Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Society of Hematology  (69)
  • 1
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 4460-4460
    Abstract: Background: Allogeneic stem cell transplant (allo-SCT) is a cornerstone in the treatment of Acute Myeloid Leukemia (AML) exerting much of its therapeutic efficacy through the graft-versus-leukemia effect. The administration of immunoregulatory cells via engrafted donor lymphocytes is essential to the eradication of residual malignancy and long-term survival. Many patients, however, either do not have an allo-SCT donor or cannot withstand the toxicity of allo-SCT. Previous studies have shown durable responses in AML patients following donor lymphocyte infusions 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). In this clinical trial we propose a role for donor leukocyte infusions (DLI) in the absence of engraftment. Without the need for engraftment patients will not need to receive high dose chemotherapy or radiation and the toxicities that accompany these therapies. Instead, allogeneic donor cells are infused into patient with relapsed/refractory (R/R) AML to serve as a potent immune stimulator. Prior to DLI, patients will receive fractionated dosing of gemtuzumab ozogamicin (GO). GO is an anti-CD-33 antibody drug conjugate approved in combination with induction therapy for de novo AML and in R/R disease. Patients who demonstrate a CR or CRi to therapy will go on to have up to 2 additional GO + DLI treatments. Bone marrow and blood samples will be obtained from patients before, during, and after treatment to determine immune effector cells (both donor and patient), cytokine release profiles, and extracellular vesicle components. Study Design and Methods: Our study, BrUOG 345 [NCT03374332], evaluates the combination of fractionated GO with non-engraftment DLI in the treatment of patients with R/R AML. Adults patients 18 years of age and older with histologically confirmed R/R AML who have had recurrence or progression after at least 1 prior standard treatment are eligible. Enrollees must have no active systemic infections and have adequate lung, liver, cardiac, and renal function with an ECOG PS 0-1. Fractionated GO 6-9mg/m2 will be administered on days 1, 4, and 7 followed by infusion of 1-2x108 CD3 cells/kg from a 0-3/6 HLA mismatched related donor cell. Patients that are in CR or CRi can receive up to 2 additional treatments with GO+DLI (Figure 1). The primary objective of the phase 1 portion is to determine the maximum tolerated dose (MTD) of DLI in combination with GO while the primary objective of Phase 2 portion is to assess the response rate after one cycle of fractionated GO followed by non-engraftment DLI in patients with relapsed/refractory AML. The study will initially utilize a 3+3 design in Phase 1 to determine if 1-2x108 CD3 cells/kg can be safely administered with GO. This study will target a response rate of 57% considered to be interesting enough to warrant further study in a randomized setting. With this hypothesis in mind, the phase 2 portion of the study will use Simon's two-stage design. The null hypothesis that the true response rate (CR and CRi) is only 29% will be tested against a one-sided alternative. In the first stage, 9 subjects will be accrued. Patients treated at MTD in the Phase 1 portion of the study will be included in this cohort. If there are 3 or fewer responses in these 9 patients, the study will be stopped for futility. Otherwise, 6 additional subjects will be accrued for a total of 15. The null hypothesis will be rejected if 7 or more responses are observed in 15 patients. This design yields a type I error rate of 0.1 and power of 80% if the true response rate is 57%. A continuous assessment of toxicity will be utilized for the Phase 2 portion of this study. Sequential boundaries will be used to monitor dose-limiting toxicity rate for patients after the initial MTD is determined. Accrual will be halted if excessive numbers of dose-limiting toxicities are seen. The primary outcome of the Phase 2 portion is the CR/CRi rate following GO and non-engraftment DLI. Secondary outcomes will include survival, both progression-free (PFS) and overall (OS) until two years post treatment, and dose limiting toxicities until 16 weeks post-infusion. Additional lab correlative studies will be performed including CD33 expression before, during, and after GO infusion and T-cell activation markers, antigen presenting cell/macrophage amounts, cytokine release profiles, and extracellular vesicle measurements (Figure 1). Disclosures Olszewski: TG Therapeutics: Research Funding; Spectrum Pharmaceuticals: Research Funding; Adaptive Biotechnologies: Research Funding; Genentech: Research Funding. Reagan:Pfizer: Research Funding; Incyte: 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: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 4589-4589
