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
    In: Zeitschrift für Medizinische Physik, Elsevier BV, Vol. 15, No. 3 ( 2005), p. 192-198
    Type of Medium: Online Resource
    ISSN: 0939-3889
    Language: German
    Publisher: Elsevier BV
    Publication Date: 2005
    detail.hit.zdb_id: 2231492-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Systematic and Applied Microbiology, Elsevier BV, Vol. 37, No. 2 ( 2014-03), p. 79-88
    Type of Medium: Online Resource
    ISSN: 0723-2020
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 283612-9
    detail.hit.zdb_id: 2046331-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 647-647
    Abstract: Abstract 647 Imatinib is a selective and very potent inhibitor of the BCR/ABL kinase. It induces ongoing complete cytogenetic remissions in the vast majority of chronic phase CML patients. However, BCR/ABL persistence is the rule despite ongoing imatinib therapy. This suggests that imatinib will not to cure CML and raises concerns about emerging imatinib resistance, long-term imatinib tolerability and compliance to therapy. We previously suggested that a combination of imatinib and immunotherapy by IFN may additionally control CML via induction of autologous cytotoxic T-cell (CTL) responses, such as those directed against the leukemia-associated antigen proteinase 3 (PR3). For example, induction of PR1-CTL which recognize PR3 on CML blasts was previously shown to be associated with IFN-, but not imatinib response. Indeed, we could recently demonstrate on a cohort of 20 newly diagnosed CML patients that low dose of IFN maintenance therapy alone was able to maintain or improve remissions obtained by a prior imatinib/IFN combination treatment (A. Hochhaus et al. , ASH 2007). After a median time of IFN maintenance therapy of 1.2 years 80% of the patients remained or improved molecular remission. Here we report a significantly longer follow up of these patients and translational studies to examine markers of IFN response. Twenty pts (14 m, 6 f; median age 45, range 23-74 yrs) with low (n=13), intermediate (n=6), and high risk (n=1) according to the Hasford score risk calculation have been investigated. Imatinib therapy had been administered for 2.4 yrs (0.2-4.9), combined with PEG-IFNa2a (Pegasys®, n=17) or IFN a2a (Roferon®, n=3). Maintenance therapy consisted of PEG-IFN (n=16) or IFN (n=4). Dose was adjusted according to response and tolerability and ranged between 135 μg PEG-IFN every 3 weeks to 180 μg PEG-IFN every week, or alternatively 2 to 5 * 3 Mill IU IFN/week. Imatinib was terminated due to side effects (n=5) or upon personal request of the patients after informed consent (n=15). At the time of imatinib withdrawal, two pts were in complete molecular remission (CMR) and 15 pts in major molecular remission (MMR). After a median observation time of 2.8 yrs (range 0.5-4.5), 15 pts were in MMR, 5 of them in CMR. Thus, the number of MMR patients increased from 2 at baseline to 5 after two years. Five patients relapsed within 0.4 years (range, 0.2-0.8) after imatinib discontinuation, but were rescued with imatinib, re-establishing molecular remission. Side effects to maintenance IFN were minor. We also studied putative markers of IFN response. IFN therapy was associated with an increase in the expression of PR3, and in the presence of auto-reactive PR1-CTL. PR1-CTL frequencies were prospectively assessed without prior in vitro amplification. In one of five assessable patients PR1-CTL were detected prior to imatinib withdrawal, but in four of seven assessable patients during IFN maintenance therapy. Longitudinal measurements of PR-1 CTL counts suggested an inhibition of the expansion of PR1-CTL by imatinib, implying that an optimal CTL expansion may occur preferentially in the absence of imatinib. This would explain the conversion to a CMR status in some patients only after imatinib withdrawal. Together, IFN maintenance after a prior imatinib/IFN induction therapy may be an effective alternative to permanent imatinib therapy, because it enables to safely discontinue imatinib even in those patients that have not achieved a CMR at the time of pausing imatinib. Induction of a PR1-specific CTL response by IFN may contribute to the particular efficacy of IFN after CML-debulking by imatinib. Disclosures: Burchert: Roche: Research Funding. Off Label Use: Pegasys(R) therapy in CML. Neubauer:Roche: Research Funding. Hochhaus:Roche, Novartis: Research Funding.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 28, No. 8 ( 2010-03-10), p. 1429-1435
