Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3025-3025
    Abstract: Abstract 3025 Poster Board II-1001 Due to its restriction to the B-cell lineage and high surface expression in B-cell malignancies, CD19 is an attractive target antigen for immunological strategies in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). While preclinical in vivo studies of CD19-specific cellular immunotherapy have generally used xenografts from human CD19+ leukemia cell lines, primary leukemia cells are likely to more closely mimic the disease in humans and allow to differentiate between standard and high risk situations. Therefore, we investigated the in vivo sensitivity of human leukemic bone marrow to adoptive immunotherapy with gene-modified CD19-specific T cells. Among 15 primary leukemias obtained from the bone marrow of pediatric patients at diagnosis, 10 were successfully engrafted in NOD/scid mice by intrafemoral injection within 6 to 20 weeks. For therapeutic experiments, we focused on one standard risk leukemia, characterized by a rapid and sustained response to multiagent chemotherapy, and on a leukemia bearing the high-risk feature of an MLL rearrangement, which was refractory to standard treatment. Titration experiments demonstrated reliable engraftment of 1×104 leukemic cells per mouse. For CD19-directed T-cell therapy, cytotoxic T cells (CTLs) with native specificity for Epstein-Barr virus antigens were expanded from 4 healthy donors and transduced to express either a codon-optimized CD19-specific chimeric antigen receptor (CAR) containing the intracellular signaling domain of the TCRz chain (CD19-z), or a control CAR directed against the neuroectodermal antigen GD2 (14.G2a-z). Costimulatory domains now commonly used to ensure sustained T-cell activation via CARs were not included, since previous studies have shown that CAR activity in virus-specific CTLs does not benefit from additional signaling elements. CTLs had a uniform CD8+ effector memory T-cell phenotype (CD45RO+, CCR7-), and CAR surface expression was 73±21%, range 32-93% (CD19-z, n=9) and 18±13%, range 6-35% (14.G2a-z, n=5). In vitro cytotoxicity experiments confirmed specific lysis of the CD19+ leukemia cell lines REH (51Cr release 59.7±7.2% at an effector target ratio of 20:1) and SupB15 (66.7±8.6) as well as primary CD19+ leukemic cells from 5 pediatric patients (47.2±13.2%), in the absence of background lysis by 14.G2a-z-transduced control CTLs. 1×104 leukemic cells per mouse from primary engrafted mice were transferred into further cohorts of NOD/scid mice by secondary intrafemoral transplantation, followed by adoptive transfer of 4 doses of 5×106 CTLs via tail vein injection on days 1, 4, 8, and 11. IL-2 (500 IU/mouse) was administered twice-weekly, and sequential murine bone marrow aspirates were analyzed for human leukemia engraftment by flow cytometry using human CD45 and CD19-specific antibodies starting 3 weeks after transplantation. CD19z CTLs prevented engraftment of the standard risk leukemia in 3 of 4 mice, while 3 of 4 control mice developed the leukemia (p = 0.158, Log Rank/Mantel-Cox Test). Moreover, while the MLL-rearranged human leukemia became detectable in the bone marrow of 4 of 5 control mice, followed by overt and fatal leukemia, 5 of 8 mice receiving transfusions of CD19-z transduced CTLs remained disease-free (p = 0.067), and 6 of 8 remained alive, one of them with detectable leukemia cells (p = 0.054) (see Figure). Thus, adoptive transfer of CD19-redirected CTLs efficiently delayed or prevented engraftment of both standard and high risk ALLs in mice and therefore provides a promising treatment option for patients with BCP-ALL refractory to standard treatment. 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: 2009
    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 ...
  • 2
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2009
    In:  Cancer Immunology, Immunotherapy Vol. 58, No. 12 ( 2009-12), p. 1991-2001
    In: Cancer Immunology, Immunotherapy, Springer Science and Business Media LLC, Vol. 58, No. 12 ( 2009-12), p. 1991-2001
    Type of Medium: Online Resource
    ISSN: 0340-7004 , 1432-0851
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2009
    detail.hit.zdb_id: 1458489-X
    detail.hit.zdb_id: 195342-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Molecular Therapy, Elsevier BV, Vol. 27, No. 5 ( 2019-05), p. 933-946
    Type of Medium: Online Resource
    ISSN: 1525-0016
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 2001818-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Gene Therapy, Springer Science and Business Media LLC
    Abstract: To enhance the potency of chimeric antigen receptor (CAR) engineered T cells in solid cancers, we designed a novel cell-based combination strategy with an additional therapeutic mode of action. CAR T cells are used as micropharmacies to produce a targeted pro-coagulatory fusion protein, truncated tissue factor (tTF)-NGR, which exerts pro-coagulatory activity and hypoxia upon relocalization to the vascular endothelial cells that invade tumor tissues. Delivery by CAR T cells aimed to induce locoregional tumor vascular infarction for combined immune-mediated and hypoxic tumor cell death. Human T cells that were one-vector gene-modified to express a G D2 -specific CAR along with CAR-inducible tTF-NGR exerted potent G D2 -specific effector functions while secreting tTF-NGR that activates the extrinsic coagulation pathway in a strictly G D2 -dependent manner. In murine models, the CAR T cells infiltrated G D2 -positive tumor xenografts, secreted tTF-NGR into the tumor microenvironment and showed a trend towards superior therapeutic activity compared with control cells producing functionally inactive tTF-NGR. In vitro evidence supports a mechanism of hypoxia-mediated enhancement of T cell cytolytic activity. We conclude that combined CAR T cell targeting with an additional mechanism of antitumor action in a one-vector engineering strategy is a promising approach to be further developed for targeted treatment of solid cancers.
    Type of Medium: Online Resource
    ISSN: 0929-1903 , 1476-5500
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 1212513-1
    detail.hit.zdb_id: 2004200-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2007
    In:  Cancer Research Vol. 67, No. 17 ( 2007-09-01), p. 8335-8343
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 67, No. 17 ( 2007-09-01), p. 8335-8343
    Abstract: T cells with grafted specificities for surface antigens provide an avenue for rapidly producing immune effector cells with tumor specificity. However, the function of chimeric receptor (chRec) gene-modified T cells is limited by lack of T-cell expansion and persistence. We propose to use varicella zoster virus (VZV)–reactive T cells as host for the chRec because these cells can be expanded both in vitro and in vivo by stimulation of their native receptor during endogenous reexposure to the virus or by administration of VZV vaccine. We obtained human T cells reactive with VZV from the peripheral blood of seropositive donors by stimulation with VZV lysate and evaluated their characteristics after genetic modification with two tumor-specific model chRecs. Cultures dominated by cytolytic CD4+ T cells (VZV-CTL) could be expanded and maintained in vitro. Gene-modified VZV-CTL recognized and lysed tumor targets in a MHC-independent manner while maintaining functional, MHC-restricted interaction with VZV antigen through their native receptor. Thus, chRec-transduced VZV-CTL may provide a source of potent tumor-reactive cells for adoptive immunotherapy of cancer. The availability of a safe and effective VZV vaccine provides the option of repeated in vivo stimulation to maintain high T-cell numbers until the tumor is eliminated. [Cancer Res 2007;67(17):8335–43]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3437-3437
    Abstract: Myxoid liposarcomas (MLS) account for 20% of malignant adipocytic tumors and are characterized by a high rate of local recurrence and development of distant metastases in approximately 40% of patients. Most MLS are driven by the FUS-DDIT3 fusion gene encoding an aberrant transcription factor. The mechanisms whereby FUS-DDIT3 mediates sarcomagenesis are incompletely understood, and strategies to selectively target MLS cells remain elusive. In this study, we employed genome-scale RNA interference (RNAi) screening to uncover that human mesenchymal stem cells engineered to express FUS-DDIT3 and MLS cell lines are dependent on YAP1, a transcriptional co-activator and central effector of the Hippo pathway involved in tissue growth and tumorigenesis. Analysis of a large cohort of primary MLS specimens (n=223) revealed that nuclear YAP1 expression was significantly more prevalent in MLS compared to other liposarcoma subtypes. In support of the concept that increased YAP1-mediated transcriptional activity represents an essential feature of MLS development, RNAi-based YAP1 depletion in cultured MLS cells resulted in suppression of cell viability, cell cycle arrest, cellular senescence, and induction of apoptosis accompanied by decreased YAP1 target gene expression, and YAP1-positive primary MLS tumors showed strong expression of YAP1 downstream effectors such as FOXM1 and PLK1. Mechanistically, FUS-DDIT3 promotes YAP1 transcription, nuclear localization, and transcriptional activity and physically associates with YAP1 in the nucleus of MLS cells, pointing to the coordinate establishment of gene expression programs that promote MLS tumorigenesis. Consistent with the hypothesis that a YAP1-directed therapeutic approach could represent a rational strategy to selectively target FUS-DDIT3-expressing MLS cells, pharmacologic inhibition of YAP1 activity with verteporfin suppressed cell viability and YAP1 target gene expression in MLS cell lines, and the growth-inhibitory effects of YAP1 knockdown or verteporfin treatment could be recapitulated in MLS cell line-based xenograft models. Collectively, our data identify dependence on aberrant YAP1 activity as specific liability of FUS-DDIT3-expressing MLS cells, and provide preclinical evidence that YAP1-mediated signal transduction represents a candidate target for therapeutic intervention that warrants further investigation. Citation Format: Marcel Trautmann, Ya-Yun Cheng, Patrizia Jensen, Ninel Azoitei, Ines Brunner, Jennifer Hüllein, Mikolaj Slabicki, Ilka Isfort, Magdalene Cyra, Eva Wardelmann, Sebastian Huss, Bianca Altvater, Claudia Rossig, Susanne Hafner, Thomas Simmet, Anders Ståhlberg, Pierre Åman, Thorsten Zenz, Undine Lange, Thomas Kindler, Claudia Scholl, Wolfgang Hartmann, Stefan Fröhling. Requirement for YAP1 signaling in myxoid liposarcoma [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 3437.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3939-3939
    Abstract: Introduction: Myxoid liposarcoma (MLS) is the second most common type of liposarcoma and characterized by a high tendency to develop metastases. The molecular hallmark of MLS (≈90%) is a pathognomonic reciprocal t(12;16) (q13;p11) translocation, leading to the specific gene fusion of FUS and DDIT3. The resulting chimeric FUS-DDIT3 fusion protein is suggested to play a crucial role in MLS tumorigenesis, although its specific biological function and mechanism of action remain to be substantiated. While radiotherapy and chemotherapy with high-dose ifosfamide and doxorubicin represent established therapeutic options, prognosis in the metastasized situation is poor. Molecularly targeted therapeutic approaches are currently not available. Aiming at the preclinical identification of novel therapeutic options, we here investigate the functional relevance of phosphatidylinositol-3'-kinase (PI3-kinase)/Akt signaling in MLS. Experimental procedures: Immunohistochemical and FISH analyses of PI3-kinase/Akt signaling effectors were performed in a large cohort of clinical MLS tumor specimens. Mutational burden was studied by targeted next-generation sequencing (NGS; Illumina MiSeq). PI3-kinase/Akt-mediated signaling transduction was modulated by specific RNAi knockdown and a pharmacological approach applying the small molecule inhibitor BKM120 (Buparlisib; NVP-BKM120). Cell proliferation and FACS assays were performed in different MLS cell lines. An established MLS chorioallantoic membrane model (CAM) was employed for in vivo confirmation of the preclinical in vitro data. Results: In a significant subset of MLS tumor specimens, immunohistochemical staining revealed elevated phosphorylation levels of various