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
  • 11
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4543-4543
    Abstract: The concept of suppressing dendritic cells (DCs) to prevent graft-versus-host disease (GvHD) following allogeneic hematopoietic stem cell transplantation (HSCT) has been rejuvenated since the introduction of proteasome inhibitors into clinical practice. Although early post-transplant administration of proteasome inhibitors ameliorates GvHD, delayed administration has been associated with alloreactivity-dependent gastrointestinal toxicity. This phenomenon is associated with a surge in interleukin-1β (IL-1β) and acceleration in donor T cell expansion. To address this issue, we sought to investigate whether in vivo depletion of donor T cells by concomitant administration of post-transplant cyclophosphamide (C) could overcome this phenomenon. Ten-week old BALB/c mice were irradiated (10 grays in two fractions) on day -1 and transplanted with 5x106 BM cells from C57BL6 mice. GvH inocula consisted of 5x106 splenocytes from C57BL6 (some post-CFSE labeling) or B6;FVB-PtprcaTg(CAG-Luc,-GFP)L2G85Chco Thy1a/J (ubiquitously transgenic for firefly luciferase for bioluminescence imaging (BLI). All mice were purchased from Jackson Laboratory and kept in a pathogen-free environment. Mice were untreated or received ixazomib (Z) (30μg, subcutaneously) on days -1, +2, and +5, C (1 mg intraperitoneally) on day +2, or both Z and C according to the same schedule. Mice were monitored for weight, GvHD score, and survival or sacrificed on day +6 for cytokine measurement and splenic cell counts. For BLI, mice receiving transgenic splenocytes were injected with 150mg/kg of D-Luciferin intraperitoneally and imaged 25 minutes later. GvHD worsening and sudden death occurred in a fraction of mice receiving Z following day +5 injection. The phenomenon was completely prevented by the addition of C. Overall, the group receiving both Z and C had significantly improved weight, GvHD score, and survival when compared to the mice left untreated or receiving either drug alone (figure 1). The addition of C to Z prevented IL-1β increase, was associated with suppression of tumor necrosis factor α (TNFα), and resulted in a profound depletion of splenic total and donor CD4+ cells. Furthermore, proliferating cells (CFSE low) were preferentially depleted as opposed to quiescent cells (CFSE high). BLI imaging showed an increase in signal intensity shortly after the administration of Z on day +5. On the other hand, the mice receiving C in addition to Z exhibited significantly lower donor T cell proliferation when compared to the other groups (figure 2). These findings suggest that C can prevent the phenomenon of GvHD acceleration associated with the extended administration of proteasome inhibitors, suppress cytokine production, and effectively reduce splenic total and donor CD4+ cell expansion with preferential depletion of proliferating as opposed to quiescent cells. The combination of Z and C seems a promising duplet for the prevention of GvHD. When Z is administered late following allogeneic HSCT for the prevention of disease relapse, the addition of C should be considered in order to avoid any unwanted aggravation of alloreactivity. Figure 1 Figure 1. Figure 2 Figure 2. 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: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 12
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4635-4635
    Abstract: High-dose melphalan (M) followed by autologous hematopoietic stem cell transplantation (AHSCT) remains the standard treatment for multiple myeloma in eligible patients, even in the era of novel agents. However, the majority of patients ultimately relapse and succumb to their disease. Several studies have explored integrating novel agents into the conditioning regimen prior to AHSCT in order to improve complete remission rates and ultimately overall survival. We aimed to assess the feasibility of adding bendamustine (B) and carfilzomib (C) to melphalan (BCM) and performed a phase I dose escalation study. Thirteen patients were enrolled between June 2014 and June 2016. All patients received C at a fixed