ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Collection
  • 1
    Publication Date: 2019-02-28
    Description: Cold agglutinin disease is a difficult-to-treat autoimmune hemolytic anemia in which immunoglobulin M antibodies bind to erythrocytes and fix complement, resulting in predominantly extravascular hemolysis. This trial tested the hypothesis that the anti-C1s antibody sutimlimab would ameliorate hemolytic anemia. Ten patients with cold agglutinin disease participated in the phase 1b component of a first-in-human trial. Patients received a test dose of 10-mg/kg sutimlimab followed by a full dose of 60 mg/kg 1 to 4 days later and 3 additional weekly doses of 60 mg/kg. All infusions were well tolerated without premedication. No drug-related serious adverse events were observed. Seven of 10 patients with cold agglutinin disease responded with a hemoglobin increase 〉2 g/dL. Sutimlimab rapidly increased hemoglobin levels by a median of 1.6 g/dL within the first week, and by a median of 3.9 g/dL (interquartile range, 1.3-4.5 g/dL; 95% confidence interval, 2.1-4.5) within 6 weeks (P = .005). Sutimlimab rapidly abrogated extravascular hemolysis, normalizing bilirubin levels within 24 hours in most patients and normalizing haptoglobin levels in 4 patients within 1 week. Hemolytic anemia recurred when drug levels were cleared from the circulation 3 to 4 weeks after the last dose of sutimlimab. Reexposure to sutimlimab in a named patient program recapitulated the control of hemolytic anemia. All 6 previously transfused patients became transfusion-free during treatment. Sutimlimab was safe, well tolerated, and rapidly stopped C1s complement–mediated hemolysis in patients with cold agglutinin disease, significantly increasing hemoglobin levels and precluding the need for transfusions. This trial was registered at www.clinicaltrials.gov as #NCT02502903.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 2
    Publication Date: 2016-12-02
    Description: Background: We recently presented that the humanized anti-complement C1s monoclonal antibody, TNT009, rapidly stoppedhemolysisand correctedanemiain patients with severe CAD in an ongoing Phase 1b clinical trial. After completion of washout from protocol treatment, these patients suffered relapse of CAD with a drop inhemoglobinto pre-dose levels. We now report on the clinical experience of their extended re-treatment under the Named Patient provision of the Austrian Drug Act, which provided the occasion: 1) to explore lower dose regimens and alternative dosing schedules; 2) to try to recapitulate the initial success of complete remission under protocol treatment; 3) to test the durability of response with a longer term maintenance treatment with TNT009. Methods: In order to test a lower dose regimen of TNT009, the Phase 1b trial dosing of 60 mg/kg IV once weekly for both loading and maintenance was reduced to 45 mg/kg IV once weekly (4x) followed by maintenance with 45 mg/kg every other week. A simplified dosing regimen was then tried, using a single priming dose of 60 mg/kg on Day 0 followed by 60 mg/kg every other week (biweekly) thereafter beginning on Day 7. This TNT009 biweekly maintenance with 60 mg/kg was continued indefinitely to test the durability of the effect, to monitor for safety, and to serially assess their hematologic response to TNT009. Results:All infusions were well tolerated without premedication and without relevant adverse effects. TNT009 re-exposure immediately decreased CH50 activity and increased complement C4 levels, confirming the rapid onset ofpharmacodynamiceffect. The concordant and sustained effect of TNT009 on hematologic parameters related tohemolysisandanemiasis illustrated in the composite Figure 1 for the first patient treated, C1001. This patient has undergone three treatment episodes with TNT009: weekly dosing at 60 mg/kg under the Phase 1b protocol (left); weekly → biweekly 45 mg/kg (center); 60 mg/kg on Day 0 and biweekly thereafter starting on Day 7 (right). Note that the CH50 serves as apharmacodynamicmarker for the effective inhibition of the classical complement pathway by TNT009. Each new episode of treatment with TNT009 induced remission ofhemolyticanemia, and relapse occurred following washout (60 mg/kg weekly x 4) orpharmacodynamicbreakthrough (45 mg/kg maintenance regimen). Although it appears that the reduced dose maintenance regimen of 45 mg/kg biweekly does not suffice to maintain full C1s inhibition, there is no evidence oftachyphylaxisas restoration of full dose treatment at 60 mg/kg biweekly was able to restore remission again. A similar pattern of results was observed