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
  • 1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 123, No. 1 ( 2014-01-02), p. 61-69
    Abstract: Zinc-finger nucleases simultaneously and permanently inactivate HIV coreceptors ccr5 and cxcr4 resulting in HIV-resistant CD4+ T cells. These HIV-resistant cells may be used to achieve a functional cure for HIV in humans.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Molecular Therapy, Elsevier BV, Vol. 29, No. 4 ( 2021-04), p. 1625-1638
    Type of Medium: Online Resource
    ISSN: 1525-0016
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2001818-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 8, No. 1 ( 2017-03-02)
    Abstract: Monoclonal antibodies are one of the fastest growing classes of pharmaceutical products, however, their potential is limited by the high cost of development and manufacturing. Here we present a safe and cost-effective platform for in vivo expression of therapeutic antibodies using nucleoside-modified mRNA. To demonstrate feasibility and protective efficacy, nucleoside-modified mRNAs encoding the light and heavy chains of the broadly neutralizing anti-HIV-1 antibody VRC01 are generated and encapsulated into lipid nanoparticles. Systemic administration of 1.4 mg kg −1 of mRNA into mice results in ∼170 μg ml −1 VRC01 antibody concentrations in the plasma 24 h post injection. Weekly injections of 1 mg kg −1 of mRNA into immunodeficient mice maintain trough VRC01 levels above 40 μg ml −1 . Most importantly, the translated antibody from a single injection of VRC01 mRNA protects humanized mice from intravenous HIV-1 challenge, demonstrating that nucleoside-modified mRNA represents a viable delivery platform for passive immunotherapy against HIV-1 with expansion to a variety of diseases.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2580-2580
    Abstract: Abstract 2580 Challenges in developing new agents for infant MLL leukemia are compounded by rarity of cases, inherent drug resistance and unique vulnerabilities of infants to treatment-related morbidity and mortality. An additional limitation is the extent to which preclinical data and adult clinical experience can predict clinical dose-exposure and exposure-response relationships and guide dosing strategies (dose, duration, combination, schedule) and study designs (sample size, sampling scheme). Pharmacometric approaches are increasingly being used to quantify drug action and clinical performance. These require a continuum of models with hypotheses and assumptions and pharmacologic rationale to be challenged by targeted experimentation and subsequent model refinement. Key assumptions and experimental conditions are reassessed to optimize experimental designs and test evolving understanding about drug behavior and disease biology. Here we establish “proof-of-concept” quantitative pharmacology relationships between data derived from in vitro obatoclax activity in cell lines and patient samples, historical adult experience and a preclinical diseased mouse model to optimize strategies to treat infant leukemia. Obatoclax in vitro activity was quantified by MTT assays in MLL-rearranged cell lines and primary infant ALL and bilineal MLL leukemia samples. Obatoclax biodistribution was characterized in MLL leukemia-bearing mice to evaluate its potential to achieve target exposures in infants with leukemia by the following steps: 1) A nonlinear mixed-effect model based on existing obatoclax toxicokinetic data in healthy Balb/C mice guided design of a pilot study in diseased NOD-scid-IL-2Rgnull (NSG) mice where target exposure requirements were based on the AUC24h associated with peak oligonucleosomal DNA release as an apoptosis biomarker in an adult Phase I CLL trial (O'Brien 2008) and single-agent MTT assays of obatoclax in MLL cell lines and primary infant leukemia samples. 2) NSG MLL-ENL bilineal leukemia bearing mice with established xenografts received a single IV bolus dose of obatoclax (1.2 or 4.8 mg/kg; 100% or 400% of predicted target exposure). 3) Obatoclax was measured in the plasma, spleen, liver, kidney and brain for 24 h after drug administration (courtesy GeminX). A physiologically-based pharmacokinetic model (PBPK) model was developed to predict exposures in mouse target tissues based on obatoclax physicochemical properties and then scaled to a human infant. Preclinical in vitro experiments suggested IC50 values between 13 and 918 nM in primary MLL leukemia cells. Population mean (% RSE) parameter estimates for plasma clearance (CL), intercompartmental clearance (Q), plasma distribution volume (Vd) and peripheral distribution volume (V2) were 5.1 (10.1) L/h/kg, 1.78 (37) L/h/kg, 9.15 (10.2) L/kg and 17.3 (23.8) L/kg, respectively. PK parameters were close to those predicted from healthy Balb/C mice except that Cmax was lower, possibly suggesting a larger Vd in mice with disease. The half-life was almost 4-fold higher suggesting slower elimination due to mouse strain or disease