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
Filter
Material
Language
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    Georg Thieme Verlag KG ; 1999
    In:  Thrombosis and Haemostasis Vol. 82, No. 08 ( 1999), p. 345-352
    In: Thrombosis and Haemostasis, Georg Thieme Verlag KG, Vol. 82, No. 08 ( 1999), p. 345-352
    Abstract: The study of genetic bleeding disorders provided the first link between platelet functions and specific membrane glycoproteins. Two examples are well known and have been the subject of numerous reviews. First, Glanzmann’s thrombasthenia is a bleeding disorder caused by a defect of platelet aggregation in which the glycoprotein αIIbβ3 (GP IIb-IIIa) is either lacking or is expressed but is defective.1 We now know that αIIbβ3 exists on the surface of unstimulated platelets in an inactive form but, through a process known as “inside-out” signaling, responds to platelet stimulation to become a receptor for soluble fibrinogen and von Willebrand factor (vWF) to mediate platelet aggregation. αIIbβ3 is also known to bind immobilized fibrinogen and, through a process known as “outside-in” signaling, to induce platelet stimulation.2 A second example is Bernard-Soulier syndrome, a bleeding disorder caused by the failure of platelets to bind to subendothelial matrices due to the lack of or defective GP Ib-IX-V.3 It is now known that GP Ib-IX-V binds to vWF to mediate the adhesion of unstimulated platelets to injured blood vessel walls.4,5 GP Ib-IX-V interactions also induce platelet stimulation, a process mediated by signaling through GP Ib-IX-V.6 The mechanisms responsible for the binding of adhesive proteins to αIIbβ3 and GP Ib-IX-V are beginning to be understood and, as such, targets for therapeutic intervention have been identified. Three parenteral αIIbβ3 antagonists have demonstrated a therapeutic benefit in large-scale clinical trials of acute coronary syndromes, including unstable angina, non Q-wave myocardial infarction, and percutaneous intervention, and are now commercially available.7 Many orally available αIIbβ3 antagonists are presently in clinical trials. Although GP Ib antagonists have not been pursued as aggressively, animal studies have shown that they do have a proven antithrombotic benefit.8 Despite these advances in the understanding of glycoprotein ligand binding and development of therapeutic antagonists of adhesive protein receptors, the mechanisms responsible for transducing signals through these receptors have remained elusive. It is now established that signal transduction reactions through αIIbβ3 and GP Ib-IX-V are not only involved in platelet aggregation to cause vessel occlusions, but also that glycoprotein signaling affects thrombus growth and stability, as well as the biology and perhaps the pathology of the vessels in which aggregates occur. In one example, platelet-derived growth factor (PDGF), secreted in response to αIIbβ3 signaling from the α-granules of aggregated platelets, is a primary smooth muscle cell mitogen and is believed to be involved not only in the response to vascular injury but also in atherosclerotic lesion progression.9,10 In another example, CD 154 (previously termed CD40 ligand) redistributes from α-granule membranes to the surface of aggregated platelets in response to αIIbβ3 signaling.11 CD 154 is an important inflammatory mediator that induces the release of cytokines from endothelial and smooth muscle cells, initiates vascular inflammation, and participates in atherosclerotic lesion progression.12 A third example involves the assembly of prothrombinase and factor Xase on the surface of aggregated platelets, enabling platelet thrombi to be procoagulant and accounting for the apparent anticoagulant activity of αIIbβ3 antagonists.13,14 In addition, platelet aggregates also display fibrinogen and vWF bound to platelet membrane glycoproteins that function to recruit additional platelets and, therefore, enhance thrombus growth.15 More recent data also indicate that platelet aggregation induces de novo protein synthesis.16,17 These and other events are secondary to the initial adhesion and aggregation reactions of platelets and are consequences of signaling reactions induced by the adhesion and aggregation receptors. Thus, characterization of the membrane glycoprotein signal transduction pathways has become essential, not only to understand platelet function, but also to determine whether there are additional ways by which platelet-mediated pathologies can be regulated. Platelet membrane glycoprotein signaling reactions either do not occur in nucleated cells normally used for transfection studies or are insufficiently characterized. Accordingly, the use of genetics to study mechanisms of platelet adhesive protein receptor signaling has been limited. The advent of technologies that facilitate genetic manipulations in the mouse genome has produced new ways to define protein function and determine the structure-function relationships of individual proteins and is proving of value in unraveling signal transduction pathways in platelets. Although one should always be cautious in extrapolating data from mouse to human platelets (as demonstrated by the PAR receptors, see below), it is impressive that much of what has been learned about platelets appears to apply to both mouse and human. Indeed, this review summarizes the status of genetic manipulations of the mouse genome that have contributed to our understanding of platelet membrane adhesion receptor signaling in platelets.