dose (20 mg/m2) on days (d) -29, -28, -22, -21, -15, -14. The conditioning regimen and doses administered for each cohort were as described in the table below. Due to excessive toxicity, the study was amended after the first 6 patients. Per oversight of a data safety monitoring board, the dose of M was reduced to 140 mg/m2 and C dose on d +6 was omitted. Median age was 58 years (39-68). There were 8 males and 5 females. Performance status was ≥ 80% in all patients. Per the International Staging System (ISS), 3 patients had stage I disease, 5 had stage II, 4 had stage III, and 1 had unknown staging. Three patients had high-risk cytogenetics: 2 with t(4;14) and 1 with deletion 17p. Three patients had received prior AHSCT. Disease status prior to study treatment was stable disease (SD) (n=2), partial response (PR) (n=8), or very good partial response (VGPR) (n=3). Median CD34+ cell dose was 3.24x106/kg (2.23-6.92x106). Median follow-up was 14.2 months (1-24.5). Median time to neutrophil engraftment was 12 d (11-15). One patient died before achieving platelet engraftment. For the remaining patients, median time to platelet engraftment was 16 d (12-20). Non-hematologic toxicities included grade 3 acute mucositis (n=1), lower GI complications (n=6), electrolyte disturbances (n=6), transaminase elevation (n=1) renal insufficiency (n=1), pulmonary edema (n=1), prolonged QTc (n=1), atrial fibrillation (n=1), and elevated troponin (n=1) and grade 4 acute sepsis (n=2), resulting in 1 death in cohort 2 on d +44. Seven patients went on to receive maintenance therapy: 3 with bortezomib, 3 with lenalidomide, and 1 with lenalidomide, dexamethasone, and C. Post-transplant disease status was assessed per protocol by SPEP, SPIF, and serum free light chains and light chain ratio. Nine patients were evaluable on d +100. One patient had SD, 6 had VGPR, and 2 had complete response (CR). Six out of 7 (86%) evaluable patients on d +365 remain disease progression-free. In summary, BCM conditioning prior to AHSCT at the doses described in cohort 3b seems feasible with manageable toxicities. Five additional patients are being enrolled at the same dose level. Table Table. 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: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 13
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 7628-7630
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 14
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1881-1881
    Abstract: Background: Targeting T cells alone has yielded limited success in the prevention of graft-versus-host disease (GvHD) following allogeneic blood and marrow transplantation (BMT). Dendritic cells (DCs) play a central role in alloreactivity and therefore represent a suitable target. Proteasome inhibitors (PI), with their ability to inhibit the function and maturation of DC, have prompted investigators to examine their potential role in the prevention of GvHD. The investigational PI, ixazomib (IXZ), dissociates rapidly from 20S and is therefore truly reversible. It is also orally bioavailable. Our aim in this study was to explore its effect on healthy volunteer peripheral blood dendritic and T cells and in a pre-clinical GvHD mouse model. Methods: To characterize the effects of IXZ on healthy volunteer peripheral blood DCs, DCs were isolated using EasySep Pan-DC Pre-Enrichment Cocktail with purity over 90% (STEMCELL Technologies). DCs were then treated with IXZ at different concentrations (10-40nM) for 4 hrs and then stimulated with lipopolysaccharide (LPS) for 16 hrs. After this treatment, DCs were surface stained with antibodies against maturation markers and analyzed by flow cytometry. DC survival was evaluated with 7AAD staining and FACS analysis. To assess the effect of IXZ on the production of pro-inflammatory cytokines, DCs were incubated with IXZ at increasing concentration before or after the addition of LPS. Total pro-inflammatory cytokines in the supernatant of tissue culture were measured using EMD Millipore cytokine arrays. Standard mixed lymphocyte reaction and T cell proliferation assays were used to evaluate T cell function. At a minimum, all experiments were done in triplicate. Unpaired