Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Am J Transplant ; 8(10): 2140-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18727701

ABSTRACT

Primary graft dysfunction (PGD) continues to be a major cause of early death after lung transplantation. Moreover, there remains a lack of accurate pretransplant molecular markers for predicting PGD. To identify distinctive donor lung gene expression signatures associated with PGD, we profiled human donor lungs using microarray technology prior to implantation. The genomic profiles of 10 donor lung samples from patients who subsequently developed clinically defined severe PGD were compared with 16 case-matched donor lung samples from those who had a favorable outcome without PGD (development set, n = 26). Selected PCR validated predictive genes were tested by quantitative reverse transcription-polymerase chain reaction in an independent test set (n = 81). Our microarray analyses of the development set identified four significantly upregulated genes (ATP11B, FGFR2, EGLN1 and MCPH1) in the PGD samples. These genes were also significantly upregulated in donor samples of the test set of patients with poor outcomes when compared to those of patients with good outcomes after lung transplantation. This type of biological donor lung assessment shows significant promise for development of a more accurate diagnostic strategy to assess donor lungs prior to implantation.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Lung Diseases/genetics , Lung Diseases/therapy , Lung Transplantation/methods , Lung/metabolism , Primary Graft Dysfunction/diagnosis , Adult , Case-Control Studies , Cluster Analysis , Female , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Primary Graft Dysfunction/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
J Clin Invest ; 100(9): 2243-53, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9410902

ABSTRACT

Optimal T cell responsiveness requires signaling through the T cell receptor (TCR) and CD28 costimulatory receptors. Previously, we showed that T cells from autoimmune nonobese diabetic (NOD) mice display proliferative hyporesponsiveness to TCR stimulation, which may be causal to the development of insulin-dependent diabetes mellitus (IDDM). Here, we demonstrate that anti-CD28 mAb stimulation restores complete NOD T cell proliferative responsiveness by augmentation of IL-4 production. Whereas neonatal treatment of NOD mice with anti-CD28 beginning at 2 wk of age inhibits destructive insulitis and protects against IDDM by enhancement of IL-4 production by islet-infiltrating T cells, administration of anti-CD28 beginning at 5-6 wk of age does not prevent IDDM. Simultaneous anti-IL-4 treatment abrogates the preventative effect of anti-CD28 treatment. Thus, neonatal CD28 costimulation during 2-4 wk of age is required to prevent IDDM, and is mediated by the generation of a Th2 cell-enriched nondestructive environment in the pancreatic islets of treated NOD mice. Our data support the hypothesis that a CD28 signal is requisite for activation of IL-4-producing cells and protection from IDDM.


Subject(s)
CD28 Antigens/metabolism , Diabetes Mellitus, Type 1/immunology , Interleukin-4/physiology , T-Lymphocytes/immunology , Animals , Animals, Newborn , Cell Survival , Clonal Anergy , Female , Glutamate Decarboxylase/immunology , Immunization, Passive , Interleukin-2/biosynthesis , Islets of Langerhans/immunology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Signal Transduction , Th2 Cells/immunology
4.
Cancer Res ; 53(5): 1043-50, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8439951

