Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 4, No. S1 ( 2016-11)
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2016
    detail.hit.zdb_id: 2719863-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5733-5733
    Abstract: The impact of tumor matrix remodeling to the generation of an “inflamed” microenvironment that modulates responses to immunotherapy is unclear. Versican (VCAN) is a chondroitin sulphate matrix proteoglycan that promotes tolerogenic polarization of intratumoral DC through Toll-like receptor 2 (TLR2). Proteolytic processing of VCAN releases a bioactive N-terminal fragment (matrikine), versikine. In contrast to the tolerogenic actions of parental VCAN, versikine triggers IRF8-dependent transcription in myeloid cells and promotes Batf3-dendritic cell (DC) generation from FLT3L-mobilized bone marrow progenitors in vitro. Consistent with the Batf3-promoting effects of versikine, VCAN proteolysis correlates with T-cell infiltration across multiple cancers.The aims were to 1. define the impact of versikine on the intratumoral myeloid repertoire in vivo and 2. to define the efficacy of versikine as a vaccine adjuvant.4T1 breast carcinoma and Lewis Lung Carcinoma (LLC) empty vector (EV)- and versikine-expressing cells were implanted subcutaneously in syngeneic recipients. 1000mm3 tumors were harvested and intratumoral DC subsets were enumerated. Versikine-expressing tumors were characterized by significantly enhanced Batf3-DC (CD11chigh,MHC IIhigh Ly6C-, CD64-, CD24high,CD11blow) (p =0.0079 for 4T1 model and & lt;0.0001 for LLC model), whereas cDC2 (CD11chigh,MHC IIhigh Ly6C-, CD64-, CD24low, CD11bhigh ) frequency was diminished (p= 0.0079 and & lt;0.0001 respectively). Monocytic-derived DC (Mo-DC: CD11chigh, MHC IIhigh, Ly6C+, CD64+) remained unchanged. To determine the impact of versikine on responses to in situ vaccination using STING agonists, EV- and versikine-replete tumors (B16 and 4T1; 150 mm3), were injected intratumorally (IT) with a single subtherapeutic dose (200 μg) of DMXAA (murine STING agonist) or vehicle. EV-tumors did not significantly respond to 200μg DMXAA, whereas many B16-versikine and 4T1-versikine tumors regressed or growth was inhibited (p & lt;0.001 and p=0.014 respectively). Necrosis was frequently observed in 4T1 versikine-secreting tumors (6/9 mice) within 24 hours after treatment. Versikine extended survival after subtherapeutic DMXAA treatment in 4T1; log rank=p=0.01. Versikine's effects were abrogated in Batf3-null mice. To quantitate antigen-specific responses in the presence or absence of versikine, EV- and versikine-replete LLC tumors were injected IT with 500 μg DMXAA (therapeutic dose) or vehicle. We observed a significant increase in the frequency of CD8+ MHCI:SIINFEKL tetramer+ splenocytes in LLC-versikine-bearing animals as well as a marked increase in central memory T splenocytes (TCM) (CD62LhighCD44high). VCAN matrikines may generate effective adjuvants for in situ vaccination strategies across diverse solid and hematopoietic tumor types. Citation Format: Athanasios Papadas, Evan Flietner, Zachary Morrow, Joshua Wiesner, Alexander Cicala, Adam Pagenkopf, Chelsea Hope, Philip Emmerich, Dustin Deming, Jing Zhang, Peiman Hematti, Natalie Callander, Alexander Rakhmilevich, Mario Otto, Christian Capitini, Fotis Asimakopoulos. Versican proteolytic fragments (matrikines) synergize with STING agonists to elicit robust anti-tumor CD8+ T cell responses [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 5733.
    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 ...
