Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    The Endocrine Society ; 2003
    In:  The Journal of Clinical Endocrinology & Metabolism Vol. 88, No. 10 ( 2003-10-01), p. 5043-5044
    In: The Journal of Clinical Endocrinology & Metabolism, The Endocrine Society, Vol. 88, No. 10 ( 2003-10-01), p. 5043-5044
    Type of Medium: Online Resource
    ISSN: 0021-972X , 1945-7197
    RVK:
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2003
    detail.hit.zdb_id: 2026217-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: The Journal of Clinical Endocrinology & Metabolism, The Endocrine Society, Vol. 91, No. 9 ( 2006-09-01), p. 3662-3666
    Abstract: Context: The epidermal growth factor receptor (EGFR), a transmembrane tyrosine kinase (TK) receptor that mediates proliferation and survival signaling, is expressed in a wide variety of normal and neoplastic tissues. EGFR inhibitors have produced objective responses in patients with non-small-cell lung carcinomas harboring activating EGFR TK domain somatic mutations. Objective and Methods: Because the EGFR pathway has been reported to be important for the pathophysiology of thyroid carcinoma, we investigated the expression and mutational status of EGFR in 14 thyroid carcinoma cell lines as well as its functional role by evaluating their in vitro sensitivity to AEE788, a new dual-family EGFR/ErbB2 and vascular endothelial growth factor receptor TK inhibitor. We also evaluated the mutational status, mRNA and protein expression, as well as phosphorylation status of EGFR in a panel of thyroid carcinoma specimens. Results: EGFR expression and phosphorylation in the thyroid carcinoma cell lines and tissue specimens were present but not stronger than in noncancerous thyroid tissue. EGFR TK domain mutations were detected in two of 62 histological specimens (3.2%) but not in cell lines. All thyroid carcinoma cell lines were significantly less sensitive (IC50 at least 25-fold higher) in vitro to AEE788 than a primary culture of EGFR-mutant lung carcinoma cells. Conclusions: Thyroid carcinoma cells overall are poorly responsive to clinically relevant concentrations of AEE788 in vitro. The presence of EGFR-activating TK domain mutations may identify a small minority of thyroid cancer patients that may benefit from EGFR inhibitors, but additional preclinical evidence of efficacy is needed.
    Type of Medium: Online Resource
    ISSN: 0021-972X , 1945-7197
    RVK:
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2006
    detail.hit.zdb_id: 2026217-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Seminars in Oncology, Elsevier BV, Vol. 30, No. 2 ( 2003-4), p. 248-252
    Type of Medium: Online Resource
    ISSN: 0093-7754
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2003
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 1571-1571
    Abstract: The genetic heterogeneity of multiple myeloma (MM) and the evolution of the disease as it progresses result in a multiplicity of proliferative/anti-apoptotic pathways that can operate in MM cells, particularly within the context of their interaction with the bone marrow (BM) microenvironment. Collectively, these factors can contribute to de novo or acquired refractoriness of MM cells to diverse conventional and/or novel anti-MM therapeutics. To counteract the multiplicity of pathways potentially implicated in the control of MM cell resistance to drug-induced apoptosis, we have explored the use of multi-targeted small-molecule inhibitors (e.g. kinase inhibitors), clinical levels of which can simultaneously suppress the expression of multiple targets. Here we describe studies on the novel, oral, multi-targeted kinase inhibitor dasatinib (BMS-354825, Bristol Myers Squibb), which inhibits BCR-ABL, SRC, c-KIT, PDGF-R, and ephrin (EPH) receptor kinases. Although BCR-ABL and c-KIT are not primary oncogenes driving MM proliferation and survival, we studied dasatinib because of: a) emerging data from our laboratory (CS Mitsiades, unpublished observations) on patterns of expression/function patterns of EPH receptors in MM cell lines and primary tumor specimens; and b) the roles of PDGF-R and SRC in tumor-microenvironment interactions, e.g. pericytes/endothelial cells in angiogenesis and osteoclast-mediated bone resorption, respectively. In vitro, we found that dasatinib significantly suppresses, at clinically achievable sub-μM concentrations, the viability of MM cell lines (including lines resistant to conventional or other novel anti-MM agents); primary tumor specimens from multi-drug resistant MM patients; as well as MM cells co-cultured with BM stromal cells. Mechanistic studies showed that dasatinib can induce caspase-8 and -12 activation and sensitize primary MM cells to agents activating caspase-9 (e.g. Dex and bortezomib). Even though IC50 values were higher in MM cells than BCR-ABL+ CML cells, the IC50 of dasatinib was & lt;100 nM in 8/15 MM cell lines tested, suggesting substantial sensitivity to dasatinib in at least a subset of MM. Interim analyses correlating the baseline transcriptional profiles of MM cells with their degree of responsiveness to low nM levels of dasatinib showed that increased responsiveness to this inhibitor correlated with increased baseline expression of diverse proliferative/anti-apoptotic genes, including transcriptional regulators (e.g. MAF, MAFF, NFYC, PML, YY1, DAXX), cell surface receptors (e.g. EPH receptor B4, CXCR4), proteasome subunits (PSMC3, PSMD12, PSME2) and regulators of apoptosis (e.g. CIAP1, IKKe). Ongoing expansion of the panel of tested MM cells is expected to further refine this molecular signature of dasatinib responsiveness. Ongoing in vivo studies are addressing the optimal dose, schedules and sequence of administration of dasatinib in combination with other anti-MM agents. These studies, coupled with our molecular profiling efforts to identify a molecular signature of dasatinib responsiveness, will inform the design of future clinical trials of dasatinib or its analogs for the treatment of MM patients.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 247-247
    Abstract: Multiple myeloma (MM) remains an incurable neoplasia and exhibits high propensity for de novo/acquired refractoriness even to novel agents, e.g. thalidomide (Thal) or proteasome inhibitors. This may be due to complex and evolving patterns of molecular lesions potentially conferring hyperactive antiapoptotic signaling with high degree of redundancy upon inhibition of isolated targets within those pathways. We thus hypothesized that, for genetically complex neoplasias such as MM, patient outcome might improved by addition, in the therapeutic armamentarium, of agents that simultaneously inhibit multiple proliferative/antiapoptotic targets. Towards this effort of multi-targeted therapies, we studied the tyrosine kinase inhibitor PKC412 (midausporin, Novartis, Basel, Switzerland). Low-nM levels of PKC412 selectively inhibit tyrosine kinase receptors, such as FLT3. But higher PKC412 concentrations (in μM range), which presumably inhibit (at least partly) other kinases, are achieved in clinical trials without catastrophic toxicities. This suggests that PKC412 can safely suppress in vivo the activity of, not just FLT3, but a broader spectrum of kinases, some of which (individually or cooperatively) might be critical for survival/proliferation of MM cells. Indeed, in vitro kinase activity assays showed that clinically achievable (high nM or low μM) PKC412 concentrations inhibit by & gt;20% important kinases, including p70S6K, IKK-a and Akt,. Consistent with such multi-targeted effect, in vitro MTT colorimetric survival assays showed activity of PKC412 (at sub-μM levels) against panel of 40 MM cell lines and 10 primary tumor cells from MM patients (including cells resistant to Dex, alkylating agents, anthracyclines, Thal or its immunomodulatory derivatives, bortezomib, and/or Apo2L/TRAIL), and cell lines from hematologic neoplasias, e.g. B- and T-ALL, CML, various non-Hodgkin’s lymphoma subtypes, and solid tumors (e.g. breast, prostate, lung, thyroid, ovarian, renal Ca, retinoblastoma and sarcomas)). Mechanistic studies confirmed that PKC412 blocks key growth/survival pathways (e.g. PI-3K/Akt, IKK-α/NF-κB), coupled with by downstream effects on suppression of diverse inhibitors of apoptosis (e.g. FLIP, XIAP, cIAP-2, survivin). These molecular sequelae explain, at least partly, the ability of PKC412 to sensitize MM cells to other anti-MM agents (such as Dex, cytotoxic chemotherapy or proteasome inhibitor bortezomib) and overcome protective effects of cytokines (e.g. IL-6) or bone marrow stromal cells. Importantly, PKC412 significantly prolonged the overall survival (p & lt;0.03, Kaplan-Meier analysis) of SCID/NOD mice in a clinically relevant model of diffuse MM bones lesions. These studies provide basis for clinical studies of PKC412 in MM and indicate that kinase inhibitors selectively blocking specific targets at low drug levels, may also have potent anti-tumor activities related to inhibition of multiple other, less specific, nonetheless important targets, thus allowing for anti-tumor activity in a much broader spectrum of tumor types than previously appreciated.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 101, No. 10 ( 2003-05-15), p. 4055-4062