signaling components, indicating that activation of PI3-kinase/Akt signaling is a frequent feature in MLS. Activating PIK3CA mutations and loss of PTEN as mechanism for PI3-kinase/Akt activation were detected in ≈15%. PI3-kinase inhibition significantly suppressed the signaling cascade, associated with reduction of MLS cell viability and induction of apoptosis in vitro and in vivo. Conclusions: Our preclinical study emphasizes the pivotal role of the PI3-kinase/Akt signaling cascade in MLS pathogenesis and indicates the occurrence of specific mutational aberrations apart from the pathognomonic FUS-DDIT3 gene fusion. Our in vitro and in vivo results suggest that targeting the PI3-kinase/Akt signaling pathway provides a rational, molecularly founded therapeutic strategy in the treatment of MLS. Citation Format: Marcel Trautmann, Magdalene Cyra, Christian Bertling, Ilka Isfort, Bianca Altvater, Claudia Rossig, Susanne Hafner, Thomas Simmet, Jessica Becker, Inga Grünewald, Pierre Åman, Reinhard Büttner, Eva Wardelmann, Sebastian Huss, Wolfgang Hartmann. Activation of phosphatidylinositol-3′-kinase/Akt signaling in myxoid liposarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3939.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 Cancer Research, American Association for Cancer Research (AACR), Vol. 24, No. 2_Supplement ( 2018-01-15), p. B04-B04
    Abstract: Introduction: Myxoid liposarcoma (MLS) is the second most common type of liposarcoma and an aggressive disease with particular propensity to develop hematogenic metastases. Ninety percent of MLS are characterized by a reciprocal translocation t(12;16) (q13;p11), leading to the pathogenic gene fusion of FUS and DDIT3. The resulting chimeric FUS-DDIT3 fusion protein is suggested to play a crucial role in MLS pathogenesis, although the specific mechanism of action remains to be substantiated. Aiming at the preclinical identification of novel therapeutic options, we here investigate the functional relevance of FUS-DDIT3 in IGF-IR/PI3K/Akt signal transduction. Experimental Procedures: Immunohistochemical analyses of IGF-IR/PI3K/Akt signaling effectors and modulators were performed in a comprehensive cohort of clinical MLS specimens. FUS-DDIT3-dependent activation of the IGF-IR/PI3K/Akt signaling cascade was analyzed by siRNA and immunoblotting in vitro. Cell proliferation and FACS assays were performed in multiple tumor-derived MLS cell lines. An established MLS chorioallantoic membrane model (CAM) was employed for in vivo confirmation of the preclinical in vitro data. Results: In a significant subset of MLS specimens, immunohistochemical staining revealed elevated phosphorylation levels of various signaling components, representing a strong indication of activated IGF-IR/PI3K/Akt signaling to be a frequent feature in MLS. IGF-IR inhibition significantly suppressed the IGF-IR/PI3K/Akt signaling cascade, associated with impairment of MLS cell viability and induction of apoptosis in vitro and in vivo. Furthermore, siRNA-mediated knockdown of FUS-DDIT3 led to dephosphorylation of IGF-IR/PI3K/Akt signaling components, implying that the FUS-DDIT3 fusion protein is involved in the IGF-IR regulated signaling cascade. Conclusions: Our preclinical study emphasizes the pivotal role of the IGF-IR/PI3K/Akt signaling pathway in MLS pathogenesis and indicates its functional dependence on the MLS-specific FUS-DDIT3 fusion protein. Furthermore, our in vitro and in vivo results demonstrate that targeting the IGF-IR/PI3K/Akt signaling pathway provides a rational, molecularly founded therapeutic strategy in the treatment of MLS. Citation Format: Marcel Trautmann, Magdalene Alice Cyra, Christian Bertling, Ilka Isfort, Jasmin Menzel, Konrad Steinestel, Inga Grünewald, Bianca Altvater, Claudia Rossig, Pierre Åman, Eva Wardelmann, Sebastian Huss, Wolfgang Hartmann. Functional characterization of IGF-IR/PI3K/Akt signaling in myxoid liposarcoma [abstract]. In: Proceedings of the AACR Conference on Advances in Sarcomas: From Basic Science to Clinical Translation; May 16-19, 2017; Philadelphia, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(2_Suppl):Abstract nr B04.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 15, No. 15 ( 2009-08-01), p. 4857-4866
    Abstract: Purpose: Novel natural killer (NK) cell–directed strategies in cancer immunotherapy aim at specifically modulating the balance between NK cell receptor signals toward tumor-specific activation. The signaling lymphocyte activation molecule–related receptor 2B4 (CD244) is an important regulator of NK cell activation. We investigated whether 2B4-enhanced activation signals can redirect the cytolytic function of human NK cells to NK cell–resistant and autologous leukemia and tumor targets. Experimental Design: In vitro–stimulated NK cells from healthy donors and pediatric leukemia patients were gene modified with CD19 or GD2-specific chimeric receptors containing either the T-cell receptor ζ or 2B4 endodomain alone or combined. Results: Chimeric 2B4 signaling alone failed to induce interleukin-2 receptor up-regulation and cytokine secretion but triggered a specific degranulation response. Integration of the 2B4 endodomain into T-cell receptor ζ chimeric receptors significantly enhanced all aspects of the NK cell activation response to antigen-expressing leukemia or neuroblastoma cells, including CD25 up-regulation, secretion of IFN-γ and tumor necrosis factor-α, release of cytolytic granules, and growth inhibition, and overcame NK cell resistance of autologous leukemia cells while maintaining antigen specificity. Conclusion: These data indicate that the 2B4 receptor has a potent costimulatory effect in NK cells. Antigen-specific 2B4ζ-expressing NK cells may be a powerful new tool for adoptive immunotherapy of leukemia and other malignancies.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4999-4999
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4999-4999
    Abstract: Preclinical in vivo studies of chimeric antigen receptor (CAR) T cells often rely on NOD-scid gamma (NSG) mouse models which lack T cells, B cells and NK cells and thereby allow for reliable engraftment of human tumor xenografts. But murine myeloid cells present in the NSG mouse strain can affect the tumor microenvironment as well as the function of adoptively transferred human immune effector cells. Long et al. (Cancer Immunol Res. 2016 Oct;4(10):869-880) reported that pediatric sarcoma xenografts in NSG mice induce in vivo expansion of murine CD11b+ myeloid-derived suppressor cells (MDSC), and that this cell population suppresses human CAR T cell proliferation in vitro. We investigated a strategy to avoid inhibitory effects of murine MDSCs on Ewing sarcoma xenografts in NSG mice by in vivo pretreatment with murine antibody against two cell surface antigens, Ly6C/Ly-6G (granulocyte-differentiation antigen-1, Gr-1), expressed on murine MDSC. Analysis of the CD11b+ myeloid cell populations in NSG mice 17-37 days after subcutaneous transplantation of the Ewing sarcoma cell line TC-71 showed a noticeable increase of CD11b+ cells in the peripheral blood compared to non-tumor bearing NSG mice (median 3.4 × 103, range 0.7-15.5 × 103 cells/ml, n=8 vs median 2.1 × 103, range 1.2-4.0 × 103 cells/ml, n=7). The increase was even more pronounced in the spleens, with a median absolute number of 2.6 × 106 CD11b+ cells (range 0.2-21 × 106 cells) per spleen in sarcoma-bearing mice versus 0.5 × 106 CD11b+ cells (range 0.2-0.8 × 106 cells) per spleen in mice without tumors. The granulocytic MDSC subset coexpressing Ly6Gpos was the most prominent subpopulation in spleen and blood. To eliminate murine MDSCs, we treated sarcoma-bearing mice twice weekly with 200 µg anti-Gr1 antibody RB6-8C5 over a period of 2 weeks, starting at tumor volumes of 100-200 mm3. While the combined percentage of the Ly6Gpos and Ly6Cpos cell populations in the spleens decreased compared to untreated mice (median 55%, range 36-75%, n=9 versus median 74%, range 72-86%, n=8), the total numbers of CD11b+ cells further increased (median 3.1 × 106, range 0.4 × 106-11 × 106 cells/spleen). Our data suggest that anti-Gr1 antibody pretreatment leads to blockade of the Ly6C/Ly6G receptors rather than eliminating MDSC subsets. To optimize preclinical pediatric sarcoma models, other methods for depletion or functional inactivation of mouse MDSCs will need to be evaluated. Citation Format: Sareetha Kailayangiri, Bianca Altvater, Katja Urban, Jutta Meltzer, Lea Greune, Nicole Farwick, Silke Jamitzky, Claudia Rossig. Evaluation of anti-Gr1 antibody for depletion of MDSC in preclinical NSG mouse models of pediatric sarcoma [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 4999.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages