GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Society of Hematology  (9)
  • Lu, Sijie  (9)
Materialart
Verlag/Herausgeber
  • American Society of Hematology  (9)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1031-1031
    Kurzfassung: Abstract 1031 Proteinase 3 (P3), a serine protease constitutively expressed in primary granules and on the membrane of some resting granulocytes, is the target of T cell-mediated autoimmunity in Wegener's granulomatosis (WG) and of anti-leukemia immunity mediated by PR1 (VLQELNVTV)-specific cytotoxic T lymphocytes (PR1-CTL). We have previously shown anti-CD3/CD28 induced proliferation of healthy donor T-cells to be significantly inhibited by peripheral blood polymorphonuclear neutrophils (PMNs) expressing membrane P3 (mP3) at a ratio of 3 PMNs to 1 PBMC. Our results indicate that mP3+ PMNs begin to exert inhibitory effects on T cell proliferation at a ratio of 2.2 mP3+ PMNs to 1 PBMC, a ratio greater than that seen in normal homeostatic conditions in peripheral blood. The inhibition was predominantly enzyme-independent and dose-dependent. Notably soluble P3 exerted similar effects on T cells as was seen with mP3. Additionally soluble P3 induced a G0 cell cycle arrest. Of significance, soluble P3 in acute myeloid leukemia (AML) patient serum can be up to 5-fold higher than that seen in healthy control serum. To confirm mP3 specificity, we FAC-sorted PMNs based on the mP3 co-expressed CD177 molecule to obtain highly purified ( 〉 98%) mP3+ and mP3− PMNs. Compared to activated PBMC alone, activated PBMC co-cultured at a ratio of 1:3 with mP3+ PMNs showed 58% and 57% inhibition of CD8+ and CD4+ T cells, respectively (CD8+ and CD4+: p 〈 0.003). PBMC co-cultured at that same ratio with mP3− PMNs showed less inhibition - only 29% and 26% inhibition of CD8+ and CD4+ T cells, respectively (CD8+: p 〈 0.05; CD4+: p=ns). Inhibition of T cell proliferation by both mP3+PMNs and soluble P3 was blocked by anti-P3 mAbs but not by isotype-matched mAb. Furthermore, P3-mediated inhibition of T-cell proliferation is reversible since removal of PMNs or soluble P3 restored the proliferative capacity of the T cells. Because P3 is over-expressed in AML and chronic myeloid leukemia (CML), we hypothesized that mP3+ leukemia may suppress T-cell proliferation. Healthy donor T cell proliferation was studied with CFSE after stimulation with anti-CD3/CD28 mAbs in the presence or absence of mP3+ AML for five days. AML mediated a dose-dependent inhibition of T-cell proliferation contingent upon the level of mP3 expression (p 〈 0.0001). Co-incubation of PBMC with AML displaying 91% mP3 positivity, reduced proliferation of CD8+ T cells to 29.6%, compared to 94.7% in the PBMC culture alone. This inhibition could be completely abrogated by addition of anti-P3 mAb, restoring the proliferation of CD8+ T cells to a level comparable to that seen in control. In contrast, no inhibition of CD8+ T cell proliferation was observed in co-cultures of T cells with AML in which only 6% of the AML cells expressed mP3. Thus, there is an inverse correlation between percent proliferation of T cells and the amount of mP3 on AML (R=0.4539, p 〈 0.0001). In addition, AML expressing 91% mP3+ induced apoptosis of 〉 70% of the T cells at a ratio of 10 AML: 1 PBMC as assessed by uptake of aqua dye. The association between the amount of mP3 on AML and percent of apoptosis was significant (R=0.7852, p 〈 0.0001), and apoptosis induced by mP3+AML appeared to be specific since T cells did not undergo apoptosis when anti-P3 mAb was added. The same correlation was not seen after PMNs from healthy donors (% mP3+: 62%±21.7, n=14) were co-incubated with PBMC. The percentage of cells undergoing apoptosis was less than 10%, regardless of the extent of PMN mP3 positivity. Of note, mP3 expression is significantly higher in bone marrow myeloid derived suppressor cells (MDSC) from leukemia patients compared to MDSC from healthy donors (79.4±5.23% (n=7), 22.4±11.55% (n=3), respectively; p= 0.0007). Because mP3 inhibited proliferation of T cells stimulated via the T cell receptor (TCR), i.e. anti-CD3/CD28, we