    Abstract: Imatinib induces sustained remissions in patients with chronic myelogenous leukemia (CML), but fails to eradicate CML stem cells. This is of major concern regarding the issues of cure, long-term imatinib tolerability, and imatinib resistance. We therefore asked whether interferon alfa-2a (IFN) alone could maintain molecular remissions achieved by a prior combination therapy with imatinib and IFN. Patients and Methods Imatinib therapy was stopped in 20 patients who had concomitantly been pretreated with imatinib and IFN for a median of 2.4 years (range, 0.2 to 4.8 years) and 2.5 years (range, 0.2 to 4.9 years), respectively. After imatinib discontinuation, remission status was monitored monthly by quantitative analysis of the peripheral-blood BCR-ABL mRNA levels using real-time polymerase chain reaction. Proteinase-3 expression and proteinase-3–specific cytotoxic T cells (CTLs) were longitudinally measured to assess putative markers of IFN response. Results With a median time of 2.4 years after imatinib withdrawal (range, 0.5 to 4.0 years), 15 (75%) of 20 patients remained in remission. The number of patients in complete molecular remission increased under IFN from two patients at baseline to five patients after 2 years. Relapses occurred in five patients within 0.4 years (range, 0.2 to 0.8 years), but patients underwent rescue treatment with imatinib, re-establishing molecular remission. IFN therapy was associated with an increase in the expression of leukemia-associated antigen proteinase 3 and induction of proteinase-3–specific CTLs. Conclusion Treatment with IFN enables discontinuation of imatinib in most patients after prior imatinib/IFN combination therapy and may result in improved molecular response. Induction of a proteinase-3–specific CTL response by IFN may contribute to this effect.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2010
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Analytical Chemistry, American Chemical Society (ACS), Vol. 94, No. 12 ( 2022-03-29), p. 4988-4996
    Type of Medium: Online Resource
    ISSN: 0003-2700 , 1520-6882
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2022
    detail.hit.zdb_id: 1483443-1
    detail.hit.zdb_id: 1508-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 36-36
    Abstract: Abstract 36 Introduction: Chronic myeloid leukemia (CML) is caused by the BCR-ABL oncogene. CML patients lack expression of IRF-8 - an interferon-regulated transcription factor that has been shown to exert tumor suppressor functions. IRF-8 is also critical for the development of a rare dendritic cell population, so called plasmocytoid dendritic cells (PDC). PDC are quantitatively significantly reduced or absent in the peripheral blood of first diagnosis CML patients. PDC are also the major producers of IFN-alpha (IFNa) in man. IFNa is a cytokine that has significant therapeutic efficiency in the treatment of CML patients. We here wished to experimentally test, whether BCR-ABL expression and loss of IRF-8 may be causally linked to a reduction of PDC in murine CML and whether there could be any functional relevance for PDC loss in CML development or treatment. Methods: PDC counts were studied from peripheral blood samples of primary CML patients at diagnosis, at the time of remission or from healthy donors. PDC function was assessed in vitro by treatment of magnetic bead-enriched PDC with Toll-like receptor 9-specific oligos (ODN 2216) and subsequent assessment of the intracellular IFNa expression in stimulated PDC. A supposed link between BCR-ABL expression, IRF-8 repression and loss of PDC counts was studied in vivo using a murine CML transduction-transplantation model (C57/Bl6 mice, 7Gy sub-lethal irradiation for conditioning). Multiparameter flow cytometry and cell sorting were performed to analyze and enrich, BCR-ABL-positive (GFP+) hematopoietic subpopulations and PDC in order to then quantitate their IRF-8 and BCR-ABL transcript level by RT-PCR. In order to also test the functional relevance of PDC during CML leukemogenesis, CML mice were injected intravenously, weekly from day +5 after transplantation with in vitro generated PDC. Mice were simultaneously also s.c.-injected weekly with ODN 2216 to stimulate IFNα secretion in adoptively transferred PDC in vivo. Results: As previously reported, newly diagnosed CML patients displayed a significantly reduced PDC count when compared to healthy donors (p 〈 0.001). Upon remission induction with imatinib, PDC counts restored partially, but to a much lesser extend in patients successfully treated with IFNa therapy. Importantly, albeit significantly reduced in number, BCR-ABL-positive first diagnosis CML PDC seem to be functionally intact: CML and healthy donor PDC produced comparable amounts of IFNa in response to Toll-like receptor 9 -specific CpG ODN 2216 stimulation. This suggested that BCR-ABL may compromise PDC function by quantitative rather than qualitative dysregulation. CML mice developed a fatal, BCR-ABL-positive myeloproliferation within 13 to 29 days with 88% penetrance. Compared to control mice (n=8), CML mice (n=14) showed a 7-fold and 3-fold reduction of the frequency of B220+mPDCA-1+ PDC in bone marrow and spleen, respectively. This was associated with a statistically significant (4-fold) suppression of IRF8 mRNA expression in sorted BCR-ABL(GFP)-positive PDC relative to BCR-ABL-negative PDC from the same mice (n=3) or from control transplantations (n=5). By RT-PCR, there was a trend also for lower IRF8 expression in CML progenitor cells (Lin− c-Kit+ Sca-1- GFP+), but not in the stem cell enriching population (Lin− c-Kit+ Sca-1+ GFP+). This implied that IRF8 expression is lost during BCR-ABL-induced leukemogenesis in more mature compartments, which supposedly include PDC precursors. Intriguingly, a once weekly adoptive transfer of in vitro generated (to 〉 30% enriched) PDC for three successive weeks combined with a once weekly subcutaneous injection of CpG ODN 2216 for three weeks was sufficient to almost double survival of CML mice. Conclusions: Using a murine model of CML, we provide first experimental evidence that BCR-ABL induced myeloproliferation is causally linked to a quantitative suppression of PDC, and that this is associated with a BCR-ABL-mediated suppression of IRF8 transcription. Since adoptively transferred PDC were capable of counteracting murine CML development, BCR-ABL may facilitate leukemogenesis in part by obstructing PDC maturation. PDC could thus be a novel immunological effector cell population that exerts and/or integrates anti-leukemic immune responses in CML. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 21 ( 2018-11-01), p. 6223-6234
    Abstract: Plasmacytoid dendritic cells (pDC) are the main producers of a key T-cell–stimulatory cytokine, IFNα, and critical regulators of antiviral immunity. Chronic myeloid leukemia (CML) is caused by BCR-ABL, which is an oncogenic tyrosine kinase that can be effectively inhibited with ABL-selective tyrosine kinase inhibitors (TKI). BCR-ABL–induced suppression of the transcription factor interferon regulatory factor 8 was previously proposed to block pDC development and compromise immune surveillance in CML. Here, we demonstrate that pDCs in newly diagnosed CML (CML-pDC) develop quantitatively normal and are frequently positive for the costimulatory antigen CD86. They originate from low-level BCR-ABL–expressing precursors. CML-pDCs also retain their competence to maturate and to secrete IFN. RNA sequencing reveals a strong inflammatory gene expression signature in CML-pDCs. Patients with high CML-pDC counts at diagnosis achieve inferior rates of deep molecular remission (MR) under nilotinib, unless nilotinib therapy is combined with IFN, which strongly suppresses circulating pDC counts. Although most pDCs are BCR-ABL–negative in MR, a substantial proportion of BCR-ABL+ CML-pDCs persists under TKI treatment. This could be of relevance, because CML-pDCs elicit CD8+ T cells, which protect wild-type mice from CML. Together, pDCs are identified as novel functional DC population in CML, regulating antileukemic immunity and treatment outcome in CML. Significance: CML-pDC originates from low-level BCR-ABL expressing stem cells into a functional immunogenic DC-population regulating antileukemic immunity and treatment outcome in CML. Cancer Res; 78(21); 6223–34. ©2018 AACR.
    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
    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...