  • 3
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 8, No. 2 ( 2020-10), p. e001262-
    Abstract: Some patients with cancer treated with anticancer monoclonal antibodies (mAbs) develop antidrug antibodies (ADAs) that recognize and bind the therapeutic antibody. This response may neutralize the therapeutic mAb, interfere with mAb effector function or cause toxicities. We investigated the potential influence of ADA to modify the tumor-binding capability of a tumor-reactive ‘immunocytokine’ (IC), namely, a fusion protein (hu14.18-IL2) consisting of a humanized, tumor-reactive, anti-GD2 mAb genetically linked to interleukin 2. We characterize the role of treatment delivery of IC (intravenous vs intratumoral) on the impact of ADA on therapeutic outcome following IC treatments in an established antimelanoma (MEL) regimen involving radiotherapy (RT) +IC. Methods C57BL/6 mice were injected with human IgG or the hu14.18-IL2 IC to develop a mouse anti-human antibody (MAHA) response (MAHA + ). In vitro assays were performed to assess ADA binding to IC using sera from MAHA + and MAHA − mice. In vivo experiments assessed the levels of IC bound to tumor in MAHA + and MAHA − mice, and the influence of IC route of delivery on its ability to bind to B78 (GD2+) MEL tumors. Results MAHA is inducible in C57BL/6 mice. In vitro assays show that MAHA is capable of inhibiting the binding of IC to GD2 antigen on B78 cells, resulting in impaired ADCC mediated by IC. When B78-bearing mice are injected intravenously with IC, less IC binds to B78-MEL tumors in MAHA + mice than in MAHA − mice. In contrast, when IC is injected intratumorally in tumor-bearing mice, the presence of MAHA does not detectibly impact IC binding to the tumor. Combination therapy with RT+IT-IC showed improved tumor regression compared with RT alone in MAHA + mice. If given intratumorally, IC could be safely readministered in tumor-bearing MAHA + mice, while intravenous injections of IC in MAHA + mice caused severe toxicity. Histamine levels were elevated in MAHA + mice compared with MAHA − mice after reintroduction of IC. Conclusions Intratumoral injection may be a means of overcoming ADA neutralization of therapeutic activity of tumor-reactive mAbs or ICs and may reduce systemic toxicity, which could have significant translational relevance.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2020
    detail.hit.zdb_id: 2719863-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 11 ( 2020-7-24)
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2020
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 15 ( 2024-7-15)
    Abstract: The majority of experimental approaches for cancer immunotherapy are tested against relatively small tumors in tumor-bearing mice, because in most cases advanced cancers are resistant to the treatments. In this study, we asked if even late-stage mouse tumors can be eradicated by a rationally designed combined radio-immunotherapy (CRI) regimen. Methods CRI consisted of local radiotherapy, intratumoral IL-12, slow-release systemic IL-2 and anti- CTLA-4 antibody. Therapeutic effects of CRI against several weakly immunogenic and immunogenic mouse tumors including B78 melanoma, MC38 and CT26 colon carcinomas and 9464D neuroblastoma were evaluated. Immune cell depletion and flow cytometric analysis were performed to determine the mechanisms of the antitumor effects. Results Tumors with volumes of 2,000 mm 3 or larger were eradicated by CRI. Flow analyses of the tumors revealed reduction of T regulatory (Treg) cells and increase of CD8/Treg ratios following CRI. Rapid shrinkage of the treated tumors did not require T cells, whereas T cells were involved in the systemic effect against the distant tumors. Cured mice developed immunological memory. Conclusions These findings underscore that rationally designed combination immunotherapy regimens can be effective even against large, late-stage tumors.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2024
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Immunology, Immunotherapy, Springer Science and Business Media LLC, Vol. 72, No. 7 ( 2023-07), p. 2459-2471
    Abstract: The in-situ vaccine using CpG oligodeoxynucleotide combined with OX40 agonist antibody (CpG + OX40) has been shown to be an effective therapy activating an anti-tumor T cell response in certain settings. The roles of tumor volume, tumor model, and the addition of checkpoint blockade in the efficacy of CpG + OX40 in-situ vaccination remains unknown. Methods Mice bearing flank tumors (B78 melanoma or A20 lymphoma) were treated with combinations of CpG, OX40, and anti-CTLA-4. Tumor growth and survival were monitored. In vivo T cell depletion, tumor cell phenotype, and tumor infiltrating lymphocyte (TIL) studies were performed. Tumor cell sensitivity to CpG and macrophages were evaluated in vitro. Results As tumor volumes increased in the B78 (one-tumor) and A20 (one-tumor or two-tumor) models, the anti-tumor efficacy of the in-situ vaccine decreased. In vitro, CpG had a direct effect on A20 proliferation and phenotype and an indirect effect on B78 proliferation via macrophage activation. As A20 tumors progressed in vivo, tumor cell phenotype changed, and T cells became more involved in the local CpG + OX40 mediated anti-tumor response. In mice with larger tumors that were poorly responsive to CpG + OX40, the addition of anti-CTLA-4 enhanced the anti-tumor efficacy in the A20 but not B78 models. Conclusions Increased tumor volume negatively impacts the anti-tumor capability of CpG + OX40 in-situ vaccine. The addition of checkpoint blockade augmented the efficacy of CpG + OX40 in the A20 but not B78 model. These results highlight the importance of considering multiple preclinical model conditions when assessing the efficacy of cancer immunotherapy regimens and their translation to clinical testing.