ABSTRACT

The adoptive immunotherapy of human malignancy requires reliable methods to sensitize and expand patients' T-cells reactive to autologous tumors. In animal studies, we have generated therapeutic effector cells against a poorly immunogenic tumor by a two-step procedure: vaccination of the host followed by the secondary stimulation of vaccine-primed lymph node (LN) cells by in vitro sensitization (IVS) with tumor in the presence of interleukin 2 (IL-2). Based on these observations, we performed a clinical trial in patients with advanced cancer to evaluate the antitumor efficacy of vaccine-primed LN cells which were similarly activated in vitro. Patients were vaccinated with irradiated autologous tumor admixed with Bacillus Calmette-Guérin and had draining LN excised 10 days later for IVS culture. During IVS culture, LN cells expanded up to 14-fold (average of 8.4-fold). A mean of 6.7 x 10(9) cells was infused in ten patients (seven melanoma, three renal cell cancer) along with the concomitant i.v. administration of IL-2 (180,000 IU/kg every 8 h for 5 days). Phenotype analysis of IVS-LN cells revealed 78 +/- 4% CD3+ T-cells which were predominantly CD4+ (67 +/- 5%) with expression of HLA-DR and IL-2 receptor. IVS-LN cells displayed relative specificity of autologous tumor lysis in four of ten cases compared to zero of seven IVS-peripheral blood lymphocytes derived from the same patients as measured by the 51Cr release assay. One mo after therapy, seven of nine patients treated with IVS-LN cells and IL-2 developed delayed-type hypersensitivity reactivity to autologous tumor compared to zero of nine patients treated with tumor vaccination and IL-2 only (P < 0.002). These observations suggest that antitumor reactivity was passively transferred with the IVS-LN cells. Major toxic side effects including fever, hepatic dysfunction, and weight gain associated with the capillary leak syndrome were associated with exogenous IL-2 administration. Tumor vaccination and cell transfer were well tolerated without significant complications. Of the ten patients treated with IVS-LN cells and IL-2, there were one partial and one minor response, and one patient has had stable disease for 27+ mo. There was no evidence of tumor response in ten patients treated with tumor vaccination and IL-2 only. Further clinical studies evaluating the antitumor reactivity of vaccine-primed LN cells are warranted.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , T-Lymphocytes/immunology , Vaccines/immunology , Adult , Aged , CD3 Complex/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Cytotoxicity, Immunologic , Female , Humans , Hypersensitivity, Delayed , Immunotherapy, Adoptive/adverse effects , Interleukin-2/therapeutic use , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Lymph Nodes/immunology , Male , Melanoma/immunology , Melanoma/therapy , Middle Aged , Phenotype , Vaccination
5.
Mucosal Immunol ; 9(6): 1528-1536, 2016 11.
Article in English | MEDLINE | ID: mdl-26838048

ABSTRACT

Understanding vaginal and rectal HIV transmission and protective cellular and molecular mechanisms is critical for designing new prevention strategies, including those required for an effective vaccine. The determinants of protection against HIV infection are, however, poorly understood. Increasing evidence suggest that innate immune defenses may help protect mucosal surfaces from HIV transmission in highly exposed, uninfected subjects. More recent studies suggest that systemically administered type 1 interferon protects against simian immunodeficiency virus infection of macaques. Here we hypothesized that topically applied type 1 interferons might stimulate vaginal innate responses that could protect against HIV transmission. We therefore applied a recombinant human type 1 interferon (IFN-ß) to the vagina of rhesus macaques and vaginally challenged them with pathogenic simian/human immunodeficiency virus (SHIV). Vaginal administration of IFN-ß resulted in marked local changes in immune cell phenotype, increasing immune activation and HIV co-receptor expression, yet provided significant protection from SHIV acquisition as interferon response genes were also upregulated. These data suggest that protection from vaginal HIV acquisition may be achieved by activating innate mucosal defenses.


Subject(s)
Antiviral Agents/administration & dosage , Interferon-beta/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/drug effects , Administration, Intravaginal , Administration, Topical , Animals , Biomarkers , CD4 Antigens/metabolism , Female , Gene Expression Regulation/drug effects , Lymphocyte Activation/immunology , Macaca mulatta , Macrophages/immunology , Macrophages/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phenotype , Receptors, CCR5/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vagina/immunology , Vagina/virology , Viral Load
6.
J Clin Oncol ; 15(2): 796-807, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9053507

ABSTRACT

PURPOSE: In preclinical studies, we have reported the ability to induce immune T cells in lymph nodes (LN) primed by in vivo vaccination with tumor cells admixed with a bacterial adjuvant. These LN cells can be activated and expanded ex vivo for the successful immunotherapy of established tumors. We have applied these methods to generate vaccine-primed LN in patients with advanced melanoma and renal cell cancer (RCC) for therapy. MATERIALS AND METHODS: Irradiated autologous tumor cells admixed with bacille Calmette-Guérin (BCG) were used to vaccinate patients. Seven days later, draining LN were removed for activation with anti-CD3 monoclonal antibody (mAb) followed by expansion in interleukin-2 (IL-2). Activated LN cells were administered intravenously (IV) with the concomitant administration of IL-2. RESULTS: A total of 23 patients were evaluated (11 melanoma and 12 RCC). Vaccine-primed LN were expanded ex vivo with a mean of 8.4 x 10(10) cells administered per patient. Among 20 patients assessed, 15 demonstrated minimal cytotoxicity of autologous tumor cells by the activated LN cells, with the remaining mediating nonspecific cytotoxicity. By contrast, a majority of the activated LN cells showed highly specific release of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interferon gamma (IFN-gamma) to autologous but not allogeneic tumor stimulation. This tumor-specific cytokine release was found to be major histocompatibility complex (MHC) class I-restricted, which indicates the involvement of CD8+ cells. Among 11 melanoma patients, one had a partial tumor response. Among 12 RCC patients, two had complete and two partial responses. A trend (P = .066) between the enhancement of delayed-type hypersensitivity (DTH) reactivity to autologous tumor after therapy and tumor regression was observed. CONCLUSION: Tumor vaccines can be used to induce immunologically specific T-cell responses against melanoma and RCC in draining LN. Anti-CD3/IL-2 activation of primed LN cells can be reliably performed for clinical therapy and appears to have activity in patients with metastatic RCC.