    Abstract: Histone acetylation modulates gene expression, cellular differentiation, and survival and is regulated by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDAC inhibition results in accumulation of acetylated nucleosomal histones and induces differentiation and/or apoptosis in transformed cells. In this study, we characterized the effect of suberoylanilide hydroxamic acid (SAHA), the prototype of a series of hydroxamic acid–based HDAC inhibitors, in cell lines and patient cells from B-cell malignancies, including multiple myeloma (MM) and related disorders. SAHA induced apoptosis in all tumor cells tested, with increased p21 and p53 protein levels and dephosphorylation of Rb. We also detected cleavage of Bid, suggesting a role for Bcl-2 family members in regulation of SAHA-induced cell death. Transfection of Bcl-2 cDNA into MM.1S cells completely abrogated SAHA-induced apoptosis, confirming its protective role. SAHA did not induce cleavage of caspase-8, -9, or -3 in MM.1S cells during the early phase of apoptosis, and the pan-caspase inhibitor ZVAD-FMK did not protect against SAHA. Conversely, poly(ADP)ribose polymerase (PARP) was cleaved in a pattern indicative of calpain activation, and the calpain inhibitor calpeptin abrogated SAHA-induced cell death. Importantly, SAHA sensitized MM.1S cells to death receptor–mediated apoptosis and inhibited the secretion of interleukin 6 (IL-6) induced in bone marrow stromal cells (BMSCs) by binding of MM cells, suggesting that it can overcome cell adhesion–mediated drug resistance. Our studies delineate the mechanisms whereby HDAC inhibitors mediate anti-MM activity and overcome drug resistance in the BM milieu and provide the framework for clinical evaluation of SAHA, which is bioavailable, well tolerated, and bioactive after oral administration, to improve patient outcome.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2003
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 101, No. 7 ( 2003-04-01), p. 2762-2769
    Abstract: Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow and their egress into peripheral blood with progression to plasma cell leukemia. Our previous study defined a functional role of CD40 activation in MM cell homing and migration. In this study, we examine signaling events mediating CD40-induced MM cell migration. We show that cross-linking CD40, using either soluble CD40L (sCD40L) or anti-CD40 monoclonal antibody (mAb), induces phosphatidylinositol 3–kinase (PI3K) activity and activates its downstream effector AKT in MM.1S cells. CD40 activation also activates the MAP kinase (MEK) pathway, evidenced by phosphorylation of extracellular signal-regulated mitogen-activated protein kinase (ERK), but not c-jun amino-terminal kinase (JNK) or p38, in a dose- and time-dependent manner. Using pharmacologic inhibitors of PI3K and MEK, as well as adenoviruses expressing dominant-negative and constitutively expressed AKT, we demonstrate that PI3K and AKT activities are required for CD40-induced MM cell migration. In contrast, inhibition of ERK/MEK phosphorylation only partially (10%-15%) prevents migration, suggesting only a minor role in regulation of CD40-mediated MM migration. We further demonstrate that CD40 induces nuclear factor (NF)–κB activation as a downstream target of PI3K/AKT signaling, and that inhibition of NF-κB signaling using specific inhibitors PS1145 and SN50 completely abrogates CD40-induced MM migration. Finally, we demonstrate that urokinase plasminogen activator (uPA), an NF-κB target gene, is induced by CD40; and conversely, that uPA induction via CD40 is blocked by PI3K and NF-κB inhibitors. Our data therefore indicate that CD40-induced MM cell migration is primarily mediated via activation of PI3K/AKT/NF-κB signaling, and further suggest that novel therapies targeting this pathway may inhibit MM cell migration associated with progressive MM.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2003
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1587-1587
