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: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3099-3099
    Abstract: Identification of unique cell surface markers on myeloma cells is important for development of targeted therapies and detection of residual disease. GPRC5D is an orphan receptor reportedly expressed at high level in bone marrow (BM) aspirates or biopsies from myeloma patients. Other studies detected GPRC5D in skin and brain but not in other tissues. The aims of the study were to assess GPRC5D gene expression and cell surface localization in myeloma cells, the changes in expression of GPRC5D and other typical myeloma cell markers following coculture of primary myeloma cells with osteoclasts and the consequences of GPRC5D gene silencing on myeloma cell growth. Global gene expression profile (GEP), qRT-PCR and immunohistochemistry revealed exclusive high expression of GPRC5D in normal and myeloma plasma cells but not in B cells, MSCs, osteoclasts or BM mononucleated cells. GPRC5D expression was higher in myeloma plasma cells from newly diagnosed patients (n=698) compared to normal plasma cells (n=26, p 〈 0.0001)). Among molecularly classified groups GPRC5D expression is higher in MS, MF and LB subgroups and lowest in CD2 and MM cell lines. Flow cytometry analysis and immunohistochemistry detected cell surface expression of GPRC5D in myeloma plasma cells while nuclear localization was also detected in certain myeloma cell lines (e.g. H929). Western blots analysis confirmed GPRC5D expression in whole cell lysate and nuclear fraction. We and other demonstrated phonotypical plasticity of myeloma plasma cells capable of altered expression of recognizable plasma cell markers (e.g. CD138, CD45) following coculture with stromal cells (Dezorella et al., 2009) or osteoclasts (Yaccoby, 2005). In coculture of primary CD138-selected myeloma cells with osteoclasts (n=8), CD138 (p 〈 0,004), CD38 (p 〈 0.001) and GPRC5D (p 〈 0.007) were commonly and significantly downregulated, CD45 (p 〈 0.02) was upregulated, and IRF4 expression was unaffected in cocultured myeloma cells compared to the control freshly obtained uncultured myeloma cells assessed by GEP and qRT-PCR. We also observed reduced expression of GPRC5D in MM cells purified from focal lesion compared to interstitial marrow in paired clinical samples (n=176, p 〈 1.21E-09), suggesting that high activity of osteoclasts in osteolytic lesions mediates phenotypical alteration in myeloma cells. Stable knockdown of GPRC5D by 70% in CAG myeloma line using lentiviral particles containing shRNA had no effect on short-term growth of these cells assessed by MTT assay. These data indicate that GPRC5D is a novel cell surface marker for myeloma plasma cells and that its expression is reduced in dedifferentiated myeloma cells. 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: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1926-1926
    Abstract: Tumor associated macrophages are reportedly accumulated in myelomatous bones and support myeloma cell survival and growth. Myeloma cells from the majority of patients express ICAM1, a cell surface receptor associated with the interaction of myeloma cells with stromal cells. BI-505, a humanized IgG1 lambda antibody against ICAM1, has been recently reported to inhibit growth of ICAM1-expressing myeloma cells by specifically activating macrophages (Veitonmäki et al, Cancer Cell, 2013) while lenalidomide is a known clinical immunomodulatory agent. The aim of the study was to test the effect of combination treatment with BI-505 and lenalidomide on growth of myeloma cells in vivo and in vitro. ICAM1-expressing myeloma cells from 4 patients with progressive disease were engrafted in SCID-hu mice and treated with control antibody, BI-505 (10 mg/kg, weekly, i.p.), lenalidomide (2 mg/kg, daily, p.o.) or BI-505 and lenalidomide for 5-8 weeks (5-6 mice/group in each set of patient’s cells). Myeloma growth was assessed by monitoring circulating human kappa light chain immunoglobulin or soluble syndecan-1 in the mice sera, or using live-animal imaging of myeloma cells infected with lentiviral particles containing EGFP/luciferase construct. Myeloma growth was inhibited by the two drugs in all experiments. At the end of each experiment, tumor burden in the BI-505, lenalidomide and drug combination groups was 13±4% (p 〈 0.0001), 21±6% (p 〈 0.0001) and 3±1% (p 〈 0.0001) relative to control groups, respectively. Myeloma burden was also significantly lower in the drug combination group compared to BI-505 (p 〈 0.03) or lenalidomide (p 〈 0.008) groups. These data are well in concordance with results from experiments with myeloma cell lines RPMI8226 and U266 in immunodeficient mice where