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: Oncotarget, Impact Journals, LLC, Vol. 6, No. 11 ( 2015-04-20), p. 8567-8578
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Cancer Research Vol. 68, No. 20 ( 2008-10-15), p. 8456-8464
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 20 ( 2008-10-15), p. 8456-8464
    Abstract: Idiotype (Id) protein, secreted by myeloma cells, is a tumor-specific antigen. Id-based immunotherapy has been explored in patients with myeloma, and results were disappointing. Although previous studies have shown that Id-specific CTLs are able to lyse myeloma cells, it is unclear whether other types of Id-specific T cells, such as type-1 T-helper (Th1) and type-2 T-helper (Th2) cells, are also able to suppress or kill myeloma cells. Using a 5T murine myeloma model, we generated T-cell clones of different subsets and examined their function in the context of myeloma cells. Id-specific CTLs specifically lysed myeloma cells via MHC class I, perforin, and Fas ligand (FasL), and Th1, but not Th2, cells lysed the myeloma cells by FasL–Fas interaction. CTL and Th1 cells also suppressed the growth and function of myeloma cells, whereas Th2 cells promoted the proliferation and enhanced the secretion of Id protein and cytokines by myeloma cells. CTL and Th1, but not Th2, cells were able to eradicate established myeloma in vivo after adoptive transfer. These results show that Id-specific CTL and Th1 are promising effector cells, whereas Th2 provide no protection and may even promote tumor progression in vivo. [Cancer Res 2008;68(20):8456–64]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Clinical Cancer Research Vol. 15, No. 3 ( 2009-02-01), p. 951-959
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 15, No. 3 ( 2009-02-01), p. 951-959
    Abstract: Purpose: We showed recently that anti-β2-microglobulin (β2M) monoclonal antibodies (mAb) have remarkably strong apoptotic effects on myeloma cells in vitro and in SCID-hu mice. However, whether the mAbs will be therapeutic and safe in the treatment of myeloma patients, in whom every tissue expresses low densities of MHC class I molecules and elevated levels of soluble β2M are present, remains to be determined. Experimental Design: In this study, human-like myeloma mouse models (HLA-A2-transgenic NOD/SCID mice) were developed, which express mature and functional human MHC class I (HLA-A2 and human β2M) on murine organs and present high levels of circulating human β2M derived from human myeloma cells. Myeloma-bearing mice were treated intraperitoneally with anti-β2M mAbs, and the distribution and effects of the mAbs on normal organs and established tumors were examined. Results: Our results show that anti-β2M mAbs were effective in suppressing myeloma growth in treated mice. The therapeutic efficacy of the mAbs in these mice are comparable with those observed in myeloma-bearing nontransgenic NOD/SCID mice in which no human MHC class I is expressed on murine organs. Furthermore, although the mAbs can be detected on different organs, no tissue damage or cell apoptosis was observed in the mice. Conclusion: Based on the antimyeloma efficacy and low toxicity in the mice, our study suggests that anti-β2M mAbs may be safe and the tissue-expressing and soluble β2M may not compromise their therapeutic effects in myeloma patients. This study provides further support for the future application of the mAbs as therapeutic agents for multiple myeloma.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Molecular Medicine, Springer Science and Business Media LLC, Vol. 90, No. 6 ( 2012-6), p. 695-706
    Type of Medium: Online Resource
    ISSN: 0946-2716 , 1432-1440
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 1462132-0
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 2337-2337
    Abstract: Introduction: Mantle cell lymphoma (MCL) has a poor outcome and is a therapeutic challenge. Preclinical evaluation of investigational agents for MCL has been limited by lack of suitable animal models that mimic the natural history of human MCL and provide the microenvironment in which MCL cells thrive. Since MCL usually involves the bone marrow, we developed an in vivo mouse model for primary human MCL cells in severe combined immunodeficient mice (SCID-hu), which have been implanted with human fetal bone. Materials and Methods: Human primary MCL cells were obtained and isolated from spleen, lymph nodes, bone marrow aspirates, or peripheral blood of six different MCL patients. Purified patient primary MCL cells were directly inoculated into human fetal bone chip or injected into mouse tail vein. Immunohistochemical staining with anti-human CD20 or cyclin D1 antibodies and detection of circulating human beta 2-microglobulin (B2M) in mouse serum were used to monitor the engraftment, growth, and immigration of human primary MCL cells in SCID-hu mice. Results: A total of 30 SCID-hu mice and 5 SCID mice were used. Twenty of SCID-hu mice received inoculation of 0.5 – 5 × 106 of patient primary MCL cells (2–5 SCID-hu mice/patient sample) into human fetal bone chips implanted in mice subcutaneously. Five of SCID-hu mice and 5 of SCID mice (without human fetal hone chips) were injected intravenously with 5 × 106 of patient MCL cells. The same number of cells were injected into human bone chips in 5 SCID-hu mice with equal volume of PBS as controls. Successful primary MCL cell engraftment was observed in 15 out of 20 SCID-hu mice after injection of these cells into human fetal bones. But only one out of 5 SCID-hu mice had successful engraftment after the intravenous injection of primary MCL cells into mouse tail vein. Importantly, none of SCID mice had successful engraftment after intravenous injection of 5 × 106 of primary MCL cells. These data indicated that human fetal bone provides a critical microenvironment for the survival and growth of primary MCL cells. Increasing levels of circulating human B2M in mouse serum were found after successful engraftment and growth of human primary MCL cells in SCID-hu mice. Immunohistochemical staining with anti-human CD20 and cyclin D1 antibodies confirmed that, similar to the human disease, primary MCL cells homed to mouse lymph nodes, spleen, bone marrow, and gastrointestinal tract but not to mouse liver. Treatment of MCL-bearing SCID-hu mice with atiprimod suppressed B2M secreted by functioning human MCL cells and induced tumor regression. Conclusion: Human primary MCL cells from patient were able to successfully engraft in SCID-hu mice, and mimiced the natural organ involvement of human disease homing to mouse lymph node, spleen, bone marrow, and gastrointestinal tract but not to liver. This in vivo model mimiced the natural biological features of human MCL and is therefore useful for preclinical evaluation of new therapeutic agents.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 740-740
    Abstract: Abstract 740 Bone destruction is common in osteolytic tumors such as multiple myeloma. Myeloma cells that reside in the bone cause osteolysis, which is characterized by severe bone pain, multiple fractures, and hypercalcemia in patients and thereby, compromises patient quality of life. While myeloma cells have been proposed to activate osteoclasts and/or inhibit osteoblasts leading to bone resorption, the mechanism underlying myeloma-induced osteolytic bone lesions is still poorly elucidated. As p38 mitogen-activated protein kinase (MAPK) is constitutively activated in myeloma, we hypothesized that p38 MAPK activation in myeloma cells might be responsible for myeloma-induced osteolytic bone lesions. Human myeloma cell lines ARP-1 and MM.1S caused bone lesions in SCID mice after intravenous injection. However, once p38 MAPK was knocked down by small hairpin RNAs (shRNAs) in these cell lines, myeloma was established but failed to cause bone lesions in SCID mice, whereas the wild-type or vector control myeloma cells established myeloma (with similar tumor burdens as myeloma cells with knocked-down p38 MAPK) and caused bone lesions in tibial bones, as measured by radiograph and peripheral quantitative m-computed tomography and further confirmed by histological examination. In vivo examination revealed that the number of bone surface-covering osteoclasts, osteoclast size, cellular nuclear numbers in per osteoclast, and levels of circulating collagen type I (bone resorption marker) and TRAP5b (active osteoclast marker) were all reduced in mice injected with myeloma cells with knocked-down p38 MAPK as compared with controls. Consistently, we found that tumor p38 MAPK had active effects on in vitro osteoclast differentiation and bone resorption, and inhibited osteoblast differentiation and function, suggesting that constitutive activation of p38 MAPK in myeloma cells induces osteoclastogenesis. By functional studies and protein array analysis, we showed that monocyte chemotactic protein-1 (MCP-1) and dickkopf-1 (DKK-1) are upregulated downstream of p38 MAPK activation in myeloma cells and are responsible for enhanced osteoclastogenesis. Our results showed that p38 MAPK activation in myeloma cells upregulate MCP-1 and DKK-1 expression and production, which are released into the microenvironment. MCP-1 enhanced RANK expression on osteoclast precursors and DKK-1 increased RANKL secretion from stromal cells, all of which led to activation of NF-kB and MAPK signaling pathways in osteoclasts. In vivo blockade of MCP-1 and DKK-1 by specific antibodies significantly abrogated tumor p38 MAPK-induced osteoclast activation and bone lesions in established myeloma-SCID mice. Thus, our results have elucidated a novel mechanism that p38 MAPK activity in myeloma cells contributes to myeloma-induced osteolytic bone lesions. This study indicates that disruption of tumor p38 MAPK may be a new therapeutic approach to treat osteolytic bone lesions in myeloma. Disclosures: No relevant conflicts of interest to declare.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 108, No. 13 ( 2006-12-15), p. 4071-4077