compared effects of mP3 on ZAP70 and ERK signaling by phosphoflow cytometry. ZAP70 phosphorylation in CD8+ and CD4+ T cells was reduced by 78% and 80%, respectively, within 5 minutes of co-incubation with mP3+ PMNs compared with T cells stimulated with anti-CD3/CD28 mAbs alone, while ERK1/2 phosphorylation was completely blocked within 10 minutes, suggesting that P3-mediated inhibition of T cell proliferation involves downstream TCR signaling pathways. Taken together, these data support an important new function of membrane-bound P3 on PMN and leukemia in controlling adaptive T cell immunity. Disclosures: No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2012
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3761-3761
    Kurzfassung: Immunotherapy targeting individual antigens in acute myeloid leukemia (AML) has shown promise. However, in view of leukemia heterogeneity and the loss of tumor antigen expression by AML, it is unlikely that any single antigen will be consistently expressed by all leukemia cells. This highlights the need to identify additional antigens in AML that can be targeted. Azurophil granule proteases have been shown to be effective immunotherapeutic targets. Proteinase 3 and neutrophil elastase, the parent proteins for the HLA-A2 restricted peptide PR1, have been targeted successfully in myeloid leukemia using immunotherapy. We recently discovered the HLA-A2 restricted peptide CG1 (FLLPTGAEA), which is derived from the azurophil granule protease cathepsin G (CG). We showed that CG is highly expressed by AML blasts and leukemia stem cells in a limited number of AML samples and showed that CG1 can be targeted in vitro using CG-specific cytotoxic T lymphocytes (CTL). We sought to determine whether CG1 can be targeted in vivo and to characterize the expression of CG in a large cohort of AML patients. To assess the efficacy of targeting CG1 in vivo, we used a NOD scid gamma (NSG) mouse model engrafted with the human HLA-A2+ AML cell line U937 (U937-A2), which is known to express CG. Mice were injected with U937-A2 (0.5 x 106) cells and on the following day were treated with either CG1-CTL (0.25 x 106), negative control HIV-CTL expanded from the same donor or were left untreated. Mice treated with CG1-CTL demonstrated a significantly greater reduction in U937-A2 in the bone marrow (BM) (8% residual AML; P 〈 0.05; Figure 1A) after 2 weeks compared with mice treated with HIV-CTL (27% residual AML) or that were left untreated (34% residual AML). A similar reduction in AML was seen in the spleens of mice treated with CG1-CTL. Since normal myeloid cells also express CG, to demonstrate the specificity of CG1-CTL for AML, we engrafted NSGS mice with human HLA-A2+ cord blood (CB). After confirming engraftment by detecting human (h) CD45+/HLA-A2+/mouse (m)CD45- cells in peripheral blood, we treated mice with CG1-CTL or HIV-CTL (0.25 x 106). A similar percentage of hCD45+/HLA-A2+/mCD45- cells was detected in the BM of CB-engrafted mice in both CG1-CTL (24% CB) and HIV-CTL (27% CB) after 6 weeks, suggesting the safety of targeting CG1. After demonstrating the efficacy of targeting CG1 in vivo, we then studied CG1 presentation and CG expression in AML patients. To determine CG1 presentation, primary AML blasts, U937-A2 and HLA-A2-trasnduced HL60 (HL60-A2) cell lines were lysed then immunoprecipitated with anti-HLA-A2. Peptides were eluted using standard acid elution methodology and then analyzed by mass spectrometry. CG1 peptide was detected in 6 of 9 primary AML samples and in HL60-A2 and U937-A2 cells. Reverse phase protein analysis (RPPA) was used to study CG protein expression in a large cohort of AML patients and normal donors (ND) to include the following: 511 newly diagnosed patients, 21 ND peripheral blood leukocyte controls, 21 ND CD34+ cell controls and 10 ND BM controls. CG expression in the 511 AML patients samples demonstrated a Gaussian distribution and was higher than CG expression by ND CD34+ cells in 230 samples, equal to normal CD34+ cells in 234 samples and less than normal CD34+ cells in 47 samples. As expected, G-CSF primed ND CD34+ cells