    Abstract: Context: The molecular chaperone hsp90 is a major anti-cancer therapeutic target because it regulates the function of proteins with pivotal roles in tumor cell proliferation, survival and drug resistance, including mutated/chimeric oncoproteins or oncogenic kinases/receptors. Our preclinical studies on the ansamycin hsp90 inhibitor tanespimycin (17-AAG) provided the rationale for clinical trials, either alone or in combination with the proteasome inhibitor bortezomib, for treatment of relapsed/refractory MM. In this study, we report preclinical studies of the new, non-ansamycin, hsp90 inhibitor NVP-AUY922. Methods/Results: We tested 36 human MM cell lines and observed with MTT colorimetric survival assays potent time- and dose-dependent anti-MM activity of NVP-AUY922. IC50 values were & lt;12.5 nM for 31 of 35 cell lines, which were also more sensitive than all non-malignant cells tested with NVP-AUY922, including bone marrow stromal cells (BMSCs), immunortalized human hepatocytes and normal donor PBMCs, indicating a differential selectivity of NVP-AUY922 against neoplastic tissues vs. non-malignant cells. Importantly, MM cell lines or primary MM tumor cells resistant to dexamethasone (Dex), melphalan, immunomodulatory thalidomide derivatives (IMIDs), bortezomib or TRAIL were sensitive to NVP-AUY922. In addition, MM-1S cells constitutively over-expressing Akt were equally sensitive to NVP-AUY922 compared to their parental cell line. These anti-MM effects were rapid, as MM cells were committed to cell death within 16 hrs of exposure to only 25 nM of NVP-AUY922. NVP-AUY922 overcame the protective effects conferred to MM cells by exogenous IL-6 and IGF-1, while BMSC co-culture attenuated NVP-AUY922 activity only at doses & lt;20 nM, as evidenced by compartment-specific bioluminescence imaging (CS-BLI). Mechanistic studies of NVP-AUY922 treatment of MM-1S cells showed early G2/M arrest followed by increased cell death, accompanied by caspase-3 and -8 cleavage (as early as 16 hrs). These events were preceded by decreased levels of Akt, B-Raf, phospho-MEK, cIAP2, and XIAP; and compensatory upregulation of hsp27 and hsp70. Given the highly single-agent activity of NVP-AUY922 at even low nM concentrations, formal statistical documentation of synergy was not observed in NVP-AUY922 combinations with conventional (e.g. Dex, doxorubicin) or novel (e.g. bortezomib) anti-MM agents. Encouragingly, no evidence of antagonism with any of these combinations was observed, indicating that NVP-AUY922 can be combined with current anti-MM agents in clinical settings. Conclusion: The new hsp90 inhibitor NVP-AUY922 has potent in vitro activity against MM cells resistant to conventional therapeutics, with selectivity for malignant compared to normal cells. Ongoing in vivo experiments and studies to identify biomarkers of pronounced sensitivity to NVP-AUY922 will help provide a framework for potential clinical trials of NVP-AUY922 in MM and other neoplasias.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 1496-1496
    Abstract: We have recently shown that tumor cell proliferation, survival and drug-resistance in multiple myeloma (MM) and a broad range of other tumors is critically influenced by insulin-like growth factors (IGFs) and their receptor (IGF-1R) (Cancer Cell2004;5:221–30). Among the pleiotropic signaling cascades downstream of IGF-1R activation, we focused on the functional implications and therapeutic targeting of the Akt/p70S6K/mTOR axis, particularly of mTOR (mammalian Target of Rapamycin), due to its regulatory role on cellular bioenergetics, a key aspect of tumor pathophysiology. Herein, we describe the in vitro and in vivo profiles of anti-tumor activity of the selective mTOR inhibitor RAD001 (Everolimus, Novartis AG). Using in vitro MTT assays, we observed that RAD001 is active (at nM concentrations) against a broad range of tumor cells, including 〉 40 MM cell lines and 〉 10 primary MM tumor cells (including cell lines or primary cells resistant to Dex, alkylating agents, anthracyclines, thalidomide (Thal), immunomodulatory Thal derivatives, bortezomib, and/or Apo2L/TRAIL), without significant impact on viability of normal hematopoietic cells or other normal tissues (e.g. bone marrow stromal cells), and its anti-MM effect was not blocked by forced overexpression of Bcl-2 or constitutively active Akt. While cytokine- or cell adhesion-mediated interactions with the bone marrow (BM) microenvironment (e.g. BM stromal cells) protects MM cells from conventional therapies (e.g. Dex or cytotoxic chemotherapy), RAD001 was able to overcome this protective effect in co-culture models of MM cells with BM stromal cells or in vitro MM cell exposure to survival factors, e.g. IL-6 or IGF-I. Furthermore, RAD001 sensitized MM cells to other anti-MM therapeutics, e.g. dexamethasone, cytotoxic