Subject(s)
BCG Vaccine/therapeutic use , CD3 Complex/pharmacology , Carcinoma, Renal Cell/therapy , Immunotherapy, Adoptive/methods , Interleukin-2/pharmacology , Kidney Neoplasms/therapy , Killer Cells, Lymphokine-Activated/drug effects , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma/therapy , Adult , Carcinoma, Renal Cell/immunology , Drug Screening Assays, Antitumor , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Hypersensitivity, Delayed , Immunity, Cellular/drug effects , Interferon-gamma/biosynthesis , Kidney Neoplasms/immunology , Male , Melanoma/immunology , Middle Aged , Phenotype , Treatment Outcome
7.
Diabetes ; 48(9): 1720-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10480600

ABSTRACT

The mechanism of protection from type 1 diabetes conferred by regulatory T-cells induced by oral insulin treatment of NOD mice is not well understood. We demonstrate that oral insulin feeding of NOD mice induces the function of insulin B-chain reactive CD4+ regulatory T-cells, which compete with diabetogenic effector T-cells for the recognition of insulin in NOD.Scid recipient mice. These effector T-cells become deprived of interleukin (IL)-2 and interferon (IFN)-gamma and are unable to expand and migrate to the pancreas. Type 1 diabetes-protective splenic regulatory T-cells secrete relatively little transforming growth factor (TGF)-beta1, suggesting that TGF-beta may not contribute to the inactivation of effector T-cells in NOD.Scid recipients. The observed preferential infiltration of insulin-reactive regulatory T-cells rather than effector T-cells in the pancreas results in a nondestructive insulitis that correlates with an increased intrapancreatic expression of macrophage inflammatory protein-1beta. Thus, oral insulin therapy overcomes a deficiency in regulatory T-cells and protects against type 1 diabetes by inducing insulin B-chain reactive regulatory T-cells to block cytokine secretion and migration of diabetogenic effector T-cells to the pancreas. Our data emphasize that continuous oral insulin feeding over a prolonged period is required to prevent type 1 diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cytokines/metabolism , Diabetes Mellitus, Type 1/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Administration, Oral , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/immunology , Insulin/chemistry , Insulin/immunology , Mice , Mice, Inbred NOD , Pancreas/immunology , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
8.
Crit Rev Immunol ; 17(5-6): 537-44, 1997.
Article in English | MEDLINE | ID: mdl-9419441

ABSTRACT

T cells from NOD mice display an age-dependent, TCR-inducible proliferative hyporesponsiveness that may be causal to IDDM. Exogenous IL-4 completely restores this hyporesponsiveness in vitro and prevents IDDM in vivo when administered to NOD mice. We therefore tested the hypothesis that stimulation of a Th2 response by either IL-4 or CD28 costimulation may block progression to IDDM. Low-dose IL-4 treatment beginning at 2 weeks of age (pre-insulitis) protects NOD mice from insulitis, sialitis, and thyroiditis, indicating that IL-4 modulates T cell migration to these inflammatory sites. Cytokine secretion profiles of stimulated T cells and assays of intrapancreatic cytokine concentrations revealed that IL-4 treatment prevents IDDM by stabilizing a protective Th2-mediated environment in the thymus, spleen, and pancreatic islets. Whereas treatment of NOD mice with an anti-CD28 mAb between 2 to 4 weeks of age inhibits destructive insulitis and protects against IDDM by enhancing IL-4 production by T cells, anti-CD28 treatment between 5 to 7 weeks of age does not prevent IDDM. Simultaneous anti-IL-4 treatment abrogates the protective effect conferred by anti-CD28 treatment. Our data demonstrate that stimulation of a Th2-cell-enriched environment in the pancreas during the inductive phase of disease development blocks progression to IDDM in NOD mice.


Subject(s)
CD28 Antigens/immunology , Diabetes Mellitus, Type 1/prevention & control , Interleukin-4/pharmacology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , CD28 Antigens/pharmacology , Cell Division , Chemokines, CC/immunology , Cytokines/immunology , Cytokines/pharmacology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Humans , Interleukin-4/immunology , Mice , Mice, Inbred NOD , T-Lymphocytes/cytology , Th2 Cells/immunology
9.
J Leukoc Biol ; 61(4): 507-16, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9103238

ABSTRACT

We have previously demonstrated that the growth of weakly immunogenic murine sarcomas leads to the induction of immunologically specific pre-effector cells in tumor-draining lymph nodes (TDLN). The in vitro activation of TDLN cells with anti-CD3 monoclonal antibodies (mAbs) and interleukin-2 (IL-2) resulted in the acquisition of effector function as measured by tumor regression in the adoptive immunotherapy of pulmonary metastases. Further studies were performed to characterize the mechanisms associated with in vivo tumor reactivity mediated by activated TDLN cells. By positive selection, CD4+ and CD8+ T cells were purified and activated by the anti-CD3/IL-2 method. CD8+, but not CD4+, cells manifested tumor-specific granulocyte-macrophage colony-stimulating factor (GM-CSF) and interferon-gamma (IFN-gamma) release in vitro, and elicited tumor regression in vivo. By contrast, only activated CD4+ were found to release significant amounts of IL-2 in response to tumor antigen but did not mediate tumor regression in vivo. Mixing the two purified populations enhanced the antitumor activity of the CD8+ T cells. In culture, IL-2 was found to augment the relative amount of tumor-specific release of GM-CSF and IFN-gamma by activated TDLN cells. We found that the tumor-specific release of GM-CSF and IFN-gamma by activated lymphocytes was strongly associated with the in vivo therapeutic efficacy of these cells. Evidence in support of this included the following: (1) cytokine release of TDLN derived after different durations of tumor growth correlated with tumor reactivity in adoptive transfer studies, (2) cytokine release of T cells derived from different lymphoid organs corresponded with tumor reactivity in adoptive transfer, and (3) in vivo administration of neutralizing mAb to IFN-gamma and GM-CSF significantly inhibited the antitumor reactivity of TDLN cells. These studies document the contributory roles of IFN-gamma, GM-CSF, and IL-2 released by activated CD4+ and CD8+ T cells involved in tumor regression.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cytokines/physiology , Fibrosarcoma/immunology , Lymph Nodes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Cytokines/metabolism , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Immunotherapy, Adoptive , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interferon-gamma/physiology , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2/physiology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL
10.
Food Chem Toxicol ; 77: 120-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25592784

ABSTRACT

A toxicity/toxicokinetic swine-adapted infant formula feeding study was conducted in Domestic Yorkshire Crossbred Swine from lactation day 3 for 28 consecutive days during the preweaning period at carrageenan concentrations of 0, 300, 1000 and 2250 ppm under GLP guidelines. This study extends the observations in newborn baboons (McGill et al., 1977) to piglets and evaluates additional parameters: organ weights, clinical chemistry, special gastrointestinal tract stains (toluidine blue, Periodic Acid-Schiff), plasma levels of carrageenan; and evaluation of potential immune system effects. Using validated methods, immunophenotyping of blood cell types (lymphocytes, monocytes, B cells, helper T cells, cytotoxic T cells, mature T cells), sandwich immunoassays for blood cytokine evaluations (IL-6, IL-8, IL1ß, TNF-α), and immunohistochemical staining of the gut for IL-8 and TNF-α were conducted. No treatment-related adverse effects at any carrageenan concentration were found on any parameter. Glucosuria in a few animals was not considered treatment-related. The high dose in this study, equivalent to ~430 mg/kg/day, provides an adequate margin of exposure for human infants, as affirmed by JECFA and supports the safe use of carrageenan for infants ages 0-12 weeks and older and infants with special medical needs.


