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
  • American Association for Cancer Research (AACR)  (9)
  • 2010-2014  (9)
Material
Publisher
  • American Association for Cancer Research (AACR)  (9)
Language
Years
  • 2010-2014  (9)
Year
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3868-3868
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3868-3868
    Abstract: The receptor-tyrosine-kinase-like orphan receptor 1 (ROR1) was identified by a polymerase chain reaction (PCR)-based search for tyrosine kinases similar to the tropomyocin receptor kinase (Trk) neurotropic receptors. ROR1 and a related protein, ROR2, were identified as orphan receptors with an extracellular Frizzled-like, cysteine-rich domain, an extracellular, membrane-proximal kringle domain, and an intracellular tyrosine-kinase-like domain. Both ROR proteins are primarily expressed during embryogenesis. In prior studies, we and others found that ROR1 was expressed by leukemia cells and some cancer cell lines, and was involved in cell survival. However, it was not known whether breast tumor cells expressed ROR1 or whether its expression had functional and clinical significance. In the present study we used a high-affinity mAb specific for ROR1 (named 4A5) to examine human breast cancers and investigate its functional role for tumor growth. We examined fresh-frozen tumor biopsy specimens or tissue microarrays of neoplastic and normal adult tissues for ROR1. The neoplastic cells of high proportions of human breast cancers (70%) expressed ROR1, which was not detected on non-neoplastic adult breast tissues. We interrogated available DNA microarray datasets on primary human breast cancers and cancer cell lines, and fount that breast cancer cell lines or primary breast cancers that expressed high-levels of ROR1 were more likely to lack expression of estrogen or progesterone receptors or HER2/Neu. Conceivably, tumors that are poorly differentiated are more likely to express ROR1. Patients who had primary breast cancers that expressed higher levels of ROR1 had a significantly shorter median survival than did patients with primary breast cancers that had low-to-negligible expression of ROR1. We silenced ROR1 expression in breast cancer cell lines to evaluate its function on tumors. When silenced for ROR1 for MDA-MB-231 cells that expressed ROR1, MDA-MB-231 cells were more sensitive to spontaneous apoptosis and had an impaired cell growth in vitro and in vivo. On the other hand, MCF-7 cells that were transduced to express ROR1 had more aggressive growth characteristics than did control MCF-7 cells. We investigated the mechanism by ROR1 induced cell survival and found that ROR1 could interact with casein kinase 1 epsilon (CK1α) to activate phosphoinositide 3-kinase-mediated AKT phosphorylation and cAMP-response-element-binding protein (CREB), which in turn induce expression of genes (CCNB1, BCL2, and CCND1) that can enhance resistance to apoptosis and/or promote tumor growth. Moreover, Wnt5a, a ligand of ROR1, could induce ROR1-dependent signaling and enhance cell growth. This study demonstrates that ROR1 is expressed in human breast cancers and has biological and clinical significance, indicating that it may be a potential target for breast cancer therapy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3868. doi:1538-7445.AM2012-3868
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 2796-2796
    Abstract: The Pim serine/threonine kinase family is composed of three highly homologous members; Pim-1, Pim-2 and Pim-3, identified by the ability of the prototype member Pim-1 to drive lymphomagenesis in mice. Upregulation of Pim-1 and Pim-2 is observed in leukemias and lymphomas, including AML, NHL and CLL, highlighting the potential of these kinases as therapeutic targets in these indications. Overexpression of Pim-1 or Pim-3 has also been observed in prostate, pancreatic, gastric, bladder and hepatocellular cancers. Pim kinases are downstream effectors of many cytokine and growth factor signaling pathways and are direct transcriptional targets of STAT transcription factors activated by these pathways. Pims can phosphorylate multiple substrates to mediate cell proliferation and survival. Here we describe the activity of AZD1208, an orally available, potent and highly selective Pim inhibitor that effectively inhibits all three isoforms. AZD1208 inhibits the growth of several AML cell lines and sensitivity correlates with the level of Pim-1 expression, STAT5 activation and presence of protein tyrosine kinase mutation. AZD1208 causes cell cycle arrest and apoptosis in MOLM-16 cells in culture. This is accompanied by a dose-dependent reduction in phosphorylation of BAD, 4EBP1 and p70S6K. AZD1208 suppresses the growth of MOLM-16 and KG-1a xenograft tumors in vivo in a dose proportional manner. In addition, AZD1208 leads to potent inhibition of colony growth of primary AML cells from bone marrow aspirates and downregulates phosphorylation of Pim targets. These results underscore the therapeutic potential of Pim kinase inhibition by AZD1208 for the treatment of AML. They also further support investigation of this inhibitor in other hematological and solid tumor malignancies where PIM signaling may play a role in tumorigenesis and survival. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2796. doi:1538-7445.AM2012-2796