t test was used for statistical analysis. A p-value 〈 0.05 was considered significant. The B6 → BALB/c pre-clinical GvHD model was adopted to evaluate the effect of IXZ on GvHD development. Mice were transplanted in 3 groups. Group 1 received a lethal dose of total body irradiation (TBI), donor bone marrow (BM) cells, and IXZ, group 2 received TBI, donor BM cells donor splenocytes, and a vehicle, and group 3 received TBI, donor BM cells, donor splenocytes, and IXZ. The dose of BM cells and splenocytes was 5 X 106 each. IXZ was given at 1.5 mg/kg subcutaneously. Two dosing schedules were tested in 2 separate experiments: day-1 and +2 or day +1 and +4. Results: IXZ inhibited the expression of 6 DC maturation markers including CD40, CD54, CD80, CD83, CD86 and CD197 (CCR-7). The inhibition started at a concentration of 10nM and was dose-related. IXZ also decreased the percentage of total DCs simultaneously expressing multiple markers. DCs viability remained unchanged in comparison to control at a concentration of 10nM and dropped to 68% and 43%, on average with concentrations of 20nM and 40nM, respectively. IXZ significantly decreased DC production of IL-6, IL-12, and IL-23 starting at the concentration of 20nM. IL-1β was decreased at the concentration of 40 nM. Importantly, there was no significant change in the cytokine production by DCs when IXZ was added 4 hrs after LPS except for IL-1β which increased at 30nM. Starting at the concentration of 10nM, IXZ dose-dependently inhibited T cell proliferation. At 40nM IXZ abolished T cells. In our in vivo study IXZ improved GvHD scores on days +7 and +11 in group 3 in comparison to group 2 when it was given on days -1 and +2. Conversely, when IXZ was given on day +1 and +4, group 3 mice had higher scores of GvHD and worse survival outcomes when compared to group 2. There was no noticeable drug toxicity in group 1 mice. Conclusion: In summary: 1) IXZ inhibits DC maturation with relative preservation of cell viability and inhibits pro-inflammatory cytokine production in DCs when added before LPS stimulation; 2) IXZ inhibits T-cell proliferation; 3) IXZ affects GvHD development in a schedule-dependent fashion with early administration improving and late administration worsening GvHD. Additional analysis of tissue and serum samples is in progress. These results provide background for careful design of clinical trials using IXZ for the prevention of GvHD. Disclosures Al-Homsi: Millennium Pharmaceuticals: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 15
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1398-1398
    Abstract: Introduction: CYAD-01 is a chimeric antigen receptor T-cell (CAR-T) product based on the receptor NKG2D which demonstrated anti-tumor efficacy through different mechanisms of action in numerous preclinical models. A comprehensive clinical program was developed with the aim to define the optimal CYAD-01 treatment in acute myeloid leukemia (AML). Methods: The ongoing Phase I THINK trial (NCT03018405) evaluates multiple CYAD-01 injections in 2 parallel cohorts: one in patients (pt) with metastatic solid tumors and the other one in relapsing/refractory hematological malignancies. The dose escalation segment of the study evaluates 3 dose levels (DL; 3x108, 1x109 and 3x109 cells per injection) of one cycle of 3 CYAD-01 administration at 2 weeks apart. As of July 30, 2018, 12 pts in the THINK hematological cohort (8 AML, 3 MM and 1 MDS) have been enrolled at the 3 DLs without prior preconditioning. No dose-limiting toxicity occurred. We evidenced promising anti-leukemic activity with 42% ORR in r/r AML with 5/7 pts having clinical benefit. Based on the initial data generated into the THINK study, two additional Phase I clinical trials have been developed to evaluate 3 DLs (1x108, 3x108 and 1x109 cells per injection) post preconditioning regimen or in association with standard of care (SoC). The DEPLETHINK study (NCT03466320) aims at evaluating the CYAD-01 treatment administered after a cyclophosphamide and fludarabine preconditioning regimen to r/r AML patients. The preconditioning chemotherapy should (i) favor the expansion and engraftment of CYAD-01, (ii) increase NKG2D ligand