chemotherapeutics, or the proteasome inhibitor bortezomib, even in cases of primary MM tumor cells refractory to these respective agents. Using hierarchical clustering analyses and relevance network algorithms, we found that the pattern of MM cell dose-response relationships to RAD001 is clearly distinct from the patterns of sensitivity or resistance to other conventional or investigational anti-MM drugs. This further supports the notion that RAD001 confers a constellation of pro-apoptotic/anti-proliferative molecular sequelae distinct from those of currently available anti-MM drugs, and also suggests that RAD001 may have anti-tumor activity even against subgroups of MM which may be resistant to other novel therapies which that are currently in clinical development. Importantly, administration of RAD001 in a SCID/NOD mice model of diffuse MM bone had in vivo anti-tumor activity, including suppression of MM tumor burden and prolongation of survival (p 〈 0.01, log-rank test). These studies highlight an important role for mTOR in growth/survival of human MM cells and provide proof-of-principle for future clinical studies of mTOR inhibitors for the treatment of MM and other plasma cell dyscrasias.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 2404-2404
    Abstract: We have previously shown that inhibitors of the hsp90 molecular chaperone (including geldanamycin, 17-allylamino-17-demethoxy-geldanamycin (17-AAG) and other members of the ansamycin family) have in vitro and in vivo activity against MM cells (including drug-sensitive and -resistant cells) and confer sensitization to several other anti-tumor agents. An extensive program of phase I clinical trials in solid tumors has documented biological activity of 17-AAG with manageable hsp90 inhibitor-related toxicities and has identified maximum tolerated doses for further clinical development. These first trials involved an intravenous formulation of 17-AAG in DMSO and egg-phospholipids. As such, it is believed that the formulation may have contributed to some of the observed side effects of 17-AAG administration; and conversely, that alternative formulations of 17-AAG in clinically inert excipients might potentially allow for administration of higher doses of 17-AAG and more favorable clinical outcomes. In this study, we describe the in vitro and in vivo clinical profile of KOS-953, a cremophor-based 17-AAG formulation. In our in vitro studies, KOS-953 exhibits comparable anti-MM activity to DMSO-based 17-AAG, including activity against a broad spectrum of MM cell lines and primary cells which are both sensitive and resistant to cytotoxic chemotherapeutics, proteasome inhibitor bortezomib, thalidomide or its derivatives. Consistent with prior experience on DMSO-based formulations of 17-AAG, KOS-953 triggered intracellular depletion of several key kinases, including Akt, Raf, IKK-a; decreased the constitutive and cytokine-induced activity of NF-κB; and suppressed expression of diverse intracellular anti-apoptotic proteins (e.g. FLIP, XIAP, survivin); leading to tumor cell sensitization to other pro-apoptotic agents (e.g. cytotoxic chemotherapy, PS-341). Importantly, we evaluated the in vivo anti-tumor activity of KOS-953 in our SCID/NOD mouse model of diffuse MM bone lesions. In this model, mice with bioluminescence imaging-confirmed diffuse skeletal lesions were randomly assigned to receive either KOS-953 (50 mg/kg i.p., twice weekly) or equal volume of vehicle. Mice in both cohorts were followed-up serially by whole-body real-time fluorescence imaging and whole-body bioluminescence imaging, models previously validated in separate studies of our group. The primary endpoint of the study was the overall survival of mice, defined as time between injection of tumor cells and sacrifice for hind limb paralysis, moribund state, or death. KOS-953 treatment was associated with prolongation of overall survival of mice (p & lt;0.05, log-rank test), and was well tolerated, without vital organ tissue damage in histopathologic analyses. These findings indicate that alternative 17-AAG formulations, such as cremophor-based ones, can deliver biologically-active doses of 17-AAG and achieve anti-tumor responses. Coupled with our accumulating experience on the role of hsp90 for MM cell proliferation, survival and drug-resistance, the current study therefore provides the preclinical framework for further clinical evaluation of hsp90 inhibitors, such as KOS-953, to improve treatment outcome in MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages