Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cancer Gene Ther ; 18(12): 850-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21869824

ABSTRACT

Carcinoembryonic antigen (CEA) is a cancer vaccines' target. Several features of recombinant adeno-associated virus (rAAV) are attractive for vaccine applications. Combining other viral vector vaccines with Toll-like receptor (TLR) agonists enhances antitumor immunity. Wild-type and CEA transgenic (Tg) mice were immunized with rAAV-expressing CEA, the TLR9 agonist, oligodinucleotide (ODN)1826 and the TLR7 agonist, imiquimod. Mice were challenged with MC38 colon tumor cells and MC38 cells expressing CEA. rAAV-CEA immunization combined with ODN1826 or imiquimod enhanced CEA-specific T-helper 1 immunity and protected against tumor challenge in wild-type but not in CEA-Tg mice. In contrast, immunization with rAAV-CEA in CEA-Tg mice could abrogate the antitumor effects of ODN1826 and promote tumor growth. Compared to wild-type, CEA-Tg mice were characterized by a greater myeloid suppressor cell and T-helper 2 response to TLR agonists and to syngeneic tumors. Depleting PDCA1(+) plasmacytoid dendritic cells and Gr1(+) myeloid cells increased anti-CEA immune responses in CEA-Tg mice to rAAV-CEA-ODN1826 immunization, whereas depleting CD25(+) T cells did not. There are differences in the response of wild-type and CEA-Tg mice to rAAV-CEA, TLR agonists and syngeneic tumor. In CEA-Tg mice, tumor growth can be promoted with rAAV-CEA and TLR agonists. Dendritic and myeloid cells play a regulatory role.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/therapy , Membrane Glycoproteins/agonists , Toll-Like Receptor 7/agonists , Toll-Like Receptor 9/agonists , Aminoquinolines/pharmacology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/genetics , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/immunology , Dependovirus , Female , Gene Expression Regulation , Humans , Imiquimod , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodeoxyribonucleotides/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
2.
Gene Ther ; 13(1): 29-39, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16136165

ABSTRACT

The potential of adeno-associated virus (AAV)-based vectors in human gene therapy is being explored for several diseases. Although sustained transgene expression and low vector-associated cellular immunity are attractive features of recombinant (r) AAV, the wider application of rAAV vectors encapsidated in serotype 2 capsid is hampered by poor transduction efficiency in many target tissues. These include ex vivo-generated dendritic cells (DC), which have demonstrated promising immunotherapeutic activity. We report here that efficient transduction of mouse bone marrow-derived DC can be achieved with self-complementary (sc) rAAV encapsidated in serotype 6 capsid. Sequential exposure of DC precursor cultures to IL-4 and GM-CSF with sc rAAV6 encoding the human tumor antigen, carcinoembryonic antigen (CEA), for 7 days followed by activation with CpG oligodeoxynucleotides (ODN) and anti-mouse CD40 antibody resulted in highly efficient transduction of DC. DC surface markers as determined by flow cytometry analysis of sc rAAV6-transduced DC were comparable to nontransduced DC. Efficiency of vector transduction and transgene expression were confirmed by immunostaining and real-time PCR. Microarray analysis of RNA from CpG ODN and CD40 antibody stimulated sc AAV6-transduced DC revealed upregulation of transcription factors and cytokines involved in immune activation and downregulation of inhibitory factors, suggesting a possible role of transcriptional activation in the observed effect. The adoptive transfer into syngeneic mice of the ex vivo-transduced and activated DC resulted in the development of CEA-specific antibody and T-helper 1-associated immune responses. Immunized mice also developed antibody to AAV6 capsid protein, which did not crossreact with AAV2 capsid protein. These studies demonstrate the potential utility of sc rAAV serotype 6-based vectors in transduction of DC for genetic vaccination approaches.


Subject(s)
Adoptive Transfer/methods , Dendritic Cells/immunology , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Animals , Antibodies, Viral/blood , Antibody Specificity , Antigen Presentation , Bone Marrow Cells/immunology , CD40 Antigens/genetics , Carcinoembryonic Antigen/genetics , Cell Line , CpG Islands , Dependovirus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Humans , Immunohistochemistry/methods , Interferon-gamma/immunology , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
3.
Gene Ther ; 9(17): 1189-93, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12170383

ABSTRACT

A promising approach to immunotherapy involves the loading of dendritic cells (DCs) with genetic material to facilitate sustained expression of a relevant antigen in this population of potent antigen presenting cells (APC). Viral vectors such as adenovirus (Ad) have been used for this purpose. Existing methods for DC infection are limited by lack of specificity and a requirement for DC exposure to high viral doses. Targeting of Ad to DCs with bispecific antibodies has significantly augmented levels of transgene expression. Genetic fusion of the extracellular portion of coxsackievirus-adenovirus receptor (CAR) to cell-specific ligands has also proved successful in targeting Ad to cells of interest. We report here the production and primary characterization of a new fusion protein comprising the ecto-domain of CAR connected to a single chain antibody (scFv) G28-5 against human CD40 present on the surface of DCs. We demonstrate that the fusion protein (CAR/G28) specifically interacts with both recombinant Ad fiber knob and the ecto-domain of human CD40 in a binding assay (ELISA). Finally, we show that the CAR/G28 fusion protein promotes highly efficient transduction of DCs of both rhesus monkey and human origin.


Subject(s)
Adenoviridae , CD40 Antigens/genetics , Dendritic Cells/immunology , Enterovirus , Genetic Therapy/methods , Immunoglobulin Fragments/genetics , Receptors, Virus/genetics , Dendritic Cells/virology , Genetic Engineering , Genetic Vectors/administration & dosage , Humans , Immunotherapy/methods , Recombinant Fusion Proteins/administration & dosage , Transduction, Genetic/methods
4.
Cancer ; 89(12): 2646-54, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11135227

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are potent initiators of immune responses, and the infiltration of DCs into tumors may confer an improved prognosis. Whether the injection of DCs directly into tumors can mediate biologic activity was examined. METHODS: Patients with metastatic dermal or subcutaneous tumors received granulocyte-macrophage-colony stimulating factor to increase the numbers of peripheral blood monocyte precursors. DCs were then generated from monocytes obtained by phlebotomy with granulocyte-macrophage-colony stimulating factor and interleukin-4 in autologous plasma. Tumors were injected at multiple sites with 30 million autologous DCs per tumor. RESULTS: Seven patients with melanoma and three patients with breast carcinoma were treated. Injections were well tolerated. Regression of the injected tumors, beginning as early as 4 days after injection, was observed in four patients with melanoma and in two patients with breast carcinoma. Biopsies of regressing lesions showed lymphocyte infiltration associated with DCs and necrosis. Neutrophils and macrophages were not evident. Lymphocytes expanded from the regressing tumors proliferated in response to heat shock proteins, HSP70 and gp96, derived from autologous tumor. The DCs injected produced interferon-alpha and expressed Fas ligand mRNA but did not exhibit cytolytic activity in vitro. Expression of the costimulatory molecule, B7-2 (CD86), decreased on DCs after intratumoral injection. CONCLUSIONS: This pilot study demonstrates that DCs derived in vitro can exist viably after intratumoral injection and can mediate biologic activity in situ. Tumor-derived heat shock proteins may be involved in the antitumor activity observed.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , Adult , Aged , Antigens, CD/analysis , B7-2 Antigen , Cell Transplantation , Dendritic Cells/transplantation , Female , Flow Cytometry , HLA-DR Antigens/analysis , Heat-Shock Proteins/analysis , Humans , Immunohistochemistry , Injections, Intralesional , Integrin alphaXbeta2/analysis , Male , Membrane Glycoproteins/analysis , Middle Aged , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , Pilot Projects , Treatment Outcome
5.
Cancer Immunol Immunother ; 48(9): 525-32, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10602890

ABSTRACT

PURPOSE: Mucins have been implicated in tumor-associated immunosuppression. The possibility that colon cancer mucin (CCM) may modulate T-helper 1 (TH1) activity was evaluated by investigating its effect on the production of interleukin-2 (IL-2) by CD4+ cells, a process that requires antigen-specific and costimulatory signals. METHODS: CCM was purified from human colorectal cancer cells by gel-exclusion fast-pressure liquid chromatography. Cytokine production of purified CD4+ cells was evaluated at the protein and gene level in the presence of a phorbol ester or an anti-CD3 monoclonal antibody (mAb) plus mAb against the CD28 costimulatory receptor to mimic two-signal activation. RESULTS: Soluble CCM, which contains mucins MUC2 as well as MUC1, inhibited IL-2 mRNA expression and secretion of CD4+ stimulated with a phorbol ester or an anti-CD3 mAb plus anti-CD28 mAb. Pretreatment of CD4+ cells with anti-CD28 mAb abrogated the suppressive effects of CCM on IL-2 production, and flow cytometry showed decreased binding of anti-CD28 mAb to its receptor in the presence of mucin. In addition, Ca2+ mobilization after T cell receptor cross-linking with anti-CD3 mAb was maintained in the presence of CCM. Although interferon gamma production was also diminished, CCM did not induce a general inhibition of cytokine production, nor did it decrease cell viability. Macrophage inflammatory protein 1alpha production was up-regulated; the production of IL-10 and transforming growth factor beta was unchanged. CONCLUSIONS: The results indicate that CCM can alter TH1 activity and suggest that the modulation of costimulatory interactions is involved. They provide another mechanism of immunosuppression mediated by these highly expressed tumor products.


Subject(s)
Adenocarcinoma, Mucinous/chemistry , CD28 Antigens/immunology , Colorectal Neoplasms/chemistry , Gene Expression Regulation/drug effects , Lymphokines/biosynthesis , Mucins/pharmacology , Neoplasm Proteins/pharmacology , Th1 Cells/drug effects , Antibodies, Monoclonal/pharmacology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2/biosynthesis , Interleukin-2/genetics , Interleukin-2/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Mucin-1/isolation & purification , Mucin-1/pharmacology , Mucin-2 , Mucins/isolation & purification , Muromonab-CD3/pharmacology , Neoplasm Proteins/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Solubility , Tetradecanoylphorbol Acetate/pharmacology , Th1 Cells/metabolism
6.
Cancer ; 86(1): 22-30, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10391559

ABSTRACT

BACKGROUND: The authors showed previously that radiolabeled monoclonal antibody (MoAb) and a hand-held, gamma-detecting probe can be used to localize tumor-reactive lymph nodes in vivo. The authors examined the feasibility, safety, and biologic effects of cellular immunotherapy using autologous cells expanded from these lymph nodes in patients with metastatic colorectal carcinoma. METHODS: Tumor-reactive lymph nodes containing radiolabeled MoAb were localized and excised from 32 patients with metastatic, unresectable colorectal carcinoma at laparotomy. Lymph nodes were dissociated, and cells were cultured ex vivo for 10-14 days. Patients received a single infusion of autologous, expanded cells with no systemic interleukin (IL)-2. RESULTS: A mean of 1.6 x 10(10) expanded autologous lymph node cells were infused with toxicity limited to occasional fevers or chills. The cells infused predominately were activated CD3+ T-cells that expressed genes for IL-4, IL-5, interferon-gamma, and granulocyte-macrophage colony stimulating factor (GM-CSF) by using reverse transcriptase-polymerase chain reaction. Indium-111 labeled cells were observed to traffic initially to the lungs, bone marrow, liver, and spleen. One patient on study achieved a partial response (>80% reduction), and mixed or minor responses were noted in 4 other patients. The responding patient's cell characteristics were notable for high levels of GM-CSF and IL-4 secretion on restimulation with immobilized anti-CD3 in vitro, and biopsies of the tumor were characterized by macrophage infiltration. The median survival of the cell-treated group compared favorably with a similar group of patients who underwent radioimmunoguided surgery without cell treatment (12.5 months vs. 5.8 months) CONCLUSIONS: The infusion of cells expanded from tumor-reactive lymph nodes localized with radiolabeled MoAb in vivo is reproducible and safe and has biologic activity, even in the absence of systemic IL-2 infusion. This approach represents a novel application of MoAb technology, in that MoAbs are used not to diagnose or treat disease directly but rather to identify lymph node cells with therapeutic potential.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/therapy , Immunotherapy, Adoptive , Interleukin-2/therapeutic use , Lymph Nodes/cytology , Radioimmunotherapy , Adult , Aged , CD3 Complex , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Female , Humans , Interleukin-2/administration & dosage , Lymph Nodes/surgery , Male , Middle Aged , Survival Analysis , Treatment Outcome
7.
J Infect Dis ; 179(1): 245-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9841848

ABSTRACT

A pilot study was undertaken in patients with human immunodeficiency virus type 1 (HIV-1) infection to examine the effects of infusing autologous lymph node lymphocytes that had been cultured ex vivo in conditions designed to maximize the specific secretion of HIV-1-suppressive factors, including beta chemokines. Ten patients with CD4 cell counts between 119 and 436/microliter on antiretroviral drugs received a single infusion of CD4 and CD8 lymph node lymphocytes. There were no serious acute or chronic adverse clinical effects. Increases in serum levels of macrophage inflammatory protein 1beta (MIP-1beta) and increases in the production of MIP-1beta by peripheral blood lymphocytes in response to HIV-1 env were observed. Increases in CD4 and CD8 cell counts and skin test reactivity to recall antigens and decreases in HIV-1 virus load were also observed. This cellular immunotherapy can modulate beta chemokine production in patients with advanced HIV-1 infection and may contribute immunorestorative and antiviral activities.


Subject(s)
Blood Transfusion, Autologous , Chemokines/biosynthesis , HIV Infections/immunology , HIV Infections/therapy , HIV-1 , Immunotherapy/methods , Blood Transfusion, Autologous/adverse effects , Blood Transfusion, Autologous/methods , CD4-CD8 Ratio , Cells, Cultured , Chemokine CCL4 , Chemokines/blood , Gene Products, env/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/isolation & purification , Humans , Immunity, Cellular , Immunotherapy/adverse effects , Lymph Nodes/immunology , Lymphocyte Transfusion/adverse effects , Lymphocyte Transfusion/methods , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/blood , Pilot Projects , Skin Tests , Time Factors
8.
AIDS Res Hum Retroviruses ; 14(8): 643-9, 1998 May 20.
Article in English | MEDLINE | ID: mdl-9618075

ABSTRACT

The chemokines RANTES, MIP-1alpha, and MIP-1beta have been identified as HIV-1-suppressive factors produced by CD8+ T cells. We examined the possibility that HIV-1-specific, chemokine-releasing T cells could be expanded from the lymph nodes of patients with advanced infection. Lymphocytes, separated from lymph nodes of patients with peripheral blood CD4 counts less than 500/microl obtained at diagnostic biopsies, were activated with anti-CD3 monoclonal antibody, and cultured in vitro for up to 12 days with IL-2. The phenotype, proliferative response, chemokine production, and anti-HIV-1 activity of the expanded cells was examined. Cells expanded 2.4- to 49-fold from patients with as few as 15 CD4+ cells/microl in their peripheral blood. Expanded cells were a mixture of CD8+CD45RO+ and CD4+CD45RO+ T cells. The CD8+ cells were also CD30+CDw60+CD11b-. When challenged with autologous B cell targets expressing HIV-1 Env protein, unseparated expanded cells, and purified CD8+ and CD4+ T cell subsets, proliferated and secreted MIP-1alpha and RANTES. Expanded cells were negative for HIV-1 by PCR and by culture. Culture supernatants inhibited the replication of HIV-1 in CD4+ cells in vitro. These studies indicate that HIV-1 can stimulate chemokine release by CD8+ and CD4+ cells expanded from infected lymph nodes, even from individuals with advanced infection. The numbers of chemokine-releasing T cells produced in these short-term cultures may be sufficient to be applied therapeutically as an autologous cellular therapy for HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokines/biosynthesis , HIV-1/immunology , Lymph Nodes/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Chemokines/genetics , Cytotoxicity, Immunologic , Gene Expression , HIV Infections/immunology , HIV-1/physiology , Humans , Immunophenotyping , Interleukin-2 , Leukocyte Common Antigens/analysis , Lymphocyte Activation , Muromonab-CD3 , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , RNA, Messenger/analysis , Virus Replication
9.
J Leukoc Biol ; 61(5): 600-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9129209

ABSTRACT

We compared dendritic cells (DC) derived from CD34+ hematopoietic progenitor cells with tumor necrosis factor alpha and granulocyte-macrophage colony-stimulating factor (GM-CSF) to DC derived from monocytes/macrophages with interleukin-4 (IL-4) and GM-CSF. Monocyte/macrophage-derived DC demonstrated higher levels of CD1a, lower levels of CD14, greater stimulatory activity in mixed lymphocyte reactions, and greater capacity to present soluble protein antigen than CD34+ cell-derived DC. Lymphocytes stimulated with antigen-pulsed, monocyte/macrophage-derived DC produced more IL-10 than those stimulated with antigen-pulsed, CD34+-derived DC. Whereas CD1a+ DC could be derived from CD34+ cells in serum-free- and human-sera-containing cultures, the derivation of CD1a+ DC from monocytes/macrophages required the presence of fetal calf serum. The spectrum of cytokine mRNA expression, the presentation of peptide antigen, and the sensitivity to human immunodeficiency virus-1 infection of CD34(+)- and monocyte/macrophage-derived DC were comparable. Although cells derived by both methods are potent antigen-presenting cells, there are differences between DC derived in vitro from hematopoietic progenitors and from monocytes/macrophages that may influence their in vivo activity.


Subject(s)
Dendritic Cells/physiology , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Monocytes/cytology , Amino Acid Sequence , Antigen Presentation , Antigens, CD34/metabolism , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/metabolism , HIV Infections/pathology , HIV-1 , Humans , Lymphocyte Culture Test, Mixed , Molecular Sequence Data , Phenotype , RNA, Messenger/metabolism
10.
Clin Cancer Res ; 3(12 Pt 1): 2355-62, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9815634

ABSTRACT

The clinical and immunological effects of a vaccine consisting of CTP37, a synthetic peptide corresponding to the COOH-terminal peptide (CTP) of beta-human chorionic gonadotropin (beta-hCG) conjugated to diphtheria toxoid, combined with CRL 1005, a novel synthetic nonionic block copolymer adjuvant, were examined. Twenty-one patients with metastatic, nontrophoblastic cancers received up to four immunizations by i.m. injection of a fixed dose of CTP37 and escalating doses of CRL 1005. Doses of CRL 1005 adjuvant as high as 75 mg were administered with 1 mg of CTP37 without evidence of significant local or systemic toxicity. Immunizations resulted in the production of IgG antibody to beta-hCG. CRL 1005 doses of 3-25 mg appeared to be optimal for antibody induction. Immunizations also resulted in increases in the cellular response of peripheral blood mononuclear cells (PBMCs) to the unconjugated CTP, hCG, and diphtheria toxoid. Responding PBMCs specifically secreted the TH1-associated cytokines IFN-gamma and interleukin (IL)-2 as well as the TH2-associated IL-5 and IL-10. Increased expression of IFN gamma and IL-5 mRNAs by PBMCs 4 h after immunization was also observed. CRL 1005 administered with CTP37 in aqueous solution is well tolerated. The CTP37-CRL 1005 subunit vaccine has the capacity to stimulate potentially beneficial humoral and cellular immune responses in patients with advanced cancer.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Chorionic Gonadotropin, beta Subunit, Human/therapeutic use , Neoplasms/therapy , Pentosan Sulfuric Polyester/pharmacokinetics , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Adult , Aged , Antibody Formation , Chorionic Gonadotropin , Chorionic Gonadotropin, beta Subunit, Human/administration & dosage , Chorionic Gonadotropin, beta Subunit, Human/adverse effects , Diphtheria Toxoid/administration & dosage , Diphtheria Toxoid/therapeutic use , Female , Humans , Hypersensitivity, Delayed , Immunity, Cellular , Immunoglobulin G/blood , Interferon-gamma/genetics , Interleukin-2/genetics , Interleukin-5/genetics , Male , Middle Aged , Neoplasms/immunology , Pentosan Sulfuric Polyester/administration & dosage , Polymers , Solutions , Th1 Cells/immunology
11.
Cancer Biother ; 10(2): 115-23, 1995.
Article in English | MEDLINE | ID: mdl-7663570

ABSTRACT

The ability to identify and expand effector cells with reactivity against tumor-associated antigens (TAA) is critical for effective adoptive cellular therapy. The purpose of this study was to assess lymph node lymphocytes sensitized in vivo to the shed TAA TAG-72 as a potential source of cells for adoptive cellular therapy. Lymph nodes containing microscopic tumor and/or shed TAG-72+ mucin were localized using radiolabeled CC49 monoclonal antibody and a gamma detector at the time of exploratory colorectal surgery. Lymph nodes containing microscopic tumor and shed mucin exhibited approximately 40-fold expansion in short-term (< 21 days) cultures with either IL-2 or IL-1 plus IL-2; the combination of IL-2/anti-CD3 monoclonal antibody (mAb) resulted in significantly higher expansion. Cultures generated with IL-2 alone favored the expansion of CD8+ and CD56+ cells, whereas addition of IL-1 or anti-CD3 mAb to IL-2 promoted outgrowth of CD4+ T-cells. The IL-2/anti-CD3 expanded cells exhibited low levels of cytolytic activity in vitro against autologous and allogeneic colon tumor targets. However, CD4+ cells expanded in IL-2/anti-CD3 retained the ability to proliferate in response to TAG-72 mucin-expressing autologous tumor as well as bovine submaxillary mucin (BSM) a soluble TAG-72+ mucin. In addition, CD4+ cells expressed mRNA for IL-2, IL-4, tumor necrosis factor-beta and IFNg, and retained the ability to secrete IL-2 after expansion. Thus, noncytolytic, cytokine-secreting, mucin-reactive T- cells can be expanded from lymph nodes of patients with colorectal cancer.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antigens, Neoplasm/analysis , Colorectal Neoplasms/immunology , Glycoproteins/analysis , Lymph Nodes/immunology , Mucins/analysis , T-Lymphocytes, Helper-Inducer/immunology , CD4-CD8 Ratio , Colorectal Neoplasms/therapy , Cytotoxicity, Immunologic , Humans , Immunotherapy, Adoptive , Interleukin-2/biosynthesis , Phenotype , RNA, Messenger/analysis , Tumor Cells, Cultured
12.
Immunopharmacology ; 28(1): 39-45, 1994.
Article in English | MEDLINE | ID: mdl-7928301

ABSTRACT

Monocytes macrophages have negative regulatory effects on many immunologic responses. Depletion of monocytes from peripheral blood using the lysosomotropic agent, L-phenylalanine methyl ester (PME), has been shown to improve lymphokine activated killer (LAK) cell expansion in vitro. A pilot study of the adoptive transfer of LAK cells expanded with PME was performed in patients with metastatic renal cell carcinoma. Patients received interleukin-2 (IL-2) by continuous infusion for 5 days. Leukopheresis was performed daily for 4 days during the second week. Cells obtained from 8 patients were depleted of monocytes using PME in an one-step procedure; < or = 3% of the remaining cells were monocytes. All cells were expanded for 10 days in air-porous plastic bags with IL-2. Cells expanded 2.7-fold when depleted with PME and 1.7-fold when not depleted (P = 0.02). Expanded cells were administered together with IL-2. Patients received up to 60 x 10(10) PME-depleted cells (mean = 26 x 10(10)) with LAK activity (% lysis) of 60 +/- 12%. Lymphocyte phenotype and cytolytic activity were not modulated by PME-depletion, and clinical toxicities and systemic immunologic effects observed in patients receiving PME-depleted cells were similar to that of 5 patients receiving cells not expanded with PME. Thus, the use of PME to deplete monocytes ex vivo can result in the yield of large number of effectors that retain immunologic activity for potential clinical use. The process is convenient, efficient, and does not add clinical toxicity.


Subject(s)
Carcinoma, Renal Cell/therapy , Immunotherapy, Adoptive/methods , Kidney Neoplasms/therapy , Killer Cells, Lymphokine-Activated/immunology , Phenylalanine/analogs & derivatives , Carcinoma, Renal Cell/secondary , Cytotoxicity Tests, Immunologic , Humans , Immunophenotyping , Killer Cells, Lymphokine-Activated/transplantation , Monocytes/drug effects , Phenylalanine/pharmacology , Pilot Projects
13.
Circulation ; 89(2): 712-7, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8313559

ABSTRACT

BACKGROUND: Intravascular ultrasound (IVUS) of arteries is limited by the inability of current instruments to visualize beyond the catheter tip. We have developed a prototype 4-mm-diameter forward-viewing IVUS catheter (Cardiovascular Imaging Systems, Sunnyvale, Calif) that has the ability to provide B-mode cross-sectional ultrasound data for a distance of up to 2 cm distal to the catheter tip. METHODS AND RESULTS: To study the utility of this device, a 20-MHz forward-viewing IVUS catheter was used to examine 13 arterial segments (5 human femoral arteries, 1 human carotid artery, 7 canine arteries) in vitro and 1 phantom. After imaging, all data were compared with histology (Histo). In all cases, the IVUS catheter provided forward-viewing images corresponding to the arterial geometry and demonstrated vascular landmarks and atherosclerotic lesions. There was a good correlation between Histo-determined luminal diameters (LD) and IVUS-determined diameters for a distance of 14 mm ahead of the catheter tip: IVUS LD = 1.0 Histo LD + 1.3 (r = .87). CONCLUSIONS: These preliminary data suggest that a forward-viewing IVUS catheter is feasible, accurate, and useful for evaluation of arterial geometry distal to the catheter tip.


Subject(s)
Arteries/diagnostic imaging , Catheterization/instrumentation , Ultrasonography/methods , Angiography , Animals , Aorta/diagnostic imaging , Carotid Arteries/diagnostic imaging , Dogs , Evaluation Studies as Topic , Humans , In Vitro Techniques
14.
Circulation ; 89(2): 718-23, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8313560

ABSTRACT

BACKGROUND: Current intravascular ultrasound (IVUS) catheters provide transverse imaging at the level of the ultrasound transducer. This limits imaging to large-diameter segments without critical atherosclerotic narrowings. We have developed a prototype 20-MHz forward-viewing IVUS catheter that provides two-dimensional sector imaging distal to the catheter tip. A present limitation of this technique is that the catheter must be manually rotated to obtain multiple longitudinal views required to integrate the segment into a three-dimensional matrix. To overcome this, we have developed an algorithm that reconstructs these multiple two-dimensional forward-viewing IVUS images into a three-dimensional matrix for more complete depiction of the segment distal to the ultrasound catheter. This algorithm allows display and multidimensional slicing of the three-dimensional reconstruction. METHODS AND RESULTS. To test our algorithms, five arterial segments (three canine aortas, two human femoral arteries) were evaluated in vitro. In each segment, 36 forward-viewing longitudinal slices were collected, digitized, processed, and reoriented to produce a three-dimensional reconstruction (3DR) matrix. The matrix data were sliced into parallel transverse sections and compared with morphometric interpretation of histological sections (Histo). As a result, image data could be reconstructed for a distance of 2.0 cm ahead of the catheter. 3DR easily demonstrated wall and luminal morphology and provided transverse IVUS images comparable to the histological specimens. A good correlation was noted between Histo- and 3DR-determined luminal diameters (LD) and luminal areas: 3DR LD = 1.4 Histo LD-0.4, r = .86; 3DR LD = 0.7 +/- 0.20 cm (mean +/- SD); and Histo LD = 0.7 +/- 0.13 cm. CONCLUSIONS: These preliminary data demonstrate the feasibility of 3DR of forward-viewing IVUS data. This method allows rapid, detailed analysis of diseased arterial segments previously unavailable with standard IVUS and may permit better targeting of interventional techniques.


Subject(s)
Arteries/diagnostic imaging , Catheterization/instrumentation , Data Display , Image Processing, Computer-Assisted , Ultrasonography/methods , Animals , Arteriosclerosis/diagnostic imaging , Dogs , Humans , In Vitro Techniques
15.
Cancer ; 73(3): 580-9, 1994 Feb 01.
Article in English | MEDLINE | ID: mdl-8299079

ABSTRACT

BACKGROUND: Lymph node lymphocytes vary in their responsiveness to tumor. A technique has been developed that uses radiolabeled monoclonal antibody (MoAb) against the tumor-associated mucin, TAG-72, and a gamma-detecting probe by which lymph nodes containing microscopic tumor and/or shed TAG-72 can be identified in vivo. The immunologic characteristics of these lymph nodes were examined. METHODS: Patients with colon cancer received 125I-labeled MoAb CC49 by intravenous injection preoperatively. During laparotomy lymph nodes that appeared normal on inspection and palpation but which contained radiolabeled MoAb were identified using a hand-held gamma-detecting probe. These lymph nodes and other lymph node and tumor specimens were resected for analysis. RESULTS: Lymph nodes identified by the probe were found by immunohistochemical studies to contain microscopic tumor and/or shed antigen associated with germinal centers. They were characterized by greater CD4+:CD8+ ratios, rates of expansion, and cytolytic activity compared with lymphocytes from lymph nodes with macroscopic tumor, noninvolved lymph nodes, and tumors. All lymph node lymphocytes identified by the probe demonstrated significant proliferative responses to autologous tumor and, in contrast to lymphocytes from noninvolved lymph nodes, significant proliferative responses to allogeneic TAG-72+ tumor cells and to soluble TAG-72+ mucin. CONCLUSIONS: By locating lymph nodes with microscopic tumor and/or shed antigen, the use of radiolabeled MoAb in vivo can be used to reproducibly identify tumor-reactive lymph node lymphocytes. This technique may be useful in identifying cells for use in adoptive immunotherapy programs and in studying the regulation of immune responses in vivo.