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 975-975
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 975-975
    Abstract: MicroRNAs (miRNAs) can influence the expression levels of genes that can affect the clinical outcome of patients with chronic lymphocytic leukemia (CLL). In a well-defined cohort of 86 patients, we observed heterogeneity in the relative leukemia-cell expression of miR-155. Cases that expressed high-levels of miR-155 more frequently expressed zeta-associated protein of 70 kD (ZAP-70), used unmutated Ig heavy-chain-variable-region genes (IGHV), and had shorter treatment-free survival (TFS) and overall survival (OS) than cases with low-level expression of miR-155. Recursive partitioning allowed us to define a threshold for “high-level” expression of miR-155 (in copy numbers per CLL cell) that best associates with adverse clinical outcome. We examined the potential basis for this association. One of the genes targeted by miR-155 encodes the phosphatidylinositol polyphosphate 5-phosphatase SHIP-1, which can dephosphorylate proteins that had been activated in response to B-cell receptor (BCR) ligation. We found that CLL cells with “high-level” miR-155 expressed significantly lower levels of SHIP-1 protein and were more sensitive to ligation of surface-µ than CLL cells with low-levels of miR-155. Transfecting CLL cells with miR-155 reduced their expression of SHIP-1 and enhanced the stimulatory-response to surface-µ ligation. Conversely, transfection of CLL cells with a miR-155 inhibitor had the opposite effects. The enhanced sensitivity to BCR-ligation of cells expressing “high-level” miR-155 may account for its association with adverse clinical outcome in patients with CLL. However, within any one patient we also find heterogeneity in the expression levels of miR-155. By GEO data analysis (GSE21029) we found that CLL cells in blood expressed lower levels of miR-155 than did CLL cells in lymphoid tissues, where they interact with supportive cells, such as Nurselike cells or activated T cells, which can provide survival- or growth-promoting signals via expression of BAFF/APRIL or CD40L, respectively. We found that stimulation of CLL cells in vitro via CD40 ligation or exogenous BAFF could induce higher expression of miR-155, leading to reduced leukemia-cell expression of SHIP-1 and enhanced sensitivity to surface-µ ligation. Moreover, we found that the “proliferative” subgroup of blood CD5brightCXCR4dim CLL cells, which represent CLL cells newly released from the tissue microenvironment, expressed higher level of miR-155 and lower levels of SHIP-1 protein, and were more sensitive to surface-µ ligation than the “resting” subgroup of blood CD5dimCXCR4bright CLL cells, which represent cells that may be due to enter the tissue microenvironment. As such, this study also demonstrates that miR-155 could be upregulated by signals in the CLL microenvironment, leading to lower-level expression of SHIP-1, and enhanced stimulation in response to BCR ligation. Citation Format: Liguang Chen, Bing Cui, Suping Zhang, Marek Mraz, Jessie-F. Fecteau, Jian Yu, Ling Zhang, Lei Bao, Laura Rassenti, Karen Messer, Carlo Croce, Thomas Kipps. MicroRNA-155 In chronic lymphocytic leukemia influences B-cell receptor signaling. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 975. doi:10.1158/1538-7445.AM2014-975
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 3_Supplement ( 2013-02-01), p. C4-C4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 3_Supplement ( 2013-02-01), p. C4-C4