burden. There was sustained drug retention in spleen, liver, kidney and brain. Brain:plasma were ∼2-10:1, higher than the 〈 1:1 typical of most drugs, indicating excellent CNS penetration. The PBPK model for obatoclax in the mouse compared well with observed biodistribution in diseased NSG mouse tissues (Fig 1). Though brain penetration was good, deviations between observed and PBPK-simulated exposure values may suggest involvement of transporters. PBPK simulation of obatoclax administration to a 1 yr, 10 kg human infant suggested that adequate drug exposure to target organs should be achievable at clinically relevant doses (Fig 2). This work links models that define obatoclax in vitro exposure targets with in vivo exposure obtained in leukemia bearing mice. Predictability of these models relative to infant leukemia will be assessed following completion of the ongoing COG phase I obatoclax trial in relapsed/refractory pediatric cancers (ADVL0816). Our analyses imply that projecting outcome in treating infant MLL disease with obatoclax will be possible once these models are further refined.Fig 1PBPK simulations relative to observed obatoclax biodistribution in the NSG MouseFig 1. PBPK simulations relative to observed obatoclax biodistribution in the NSG MouseFig 2PBPK-simulated obatoclax plasma and tissue exposure in a 1 year old receiving 20 mg/m2 infusion over 3 hFig 2. PBPK-simulated obatoclax plasma and tissue exposure in a 1 year old receiving 20 mg/m2 infusion over 3 h 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: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 673-673
    Abstract: Abstract 673 Inhibition of lymphocyte trafficking early after allogeneic stem cell transplantation (SCT) could limit T cell interactions with antigen-presenting cells and migration to target tissues. This represents a novel strategy to prevent GvHD without interfering with GvL activity. CCR5 is a chemokine receptor expressed on effector T-cells and immature dendritic cells and binds 3 ligands - CCL3, CCL4 and RANTES (CCL5). Accumulating evidence from animal models and clinical observations implicates CCR5 as pivotal in the pathogenesis of GvHD. Genomic analyses suggest that the same CCR5 polymorphisms that confer resistance to HIV infection also correlate with a lower susceptibility to acute GvHD. Maraviroc (MVC; Selzentry®, Pfizer) is the first oral CCR5 antagonist in clinical use. We hypothesized that modulating T-cell trafficking early after allogeneic SCT via CCR5 blockade would limit GvHD. We therefore performed preclinical and clinical testing of MVC as GvHD prophylaxis. Our goals were to 1) determine in vitro activity of MVC on chemotaxis, 2) determine the feasibility, safety and appropriate dose of MVC as part of GvHD prophylaxis, and 3) demonstrate biological activity of MVC through immune pharmacodynamic assays. In vitro, MVC fully inhibited CCR5 internalization by CCL3 and RANTES even at concentrations as low as 1 μM. Using RANTES as a chemotactic trigger, MVC caused dose-dependent inhibition of lymphocyte chemotaxis by up to 53% at MVC 1mM. To address concerns that MVC might impair hematopoiesis, we demonstrated that CCR5 was not expressed on the surface of bone marrow- and peripheral blood-derived CD34+ cells. Moreover, when CD34+ cells were plated in methylcellulose, formation of CFU-GEMM and CFU-GM was not affected by the presence of MVC 1μM; CFU-E and BFU-E were slightly decreased compared to controls. Based on these and other data, we enrolled 19 pts in a phase I/II study of reduced intensity conditioned allogeneic SCT with MVC GvHD prophylaxis. Pts received fludarabine 120mg/m2 and IV busulfan 6.4 mg/kg followed by peripheral blood stem cells from matched related (n=6), matched unrelated (n=10) and 1-antigen mismatched unrelated (n=3) donors. In addition to standard GvHD prophylaxis with tacrolimus and methotrexate, MVC at escalating dose levels was given from day -2 to +30. Median age was 63 (range 21–74). Indications for SCT were AML/MDS (9), NHL (4), myelofibrosis (2), CLL, aplastic anemia, Hodgkin lymphoma and myeloma (1 each). Pharmacokinetic analysis on 6 pts at each dose revealed that the 300 mg and 150 mg bid dose levels resulted in mean Cavg of 536 and 118 ng/ml, respectively. 3/6 patients at 150mg did not reach the targeted minimum Cavg (100 ng/ml), while the 300mg dose level resulted in adequate Cavg in 6/6 patients and was used as the phase II dose. MVC was well tolerated; 3 pts did not complete the entire course because of transient LFT abnormalities (1) or mucositis (2). The median time to ANC 〉 500/μL was 15 d (range 10–21) and to platelets 〉 20k/μL was 13 d (range 11–24) with no graft rejections. The median donor chimerism at day 100 was 97% (range 83–100%). A day 100-landmark analysis in evaluable pts demonstrated that the cumulative incidence of acute GvHD grade 2–4 was 27% (grade 3–4; 9%) in this high-risk population. Importantly, by day 100 all cases of acute GvHD involved only the skin without liver or intestinal involvement. At a median follow up of 186 days, 3/19 patients relapsed (2 AML, 1 NHL) and 6/19 patients died (3 disease-related, 1 neutropenic sepsis, 1 SOS, 1 unrelated). There were no GvHD-related deaths. To explore potential mechanisms, we tested the capacity of patient serum to inhibit CCR5 internalization and chemotaxis. Patient serum from multiple time points (trough, 1, 2, 3, 4, 6 hr post dose) effectively prevented internalization of CCR5 by RANTES. In addition, in vitro chemotaxis of normal donor T-cells in response to RANTES was significantly impaired in the presence of patient serum from day 0 (on MVC) as compared to day 60 (off MVC). In summary, inhibition of lymphocyte trafficking to peripheral tissues represents a novel strategy to modulate and possibly reduce acute GvHD in allogeneic SCT. MVC at 300mg bid was well tolerated and biologically active in pharmacodynamic assays. Patients receiving MVC exhibited limited GvHD by day 100 without excessive relapses. The phase II portion of the trial is ongoing. Disclosures: Off Label Use: Off label use of maraviroc (Selzentry) will be discussed. Frey:Pfizer, Inc.: Speakers Bureau. Vonderheide:Pfizer, Inc.: Research Funding. Porter:Pfizer, Inc.: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 24, No. 36 ( 2006-12-20), p. 5725-5734
    Abstract: Tumor immunosurveillance influences oncogenesis and tumor growth, but it remains controversial whether clinical failure of immunosurveillance is a result of lymphocyte dysfunction or tumor escape. In this study, our goal was to characterize the physiology of tumor immunosurveillance in children with high-risk neuroblastoma (HR-NBL). Patients and Methods Immunohistopathologic studies were carried out on 26 tumor samples from a cohort of HR-NBL patients diagnosed at Children's Hospital of Philadelphia for the 2-year period from May 2003 to May 2005. Blood from nine HLA-A2 + patients in this cohort was analyzed for T cells specific for the antiapoptotic protein survivin. Results Survivin protein was expressed by 26 of 26 tumors. In HLA-A2 + patients, circulating cytotoxic T lymphocytes (CTLs) specific for survivin were detected by peptide/major histocompatibility complex tetramer analysis in the blood of eight of nine children with HR-NBL at the time of diagnosis. Rather than being selectively rendered anergic in vivo, circulating survivin-specific CTLs were highly functional as shown by cytotoxicity and interferon gamma enzyme-linked immunospot assays in six of nine patients. Survivin-specific CD107a mobilization by T cells was found in five of five patients. By immunohistochemistry, tumor-infiltrating T cells were few or absent in 26 of 26 tumors. Conclusion Children with HR-NBL harbor robust cellular immune responses to the universal tumor antigen survivin at the time of diagnosis, but intratumoral T cells are strikingly rare, suggesting a failure of cellular immunosurveillance. Efforts to develop novel therapies that increase T-cell trafficking into tumor nests are warranted.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2006
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2021-01-12)
    Abstract: Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34 + cells and implanting autologous thymus in immune-deficient NOD- scid IL2Rγ null (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3 + Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8 + /Granz B + T cells homeostasis are observed in tumor regions where FOXP3 + Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 23, No. 8 ( 2017-04-15), p. 1955-1966
    Abstract: Purpose: Immunotherapy is an emerging paradigm for the treatment of cancer, but the potential efficacy of many drugs cannot be sufficiently tested in the mouse. We sought to develop a rational combination of motolimod—a novel Toll-like receptor 8 (TLR8) agonist that stimulates robust innate immune responses in humans but diminished responses in mice—with pegylated liposomal doxorubicin (PLD), a chemotherapeutic that induces immunogenic cell death. Experimental Design: We followed an integrative pharmacologic approach including healthy human volunteers, non-human primates, NSG-HIS (“humanized immune system”) mice reconstituted with human CD34+ cells, and patients with cancer to test the effects of motolimod and to assess the combination of motolimod with PLD for the treatment of ovarian cancer. Results: The pharmacodynamic effects of motolimod monotherapy in NSG-HIS mice closely mimicked those in non-human primates and healthy human subjects, whereas the effects of the motolimod/PLD combination in tumor-bearing NSG-HIS mice closely mimicked those in patients with ovarian cancer treated in a phase Ib trial (NCT01294293). The NSG-HIS mouse helped elucidate the mechanism of action of the combination and revealed a positive interaction between the two drugs in vivo. The combination produced no dose-limiting toxicities in patients with ovarian cancer. Two subjects (15%) had complete responses and 7 subjects (53%) had disease stabilization. A phase II study was consequently initiated. Conclusions: These results are the first to demonstrate the value of pharmacologic approaches integrating the NSG-HIS mouse, non-human primates, and patients with cancer for the development of novel immunomodulatory anticancer agents with human specificity. Clin Cancer Res; 23(8); 1955–66. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 3_Supplement ( 2016-02-01), p. PR07-PR07
    Abstract: Non-small cell lung cancer (NSCLC) is one the most prevalent forms of lung cancer. Lung tumors consist of a heterogeneous population of tumor cells, immune cells and stromal cells. It is critical to assess the nature and localization of immune effector cells and their expression of immune checkpoint molecules to develop more effective therapeutic strategies. In this study, we used immunohistochemistry (IHC) to assess the localization of human immune cells and expression of immune checkpoint molecules to define the microenvironment composition. Furthermore, we developed a mouse xenograft model of human lung tumor using BLT-NSG mice with a reconstituted human immune system and A549 cells. Formalin-fixed paraffin embedded NSCLC lung tumor sections (n=28 adenocarcinoma, n=16 squamous) were processed by IHC to assess expression and localization of CD4, CD8, MPO, Foxp3, CD68 and cytokeratin, as well as immune checkpoint molecules (PD-1, Tim-3, LAG-3) and ligands (PD-L1 and VISTA). For the xenograft model, BLT-NSG mice (NSG mice transplanted with fetal liver CD34+ cells and autologous thymus) at 12-16 weeks post-transplant were injected with A549-luciferase (A549-Luc) cells via transpleural or intravenous injection. Weekly bioluminescence imaging was done to assess tumor growth. Lungs and spleens were harvested and enzymatically dissociated for flow cytometric analysis of human effectors and immune checkpoint molecules. We found that significantly more human effectors are localized to the stromal compartment (cytokeratin-) than within the cytokeratin+ tumor nests. The stromal compartment contained 12-fold more CD8+ T-cells, 23-fold more CD4+ T-cells, and 11-fold more regulatory T-cells (Foxp3+). Two-fold more neutrophils (MPO+) and macrophages (CD68+) were found in stroma outside of tumor nests. There were significantly more CD8+ T-cells (p & lt;0.05) and MPO+ neutrophils (p & lt;0.01) localized within the cytokeratin+ areas of squamous compared to adeno tumors. PD-1, Tim-3 or LAG-3 IHC showed that 100-fold more PD-1+ cells are found within the tertiary lymphoid structures in the stromal compartment. 19-fold more Tim-3+ cells, both lymphocytes and macrophages, are found in the stromal compartment. LAG-3+ cells are lymphocytes, and localized mostly to the stromal compartment (11-fold more than in cytokeratin+ areas). PD-L1 IHC showed that 12/16 (75%) of squamous tumors and 14/28 (50%) of adeno tumors express PD-L1 on tumor cells, and H-Scores of the PD-L1 staining from squamous tumors were significantly higher than the adeno tumors (p=0.019). A majority of macrophages expressed PD-L1, but the expression level varied from sample to sample (1+ to 3+ scoring). VISTA IHC showed that lymphocytes, neutrophils and macrophages expressed VISTA, and 26-fold more VISTA+ cells are found in the stromal compartment. In our A549 xenograft model study, NSG and BLT-NSG mice with IV-injected A549 cells showed tumor growth differences by bioluminescence imaging. Flow cytometric analyses showed an increased percentage of non-naïve CD8+ (88%) and non-naïve CD4+ (97%) cells compared to spleens (44% non-naïve CD8+ cells and 74% non-naïve CD4+ cells). There was an increased expression of LAG-3 in both human CD8+ (19-90%) and human CD4+ (9-61%) non-naïve T-cells in the lungs, but LAG-3+ cells in the spleen were minimal ( & lt;6%). PD-1 expression was high in non-naïve T-cell population in both lungs and spleen ( & gt;70%). In addition, there was an up-regulation of PD-L1+(4%) and VISTA+ (12%) in the human CD14+ population in the lungs. In conclusion, the expression of various immune checkpoint molecules in effector cells may limit their localization within the stromal compartment. Furthermore, our xenograft model was able to recapitulate our observations with human patient samples, and would be a valuable in vivo mouse tool to test new therapeutic agents. Citation Format: Hyun-Jeong Ra, Gisela Brake-Silla, Evgeniy Eruslanov, Michael Sharp, Charuhas Deshpande, Michael D. Feldman, Amy Ziober, Li-Ping Wang, Manuel A. Sepulveda, Linda A. Snyder, Gwenn Danet-Desnoyers. Characterization of human effectors in the tumor microenvironment of NSCLC patients and lung tumor xenograft models. [abstract]. In: Proceedings of the Fourth AACR International Conference on Frontiers in Basic Cancer Research; 2015 Oct 23-26; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2016;76(3 Suppl):Abstract nr PR07.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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...