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 4589-4589
    Abstract: Abstract 4589 Introduction: Spontaneous remission of chronic lymphocytic leukemia (SRCLL) has occassionaly been reported since the 1950s. About 40–50 cases have been reported in the English literature. The often quoted incidence of such a phenomena is estimated to be 1% of all CLL cases (Table 1). However, a review of these reports which have determined this incidence suggests that many patients did not have a true SRCLL. The authors have conducted a systemic search within their own institution database to determine the true incidence of SRCLL. Methods: After obtaining appropriate IRB approval, a systemic search was performed within the our institution's hematopathology databases for the diagnosis of chronic lymphocytic leukemia from year 2000 to 2012. Records and laboratory findings were reviewed for all cases. Records for patients with laboratory data suggestive of remission were reviewed to determine if they had received any form of therapy. All cases were included in the review if the diagnosis of CLL was firmly established by flow cytometry. SRCLL was defined as: 1) the patient's elevated lymphocyte number at diagnosis was 〉 5 × 109/L and persisted for 〉 6 months confirming the initial diagnosis of CLL, 2) the lymphocyte number subsequently normalized to 〈 5 × 109/L, the lymphocyte count remained normal for greater than 9 months, 3) resolution of other features of CLL such as lymphadenopathy, and 4) no intervening treatment was given to patients, including radiation therapy or steroids. Results: After reviewing 167 cases, only one case met our inclusion criteria, with an estimated incidence of 0.6%. Conclusion: Spontaneous remission of CLL is a rare phenomenon. This review suggests that the incidence of SRCLL is less then the previously often quoted incidence of 1% and may more appropriately approximate 0.6%. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 4187-4187
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 4187-4187
    Abstract: Directed differentiation is defined as the ability to program a stem cell at the most primitive level while it still has its reproductive and full proliferative potential. This is in contrast to ex-vivo expansion where the stem cells are forced into specific lineage commitments, limiting the overall therapeutic utility. We have reproducibly induced directed stem cell differentiation towards megakaryopoiesis by capitalizing on inherent changes in sensitivities to inductive cytokine signals in the context of cell cycle position. Murine experiments have been performed on highly purified quiescent G0–1 lineagenegative rhodaminelowHoeschtlow (LRH) marrow stem cells. When exposed to thrombopoietin, FLT3-ligand and steel factor (TFS), they synchronously pass through cell cycle. Megakaryopoiesis is focused at early to mid S-phase, returning to baseline before initial cell division. Population based differentiation cultures after 14-days produced up to 49% megakaryocytes with stem cells sub-cultured during early-mid S-phase with little to no production with colonies cultured from stem cells in G0–1 or G2 phase at time directed differentiation signaling. Gene expression showed over 2 fold increases in FOG, Nfe2 and Fli1. Clonal studies confirm the results. In one experiment, 33% of clonally derived colonies that grew from early-S phase cells and 10% of colonies that grew from mid-S phase cells had megakaryocytes present compared with 0% for G0–1 and G2 cells. We have now worked with human lineagenegative double-effluxed-rhodaminelow double-effluxed-Hoeschtlow G0–1 stem cells. When expose to TFS cytokines, there initial cell cycle lasts more than 80 hours opposed to CD34+ cells and murine LRH cells which have divided by 44–48 hours. This human population of stem cells comprises approximately 0.01% of CD34+ cells and has tremendous promise in replicating our murine work, elucidating opportunities for human translational work targeting patients that have a block of differentiation toward megakaryopoiesis i.e. sub-sets of autologous transplant or myelodysplastic syndrome patients.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 132, No. 19 ( 2018-11-08), p. 2053-2066
    Abstract: Although the pathogenesis of primary myelofibrosis (PMF) and other myeloproliferative neoplasms (MPNs) is linked to constitutive activation of the JAK-STAT pathway, JAK inhibitors have neither curative nor MPN-stem cell-eradicating potential, indicating that other targetable mechanisms are contributing to the pathophysiology of MPNs. We previously demonstrated that Abelson interactor 1 (Abi-1), a negative regulator of Abelson kinase 1, functions as a tumor suppressor. Here we present data showing that bone marrow-specific deletion of Abi1 in a novel mouse model leads to development of an MPN-like phenotype resembling human PMF. Abi1 loss resulted in a significant increase in the activity of the Src family kinases (SFKs), STAT3, and NF-κB signaling. We also observed impairment of hematopoietic stem cell self-renewal and fitness, as evidenced in noncompetitive and competitive bone marrow transplant experiments. CD34+ hematopoietic progenitors and granulocytes from patients with PMF showed decreased levels of ABI1 transcript as well as increased activity of SFKs, STAT3, and NF-κB. In aggregate, our data link the loss of Abi-1 function to hyperactive SFKs/STAT3/NF-κB signaling and suggest that this signaling axis may represent a regulatory module involved in the molecular pathophysiology of PMF.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 2150-2150
    Abstract: The quest for effective, yet less toxic means of treating advanced diseases has led the bone marrow transplantation (BMT) researchers and clinicians searching for the optimal balance between host immune suppression, stable chimerism, effective graft versus tumor response and overall survival. We asked the question “how low can you go?” and approached this from the minimalist view. Born out of murine studies, mini-HLA-identical bone marrow transplantation (BMT) using 100cGy as host conditioning followed by non-mobilized peripheral blood lymphocyte infusion of 1×108 CD3 cells/kg was successful in achieving a complete responses (CR) in 4 of 11 refractory hematologic malignancy patients (Blood100:442, 2002). This study has been extended using HLA-haploidentical donors. We have evaluated CD3+ cell dose escalation with HLA-haploidentical transplantation in patients with refractory malignancies. We have performed 42 HLA-haplo transplants with escalation of the CD3 dose from 1x106–2x108cells/kg using G-CSF primed peripheral blood stem cells, with a conditioning regimen of 100cGy TBI. Twenty patients were treated at the highest level. The CD34 dose infused was always 〉 2x106cell/kg. Ages ranged from16–82 years. We have treated 15 patients with solid tumors and 27 patients with hematologic malignancies consisting of AML (13), NHL (5), Multiple Myeloma (5) CML (3), and HD (1). One treatment related death occurred from grade IV aGVHD. Most patients had a transient febrile engraftment syndrome believed to be immunologically based and represents neither hyperacute nor acute graft versus host disease. There were no objective responses in the patients with solid tumors. One of 5 patients with NHL had a partial response. We observed several encouraging responses in patients with refractory AML. We treated 13 patients with AML and have obtained a complete response in three patients and transient responses in 7 of 12 evaluable patients (survival 〉 2wks after BMT). The median time of transient response was19 days± 5 days and was defined as a persistent loss of peripheral blood blasts and/or more than 50% reduction in marrow blasts as seen on biopsy. In these patients, improvement in blood counts was seen in 3 patients and an improved performance status in 6 of 12. All responses occurred at CD3 levels of 1–2x108 cells/kg. All complete and transient responses occurred in the absence of measurable donor chimerism ( 〈 5%). Donor cells labeled with indium111 analyzed 1,2,3 and six days following BMT in one patient showed persistent signal in the bone marrow and spleen. The kinetics of detectable donor cells in the peripheral blood and marrow show near complete homing to the marrow by 24 hours with a subsequent loss of detectable chimerism 6 days after BMT. Serial bone marrow biopsies performed in several patients show evidence for large tumor reduction and early resumption of normal hematopoiesis. In summary, TBI of 100cGy followed by HLA-haplo transplant is a biologically active therapy for refractory AML. The responses outside of chimersim are intriguing and warrant further investigation. This well tolerated treatment produced minimal toxicity for the majority of patients. Theories on biological effect include an initial graft vs. tumor cell kill, altered host immune response breaking host tumor tolerance, persistent non-detectable microchimerism or a combination of any of the three.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 1677-1677
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1677-1677
    Abstract: Transplanted bone marrow donor cells with tissue specific phenotypes have been found in the brain, liver, heart, skin, lung, kidney, and gut of transplanted humans and mice. Such observations have led to the controversial hypothesis that hematopoietic stem cells (HSC) might be intrinsically plastic, and through transdifferentiation or fusion lead to the repair of damaged tissues throughout the body. Alternately, it is suggested that fusion of macrophages to the recipient cells may explain this phenomenon. We have shown recently that purified HSC are the cells responsible for GFP positive donor-derived muscle fibers in the recipient mice post bone marrow transplantation. However, further studies sorting for macrophage markers Mac-1 and F4/80 also resulted in donor-derived muscle fibers in the host. To address this discrepancy, we investigated subpopulations of Mac-1 and F4/80 positive cells, in the presence or absence of stem cell markers (Sca-1 and C-kit). We demonstrate that only the subpopulations of Mac-1 and F4/80 positive cells harboring stem cell markers, Sca-1 or c-kit, were capable of contributing to the regenerating muscle post transplantation. Furthermore, these same subpopulations demonstrated single cell High Proliferative Potential (HPP) (6–26%) in a 7 factor cytokine cocktail, compared to the Mac-1 or F4/80 cells with no stem cell markers (0%). Additionally, they demonstrated long-term engraftment in all three lineages at 1-year (average chimerism of 55% versus 0% in stem cell marker negative groups). These subpopulations were also evaluated for morphology using Hematoxylin/Eosin (H/E), Wright-Giemsa, and Nonspecific Esterase staining. In the Mac-1 and F4/80 positive groups, those negative for stem cell markers resembled differentiated cells of the myeloid origin (macrophages, granulocytes), while those with positive stem cell markers demonstrated stem cell characteristics. We did not observe any engraftability, donor-derived muscle fibers, or HPP potential for CD14 or cfms positive cells coexpressing stem cell markers, indicating that these markers are more appropriate for identifying macrophages. In conclusion, our studies demonstrate that both Mac-1 and F4/80 surface markers are present on HSC and therefore caution must be taken in the interpretation of data using these macrophage markers. It is reasonable to believe that the use of Mac-1 and/or F4/80 surface markers in a lineage depletion process may result in the loss of a subpopulation of stem cells, and other markers such as CD14 or c-fms may be more appropriate for eliminating differentiated macrophages.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 100, No. 2 ( 2002-07-15), p. 442-450
    Abstract: Allogeneic stem cell transplantation is curative for certain cancers, but the high doses of chemotherapy/radiotherapy lead to toxicity. Here, we treat patients with refractory cancer with 100 cGy total body irradiation (TBI) followed by infusion of nonmobilized pheresed allogeneic peripheral blood cells. Twenty-five patients, with a median age of 47 years, with refractory cancers were enrolled. Eighteen patients received sibling and 7 received unrelated cord blood cells. Donor chimerism was assessed at weeks 1, 2, 3, 4, and 8 after transplantation. Seven patients with solid tumors received a sibling transplant and 6 received a cord blood transplant; none achieved donor chimerism, but 1 treated at the higher dose level of 1 × 108 CD3+ cells/kg had a transient nodal response. Twelve patients with hematologic malignancies were treated; 1 received a cord blood transplant and 11 received sibling donor cells. Nine of these 11 patients achieved donor chimerism, ranging from 5% to 100%. Four patients had sustained complete remission of their cancers, including one patient with transient 5% donor chimerism. The development of chimerism correlated with hematologic malignancy (P  & lt; .001), total previous myelotoxic chemotherapy (P  & lt; .001), T-cell dose (P = .03), and graft-versus-host disease (P = .01). Tumor response correlated with donor chimerism (P = .01). Engraftment was achieved in patients with hematologic malignancies who had been heavily pretreated, suggesting the degree of immunosuppression may be a determinant of engraftment. Low-dose TBI and allogeneic lymphocyte infusion may induce remission in patients with refractory hematologic malignancy.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2002
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 4194-4194
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 4194-4194
    Abstract: Hierarchical models of hematopoiesis suppose an ordered system in which stem cells and progenitors with specific fixed differentiation potentials exist. We show that the potential of marrow stem cells to differentiate changes reversibly with cytokine-induced cell cycle transit. To address whether the cell cycle plays a role in the differentiation of stem cells, we co-cultured murine bone marrow Lin- Sca-1+ cells, at different points in their cycle, with the OP9-DL1 system. OP9-DL1 stromal cell layer has been transduced to allow T-cell differentiation in culture. We first induced cell cycle synchrony by exposing the isolated cells to a cytokine cocktail of TPO, Flt-3 and Stem Cell Factor. The cells were exposed to this primary culture for 0, 6, 24, 32 and 40 hours and were subsequently cultured on an OP9-DL1 stromal cell layer grown in 6-well plates. Cells were co-cultured for 8 days and 21 days, in the presence of IL-7 and Flt-3. Cultured cells were evaluated for CD4, CD8, B220, CD19, NK1.1, and Mac-1 surface markers, using flow cytometry. On Day 8, we found a significant hotspot at 32-hours (early-S phase) for B220+ cells (34.3 %), while Mac-1 positive cells demonstrated a 24-hour hotspot (18.1 %). As expected, terminal T and B-cell differentiation (CD 4, CD8, and CD19) was undetectable at 8 days. Three separate short-term (8 day) experiments have confirmed these data. Cells in culture for 21 days similarly show variation in differentiation outcome. CD4 cells demonstrate a peak at the 40 hour time point (mid-S phase) (69.9%), while CD8 positive cells were significantly increased at the 32 hour time point (34.4%). These data indicate both B and T cells show reversible differentiation fluxes linked to cell cycle. This work supports previous evidence that marrow hematopoiesis at the stem cell level is regulated on a continuum and that stem cells have reversible, cycle-related differentiation capacity.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 5244-5244
    Abstract: Introduction Extracellular vesicles (EVs) form a unique class of messengers for intercellular communication. Depending on their cell of origin, EVs have the ability to induce a phenotypic change in the recipient cell. For example, EVs from explant prostate cancer induce a neoplastic phenotype in normal prostate cell lines. Conversely, EVs from human mesenchymal stem cells (hMSC) reverse the malignant phenotype in prostate and colorectal cancer and mitigate radiation damage to the marrow. Characterization of EVs as "good" or "bad" has the potential to be a very important diagnostic tool in regard to direct therapy and biomarker identification. Currently, there is no way of characterizing the "goodness" of an EV sample. We leveraged advances in the area of machine learning to develop a novel therapeutic tool that can classify the goodness of an EV particle distribution in a serum sample. Methods EVs were harvested from three sources: hMSC primary progenitor cells, Kasumi Acute Myeloid Leukemia (AML) cells lines, and patient samples. Using a standard centrifugation isolation, EVs were isolate, resuspended in 1% DMSO, and frozen. All samples were analyzed using the NanoSight N500. We collected biophysical properties of the EVs such as diameter and diffusion coefficient. The results were summarized in a distribution based on either size or diffusion coefficient. Summary statistics from each distributions were calculated. Summary statistics included mean, mode, and the diameter at which 10%, 50%, and 90% of size or diffusion coefficient is comprised of smaller particles (D10, D50, and D90, respectively). These served as inputs into a softmax Multilayer Perceptron. This neural network classifier was trained on only the hMSC-derived EVs and Kasumi AML-derived EVs, which served to represent a healthy patient and leukemic patient respectively. Results/Conclusion The mean accuracy after 10 fold cross validation was 90.16% ± 9.26%. For each validation run, a Receiver Operating Characteristic (ROC) curve was drawn and the area under the curve (AUC) was calculated. The mean AUC (after 10 fold cross validation) was 95.97% ± 5.38%. We programmed the algorithm, when given a patient sample, to calculate and return similarity to the Kasumi AML-derived EVs. The algorithm was given three patient sample representing three leukemic disease processes: AML, Chronic Myelomonocytic Leukemia (CMML), and Multiple myeloma (MM). The result was a calculate % similarity of 100%, 100%, and 66% for AML, CMML, and MM respectively. These results are promising and have prompted us to begin collecting and test the algorithms on normal, active leukemic, and recovered leukemic patients. We endeavor to evolve our predictive algorithms to include disease and patient specific information, allowing us to adapt our learning models towards clinically relevant endpoints. Disclosures Reagan: Alexion: Honoraria; Pfizer: Research Funding; Takeda Oncology: Research Funding. Olszewski:Spectrum Pharmaceuticals: Consultancy, Research Funding; TG Therapeutics: Research Funding; Genentech: Research Funding.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages