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: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 29-30
    Abstract: Background: Acute Myeloid Leukemia (AML) has a five-year survival rate of 25% and its high mortality is linked to poor response to treatment and relapse. Our understanding of the molecular mechanisms controlling relapse and AML progression is limited. Animal models indicate that AML cells significantly modulate their bone marrow microenvironment inducing gradual loss of endosteal and vascular niches, both playing critical roles in support and maintenance of normal hematopoiesis. The goal of this study was to determine microenvironmental factors driving the gradual retraction of endosteal and vascular niches directly in the AML core bone marrow biopsies, and assess the treatment effect on hematopoietic and non-hematopoietic cells. Methods: Transcriptomics and histopathologic evaluations of matched human AML core bone marrow biopsies obtained at diagnosis (n=12) and day 14 post-induction therapy (n=12) with daunorubicin and cytarabine (7+3) were performed. Based on post-treatment frequency of blasts in the AML bone marrow aspirate, patients were classified as responders ( & lt;5% blasts) or non-responders ( & gt; 5% blasts). Three of 6 responders (3 men, 3 women, average age 59 yrs) had normal karyotype, and three of 6 non-responders (1 man, 5 women, average age 52.6 yrs), had normal karyotype. RNA was isolated from the core bone marrow biopsies and subjected to Clariom D Human Affymetrix arrays. Transcriptomics data were analyzed using Affymetrix Transcriptome Analysis Console with LIMMA R package and Gene Set Enrichment Analysis (GSEA). H & E stained bone marrow biopsy slides were subjected to blinded histopathological assessment. Results: Transcriptomic data analysis of responder vs. non-responder samples at diagnosis indicated significant loss of transcripts associated with heme metabolism (HBB, HBD, GYPE, CA1) suggesting decrease in frequency of erythroid progenitors (Fig.A). Trends of decreased frequency of erythroid progenitors were noted in both bone marrow biopsies and aspirates of diagnostic non-responder samples (Fig.B). Decreased frequency of lymphoid cells was also noted (Fig.B). Interestingly, while post-treatment we noted a relative increase in frequencies of lymphoid cells in both responder and non-responder samples, the increase was more prominent in responders (Fig.B). Trilineage hematopoiesis appeared affected more in diagnostic and post-treatment responder samples. Transcriptome analyses of diagnostic vs. post-treatment responder samples indicated significant increase in transcripts associated with activity within endosteal niche (SPARC, SPP1, DCN, VCAN, BGN) and significant loss of transcripts associated with DNA replication (TOP2, HELLS, E2F8) (Fig.C), the latter was consistent with treatment-related loss of cellularity. Only modest increase in SPARC, SPP1 or BGN levels and no significant decrease in DNA-replication associated transcripts were noted in non-responder post-treatment samples (Fig.1D). These data indicate greater loss of AML cells and greater activity within the endosteal niche in responder in comparison to non-responder samples. Finally, analyses performed on post-treatment responder vs. non-responder samples showed significant decrease in SPARC, SPP1, DCN, VCAN, BGN in non-responder post-treatment samples (Fig.E, F). Endosteal niche in histopathologic evaluation at diagnosis was generally unremarkable in both responder and non-responder samples with only rare osteoblasts present. In contrast, post-treatment, we found an elevated number of osteoblasts in responders in comparison to non-responder samples (Fig.G, H). Conclusions: Transcriptomic and histopathologic analyses of AML bone marrow biopsies procured at diagnosis and post-treatment from responder or non-responders indicate inverse correlation between the activity of endosteal niche and levels of transcripts involved in osteoblast maturation and homeostasis. Significant suppression of mesenchymal/osteoblast component of the niche is observed in non-responder samples. To our knowledge this is a first report showing the correlation between levels of osteopontin (SPP1), osteonectin (SPARC) and biglycan (BGN) and response to chemotherapy directly in the AML core bone marrow biopsies. Our data suggest that osteo-stimulatory factors could be used to achieve better therapeutic outcomes in AML. 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: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2021
    In:  Blood Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1100-1100
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1100-1100