    Type of Medium: Online Resource
    ISSN: 0340-6245 , 2567-689X
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 1999
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Georg Thieme Verlag KG ; 2000
    In:  Thrombosis and Haemostasis Vol. 84, No. 08 ( 2000), p. 312-318
    In: Thrombosis and Haemostasis, Georg Thieme Verlag KG, Vol. 84, No. 08 ( 2000), p. 312-318
    Abstract: Lesions in the genes for GPIbα, GPIbβ or GPIX result in a bleeding diathesis, the Bernard-Soulier syndrome (BSS), which associates a platelet adhesion defect with thrombocytopenia, giant platelets and abnormal megakaryocytes (MK). The role of GPV, also absent in BSS, was recently addressed by gene targeting in mice. While a negative modulator function for GPV on thrombin-induced platelet responses was found in one model, the absence of GP V had no effect on GPIb-IX expression or platelet adhesion. Our study extends previous results and reports that electron microscopy of bone marrow from the GPV knockout mice revealed a normal MK ultrastructure and development of the demarcation membrane system (DMS). There was a usual presence of MK fragments in the bone marrow vascular sinus. Immunogold labelling of MK from the knockout mice showed a normal distribution of GPIb-IX in the DMS and on the cell surface. The distribution of fibrinogen, vWF and P-selectin was unchanged with, interestingly, P-selectin also localised within the DMS in both situations. Thus GPV is not crucial to MK development and platelet production, consistent with the fact that no mutation in the GPV gene has as yet been described in BSS.
    Type of Medium: Online Resource
    ISSN: 0340-6245 , 2567-689X
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2000
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Growth Factors, Informa UK Limited, Vol. 8, No. 4 ( 1993-01), p. 253-265
    Type of Medium: Online Resource
    ISSN: 0897-7194 , 1029-2292
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 1993
    detail.hit.zdb_id: 2076728-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood Cancer Journal, Springer Science and Business Media LLC, Vol. 9, No. 2 ( 2019-02-04)
    Abstract: FcRH5 is a cell surface marker enriched on malignant plasma cells when compared to other hematologic malignancies and normal tissues. DFRF4539A is an anti-FcRH5 antibody-drug conjugated to monomethyl auristatin E (MMAE), a potent anti-mitotic agent. This phase I study assessed safety, tolerability, maximum tolerated dose (MTD), anti-tumor activity, and pharmacokinetics of DFRF4539A in patients with relapsed/refractory multiple myeloma. DFRF4539A was administered at 0.3–2.4 mg/kg every 3 weeks or 0.8–1.1 mg/kg weekly as a single-agent by intravenous infusion to 39 patients. Exposure of total antibody and antibody-conjugate-MMAE analytes was linear across the doses tested. There were 37 (95%) adverse events (AEs), 8 (21%) serious AEs, and 15 (39%) AEs ≥ grade 3. Anemia ( n  = 10, 26%) was the most common AE considered related to DFRF4539A. Two cases of grade 3 acute renal failure were attributed to DFRF4539A. There were no deaths; the MTD was not reached. DFRF4539A demonstrated limited activity in patients at the doses tested with 2 (5%) partial response, 1 (3%) minimal response, 18 (46%) stable disease, and 16 (41%) progressive disease. FcRH5 was confirmed to be expressed and occupied by antibody post-treatment and thus remains a valid myeloma target. Nevertheless, this MMAE-based antibody-drug-conjugate targeting FcRH5 was unsuccessful for myeloma.
    Type of Medium: Online Resource
    ISSN: 2044-5385
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2600560-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 9269-9271
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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. 112, No. 11 ( 2008-11-16), p. 4974-4974
    Abstract: Antibody-based therapeutic approaches have revolutionized the treatment of non- Hodgkin’s lymphomas (NHL) as well as other hematological malignancies. However, given the biological and clinical heterogeneity of the diseases, a large variability in clinical response has been observed. Currently, patients with refractory or relapsed lymphomas have limited therapeutic options, and therefore the need to develop effective new treatments remains urgent. Antibody drug conjugates (ADC) may prove valuable in this respect. The B cell surface antigen, CD79b, is an attractive target for this approach as it is rapidly internalized upon antibody engagement and has an appropriate expression pattern, being expressed only on normal and malignant B-cells. Anti-CD79b-MCC-DM1 is an ADC consisting of a chimeric monoclonal anti-CD79b antibody, 10D10, conjugated via a thioether linker to a cytotoxic drug, DM1. DM1, a maytansinoid derivative, is a potent tubulin polymerization inhibitor. Anti-CD79b ADCs have demonstrated strong therapeutic activity against B-cell lymphoma xenografts (Polson, 2007). To investigate the pharmacodynamics (PD), pharmacokinetics (PK), and tolerability of 10D10-MCC-DM1 in nonhuman primates, we administered 2 intravenous ~30 mg/kg doses (q 3 weeks) of 10D10-MCC-DM1 or unconjugated antibody 10D10 to cynomolgus monkeys, along with vehicle controls. 10D10-MCC-DM1 and 10D10 resulted in sustained depletion of B-cells and B cell subsets in peripheral blood, with more substantial depletion in the group given 10D10- MCC-DM1. These findings correlated well with the depletion of B-cells observed in lymphoid tissues (bone marrow, lymph nodes, and spleen). We observed an initial rapid depletion of B-cells, likely attributed to probable antibody-mediated clearance by the reticuloendothelial system. This was followed by a greater sustained B-cell depletion by the ADC, attributed to the anti-mitotic pharmacology expected following B-cell receptorbound ADC internalization. In addition, unconjugated 10D10 showed substantially less depletion than 10D10-MCC-DM1 of the CD20+CD21− B cell subset, a subgroup of B cells which is phenotypically similar to human germinal center cells. There was no substantial difference in T cells or NK cells. The PD profiles are particularly encouraging from a therapeutic perspective, as the ADC may be expected to target proliferating NHL cells in a manner similar to dividing B-cells in germinal centers. The pharmacokinetics are described by the plasma measures of total antibody, conjugate, and DM1. 10D10-MCC-DM1 was cleared slowly from the plasma with a terminal half-life of 8.98 ± 1.7 days. The apparent central volume of distribution for 10D10-MCC-DM1 was 47.7 mL/kg, which approximated the physiological plasma volume in monkeys. Modest and expected accumulation was observed following the second dose. At the similar dose level of 10D10 and 10D10-MCC-DM1, the exposure of the total antibody was similar. These PK data, when compared alongside the PD results, point to the increased effectiveness of the drug conjugate in depleting B-cells. Free DM1 concentration in plasma following administration of 10D10-MCC-DM1 exhibited a profile similar to that of 10D10-MCC-DM1 but concentrations were much lower than the corresponding 10D10-MCC-DM1 with a Cmax of 189 ± 23 ng/mL. Based on the species-invariant method, it is projected that the half-life in humans for the conjugate is approximately 19.3 ± 3.4 days. Thus the pharmacokinetic profiles of the conjugate appear to support a dosing schedule of every three weeks in clinical studies. Both 10D10-MCC-DM1 and 10D10 were well tolerated. In monkeys administered 10D10-MCC-DM1, in addition to changes in the lymphoid and hematopoietic systems (B-cell depletion), minimal axonal degeneration of the sciatic nerve was observed. Trastuzumab (Herceptin)-MCC-DM1, an ADC which has shown promising safety and efficacy profiles in clinical trials, displayed similar non-B-cell related effects in cynomolgus monkeys with dose-limiting thrombocytopenia in humans. Thus it appears that targeting B-cells by an anti-CD79b ADC does not have any additional target-independent toxicity. Taken together, these preclinical data suggest that targeting CD79b with ADCs may provide a safe and effective therapy for B-cell malignancies in humans.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2001
    In:  Blood Vol. 98, No. 2 ( 2001-07-15), p. 368-373
    In: Blood, American Society of Hematology, Vol. 98, No. 2 ( 2001-07-15), p. 368-373
    Abstract: The glycoprotein (GP) Ib-V-IX complex plays a critical role in initiating platelet adhesion to von Willebrand factor (vWF) at the site of vascular injury. The complex also forms a high-affinity binding site for thrombin. Using an intravital microscopy mouse model, it was previously established that vWF plays a critical role in mediating platelet adhesion and thrombus formation following mesenteric arteriolar injury induced by ferric chloride. Further characterization of this model showed that these thrombotic events were also thrombin dependent. Using this vWF- and thrombin-dependent model, this study shows that GP V gene deficiency significantly accelerates both platelet adhesion and thrombus formation in mice following arteriolar injury. The time required for vessel occlusion in GP V–deficient (GP V−/−) mice was significantly shorter than that in wild-type mice. Interestingly, large emboli were also produced in GP V−/− mice, but not in wild-type mice, causing frequent downstream occlusion. However, when the 2 genotypes were compared in the in vitro perfusion chamber where thrombin was inhibited by heparin, no significant differences were found in either initial single-platelet adhesion or thrombus volume. These results demonstrate that GP V−/− mice have accelerated thrombus growth in response to vascular injury and suggest that this is caused by enhanced thrombin-induced platelet activation rather than enhanced binding of GPIb-V-IX to vWF. Absence of GP V also compromises thrombus stability.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2001
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2023
    In:  Journal of Clinical Oncology Vol. 41, No. 6_suppl ( 2023-02-20), p. 537-537
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 6_suppl ( 2023-02-20), p. 537-537
    Abstract: 537 Background: Tislelizumab, a humanized monoclonal antibody that targets programmed cell death protein 1 (PD-1), has shown promising activity in the treatment of advanced urothelial bladder cancer (UBC). Recent studies suggest that liver metastases (LM) are associated with reduced effectiveness of PD-1/PD-L1 therapies (Yu, et al. Nat Med. 2021;27[1]:152-164). We evaluated how LM correlate with survival outcomes and the tumor immune microenvironment in UBC patients treated with tislelizumab in the BGB-A317-204 trial (NCT04004221). Methods: Cox regression was used to evaluate the effect of LM on overall survival (OS). Other key baseline characteristics were further included as covariates in the model to investigate the adjusted effect of LM and the interactions of LM with them. Gene expression profiling and multiplex immunohistochemistry (mIHC) analysis were performed on baseline tumor samples. Gene expression differences between LM positive (LM+) and LM negative (LM–) patients were compared by Wilcoxon rank-sum test for continuous biomarkers, and Fisher’s exact test for categorical biomarkers. All P-values reported in this post-hoc exploratory analysis were descriptive, without multiplicity adjustment. A result of P 〈 0.05 was considered statistically significant. Results: A total of 113 patients were included in the analysis. LM were present in 27/113 (23.9%) of them. Cox regression analysis showed the presence of LM was a negative prognostic factor for overall survival (OS) in UBC patients treated with tislelizumab, OS (hazard ratio (HR), 3.7; 95% CI, 2.3-6.2; P 〈 0.001). The LM negative prognostic value remained unaffected after adjustment for other baseline covariates. Next, we investigated immune cell infiltration and gene expression as potential indicators of impaired response in LM+ patients included in this analysis. Significantly fewer circulating lymphocytes at baseline were found in LM+ compared to LM– patients ( P 〈 0.05), while no differences were observed in other immune cell types. Reduced expression of gene signatures of CD4 Th1 ( P 〈 0.05), CD8+T cells ( P 〈 0.05) and NK cells functions ( P 〈 0.05) within tumor tissues was observed in samples from LM+ patients. Reduced CD8+ T-cell infiltration within the tumor and the intra-tumor stroma was also observed in LM+ patients based on mIHC analysis. No significant differences were observed in tumor mutational burden level between LM+ and LM– patients. Conclusions: LM were associated with worse outcomes of UBC patients treated with tislelizumab in the A317-204 study. Diminished number and impaired function of CD8+ T cells and NK cells in the tumor microenvironment of LM+ patients may have contributed to the worse survival observed in this subset of patients. Clinical trial information: NCT04004221 .
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2008
    In:  Investigational New Drugs Vol. 26, No. 1 ( 2008-02), p. 7-12
    In: Investigational New Drugs, Springer Science and Business Media LLC, Vol. 26, No. 1 ( 2008-02), p. 7-12
    Type of Medium: Online Resource
    ISSN: 0167-6997 , 1573-0646
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2008
    detail.hit.zdb_id: 2009846-7
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 16, No. 2 ( 2010-01-15), p. 497-508
    Abstract: Purpose: Targeted therapeutics have significantly changed the outcome for patients diagnosed with cancer. Still, effective therapeutic intervention does not exist for many cancers and much remains to be done. The objective of this study was to identify novel genes that potentially regulate tumor growth, to target these gene products with monoclonal antibodies, and to examine the therapeutic potential of these antibodies. Experimental Design: Using cDNA microarray analysis, we identified genes overexpressed in several solid malignancies. We generated a mouse monoclonal antibody, 19.2.1, and its humanized counterpart, PDL192, to one such target, TweakR (TWEAK receptor, Fn14, TNFRSF12A, CD266), and characterized the antitumor activities in vitro and in mouse xenograft models. Results: Both 19.2.1 (mouse IgG2a) and PDL192 (human IgG1), like TWEAK, the natural ligand of TweakR, inhibited the growth of several TweakR-expressing cancer cell lines in anchorage-dependent and anchorage-independent assays in vitro. Both antibodies showed significant antitumor activity in multiple mouse xenograft models. PDL192 and 19.2.1 also induced antibody-dependent cellular cytotoxicity (ADCC) of cancer cell lines in vitro. A chimeric version of 19.2.1 containing the mouse IgG1 Fc region (19.2.1×G1) exhibited significantly less ADCC than 19.2.1. However, 19.2.1×G1 showed differential activity in vivo, with activity equivalent to 19.2.1 in one model, but significantly less efficacy than 19.2.1 in a second model. These results indicate that PDL192 and 19.2.1 mediate their antitumor effects by signaling through TweakR, resulting in reduced tumor cell proliferation, and by ADCC. Clin Cancer Res; 16(2); 497–508
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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...