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
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2012
    In:  European Heart Journal Vol. 33, No. suppl 1 ( 2012-08-02), p. 339-653
    In: European Heart Journal, Oxford University Press (OUP), Vol. 33, No. suppl 1 ( 2012-08-02), p. 339-653
    Type of Medium: Online Resource
    ISSN: 0195-668X , 1522-9645
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2012
    detail.hit.zdb_id: 2001908-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Molecular Cancer Therapeutics Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. B94-B94
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. B94-B94
    Abstract: Immune checkpoint modulators, such as antibodies targeting CTLA-4 or PD-1, are now being approved for treatment of patients with unresectable or metastatic melanoma and advanced squamous non-small cell lung cancer (NSCLC) who have progressed on or after platinum-based chemotherapy. Efficacy was also evidenced on other tumor types (renal cell carcinoma, bladder, Hodgkin lymphoma, colorectal carcinoma (CRC) ⋯). However, there is still needs to identify predictive biomarkers of response in order to select patients who will benefit from treatments. PD-L1 expression was proposed to be a good candidate for NSCLC, even if PD-L1 expression is a difficult parameter due to its expression on both tumor cells and immune cells as well as technical challenges to use immunohistochemical detection. The dynamic of the immune system as well as the site and time where interactions between tumor cells and immune cells take place, increase the complexity of having a solid biomarker identified. In addition, for other pathologies like colorectal carcinoma, genomic biomarkers were evidenced. For example, CRC patients with mismatch repair (MMR) deficiencies have an objective response rate of 62% compared with 0% in patients with MMR-proficient tumors. We propose here the use of syngeneic models to address mechanism of action and biomarker related questions for agent targeting PD-1 / PD-L1 axis. Syngeneic model systems remain one of the only options to analyse physiologically relevant interactions between tumor and immune cells. Up to now, eight models were characterized for response to either PD-1 and/or PD-L1 targeting antibodies. Among them, 4T1, LLC and Renca were identified as non-responders and B16-F10, CT-26, EMT-6, MBT-2 identified as partial responders. Most of the time, targeting PD-1 is more effective than targeting PD-L1, even if there is exception (e.g. B16-F10). Attempting to identify key parameters that could help us understand efficacy of PD-1 /PD-L1 axis disruptors, intratumoral immune infiltrate was analyzed in depth using flow cytometry: regulatory T cells (Treg), effector T cells (Teff), M-MDSCs, G-MDSCs, TAMs were phenotyped and quantified. In contrast to CTLA-4 targeting therapy, where Teff/Treg ratio was correlated to treatment efficacy, this is not the case for PD-1 or PD-L1 targeting therapies. It is now hypothesized that a more complex signature (e.g. detailed phenotype of CD8 positive T cells, tumor neoantigen expression⋯) will be needed to identify valuable biomarkers of response. Preliminary results using syngeneic models, both subcutaneously or orthotopically engrafted with tumors, will be presented. Citation Format: MARC HILLAIRET DE BOISFERON, Francis Bichat, CAROLINE MIGNARD, XAVIER TIZON, DAMIEN FRANCE, SYLVIE MAUBANT, Jean-Francois Mirjolet. Efficacy of PD-1 - PD-L1 pathway disruptors in syngeneic models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B94.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2790-2790
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2790-2790
    Abstract: The humanization process of mice with various tissues named Chi-mice aimed to reproduce better the human situation and to be more predictive than conventional models. In order to evaluate new targeting therapies or adverse side effects involving skin components, pre-clinical studies need to be assayed with new molecules in adequate and validated mouse models bearing human skin. The collection of skin samples was done under ethically approved master agreements and with the signed consent of each patient. The patient's clinical history, the serology results (HIV, HBV and HCV) and tissue banking were centralized in our internal approved biological resource center. To develop such models, we used skins of different origins including foreskin, breast and abdomen that were isolated by different methods (full recovery of about 1 cm depth sample or dermatomization of the 1-2 mm epidermal/dermal layer). These samples were xenografted on immune deficient NOD-SCID mice under different conditions including matrigel grafting. After one month implantation, skingrafts were collected and observed microscopically to confirm the preservation of human organization of skin by full histology. Also, the species nature of vessels was characterized by immunochemistry to evaluate the host penetration within human skingraft and both CD-31 positive mouse and human vessels were found in the skingraft. In the context of vascular leak syndrome (VLS) induction by IL2 treatment, we have used the wet/dry ratio of skingraft to measure the induced-edema and we have measured the blue evans uptake in those injured skingraft to appreciate the epidermal vasculature leaking. Anapathological comparisons were also performed to reveal the histological modifications observed during VLS. These different characterizations performed during development of the skingraft model enabled us to obtain human skin xenografted on mice that retained the human characteristics of the primary material. The co-implantation of other human tissue (tumor) with skin in same mice which refined better the humanized model will be presented. The use of fresh skin and various tissues in drug discovery and early preclinical development of new therapies aimed at corroborating results with clinical reality. Altogether, these processes from the clinical sample collection to the in vivo drug efficacy study through ex vivo assays should help the preclinical drug selection, development and clinical positioning as well as companion biomarker identification. Citation Format: Guillaume Serin, Loïc Morgand, Marie Leblanc, Francis Bichat. Establishment and characterization of human skingraft model in immunodeficient mice. [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 2790. doi:10.1158/1538-7445.AM2013-2790
    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 ...
  • 4
    In: Journal of Medicinal Chemistry, American Chemical Society (ACS), Vol. 59, No. 7 ( 2016-04-14), p. 2918-2927
    Type of Medium: Online Resource
    ISSN: 0022-2623 , 1520-4804
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2016
    detail.hit.zdb_id: 1491411-6
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 394-394
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 394-394
    Abstract: Background: Humanized mice are a promising translational research model for evaluation of pharmacological compounds efficacy and safety in Oncology. Their use has been enhanced by the development of new stocks of immunodeficient hosts, most notably mouse strain such as NOD-scid/IL2rγ null mice (NSG). As previously described (L. Schultz et al, J. Immunol. 2005) the NSG mice could also be successfully humanized after engraftment of human hematopoietic stem cells (HSC). NSG mice have also been shown to be superior to other immunodeficient mice (BALB/c nu/nu, NOD/Shi-scid) for xenograft of tumour material. These models are particularly needed in preclinical development where there is no appropriate small animal model combining the human immune system and human tumour. Methods: Freshly collected umbilical cord blood (UCB) samples were provided by the “établissement français du sang, EFS” following the French ethical rules. The UCB were selected for their HSC CD34+ content and T cells depleted before being intracardiacally injected in whole body irradiated newborn NSG mice. The reconstitution of a human immune system within mouse was analyzed in peripheral blood and in primary and secondary lymphoid organs (bone marrow, spleen and thymus) using a 7-color FACS analysis. The mice were then xenografted with tumour cell lines, fresh primary human solid tumours and haematological malignancies to combine the development of human cancer and human immune system within the same mouse. Results and conclusions: Fifteen weeks after stem cells injection, the peripheral blood contains about 50% of human leukocytes distributed as followed: about 50% of T lymphocytes, about 40% of B lymphocytes, about few percent of monocytes, macrophages, granulocytes, natural killer cells and platelets. At same time, the central chimerism within lymphoid organs for human leucocytes has represented about 80%, 85% and 95% in bone marrow, spleen and thymus cells, respectively. This NSG mouse model opens new in vivo perspectives to study the complex relationships between the human immune system and human tumour cells under therapeutic treatments with biologics (antibody-dependent cell-mediated cytotoxicity, antitumour vaccination strategies . . .). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 394.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 328-328
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 328-328
    Abstract: Cationic antimicrobial peptides (CAPs) are naturally occurring molecules found in a number of species as a defense mechanism for eukaryotic cells against pathogens and are an integral part of the innate immune system. CAPs have a broad spectrum of activities including antimicrobial and anticancer while being less cytotoxic toward non-malignant cells. The potential therapeutic application against cancer has spawned an interest in developing oncolytic agents that display a new mode of action to overcome the potential drug resistance associated with other current therapeutics. The anticancer effects of CAPs are still under investigation, but several peptides have already exhibited a promising potential with cytotoxic activities against a broad spectrum of tumor cells. Oncolytic peptides exert their activity through either a membranolytic mode of action or an interaction with intracellular targets, or a combination of both. LTX-315 (K-K-W-W-K-K-W-Dip-K-NH2), a novel oncolytic peptide developed by Lytix Biopharma AS has the potential to adopt an amphipathic helical coil structure. In vitro studies have demonstrated that it was highly effective against both drug-resistant and drug-sensitive cancer cells from several organ origins, with lower toxicity toward normal cells. LTX-315 was designed for intratumoral treatment of transdermal lesions. Previously, LTX-315 has been shown to induce complete regression of B16 melanomas and long lasting antitumor immune responses. Histological analyses of treated tumors revealed extensive hemorrhagic necrosis and infiltration of CD3+ T cells. Moreover, mRNA levels of inflammatory cytokines such as IL1β, IL6 and IL18 were found to be increased in the tumor tissue after LTX-315 treatment. The treatment did also prevent lung metastasis in mice re-challenged with B16F1 cells intravenously. Due to its oncolytic mode of action, LTX-315 induces immunogenic cell death through the release of danger-associated molecular pattern molecules and tumor antigens. Recently, we have demonstrated that when subcutaneously established EMT-6 tumors (inoculated into both flanks of the animal) were treated intratumorally with LTX-315, an antitumor response was observed with a T/C ratio of 17% 19 days post start of treatment. Furthermore, an abscopal effect of LTX-315 on the untreated tumor was also reported but only when it was combined with anti-PD-L1 antibody. At the end of study, 50% of mice that had received the combination therapy were still alive vs 30% and 40% in the groups treated with LTX-315 or anti-PD-L1 antibody alone, respectively. In conclusion, LTX-315 seems to be an ideal combinations partner for immune checkpoint inhibitors. Citation Format: Ketil André Camilio, Baldur Sveinbjørnsson, Sylvie Maubant, Guillaume Serin, Jean-François Mirjolet, Francis Bichat, Øystein Rekdal. Antitumor activity of the oncolytic peptide LTX-315 in syngeneic tumor models. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 328.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1567-1567
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1567-1567
    Abstract: Immune cells within the tumor microenvironment are now well recognized as to be targets of interest for cancer treatment. However there are no available pre-clinical models that will accurately support the pre-clinical development of such therapeutic approaches. Humanized NOG (NOD/shi-scid/IL-2Rγnull) mice, bearing human immune cells with or without human target tumor cells, are relevant models to test various therapeutic strategies (e.g. antibody dependent cell cytotoxicity, Treg targeting antibodies, TLR agonists, vaccines, adoptive T cells transfer, …) in various pathologies (e.g. oncology, autoimmune disease, inflammation, transplantation, …). Humanization of NOG mice was already characterized and validated using freshly collected human PBMCs or hematopoietic stem cells (HSCs) in both newborn and adult NOG mice. The complete characterization of the reconstituted NOG mice was done in peripheral blood as well as central lymphoid organs such as bone marrow, spleen and thymus, using FACS analysis. Moreover, the human immune reconstitution and function in NOG mice was also evidenced with human cytokine and immunoglobulin quantification in mouse plasma samples. The injection of mature human PBMC in adult NOG mice leads to the consistent development of a xenogeneic graft-versus-host disease (GvHD), which mimics human GvHD (modification of CD4/CD8 ratio, expression of T cells activation markers and cytokine production). Growth characteristics of human tumors models on humanized mice will be presented either as subcutaneous or as disseminated intravenous model. Randomization parameters were selected regarding both tumor and immune cell sides. Required readouts to understand immune cell modulation (Cell phenotyping and functionality) and related antitumor efficacy will be described. Finally, such models, i.e. humanized NOG mice bearing or not human tumor cells in the context of evaluation of new therapies will be detailed in various pathologies. Citation Format: Caroline Mignard, Jean-Francois Mirjolet, Holger Kissel, Francis Bichat, Olivier Duchamp. Use of humanized NOG mice within the pre-clinical development of new human specific therapies. [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 1567. doi:10.1158/1538-7445.AM2013-1567
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 624-624
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 624-624
    Abstract: Mice with a humanized immune system, so called “humanized” mouse models, have been established to study the complex interaction of the human immune system during human disorders. In case of cancer research, the in vivo model ideally should recapitulate the biological characteristics of the human tumor and of the related tumor microenvironment in patient such as immune system. Human immune system is reconstituted in immunodeficient mice using either human PBMCs or hematopoietic stem cells (HSCs). Humanized mice bearing human target tumor cells constitute relevant models for evaluation of cancer therapeutics such as bispecific antibodies, immune cell targeting antibodies. In vivo proof of concept studies were performed with humanized mice xenografted with either human disseminated lymphoma or with human solid tumors (tumor cell line and Patient Derived Xenograft model, Her2 positive breast, colon, ovarian and lung cancers) and treated with therapeutic antibodies. Biomarkers of response to immune cell targeting therapies were evaluated by flow cytometry as well as immunohistochemistry analyses. In case of disseminated B-cell lymphoma grafted into mice reconstituted with human PBMC, efficacy of antibody was evidenced by an increased survival and a high decrease of human B cells in mouse bone marrow. Lung and ovarian PDX developed in HSC reconstituted BRGS mice induced an increase in Tregs and a change in T/B ratio in blood and spleen samples. Furthermore, we evidenced the presence of Tregs within human PDX tumor by immunohistochemistry. Citation Format: Olivier Duchamp, Francis Bichat, Caroline Mignard, François Ghiringhelli, Jean-François Mirjolet, IMODI Consortium. Humanized mouse models for evaluation of cancer therapies. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 624.
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1325-1325
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1325-1325
    Abstract: Immunotherapy based on monoclonal antibodies targeting cancer cells is now developed as a valid approach to treat cancer. Modulation of novel immune checkpoints and other targets are highly promising approaches against cancer and many other diseases. They have the potential to activate the immune system and to establish an active defense against pathological conditions. In addition, antibodies, antibody fragments, and other biologics can also have a strong impact on the immune system which needs to be evaluated early on. In order to develop and accurately evaluate these immunology linked approaches, appropriate preclinical models with relevant immunological readouts are needed at different stages of therapy development. Ideally, methods should be available that allow predictive readouts in vivo and ex vivo. A comprehensive panel of tools was constructed and validated aiming at evaluating the modulation of the immune system by new therapies. In immunocompetent mice, immune cells were studied for the detection of their cell surface markers, induction of proliferative phenotype, antigen-specific T lymphocyte detection, secretion of soluble mediators using FACS phenotyping, cytometric bead assay (CBA) or Luminex multiplex technologies and ELISPOT. We report on a panel of syngenic tumor models (4T1, B16-F10, CT26, EMT6, AY27, LLC1, MBT-2 and Renca) our capacity to correlate subpopulation of immune infiltrating cells (as T cells, macrophages, NK cells, granulocytic and monocytic cells) and the therapeutic effects of critical antibodies directed against Cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death 1 membrane protein (PD-1). In immunodeficient mice, the reconstitution of the mature human immune system with PBMC or naïve human immune system using hematopoietic stem cells was used to evaluate the modulation of those immune cells by therapies through human cytokine release and onset of graft versus host disease. All of these tools were used in the context either of rodent syngenic models or humanized mouse models. In addition to their use in immune system modulating anti-cancer therapies, these humanized or syngenic models also have a potential application in many other therapeutic areas such as autoimmune diseases, inflammation, anti-infectives. Citation Format: Marc Hillairet de Boisferon, Francis Bichat, Caroline Mignard, Xavier Tizon, Damien France, Jean-François Mirjolet. Evaluation of immuno-oncology related treatment in syngenic and human reconstituted immune systems. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1325. doi:10.1158/1538-7445.AM2015-1325
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 10
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 11 ( 2018-11-01), p. 2389-2398
    Abstract: Carboxylesterase 1c (CES1c) is responsible for linker-drug instability and poor pharmacokinetics (PK) of several antibody–drug conjugates (ADC) in mice, but not in monkeys or humans. Preclinical development of these ADCs could be improved if the PK in mice would more closely resemble that of humans and is not affected by an enzyme that is irrelevant for humans. SYD985, a HER2-targeting ADC based on trastuzumab and linker-drug vc-seco-DUBA, is also sensitive to CES1c. In the present studies, we first focused on the interaction between CES1c and SYD985 by size- exclusion chromatography, Western blotting, and LC/MS-MS analysis, using recombinant CES1c and plasma samples. Intriguingly, CES1c activity not only results in release of the active toxin DUBA but also in formation of a covalent bond between CES1c and the linker of vc-seco-DUBA. Mass spectrometric studies enabled identification of the CES1c cleavage site on the linker-drug and the structure of the CES1c adduct. To assess the in vivo impact, CES1c−/− SCID mice were generated that showed stable PK for SYD985, comparable to that in monkeys and humans. Patient-derived xenograft (PDX) studies in these mice showed enhanced efficacy compared with PDX studies in CES1c+/+ mice and provided a more accurate prediction of clinical efficacy of SYD985, hence delivering better quality data. It seems reasonable to assume that CES1c−/− SCID mice can increase quality in ADC development much broader for all ADCs that carry linker-drugs susceptible to CES1c, without the need of chemically modifying the linker-drug to specifically increase PK in mice. Mol Cancer Ther; 17(11); 2389–98. ©2018 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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...