GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 10, No. 14 ( 2004-07-15), p. 4699-4708
    Abstract: Purpose: Dendritic cells (DCs) are potent antigen-presenting cells that are uniquely capable of inducing tumor-specific immune responses. We have conducted a Phase I trial in which patients with metastatic breast and renal cancer were treated with a vaccine prepared by fusing autologous tumor and DCs. Experimental Design: Accessible tumor tissue was disrupted into single cell suspensions. Autologous DCs were prepared from adherent peripheral blood mononuclear cells that were obtained by leukapheresis and cultured in granulocyte macrophage colony-stimulating factor, interleukin 4, and autologous plasma. Tumor cells and DCs were cocultured in the presence of polyethylene glycol to generate the fusions. Fusion cells were quantified by determining the percentage of cells that coexpress tumor and DC markers. Patients were vaccinated with fusion cells at 3-week intervals and assessed weekly for toxicity, and tumor response was assessed at 1, 3, and 6 months after completion of vaccination. Results: The vaccine was generated for 32 patients. Twenty-three patients were vaccinated with 1 × 105 to 4 × 106 fusion cells. Fusion cells coexpressed tumor and DC antigens and stimulated allogeneic T-cell proliferation. There was no significant treatment-related toxicity and no clinical evidence of autoimmunity. In a subset of patients, vaccination resulted in an increased percentage of CD4 and CD8+ T cells expressing intracellular IFN-γ in response to in vitro exposure to tumor lysate. Two patients with breast cancer exhibited disease regressions, including a near complete response of a large chest wall mass. Five patients with renal carcinoma and one patient with breast cancer had disease stabilization. Conclusions: Our findings demonstrate that fusion cell vaccination of patients with metastatic breast and renal cancer is a feasible, nontoxic approach associated with the induction of immunological and clinical antitumor responses.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1148-1148
    Abstract: Abstract 1148 Poster Board I-170 Graft versus host disease (GVHD) remains a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HCT). In vivo quantitative T-cell depletion using CAMPATH-1h (anti-CD52) has been explored in an effort to prevent acute GVHD. More recently, a regimen consisting of total lymphoid irradiation and anti-thymocyte globulin (ATG) has been shown to polarize T cells towards an inhibitory phenotype potentially reducing the associated risk for GVHD. However, these strategies may be associated with impaired post-transplant immune reconstitution, increased risk of tumor relapse and opportunistic infection. In this study we examined the pattern of cellular immune recovery following T cell depletion with CAMPATH-1h and compared results with an initial cohort of patients undergoing reduced intensity conditioning with TLI and ATG. Immunologic analyses were performed on twenty patients undergoing reduced intensity conditioning in conjunction with low dose CAMPATH -1h (50 mg) and an initial cohort of 5 patients treated with TLI/ATG. Conditioning with CAMPATH-1h resulted in the significant depletion of CD3, CD4, and CD8 T cells in the early post-transplant period and persistence of CD4 T cell depletion ( 〈 200 cells /uL) for more than 6 months. Following TLI/ATG, persistent depletion of CD4+ T cells was also observed but no significant decrease in CD8 T cells was seen. A two-fold increase in circulating CD56+ NK cells, 21.8 to 41.6% (p=0.004), was seen following TLI-ATG, which was not noted following Campath conditioning. CAMPATH-1h conditioning was associated with a significant decrease in mean CD45RO+ memory T cells in the early post-transplant period (27.2 to 5.7% of the total population of nonadherent peripheral blood mononuclear cells, p=0.034). Relative percentages of naïve T cells (CD45RA+), central memory (CD45RO+CD62L+CCR7+) (CM), and effector memory (CD45RO+CD62L-CCR7-) (EM) T cells remained stable in the pre- and post-transplantation period. The CM:EM was 0.6 pre-transplant and at day 60, respectively. In contrast, T cell recovery in early post-transplant following the TLI/ATG regimen was associated with no reduction in CD45RO+ memory T cells. A significant rise in the relative percentages of naïve T cells from 39 to 61.3% (p=0.04), CM cells from 12 to 32.8% (p=0.05), a corresponding fall in EM cells from 57.9 to 32.5% (p=0.10), and a significant change in the CM:EM levels (0.2 pre-transplant, 1.0 day 60 post-transplant) was noted after TLI/ATG. The mean percentage of regulatory T cells as defined by the percentage of CD4+/CD25+ cells that express FoxP3 rose in the early post-transplant period following both regimens (8 to 20.7% at Day 30, p=0.003 in the CAMPATH group; 5.6 to 16.9% at Day 30, p=0.03 in the ATG/TLI group). Functional analyses demonstrated that the T cell proliferative response to the mitogen, Phytohemagglutinin (PHA), was profoundly depressed following CAMPATH-1h with mean SI decreasing from 34 pre-transplant to 1.4 at Day 30. In contrast, treatment with TLI/ATG resulted in no significant change in T cell proliferation in response to PHA with SI only decreasing from 45 pre-transplant to 36 at Day 30. Assessment of T cell polarization following stimulation with PHA or phorbol-ester (PMA)/ionomycin, recipient derived dendritic cells (DCs) or third party DCs demonstrated a rise of CD8+ T cells expressing, IL-4 and IL-10 consistent with a suppressor phenotype. Minimal T cell proliferation was observed following stimulation with patient derived DCs, which is consistent with suppression of the expansion of alloreactive T cells. In summary, both CAMPATH and TLI/ATG result in CD4+ T cell depletion but TLI/ATG resulted in relative preservation of CD8+ T cells, persistence of memory cells, relative preservation of central memory as compared to memory effector cells and intact response to mitogens. TLI/ATG therapy was also associated with the polarization of CD8+ T cells towards a Tc2 phenotype and lack of proliferation in response to recipient derived DCs. As such, TLI/ATG appears to be associated with more modest level of functional T cell depletion characterized by Tc2 polarization and suppression of host/donor alloreactivity. Disclosures Spitzer: Genzyme: Consultancy. Avigan:Genzyme: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 5209-5209
    Abstract: In pre-clinical models, vaccination with attenuated vaccinia and fowl pox virus expressing tumor antigens and costimulatory molecules (CD54/ICAM-1; CD58/LFA-1 and CD80/B7.1-TRICOM) potently stimulates anti-tumor immune responses. However, vaccine efficacy may be limited by intrinsic deficiencies of native dendritic cells (DC) populations in patients with malignancy that are required to process and present the virally introduced antigens. An alternative strategy involves the transduction of ex vivo generated activated DCs. We have examined the capacity of DCs transduced with a fowl pox vector expressing MUC-1, CEA, and TriCOM (PANVAC-F) to elicit antigen specific responses and expand activated as compared to regulatory T cell populations. Partially mature DCs were generated from leukopak preparations obtained from normal volunteers by culturing adherent peripheral blood mononuclear cells for 5 days with GM-CSF and IL-4. DCs were transduced with PANVAC-F vector and matured with either TNFa or the combination of PG-E2, TNFa, IL-6 and IL-1b. In 5 serial studies, transduction with PANVAC-F resulted in mean MUC1 expression in 64.6% (SEM: ±1.6) of cells with a mean fluorescent intensity (MFI) of 249.8 (SEM: ±45.7). In addition, transduced DCs also demonstrated high levels of expression of class II (99%; MFI:250), CD54 (98.4%;MFI:516), CD58 (98.8%;MFI:155) and CD80 (78.2%; MFI:115). Of note, transduced DCs demonstrated higher levels of the maturation marker CD83 (40.3%; SEM: ±1.6, n=3) as compared to untransduced DCs 19.7% (SEM: ±2.9) (p=0.02) suggesting that transduction enhanced DC maturation and activation. Transduced DCs matured with PGE2, TNFa, IL-6 and IL-1b as compared to TNFa alone demonstrated higher levels of CD83 and CCR7 and a more stable phenotype following withdrawal of cytokine support. To assess the ability of PANVAC-F to stimulate tumor antigen specific responses, the presence of T cells binding the MUC-1 specific tetramer was quantified following stimulation of autologous T cells derived from HLA*0201 healthy donors. An increase in CD8+ and MUC1+ cells were observed (8.3%) with stimulation with PANVAC-F transduced DCs as compared to untransduced DCs (2.5%). The capacity of DCs transduced with PANVAC-F to stimulate interferon gamma producing activated T cells, as compared to CD4+/CD25+/Foxp3+ regulatory T cell populations is being assessed. In summary, PANVAC-F transduced DCs stimulate expansion of antigen specific T cell populations suggesting their potential role as tumor vaccines for MUC-1/CEA expressing tumors. We are initiating a trial for patients with ovarian carcinoma in early (marker only) relapse in which patients will be randomized to undergo serial vaccination with PANVAC-V/PANVAC-F or DCs transduced with PANVAC-F. The capacity to generate anti-tumor immunity in vivo will be assessed as a primary endpoint.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 578-578
    Abstract: Autologous transplantation results in the transient reversal of tumor mediated tolerance due to the reduction in disease bulk, the depletion of regulatory T cells, and in the increased presence of tumor reactive lymphocytes during the period of lymphopoietic reconstitution. As a result, cancer vaccines are being explored as a means of targeting residual myeloma cells following stem cell transplant. We have developed a cancer vaccine in which patient derived tumor cells are fused with autologous dendritic cells (DCs). In this way multiple tumor antigens are presented in the context of DC mediated costimulation. We are conducting a study in which patients with multiple myeloma (MM) undergo stem cell transplantation followed by vaccination with 3 doses of DC/MM fusions. DCs were generated from adherent mononuclear cells cultured with GM-CSF and IL-4 for 5–7 days and matured with TNFa. DCs strongly expressed costimulatory and maturation markers. Myeloma cells were isolated from bone marrow aspirates and were identified by their expression of CD38, CD138, and/or MUC1. DC and MM cells were fused with polyethylene glycol as previously described and fusion cells were quantified by determining the percentage of cells that coexpress unique DC and myeloma antigens. To date, 19 patients have been enrolled and 18 have completed vaccine generation. Mean yield of the DC and myeloma preparations was 1.84 × 108 and 8.3 × 107 cells, respectively. Mean fusion efficiency was 40% and the mean cell dose was 4.3 × 106 fusion cells. As a measure of their potency as antigen presenting cells, fusion cells prominently stimulated allogeneic T cell proliferation in vitro. Mean stimulation indexes were 12, 57, and 31 for T cells stimulated by myeloma cells, DCs, and fusion cells, respectively. Adverse events judged to be potentially vaccine related included injection site reactions, pruritis, myalgias, fever, chills, and tachycardia. Six patients have completed the follow up period and 3 patients are currently undergoing vaccination. All patients achieved a partial response to transplant. Three patients demonstrated resolution of post-transplant paraprotein levels following vaccination. One patient with highly aggressive disease who experienced disease progression in the early post-transplant period, demonstrated initial response and then stabilization of disease with vaccination. We are examining the effect of transplant and vaccination on measures of cellular immunity, anti-tumor immunity and levels or activated as compared to regulatory T cells. T cell response to PHA mitogen was transiently depressed post-transplant. In contrast, a transient increase was noted post-transplant in mean T cell expression of IFNγ in response to autologous myeloma cell lysate. In preliminary studies, a relative increase in the ratio of activated (CD4/CD25low) to regulatory (CD4/CD25high) T cells was observed. To date, all evaluable patients demonstrated evidence of vaccine stimulated anti-tumor immunity as manifested by a rise in IFNγ expression by CD4 and/or CD8+ T cells following ex vivo exposure to autologous tumor lysate. In this ongoing study, fusion cell vaccination in conjunction with stem cell transplantation has been well tolerated, induced anti-tumor immunity and clinical responses in patients with multiple myeloma.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 117, No. 2 ( 2011-01-13), p. 393-402
    Abstract: We have developed a tumor vaccine in which patient-derived myeloma cells are chemically fused with autologous dendritic cells (DCs) such that a broad spectrum of myeloma-associated antigens are presented in the context of DC-mediated costimulation. We have completed a phase 1 study in which patients with multiple myeloma underwent serial vaccination with the DC/multiple myeloma fusions in conjunction with granulocyte-macrophage colony-stimulating factor. DCs were generated from adherent mononuclear cells cultured with granulocyte-macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor-α and fused with myeloma cells obtained from marrow aspirates. Vaccine generation was successful in 17 of 18 patients. Successive cohorts were treated with 1 × 106, 2 × 106, and 4 × 106 fusion cells, respectively, with 10 patients treated at the highest dose level. Vaccination was well tolerated, without evidence of dose-limiting toxicity. Vaccination resulted in the expansion of circulating CD4 and CD8 lymphocytes reactive with autologous myeloma cells in 11 of 15 evaluable patients. Humoral responses were documented by SEREX (Serologic Analysis of Recombinant cDNA Expression Libraries) analysis. A majority of patients with advanced disease demonstrated disease stabilization, with 3 patients showing ongoing stable disease at 12, 25, and 41 months, respectively. Vaccination with DC/multiple myeloma fusions was feasible and well tolerated and resulted in antitumor immune responses and disease stabilization in a majority of patients.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3080-3080
    Abstract: Multiple myeloma expresses unique antigens that potentially serve as targets for tumor specific immunotherapy. Dendritic cells (DCs) are the most potent antigen presenting cells that prominently express costimulatory molecules and are uniquely able to stimulate anti-tumor immune responses. We have developed a promising cancer vaccine in which patient derived myeloma cells are fused with autologous DC resulting in the presentation of a broad array of tumor antigens in the context of DC mediated costimulation. DC/myeloma fusions potently stimulate anti-tumor immune responses in vitro as manifested by the lysis of autologous tumor targets. We are currently conducting phase I clinical trials in which patients with myeloma undergo serial vaccination with DC/myeloma fusions alone or in conjunction with stem cell transplantation. GM-CSF (100 μg) was administered subcutaneously on the day of vaccination and for 3 days thereafter. To date, 18 patients have been enrolled (11- vaccine alone, 7 vaccine transplant). To generate mature DCs, adherent mononuclear cells were isolated from a leukapharesis collection, cultured for 5 days with GM-CSF and IL-4 and terminal maturation was induced by exposure to TNFa for 48–72 hours. DCs prominently expressed HLA class II, costimulatory and maturation markers. The mean yield and viability of the DC preparations was 1.5 x 108 cells and 88%, respectively. Patient derived myeloma cells were isolated from bone marrow aspirates and were quantified by the expression of CD38 and/or CD138. The mean yield and viability of the myeloma cell collections was 7.3 x 107 cells and 89%, respectively. Fusion cells were generated by coculture of DCs with myeloma cells at a 3:1–10:1 ratio in the presence of 50% polyethylene glycol. Fusion cells were quantified by determining the percentage of cells that co-expressed unique DC and myeloma antigens. Mean fusion efficiency and viability of the fusion cell preparation was 40% and 84%, respectively. As a measure of immunologic potency, fusion cells prominently stimulated allogeneic T cell proliferation. To date, 13 patients have completed vaccination at a dose of 1–5 x 106 fusion cells. Adverse events judged to be potentially vaccine related have included vaccine injection site reactions, edema, rash, fever (infection), chills, fatigue, muscle aches, pruritis, and diarrhea. One patient with a history of prior deep venous thrombosis (DVT) developed a DVT and pulmonary embolus of uncertain relation to the vaccine. To date, 4/6 evaluable patients have demonstrated evidence of vaccine induced anti-myeloma immunity as demonstrated by at least 2 fold increase in IFNγ expression by CD4 and/or CD8 T cells in response to ex vivo exposure to autologous tumor lysate. Of patients undergoing vaccine therapy alone, 5 patients demonstrated stabilization of the myeloma paraprotein for 2–6 months following initiation of vaccination. Of 3 patients completing post-transplant vaccination, 1 patient demonstrated resolution of the persisting myeloma protein post-transplant, 1 patient exhibited stable post-transplant paraprotein levels for 6 months, and 1 patient demonstrated a transient increase followed by a progressive decline in paraprotein levels post-transplant.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1794-1794
    Abstract: The epithelial mucin antigen (MUC1) is aberrantly expressed in many epithelial tumors and hematologic malignancies and promotes oncogenesis and tumor progression. MUC1 is recognized by the T cell repertoire and has served as a target for cellular immunotherapy. In the present study, we examined MUC1 as a marker for myeloid leukemia cells and their progenitors and its capacity to serve as a target for leukemia stem cells. Myeloid leukemia cells were isolated from bone marrow aspirates or peripheral blood in patients with high levels of circulating disease. MUC1 was not expressed on unselected leukemia samples (mean expression 3%, n=12). Similarly, low levels of MUC1 expression were seen in leukemic blasts with monocytoid differentiation (mean expression 2.7%, n=5). A subset of leukemia specimens underwent CD34 selection by magnetic bead separation. In contrast to unselected cells, 38% of CD34+ leukemia cells expressed MUC1 (n=5). The leukemia stem cell compartment was isolated by separating CD34+/CD38−/ lineage- fractions by flow cytometric sorting. Leukemia stem cells demonstrated strong expression of MUC-1 by immunohistochemical staining and FACS analysis. Similarly, we examined MUC1 expression on progenitor cells derived from chronic phase chronic myeloid leukemia and following blast transformation. MUC1 was seen in only 4% of CD34+ cells obtained from chronic phase CML samples (n=4) while uniform expression was observed in samples derived from patients with accelerated/blastic phase disease. These data suggest that MUC1 serves as a marker for early leukemia progenitors and is associated with blastic transformation. We assessed the capacity of a cancer vaccine consisting of dendritic cell (DC)/myeloid leukemia fusions to stimulate immune responses that target MUC1 and other antigens expressed by the stem cell compartment. DCs were generated from adherent mononuclear cells that were cultured with GM-CSF and IL-4 and matured with TNFa. DCs were fused with patient derived myeloid leukemia cells using polyethylene glycol as previously described. Fusion cells were quantified by determining the percentage of cells that expressed unique DC and leukemia antigens. DC/AML fusions induced the expansion of MUC1 specific T cells. Stimulation of autologous T cells with DC/AML fusions resulted in a mean 3 fold increase in CD8+ cells binding the MUC-1 tetramer (N=4). DC/AML fusions stimulated anti-tumor immune responses that targeted leukemia stem cells. Fusion stimulated T cells demonstrated increased expression of IFNγ following exposure to lysate generated from unselected leukemia cells (29 fold) and leukemia stem cells (28 fold). In contrast, exposure to renal carcinoma lysate generated only a 5 fold increase in IFNγ. In summary, these findings suggest that leukemic progenitors in AML and accelerated/blast phase CML express MUC-1. DC/tumor fusion vaccines target the MUC-1 protein and the stem cell compartment, and may be a potent immunotherapeutic strategy to eliminate the malignant stem cell clone in AML.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of Immunotherapy, Ovid Technologies (Wolters Kluwer Health), Vol. 35, No. 7 ( 2012-09), p. 555-569
    Type of Medium: Online Resource
    ISSN: 1524-9557
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2012
    detail.hit.zdb_id: 2048797-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 93, No. 5 ( 1999-03-01), p. 1487-1495
    Abstract: Multiple myeloma (MM) cells express idiotypic proteins and other tumor-associated antigens which make them ideal targets for novel immunotherapeutic approaches. However, recent reports show the presence of Kaposi’s sarcoma herpesvirus (KSHV) gene sequences in bone marrow dendritic cells (BMDCs) in MM, raising concerns regarding their antigen-presenting cell (APC) function. In the present study, we sought to identify the ideal source of DCs from MM patients for use in vaccination approaches. We compared the relative frequency, phenotype, and function of BMDCs or peripheral blood dendritic cells (PBDCs) from MM patients versus normal donors. DCs were derived by culture of mononuclear cells in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4. The yield as well as the pattern and intensity of Ag (HLA-DR, CD40, CD54, CD80, and CD86) expression were equivalent on DCs from BM or PB of MM patients versus normal donors. Comparison of PBDCs versus BMDCs showed higher surface expression of HLA-DR (P = .01), CD86 (P = .0003), and CD14 (P = .04) on PBDCs. APC function, assessed using an allogeneic mixed lymphocyte reaction (MLR), demonstrated equivalent T-cell proliferation triggered by MM versus normal DCs. Moreover, no differences in APC function were noted in BMDCs compared with PBDCs. Polymerase chain reaction (PCR) analysis of genomic DNA from both MM patient and normal donor DCs for the 233-bp KSHV gene sequence (KS330233) was negative, but nested PCR to yield a final product of 186 bp internal to KS330233 was positive in 16 of 18 (88.8%) MM BMDCs, 3 of 8 (37.5%) normal BMDCs, 1 of 5 (20%) MM PBDCs, and 2 of 6 (33.3%) normal donor PBDCs. Sequencing of 4 MM patient PCR products showed 96% to 98% homology to the published KSHV gene sequence, with patient specific mutations ruling out PCR artifacts or contamination. In addition, KHSV-specific viral cyclin D (open reading frame [ORF] 72) was amplified in 2 of 5 MM BMDCs, with sequencing of the ORF 72 amplicon revealing 91% and 92% homology to the KSHV viral cyclin D sequence. These sequences again demonstrated patient specific mutations, ruling out contamination. Therefore, our studies show that PB appears to be the preferred source of DCs for use in vaccination strategies due to the ready accessibility and phenotypic profile of PBDCs, as well as the comparable APC function and lower detection rate of KSHV gene sequences compared with BMDCs. Whether active KSHV infection is present and important in the pathophysiology of MM remains unclear; however, our study shows that MMDCs remain functional despite the detection of KSHV gene sequences.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1999
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2000
    In:  Proceedings of the National Academy of Sciences Vol. 97, No. 6 ( 2000-03-14), p. 2715-2718
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 97, No. 6 ( 2000-03-14), p. 2715-2718
    Abstract: We have reported that fusions of murine dendritic cells (DCs) and murine carcinoma cells reverse unresponsiveness to tumor-associated antigens and induce the rejection of established metastases. In the present study, fusions were generated with primary human breast carcinoma cells and autologous DCs. Fusion cells coexpressed tumor-associated antigens and DC-derived costimulatory molecules. The fusion cells also retained the functional potency of DCs and stimulated autologous T cell proliferation. Significantly, the results show that autologous T cells are primed by the fusion cells to induce MHC class I-dependent lysis of autologous breast tumor cells. These findings demonstrate that fusions of human breast cancer cells and DCs activate T cell responses against autologous tumors.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2000
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...