    Type of Medium: Online Resource
    ISSN: 0340-7004 , 1432-0851
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    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 ...
  • 7
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 12 ( 2021-11-15)
    Abstract: Combining CpG oligodeoxynucleotides with anti-OX40 agonist antibody (CpG+OX40) is able to generate an effective in situ vaccine in some tumor models, including the A20 lymphoma model. Immunologically “cold” tumors, which are typically less responsive to immunotherapy, are characterized by few tumor infiltrating lymphocytes (TILs), low mutation burden, and limited neoantigen expression. Radiation therapy (RT) can change the tumor microenvironment (TME) of an immunologically “cold” tumor. This study investigated the effect of combining RT with the in situ vaccine CpG+OX40 in immunologically “cold” tumor models. Methods Mice bearing flank tumors (A20 lymphoma, B78 melanoma or 4T1 breast cancer) were treated with combinations of local RT, CpG, and/or OX40, and response to treatment was monitored. Flow cytometry and quantitative polymerase chain reaction (qPCR) experiments were conducted to study differences in the TME, secondary lymphoid organs, and immune activation after treatment. Results An in situ vaccine regimen of CpG+OX40, which was effective in the A20 model, did not significantly improve tumor response or survival in the “cold” B78 and 4T1 models, as tested here. In both models, treatment with RT prior to CpG+OX40 enabled a local response to this in situ vaccine, significantly improving the anti-tumor response and survival compared to RT alone or CpG+OX40 alone. RT increased OX40 expression on tumor infiltrating CD4+ non-regulatory T cells. RT+CpG+OX40 increased the ratio of tumor-infiltrating effector T cells to T regulatory cells and significantly increased CD4+ and CD8+ T cell activation in the tumor draining lymph node (TDLN) and spleen. Conclusion RT significantly improves the local anti-tumor effect of the in situ vaccine CpG+OX40 in immunologically “cold”, solid, murine tumor models where RT or CpG+OX40 alone fail to stimulate tumor regression.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. 6 ( 2021-06), p. e002715-
    Abstract: Current clinical trials are using radiation therapy (RT) to enhance an antitumor response elicited by high-dose interleukin (IL)-2 therapy or immune checkpoint blockade (ICB). Bempegaldesleukin (BEMPEG) is an investigational CD122-preferential IL-2 pathway agonist with prolonged in vivo half-life and preferential intratumoral expansion of T effector cells over T regulatory cells. BEMPEG has shown encouraging safety and efficacy in clinical trials when used in combination with PD-1 checkpoint blockade. In this study, we investigated the antitumor effect of local RT combined with BEMPEG in multiple immunologically ‘cold’ tumor models. Additionally, we asked if ICB could further enhance the local and distant antitumor effect of RT+BEMPEG in the setting of advanced solid tumors or metastatic disease. Methods Mice bearing flank tumors (B78 melanoma, 4T1 breast cancer, or MOC2 head and neck squamous cell carcinoma) were treated with combinations of RT and immunotherapy (including BEMPEG, high-dose IL-2, anti(α)-CTLA-4, and α-PD-L1). Mice bearing B78 flank tumors were injected intravenously with B16 melanoma cells to mimic metastatic disease and were subsequently treated with RT and/or immunotherapy. Tumor growth and survival were monitored. Peripheral T cells and tumor-infiltrating lymphocytes were assessed via flow cytometry. Results A cooperative antitumor effect was observed in all models when RT was combined with BEMPEG, and RT increased IL-2 receptor expression on peripheral T cells. This cooperative interaction was associated with increased IL-2 receptor expression on peripheral T cells following RT. In the B78 melanoma model, RT+BEMPEG resulted in complete tumor regression in the majority of mice with a single ~400 mm 3 tumor. This antitumor response was T-cell dependent and supported by long-lasting immune memory. Adding ICB to RT+BEMPEG strengthened the antitumor response and cured the majority of mice with a single ~1000 mm 3 B78 tumor. In models with disseminated metastasis (B78 primary with B16 metastasis, 4T1, and MOC2), the triple combination of RT, BEMPEG, and ICB significantly improved primary tumor response and survival. Conclusion The combination of local RT, BEMPEG, and ICB cured mice with advanced, immunologically cold tumors and distant metastasis in a T cell-dependent manner, suggesting this triple combination warrants clinical testing.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4440-4440
    Abstract: Brain metastases develop in many patients with advanced metastatic cancers, and this challenging clinical scenario demands improved treatment approaches. We previously observed that an in situ vaccination (ISV) regimen combining local radiation (RT) and intratumoral (IT) injection of the radiated tumor site with interleukin-2 (IL2) resulted in enhanced response to immune checkpoint inhibition (ICI: αCTLA4 Ab) in immunologically cold murine models of melanoma. This ISV treatment resulted in complete response (CR) at both the ISV-targeted tumor and at distant extracranial tumor sites in most mice. However, in a model of melanoma brain metastases, this same ISV provided only a modest improvement in survival with all mice succumbing to progressive tumor burden in the brain (Sriramaneni et al., 2020 AACR submitted abstract). Bempegaldesleukin (NKTR-214, Nektar Therapeutics) is a PEGylated derivative of IL2 that can be administered systemically with minimal toxicity, has a longer half-life (~20 hours), and preferentially activates cytotoxic (CD8+) T cells and NK cells over regulatory T-cells within the tumor microenvironment. We hypothesized that these qualities of NKTR-214 could enhance immune response and efficacy of ISV against brain melanoma metastases, and therefore we tested in combination with local RT targeting an extracranial tumor site. Immunologically cold B78 melanoma tumor cells were implanted into the right flank and the right striatum of the brain in syngeneic C57BL/6 mice. Right flank tumors (150-200 mm3) were irradiated (12 Gy) on treatment day 1. NKTR-214 was administered intravenously on treatment days 6, 15, 24. NKTR-214 combined with RT significantly extended survival in these mice, compared to RT or NKTR-214 alone (p=0.008). Twenty percent of these mice treated with right flank RT + NKTR-214 exhibited durable CR at all tumor sites, whereas no CR was observed in mice treated with RT or NKTR-214 alone. Using this same regimen, we treated mice bearing a right flank B78 melanoma tumor (targeted by local RT) and non-radiated B78 melanoma tumors on the left flank and in the brain. In these mice, we compared the immune response in the brain and left flank tumors following treatment with right flank RT + systemic NKTR-214. Analysis of these tumors by microscopy at treatment day 15 demonstrated significantly increased CD8+ T cell infiltrate and an increased ratio of CD8+ T cells: FOXP3+ regulatory T cells in both the left flank and brain tumors compared to untreated control mice. In contrast to our prior observations with RT + IT-IL2, the combination of RT + NKTR-214 triggered a comparable immune response in both the brain and left flank tumors with respect to the degree of CD8+ T cell infiltration and the ratio of CD8+ effector: FOXP3+ regulatory T cells. These results suggest that a combination of RT + NKTR-214 may elicit an ISV effect that is capable of conferring enhanced anti-tumor immune response against brain metastases, and could potentially abrogate the need for direct brain radiation or surgery. Citation Format: Paul A. Clark, Raghava N. Sriramaneni, Alexander Pieper, Amber M. Bates, Bryce R. Anderson, Wonjong Jin, Justin C. Jagodinsky, Alexander L. Rakhmilevich, Zachary S. Morris. Bempegaldesleukin (NKTR-214), a CD122 preferential IL-2 pathway agonist, augments the in situ vaccine response to radiation of an extracranial tumor in a murine melanoma model, conferring response at non-radiated tumor sites in the brain [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 4440.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1210-1210