    Abstract: Cytokine storm or cytokine release syndrome is a systemic inflammatory response to different triggers which leads to excessive activation of immune cells with the release of a large amount of pro-inflammatory cytokines. These uncontrolled and excessive releases of cytokines accompany multisystem organ failure and death. Acute radiation exposure leads to acute hematopoietic and acute gastrointestinal syndromes with early mortality. Cytokine storm accompanies these syndromes and significantly complicates the clinical outcomes. Recent data has suggested that the inflammasome and associate cytokine storm may play a major role in various injuries and tissue problems after irradiation. Inflammasome activates caspase-1 which give rise to the proinflammatory cytokines such as interleukin-18 (IL-18) and interleukin-1 beta (IL-1β) into their biologically active forms and further induce other cytokines release. Extracellular vesicles (EVs) are nanosized lipid bilayer vesicles, released from cells in the whole body. EVs play an important role in intercellular communication. EVs have been shown to transfer protein, lipid, DNA, mRNAs, and microRNAs to recipient cells, thereby mediating a variety of biological responses. We have been investigating the capacity of marrow-derived mesenchymal stem cell extracellular vesicles (MSC-EV) to reverse radiation damage to murine bone marrow stem/progenitor cells. In this study, we have evaluated the effect of MSC-EVs on reversal of high dose radiation injury to bone marrow cells. C57BL/6 mice received 0, 700, and 950-1000cGy WBI (either single or split dose). After 24 hrs. post-irradiation, mice received an IV infusion of 2X10 9 hMSC EVs daily for three days with a control group receiving vehicle only. Marrow was harvested from 700 cGy exposed mice which were vesicle treated or not. These marrow cells were evaluated for engraftment into 950 cGy exposed mice. The engraftment rates in mice transplanted with marrow from irradiated EV injected animals was 34.66±14.19% at 4 months post transplantation and marrow from the group not treated with EV gave an engraftment rate of 6.92±3.53%. We further evaluated the effect of MSCs-EVs on decreasing the mortality in mice exposed to 950cGy WBI. The MSC-EV untreated mice were dead between 12-18 days post radiation, but MSC-EV treated mice maintained a 60% survival rate at 130 days post radiation, suggesting that MSC-EV treatment could significantly extend the survival rate of mice after exposure to lethal radiation. We evaluated the circulating inflammatory markers in 1000 cGy whole body irradiated mice with/without MSC-EV treatment. Twenty-four hours post-irradiation, mice received an IV infusion of 2x10 9 hMSC EVs every day for three days with a control group receiving vehicle only. The serum was collected after 8 days post radiation for inflammatory response analysis by ProcartaPlex multiplex immunoassays. There were dramatic increases in cytokines secretion in serum including IFN gamma, IL-12p70, IL-13, IL-1 beta, IL-2, IL-5, IL-6, TNF alpha, GM-CSF and IL-18 in irradiated mice compared to non-irradiated mice and a significant reduction in cytokine levels after MSC-EVs treatment. We further found significantly higher levels of IL-1 beta and activated caspase-1 p20 protein expression in the spleen after 1000cGy radiation compared to non-radiated mice. Moreover, we found that with MSC-EV treatment, the expression level of IL-1 beta and active caspase-1 p20 was significantly inhibited in spleen compared to untreated irradiated mice, indicating that the inhibition of cytokine storm post-radiation by MSC-EVs might be mediated by inhibition of inflammasome activation. Our study suggests that EV Inhibition of an inflammasome mediated cytokine storm could be therapeutically important in many disease entities. 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: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 11402-11402
    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 ...
  • 4
    In: American Journal of Respiratory Cell and Molecular Biology, American Thoracic Society, Vol. 62, No. 5 ( 2020-05), p. 577-587
    Type of Medium: Online Resource
    ISSN: 1044-1549 , 1535-4989
    RVK:
    Language: English
    Publisher: American Thoracic Society
    Publication Date: 2020
    detail.hit.zdb_id: 1025960-0
    detail.hit.zdb_id: 1473629-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 37-37
