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)  (4)
  • 2010-2014  (4)
Material
Publisher
  • American Association for Cancer Research (AACR)  (4)
Language
Years
  • 2010-2014  (4)
Year
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Molecular Cancer Therapeutics Vol. 13, No. 7 ( 2014-07-01), p. 1803-1812
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 13, No. 7 ( 2014-07-01), p. 1803-1812
    Abstract: Bispecific antibodies (BsAb) have proven to be useful targeting vectors for pretargeted radioimmunotherapy (PRIT). We sought to overcome key PRIT limitations such as high renal radiation exposure and immunogenicity (e.g., of streptavidin–antibody fusions), to advance clinical translation of this PRIT strategy for diasialoganglioside GD2-positive [GD2(+)] tumors. For this purpose, an IgG-scFv BsAb was engineered using the sequences for the anti-GD2 humanized monoclonal antibody hu3F8 and C825, a murine scFv antibody with high affinity for the chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) complexed with β-particle–emitting radiometals such as 177Lu and 90Y. A three-step regimen, including hu3F8-C825, a dextran-based clearing agent, and p-aminobenzyl-DOTA radiolabeled with 177Lu (as 177Lu-DOTA-Bn; t1/2 = 6.71 days), was optimized in immunocompromised mice carrying subcutaneous human GD2(+) neuroblastoma (NB) xenografts. Absorbed doses for tumor and normal tissues were approximately 85 cGy/MBq and ≤3.7 cGy/MBq, respectively, with therapeutic indices (TI) of 142 for blood and 23 for kidney. A therapy study (n = 5/group; tumor volume, 240 ± 160 mm3) with three successive PRIT cycles (total 177Lu: ∼33 MBq; tumor dose ∼3,400 cGy), revealed complete tumor response in 5 of 5 animals, with no recurrence up to 28 days after treatment. Tumor ablation was confirmed histologically in 4 of 5 mice, and normal organs showed minimal overall toxicities. All nontreated mice required sacrifice within 12 days ( & gt;1.0-cm3 tumor volume). We conclude that this novel anti-GD2 PRIT approach has sufficient TI to successfully ablate subcutaneous GD2(+)-NB in mice while sparing kidney and bone marrow. Mol Cancer Ther; 13(7); 1803–12. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 18_Supplement ( 2011-09-15), p. PR7-PR7
    Abstract: To assess therapeutic response and potential toxicity of oncolytic virotherapy, a noninvasive, deep-tissue imaging modality is needed. This study aimed to assess the feasibility, parameters, determining factors of serial imaging and long-term monitoring of systemic virotherapy together with radiotherapeutic response of pancreatic cancer xenografts treated with a vaccinia virus encoding the human sodium iodide symporter (hNIS), GLV-1 hi 53. hNIS, an intrinsic plasma membrane protein, mediates the active transport, and trapping of iodine mainly in the thyroid and some extrathyroidal tissues such as the stomach. Pancreatic cancer xenografts (PANC-1) in nude mice were treated systemically or intratumorally with GLV-1h153 and serially imaged using 124I-PET at 1, 2, 3, and 5 weeks post virus injection and 4 hours post radiotracer injection. At week 1 and 2 post virus injection, mice were also imaged at 1, 8, 24, 48, and 72 hours post radiotracer injection in order to obtain time-activity curves for dosimetry calculations of radioiodine uptake. The PET signal intensity was compared with tumor therapeutic response and optical imaging. Tumors were histologically analyzed for morphology and presence of virus particles. Autoradiography was performed utilizing technecium-pertechtenate and gamma-scintigraphy to assess determining factors for radiouptake in tumors. Combination therapy with GLV-1h153 and systemic radioiodine was also explored. GLV-1h153 successfully facilitated serial long-term imaging of virotherapy with PET signal intensity correlating to antitumor response. Colonization of tumors with GLV-1h153 mediated radioiodine uptake at potentially therapeutic doses. Successful radiouptake required presence of virus, adequate blood flow, and viable tissue, while loss of signal intensity was linked to tumor death and necrosis. Finally, combining systemically administered GLV-1h153 and 131I led to enhanced tumor kill when compared to virus or 131I treatment alone (P & lt;0.001). GLV-1h153 is thus a promising oncolytic agent for the treatment, long-term imaging, and monitoring of the therapeutic response of cancer. GLV-1h153 provided insights into tumor biological activity and facilitated enhanced tumor kill when combined with systemic targeted radiotherapy. Further investigation into parameters and potential synergistic effects of combination therapy is warranted. This abstract is also presented as Poster C7. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the Second AACR International Conference on Frontiers in Basic Cancer Research; 2011 Sep 14-18; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2011;71(18 Suppl):Abstract nr PR7.
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2069-2069
    Abstract: Introduction: STEAP1 (Six Transmembrane Epithelial Antigen of Prostate 1) is a cell-surface therapeutic target overexpressed in 85% of prostate cancers. Zr-89 MSTP2109A, a humanized antibody targeting STEAP1 conjugated with desferrioxamine and radiolabeled with positron emitter Zr-89 (T1/2 78 h) was developed to enable imaging of STEAP1 expressing tumors. Methods: To evaluate safety, pharmacokinetics (PK), biodistribution and tumor uptake a prospective Phase I/II PET imaging study in mCRPC patients is ongoing. Patients with STEAP1 positive immunohistochemistry (IHC) in archival tissue were studied following 5.0 + 0.2 mCi IV injection of Zr-89 MSTP2109A (immunoreactivity 96 + 2%) . PET/CT images were compared with contemporaneous bone, CT and when available FDG scan findings on a lesional basis. Results: 15 patients have been studied. Two drug related adverse events included mild rigors and/or chills. The first 6 patients were imaged at four timepoints over a 7 day period and showed that PET imaging tumor uptake increased over time and was optimal at 6d (range 5-9). Based on this, all subsequent patients were imaged on day 6. Median PK showed: Plasma concentration at zero time (Co) 28.2%ID/L; T1/2 beta 197.8h; volume of distribution 3.543L; clearance 19.7 ml/h. All patients had definite Zr-89 MSTP2109A uptake in multiple sites of known disease (bone scan or CT standard-of-reference). All patients had bone and 7 had soft tissue metastases on Zr-89 MSTP2109A. In 10 patients, the Zr-89 MSTP2109A and bone scans detected a similar number of lesions; in 5 patients, the bone scan detected many more lesions. This discordance may represent treated non viable disease, as contemporaneous FDG PET scans tended to concur with the Zr-89 MSTP2109A scans rather than bone scans. In 4 of 7 pts Zr-89 MSTP2109A detected more soft tissue lesions than CT. The mean maximal standardized uptake values (SUVmax) in bone and soft tissue metastasis were 22.5 + 13.5 (range 4.4 to 59.3) and 16.4 + 5.6 (range 9.0 to 24.0) respectively. Correlation of tumor SUVmax and IHC is in progress. 11 of 15 patients underwent 11 biopsies within 30d of study (mean 18 + 9d), 7 were performed in PET positive sites after Zr-89 MSTP2109A imaging, 4 were performed prior to PET imaging; tumor was present in 10 of the 11 biopsies and they were all Zr-89 MSTP2109A positive. Conclusion: Zr-89 MSTP2109A is well tolerated. It shows high and progressively increasing tumor contrast in patients with mCRPC. All sites biopsied (11/11) had increased Zr-89 MSTP2109A uptake, only one biopsy was negative for tumor. Given its excellent tumor uptake additional studies to further characterize Zr-89 MSTP2109A may be pursued, for example, selecting patients for STEAP1 directed therapies, assessing treatment response to STEAP1 directed therapy, or as an early biomarker for metastatic disease. Citation Format: Jorge A. Carrasquillo, Daniel C. Danila, Volkan Beylergil, Joseph A. O'Donoghue, Sarah M. Cheal, Shutian Ruan, Neeta Pandit-Taskar, Josef J. Fox, Stephen E. Fleming, Pat B. Zanzonico, Govind Ragupathi, Serge K. Lyashchenko, Simon P. William, Steven M. Larson, Howard I. Scher, Bernard M. Fine, Michael J. Morris. Initial PET imaging and pharmacokinetic results from a Phase I/II study of Zr-89-labeled anti-STEAP1 antibody in metastatic castrate-resistant prostate cancer (mCRPC) patients. [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 2069. doi:10.1158/1538-7445.AM2014-2069
    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) ; 2014
    In:  Clinical Cancer Research Vol. 20, No. 8 ( 2014-04-15), p. 2182-2191
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 8 ( 2014-04-15), p. 2182-2191
    Abstract: Purpose: The norepinephrine transporter (NET) is a critical regulator of catecholamine uptake in normal physiology and is expressed in neuroendocrine tumors like neuroblastoma. Although the norepinephrine analog, meta-iodobenzylguanidine (MIBG), is an established substrate for NET, 123I/131I-MIBG has several clinical limitations for diagnostic imaging. In the current studies, we evaluated meta-[18F]-fluorobenzylguanidine ([18F] -MFBG) and compared it with 123I-MIBG for imaging NET-expressing neuroblastomas. Experimental Design: NET expression levels in neuroblastoma cell lines were determined by Western blot and 123I-MIBG uptake assays. Five neuroblastoma cell lines and two xenografts (SK-N-BE(2)C and LAN1) expressing different levels of NET were used for comparative in vitro and in vivo uptake studies. Results: The uptake of [18F]-MFBG in cells was specific and proportional to the expression level of NET. Although [18F] -MFBG had a 3-fold lower affinity for NET and an approximately 2-fold lower cell uptake in vitro compared with that of 123I-MIBG, the in vivo imaging and tissue radioactivity concentration measurements demonstrated higher [18F]-MFBG xenograft uptake and tumor-to-normal organ ratios at 1 and 4 hours after injection. A comparison of 4 hours [18F] -MFBG PET (positron emission tomography) imaging with 24 hours 123I-MIBG SPECT (single-photon emission computed tomography) imaging showed an approximately 3-fold higher tumor uptake of [18F]-MFBG, but slightly lower tumor-to-background ratios in mice. Conclusions: [18F]-MFBG is a promising radiopharmaceutical for specifically imaging NET-expressing neuroblastomas, with fast pharmacokinetics and whole-body clearance. [18F] -MFBG PET imaging shows higher sensitivity, better detection of small lesions with low NET expression, allows same day scintigraphy with a shorter image acquisition time, and has the potential for lower patient radiation exposure compared with 131I/123I-MIBG. Clin Cancer Res; 20(8); 2182–91. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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...