Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Type of Medium
Language
  • 1
    In: Cancer Cell, Elsevier BV, Vol. 33, No. 4 ( 2018-04), p. 649-663.e4
    Type of Medium: Online Resource
    ISSN: 1535-6108
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 2074034-7
    detail.hit.zdb_id: 2078448-X
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Immunity, Elsevier BV, Vol. 46, No. 4 ( 2017-04), p. 577-586
    Type of Medium: Online Resource
    ISSN: 1074-7613
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 2001966-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 383-383
    Abstract: Introduction Patients with acute lymphoblastic leukaemia (ALL) relapsing after allogeneic stem cell transplantation (SCT) have a dismal prognosis. Recent clinical trials with T cells engineered to express 2nd generation CD19 chimeric antigen receptors (CARs) incorporating co-stimulatory domains for improved persistence and expansion report unprecedented anti-leukemic responses. However, responses are associated with Cytokine Release Syndrome (CRS) due to supra-physiological activation of the redirected T-cells. As an alternative, we studied use of donor-derived Epstein Barr virus (EBV)-specific T cells (CTL) transduced with a 1st generation CD19CAR as effectors, relying on signalling through the endogenous T cell receptor (TCR) to drive more physiological proliferation and persistence. This has enabled us to investigate a novel strategy to facilitate the expansion/persistence of CD19CAR T cells by vaccination with irradiated donor-derived, EBV transformed lymphoblastoid cell lines (LCL). We are conducting a European multi-centre phase I/II study of this approach in patients with pediatric ALL relapsing after SCT and report our interim findings. Methods Donor-derived EBV-specific CTL were generated from 80mls donor blood by repetitive stimulation with LCL, followed by transduction with an SFG retroviral vector encoding a CD19CAR consisting of the FMC63 single chain Fv linked to a CD3ζ endodomain. Patients were eligible for CD19CAR CTL therapy either pre-emptively if they became MRD-positive ( 〉 5 x 10-4 in BM) within the 1st year post-SCT or prophylactically at day 60-70 post-2nd SCT. All patients had early withdrawal of immunosuppression and received lymphodepletion with fludarabine 90 mg/m2. Patients with detectable residual disease also received cytoreduction with vincristine/dexamethasone prior to infusion of cryopreserved CD19CAR CTL. Persistence of CAR CTL was measured by quantitative PCR and flow cytometry of blood. Disease status was assessed by morphology and IgH MRD analysis on bone marrow samples. The study design incorporated an interim analysis, allowing for vaccination with irradiated LCL if CD19CAR CTL were not detectable in 50% of patients at 2 months post-infusion. Results So far, 20 patients have been recruited (14 pre-emptive, 6 prophylactic arm) and 7 patients treated (3 pre-emptive, 4 prophylactic). The infused cell dose was 2 x 108/m2 in 6 patients and 4 x 107/m2 in the other. CD19CAR expression varied from 12.1-58.9%. No grade 3-5 toxicity was noted. In particular, no CRS, neurotoxicity or graft versus host disease (GVHD) attributable to CD19CAR CTL was seen. B-cell depletion was transient, lasting 1-2 months. In terms of disease response, 2 patients treated prophylactically remain in MRD negative remission after 3 and 17 months’ follow-up. A further patient showed transient clearance of BM MRD following immunotherapy in association with EBV viremia. He subsequently relapsed but has stable disease after retreatment with CD19CAR CTL with LCL vaccination. The other 4 patients had disease progression between 2 weeks and 3 months post-CD19CAR CTL infusion. At a median follow-up of 8 months, 2 patients have died of relapse, 3 are alive with disease and 2 remain disease-free. A planned interim analysis of the initial 6 patients treated with CD19CAR CTL alone showed poor expansion/persistence of CD19CAR CTL which were only detectable in the blood in 1 patient up to 28 days post-infusion. This may reflect that only 1 patient had EBV viremia at the time CD19CAR CTL were infused. In view of this, a second trial cohort received subcutaneous vaccination with irradiated, donor-derived LCL at 2 days before and at 1 and 2 months following CD19CAR CTL infusion to provide signalling through the endogenous EBV-specific TCR. So far, 2 patients have been vaccinated and a 3rd is planned shortly. Data on the effect of vaccination on CD19CAR CTL expansion/persistence will be presented. Conclusions This ongoing study shows safety of adoptive immunotherapy with donor EBV CTL transduced with a 1st generation CD19CAR in paediatric patients with ALL relapsing post allo-SCT. However, in the absence of a co-stimulatory domain in vivo expansion and persistence of transferred CTL is poor. We are investigating whether vaccination with irradiated, donor-derived EBV LCL improves persistence and efficacy of CAR transduced T cells and initial data on this approach will be presented Disclosures Pule: Cellectis: Martin Pule's laboratory receives funding for contract research from Cellectis Therapeutics Other.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 1999
    In:  Irish Journal of Medical Science Vol. 168, No. 2 ( 1999-4), p. 151-154
    In: Irish Journal of Medical Science, Springer Science and Business Media LLC, Vol. 168, No. 2 ( 1999-4), p. 151-154
    Type of Medium: Online Resource
    ISSN: 0021-1265 , 1863-4362
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 1999
    detail.hit.zdb_id: 2275855-0
    detail.hit.zdb_id: 2468506-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 115, No. 2 ( 2010-01-14), p. 396-407
    Abstract: Immunotherapy with allodepleted donor T cells (ADTs) improves immunity after T cell–depleted stem cell transplantation, but infection/relapse remain problematic. To refine this approach, we characterized the expression of surface markers/cytokines on proliferating alloreactive T cells (ATs). CD25 was expressed on 83% of carboxyfluorescein diacetate succinimidyl esterdim ATs, confirming this as an excellent target for allodepletion. Seventy percent of CD25− ATs expressed CD71 (transferrin receptor), identifying this as a novel marker to target ATs persisting after CD25 depletion. Comparison of residual alloreactivity after combined CD25/71 versus CD25 immunomagnetic depletion showed enhanced depletion of alloreactivity to host with CD25/71 depletion in both secondary (2°) mixed lymphocyte reactions (P 〈 .01) and interferon-γ enzyme-linked immunospot assays (P 〈 .05) with no effect on third-party responses. In pentamer/interferon-γ enzyme-linked immunospot assays, antiviral responses to cytomegalovirus, Epstein-Barr virus, and adenovirus were preserved after CD25/71 allodepletion. CD25/71 ADTs can be redirected to recognize leukemic targets through lentiviral transfer of a chimeric anti-CD19ζ T-cell receptor. Finally, we have established conditions for clinically applicable CD25/71 allodepletion under European Union Good Manufacturing Practice conditions, resulting in highly effective, reproducible, and selective depletion of ATs (median residual alloreactivity to host in 2° mixed lymphocyte reaction of 0.39% vs third-party response of 62%, n = 5). This strategy enables further clinical studies of adoptive immunotherapy with larger doses of ADTs to enhance immune reconstitution after T cell-depleted stem cell transplantation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 3008-3008
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3008-3008
    Abstract: Abstract 3008 In vivo selection of mycophenolate mofetil-resistant T cells for adoptive immunotherapy. Background Following allogeneic solid organ or hematopoietic stem cell transplantation, adoptive transfer of therapeutic T cells may be hindered by the requirement for immune suppressive drugs to prevent rejection or graft-versus-host disease. Mycophenolate mofetil (MMF) is a non-competitive inhibitor of inosine-5'-monophosphate dehydrogenase 2 (IMPDH2), an inducible enzyme that generate guanine nucleotides for DNA and RNA synthesis in T cells. In this study, we have evaluated the potential for gene transfer to T cells of a mutated IMPDH2 that confers 〉 2000-fold resistance to MMF (IMPDH2R; T333I, S351Y). Methods Wild type IMPDH2WT, IMPDH2R and IMPDH2 with a catalytic site mutation (IMPDH2CS; C331A) were cloned into SFG retroviral vectors as fusions to eGFP reporter sequences. Murine thymoma (BW 5147) or CD8 T cells were transduced with each vector and their phenotype and function evaluated in the presence or absence of mycophenolic acid (MPA), the active metabolite of MMF. Results BW thymoma cells transduced with IMPDH2R exhibited less apoptosis than cells transduced with IMPDH2CS in response to MPA (ratio % Annexin V+ MPA: no MPA- IMPDH2R = 1.2; IMPDH2CS = 2.9, p=0.01). Cells transduced with IMPDH2R were also able to overcome the G1 cell-cycle arrest induced by MPA when compared to control IMPDH2CS cells (Ratio %cells in S-G2/M phases MPA: no MPA- IMPDH2R = 1.0; IMPDH2CS = 0.3, p=0.03). This led to selective enrichment of IMPDH2R transduced cells in the presence of MPA. At low dose MPA (450nM), IMPDH2R transduced cells enriched compared to IMPDH2CS but not IMPDH2WT (ratio % GFP MPA: no MPA- IMPDH2R = 1.6 vs IMPDH2CS = 1.1, p=0.04 and IMPDH2WT = 1.5 p= 0.14). However, at high dose (4500nM) IMPDH2R exhibited enhanced enrichment (ratio % GFP MPA: no MPA- IMPDH2R = 2.8 vs IMPDH2CS = 1.0 p=0.03 and vs IMPDH2WT= 1.5 p=0.03). Gene transfer of IMPDH2R into murine CD8 T cells also led to selective enrichment compared to controls in the presence of MPA when cultured with proliferation-inducing common gamma-chain cytokines (ratio MPA: no MPA IMPDH2R = 4.4 vs. IMPDH2CS = 1.10, p=0.02). Strong selection for IMPDH2R-tranduced CD8 OT-1 T cells in the presence of MPA was also observed under conditions of antigen-induced proliferation (ratio IMPDH2R= 7.1, control = 0.8, p=0.002). To assess in vivo selection, sub-lethally irradiated (2Gy) B6.PL (Thy1.1) mice were injected with a 1:1 mix of OT1 TCR transgenic Thy1.2 CD8 T cells transduced with IMPDH2R or IMPDH2CS that could be differentiated by the congenic markers CD45.1 and CD45.2. Transferred cells were stimulated by s.c. injection with cognate peptide (SIINFEKL) in IFA and MMF (200mg/kg/day) was given by daily ip injection. As in vitro, IMPDH2R-transduced OT1 cells were preferentially selected over IMPDH2CS-transduced cells following MMF treatment (day 14 ratio IMPDH2R to IMPDHCSwas 19.3 vs 1.2 in the absence of MMF). Conclusions T cells transduced with IMPDH2R are resistant to the anti-proliferative and apoptotic effects of MPA in vitro and demonstrate strong selection in vivo compared to controls at therapeutic levels of MMF. These data support the potential of conferring MMF resistance as a strategy to permit the survival of therapeutic T cells in immunosuppressed allograft recipients. Disclosures: Stauss: Cell Medica: Scientific Advisor Other.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 114, No. 23 ( 2009-11-26), p. 4792-4803
    Abstract: Epstein-Barr virus (EBV)–driven posttransplantation lymphoproliferative disease (PTLD) is a serious complication of immunosuppression after either stem cell transplantation (SCT) or solid organ transplantation (SOT). Adoptive transfer of EBV-specific cytotoxic T lymphocytes (EBV-CTLs) is an effective prophylaxis and treatment for PTLD after SCT, but not for PTLD after SOT when pharmacologic immunosuppression cannot be discontinued. We report the generation of calcineurin (CN) mutants that render EBV-CTL resistant to the immunosuppressants tacrolimus (FK506) and cyclosporin A (CsA): mutant CNa12 confers resistance to CsA but not FK506, and mutant CNa22 confers resistance to FK506 but not CsA, whereas mutant CNb30 renders CTLs resistant to both calcineurin inhibitors. Untransduced EBV-CTLs do not proliferate in the presence of FK506/CsA. However, EBV-CTLs transduced with a retroviral vector coding for these mutants retain the ability to both proliferate and secrete normal levels of interferon-γ in the presence therapeutic levels of FK506 (CNa12), CsA (CNa22), or both (CNb30). The cytotoxicity and phenotype of EBV-CTL lines were unaffected by expression of these mutant CNs. This approach should allow effective immunotherapy with EBV-CTLs in the SOT setting without risking the graft by reduction in immunosuppression, and represents a generic approach to improving immunotherapy in the face of immunosuppression.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 115, No. 5 ( 2010-02-04), p. 925-935
    Abstract: T-cell immunotherapy that takes advantage of Epstein-Barr virus (EBV)–stimulated immunity has the potential to fill an important niche in targeted therapy for EBV-related cancers. To address questions of long-term efficacy, safety, and practicality, we studied 114 patients who had received infusions of EBV-specific cytotoxic T lymphocytes (CTLs) at 3 different centers to prevent or treat EBV+ lymphoproliferative disease (LPD) arising after hematopoietic stem cell transplantation. Toxicity was minimal, consisting mainly of localized swelling at sites of responsive disease. None of the 101 patients who received CTL prophylaxis developed EBV+ LPD, whereas 11 of 13 patients treated with CTLs for biopsy-proven or probable LPD achieved sustained complete remissions. The gene-marking component of this study enabled us to demonstrate the persistence of functional CTLs for up to 9 years. A preliminary analysis indicated that a patient-specific CTL line can be manufactured, tested, and infused for $6095, a cost that compares favorably with other modalities used in the treatment of LPD. We conclude that the CTL lines described here provide safe and effective prophylaxis or treatment for lymphoproliferative disease in transplantation recipients, and the manufacturing methodology is robust and can be transferred readily from one institution to another without loss of reproducibility. The current trial was registered at www.clinicaltrials.gov as #NCT00058812.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 5747-5747
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 5747-5747
    Abstract: Background: CD22 is an attractive target for Chimeric Antigen Receptor (CAR) since it is expressed by most B-cell malignancies. However, CAR targeting of CD22 is challenging since CD22 has a long ectodomain of 300 Å, which is several times wider than the optimal immune synapse. Further, the ectodomain is rigid, so even targeting of membrane proximal domains may not allow effective synapse formation. Further still, CD22 is expressed at low density and dropping of antigen density has been described as a mode of escape. A previously described CD22 CAR based on the M971 antibody required a very short linker between single-chain VH-VL for sensitive function, presumably to allow CAR concatenation but which comes at the cost of basal signalling. Our aim was to engineer an anti-CD22 CAR which triggered not only killing, but proliferation in response to less than 1000 CD22 molecules per cell, without basal signalling. We conducted a functional screen for stable / high affinity binders recognizing the CD22 membrane proximal domains that function in a CAR against cells expressing low levels of CD22 target. Methods: Binders were generated in two campaigns. Hyperimmune mice were vaccinated with recombinant CD22 protein lacking the highly immunogenic N-terminal V-type domain. Similarly, Wistar rats were immunized via genetic vaccination using a modified gene encoding the 4 membrane proximal domains of CD22, cloned into a DNA vaccine plasmid designed to stimulate a humoral immune response. A total of 18 binders passed biophysical screening properties of stability, affinity and proximal binding. Binding affinity ranged between 1-36.5nM; the binders were cloned into 41BB-Zeta signalling CARs. We generated a panel of CD22 expressing SupT1 cell lines. Using expression cassettes with amber stop codons preceding the CD22 frame, we generated stable SuptT1 cell lines which express CD22 below the limit of amplified flow cytometry detection ( 〈 100 copies, CD22Und) and a cell line with 255 copies / cell (CD22Low). In addition, using conventional means, we generated cell lines expressing intermediate and high levels of CD22 expression (CD22Mid=3,444, CD22High=78,916 copies/cell). Results: The 18 CAR constructs went through an initial round of screening using primary T-cells as effectors and engineered SupT1 cells as targets. A threshold of 〉 50% killing of CD22Und target cells at an effector:target (E:T) ratio of 1:4 at 72 hour was set for further characterization. Only one CAR derived from the hyperimmune murine binders, and two derived from the rat binders met this threshold. Further characterization of killing at lower E:T ratios, cytokine release and proliferation revealed that CAR based on the rat binder 9A8 had superior function in response to low density target cells compared with the other two CARs and was characterized further and compared with the M971 CAR. This comparison showed approximately 2-fold better killing of CD22Und 9A8, and approximately 4-fold increased proliferation with no basal signalling. Next, we tried to correlate binder characteristics with sensitivity to low CD22 density. No correlation was found between either affinity, epitope, or stability and sensitivity. In fact, two of the best candidates could be paired with CARs which recognized the same CD22 domain and had similar binding kinetics and stability but had much lower sensitivity to CD22. This suggests that some other as yet unappreciated binder characteristic leads to sensitivity and we are currently investigating this via a structural analysis of binder / target interaction. Finally, we are currently testing 9A8 co-expressed with the CAT19 CD19 CAR (AUTO1) (Blood 2017 130:806) in small animal models of B-ALL. Conclusion: A highly sensitive CD22 CAR could be selected by empiric screening of CARs derived from a set of binders which target the proximal domains of CD22. CAR concatenation was not required for a high-degree of sensitivity to CD22 antigen. No correlation was found between binder characteristics and function. Disclosures Thomas: Autolus: Employment, Equity Ownership. Cordoba:Autolus: Employment, Equity Ownership. Pule:Autolus: Employment, Equity Ownership, Patents & Royalties.
    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