the combination of BI-505 and lenalidomide showed significantly more anti-tumor effect compared to either treatment alone. In vitro, we established a coculture system in which whole donor bone marrow (BM) cells were cultured for 7 days with serum pooled from myeloma patients, followed by coculture with CD138-selected myeloma cells for an additional 7 days (BM:myeloma cell ratio 4:1). Phenotypically, the cultured BM contained cells of various hematopoietic lineages including macrophages. Growth of myeloma cells was assessed by CD45/CD38 flow cytometry or bioluminescence analysis of luciferase-expressing myeloma cells in coculture. In this system, primary myeloma cells survived and propagated regardless of their molecular risk signature or subsets while BI-505 (1-5 μg/ml) inhibited growth of ICAM1-expressing myeloma cells by 54±6% (n=5, p 〈 0.003) but had no effect on myeloma cells that do not express ICAM1. In the coculture setting, combination of BI-505 (2.5 μg/ml) and lenalidomide (0.5 μM) had higher anti-tumor efficacy than each of the agents alone. These data indicate that macrophages can be activated by BI-505 to induce anti-myeloma response and that the anybody activity is enhanced in combination with lenalidomide. Disclosures: Teige: BioInvent International: Employment. Frendéus:BioInvent International: Employment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 2944-2944
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2944-2944
    Abstract: Abstract 2944 Multiple myeloma (MM) bone disease is characterized by increased activity of osteoclasts and reduced osteoblast numbers. We recently reported that cytotherapy with mesenchymal stem cells (MSCs) promotes bone formation, inhibits bone disease and reduces MM growth (Yaccoby et al., 2006; Li et al., 2011) and that MSCs secrete bone remodeling associated factors such as decorin that directly inhibits osteoclastogenesis and promotes osteoblastogenesis (Li et al., 2012). To shed light on molecular mechanisms associated with the cytotherapeutic effects of MSCs we exploited the SCID-hu model engrafted with MM cells from various patients and cytotherapeutically treated with fetal or healthy donors MSCs. Human global gene expression profile was performed in nonmyelomatous implanted bones (n=5) and in myelomatous implanted bones injected with human MSCs (1×106 cells/bone) and analyzed immediately (control group, n=17) or 24 hours later (n=16). We indentified heme oxygenase 1 (HMOX1) as one of the top significant upregulated genes consistently induced by 24 hours cytotherapy using fetal MSCs or MSCs from 3 different donors. HMOX1 induction was confirmed by qRT-PCR and immunohistochemistry and was also found to be consistently induced by MSCs cytotherapy in myelomatous bones engrafted with MM cells from different patients (n=4). Further analysis revealed lower expression of HMOX1 in myelomatous versus nonmyelomatous bones. HMOX1, an inducible antioxidant, degrades intracellular heme into carbon monoxide, free iron and biliverdin and is involved in oxidative stress response and intracellular iron homeostasis both of which are known to regulate osteoclastogenesis. Culture of human blood mononucleated cells or committed osteoclast precursors with MSCs (non-contact co-culture) in osteoclastogenic medium supplemented with RANKL and M-CSF inhibited multinucleated osteoclast formation by 70% (p 〈 0.0001) and 97% (p 〈 0.0001), respectively. MSCs induced HMOX1 expression in osteoclast precursors by 3 folds (p 〈 0.02) and HMOX1 levels remained higher in these cells during osteoclast formation. Concurrently, RANK (RANKL receptor) and NFATC1 (osteoclast main transcription factor) were constantly downregulated in osteoclast precursors by MSCs by 65±10% (p 〈 0.01) and 42±11% (p 〈 0.05), respectively, followed by reduced expression of the osteoclastic markers cathepsin K (p 〈 0.0002), acid phosphatase 5 (p 〈 0.01) and vitronectin receptor (p 〈 0.02) at the end of the differentiation process (7 days). Similar to MSCs, overexpression of HMOX1 in osteoclast precursors using lentiviral vector markedly reduced their ability to form osteoclasts while HMOX1 inducer, hemin (50 μM), induced HMOX1 in osteoclast precursors and inhibited their differentiation into osteoclasts by 49±2% (p 〈 0.0001). In MM-bearing SCID-rab mice (9 mice/group) bone mineral density (BMD) was reduced by 17±3% from pretreatment levels in control group whereas in hosts treated with hemin for 3 weeks BMD was reduced by 2±3% (p 〈 0.005). Histological bone sections revealed reduced number of TRAP-expressing osteoclasts in myelomatous bones from hemin-treated hosts (p 〈 0.01). These data suggest that HMOX1 is suppressed in myelomatous bone and that therapeutic induction of HMOX1 is a promising approach to control MM-induced osteoclastogenesis and osteolytic bone disease. 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: 2012