in the other patients offered Named Patient treatment with TNT009, but with two instructive exceptions. Patient C1004 had unsuspected erythropoietin deficiency and did not achieve complete normalization ofhemoglobinuntil concomitant treatment with erythropoietin was begun. Patient C1003 did not have CAD but Cold Agglutinin Syndrome (CAS) secondary to an extensivelymphoplasmacyticlymphoma (LPL). Initially she was treated with TNT009 monotherapy and did not respond. TNT009 was then stopped and her LPL was treated withibrutinib, resulting in a gradual amelioration of heranemiauntil anintercurrentinfection triggered relapse of herhemolyticanemia. At that time re-treatment with TNT009 (whileibrutinibwas continued) quickly normalized her hematologic parameters. Eventually TNT009 was able to increasehemoglobinlevels to 〉12 g/dLin all 5 of 5 treated patients. Conclusion:TNT009 was previously shown to induce rapid correction ofhemolyticanemiain severe CAD patients in a Phase 1b trial. It has now been shown that subsequent washout of TNT009 leads to relapse in all patients, and that re-induction of remission by TNT009 is possible in all. Long-term maintenance of remission can be achieved by dosing TNT009 at 60 mg/kg biweekly, the dose regimen to be tested in larger-scale clinical trials of CAD. Figure Figure. Disclosures Gilbert: Truenorth Therapeutics, Inc.: Employment, Equity Ownership. Panicker:Truenorth Therapeutics, Inc.: Employment. Parry:Truenorth Therapeutics, Inc.: Employment, Equity Ownership. Jaeger:Janssen: Consultancy, Honoraria, Other: Travel, Accommodations, Expenses.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 3
    Publication Date: 2005-11-16
    Description: Introduction: After intensive treatment regimens have been established, the survival rate for patients with advanced Hodgkin’s disease is approximately 91 % after five years and 13 % of the patients have a relapse or have primary progressive disease (2 %) within the first five years. For patients with relapse after conventional chemotherapy +/− radiotherapy, however, there is a real chance of achieving remission again. Since it is often difficult to harvest autologous stem cells following an intensive pretreatment, our center embarks on the strategy to harvest autologous blood stem cells in high-risk patients, defined according to the risk stratification of the German Hodgkin Study Group, already as part of the initial polychemotherapy. Results: Between 9/2003 and 5/2005, we analyzed the results of the stem cell harvest of 12 consecutive patients with Hodgkin’s disease who were mobilized with the escalated BEACOPP regimen. There were 7 female and 5 male patients. Escalated BEACOPP was the primary therapy in ten patients and a relapse was treated in two patients; the previous treatment was 4 or 6 cycles of the ABVD regime + involved field radiation. The ten patients who did not receive previous treatment were classified as having Ann Arbor stage IIA/2 IIB/5, IIIB/2 and IVB/1 and all of them had a large mediastinal bulk as an additional risk factor. The two patients who did receive a previous treatment were classified as having an initial Ann Arbor stage IIA or IIB, without an additional risk factor. The stem cells were collected in 1 patient from cycle 2, in 8 patients from cycle 3 and in 3 patients from cycle 4 of the escalated BEACOPP regimen. A total of 11 patients received a standard dose of filgrastim, 5μg/kg body weight s.c., from day 8 up to the last apheresis and 1 patient received pegfilgrastim 6mg s.c. All aphereses were performed using an Amicus cell separator® (Baxter, MNC set, closed two-arm). 7 patients required only 1 apheresis and the remaining 5 patients required 2 aphereses. An apheresis result sufficient for a possible reinfusion could be achieved in all patients (4.26 – 14.4 x10e6 CD34 pos. cells/kg/body weight, mean: 7.7). Summary: According to our experience, escalated BEACOPP regimen is very suitable for the harvesting of stem cells in high-risk patients with Hodgkin’s disease even though they are receiving procarbazine. A sufficient quantity of stem cells can also be collected from pretreated patients. The stem cell mobilization can be integrated into the escalated BEACOPP regimen safely and without a delay in treatment and thus creates, already at an early stage, the pre-condition for a high-dose therapy, which might be required in high-risk patients.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 4
    Publication Date: 2004-11-16
    Description: Background: In contrast to other human endogenous retrovirus families which are highly defective, HERV-K proviruses possess all viral gene types of exogenous retroviruses. In malignancy (tumor cell lines, breast cancer, melanoma as well as leukemias and lymphomas) but also in many normal tissues, including placenta HERV-K mRNA expression has been detected. In hematopoietic stem cells, an interaction of HERV-K with elements from vectors which are used for gene therapy may lead to unforeseen consequences. The purpose of this study was to localize HERV-K mRNA expression in hematopoietic stem cells of different sources and to evaluate a potential association of HERV-expression with hematologic diseases and to define a possible risk of HERV-expression for transplantation. Methods: Quantitative real time PCR (RTQPCR) was used for detecting HERV-K expression in MNC from blood and/or bone marrow samples of patients with leukemias ( 10 MDS, 16 MPD, 14 AML and 1 CML blast crisis) and MNC from blood (n=9) and lymph nodes (n=3) from lymphoma patients. In addition, 8 blood samples and 2 bone marrow samples from healthy donors as well as 9 cord blood samples and 20 apheresates from lymphoma patients in remission containing PBSC were analyzed. Analysis of the same HERV-K gene (pol) in placenta and maternal blood as well as in genomic DNA from all above mentioned sources served as control. In HERV-K overexpressing samples expression of HERV in specific cells was tested by RNA in situ hybridisation (RISH), and combined staining with a CD34 specific monoclonal antibody. Results: Relative mRNA levels of HERV-K in apheresates as well as in blood and bone marrow samples of patients with MDS, MPD and AML from subtypes with less than 1% CD34 positive cells were in the range of less than 2% in relation to housekeeping genes. However, in bone marrow samples of two AML and a CML blast crisis (all of them with more than 50% CD34+ cells) as well as in CD34+ cells from cord blood (but not from apheresates or normal bone marrow) as well as in three lymph nodes of patients with B-cell lymphomas, an overexpression of HERV-K was detected and confirmed by RISH. Enhanced expression of HERV-K was detected in cord blood derived apoptotic CD34+ cells, whereas the relative amount of HERV in genomic DNA did not change in samples from different sources. Conclusions. Our data indicate that HERV-K may be activated in lymph nodes of lymphomas and in CD34+ cells from bone marrow of leukemia patients and from cord blood but not in stem cell apheresates from peripheral blood. In cord blood and malignancy expression appears to be associated with apoptosis. Similar amounts of HERV-K in different samples of genomic DNA of the same cells and the absence of HERV-K activation in normal bone marrow and PBSC indicate that activation of HERV-K is the result of regulation of gene expression rather than amplification. A possible specific role of HERV in the pathogenesis of hematologic diseases and in hematopoiesis (cord blood) remains to be established. Differences between peripheral blood stem cells and cord blood may support a critical stance regarding the use of cord blood derived stem cells for transplantation and gene therapy.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 5
    Publication Date: 2020-11-05
    Description: Myeloproliferative neoplasms (MPN) are characterized by clonal hematopoiesis, hyperproliferation of myeloid cells, hyperinflammation and immune deregulation. The three classical BCR-ABL1-negative MPN are essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF). The disease is driven by JAK2, CALR or MPL somatic mutations in most patients. Drug resistance is a major problem in MPN. Recent data suggest that MPN cells display certain immune checkpoint molecules that may contribute to resistance, including PD-L1. Antibodies targeting the PD1/PD-L1 axis are highly promising anti-cancer drugs. Their potential use in MPN is being explored but it is unclear which MPN subtypes are most suitable for testing in clinical trials. The aim of our project was to assess PD-L1 expression in disease-initiating neoplastic stem cells (SC) and differentiated cells of MPN patients and to develop therapeutic approaches capable of blocking PD-L1 expression in MPN SC. In a first step, PD-L1 expression was assessed by RNA-sequencing of granulocytes of 106 MPN patients and 15 healthy donors (HD). The cohort included 56 PMF, 33 ET and 17 PV patients. For 102 patients data from Human Genome-wide Affymetrix 6.0 SNP arrays were available. We observed a ~5-fold higher expression of PD-L1 mRNA in patients with PV compared to other MPN (P
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 6
    Publication Date: 2017-12-07
    Description: Background: Cold agglutinin disease is an autoimmune hemolytic anemia with limited treatment options and no established standard of care. The pathophysiology is driven by the classical complement pathway in which IgM auto-antibodies bind erythrocytes and fix complement via initial binding and activation of the C1 complex generating active C1s protease. Anemia results from extravascular hemolysis of complement opsonized erythrocytes, primarily in the liver. The anti-C1s antibody BIVV009 inhibits C1s activity, and specifically blocks the classical complement pathway, leaving the alternate and lectin complement pathways intact. We hypothesized that classical complement pathway blockade using BIVV009 would prevent hemolysis, correct anemia, and obviate the need for transfusions in patients with primary cold agglutinin disease. Methods: Six patients primary cold agglutinin disease patients were enrolled in an open label Ph1/1b trial. The study was conducted in three parts: Part A, single ascending doses in healthy volunteers (HV); Part B, multiple ascending doses in HV; and Part C, multiple doses in patients with four classical complement mediated diseases including cold agglutinin disease. Patients in Part C received a test dose of 10 mg/kg BIVV009, followed by a full dose of 60 mg/kg 1-4 days later, and three additional weekly doses of 60 mg/kg. Biweekly fixed doses of 5.5g were used for maintenance therapy in a subsequent Named Patient Program. Results: All infusions were well tolerated without need for pre-medication, and pharmacokinetic data demonstrated that BIVV009 infusions supported biweekly treatment. BIVV009 concentrations 〉18µg/mL inhibited the classical pathway of complement activation (as assessed by the Wieslab-CP assay). BIVV009 infusion subsequently raised endogenous C4 levels 3.2-fold (95%CI: 2.4-4.0 fold; p3.5 g/dL (mean 4.3g/dl; 95%CI: 3.8-4.9 g/dL; individual best response; p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 7
    Publication Date: 2020-11-05
    Description: Introduction Carfilzomib-based induction therapy is highly effective in newly diagnosed pts with multiple myeloma (NDMM), but information on quality of life (QoL) during first line and maintenance therapy is not available. Here, we analyze patient-reported QoL in transplant-non eligible (TNE) NDMM pts randomized to either KTd or KRd induction therapy followed by second randomization to carfilzomib maintenance, or control. Patients and Methods At time of analysis, 101 TNE pts with NDMM had been enrolled, but QoL data documented for ≥ 2 cycles were available in 78 pts so far (median age: 75 years, ISS stage I: 26.6%, II: 35.4%, III: 38.0%, ECOG status 0: 51.9%, 1: 48.1%). Patients were randomized to either 9 cycles KTd or KRd, and subsequently (46 pts with ≥PR) to either carfilzomib maintenance therapy (d1 and 15) or to control for 12 months. Carfilzomib was administered twice weekly (27mg/m2) during cycles 1 and 2, and thereafter weekly at a dose of 56mg/m2. Thalidomide was given daily at 100mg (50mg in pts ≥75 years), lenalidomide at 25mg, d1-21 per 4 week cycle, and dexamethasone at 40 mg (20mg in pts ³75 years) once weekly. QoL was assessed by the QoL questionnaire EORTC QLQ-C30. Assessments were done at baseline, and monthly thereafter for 21 months. A two-sided t-test was used for comparison with the general population. Wilcoxon signed-rank tests were applied to evaluate differences in QoL dimensions within the treatment groups. A clinically meaningful improvement has been defined as a change of ≥10 score points. Results Comparison with the general population : Mean scores for health-related global QoL (54.1±22.6) and physical functioning (61.2±25.0) were significantly lower in pts compared to those reported for the general population of similar age (67.2±20.6, p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 8
    Publication Date: 2020-11-05
    Description: The classical BCR-ABL1-negative myeloproliferative neoplasms (MPN) are characterized by over-production of myeloid cells, disease-related mutations in certain driver-genes (JAK2, CALR, MPL) and an increased risk to transform to secondary acute myeloid leukemia (sAML). Although considered stem cell-derived neoplasms, little is known about the phenotype and functional properties of disease-initiating neoplastic stem cells (NSC) in MPN and sAML. Recent data suggest that MPN NSC reside in a CD34+ fraction of the malignant clone. Therefore, these cells are considered most critical target populations to be examined for expression of molecular and immunological targets with the aim to develop improved or even curative NSC-eliminating therapies, such as antibody-based or CAR-T cell approaches. Using a panel of monoclonal antibodies (n=40) and multicolor flow cytometry, we established the immunological phenotype and target expression profiles of putative CD34+/CD38─ NSC and CD34+/CD38+ progenitor cells in patients with polycythemia vera (PV, n=18), essential thrombocythemia (ET, n=29), primary myelofibrosis (PMF, n=38) and post-MPN sAML (n=11). In almost all patients, the putative MPN stem cells expressed the stem cell invasion receptors Hermes (CD44) and ADGRE5 (CD97), C1qR1 (CD93), the migration/adhesion receptor MIC2 (CD99), and the stem cell antigen AC133 (CD133). Contrasting normal stem cells, MPN NCS and sAML stem cells failed to express Thy-1 (CD90). Among the cytokine receptors tested, MPN NSC invariably displayed the TGFßR-related antigen endoglin (CD105), TPOR (CD110), SCFR KIT (CD117), IL-3RA (CD123), CXCR4 (CD184) and IGF-1R (CD221). NSC expressed particularly high levels of KIT and low levels of TPOR and IGF-1R. The IL-2RA (CD25) was identified on NSC in most patients with PMF and sAML, and in a few with ET, but not in patients with PV. Similarly, the GM-CSFR (CD116) was found to be expressed on NSC in most patients with PMF, a few with ET and no with PV. MPN NSC did not exhibit substantial amounts of M-CSFR (CD115), IL-3RB (CD131), FLT3 (CD135), NGFR (CD271) VEGFR-2 KDR (CD309), EPOR, MET or OSMRB. The CD34+/CD38+ MPN progenitor cells displayed a similar profile of cytokine receptors. In addition, MPN and sAML progenitor cells expressed IL-1RAP and CLL-1 in most donors examined. We next examined the expression of various immunological targets and resistance-mediating immune checkpoint antigens on NSC and MPN progenitor cells. In all MPN patients and all sAML patients tested, NSC were found to express substantial amounts of Siglec-3 (CD33) and low levels of Campath-1 (CD52) and MDR-1 (CD243). In addition, MPN NSC and sAML stem cells invariably displayed the "don't eat" me checkpoint IAP (CD47) and the classical checkpoint PD-L1 (CD274). Exposure to interferon-gamma (200 U/ml, 24 hours) resulted in an upregulation of PD-L1 on NSC. In a subset of patients, MPN NSC expressed low levels of HB15 (CD83). In contrast, MPN NSC and sAML stem cells failed to express B7-1 (CD80), B7-2 (CD86), PD-L2 (CD273) and PD1 (CD279). MPN progenitor cells and sAML progenitors expressed an identical profile of cell surface targets and checkpoint antigens. Finally, we confirmed the disease-initiating capacity of MPN stem- and progenitor cells (CD34+ cells) using primary PMF cells in xenotransplantation experiments employing NSGS mice expressing human interleukin-3 (IL-3), granulocyte/macrophage colony-stimulating factor (GM-CSF) and stem cell factor (SCF). After 28 weeks post injection, engraftment of human CD45+ cells in the bone marrow of NSGS mice was found in 15/15 mice injected with bulk mononuclear cells (MNC) containing CD34+ cells and in 0/15 NSGS mice injected with MNC depleted of CD34+ cells. Together, MPN NSC reside in a CD34+ fraction of the malignant clone and display a unique phenotype, including cytokine receptors, immune checkpoint molecules and other target antigens. The phenotypic characterization of neoplastic stem cells should facilitate their enrichment and the development of NSC-eradicating treatment concepts in MPN. Disclosures Valent: Allcyte GmbH: Research Funding; Pfizer: Honoraria; Cellgene: Honoraria, Research Funding.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 9
    Publication Date: 2020-11-05
    Description: Background The depth of response to myeloma therapy shows significant variation between individual patients. Responsiveness depends on the activity of the treatment regimen, the genetic aberrations of the predominant myeloma clone(s), and on the composition of the bone marrow environment, particularly of the immune compartment, in patients treated with drugs having broad immune effects. Here, we evaluated possible relationships between multiple immune cells and response to therapy in a series of newly diagnosed patients randomized to either carfilzomib-lenalidomide-dexamethasone or to carfilzomib-thalidomide-dexamethasone. Patients and Methods Forty-one patients with newly diagnosed multiple myeloma enrolled in a randomized phase II trial (AGMT_MM 02) with baseline bone marrow phenotyping were enrolled. Median age: 74 years (range: 58-84 years), R-ISS I: 23.8%, R-ISS II: 33.3%, and R-ISS III: 42.9%, IgG: 57.1%, IgA: 21.4%, light chain only: 19.0%, IgM: 2.4%, ECOG Status 0: 42.9%, 1: 57.1%. Treatment: Carfilzomib : Cycle 1 day 1+2: 20mg/m2, days 8+9 and 15+16: 27 mg/m2 ; Cycle 2 day 1,2,8,9,15 + 16: 27 mg/m2 Cycle 3-9: 56mg/m2 on days 1, 8 and 15. Lenalidomide: 25mg p.o. on days 1-21/cycle or thalidomide: 100 mg p.o. on days 1-28; in patients ≥75 years of age 50mg p.o. on days 1-28, dexamethasone: 40 mg p.o. on days 1, 8, 15, 22 (± 1 day), in patients ≥ 75 years of age 20 mg p.o. this treatment was administered for nine cycles. Thereafter, patients were randomized to carfilzomib maintenance at last tolerated dose on days 1+15 or to control for 12 cycles. BM samples were stained with 3 different antibody combinations for the analysis of T, B and NK cells (T cells: CD45RA, CD127, CD8, TCRgd, CD25, CD197, CD4, CD3, B/NK cells: CD57, CCR7, CD314, CD85j, CD62L, CD3, CD16, CD56, CD335, HLADR, CD337) using sample preparation protocols standardized by EuroFlow. We used a semi-automated pipeline to unveil full cellular diversity based on unbiased clustering. The median and range of each cellular subgroup was calculated in patients with CR and compared with results obtained in non-CR patients. Statistical significance of these comparisons was calculated using the Wilcoxon test. Results Median follow up is 15.9 months. Thirteen of the 41 patients achieved a CR (31.7%) and 9 (75%) of the 12 who had MRD testing were shown to be MRD negative at a sensitivity of 10-6. Nine (22.0%) of the 41 patients did already progress. High numbers of total B cells (p=0.011), B-cells expressing CD62L (p=0.027), CD4+ CD127+ central memory T cells (p=0.009), and CD337+ (p=0.012), or adaptive CD57lo (p=0.002) cytotoxic NK cells were significantly associated with achievement of CR. By contrast, lower numbers of T-NK cells (CD3+CD56+) (p=0.006), CD127- double-negative T cells (CD4- CD8-) (p=0.013), CD85j+ tissue resident immunoregulatory NK cells (p=0.002) and CD56+CD57+ (p=0.002) and CD56+ HLADR+ (p=0.003) T cells correlated significantly with achievement of CR. Significant correlations were also noted when NK cell subsets were analyzed for a possible association with progressive disease. Higher numbers of CD85j+ tissue resident immunoregulatory NK cells (p=0.001) were associated with progressive disease. In contrast, higher numbers of canonical cytotoxic NK cells (p=0.015) and CD314+ circulating immunoregulatory NK cells (p=0.033) correlated with sustained response. Conclusion Our data show significant correlations between the composition of bone marrow immune cells with achievement of CR and sustained response in newly diagnosed patients uniformly treated with carfilzomib-based therapy. High cytotoxic NK cell and B-cell, and low CD56+ T cell numbers among others were associated with deep response to carfilzomib-based therapy. Furthermore, higher numbers of specific NK cell subsets (CD85j+, CD57lo), correlated with disease progression while for other NK cell subtypes (canonical cytotoxic NK cells and CD314+ circulating immunoregulatory) a correlation with sustained response was noted. This study paves the way towards clinically relevant immune monitoring, whereby a holistic and unbiased assessment of the immune tumor microenvironment is connected with patients' treatment, depth of response and outcome. Disclosures Greil: Daiichi Sankyo, Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; MSD Merck: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Astra zeneca: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; F. Hoffmann-La Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; BMS/celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding. Podar:Amgen, BMS-Celgene, Janssen, Roche: Consultancy, Honoraria, Research Funding. Petzer:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Kite-Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Agis:Janssen: Honoraria, Research Funding; Amgen: Honoraria; BMS: Honoraria; Celgene: Honoraria; Takeda: Honoraria. Schreder:BMS-Celgene, Amgen: Consultancy. Paiva:Janssen: Consultancy, Honoraria; Karyopharm: Consultancy, Honoraria; Kite: Consultancy; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Sanofi: Consultancy, Honoraria, Research Funding; SkylineDx: Consultancy; Takeda: Consultancy, Honoraria, Research Funding; Roche: Research Funding; Adaptive: Honoraria; Amgen: Honoraria. Ludwig:Bristol Myers: Other: Advisory Boards, Speakers Bureau; Sanofi: Other: Advisory Boards, Speakers Bureau; Seattle Genetics: Other: Advisory Boards; Takeda: Research Funding; Amgen: Other: Advisory Boards, Research Funding, Speakers Bureau; Celgene: Speakers Bureau; Janssen: Other: Advisory Boards, Speakers Bureau. OffLabel Disclosure: Use of Carfilzomib in myeloma first-line treatment
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...