    Abstract: Extracellular vesicles (EVs) are critical mediators of intercellular communication within the bone marrow niche and have been implicated in numerous features of aging. However, their role in natural hematopoietic stem cell (HSC) aging has not been fully elucidated. The goal of this work was to test the hypothesis that EVs from whole bone marrow (BM-EVs) can modulate the HSC aging phenotype in vivo. With respect to HSC aging, our prior work showed that, in contrast to the well-known reduced functional capacity and prominent myeloid skewing displayed by old immunophenotypically-defined HSCs, old unseparated whole bone marrow (WBM) (24-26-mo-old) had a 4-fold increase in functional HSCs compared to young (6-8-wk-old) WBM in limit dilution competitive bone marrow transplantation, and showed minimal to no myeloid skewing. In order to test the ability of BM-EVs derived from this total WBM population to alter HSC aging phenotype, we first isolated EVs from WBM flushed from old (24-26-month old) and young (6-8-week old) C57/BL6 (CD45.2) mice by differential centrifugation (2000 × g for 30 min, supernatant centrifuged 100,000 × g for 1 hour, BM-EV pellet collected). Utilizing nanoparticle tracking analysis, we found no difference in mean particle size between old and young BM-EVs, but there was an approximately 2-fold increase in the number of EVs from old WBM compared to young WBM. To test the ability of these BM-EVs to alter HSC function in vivo, we injected old CD45.2 mice with 2 x 109 young BM-EVs and young CD45.2 mice with 2 x 109 old BM-EVs mice via tail vein, daily x 3 days. Control mice were injected with age-matched BM-EVs or vehicle alone. At one-month post injection, we isolated total WBM and Lineage negative/c-Kit+/Sca-1+/CD150+ cells (LSK-SLAM) from the EV-exposed or vehicle control mice. We injected either 3 x 105 WBM cells or 400 LSK-SLAM mixed with 3 x 105 healthy WBM competitor cells from young B6.SJL (CD45.1) mice into lethally irradiated young CD45.1 hosts and measured peripheral blood chimerism and lineage contribution by flow cytometry up to 6 months post-transplant. For the young marrow, exposure to old BM-EVs had no appreciable effects on engraftment capacity or lineage distribution. However, old WBM exposed to young BM-EVs exhibited a significant decrease in engraftment (15% ± 5%) when compared to old WBM exposed to age-matched old BM-EVs (61% ± 14%) or vehicle control (47% ± 7%) (% average donor chimerism ± SEM, n=4-5 mice/group, p & lt;0.04). Similarly, there was a trend toward decreased engraftment capacity by old LSK-SLAM after young BM-EV exposure (7% ± 4%) and increased engraftment capacity by the old LSK-SLAM after old BM-EV exposure (27% ± 10%) compared to vehicle control (15% ± 2%) (% average donor chimerism ± SEM, n=4-5 mice/group, p=not significant (ns)). These results are consistent with our prior data discussed above in which old un-separated WBM, the source of the old BM-EVs, had increased engraftment capacity compared to young WBM. Interestingly, there was also a trend toward reversal of classic myeloid skewing when old LSK-SLAM were exposed to young BM-EVs (36% ± 11%) compared to vehicle control (64% ± 3%) (average myeloid % of donor-derived peripheral blood ± SEM, n=4-5mice/group, p=ns). In addition, consistent with the known increase in LSK-SLAM with age, our preliminary data showed that old mice exposed to young BM-EVs had an approximately 7-fold decrease in the number of LSK-SLAM in marrow, indicating that BM-EVs within the young bone marrow microenvironment may modulate HSC population size. Finally, microRNA (miR) expression profiling (NanoString Technologies) indicated that a number of miRs known to be involved in hematopoiesis, proliferation, self-renewal, differentiation, senescence and inflammation were differentially represented in old and young BM-EVs, with miR-29a, miR-24, and miR-21 significantly increased and miR-105 significantly decreased in old BM-EVs compared to young BM-EVs. Together, these data indicate that young BM-EVs, via transfer of differentially age-related cargo, may alter engraftment capacity, modify the lineage commitment and may regulate LSK-SLAM population size during natural aging. Future studies more clearly delineating age-associated BM-EV effects on HSCs and defining the molecular mechanisms underlying these effects could yield key insights into the natural aging of HSCs and facilitate restoration of healthy hematopoiesis in the elderly. 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: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 142, No. Supplement 1 ( 2023-11-02), p. 6874-6874
    Abstract: Background: Previous studies have shown durable AML patient responses following donor lymphocyte infusions (DLI) in the absence of engraftment in both the frontline and relapsed setting (Dey et al, BJH 2005, Colvin et al, BBMT 2009, Ai et al, Blood 2010). Herein we report results from the completed dose-escalation phase from our trial of fractionated dosing of gemtuzumab ozogamicin (GO) followed by non-engraftment mismatched allogeneic donor cells infusion in patients with relapsed/refractory (R/R) AML. In this trial, instead of engraftment, it is postulated that donor cells serve as a potent immune stimulator. Study Design and Methods: In this phase I/II study, GO 6-9mg/m 2 total dose was administered as a fractionated 2-3mg/m 2 dose (capped at 4.5mg) on days 1, 4, and 7, followed by infusion of HLA-mismatched (0-3/6 antigens) related donor cells on day 8 [NCT03374332]. The dose-escalation phase 1 cohort used a 3+3 design combining GO with donor leucocyte infusion (DLI) at dose 1-2x10 7 CD3+ cells/kg (Dose Level 1) or 1-2x10 8 CD3+ cells/kg (Dose Level 2). Patients enrolled in the phase II portion were treated at the maximum tolerated dose (MTD) level. Lab correlative studies were drawn pre and post DLI to assess immune effector cells and leukemic blast surface expr ession as well as cytokine release profiles. Patients 18 years of age and older with histologically confirmed R/R AML who had recurrence or progression after at least 1 prior standard treatment were eligible. Enrollees must have had no active systemic infections and adequate lung, liver, cardiac, and renal function with an ECOG PS 0-1. Patients in CR or CRi after initial treatment could receive up to 2 additional treatments with GO+DLI. Results: Eleven patients (7 men and 4 women) were enrolled into the study. The median age was 72 years (54-81 years) and patients had received 1-4 previous lines of treatment. All patients were refractory to hypomethylating agents and venetoclax. No patient was a candidate for allogeneic stem cell transplant at the time of study enrollment. Ten patients received 3/6 and one patient received 1/6 HLA mismatched donor cells from a first- or second-degree relative. The MTD was determined to be 2x10 8 CD3+ cells/kg. Three patients were enrolled on dose level 1 and eight patients were enrolled on dose level 2. Treatment was well tolerated with minimal Grade 3 or 4 non-hematologic adverse events attributable to the study ( Table 1). One patient at dose level 2 developed septic shock and died prior to DLI while another patient at dose level 2 developed cryptococcal fungemia and died within one week of DLI. No donor experienced a Grade 3 or 4 adverse event. All patients had fever post DLI that fit criteria for Grade 1 or 2 CRS while one patient developed Grade 3 CR. Two patients with Grade 2 CRS and one patient with Grade 3 CRS received tociluzimab. One patient attained a CR after one cycle of GO+DLI. All other patients had progressive disease. Ultimately the study was stopped early due to futility. Median IL-6 and IL-8 levels were higher in the first 3 patients at dose level 2 (605.5pg/mL and 218.5 pg/mL) compared to the three patients in dose level 1 (79pg/mL and 57.8 pg/mL) but the difference was not statistically significant. IL-10 levels were significantly higher in dose level 2 than dose level 1(125 vs 14.8 pg/mL, p=0.05) Conclusions: The MTD of DLI in combination with GO for relapsed/refractory AML patients is 2x10 8 CD3+ cells/kg. Treatment in an older relapsed/refractory population was well tolerated. Patients reliably developed fevers post DLI with Grade1 or 2 CRS. However, there was no evidence that fever development corresponded to a clinical benefit or that the addition of non-engraftment DLI augmented the effect of single agent gemtuzumab. Additional analysis of cytokine release profiles for the remaining patients as well as immune checkpoint inhibition on leukemic blasts for all patients will be presented at the meeting.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2023
    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: British Journal of Haematology, Wiley, Vol. 200, No. 6 ( 2023-03), p. 740-754
    Abstract: While the bone marrow (BM) microenvironment is significantly remodelled in acute myeloid leukaemia (AML), molecular insight into AML‐specific alterations in the microenvironment has been historically limited by the analysis of liquid marrow aspirates rather than core biopsies that contain solid‐phase BM stroma. We assessed the effect of anthracycline‐ and cytarabine‐based induction chemotherapy on both haematopoietic and non‐haematopoietic cells directly in core BM biopsies using RNA‐seq and histological analysis. We compared matched human core BM biopsies at diagnosis and 2 weeks after cytarabine‐ and anthracycline‐based induction therapy in responders ( 〈 5% blasts present after treatment) and non‐responders (≥5% blasts present after treatment). Our data indicated enrichment in vimentin ( VIM ), platelet‐derived growth factor receptor beta ( PDGFRB ) and Snail family transcriptional repressor 2 ( SNAI2 ) transcripts in responders, consistent with the reactivation of the mesenchymal population in the BM stroma. Enrichment of osteoblast maturation‐related transcripts of biglycan ( BGN ), osteopontin ( SPP1 ) and osteonectin ( SPARC ) was observed in non‐responders. To the best of our knowledge, this is the first report demonstrating distinct osteogenic and mesenchymal transcriptome profiles specific to AML response to induction chemotherapy assessed directly in core BM biopsies. Detailing treatment response‐specific alterations in the BM stroma may inform optimised therapeutic strategies for AML.
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 80077-6
    detail.hit.zdb_id: 1475751-5
    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...