    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. 118, No. 21 ( 2011-11-18), p. 3943-3943
    Abstract: Abstract 3943 Myeloma plasma cell high labeling index and molecular signature of proliferation are strong adverse prognostic factors often characterize patients with high risk disease. The overall aim of the study was to identify cell proliferation associated genes implicating highly proliferating myeloma cells in the supportive bone marrow environment. To shed light on molecular factors associated with rapid growth of myeloma cells, primary myeloma cells from 10 patients, molecularly classified as high risk were engrafted in SCID-rab mice. Growth rate of myeloma varied between patients' cells but in all cases myeloma propagated within and surrounding the supportive implanted bone but not in any murine organs. We performed global gene expression profiling (GEP) on myeloma plasma cells recovered from mice and compared their GEP with the baseline, pre-injected myeloma cells. Based on stringent criteria (e.g. p 〈 0.05, 〉 2 folds) approximately 127 probe sets were commonly overexpressed and 36 probe sets underexpressed in myeloma cells from SCID-rab mice than baseline myeloma cells. Genes whose expression altered were mainly associated with proliferation, survival, metabolism, transcription and immunity. Among genes involved in cell proliferation we indentified stearoyl CoA desaturase 1 (SCD1), which was upregulated in 7 of 10 cases by overall 2.3±0.6 folds (p 〈 0.01). In coculture of primary myeloma cells with the supportive osteoclasts (n=8), SCD1 was upregulated in 6 of 8 cocultures by 5.6±2.4 folds (p 〈 0.02). SCD1 upregulation in vivo and in cocultures was consistently observed in 3 different GEP probe sets. SCD1 is a rate-limiting enzyme responsible for synthesis of monounsaturated fatty acids and is activated in highly proliferating tumor cells to sustain the increasing demand of new membrane phospholipids and energy storage, and reducing intracellular content of cytotoxic saturated fatty acids. Various SCD1 inhibitors are currently being evaluated for metabolic diseases. In vitro, small molecule SCD1 inhibitor (BioVision) dose dependently (0.1–10 μM, 96 hrs) inhibited growth of rapidly growing myeloma cell lines (n=5) but had moderate inhibitory effect on their survival. Compared to control vehicle-treated cultures, numbers of viable myeloma cells were reduced by 76±5% (p 〈 0.008) and 51±3% (p 〈 0.0001) following treatment with 0.1 μM and 5 μM of SCD1 inhibitor, respectively. Cell viability was reduced from 91±0.5% in control groups to 82±3% (p 〈 0.05) and 73±5% (p 〈 0.03) in cultures treated with 0.1 μM and 5 μM of SCD1 inhibitor, respectively. In vivo, luciferase-expressing H929 myeloma cells were engrafted in SCID-rab mice. Myeloma growth was monitored by live-animal bioluminescence imaging. Upon establishment of myeloma hosts were treated with SCD1 inhibitor using Alzet osmotic pump directly connected to the open side of the implanted bone and constantly released drug (1.25 μg/hour) or vehicle over a period of 2 weeks. At experiment's end myeloma burden was increased from pretreatment levels by 49±3 folds and 30±3 folds in control vehicle- and SCD1 inhibitor-treated hosts, respectively (p 〈 0.01). We conclude that SCD1 is highly activated in proliferating myeloma cells and is essential for their rapid growth. 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: 2011
    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. 118, No. 21 ( 2011-11-18), p. 3941-3941
    Abstract: Abstract 3941 Iron overload is a significant clinical feature in multiple myeloma (MM) and has been implicated in osteoporosis. MM patients also frequently suffer from anemia presumably due to elevated hepcidin secretion and dysfunctional erythropoiesis. The aims of the study were to shed light on molecular mechanisms associated with iron overload in MM cells and study the effect of the novel iron chelator, Dp44mT, on MM cell growth, osteoclastogenesis and MM bone disease in vitro and in vivo. In our clinical global gene expression profiling (GEP) data the main iron transporter gene TFRC (transferrin receptor) was 〉 3 folds higher (p 〈 0.0001) in newly diagnosed MM cells (n=556) than normal plasma cells (n=25) while the iron exporter ferroportin was downregulated in MM cells by 〉 4 folds (p 〈 0.0001). Deregulated TFRC and ferroportin expression were more profound in the molecularly classified proliferation (PR) subtype. Osteoclasts which are known to have abundant mitochondria due to high energy consumption express excessive TFRC ( 〉 5 folds higher than highly proliferating MM cells). In primary MM cell-osteoclast cocultures (n=8) TFRC expression was upregulated in cocultured MM cells than baseline MM cells (p 〈 0.03) while ferroportin was lower in cocultured osteoclasts than control osteoclasts (p 〈 0.04). Our GEP, qRT-PCR and immunohistochemistry analyses revealed expression of hepcidin by osteoclasts but not MM cells. Hepcidin was not detected in conditioned media from osteoclasts cultured alone or cocultured with MM cells using ELISA, suggesting an autocrine role of hepcidin in maintaining excess iron in osteoclasts. In vitro, Dp44mT dose dependently inhibited growth of MM cell lines (n=3) at low nanomolar levels (IC50 at 3±0.8 nM, p 〈 0.03, 48 hrs). In contrast, known chelators such as Deferoxamine and Deferasirox inhibited myeloma cell growth at 10–50 micromoles range. At 1nM Dp44mT also suppressed formation of multinucleated osteoclasts by 87% (p 〈 0.001) and bone resorbing activity of mature osteoclasts on dentine slices by 94% (p 〈 0.03). Dp44mT induced upregulation of BMP2 expression in osteoblast precursors and promoted osteoblast differentiation. In vivo, SCID-rab mice engrafted with luciferase-expressing U266 MM line (6 mice/group) or the Hg MM line (maintained through in vivo passaging, 10 mice/group) were subcutaneously treated with vehicle or Dp44mT (1 mg/kg/day) for 2–3 weeks. Using live-animal imaging, Dp44mT reduced growth of U266 cells by 3 folds from pretreatment levels (p 〈 0.01) while in control group tumor burden was increased by 52 folds from pretreatment levels (p 〈 0.002). Dp44mT also inhibited growth of Hg MM cells determined by measurement of circulating human immunoglobulins in mice sera (p 〈 0.01). Osteoclasts numbers were lower by 36% (p 〈 0.003) while osteoblasts numbers were higher by 59% (p 〈 0.017) in myelomatous bones from hosts treated with Dp44mT than control vehicle. Our data suggest that interaction of myeloma cells with osteoclasts alters expression of distinct iron metabolism associated factors which elicit iron overload in both cell types, resulting in increased myeloma cell proliferation and osteoclast activity. This study also suggests that Dp44mT is an effective iron chelator with marked anti-MM activity. Disclosures: Barlogie: Celgene, Genzyme, Novartis, Millennium: Consultancy, Honoraria, Patents & Royalties. Shaughnessy:Myeloma Health, Celgene, Genzyme, Novartis: Consultancy, Employment, Equity Ownership, Honoraria, Patents & Royalties.
    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: American Journal of Hematology, Wiley, Vol. 88, No. 6 ( 2013-06), p. 463-471
    Type of Medium: Online Resource
    ISSN: 0361-8609
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 1492749-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1648-1648
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1648-1648
    Abstract: Multiple myeloma (MM) is a neoplasm of low proliferative plasma cells that are highly dependent on bone marrow (BM) microenvironment for survival and growth. Primary human MM cells do not survive in conditions designed for cell lines and cell lines do not represent the primary cell heterogeneity and are microenvironment independent. Current in vitro and in vivo systems for primary human MM are limited to co-culture with specific BM cell type or growth in immunodeficient animal model. We and others have demonstrated the ability of the recognizable mature, C138+ or CD45-/CD38+ MM cells to produce MM in those experimental systems. The aim of the study was to establish and test the survival and growth of primary MM plasma cells in long term culture with a complete adult normal BM. This system is different from the autologous BM culture that is already affected by the disease. Whole BM cells from healthy donors were cultured in αMEM medium supplemented with 10% FBS and 10% serum from MM patients. Following 7-9 days the cultures were composed of adherent and nonadherent cellular compartments. The nonadherent compartment contained typical BM hematopoietic cells such monocytes, B and T lymphocytes and NK cells as assessed by flow cytometry, while the adherent compartment contained cells that morphologically resemble macrophages, osteoclasts, megakaryocytes and fibroblast-like cells. At this culture stage, CD138-selected MM cells from 13 patients were added to the BM cultures and survival and growth of MM cells were determined after 7 days by assessing proportion of CD45low/intermediate/CD38high MM plasma cells among total number of nonadherent cells. Low proportions of normal plasma cells (0.1-1.7%) were also detected in the cultures throughout the duration of experiments. In additional study, we used luciferase-expressing IL6-dependent INA6 MM cells and stroma-dependent MM line to assess MM growth within the whole BM cultures using bioluminescence analysis. Cell viability was constantly high ( & gt;90%) in the primary MM cell-normal BM cultures. Subsets of primary MM plasma cells were survived and detected in all culture cases while in 9 of 13 experiments, number of primary MM plasma cells was heterogeneously increased by 63±19% (p & lt;0.01). INA6 and stroma-dependent line had growth advantage in this culture compared to their standard growth conditions with IL6 or stromal cells, respectively. Patient serum but not healthy donor serum was essential for establishing primary MM growth. This culture system demonstrates the ability of MM plasma cells to grow in a normal, adult whole human BM microenvironment and is appropriate for studying MM-microenvironment interaction, characterization of MM cell subpopulations capable of long term survival and targeted therapy. Citation Format: Rakesh Bam, Sathisha Upparahalli Venkateshaiah, Xin Li, Sharmin Khan, Wen Ling, Bart Barlogie, Joshua Epstein, Shmuel Yaccoby. Primary myeloma plasma cells are capable of growth in adult, normal whole human bone marrow environment . [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1648. doi:10.1158/1538-7445.AM2013-1648
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 8
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 15, No. 1 ( 2015-12)
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2041352-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: International Journal of Molecular Sciences, MDPI AG, Vol. 24, No. 10 ( 2023-05-20), p. 9048-
    Abstract: The main aim of this study is to synthesize contrast microbubbles (MB) functionalized with engineered protein ligands using a microfluidic device to target breast cancer specific vascular B7-H3 receptor in vivo for diagnostic ultrasound imaging. We used a high-affinity affibody (ABY) selected against human/mouse B7-H3 receptor for engineering targeted MBs (TMBs). We introduced a C-terminal cysteine residue to this ABY ligand for facilitating site-specific conjugation to DSPE-PEG-2K-maleimide (M. Wt = 2.9416 kDa) phospholipid for MB formulation. We optimized the reaction conditions of bioconjugations and applied it for microfluidic based synthesis of TMBs using DSPE-PEG-ABY and DPPC liposomes (5:95 mole %). The binding affinity of TMBs to B7-H3 (MBB7-H3) was tested in vitro in MS1 endothelial cells expressing human B7-H3 (MS1B7-H3) by flow chamber assay, and by ex vivo in the mammary tumors of a transgenic mouse model (FVB/N-Tg (MMTV-PyMT)634Mul/J), expressing murine B7-H3 in the vascular endothelial cells by immunostaining analyses. We successfully optimized the conditions needed for generating TMBs using a microfluidic system. The synthesized MBs showed higher affinity to MS1 cells engineered to express higher level of hB7-H3, and in the endothelial cells of mouse tumor tissue upon injecting TMBs in a live animal. The average number (mean ± SD) of MBB7-H3 binding to MS1B7-H3 cells was estimated to be 354.4 ± 52.3 per field of view (FOV) compared to wild-type control cells (MS1WT; 36.2 ± 7.5/FOV). The non-targeted MBs did not show any selective binding affinity to both the cells (37.7 ± 7.8/FOV for MS1B7-H3 and 28.3 ± 6.7/FOV for MS1WT cells). The fluorescently labeled MBB7-H3 upon systemic injection in vivo co-localized to tumor vessels, expressing B7-H3 receptor, as validated by ex vivo immunofluorescence analyses. We have successfully synthesized a novel MBB7-H3 via microfluidic device, which allows us to produce on demand TMBs for clinical applications. This clinically translatable MBB7-H3 showed significant binding affinity to vascular endothelial cells expressing B7-H3 both in vitro and in vivo, which shows its potential for clinical translation as a molecular ultrasound contrast agent for human applications.
    Type of Medium: Online Resource
    ISSN: 1422-0067
    Language: English
    Publisher: MDPI AG
    Publication Date: 2023
    detail.hit.zdb_id: 2019364-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Institute of Electrical and Electronics Engineers (IEEE) ; 2020
    In:  IEEE Transactions on Medical Imaging Vol. 39, No. 10 ( 2020-10), p. 3079-3088
    In: IEEE Transactions on Medical Imaging, Institute of Electrical and Electronics Engineers (IEEE), Vol. 39, No. 10 ( 2020-10), p. 3079-3088
    Type of Medium: Online Resource
    ISSN: 0278-0062 , 1558-254X
    RVK:
    Language: Unknown
    Publisher: Institute of Electrical and Electronics Engineers (IEEE)
    Publication Date: 2020
    detail.hit.zdb_id: 2068206-2
    detail.hit.zdb_id: 622531-7
    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...