    Abstract: Previous studies demonstrated that circulating dendritic cells (DCs) in myeloma patients were functionally abnormal. However, the phenotype and function of patients' monocyte-derived DCs (MoDCs), which are commonly used for immunotherapy, were poorly defined. This study was undertaken to examine the quality of MoDCs from myeloma patients compared with cells from healthy donors. We found that patient-derived MoDCs are phenotypically and functionally defective. Compared with their normal counterparts, patient-derived, mature MoDCs expressed significantly lower levels of CD1a, CD40, CD80, and HLA-DR and were poor at activating alloreactive T cells, presenting recall antigen, and activating autologous antigen- and myeloma-specific T cells. These abnormalities may be attributed to elevated production of autocrine cytokines such as IL-6, activated p38 and STAT3, and inhibited MEK/ERK signaling pathways in the progenitor cells. Treatment with neutralizing IL-6–specific antibody and, more importantly, p38 inhibitor, or both, could correct these abnormalities. Treating patient-derived cells with these agents not only significantly increased cell yield but also produced MoDCs that were as functional as their normal counterparts. Thus, this study has delineated the mechanistic defects of MoDCs from myeloma patients and identified ways for restoring the function of the cells to improve the efficacy of DC-based immunotherapy in this disease.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 110, No. 8 ( 2007-10-15), p. 3028-3035
    Abstract: We recently showed that monoclonal antibodies (mAbs) against β2-microglobulin (β2M) have a remarkably strong apoptotic effect on myeloma cells. The mAbs induced apoptosis by recruiting major histocompatibility complex (MHC) class I to lipid rafts, activated c-Jun N-terminal kinase (JNK), and inhibited phosphatidylinositol 3-kinase (PI3K)/Akt and extracellular signal–regulated kinase (ERK) pathways. Growth and survival cytokines such as interleukin-6 (IL-6) and insulin-like growth factor-I (IGF-I), which could protect myeloma cells from dexamethasone-induced apoptosis, did not affect mAb-mediated cell death. This study was undertaken to elucidate the mechanisms underlying anti-β2M mAb–induced PI3K/Akt and ERK inhibition and the inability of IL-6 and IGF-I to protect myeloma cells from mAb-induced apoptosis. We focused on lipid rafts and confirmed that these membrane microdomains are required for IL-6 and IGF-I signaling. By recruiting MHC class I into lipid rafts, anti-β2M mAbs excluded IL-6 and IGF-I receptors and their substrates from the rafts. The mAbs not only redistributed the receptors in cell membrane, but also abrogated IL-6– or IGF-I–mediated Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3), PI3K/Akt, and Ras/Raf/ERK pathway signaling, which are otherwise constitutively activated in myeloma cells. Thus, this study further defines the tumoricidal mechanism of the mAbs and provides strong evidence to support the potential of these mAbs as therapeutic agents for 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3040-3040
    Abstract: Abstract 3040 Dickkopf-1 (DKK1), a secreted protein and Wnt signaling pathway inhibitor, is highly expressed by the tumor cells of almost all patients with multiple myeloma (MM) and may be responsible for suppressed osteoblast formation. In our previous studies, we demonstrated that DKK-1 is a potent tumor-associated antigen in MM recognized by cytotoxic T lymphocytes (CTLs), which can effectively lyse autologous myeloma tumor cells in vitro (Qian et al., Blood 2007;110:1587-1594) and eradicate established patient-derived primary myeloma in SCID-hu mice upon adoptive transfer. To examine the potential of DKK1-based immunotherapy in MM, we investigated the efficacy of active vaccination with (murine) DKK1 DNA vaccine in a murine (MOPC-21) myeloma model. A plasmid DNA construct encoding defensin2-sFv was generated, and DKK1 full-length cDNA was cloned from DKK1-expressing mouse stromal cells by reverse transcription–polymerase chain reaction and genetically fused with defensin2 (DNA-vac). We first examined the ability of the vaccines to protect mice from developing myeloma. While 100% of mice vaccinated with vector control or PBS (10 mice for each group) developed tumors, 70% and 40% mice vaccinated with DNA-vac or DNA-vac with CpG (ODN 1826), respectively, developed tumors. These results clearly show that active vaccination with DKK1 DNA vaccines was able to protect mice from tumor challenge, and that combination with CpG was more effective than DNA vaccine alone. On day 90 after tumor challenge, all surviving tumor-free mice were rechallenged with the same myeloma cells and followed for tumor development. By day 180, All surviving mice that were vaccinated with DNA-vac or DNA-vac with CpG have no developed tumors, indicating that active vaccination with DNA vaccines induced strong tumor-specific memory immune responses to protect mice from tumor rechallenge. Next, we examined the therapeutic effects of DNA vaccines in our myeloma mouse model. DNA vaccine alone eradicated established myeloma in 1 out of 5 mice, while DNA vaccine plus CpG eradicated myeloma in 3 out of 5 mice bearing intermediate tumors (≥ 5 mm in diameter). These results indicate that DNA vaccine plus CpG was much more effective at eradicating established myeloma than DNA vaccine alone. Finally, the mechanisms of tumor protection induced by DNA vaccines were investigated. By depleting CD4+ or CD8+ T cells, we showed that CD8+ T cells are required for DNA-induced antimyeloma responses. In DNA-vaccinated mice, splenocytes contained increased numbers of DKK1-specific, IFN-g-secreting and proliferative T cells. The splenic CD8+ T cells exhibited enhanced cytotoxicity against myeloma cells. Furthermore, DKK1-specific CD8+ T cells were shown to be increased in DKK1-DNA vaccinated mice by using DKK1-peptide (P11, P15 and P210) tetramer staining. These results demonstrate the presence of myeloma-specific CTLs in vaccinated mice and show that DKK1 DNA vaccine can induce a potent CTL response capable of killing myeloma cells. Together, our study lays a basis for future clinical trials in MM by using DKK1 as a vaccine for all patients. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 657-657
    Abstract: Human C-reactive protein (CRP) is an acute-phase protein, and elevated levels of CRP are present in patients with infections, inflammatory diseases, necrosis such as myocardial infarction, or malignancies including multiple myeloma (MM), lymphoma, and carcinoma. CRP is able to bind a variety of ligands and receptors, activating the classical complement pathway. Accumulating evidence strongly suggests that in cardiovascular disease CRP is not only a marker of inflammation, but also contributes to pathogenesis of the disease. These findings led to our hypothesis that CRP may have a functional effect on tumor cells. The present study was undertaken in a myeloma setting to determine whether CRP might affect tumor cell growth and survival. When added to the culture of primary myeloma cells isolated from patients, CRP promoted cell proliferation and reduced cell apoptosis in a dose-dependent manner. To confirm this result, we examined the effects of CRP on myeloma cell lines under stressed conditions and showed that CRP protected myeloma cells from apoptosis induced by serum starvation or IL-6 deprivation. More importantly, CRP also protected myeloma cells from apoptosis induced by dexamethasone or melphalan, two common chemotherapy drugs for MM. The protection was significant since CRP reduced cell death by 50 to 60%, and may be clinically relevant because the results were reproduced in myeloma-SCID mouse models. Injection of CRP prior to treatment of myeloma-bearing mice with dexamethasone or melphalan significantly undermined the therapeutic effects of these chemotherapy drugs. Mice receiving CRP with dexamethasone or melphalan had significantly larger tumor burdens compared with mice treated with dexamethasone or melphalan alone, whereas treatment with CRP alone had no effects on the tumor cells. CRP protected tumor cells from apoptosis by downregulating Bax expression, inhibiting phosphorylation of Bcl-2, and upregulating phosphorylation of Bad, which led to inhibited caspase-9, caspase-3 and PARP activation induced by dexamethasone. We next examined cell surface receptors for CRP and found that CRP bound Fcγ RII CD32A and/or CD32C, but not CD32B. Specific siRNAs that inhibited CD32A or CD32C but not CD32B expression, and antibodies against CD32A/C that blocked receptor-ligand interaction abrogated CRP-mediated protection of cell apoptosis. These results indicate that CRP mediated its effects via activating immunoreceptor tyrosine-based activation motif (ITAM)-containing Fcγ RII. By binding to these receptors, CRP increased the level of phosphorylated Akt, ERK1/2, and IkBa; relocalized NFkB p65 to the nucleus; and inhibited p38 kinase activity. Inhibitors against these signaling molecules blocked the activity of these pathways and abrogated CRP-mediated protection of myeloma cell apoptosis induced by dexamethasone. Therefore, our results provide strong evidence for a novel effect of CRP on myeloma cells. This study also implicates CRP as a potential target for MM therapy.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...