had high expression of CG. There were no significant differences in CG expression within the different AML FAB subtypes, although M4 eos and M5b subtypes showed the lowest expression levels. In multivariate Cox model analysis, higher CG expression significantly predicted shorter overall survival (OS) in all patients (P=0.04) (Figure 1B). The association of CG with shorter OS was most significant in patients who had intermediate cytogenetics and FLT3 mutation (P=0.02). Moreover, CG levels were determined in paired samples taken at diagnosis and relapse, and the levels were compared using paired t test and Pearson product-moment correlation analysis. Among 47 cases with paired diagnosis and relapse samples, there was significantly higher CG protein expression in relapse samples (P=0.0001). In summary, we found CG to be an important antigen in AML. CG is associated with worse outcomes in AML patients, however in vivo mouse data show that targeting CG eliminates AML while sparing normal hematopoiesis. Together these data indicate that CG has a promising role as a novel immunotherapeutic target in AML. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2014
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2986-2986
    Kurzfassung: Abstract 2986 Background: Immunotherapy targeting aberrantly expressed leukemia associated antigens (LAA) has shown promising results in the management of myeloid leukemia. However, because of the heterogeneity and clonal evolution that is a feature of myeloid leukemia, targeting single peptide epitopes has had limited success, highlighting the need for novel antigen discovery. PR1 is a well-characterized LAA, is derived from the azurophil granule proteases neutrophil elastase (NE) and proteinase-3 (P3), and was effectively targeted in myeloid leukemia. We have previously shown that NE and P3 are aberrantly localized outside azurophil granules in myeloid leukemia and that their aberrant expression facilitates PR1 antigen presentation. Similar to NE and P3, cathepsin G (CG) is a serine protease located within azurophil granules in neutrophils. Because azurophil granules are disrupted in myeloid leukemia and since CG was shown to be immunogenic through its association with autoimmune disease, we hypothesized that CG may be aberrantly expressed in myeloid leukemia, thereby facilitating its presentation on HLA class I molecules and rendering it a novel target for myeloid leukemia immunotherapy. Methods: SYFPEITHI and IEDB binding algorithms were used to identify CG peptides with highest binding affinities. Flow cytometry based binding assays were performed using the T2 cell line followed by HLA-A0201 staining to confirm peptide binding and determine affinities of CG-derived peptides for HLA-A0201. Peptide/HLA-A0201 complexes were released from HLA-A0201 transfected U937 cells and purified using immunoaffinity chromatography with the anti-HLA-A0201 antibody BB7.2. The peptides were dissociated from HLA-A0201 by weak acid elution and analyzed using reverse-phase HPLC-tandem mass spectrometry. Tandem mass spectra were analyzed using the Mascot sequencing algorithm. Western blots (WB) and immunoflouresence confocal microscopy were used to determine CG expression by primary leukemia. Peptide-pulsed T2 cells were used to expand CG1- and PR1-cytotoxic T lymphocytes (CTLs) for use in calcein AM cytotoxicity assays. Peptide-pulsed T2 cells and primary leukemia from patients were used as target cells. Tetramer staining and cytokine flow cytometry (CFC) assays were used to show frequency and function of CG1-CTLs, respectively. CG1- and PP65- pulsed T2 cells were used as stimulators in CFC assays. Patient samples were collected after informed consent. Results: Five CG derived nonameric peptides were identified using SYFPEITHI and IEDB binding algorithms. CG1 peptide (FLLPTGAEA) was identified as having the highest binding affinity to HLA-A0201 in comparison with other CG-derived peptides and with PR1 peptide (CG1 IC50=1 uM vs. PR1=9 uM). The dissociation t1/2 for CG1 and PR1 peptides was 〉 8 hours. CG1 peptide was eluted from the surface of U937-A2 cell line, and using reverse-phase HPLC-tandem mass spectrometry, we confirmed CG1 antigen presentation by leukemia. WB analysis for CG showed high CG expression in 9 of 11 AML patient samples and along with immunofluorescent cell imaging, showed localization of CG outside granules in compartments that could render CG susceptible to proteasomal degradation and antigen presentation. Killing of primary AML blasts by CG1-CTL was shown in 6 of 8 AML patient samples and directly correlated with CG expression and HLA-A0201 status. Blocking HLA-A0201 with BB7.2 antibody abrogated CG1-CTL mediated cytotoxicity, indicating HLA-A0201 dependent killing by CG1-CTLs. CG1-specific CTLs were detected in AML patient samples at frequencies ranging between 0.6% to 1.4% of total CD8+ cells. Furthermore, following stem cell transplant (SCT), 4–10 fold higher frequencies of functional CG1-CTLs were detected in patient samples when compared with pre-SCT samples from the same patient, as measured by IFN-gamma and/or TNF-alpha CFC. Conclusion: We demonstrate that CG is a novel immunotherapeutic target in myeloid leukemia. We show that CG is a LAA that is aberrantly expressed in leukemia, is presented on HLA-A0201 molecule and can be effectively targeted by CG1-CTLs. Since immunity to CG has been previously reported in autoimmune diseases, our findings show that the immunogenic potential of CG can be redirected therapeutically to target leukemia. Cumulatively, our data suggest that further development of CG-targeting therapies in myeloid leukemia is warranted. Disclosures: No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2011
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 4973-4973
    Kurzfassung: Abstract 4973 PR1 (VLQELNVTV) is a human leukocyte antigen (HLA)-A2 restricted nonameric peptide that has been effectively targeted in myeloid leukemia using PR1-peptide vaccine and PR1-specific cytotoxic T cells (CTL). A phase I/II clinical trial has been initiated with PR1 peptide vaccine, which demonstrated complete remission and immunologic responses in patients with acute (AML) and chronic (CML) myeloid leukemia, as well as myelodysplastic syndrome. We recently developed an anti-PR1/HLA-A2 antibody (8F4) and have demonstrated its activity against myeloid leukemia blasts in vitro and in a human leukemia xenograft mouse model. PR1 is derived from the serine proteases proteinase-3 (P3) and neutrophil elastase (NE), which are normally found within neutrophil azurophil granules and are released into the inflammatory milieu. We have shown that through cross-presentation, a process whereby exogenous antigen is taken up, processed and expressed on cell surface in association with human leukocyte antigen class I molecules, PR1 can be expressed on the surface of cells that do not endogenously express NE and P3. Specifically, we showed that P3 and NE are taken up and cross-presented by antigen presenting cells (APC), including B cells, as well as breast cancer and melanoma cells. Furthermore, cross-presentation by these non-myeloid cells led to their killing by PR1-CTL and 8F4 antibody. Since multiple myeloma is derived from B cells, which are deficient in NE and P3 but have intrinsic APC functions including cross-presentation, we hypothesized that P3 and/or NE may be taken up and cross-presented by multiple myeloma cells, hence rendering them targets for PR1-CTL and 8F4 antibody. Using RT-PCR and western blotting, we first show that NE and P3 are absent in myeloma cell lines including ARK, ARP-1, OMP-2, LP-1, U-266, IM9, and RPMI 8226. Intracellular staining for P3 and NE after culturing myeloma cell lines with P3 (10 ug/mL) or NE (10 ug/mL) confirmed uptake of P3 and NE by multiple myeloma cell lines; uptake is time dependent with variable plateauing between the cell lines over a 30-hour time course. Using 8F4, the novel PR1-HLA-A2 monoclonal antibody, we demonstrate that P3 and NE are cross-presented by the HLA-A2 positive U-266 myeloma cell line. Peak P3 and NE cross-presentation occurs at 24 hours and 6 hours, respectively. Next, we studied whether PR1 cross-presentation causes cells to be susceptible to