/drug effects , Transplantation, Autologous/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Adult , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Female , Humans , Injections, Subcutaneous , Interleukin-2/administration & dosage , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphoma/immunology , Lymphoma/therapy , Male , Middle Aged , Self Administration , Treatment Outcome , Tumor Cells, Cultured
16.
Antiviral Res ; 32(2): 99-115, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8891169

ABSTRACT

RNA decoys are oligonucleotides corresponding to the TAR and RRE sequences of HIV which inhibit the HIV-encoded regulatory proteins Tat and Rev, respectively. Adeno-associated viral vectors encoding RNA decoys stably transduced into the human T-cell line CEM-SS expressed transactivating region (TAR) and Rev-responsive element (RRE) RNA decoys from tRNA polIII promoters at high levels, without any apparent deleterious effects on cell growth or expression of CD4. DNA blot analysis indicated that RNA decoy-encoding vectors were not rearranged and were integrated into the genomic DNA of selected cell lines. Vector DNA with the appropriate TAR and RRE sequences was isolated from transduced cell lines after prolonged growth in culture, further confirming that the vector DNA was present in a stable form through multiple cell cycles. Cell lines expressing TAR and RRE decoys transiently inhibited HIV gene expression and replication by 70-99% as determined by measurement of intracellular and extracellular HIV p24 production. Adeno-associated vectors encoding RNA decoys may be useful for gene therapy of HIV infection.


Subject(s)
Dependovirus/genetics , Genetic Vectors , HIV Long Terminal Repeat/genetics , HIV-1/physiology , RNA, Viral/genetics , T-Lymphocytes/virology , Cell Line , Gene Products, rev/antagonists & inhibitors , Gene Products, rev/metabolism , Gene Products, tat/antagonists & inhibitors , Gene Products, tat/metabolism , Gene Transfer Techniques , Genetic Therapy , HIV Core Protein p24/biosynthesis , Humans , RNA Processing, Post-Transcriptional , RNA, Transfer/genetics , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
17.
Biotechniques ; 20(6): 1098-103, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8780881

ABSTRACT

We have established rapid procedures that negatively deplete and positively select for specific murine cell populations. By using polystyrene tissue culture flasks containing a covalently bound mouse anti-rat antibody and specific anti-mouse, cell-surface antigen antibodies, we easily and efficiently depleted greater than 90% of the mature lineage cells from murine bone marrow. This selection procedure resulted in an enrichment of progenitor colonies (CFU-Cs) in murine bone marrow. Using the same polystyrene tissue culture devices, we can directly isolate CD117+ (c-kit+) murine hematopoietic cells. As few as 2000 of these CD117+ cells rescued and reconstituted lethally irradiated recipients in a murine bone marrow transplant model.


Subject(s)
Cell Separation/methods , Culture Techniques/methods , Hematopoietic Stem Cells , Animals , Bone Marrow Transplantation , Cell Lineage , Culture Techniques/instrumentation , Flow Cytometry/methods , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/immunology
19.
Clin Cancer Res ; 2(1): 59-68, 1996 Jan.
Article in English | MEDLINE | ID: mdl-9816091

ABSTRACT

We have previously shown that cationic liposomes facilitate adeno-associated virus (AAV) plasmid transfections of primary and cultured cell types. To test the clinical feasibility of using genetically modified tumor vaccines for the treatment of breast and ovarian cancers, we have constructed an expression plasmid pMP6IL2 and investigated the use of liposome-mediated gene delivery into primary, uncultured human breast and ovarian tumor cells to produce interleukin 2 (IL-2)-secreting tumor cells. We have demonstrated significant levels of IL-2 expression in tumor cell lines and primary breast and ovarian tumor cells using this AAV-based expression plasmid complexed to cationic liposomes. Transfections with the non-AAV plasmid containing the identical expression cassette as the AAV plasmid induced IL-2 expression in the tumor cell line but failed to produce IL-2 in primary tumor cells. Significant levels of IL-2 were induced with the AAV plasmid regardless of liposome compositions used for transfection. The transfected breast cell line and primary tumor cells were able to express the transgene product for up to 28 days after lethal radiation. The transfection efficiency was comparable for both the tumor cell line and primary tumor cells and ranged from 20 to 50% for both cell types as assessed by intracellular IL-2 staining. Although the primary tumor cell preparations consist of mixed population of cells, at least 40% of the tumor cells expressed the transgene as assessed by immunostaining for IL-2. The ability to efficiently express transgenes in freshly isolated, nondividing tumor cells may potentiate active immunotherapy strategies for gene-based cancer treatment.


Subject(s)
Breast Neoplasms/therapy , Dependovirus/genetics , Genetic Therapy , Immunotherapy, Active , Interleukin-2/genetics , Ovarian Neoplasms/therapy , Blotting, Southern , Female , Gene Transfer Techniques , Humans , Plasmids , Tumor Cells, Cultured
20.
Stem Cells ; 13(6): 607-12, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8590862

ABSTRACT

The development of serum-free systems for the maintenance and expansion of both primitive and committed hematopoietic progenitors has numerous applications in both basic and clinical research. Many different media have been tested and refined over the years, and current formulations now yield results similar to those observed with fetal bovine serum-based medias. Using these serum-free culture systems, the impact of the cell microenvironment and individual growth factors on primitive and maturing stem cells have both been studied. The utility of progenitor populations expanded ex vivo under serum-free conditions is under investigation.


Subject(s)
Hematopoietic Stem Cells/physiology , Animals , Cattle , Cells, Cultured , Culture Media, Serum-Free , Humans , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...