Subject(s)
Carrageenan/pharmacokinetics , Gastrointestinal Tract/drug effects , Immune System/drug effects , Infant Formula/chemistry , Animals , Animals, Newborn , Body Weight/drug effects , Carrageenan/adverse effects , Carrageenan/blood , Dose-Response Relationship, Drug , Female , Interleukin-1beta/blood , Interleukin-6/blood , Interleukin-8/blood , Male , Organ Size/drug effects , Swine , Toxicity Tests , Tumor Necrosis Factor-alpha/blood
11.
Hum Gene Ther ; 11(12): 1647-56, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10954899

ABSTRACT

We tested the efficacy of biolistic-mediated gene transfer as a noninvasive therapy for type 1 diabetes (T1D) in nonobese diabetic (NOD) mice by expression of murine interleukin 4 (mIL-4) cDNA. Epidermal delivery of 2 microg of DNA yielded transient detection of serum mIL-4, using a conventional cDNA expression vector. A vector stabilized by incorporation of the Epstein-Barr virus (EBV) EBNA1/oriP episomal maintenance replicon produced higher levels of serum mIL-4 that persisted for 12 days after inoculation. Although biolistic inoculation of either vector reduced insulitis and prevented diabetes, the protracted mIL-4 expression afforded by the EBV vector resulted in Th2-type responses in the periphery and pancreas and more significant protection from the onset of diabetes. Our studies demonstrate the efficacy of biolistic gene delivery of stabilized cytokine expression as a viable therapeutic approach to prevent the onset of T1D.


Subject(s)
Biolistics , Diabetes Mellitus, Type 1/prevention & control , Interleukin-4/genetics , Animals , Cytokines/analysis , Cytokines/metabolism , DNA, Complementary/genetics , Female , Flow Cytometry , Genetic Vectors , Herpesvirus 4, Human/genetics , Immunoglobulin E/blood , Interleukin-4/metabolism , Mice , Mice, Inbred NOD , Pancreas/metabolism , Pancreas/pathology , T-Lymphocytes/metabolism
12.
J Clin Endocrinol Metab ; 62(6): 1116-23, 1986 Jun.
Article in English | MEDLINE | ID: mdl-3457797

ABSTRACT

Endometrial stromal cells and isolated endometrial glands obtained from women during days 6-26 of the ovarian cycle were cultured for 24 h in the presence of the progesterone antagonists 17 beta-hydroxy-11 beta-[4-dimethylaminophenyl]17 alpha-[1-propynyl] estra-4,9-dien-3-one (RU486) and 17 beta-hydroxy-11 beta-[4-dimethylaminophenyl] 17 alpha-[3-hydroxy-1-propenyl]estra-4,9-dien-3-one (ZK 98734). Both steroids stimulated prostaglandin F2 alpha (PGF2 alpha) production by stromal cells in a dose-dependent manner, in doses ranging from 10-1000 nM. Progesterone (100 nM) inhibited RU486 stimulation, except at the highest dose of antiprogestin. PGE2 was produced in smaller amounts than PGF2 alpha, but, when measurable, it also increased in the presence of RU486. In contrast, RU486 did not increase PG production by endometrial glands. In an experiment to determine the effect of pretreatment, stromal cells were incubated for 24 h with 1000 nM progesterone or RU486 (all with 100 nM 17 beta-estradiol) with either 30 or 6 microM arachidonic acid. These six batches of cells were incubated for a second 24 h with either progesterone or antiprogestin. Cells pretreated with the higher dose of arachidonic acid had a marked increase in PGF2 alpha production during the second 24 h only when also pretreated with progesterone. This finding suggests that progesterone allows an accumulation of PG precursor in a suitable accessible pool. Pretreatment with progesterone also allowed a greater conversion of PG to its 13,14-dihydro-15-keto metabolite. These results suggest that antiprogesterone steroids may act as menstrual regulators by: stimulating endogenous PG production within the endometrial stromal cells and inhibiting PG catabolism.


Subject(s)
Endometrium/metabolism , Estrenes/pharmacology , Progesterone/antagonists & inhibitors , Prostaglandins/biosynthesis , Dinoprost , Dinoprostone , Endometrium/drug effects , Fatty Acids/metabolism , Female , Humans , In Vitro Techniques , Mifepristone , Progesterone/pharmacology , Prostaglandins E/biosynthesis , Prostaglandins F/biosynthesis
13.
Cancer Gene Ther ; 3(1): 39-47, 1996.
Article in English | MEDLINE | ID: mdl-8785710

ABSTRACT

We examined the host immune response to the poorly immunogenic B16-BL6 melanoma, which was transduced to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) (450 ng/10(6)/24 h). Tumor growth after subcutaneous inoculation was not significantly altered, although an influx of neutrophils and monocytes/macrophages was evident within tumors and draining lymph nodes (LNs). Immunization with irradiated transduced cells did not induce systemic immunity to the parental tumor. However, vaccination with transduced tumors significantly augmented in vivo sensitization of draining LN cells. These tumor-draining LN (TDLN) cells, when secondarily stimulated in vitro with anti-CD3 monoclonal antibodies and expanded in interleukin-2 (10 U/ml), exhibited greater release of GM-CST and interferon-gamma against tumor compared with TDLN cells from animals with parental tumor. In adoptive immunotherapy, activated LN cells draining transduced tumors mediated significant reductions of the numbers of established pulmonary metastases compared with LN cells draining parental tumor, which were ineffective. In addition, the therapeutic efficacy of LN cells draining transduced tumors was significantly better than LN cells primed in vivo with tumor cells admixed with Corynebacterium parvum, which we have previously described as an approach to generate immune cells. Thus, GM-CSF appears to be an important adjuvant in the induction of tumor immunity.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lymph Nodes/immunology , Melanoma, Experimental/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Base Sequence , CD3 Complex/immunology , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-2/immunology , Major Histocompatibility Complex/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Tumor Cells, Cultured
14.
J Immunol Methods ; 183(2): 239-50, 1995 Jun 28.
Article in English | MEDLINE | ID: mdl-7602147

ABSTRACT

Augmentation of tumor immunogenicity has been increasingly studied as a strategy to develop host immunity against established malignancies. Genetic modification of tumors to secrete immunoregulatory peptides such as IL-4 has been demonstrated to augment tumor immunogenicity and enhance the induction of tumor reactive lymphoid cells in animal models. To explore the ability of IL-4 to augment the immunogenicity of melanoma cells, we constructed a recombinant retrovirus vector encoding for human IL-4 and used it to transduce human melanomas. After optimizing retrovirus transduction conditions using a reporter virus, an IL-4 encoding retrovirus vector was used to transduce early and late passage melanoma cells. IL-4 production rates of up to 2000 pg/ml per 24 h per 10(6) cells were achieved, and provirus could be detected by Southern blot of the transduced cells at 0.1 copies per cell. The IL-4 produced by the melanoma cells was biologically active. Irradiated transduced melanoma cells continued to produce IL-4 for at least two weeks of observation. Thus melanoma cells can be efficiently modified to secrete biologically active IL-4, and may be suitable substrates for autologous tumor cell vaccines.


Subject(s)
Interleukin-4/genetics , Interleukin-4/metabolism , Melanoma/therapy , Cryopreservation , Genetic Therapy , Humans , Immunotherapy, Active , Melanoma/metabolism , Retroviridae/genetics , Transfection , Tumor Cells, Cultured
15.
J Autism Dev Disord ; 24(5): 619-29, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7814310

ABSTRACT

Treated persistent ruminative vomiting of a 15-year-old boy with autism using a multicomponent behavioral medicine program within a residential facility. Preceding intervention the boy had lost 15 pounds associated with high-rate ruminating. The treatment program included a combination of dietary, nutritional, and behavioral procedures that emphasized food restrictions, satiation, and setting condition manipulations. Ruminative vomiting was reduced to near-zero levels and weight gain was achieved following treatment implementation. These therapeutic gains were sustained during a maintenance programming phase and at 1- through 4-month follow-up assessments. Issues related to functional assessment and treatment formulation in behavioral medicine intervention for ruminative vomiting are discussed.


Subject(s)
Autistic Disorder/therapy , Behavior Therapy/methods , Stereotyped Behavior , Vomiting/therapy , Weight Loss , Adolescent , Autistic Disorder/psychology , Combined Modality Therapy , Feeding Behavior/psychology , Humans , Lead Poisoning/psychology , Lead Poisoning/therapy , Male , Patient Care Team , Residential Treatment , Vomiting/psychology
16.
Arch Otolaryngol Head Neck Surg ; 119(12): 1289-95, 1993 Dec.
Article in English | MEDLINE | ID: mdl-17431982

ABSTRACT

OBJECTIVE: The genetic modification of tumors offers an approach to modulate the host immune response to relatively weak native tumor antigens. We examined the immunobiologic effects of various cytokine genes transferred into the poorly immunogenic B16-BL6 murine melanoma. DESIGN: Retroviral expression vectors containing cDNAs for interleukin 2, interleukin 4, interferon gamma, or a neomycin-resistant control were electroporated into a B16-BL6 tumor clone. Selected transfected clones were examined for in vitro cytokine secretion and in vivo tumorigenicity. RESULTS: When cells from individual clones were injected intradermally into syngeneic mice, the interleukin 4-secreting clone grew significantly slower than did the neomycin-resistant transfected control, while the growth of the interleukin 2- and interferon gamma-expressing clones was not affected. Despite minimal cytokine secretion by interferon gamma-transfected cells, these cells expressed upregulated major histocompatibility class I antigen and were more susceptible to lysis by allosensitized cytotoxic T lymphocytes compared with parental or neomycin-resistant transfected tumor targets. CONCLUSIONS: We observed diverse immunobiologic effects associated with cytokine gene transfer into the B16-BL6 melanoma. Interleukin 4 transfection of tumor resulted in decreased in vivo tumorigenicity that may be related to a host immune response. Further studies to evaluate the host T-cell response to these gene-modified tumors are being investigated.


Subject(s)
Cytokines/genetics , Cytokines/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Clone Cells , Female , Histocompatibility Antigens Class I/metabolism , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Skin Neoplasms/metabolism , Transfection
17.
Behav Modif ; 17(2): 136-47, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8471009

ABSTRACT

Persons with developmental and physical disabilities who are enrolled in educational programs often participate in adaptive physical education classes. Primarily, these classes are designed to provide individuals with the opportunity to develop their physical abilities. However, they can also serve as a training ground for the Special Olympics. Teaching the motor skills that are prerequisite to participation in many adaptive physical education and Special Olympics activities can be a formidable objective. This study demonstrates how a person with developmental disabilities was taught, by way of stimulus control shaping, the necessary motor skills to enable him to participate in the hurdling event at the Special Olympics.


Subject(s)
Disabled Persons , Down Syndrome/rehabilitation , Education, Special/methods , Physical Education and Training/methods , Adult , Disabled Persons/psychology , Down Syndrome/psychology , Humans , Male , Motor Skills , Physical Fitness
18.
Res Dev Disabil ; 17(3): 203-15, 1996.
Article in English | MEDLINE | ID: mdl-8743553

ABSTRACT

A treatment package that included two setting condition manipulations and visual occlusion was implemented to gain control over the high-intensity screaming and whining of a 16-year-old female with developmental disabilities. The study included an analysis of the individual and combined components of the treatment package and a stimulus control analysis of three salient features of the visual occlusion apparatus (i.e., opaque screen, secured helmet, and cranial pressure). Results showed that the treatment package occasioned a deceleration in the two targeted vocal behaviors and a reduction in the amount of time the participant was required to wear the occlusion apparatus. An analysis of the apparatus suggested that the critical element needed to control inappropriate vocalizations appeared to be cranial pressure, which was naturally produced by the helmet. Consequently, the helmet was eliminated and cranial pressure was produced by a woman's headband. Follow-up data, collected 1, 2, and 3 months after termination of systematic intervention, revealed near-zero levels of screaming and whining behaviors. A 9-month follow-up investigation involving the removal and subsequent reinstatement of the headband procedure supported the use of the headband for the maintenance of behavioral gains.


Subject(s)
Behavior Therapy/methods , Disabled Persons/psychology , Intellectual Disability/therapy , Adolescent , Combined Modality Therapy , Crying , Female , Follow-Up Studies , Humans , Intellectual Disability/psychology , Language Development Disorders/psychology , Language Development Disorders/therapy , Reinforcement Schedule , Social Behavior
SELECTION OF CITATIONS
SEARCH DETAIL