expression on tumor cells and (iii) help to overcome the strong immunosuppressive microenvironment. The EPITHINK study (EudraCT 2018-000745-39) aims at assessing the safety of the CYAD-01 treatment administered concurrently with SoC 5-azacytidine treatment in treatment-naïve AML pts ineligible for intensive treatment. Similar to the preconditioning treatment, the epigenetic treatment could contribute to favor engraftment of CYAD-01 cells and increase target antigen expression while better controlling the disease progression of patients early in the treatment. Results: Description of the study design and preliminary clinical data from the first cohorts of the EPITHINK and DEPLETHINK studies will be presented. The preliminary data in term of CYAD-01 engraftment kinetics, NKG2D ligand expression (including blasts and soluble ligands), and the kinetics of cytokine induction will be correlated with the concurrent treatment (preconditioning or epigenetic treatment) versus the stand alone therapeutic approach. Conclusions: We have demonstrated the feasibility and safety of multiple injections of CYAD-01 without preconditioning chemotherapy (THINK). Preliminary safety, activity and correlative science data will be presented according a prior preconditioning (DEPLETHINK) and concurrent administration with SoC epigenetic treatment (EPITHINK). Disclosures Sallman: Celgene: Research Funding, Speakers Bureau. Kerre:BMS: Consultancy; Celgene: Consultancy, Research Funding; Celyad: Consultancy. Wang:Novartis: Speakers Bureau; Amgen: Consultancy; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz: Speakers Bureau; Novartis: Speakers Bureau; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz: Speakers Bureau. Selleslag:Kiadis Pharma: Other: Financial support for study-related issues. Beguin:Kiadis Pharma: Consultancy. Al-Homsi:Celyad: Membership on an entity's Board of Directors or advisory committees. Dekker:Celyad: Employment. Breman:Celyad: Employment. Sotiropoulou:Celyad: Employment. Snykers:Celyad: Employment. Braun:Celyad: Employment. Lonez:Celyad: Employment. Verma:Celyad: Employment. Lehmann:Celyad: Employment, Honoraria, Patents & Royalties; GSK: Patents & Royalties. Davila:Celyad: Consultancy, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 16
    Online Resource
    Online Resource
    American Society of Hematology ; 2010
    In:  Blood Vol. 116, No. 21 ( 2010-11-19), p. 1400-1400
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1400-1400
    Abstract: Abstract 1400 Introduction: Hemostatic abnormalities are frequently encountered during initial presentation of acute myeloid leukemia (AML) as well as during induction treatment. Patients often present with decreased platelet counts and sometimes with abnormalities of the coagulation parameters with or without bleeding manifestations. In the absence of overt disseminated intravascular coagulopathy (DIC), transfusion practices during treatment are essentially based on the platelet count. Fibrinogen is an important element of the clotting mechanisms, constituting a template for both thrombin binding and the fibrinolytic system. We sought to determine the prevalence of altered serum fibrinogen levels during induction treatment in AML and its clinical significance. Methods: We prospectively measured serum fibrinogen on a daily basis in patients with newly diagnosed AML treated with induction anti-tumor chemotherapy at our institution from February 2007 to July 2010. Inclusion criteria included the diagnosis of AML and no overt DIC. Results: The study population included 17 patients (47% females) with mean age of 55.3 years (range 18 –80 years). At presentation, the mean platelet count was 62 × 109/L (range 3 – 252 × 109/L), the mean prothrombin time was 11.4 seconds (range 9.6–13.4 secs, normal range 10—13.7 secs), the mean international normalized ratio was 1.1 (range 0.9–1.3; normal range 0.9–1.1) and the mean activated partial thromboplastin time was 26.56 seconds (range 20.1–41 secs, normal range 22.3–34.0 secs). All patients had normal serum fibrinogen levels at presentation (mean 380.6 mg/dl; range 258–567 mg/dl; normal range 200–400 mg/dl). Thirteen patients were treated with idarubicin and cytarabine, 3 patients received a FLAG (fludarabine, cytarabine and G-CSF) regimen and one patient had amonafide and cytarabine. Serum fibrinogen levels were recorded on all patients on a daily basis (Figure 1). Nine (53%) developed hypofibrinogenemia on the fourth day of induction, 2 (12%) on day 5, 3 (17%) on day 6, 2 (12%) on day 7, and one (6%) on day 8 of the induction. Eight patients (47%) received prophylactic cryoprecipitate when the serum fibrinogen levels fell below 150 mg/dl. We did not observe a significant trend difference in serum fibrinogen levels between patients who received cryoprecipitate and those who did not. Serum fibrinogen levels were back to normal without transfusion support by day 6 for one patient (5.88 %), day 7 for 2 patients (11.76 %), day 8 for 3 patients (17.65 %), day 9 for one patient (5.88 %), day 10 for one patient (5.88 %), day 11 for 3 patients (17.65 %), day 12 for 6 patients (35.3 %). Conclusion: Patients with acute myeloid leukemia receiving induction chemotherapy may frequently develop isolated hypofibrinogenemia without evidence of disseminated intravascular coagulation. This finding is usually self-limited and disappears shortly after the completion of antitumor chemotherapy, usually by day 12. The mechanism of this under-recognized phenomenon is unclear. Its common occurrence raises questions about the appropriateness of transfusion practices in AML based solely on the platelet count and argues in favor of the need of more global tests such as thromboelastography. 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: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 17
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 875-875
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 875-875
    Abstract: Medically-ill patients are at increased risk for developing venous thromboembolism (VTE) due to multiple risk factors. Evidence-based practice guidelines recommend pharmacologic modalities such as unfractionated heparin (UFH), low molecular weight heparins (LMWH; i.e.enoxaparin, dalteparin, nadroparin) and pentasaccharide fondaparinux for the prevention of VTE. We conducted systematic review and meta-analysis to evaluate the role of different interventions in prevention of VTE in medically-ill patients. We searched MEDLINE, EMBASE and the Cochrane Controlled Trials Register from 1981–2006 for randomized controlled trials reflecting contemporary antithrombin strategies against in-hospital VTE. Search terms included heparin, enoxaparin, dalteparin, nadroparin, and fondaparinux. Eligible studies included medically-ill patients with risk factors for VTE followed for 7–21 days. We constructed a meta-analysis using Comprehensive Meta Analysis, version 2.0 using the Der-Simonian-Laird random effects model as a sensitivity analysis. Primary end points included the incidence of DVT or PE. Secondary end points included minor and major bleeding, and death from VTE. Eight trials representing 9914 patients were selected for analysis. Four trials compared a LMWH to placebo and four trials compared a LMWH to UFH. Fondaparinux was considered an ultra LMWH. The mean age of represented patients was 72.7±4.89 years, with a mean weight of 68±2.57 kg. The results of primary end points are; 1. DVT-LMWH compared to placebo reduced the odds of DVT by 42% (OR=0.58, 95% CI [0.45–0.75], p≤0.001) with an absolute risk reduction (ARR) of 2.21% and number needed to treat (NNT) of 46. Compared to either placebo or UFH, LMWH reduced the odds of DVT by 37% (OR=0.63, 95% CI [0.50–0.79] , p≤0.001) with an ARR of 1.28% and NNT of 78. 2. PE-There was no difference in the incidence of PE with either LMWH, placebo (p=0.16) or UFH (p=0.73). A 36% reduction in the odds PE or DVT (OR=0.64, 95% CI [0.49–0.82], p≤0.01, ARR=1.44%, NNT=70) was observed using a LWMH instead of UFH or placebo. The latter results were driven by the PRIME and PREVENT studies using enoxaparin and dalteparin respectively. The results of secondary end points; 1. Minor bleeding-LMWH use was associated with an increased risk of minor bleeding (OR=1.64, 95% CI [1.18–2.29] , p=0.003) compared to placebo with an absolute risk increase (ARI) of 2.24% and a number needed to harm (NNH) of 45; where MEDNOX (enoxaparin study) was responsible for this difference. 2. Major bleeding-There was no difference in major bleeding with LMWH compared to placebo (p=0.18) or UFH (p=0.42). 3. VTE mortality-LMWH was similar to placebo (p=0.63) and UFH (p=0.47) for the reduction of VTE related death. LMWH also increased the risk of any bleeding or VTE-related death (OR=1.57, 95% CI [1.17–2.10], p=0.003, ARI=2.45%, NNH=41) compared to placebo which also was driven by an increase in minor bleeding from enoxaparin use. In conclusion, although there is a significant reduction in DVT when utilizing a LMWH rather than UFH or placebo, there is however, no difference in PE events. Furthermore, it is important to consider the increased incidence of minor bleeding with LMWH (NNH 43) which may offset the reduction in DVT (NNT 45). The objective of future studies should focus on the identification of potential subgroups of patients with higher risk of VTE and lower risk of bleeding to improve outcomes and optimize safety.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 18
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1180-1180
    Abstract: Background: HuLuc63 is a humanized monoclonal antibody directed against CS1 that has demonstrated potent in vivo and in vitro activity against myeloma cells in pre-clinical models. CS1 is a cell surface glycoprotein expressed at high levels on myeloma cells, plasma cells and to a lesser extent on a subset of CD8 + lymphocytes and natural killer (NK) cells. A Phase I study was initiated to evaluate the maximum tolerated dose (MTD) of HuLuc63 in a population of relapsed refractory multiple myeloma patients who have been exposed to at least two prior therapies. In a traditional phase 1 design, six potential dosing cohorts are planned with 3 to 6 patients enrolled per cohort to evaluate safety, pharmacokinetics (PK), biologic activity and clinical response. Each course of HuLuc63 is given IV at the same dose every 2 weeks for a total of 4 doses at a level of 0.5, 1, 2.5, 5, 10, 20 mg/kg. An additional 4 doses may be administered if the patient has demonstrated at least stable disease during the initial course of therapy. To date, a total of 7 patients have been treated at 2 dose levels; 3 in the 0.5 mg/kg cohort and 4 in the 1.0 mg/kg cohort. Median patient age is 64 years and the median number of prior therapies is five. An additional patient was enrolled in the 1.0 mg/kg cohort as a patient in that cohort had a 2-week delay in receiving his second dose due to worsening pain at the site of a pre-existing lytic bone lesion. No dose-limiting toxicities have occurred. Infusions were well tolerated, with grade 1 and 2 toxicities occurring during or after the first dose including chills (2 patients), flushing, pyrexia, rigors, dyspnea and fatigue. Patients who reported first dose reactions were pretreated with acetaminophen and diphenhydramine prior to subsequent doses and only one patient reported fatigue after the second dose. Four serious adverse events have occurred in 2 patients which were considered unrelated to HuLuc63. These include migraine headache (twice in the same patient), congestive heart failure and hypercalcemia. Two patients discontinued therapy prematurely due to progressive disease but all other patients have received 4 doses of HuLuc63. No clinical responses have been observed. In xenograft mouse models, a consistent drug level of 10 mcg / mL was required to show minimal biologic activity. Preliminary PK data reveals that peak serum drug levels for the 0.5 mg/kg dosing cohort reached 10 mcg / mL, which was sufficient to achieve CS 1 saturation of at least 70% on the antigen rich NK cell subset. Drug levels dropped below 1 mcg/mL by day 7, however, coinciding with a decrease in saturation. This indicates that the higher doses to be used in subsequent cohorts may achieve and surpass sustained concentrations in patients above this level. Thus far, HuLuc63 appears to be well tolerated in patients with multiple myeloma. Enrollment is continuing to determine the MTD.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    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...