    Abstract: The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an evolutionarily-conserved, type-I membrane glycoprotein that is expressed in embryonic development, but not by virtually all normal post-partum tissues. Recent studies have found the ROR1 protein expressed by a variety of different cancers, particularly those that appear less differentiated and/or have high-grade histology (Am J Pathol. 2012, PMID 23041612). Because there appeared to be a tight association between gene and protein expression, we interrogated the gene-expression data on breast adenocarcinomas from each of 582 patients (pt) for ROR1. Because of our earlier observation that approximately a third of breast cancer pt in a different cohort had tumors' with high-level expression of ROR1 protein (PLoS One, 2012, PMID 22403610), we divided the cohort of 582 pt into tertiles based upon their tumors' relative expression of ROR1. Pt in the tertile with cancers having the highest expression of ROR1 (ROR1H) had a significantly higher rate of metastases than did pt with tumors having the mid (ROR1M) or lowest (ROR1L) expression of ROR1 (e.g. 61% vs. 35% or 41%, respectively, p & lt; 0.0001). Moreover, pt with ROR1H tumors had a higher rate of early relapse and poorer survival than did pt with ROR1L/M tumors, irrespective of ER or HER2 status (HR=2.3, p & lt;0.0001). We also examined for ROR1 expression in 14 distinct breast-cancer epithelial cell-lines, including 6 basal-type breast cancer cell-lines and 8 luminal-type breast cancer cell-lines. All 6 basal-type, metastases-prone, cell-lines expressed ROR1, which was not the case for low-invasive, luminal-type cell-lines. Silencing ROR1 in the basal cell-lines attenuated expression of proteins associated with epithelial-mesenchymal transition (EMT) (e.g. vimentin, SNAIL-1/2, and ZEB1), enhanced expression of epithelial cytokeratins and tight-junction proteins (e.g. CK-19 and ZO-1), and impaired each cell-line's migration/invasion capacity in vitro and metastatic potential in immune-deficient mice. Moreover, silencing ROR1 in parental MDA-MB-231 cells (or its derivatives LM2-4175 and BoM-1833, which preferentially metastasize to the lung or bone, respectively) inhibited each cell-line's capacity to generate orthotopic lung metastases or experimental lung and bone metastases in vivo. Treatment of MDA-MB-231 or other basal-type cell-lines with a monoclonal antibody (mAb) specific for ROR1 down-modulated vimentin and inhibited each cancer-cell-line's migration/invasion capacity in vitro. This mAb also significantly inhibited tumor metastasis when given to immune-deficient mice engrafted with MDA-MB-231 cells. Collectively, this study indicates that ROR1 may regulate EMT and metastasis, and that antibodies targeting ROR1 can inhibit cancer progression and metastasis. Citation Format: Bing Cui, Suping Zhang, Liguang Chen, Jianqiang Yu, George F. Widhopf, II, Jessie-F Fecteau, Laura Z. Rassenti, Thomas J. Kipps. Targeting ROR1 inhibits epithelial-mesenchymal transition and metastasis. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr C4.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4586-4586
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4586-4586
    Abstract: The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an evolutionarily-conserved, type-I membrane glycoprotein that is expressed in embryonic development, but not by virtually all normal post-partum tissues. Recent studies have found the ROR1 protein expressed by a variety of different cancers, particularly those that appear less differentiated and/or have high-grade histology (Am J Pathol. 2012, PMID 23041612). Because there appeared to be a tight association between gene and protein expression, we interrogated the gene-expression data on breast adenocarcinomas from each of 582 patients (pt) for ROR1. Because of our earlier observation that approximately a third of breast cancer pt in a different cohort had tumors’ with high-level expression of ROR1 protein (PLoS One, 2012, PMID 22403610), we divided the cohort of 582 pt into tertiles based upon their tumors’ relative expression of ROR1. Pt in the tertile with cancers having the highest expression of ROR1 (ROR1H) had a significantly higher rate of metastases than did pt with tumors having the mid (ROR1M) or lowest (ROR1L) expression of ROR1 (e.g. 61% vs. 35% or 41%, respectively, p & lt; 0.0001). Moreover, pt with ROR1H tumors had a higher rate of early relapse and poorer survival than did pt with ROR1L/M tumors, irrespective of ER or HER2 status (HR=2.3, p & lt;0.0001). We also examined for ROR1 expression in 14 distinct breast-cancer epithelial cell-lines, including 6 basal-type breast cancer cell-lines and 8 luminal-type breast cancer cell-lines. All 6 basal-type, metastases-prone, cell-lines expressed ROR1, which was not the case for low-invasive, luminal-type cell-lines. Silencing ROR1 in the basal cell-lines attenuated expression of proteins associated with epithelial-mesenchymal transition (EMT) (e.g. vimentin, SNAIL-1/2, and ZEB1), enhanced expression of epithelial cytokeratins and tight-junction proteins (e.g. CK-19 and ZO-1), and impaired each cell-line's migration/invasion capacity in vitro and metastatic potential in immune-deficient mice. Moreover, silencing ROR1 in parental MDA-MB-231 cells (or its derivatives LM2-4175 and BoM-1833, which preferentially metastasize to the lung or bone, respectively) inhibited each cell-line's capacity to generate orthotopic lung metastases or experimental lung and bone metastases in vivo. Treatment of MDA-MB-231 or other basal-type cell-lines with a monoclonal antibody (mAb) specific for ROR1 down-modulated vimentin and inhibited each cancer-cell-line's migration/invasion capacity in vitro. This mAb also significantly inhibited tumor metastasis when given to immune-deficient mice engrafted with MDA-MB-231 cells. Collectively, this study indicates that ROR1 may regulate EMT and metastasis, and that antibodies targeting ROR1 can inhibit cancer progression and metastasis. Citation Format: Bing Cui, Suping Zhang, Liguang Chen, Jianqiang Yu, George F. Widhopf, Jessie-F. Fecteau, Laura Z. Rassenti, Thomas J. Kipps. Targeting ROR1 inhibits epithelial-mesenchymal transition and metastasis. [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 4586. doi:10.1158/1538-7445.AM2013-4586
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. LB-40-LB-40
    Abstract: Leukemia has long been considered unpreventable, and how leukemogenesis is initially triggered remains largely unknown. We observed that autophagy is frequently downregulated in the hematopoietic stem cells of various types of leukemia patients. Using a conditional Becn1 knockout mouse model, we found that impaired autophagy results in rapid accumulation of reactive oxygen species (ROS) that causes DNA damages, genome instabilities and chromosomal translocations of hematopoietic stem cells, leading to their transformation to leukemia stem cells, ultimately attributed to various leukemogenenesis. Restoration of autophagy in the hematopoietic stem cells of the autophagy defective mice by constitutive expression of Becn1 could clean up ROS, inhibit DNA damages and chromosomal translocations, and block the transformation of hematopoietic stem cells to leukemia stem cells, ultimately inhibiting leukemogenesis. Furthermore, enhancement of autophagy by rapamycin could effectively remove the ROS experimentally induced in hematopoietic stem cells and eliminate the leukemia stem cells either transplanted to wild-type recipient mice or transformed from normal hematopoietic stem cells due to pharmacological inhibition of autophagy. Our data thus suggest that autophagy prevent leukemogenesis by protecting normal hematopoietic stem cells from transforming to leukemia stem cells and that leukemia prevention is possible by securing autophagy machinery in hematopoietic stem cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-40. doi:10.1158/1538-7445.AM2011-LB-40
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5472-5472
    Abstract: Chronic lymphocytic leukemia (CLL) cells express high levels of CD44, a cell-surface glycoprotein receptor of hyaluronic acid (HA). We evaluated the cytotoxic potential of a humanized anti-CD44 mAb (RG7356), which currently is undergoing clinical evaluation for the treatment of patients with solid-tumor malignancies. We found that the mAb was directly cytotoxic for leukemia B cells, but had little effect on normal B cells. Moreover, independent of compliment or cytotoxic effector cells, RG7356 could induce leukemia cells to undergo caspase-dependent apoptosis, particularly those CLL cells that expressed the zeta-associated protein of 70 kD (ZAP-70). The cytotoxic effect of this mAb was not mitigated when the CLL cells were co-cultured with mesenchymal stromal cells (MSC) or HA, or stimulated via ligation of the B-cell receptor (BCR) with anti-μ. RG7356 induced rapid internalization of CD44 on CLL cells at 37oC, resulting in reduced expression of ZAP-70, which we found complexed with CD44. Administration of 1 mg/kg of this mAb to immune-deficient mice engrafted with human CLL cells resulted in complete clearance of engrafted leukemia cells. These studies indicate that this mAb might have therapeutic activity, particularly in patients with CLL that express ZAP-70. Citation Format: Suping Zhang, Christina Wu, Jessie Farrah-Fecteau, Bing Cui, Liguang Chen, Ling Zhang, Rongrong Wu, Laura Rassenti, Fitzgerald S. Lao, Stefan Weigand, Thomas J. Kipps. Targeting chronic lymphocytic leukemia cells with a humanized monoclonal antibody specific for CD44. [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 5472. doi:10.1158/1538-7445.AM2013-5472
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 950-950
    Abstract: Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the western world. It is characterized by the clonal expansion of CD5+CD23+ B cells in blood, marrow, and second lymphoid tissues. Survival of CLL cells are supported by cells within the tissue microenvironment and by signals from the extracellular matrix, which in part are mediated via interactions with CD44, a surface glycoprotein that is expressed by CLL B cells and by putative cancer stem cells. CD44 interacts with other surface and cytosolic proteins expressed by CLL cells such as CD38, CD49d, MMP9 and ZAP-70, which enhanced signaling leading to growth or survival of CLL cells. We found that a mAb specific for CD44 was directly cytotoxic for leukemia B cells, but had little effect on normal B cells. It could induce CLL cells that expressed the zeta-associated protein of 70 kDa (ZAP-70) to undergo caspase-dependent apoptosis (Proc Natl Acad Sci, USA 2013, PMID: 23530247). The cytotoxic effect of this mAb was not mitigated when the CLL cells were co-cultured with mesenchymal stromal cells (MSCs), suggesting that targeting CD44 may provide an effective approach for anti-cancer therapy. A6 is an 8-amino acid peptide (Ac-KPSSPPEE-Am) that binds to the CD44 receptor within a region of the ligand-binding domain and induces anti-metastatic and anti-migration activity against a variety of cancers including ovarian, lung, and breast cancers. In this study we examined the cytotoxic effects and explored mechanism of A6 peptide against multiple CLL patient samples. We found that A6 peptide exhibits a significant cytotoxic effect against CLL patient samples (N=22) in vitro while having negligible toxicity against healthy patient B cell samples (N=6). A6 also exhibits cytotoxic effects against CLL cells in a dose dependent manner. More importantly, we found that A6 is able to down-modulate CD44 expression, leading to down regulation of CD44-associated proteins (e.g. ZAP-70). Because ZAP-70 can directly enhance BCR signaling, reduction of ZAP-70, following A6 treatment resulted in attenuation of intracellular [Ca2++] flux after IgM ligation, which is an indicator of decrease in BCR signaling. Moreover, A6 exhibits significantly greater direct cytotoxicity for CLL cells (p=0.029) that express ZAP-70 than for CLL cells that were ZAP-70 negative. We transplanted primary ZAP-70 positive CLL patient samples (N=2) into the peritoneal cavity of immunodeficient mice. Intraperitoneal treatment with 300 mg/kg of A6 peptide twice daily for 7 days resulted in a significant reduction of CLL burden (90% and 64.2%) compared to PBS treated control groups. No significant adverse effects were observed. A6 peptide has been very well-tolerated in clinical trials with no clearly identifed systemic adverse events in patients with solid tumors who have been treated to date (N = 40). Thus, A6 presents a promising therapeutic agent not only against CLL but also against CD44 expressing cancer stem cells. Citation Format: Hsien Lai, Suping Zhang, Christina Wu, Liguang Chen, Grace Liu, RongRong Wu, Fitzgerlad Lao, Jian Yu, Laura Rassenti, Michael Choi, Stephen Howell, Malcolm Finlayson, Thomas Kipps. Selective cytotoxicity of A6 peptide against ZAP-70 expressing CLL B-cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 950. doi:10.1158/1538-7445.AM2014-950
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 12 ( 2013-06-15), p. 3649-3660
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 12 ( 2013-06-15), p. 3649-3660
    Abstract: Metastasis is responsible for 90% of cancer-related deaths. Strategies are needed that can inhibit the capacity of cancer cells to migrate across the anatomic barriers and colonize distant organs. Here, we show an association between metastasis and expression of a type I receptor tyrosine kinase–like orphan receptor, ROR1, which is expressed during embryogenesis and by various cancers, but not by normal postpartum tissues. We found that expression of ROR1 associates with the epithelial–mesenchymal transition (EMT), which occurs during embryogenesis and cancer metastasis. Breast adenocarcinomas expressing high levels of ROR1 were more likely to have gene expression signatures associated with EMT and had higher rates of relapse and metastasis than breast adenocarcinomas expressing low levels of ROR1. Suppressing expression of ROR1 in metastasis-prone breast cancer cell lines, MDA-MB-231, HS-578T, or BT549, attenuated expression of proteins associated with EMT (e.g., vimentin, SNAIL-1/2, and ZEB1), enhanced expression of E-cadherin, epithelial cytokeratins (e.g., CK-19), and tight junction proteins (e.g., ZO-1), and impaired their migration/invasion capacity in vitro and the metastatic potential of MDA-MB-231 cells in immunodeficient mice. Conversely, transfection of MCF-7 cells to express ROR1 reduced expression of E-cadherin and CK-19, but enhanced the expression of SNAIL-1/2 and vimentin. Treatment of MDA-MB-231 with a monoclonal antibody specific for ROR1 induced downmodulation of vimentin and inhibited cancer cell migration and invasion in vitro and tumor metastasis in vivo. Collectively, this study indicates that ROR1 may regulate EMT and metastasis and that antibodies targeting ROR1 can inhibit cancer progression and metastasis. Cancer Res; 73(12); 3649–60. ©2013 AACR.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...