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)  (7)
  • 1
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 4, No. 1_Supplement ( 2016-01-01), p. B120-B120
    Abstract: Previous work from our lab has demonstrated that a combination of anti-tumor antibody and an IL-2 fusion protein that exhibits extended serum half-life elicits an immune response that can effectively control a wide variety of tumor models. We have since then combined this therapeutic regimen with a vaccine exhibiting efficient lymph node trafficking that can generate an impressive population of tumor-antigen specific CD8+ T-cells but by itself does not provide good anti-tumor efficacy. The efficacy of this combination immunotherapy is further boosted by immune checkpoint blockade, leading to a robust four-component therapy: 1) anti-tumor antigen antibody; 2) IL-2 fusion protein; 3) anti-tumor antigen vaccine; 4) anti-PD-1 antibody. Although this four-pronged approach is demonstrably effective in the syngeneic subcutaneous melanoma model B16F10, we wish to test its efficacy in a more physiological model. To this end, we have turned to a model developed by the Jacks Lab, known as the KP model: an inducible lung tumor model where lentivirus-driven integration and expression of Cre is able to activate oncogenic Kras and completely remove p53 function. Because our therapeutic regimen requires a targetable tumor-associated antigen with respect to both the antibody and vaccine, we chose to induce expression of human carcinoembryonic antigen (CEA) in these tumors, as CEA has a well-studied structure and biology, and frequently expresses aberrantly in many forms of human adenocarcinomas. Additionally, our lab has previously engineered an antibody targeting CEA possessing picomolar affinity. Finally, to remove any endogenous immunological response against human CEA as a foreign antigen in our mouse system, we have crossed the KP model with a mouse model transgenic for human CEA, which in the literature has been described to have identical spatiotemporal expression of CEA as found in humans and should allow for central tolerance of this antigen. In the course of this work, we have successfully introduced human CEA into our lentivirus constructs and shown tumorigenesis by these constructs in the KP model coincides with expression of tumor-associated CEA, as detected by qPCR. On the therapeutic side, we have tailored the vaccine to successfully drive an anti-CEA CD8+ T-cell response. Performing preliminary therapeutic experiments in a transplant model of the KP tumor with our four-component therapy, we saw tumor control compared to untreated tumors. Upon interrogating the CD8+ T-cell response against CEA, we found 1-15% of CD8+ T-cells in the blood respond to CEA stimulation by intracellular cytokine staining. With regards to the lung tumor model, in the course of establishing the system we have also observed that the growth kinetics of tumors expressing CEA lags behind those tumors without CEA, even in the transgenic background. Preliminary immunophenotyping work by flow cytometry suggests that tumors with CEA seem to have a reduced myeloid-derived suppressor cell (MDSC) population and a higher CD8a+ dendritic cell (DC) population compared to tumors without CEA, suggesting that the former may have a less immunosuppressive tumor microenvironment that is better able to prime an anti-tumor CD8+ T-cell response. We will be planning to conduct therapeutic trials in the more physiological lung tumor KP model in the near future, as well as investigate the differences in the immune response with tumors expressing or lacking CEA. Citation Format: Eric F. Zhu, Kavya Rakhra, Naveen Mehta, Kelly D. Moynihan, Cary F. Opel, Darrell J. Irvine, K. Dane Wittrup. Characterization and combination immunotherapy treatment of an inducible autochthonous murine lung cancer model expressing human carcinoembryonic antigen (CEA) as a tumor-associated self-antigen. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B120.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 24_Supplement ( 2012-12-15), p. OT1-1-08-OT1-1-08
    Abstract: Background: Lapatinib in combination with capecitabine is a standard of care treatment for ErbB2+ metastatic breast cancer (MBC) patients who have progressed after anthracyclines, taxanes and trastuzumab treatment. Results from the lapatinib pivotal trial showed that the addition of lapatinib to capecitabine increased median time to progression (TTP) even among heavily pre-treated patients (median of 4 prior lines of therapy). A post-hoc exploratory sub-group analysis of this trial suggested that earlier administration of lapatinib-capecitabine in MBC patients who progress after trastuzumab may produce better clinical outcomes. The TYCO study was designed to evaluate if early initiation of lapatinib-capecitabine in patients with ErbB2+ MBC who have progressed on trastuzumab-containing regimen improves TTP in comparison with a delayed start of the therapy. Trial design: TYCO is an international, multicenter, prospective, observational study in 269 ErbB2+ MBC patients whose disease has progressed after treatment with trastuzumab in the metastatic setting. Two cohorts will be compared; Group 1: patients receiving lapatinib-capecitabine just after the first trastuzumab progression, and Group 2: patients receiving lapatinib-capecitabine after two or more lines of treatment after first trastuzumab progression. The study duration is of 12 months with data collection at baseline and approximately every 3 months thereafter. Major Eligibility Criteria: 1. Females ≥18y with confirmed ErbB2+ MBC who have progressed after a previous trastuzumab-containing regimen,2. Pts eligible for standard therapy with lapatinib-capecitabine at approved conventional doses, as per local approved label.3. Pts eligible to start standard treatment with Lapatinib-capecitabine at conventional doses, or receiving standard treatment with Lapatinib-capecitabine at conventional doses, for no longer than 10 weeks from the start of the treatment to the date of inclusion in the study; Aims: Primary objective of this study is to determine if early switch to lapatinib-capecitabine in patients with ErbB2+ metastatic breast cancer who have progressed on trastuzumab containing regimen improves time to disease progression as determined by treating physician either clinically or radiologically. Secondary objectives include overall response rate and overall survival. Statistical Methods: Kaplan-Meier plots will be used to describe the median TTP after start of lapatinib-capecetabine. Cox proportional hazard model will be developed to estimate the adjusted hazard ratio (and 95% confidence intervals) comparing TTP for the two treatment group using propensity score methods (trimmed sample, adjustment for the continuous propensity score measure, and doubly robust adjustment) to adjust for potential confounding by indication that may arise due to the non-randomised design. Present and Target Accrual: Enrollment began in February 2010, and as per May 2012, 266 patients have been included from Turkey, Venezuela, Argentina, Saudi Arabia and Colombia. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr OT1-1-08.
    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) ; 2019
    In:  Cancer Research Vol. 79, No. 4_Supplement ( 2019-02-15), p. P4-06-29-P4-06-29
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 4_Supplement ( 2019-02-15), p. P4-06-29-P4-06-29
    Abstract: Background: Breast cancer is still not curable with a substantial resistance rate in all subgroups. Alterations in immunological mechanisms are assumed to play a role in pathophysiology and potential efficiency of immunotherapy approaches. Understanding the immunological changes in these patients may have major implications as predictive biomarkers for disease progression. The aim of our study was to determine immune subsets and functions in patient blood between treatment naïve locally advanced and metastatic breast cancer at diagnosis and to compare it with multiple time points during and after different treatments. Subjects and Methods: The immunological profile of 25 stage II-III patients who were candidates for neoadjuvant treatment and 27 stage IV treatment naïve patients in two comprehensive oncology clinics (Marmara University and Medeniyet University, Istanbul, Turkey) were analyzed. Age-sex-matched healthy samples (n=26) were collected from volunteers. Peripheral blood mononuclear cells (PBMC) isolated from blood samples were frozen. PBMC were thawed and stained using multi parameter antibodies for immune profiling using flow cytometry in Jackson Laboratory, Farmington, CT. Results: Differences between T cell subsets among patients (metastatic and locally advanced group separately) and healthy controls were assessed. We found significant differences (all p values & lt;0.01) in inflammatory and regulatory T cell subsets both among the two patient groups (metastatic vs locally advanced untreated)and vs healthy controls, at first time point blood samples: 1) Increase in memory CD4+ T cells (CD45RO+) proportions in both metastatic and locally advanced groups (2) Increase in central and effector CD8+ memory (CD45RO+ or CD45RO-CCR7-) T cells only in metastatic group compared to healthy and locally advanced group, 3) Increase regulatory T cells (Tregs) only in locally advanced group compared to healthy and metastatic patients, 4) Perturbations in proinflammatory Th17 cells in both patient groups compared to healthy controls. More extensive immune profiling of these groups and comparison of different time points during- and post-treatment and correlation with clinical data will be presented. Conclusions: Our results reveal significant differences in potential T cell activation and regulation in locally advanced and metastatic breast cancer patients, suggesting complex immune response at different disease stages. These findings have implications for as predictive indicators for disease progression for development of future immunotherapy strategies. Citation Format: Sezen BA, Koca S, Alan O, Renzullo S, Ozdemir FT, Kozhaya L, Telli TA, Karhan E, Ugurlu U, Gulluoglu B, Dane F, Yumuk FP, Unutmaz D. Prospective immune-profiling of locally advanced and metastatic breast cancer patients [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P4-06-29.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2696-2696
    Abstract: E3 ubiquitin ligases play critical roles in directing cellular protein fate by controlling the specificity of ubiquitin conjugation to substrate proteins and targeting them for cellular relocalization or degradation by the ubiquitin proteasome system. The E3 ubiquitin ligase CBL-B is expressed in immune cell lineages and negatively regulates activity of the T-cell receptor (TCR) by imposing a requirement for a costimulatory signal to mount a productive immune response upon TCR engagement. Mice deficient in Cbl-b, and more specifically in the RING Zn-finger ligase domain of Cbl-b, demonstrate a tumor rejection phenotype mediated by CD8+ T cells (Paolino et al., JI, 2011). We have reproduced these results and demonstrate that Cbl-b deficient mice show enhanced anti-tumor activity. In addition, we show that CD4+ and CD8+ T cells from mice deficient in Cbl-b have 5 to 10-fold enhanced secretion of IL-2 and IFN γ when stimulated ex vivo. These data provide a genetic rationale for the development of a small molecule inhibitor of CBL-B ligase activity for use in patients with tumor-mediated immune suppression of effector T cells. We have identified a series of small molecule inhibitors of CBL-B activity with biochemical potency at low nanomolar concentrations. CBL-B inhibitors increased cytokine secretion in vitro at low nanomolar concentrations, as measured by IL-2 and IFN γ secretion, in primary human and mouse T cells stimulated with CD3/CD28 or CD3 alone. The compounds also stimulated proliferation and elevated levels of the T cell surface activation markers CD25 and CD69. CBL-B inhibitors enhanced an antigen recall response in human PBMCs ex vivo, as measured by approximately 5-fold higher secretion of GM-CSF, TNF-α and RANTES, and demonstrated effects in an ex vivo model of exhausted T cell function. Oral dosing of an optimized CBL-B inhibitor enhanced anti-CD3 stimulated T cell activation in mouse CD4+ and CD8+ T cells, demonstrating a dose proportional pharmacodynamic effect. Oral administration over 28 days in the syngeneic CT-26 tumor model was well tolerated and resulted in single agent tumor growth inhibition. These data support the continued advancement of small molecule oral CBL-B inhibitors for future development in immuno-oncology. Citation Format: Jennifa Gosling, Ryan Rountree, Asad Taherbhoy, Chenbo Wang, Thomas Cummins, Frederick Cohen, Hiroko Tanaka, Dahlia Weiss, Mario Cardozo, Christopher Karim, May Tan, Joseph Juan, Austin Tenn-McClellan, Szerenke Kiss von Soly, Julie Sheung, Kathleen Boyle, Ketki Dhamnaskar, Katherine Kurylo, Jilliane Bruffey, Jennifer McKinnell, Dane Karr, Andria Christianson, Anne-Renee Van Der Vuurst de Vries, Pallavur Sivakumar, Mark Gallop, Paul A. Barsanti, Anjanabha Saha, Neil F. Bence, Christoph W. Zapf. Genetic and pharmacologic evaluation of the ubiquitin ligase CBL-B as a small-molecule, tumor immunotherapy target [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2696.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 11, No. 6 ( 2023-06-02), p. 763-776
    Abstract: Glioblastoma (GBM) is the most common malignant brain tumor in adults, responsible for approximately 225,000 deaths per year. Despite preclinical successes, most interventions have failed to extend patient survival by more than a few months. Treatment with anti—programmed cell death protein 1 (anti–PD-1) immune checkpoint blockade (ICB) monotherapy has been beneficial for malignant tumors such as melanoma and lung cancers but has yet to be effectively employed in GBM. This study aimed to determine whether supplementing anti–PD-1 ICB with engineered extended half-life IL2, a potent lymphoproliferative cytokine, could improve outcomes. This combination therapy, subsequently referred to as enhanced checkpoint blockade (ECB), delivered intraperitoneally, reliably cures approximately 50% of C57BL/6 mice bearing orthotopic GL261 gliomas and extends median survival of the treated cohort. In the CT2A model, characterized as being resistant to CBI, ECB caused a decrease in CT2A tumor volume in half of measured animals similar to what was observed in GL261-bearing mice, promoting a trending survival increase. ECB generates robust immunologic responses, features of which include secondary lymphoid organ enlargement and increased activation status of both CD4 and CD8 T cells. This immunity is durable, with long-term ECB survivors able to resist GL261 rechallenge. Through employment of depletion strategies, ECB's efficacy was shown to be independent of host MHC class I–restricted antigen presentation but reliant on CD4 T cells. These results demonstrate ECB is efficacious against the GL261 glioma model through an MHC class I–independent mechanism and supporting further investigation into IL2-supplemented ICB therapies for tumors of the central nervous system.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. P2-16-18-P2-16-18
    Abstract: Background: Breast cancer is the most common type of cancer among women in the world. Neoadjuvant chemotherapy is the standard treatment modality in locally advanced disease. Pathological complete response (pCR) is a significant prognostic factor for prolonged disease-free and overall survival. Insulin resistance is defined as a pathological condition in which insulin effect is impaired in peripheral target tissues such as skeletal muscle, liver, and adipose tissue. The relationship between breast cancer and insulin resistance is controversial. In this study, our aim is to evaluate the role of insulin resistance to predict complete response in locally advanced breast cancer patient who underwent neoadjuvant treatment. Method: Data from 55 non-diabetic, locally advanced breast cancer patients, treated with neoadjuvant chemotherapy between 2015-2017, were retrospectively evaluated. Demographic and clinicopathologic findings were analyzed. HOMA-IR was calculated by using obtained insulin and fasting blood glucose values before neoadjuvant chemotherapy (Fasting insulin x Fasting glucose / 405). We considered cut-off of 2.5 for insulin resistance. All patients received taxane and anthracycline-based neoadjuvant chemotherapy and then were operated within an average of 4-10 weeks. Findings were analyzed using SPSS. BMI, menopausal status, clinical stage, pathological receptor subtype, Ki-67 index, grade, and insulin resistance have been assessed the effects on pathological complete response status by the Logistic Regression. Results: At the time of diagnosis, the median age was 48.5 (min 27-max 80) years. Twenty-five patients had no insulin resistance. Most common pathologic subtype was hormone receptor positive, Her-2 negative invasive ductal carcinoma. Baseline demographics and disease characteristics are outlined in Table 1. Sixteen (29%) patients had pCR. Of these 16 patients with pCR, 5 (16%) had insulin resistance and 11 (44%) did not. There was a statistically significant difference between the two groups. Insulin resistance was significantly associated with lower pCR rate. We found that patients with insulin resistance had a 3.9 times lower probability of pCR than patients without insulin resistance [OR: 3.9, 95%CI:(1.1 - 13.6); p=0.02] . Conclusion: Our results reveal that insulin resistance might be a predictive biomarker for pCR in breast cancer who underwent neoadjuvant treatment. Molecular mechanisms for these associations are not well known, therefore more comprehensive and prospective studies are needed. Table 1: Baseline demographic and clinical findingsAll patients (n=55)Insulin ResistancepYes (n=30)No (n=25)Age (median) (min-max)48.5 (27-80)52 (29-80)45 (27-66)0.06BMI (median) (min-max)29.6 (18.92-43.4)33.25 (18.92-43.4)27.01 (21-35.2)0.015Waist circumference100 (56-116)100 (56-116)89 (56-106)0.7Hip circumference108 (58-134)110 (58-134)106 (95-120)0.5Fasting Glucose Level99 (72-126)103 (82-126)91 (72-115)0.003HDL57 (24-96)55 (24-83)59 (40-96)0.07LDL124 (51-219)120 (51-198)139 (61-219)0.6Total Cholesterol215 (117-314)205 (117-298)218 (127-314)0.5Trigliseride98 (56-467)121 (58-467)89 (56-164)0.007White Blood Cell6700 (3100-11100)7550 (4100-11100)6100 (3100-8900)0.004Sedimentation Rate31 (8-78)33 (9-58)27 (8-78)0.2CRP3.4 (1.59-43)4.3 (2-43)3.3 (1.5-12.5)0.01Menopause Status (n) (%)Premenopause32 (58%)16 (53%)16 (69%)0.4Postmenopause23 (42%)14 (47%)9 (31%)Clinical Stage2A9 (16%)7 (23%)2 (8%)0.22B18 (32%)10 (33%)8 (32%)3A20 (36%)8 (27%)12 (48%)3B7 (14%)5 (17%)2 (8%)3C1 (2%)01 (4%)Pathologic subtypeInvaziv ductal44 (80%)26 (87%)18 (72%)0.8Invaziv lobular5 (9%)1 (3%)4 (16%)Others6 (11%)3 (10%)3 (12%)Receptor StatusHR+, HER2-31 (56%)17 (56%)14 (56%)0.7HR-, HER 2 +7 (13%)5 (16%)2 (8%)HR+, HER2+9 (16%)4 (14%)5 (20%)HR-, HER2-8 (15%)4 (14%)4 (16%)Ki-67 index≤ 1513 (23%)11 (37%)2 (8%)0.01 & gt; 1537 (67%)19 (63%)23(92%)GradeGrade 111 (20%)11 (37%)0 & lt;0.01Grade 228 (%51)16 (53%)12 (48%)Grade 316 (29%)3 (10%)13 (52%)Pathological responseCR16 (29%)5 (16%)11 (44%)0.02Non-CR39 (71%)25 (87%)14 (56%) Citation Format: Ozkan Alan, Tugba Akin Telli, Bilge Aktas, Sinan Koca, Ilker N Ökten, Tugba Basoglu Tuylu, Nazim C Demircan, Rukiye Arikan, Ozlem Ercelep, Mustafa U Ugurlu, Handan Kaya, Serap Kaya, Nalan Akgul Babacan, Faysal Dane, Perran F Yumuk. Does insulin resistance predict complete response in breast cancer patients who underwent neoadjuvant treatment? [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P2-16-18.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1250-1250
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1250-1250
    Abstract: Activating and inhibitory Fc gamma receptors (FcγRs) can play a large role in the efficacy of certain antibody therapeutics. The Fc regions of the antibody bind to the FcγRs expressed on immune cells and modulate responses against antibody-coated tumor cells. Previous studies with FcγR knock-out mice have shown that antibody-mediated tumor clearance is improved when activating FcγRs are bound preferentially over inhibitory FcγRs. The ectodomains of FcγRs are highly homologous, making it challenging to engineer Fc regions to specifically bind individual FcγRs with high affinity and lack binding to the other FcγRs. To address this challenge, we engineered the human tenth type III fibronectin (Fn3) domain scaffold to bind individual murine FcγRs at epitopes that are distinct from the Fc binding region for testing in a mouse model. The most established pre-clinical models of cancer are mouse models; therefore studying the effect of modulating individual murine FcγRs should provide insight into engaging specific FcγRs as an anti-cancer therapeutic for humans. Using directed evolution with yeast surface display, we isolated Fn3 clones specific for murine FcγRI, FcγRII, FcγRIII, and FcγRIV through a combination of depletion, enrichment, and mutagenesis steps. Cell titrations measured with flow cytometry confirmed the binding specificity of the Fn3s. Binding epitopes were determined by fluorescence activated cell sorting of a single-point mutant yeast library of FcγR and competition studies. To confirm the biological activity of the Fn3 clones, a tumor-targeting single-chain variable fragment (scFv) was fused to each Fn3 clone. Each scFv-Fn3 construct was antigen specific and bound specifically to the FcγR that it was designed to target. The murine colon carcinoma cell line MC38 stably transfected with carcinoembryonic antigen (CEA) was used for our studies. Tumor cells were pre-incubated with scFv-Fn3 constructs targeting CEA and then combined with thioglycollate- induced peritoneal macrophages pre-treated with interferon gamma. The phagocytic activity of the macrophages was measured using flow cytometry. Pre-incubation with scFv-Fn3 constructs specific for FcγRI or FcγRIV significantly increased phagocytosis compared with pre-incubations with the control scFv-Fn3 construct. Pre-incubation with a combination of both the FcγRI- and FcγRIV-specific constructs resulted in a phagocytosis level close to that of murine IgG2a, which can interact with all of the FcγRs. These Fn3 tools will allow us to conduct future studies on the immune response that is generated by triggering individual FcγRs in other in vitro assays and in vivo models of cancer. This work was supported by a Sanofi Aventis Biomedical Innovation Award and the NIH/NIGMS Biotechnology Training Program. Citation Format: Tiffany F. Chen, Kevin Li, K. Dane Wittrup. Fibronectin domains engineered for specificity to individual murine Fc gamma receptors modulate tumoricidal activity of immune cells. [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 1250. doi:10.1158/1538-7445.AM2013-1250
    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...