    Abstract: Regulated proteolysis of the tolerogenic matrix proteoglycan versican (VCAN) through the actions of ADAMTS-proteases, is associated with enhanced CD8+ infiltration in both hematopoietic and solid tumors. However, it is unclear whether the enhanced CD8+ infiltration results from proteolysis-mediated depletion of precursor VCAN at the tumor site or from generation of bioactive proteolytic fragments ("matrikines") (e.g., the 441-aa N-terminal fragment of V1-VCAN isoform, versikine). We have previously shown that versikine promotes Batf3-dendritic cell (DC) generation from FLT3L-mobilized bone marrow (BM) progenitors in vitro. However, the effects of versikine in DC homeostasis in the tumor microenvironment in vivo are unknown. To investigate the effects of versikine in DC homeostasis in vivo, we utilized the first Ras-driven myeloma (MM) model (VQ model- Rajagopalan et al., Blood 132:1006, 2018) as well as transplantable solid tumor models in both C57BL/6J (LLC lung carcinoma) and Balb/c (4T1 mammary carcinoma) backgrounds. Tumor cells were stably engineered to secrete HA-tagged versikine vs. empty-vector (EV) controls. EV-VQ or versikine-VQ myeloma cells were implanted intracardiacally into C57BL/6J syngeneic recipients and mice were monitored until they developed myeloma-related end-organ damage (hindlimb paralysis). Both groups of mice were paralyzed at similar rates. Intratumoral conventional DCs (CD138-CD45+, CD11chi,MHC IIhiLy6C-, CD64-) clustered into two populations: cDC1 (Batf3-DC: CD24hi,CD11blo), a subset with crucial activity in cross-priming anti-tumor CD8+ T cells, and cDC2 (CD24lo, CD11bhi). Versikine enhanced intratumoral Batf3-DC frequency/infiltration, while cDC2 levels were diminished in versikine-VQ BM (Figure 1A)(Batf3-DC: 48% in EV-VQ vs. 72% in versikine-VQ, p-value= 0.0246; cDC2: 52% in EV-VQ vs. 28% in versikine-VQ, p=0.0312). Monocytic-derived DC (Mo-DC: CD11chi, MHC IIhi, Ly6C+, CD64+) frequency remained unchanged. Versikine's effects were replicated in 2 solid tumor models. Versikine-expressing tumors were characterized by significantly enhanced Batf3-DC infiltration (Fig. 1A, p-value= 0.0079 for 4T1 model and 〈 0.0001 for LLC model), whereas cDC2 numbers were diminished (p-value: 0.0079 and 〈 0.0001 respectively). Adoptive transfer of CD45.2+ pre-DC (SIRPaint, FLIT3+, CD11c+, MHC II-, Celltrace+) in LLC-EV and LLC-versikine tumors in CD45.1+ recipients did not show any differences in 3-day differentiation potential of DC precursors, implicating other mechanisms to explain the steady-state imbalance in DC subset frequencies. To examine whether versikine's effects on the intratumoral DC milieu in vivo could be therapeutically harnessed, we compared responses to STING agonist therapy between versikine-expressing and EV tumors. LLC-EV-OVA and LLC-versikine-OVA (ovalbumin, a model antigen) -expressing tumors received therapeutic intratumoral injections of DMXAA, a murine STING agonist. Analysis of splenocytes 5 days later showed a significant increase in the frequency of OVA antigen-specific, CD8+ (MHCI:SIINFEKL tetramer+) splenocytes in LLC-versikine-bearing animals (Figure 1B). Interestingly, there was a marked increase in total central memory T splenocytes (TCM) (CD62LhiCD44hi) harvested from LLC-versikine tumor-bearing mice. We conclude that versikine influences the DC milieu in the tumor bed with promotion of intratumoral cross-presenting Batf3-DC and depletion of the cDC2 subset. Our findings highlight an unappreciated facet of immune regulation of the tumor microenvironment through matrix proteolytic fragments ("matrikines"). Whereas detection of native VCAN proteolysis on myeloma biopsies (see abstract by Dhakal et al.in this meeting) portends adverse outcomes likely due to the tolerogenic effects of accumulated precursor VCAN at the tumor site, therapeutic use of the isolated, purified fragments may promote tumor innate sensing and effector priming. VCAN-matrikines, through their effects on intratumoral Batf3-DC and antigen-specific CD8+ T cell infiltration, may potentiate in situ vaccination strategies across diverse hematopoietic and solid tumor types. Figure 1 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: 2019
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages