Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2019
    In:  Journal of Cancer Research and Clinical Oncology Vol. 145, No. 6 ( 2019-6), p. 1461-1469
    In: Journal of Cancer Research and Clinical Oncology, Springer Science and Business Media LLC, Vol. 145, No. 6 ( 2019-6), p. 1461-1469
    Type of Medium: Online Resource
    ISSN: 0171-5216 , 1432-1335
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 1459285-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Current Eye Research, Informa UK Limited, Vol. 47, No. 3 ( 2022-03-04), p. 426-435
    Type of Medium: Online Resource
    ISSN: 0271-3683 , 1460-2202
    RVK:
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2022
    detail.hit.zdb_id: 1483048-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2680-2680
    Abstract: The protection afforded to leukemic blasts by the bone marrow microenvironment has been identified as an important mechanism of chemoresistance. Interaction between stroma-derived growth factor-1 alpha (SDF-1α) and its receptor, CXC chemoreceptor 4 (CXCR4) are implicated in chemotaxis, homing, and survival/apoptosis of normal and malignant hematopoietic cells in the bone marrow. Preclinical data demonstrates that plerixafor (AMD3100), a CXCR4 antagonist, disrupts tumor-stroma interactions and mobilizes leukemia cells from their protective stromal environment. Combinations of CXCR4 antagonists with chemotherapy have demonstrated preclinical synergy. Chemosensitization using plerixafor prior to cytotoxic chemotherapy has been tested in adults with acute leukemia. We report the first Phase I study of plerixafor (NCT01319864) delivered prior to chemotherapy in children with relapsed/refractory acute leukemia and MDS. Study Design Patients 〉 3 and 〈 30 years of age with relapsed or refractory AML, ALL, MDS or mixed phenotype acute leukemia were eligible for enrollment. Plerixafor was administered intravenously (IV) once daily followed 4 hours later by cytarabine (1 gm/m2 every 12 hours) and IV etoposide (150 mg/m2 daily) for a total of 5 days of therapy. Plerixafor pharmacokinetic studies were performed on days 1 and 5. Correlative biology studies included measurement of peripheral blood mobilization of leukemic blasts by flow cytometry, quantitative expression of CXCR4 on leukemic blasts, and the change in surface expression of CXCR4 on residual blasts after course 1 of therapy. Results Eighteen evaluable patients (11 AML, 6 ALL, 1 MDS) were treated at 4 dose levels of plerixafor (6, 9, 12, and 15 mg/m2/dose) utilizing a Rolling 6 design. The median number of prior regimens was 2.8 (range 1-7) for ALL and 2.1 (range 1-4) for AML. Six patients had high risk cytogenetics (3 ALL, 2 AML, 1 MDS). Three patients with ALL and 4 with AML had prior hematopoietic stem cell transplant (HSCT). Toxicities were consistent with intensive relapsed leukemia regimens. The most common Grade 1 and 2 toxicities attributed to plerixafor occurring in 〉 10% of patients were anorexia, nausea, vomiting, diarrhea, fatigue, and dizziness. There were no dose limiting toxicities and no delay in count recovery attributable to plerixafor. There were responses in 3 (2 complete response (CR), 1 complete response with incomplete hematologic recovery (CRi)) of 11 AML patients (27%) and no responses in those with ALL or MDS. Peripheral leukemia-specific blast counts (measured by flow cytometry before and 4 hours after the first dose of plerixafor) demonstrated mobilization of leukemic blasts in 14 of 16 patients with samples available, with median fold increase of 3.4 (range 1.3 to 17). The degree of leukemic blast mobilization correlated positively with quantitative leukemia blast surface CXCR4 protein expression (expressed as median fluorescence index relative to isotype control), with a Pearson’s correlation co-efficient of 0.56, p=0.02. Mean ± SD plerixafor AUC values at 12 and 15 mg/m2 were 5074 ± 380 and 5732 ± 573 ng*h/mL, respectively. Drug clearance was similar between days 1 and 5 (p=0.195). Conclusion The favorable safety profile of plerixafor and biologic rationale demonstrated in this clinical trial support further clinical study of chemosensitization using CXCR4 antagonists in overcoming chemoresistance. Disclosures: Off Label Use: Plerixafor is not approved for chemosensitization in the treatment of acute leukemia.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 2725-2725
    Abstract: Introduction: Polyamines (PAs) are cationic metabolites that enhance pro-tumorigenic cellular processes including the stimulation of cell division and proliferation, pro-survival gene expression, DNA and protein synthesis, regulation of apoptosis, oxidative stress and angiogenesis. Spermidine is a particularly important polyamine because it acts as an essential substrate of hypusine biosynthesis. Hypusination is required for the post-transcriptional activation of eukaryotic initiation factor 5A (eIF5A) which is critical to cell growth and protein synthesis. Spermidine production is highly regulated through the rate-limiting enzyme ornithine decarboxylase (ODC). Polyamines and eIF5A have been shown to be critical growth promoters in various pediatric cancers. Thus, targeted inhibition of ODC has therapeutic potential for this unique group of currently incurable pediatric malignancies. The anti-protozoan drug α-difluoromethylornithine (DFMO) is a suicide inhibitor of ODC and its effect may be potentiated by GC7 and Aurothioglucose. Previous mechanism based studies indicate that DFMO effectively reduces polyamine levels in neuroblastoma (NB) and provides rationale for ongoing Phase I and II clinical trials. To our knowledge, the effects of DFMO, GC7 and aurothioglucose have not yet been tested in pediatric leukemias. In this report, we evaluate the effectiveness of these therapeutic agents as a novel treatment of refractory hematological malignancies in the pediatric population. Methods: The recently published TARGET 2018 dataset was analyzed to determine the relationship between ODC expression and patient survival. Demographic data from this study was used to identify patients populations who may benefit from an ODC targeted therapy. Western blot analyses were then used to screen a comprehensive panel of pediatric cancer cell lines for ODC expression. Infant and pediatric leukemia cell lines were selected to represent the various subtypes including AML (TIB202, KASUMI) as well as B ALL (SUP-B15, MV411, RS4-11, CCRF-SB ) and T ALL (CEM/C1, MOLT-3). These cells were derived from children with relapsed disease and have varying degrees of resistance to conventional chemotherapy. Drug combination studies were used to define drug candidates having potential synergistic activity when combined with DFMO. Results: A Kaplan-Meier evaluation determined that high expression of ODC was associated with significantly lower patient survival and this finding was confirmed via cox regression analysis. The demographic analysis determined that females were significantly overrepresented in the high expression group whereas no variation was seen between race and ethnicity groups. Furthermore, a correlation was observed between high expression of eIF5A and younger diagnosis, indicating that ODC targeted therapy may be more effective in infant populations. DFMO-induced cytotoxicity was detected in many of the tumor cells including infant AML (TIB202) at an approximate physiologically achievable IC50 value of 200 uM. This was representative of values previously published for neuroblastoma. In infant AML, a western blot analysis determined that DFMO increased the abundance of cleaved PARP, cleaved caspase 3 and cleaved caspase 7 which serve as mechanistic markers of apoptosis. Probing for caspase 8 and caspase 9 showed no difference between treated cells and control. A combination screen of over 80 biologically active compounds was conducted to identify potential synergistic treatment partners of DFMO where GC7 and aurothioglucose (AuTG) ranked among the most promising. Further experimentation determined that doses of GC7 and AuTG, which were not effective as a singular treatment, were able to enhance the ability of DFMO to kill tumor cells. Discussion: The preclinical studies discussed here offer the first proof-of-concept data on a novel treatment approach for refractory leukemia in children. We provide mechanistic evidence to show the ability of DFMO to effectively kill polyamine-dependent tumor cells at physiologically attainable concentrations. In addition, effective drug combinations have been identified to further enhance their clinical utility. Our findings provide key preclinical information on the utility of these drugs as treatment for refractory hematological malignancies in the pediatric population. 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: 2018
    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
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2020
    In:  Neurology Vol. 95, No. 9 ( 2020-09-01), p. e1163-e1173
    In: Neurology, Ovid Technologies (Wolters Kluwer Health), Vol. 95, No. 9 ( 2020-09-01), p. e1163-e1173
    Abstract: To examine the relationship between neonatal inflammatory cytokines and perinatal stroke using a systems biology approach analyzing serum and blood-spot cytokines from 47 patients. Methods This was a population-based, controlled cohort study with prospective and retrospective case ascertainment. Participants were recruited through the Alberta Perinatal Stroke Project. Stroke was classified as neonatal arterial ischemic stroke (NAIS), arterial presumed perinatal ischemic stroke (APPIS), or periventricular venous infarction (PVI). Biosamples were stored blood spots (retrospective) and acute serum (prospective). Controls had comparable gestational and maternal ages. Sixty-five cytokines were measured (Luminex). Hierarchical clustering analysis was performed to create heat maps. The Fisher linear discriminant analysis was used to create projection models to determine discriminatory boundaries between stroke types and controls. Results A total of 197 participants were analyzed (27 with NAIS, 8 with APPIS, 12 with PVI, 150 controls). Cytokines were quantifiable with quality control measures satisfied (standards testing, decay analysis). Linear discriminant analysis had high accuracy in using cytokine profiles to separate groups. Profiles in participants with PVI and controls were similar. NAIS separation was accurate (sensitivity 77%, specificity 97%). APPIS mapping was also distinguishable from NAIS (sensitivity 86%, specificity 99%). Classification tree analysis generated similar diagnostic accuracy. Conclusions Unique inflammatory biomarker signatures are associated with specific perinatal stroke diseases. Findings support an acquired pathophysiology and suggest the possibility that at-risk pregnancies might be identified to develop prevention strategies. Classification of evidence This study provides Class III evidence that differences in acute neonatal serum cytokine profiles can discriminate between patients with specific perinatal stroke diseases and controls.
    Type of Medium: Online Resource
    ISSN: 0028-3878 , 1526-632X
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2020
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5393-5393
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5393-5393
    Abstract: Introduction: The activation of stimulator of interferon (IFN) genes (STING), an intracellular receptor system located in the endoplasmic reticulum, has been shown to augment antitumor immunity through the induction of pro-inflammatory cytokines. Currently, a number of STING agonists have been developed to treat refractory malignancies. Our previous studies have identified PV-10 (4,5,6,7-tetrachloro-2',4',5',7'-tetraiodofluorescein) as a novel therapeutic agent with potent activity following intra-tumoral injection. Here we describe a previously unidentified mechanism by which PV-10 may facilitate sustained immune activation and therapeutic antitumor activity. Methods: The well-established acute monocytic leukemia (AML) cell line THP-1 was used as a model to study STING activation in vitro. Cells were treated with PV-10 and the induction of STING was evaluated by Western blot analysis using cGAMP as a positive control. Proteins that associate with STING in the presence of PV-10 were purified by immunoprecipitation and analyzed by mass spectrometry (LC-MS/MS, Mascot database). The culture supernatants from PV-10 treated cells were probed for a panel of 42 immune cytokines using the Bio-Plex multiplex bead-based assay system. Results: We show that the exposure of THP-1 cells to PV-10 leads to the appearance of a new 70-KD STING dimer band detected by specific antibodies. Compared to cGAMP controls no induction of PDL-1 was noted. Mass spectrometric analysis of immuno-precipitates of STING in these cells showed the presence of Heat Shock Proteins (HSP) 60, 70 and 90 as well as Polyadenylate Binding Protein 1 (PABP1) to the dimerized STING complex. The chemokine assays showed specific upregulation of a distinct set of pro-inflammatory and cytotoxic T-cell recruitment cytokines. A peak in the induction of MCP-3 and IFN gamma was seen at 24 hours (2 fold) and an approximately 10-fold increase in IL-6, IL-8 and IP-10 was seen in the 24 hours following exposure to PV-10. A significant increase in MCP-1 levels was also noted. Discussion: The compound PV-10 has been shown to induce tumor necrosis following intra-tumoral injection. Our present data show a possible mechanism of this agent through STING dimerization and HSP association leading to a sustained pro-inflammatory and immune response. We provide experimental data, for the first time, for a role of HSPs in STING-mediated immune activation pathways and the description of an agent that may play a role in effective single-agent immunotherapy or drug combination therapy approaches in future clinical studies. Citation Format: Satbir Thakur, Chunfen Zhang, Laurent Brechenmacher, Luis Murgia-Favela, Aru Narendran. Association of heat shock proteins as chaperone for STING: A potential link in a key immune activation mechanism revealed by the novel anti-cancer agent PV-10 [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5393.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. LB-140-LB-140
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. LB-140-LB-140
    Abstract: BACKGROUND: Brain tumour initiating cells (BTICs) are stem-like cells hypothesized to mediate tumorigenesis and recurrence in glioblastoma multiforme (GBM). Patient-derived BTICs represent highly relevant preclinical GBM models, forming aggressive, infiltrative tumours in nude mice that closely recapitulate the phenotypic heterogeneity of patient GBMs. BTICs have also been shown to possess mechanisms mediating both chemo- and radio-resistance. Recently, success has been demonstrated in treating BTICs with oncolytic virotherapy (OV), which uses replication-competent viruses to specifically target and kill malignant cells. Myxoma virus (MyxV) is a promising oncolytic candidate, which our lab has shown to be highly efficacious in preclinical GBM models, effectively curing GBM xenografts with a single intra-cranial injection. By contrast, long-term survival is not obtained in MyxV-treated, BTIC-tumour-bearing mice, though survival is prolonged. HYPOTHESIS: We hypothesize that MyxV treatment of BTICs can be improved by utilizing clinically relevant chemotherapeutics identified via high-throughput pharmacoviral screens. RESULTS: We utilized a diverse, comprehensive panel of 80 small molecule inhibitors with preclinical and clinical anti-cancer efficacy to screen for synergy with MyxV treatment in vitro. Multiple candidates have been identified and are currently being validated for synergy (Chou-Talalay method) using a panel of genetically distinct, patient-derived BTICs. Our results implicate multiple potential targets for OV combination therapy, including topoisomerase I and the PI-3K/Akt/mTOR pathway. Target validation is currently underway using shRNA knockdowns and additional targeted inhibitors of these pathways. Further, the mechanism of synergistic cell death during these combination treatments is being explored. In vivo experiments utilizing the dual-pronged pharmacoviral approach are also underway. SIGNIFICANCE: We describe the nature and identity of compounds that carry the potential to sensitize BTICs to MyxV infection. Our findings offer an effective avenue to elucidate resistance mechanisms and develop efficacious combination therapies for targeting disease reservoirs within highly refractory GBMs. 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 LB-140. doi:1538-7445.AM2012-LB-140
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Clinical Immunology, Elsevier BV, Vol. 119 ( 2006-1), p. S78-
    Type of Medium: Online Resource
    ISSN: 1521-6616
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2006
    detail.hit.zdb_id: 1462862-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 19, No. 13 ( 2013-07-01), p. 3649-3658
    Abstract: Purpose: Ridaforolimus (MK-8669, AP23573) is a potent and selective mammalian target of rapamycin (mTOR) inhibitor. Preclinically, ridaforolimus displays antiproliferative activity against a variety of human tumors in vitro and tumor xenograft models in vivo, with additive or synergistic activity when combined with other anticancer agents. Antitumor activity has been confirmed in adults. This phase I study determined the safety, pharmacological, biologic, and toxicity profiles of ridaforolimus in pediatric patients with refractory malignancies. Experimental Design: Eligible children ages 1 to 18 years with advanced solid tumors were enrolled in a 3 + 3 dose escalation design, to determine the safety, tolerability, and maximum tolerated dose (MTD)/dose-limiting toxicity (DLT) of ridaforolimus. Toxicities, pharmacokinetics, and pharmacodynamics were characterized. Results: Fifteen patients were treated. No DLT was observed at any dose level tested; therefore, an MTD was not identified. Most adverse events were mild to moderate; the most common grades 3 and 4 adverse events were hematologic, including thrombocytopenia and anemia. Nonhematologic adverse events were mostly electrolyte disturbances. The observed pharmacokinetic profile of ridaforolimus in children was consistent with that previously showed in adults. Pharmacodynamic confirms that the dose range tested has pharmacological/pharmacodynamic activity. Forty percent of patients achieved stable disease including four of six with central nervous system tumors and two of eight with sarcomas. Conclusions: This first-in-pediatrics study shows that the second-generation mTOR inhibitor ridaforolimus is well tolerated in heavily pretreated children with refractory solid tumors. No DLTs were observed over the dose range tested. Ridaforolimus may represent a therapeutic option for use in pediatric malignancies. Clin Cancer Res; 19(13); 3649–58. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 13, No. 6 ( 2007-03-15), p. 1783-1788
    Abstract: Purpose: Heat shock protein 90 (Hsp90) is essential for the posttranslational control of many regulators of cell growth, differentiation, and apoptosis. 17-N-Allylamino-17-demethoxygeldanamycin (17-AAG) binds to Hsp90 and alters levels of proteins regulated by Hsp90. We conducted a phase I trial of 17-AAG in pediatric patients with recurrent or refractory neuroblastoma, Ewing's sarcoma, osteosarcoma, and desmoplastic small round cell tumor to determine the maximum tolerated dose, define toxicity and pharmacokinetic profiles, and generate data about molecular target modulation. Experimental Design: Escalating doses of 17-AAG were administered i.v. over 1 to 2 h twice weekly for 2 weeks every 21 days until patients experienced disease progression or toxicity. harmacokinetic and pharmacodynamic studies were done during cycle 1. Results: Fifteen patients were enrolled onto dose levels between 150 and 360 mg/m2; 13 patients were evaluable for toxicity. The maximum tolerated dose was 270 mg/m2. DLTs were grade 3 transaminitis and hypoxia. Two patients with osteosarcoma and bulky pulmonary metastases died during cycle 1 and were not evaluable for toxicity. No objective responses were observed. 17-AAG pharmacokinetics in pediatric patients were linear; clearance and half-life were 21.6 ± 6.21 (mean ± SD) L/h/m2 and 2.6 ± 0.95 h, respectively. Posttherapy increases in levels of the inducible isoform of Hsp70, a marker of target modulation, were detected in peripheral blood mononuclear cells at all dose levels. Conclusion: 17-AAG was well tolerated at a dose of 270 mg/m2 administered twice weekly for 2 of 3 weeks. Caution should be used in treatment of patients with bulky pulmonary disease.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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