PR1-specific killing by PR1-CTL and 8F4 monoclonal antibody. We show that cross-presentation increases susceptibility of U-266 myeloma cell line to killing by PR1-CTL. Specifically, following incubation with P3 (10 ug/mL) and NE (10 ug/mL), we show 30% and 25% killing, respectively, of U-266 cells by PR1-CTL versus unpulsed U-266 cells (10% killing). Furthermore, we show dose-dependent killing of the U-266 myeloma cell line by 8F4 antibody in a complement dependent cytotoxicity assay, with maximum 30% and 20% killing of U-266 cells after incubation with P3 (10 ug/mL) or NE (10 ug/mL), respectively, versus no killing in the unpulsed group. Finally, using PR1/HLA-A2 dextramer staining, we show PR1-CTL in peripheral blood from patients with multiple myeloma following stem cell transplantation (Median, 0. 06%; range, 0. 03%-0. 1%). Our data therefore show that if the uptake of P3 or NE, present in the inflammatory milieu of multiple myeloma bone marrow and extramedullary tumors, leads to PR1 cross-presentation, then PR1-based immunotherapy may be useful to treat patients with multiple myeloma. These results also support a new paradigm linking inflammation and innate immunity to adaptive immune responses to cancer. Disclosures: No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2012
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2133-2133
    Kurzfassung: PR1 is an HLA-A2-retricted, nonameric peptide that is derived from the azurophil granule proteases neutrophil elastase (NE) and proteinase 3 (P3). PR1 has been targeted successfully in acute (AML) and chronic (CML) myeloid leukemia using anti-PR1/HLA-A2 antibody (8F4), PR1-peptide vaccine and PR1-specific cytotoxic T lymphocytes (PR1-CTL). We have previously reported that NE and P3 are cross-presented by normal B cells and dendritic cells (DC), leading to PR1 expression by HLA-A2. Since multiple myeloma (MM) is a B cell malignancy, we investigated whether MM cells can cross-present PR1 as a possible target for immunotherapy. To study whether PR1 is presented by MM cells, patient bone marrow (BM) was stained with 8F4 antibody and then imaged using confocal microscopy. PR1/HLA-A2 was detected on the surface of CD138+ MM cells in BM samples from 3 of 6 HLA-A2+ MM patients (Fig. 1). We then investigated whether cellular immunity to PR1 is detected in peripheral blood (PB) from patients with MM. PB samples from MM patients who had undergone allogeneic (allo) (n=9) and autologous (auto) (n=2) stem cell transplantation (SCT) were stained with PR1/HLA-A2 dextramer in addition to standard lineage markers. PR1-specific CD8+CTL were detected in PB of 10 of 11 patients (range=0.02%-2.9%). Because P3 and NE expression is limited to myeloid cells, we sought to determine the mechanism of PR1 presentation in MM. We performed RT-PCR and western blotting on seven MM cell lines, including U266, ARK, ARP-1, OPM-2, LP-1, IM-9 and RPMI 8226. Neither NE nor P3 were detected in the MM cell lines studied at either the mRNA or proteins levels. We then investigated whether MM cells took up NE and P3. We cultured MM cells for 30 hours with 10 ug/mL of soluble NE and P3 or irradiated HLA-A2 negative PMN, the latter as a source for cell-associated NE and P3. Cells were then stained intracellularly for NE and P3 at different time points. Flow cytometry analysis showed that all the cell lines analyzed took up NE and P3. Uptake was seen as early as 1 hour after co-culture with soluble NE and P3 and was higher for soluble P3. Additionally, more uptake was seen in the cells that were co-cultured with irradiated PMN in comparison with soluble NE and P3. We then investigated whether PR1 expression in MM was through NE and P3 cross-presentation. We focused our studies on the HLA-A2+ U266 MM cell line. U266 cells were co-cultured with soluble NE, P3 or irradiated PMN, as described in the previous section, and then surface stained with 8F4. We detected PR1/HLA-A2 on the surface of U266 cells by flow cytometry as early as 6 and 24 hours after co-culture with soluble NE and P3, respectively. Cross-presentation was also seen in the cells that were co-cultured with irradiated PMN to a similar extent in comparison with the cells that were cultured with soluble NE and P3, however, cross-presentation occurred at an earlier time point (1 hour) in the cells that were cultured with PMNs. Cross-presentation was abrogated by lactacystin, a proteasome inhibitor, and brefeldin A, an ER/Golgi transport inhibitor, indicating that NE and P3 cross-presentation occurs through conventional cross-presentation mechanisms. Furthermore, because immune modulatory drugs and proteasome inhibitors are part of the standard of care therapy for MM, and since both have been shown to affect cross-presentation by DC, we tested whether lenalidomide and bortezomib altered PR1 cross-presentation by U266 and normal DC. U266 and DC were cultured with irradiated PMN in the presence of increasing doses of bortezomib or lenalidomide and then stained with PR1/HLA-A2. Flow-cytometry analysis showed a significant inhibition of PR1-cross-presentation by U266 after addition of bortezomib, but not lenalidomide. PR1 cross-presentation by DC was not affected by either drug. Finally, we tested whether PR1 cross-presentation caused MM cells to become susceptible to PR1-targeting immunotherapies. U266 cells were cultured with NE or P3 for 24 hours, the time point that showed the maximal cross-presentation, followed by addition of 8F4 antibody or co-culture with PR1-CTL. Using calcein AM cytotoxicity assays, we showed dose dependent killing of U266 by 8F4 and PR1-CTL following PR1 cross-presentation (Fig. 2). Together our data show that PR1 is cross-presented by primary MM cells and cell lines. These findings lay the foundation for the future applications of PR1-targeting immunotherapies in MM. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2014
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Society of Hematology ; 2022
    In:  Blood Vol. 140, No. Supplement 1 ( 2022-11-15), p. 10269-10270
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 10269-10270
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2022
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 9061-9062
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2022
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    In: Blood, American Society of Hematology, Vol. 117, No. 16 ( 2011-04-21), p. 4262-4272
    Kurzfassung: PR1 (VLQELNVTV) is a human leukocyte antigen-A2 (HLA-A2)–restricted leukemia-associated peptide from proteinase 3 (P3) and neutrophil elastase (NE) that is recognized by PR1-specific cytotoxic T lymphocytes that contribute to cytogenetic remission of acute myeloid leukemia (AML). We report a novel T-cell receptor (TCR)–like immunoglobulin G2a (IgG2a) antibody (8F4) with high specific binding affinity (dissociation constant [KD] = 9.9nM) for a combined epitope of the PR1/HLA-A2 complex. Flow cytometry and confocal microscopy of 8F4-labeled cells showed significantly higher PR1/HLA-A2 expression on AML blasts compared with normal leukocytes (P = .046). 8F4 mediated complement-dependent cytolysis of AML blasts and Lin−CD34+CD38− leukemia stem cells (LSCs) but not normal leukocytes (P 〈 .005). Although PR1 expression was similar on LSCs and hematopoietic stem cells, 8F4 inhibited AML progenitor cell growth, but not normal colony-forming units from healthy donors (P 〈 .05). This study shows that 8F4, a novel TCR-like antibody, binds to a conformational epitope of the PR1/HLA-A2 complex on the cell surface and mediates specific lysis of AML, including LSCs. Therefore, this antibody warrants further study as a novel approach to targeting leukemia-initiating cells in patients with AML.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2011
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 2775-2775
    Kurzfassung: Background Pembrolizumab, an antibody that blocks programmed cell death protein 1 (PD-1), has been FDA approved for several solid tumors and hematologic malignancies. Currently, pembrolizumab is under investigation for acute myeloid leukemia (AML) in combination with hypomethylating agents. It is established that AML is highly responsive to immunotherapy, as seen with the anti-leukemic effect of allogeneic hematopoietic stem cell transplantation (alloHSCT). However, response to cytotoxic T lymphocytes (CTLs) that target leukemia-associated antigens (LAAs) has been less reliable in eradicating disease. This insufficient response to LAA-specific CTLs is likely partially accounted for by the immune dysregulation seen in AML. Because of promising murine data that blockade of the PD1/PD-L1 pathway enhances the graft versus leukemia effect of alloHSCT, we investigated if adding pembrolizumab to CTLs that target the two LAAs CG1 and PR1 will enhance CTL antileukemia activities. CG1 and PR1are two HLA-A2 restricted nonameric peptides that we validated as promising AML targets derived from cathepsin G (i.e. CG1), and proteinase 3 (P3) and neutrophil elastase (NE) (i.e. PR1). We hypothesized that pembrolizumab added to CG1-CTLs and PR1-CTLs, will enhance their anti-leukemic effects with minimal off target toxicities. Methods Using a standard calcein AM in vitro cytotoxicity assay, we co-cultured AML targets, including U937 HLA-A2 + (U937-A2) AML cell line and primary patient HLA-A2 + AML samples, with CG1-CTL and PR1-CTL. AML cells were loaded with calcein AM and then incubated with CG1- and PR1-CTLs at increasing effector to target ratios. Pembrolizumab or isotype antibody were added to the cultures. After 4 hours, calcein AM was measured to determine cell viability. T2 cells pulsed with PR1 or CG1, were used as a positive control and non-pulsed T2 cells were used as negative control cells. For in vivo experiments, we used a human established AML xenograft mouse treatment model to determine the effect of pembrolizumab when added to CG1- or PR1- CTL in vivo. CG1-CTL and PR1-CTLs were expanded from the same donor and used as the effector cells. NOD/SCID gamma (NSG) mice (4-6-week-old females) were engrafted with AML samples by tail vein injection. After confirming engraftment, CG1- and/or PR1-CTL were administered to mice and pembrolizumab or isotype antibody [100ug/mouse] were given three times over two weeks. Mice were monitored for clinical GVHD and AML three times/week. Mice were sacrificed at approximately two weeks following treatment. To assess for GVHD, mouse tissues including spleen, liver, kidney, BM, intestine, brain, heart, and lung were harvested and fixed in 10% formalin. The fixed tissue samples were embedded in paraffin, sectioned, and stained with hematoxylin and eosin prior to histologic examination. Bone marrow (BM) was processed using standard methodology and analyzed for residual AML by flow cytometry. Results: In vitro our data demonstrate that U937-A2 and two primary patient HLA-A2 AML samples, have enhanced cell lysis when treated with pembrolizumab (CTL+ pembrolizumab) in comparison with isotype (Iso) + CTL, pembrolizumab only, iso only or CTL only groups (Figure 1). In vivo data show a decrease in U937-A2 disease burden and primary patient AML after treatment with CG1and/or PR1-CTL (CTL-treated) . This decrease was enhanced when pembrolizumab was added to the CTL-treated mice (CTL+ pembrolizumab) in comparison with mice treated with pembrolizumab only (pembrolizumab-treated), or CTL only (CTL-treated) (Figure 2). Pathology data to assess toxicity in mice treated with CTL +/- pembrolizumab showed an enhanced pulmonary (perivascular and parabronchial) lymphocytic infiltration in mice treated with the combination of CTL and pembrolizumab. The other organs investigated, showed no change when combination therapy was used. Conclusion: We have validated in vitro and in vivo the enhanced killing of AML (cell lines and primary patient samples) by CG1-CTL and/or PR1-CTL after addition of pembrolizumab. Toxicity data show mild enhancement of lymphocytic infiltrate in the lungs after addition of pembrolizumab to CTL. Our data suggest that the strategy of combining LAA-specific CTL with immune checkpoint blockade could prove beneficial in the setting of adoptive T cell therapy and allogeneic stem cell transplantation for AML. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2021
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...