Subject(s)
Colonic Neoplasms/immunology , Lymph Nodes/pathology , Lymphocytes/immunology , Antibodies, Monoclonal , Antigens, Neoplasm/analysis , Antigens, Neoplasm/immunology , CD4-CD8 Ratio , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Glycoproteins/analysis , Glycoproteins/immunology , Humans , Immunotherapy, Adoptive , Iodine Radioisotopes , Lymph Nodes/immunology , Lymphocytes/pathology , Tumor Cells, Cultured
16.
Int J Oncol ; 5(6): 1447-53, 1994 Dec.
Article in English | MEDLINE | ID: mdl-21559734

ABSTRACT

We treated 23 patients with non-trophoblastic cancers with escalating doses of a synthetic vaccine consisting of the carboxy-terminal peptide of beta human chorionic gonadotropin conjugated to diphtheria toroid (CTP37), a muramyl dipeptide as an adjuvant, and squalene/mannide monooleate as a vehicle. Toxicity consisted of pain and sterile abscess formation at the injection site and of constitutional symptoms. Diphtheria toxoid hypersensitivity developed in one patient. Immunizations elicited anti-beta hCG IgG antibody which persisted for more than 10 months. Disappearance of circulating hCG present pre-immunization and tumor regressions were observed. Active specific immunotherapy with CTP37 vaccine is well-tolerated and has biological activity in patients with cancer.

18.
Mol Biother ; 4(2): 77-82, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1515098

ABSTRACT

To determine if intensive chemotherapy consisting of cyclophosphamide (C), etoposide (E), and cisplatin (P) (CEP) may be usefully combined with recombinant human interleukin-2 (rhIL-2), we examined a murine tumor model designed to approximate a common clinical situation: macroscopic, drug-resistant cancer. Using C57BL/6 mice with extensive tumor burden 10 days after intravenous B16 melanoma cell injection, we observed (1) C, E, and P synergize to enhance survival but do not cure mice at the highest tolerable dose (C = 200 mg/kg, E = 60 mg/kg, and P = 3 mg/kg); (2) rhIL-2 at 3 x 10(5) U (subcutaneously) daily for 4 days administered 10-18 days after B16 injection significantly improves survival; (3) CEP plus rhIL-2 is more effective than CEP alone only when rhIL-2 is administered before CEP; (4) CEP suppresses IL-2-induced lymphokine-activated killer cell activity in the spleen; and (5) rhIL-2 protects mice incompletely from the immunologic and hematologic suppression of CEP. Our results suggest that intensive chemotherapy combined with rhIL-2 may be beneficial. The success of any such combination may be schedule dependent.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Interleukin-2/administration & dosage , Melanoma, Experimental/drug therapy , Melanoma, Experimental/therapy , Animals , Cisplatin/administration & dosage , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Drug Synergism , Etoposide/administration & dosage , Female , Immunotherapy , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/immunology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL
19.
Mol Biother ; 3(1): 22-5, 1991 Mar.
Article in English | MEDLINE | ID: mdl-2069757

ABSTRACT

Depleting monocytes from human peripheral blood mononuclear cells (PBMC) enhances the in vitro activation of lymphokine-activated killer (LAK) cells. To determine if monocytes also altered LAK-cell expansion, we evaluated two methods of depleting monocytes from PBMC: nylon wool adherence (NWA) and phenylalanine methyl ester (PME) treatment. Both methods of depleting monocytes enhanced interleukin-2 (IL-2) driven, LAK-cell expansion; LAK expansion, however, was significantly greater after depletion with NWA than after PME. LAK cytotoxicity after NWA and PME depletion was equivalent. The degree of monocyte depletion, determined by evaluating morphology and the number of Leu-M3 (CD14) positive cells, and the proliferation of Leu 19 (CD56), OKT-3 (CD3), Leu2 (CD8), and Leu 3a (CD4) positive cells was also equivalent. Exposure of IL-2 activated cells to PME did not alter their cytotoxic activity. However, sequential treatment of PBMC with NWA, then PME, or with PME and then NWA, resulted in reduced expansion. This reduction in expansion was similar to PBMC treated with PME alone. Exposure of PME-depleted cells to nylon wool or to supernatants obtained from cells adherent to nylon wool further decreased LAK expansion relative to cells treated with NWA alone. We conclude that even at relatively low cell density, human monocytes markedly inhibit LAK-cell expansion in IL-2 driven PBMC cultures. Further, depletion of monocytes by NWA adherence is more effective than by treatment with PME, possibly due to subtle cellular damage induced by this latter treatment. These findings have implication for the in vitro and in vivo generation of LAK-cells by IL-2.


Subject(s)
Killer Cells, Lymphokine-Activated/immunology , Monocytes/immunology , Cell Separation/methods , Cytotoxicity, Immunologic , Humans , In Vitro Techniques , Interleukin-2/pharmacology , Lymphocyte Activation , Phenylalanine/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL