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
Filter
Material
Language
Subjects(RVK)
  • 1
    In: The Lancet Oncology, Elsevier BV, Vol. 11, No. 5 ( 2010-05), p. 421-428
    Type of Medium: Online Resource
    ISSN: 1470-2045
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2049730-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 314-314
    Abstract: Abstract 314 We have previously reported mobilization data of hematopoietic stem and progenitor cells (HSPCs) in donors treated with IV plerixafor in a phase I/II open label trial (Rettig et al. Blood 2010 116: a2261). Here we report the final outcomes of donors and recipients of the phase II portion of the trial evaluating the use of 0.32 mg/kg plerixafor IV as a single agent for stem cell mobilization. We hypothesized, based on preclinical studies in the mouse, that IV plerixafor would have more rapid and prolonged mobilization and increase stem cell yields so that failure to collect ≥2×106 CD34+ /Kg recipient weight in one apheresis would be reduced from 33% (seen with 240 ug SC plerixafor) to 11%. We also hypothesized based on our prior observations of increased plasmacytoid dendritic cells (pDCs) in plerixafor mobilized products (Rettig et al. Blood 2009 114: a32), that peripheral blood stem cell products would be enriched in pDCs which may lead to improved immunity in the recipient after transplant and reduced incidence of CMV. Methods. In this phase II open label trial donors were mobilized with 0.32 mg/Kg IV plerixafor (over 30 minutes), followed by a 20 L apheresis starting 4 hours after injection. The primary outcome was failure to collect ≥2×106 CD34+/Kg actual recipient weight on day 1. Secondary outcomes included donor toxicity, successful collection in 2 aphereses, pharmacodynamics of mobilization, engraftment, graft-versus-host disease (GvHD), treatment related mortality (TRM), incidence of CMV, relapse free survival, and overall survival. The study was powered to detect a difference in failure rate from 33% to 11%. Sibling donors were 18 – 70 years of age and 6/6 HLA matched with recipients. Recipients were 18 – 67 years of age with a hematologic malignancy (AML n=14, NHL n=9, CLL n=3, MDS n=3, ALL n=2, CML n=1, and HD n=1). 33 recipients were transplanted (5 reduced-intensity and 28 myeloablative conditioning). A total of 27 donor recipient pairs were initially enrolled and 7 additional pairs of subjects were enrolled to replace 6 donors who failed to collect goal apheresis volume ( 〉 17.5L) due to technical problems related to initial apheresis collections. Median follow up of recipients is 248 days (range 37–695 days). Results. The primary outcome of failure to reach ≥2×10e6 CD34+/Kg recipient weight was analyzed on a per protocol basis. CD34 HSPC mobilization kinetics were previously reported for the phase I cohorts (ASH 2010). 29 donors were evaluable for the primary outcome. 34% (10/29) failed to reach the day 1 collection goal of ≥2×106 CD34/Kg recipient weight. 10% (3/29) failed to reach goal after 2 apheresis procedures. Toxicity in donors was mild with the most common reported side effects being grade 1 GI toxicity of abdominal bloating (30%) and grade 1 bradycardia (30%). All recipients engrafted and there was no graft failure. The median time to neutrophil engraftment (ANC 〉 500K/uL × 2 days) was 14 days, with 31/33 engrafting by day 21. The median time to platelet engraftment (Platelets 〉 50K/uL × 2 days) was 25 days, with 23/33 engrafting by day 30. The incidence of grade II-IV acute GvHD was 7/33 (21%). The incidence of grade III-IV acute GvHD was 4/33 (12%). The cumulative incidence of chronic GvHD in those living beyond 100 days was 7/25 (28%) and 4/25 (16%) had severe disease. The incidence of CMV viermia 〉 10,000 copies in at risk recipients (defined as serology positive in donor or recipient) was 3/20 (15%). The incidence of CMV disease was 1/20 (5%). Median overall survival was 258 days, with 43% one-year survival. Median relapse free survival was 238 days. The day 100 TRM was 1/33 (3%). Summary. IV administration of plerixafor was well tolerated by donors. However, based on our prior experience with subcutaneous dosing with 0.24mg/kg (Devine et al Blood, 2008) there was no improvement in the percent of normal donors who successfully collected ≥2×106 CD34+/Kg after a single IV infusion of 0.32 mg/Kg plerixafor (67% vs. 66% respectively). Failure to reach collection goal in one apheresis procedure remains high with single agent plerixafor regardless of route of administration. Incidence of CMV viremia was low and could possibly be related to increased plasmacytoid dendritic cells in the product. The incidence of aGvHD was lower than historical controls for G-CSF mobilized peripheral blood transplantation and deserves further evaluation in future prospective trials involving CXCR4 antagonists such as plerixafor. Disclosures: Schroeder: Genzyme: Research Funding. Rettig:Genzyme: Honoraria. Uy:Genzyme: Consultancy, Speakers Bureau. DiPersio:Genzyme: Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    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. 114, No. 22 ( 2009-11-20), p. 3693-3693
    Abstract: Abstract 3693 Poster Board III-629 Background Relapsed or refractory (rel/ref) classical Hodgkin lymphoma (cHL) remains a clinical challenge with limited effective treatments after high dose therapy with autologous stem cell transplantation (ASCT). Lenalidomide (Revlimid®) is an approved anti-neoplastic therapy for multiple myeloma and myelodysplastic syndrome with del(5q). In ongoing investigations it has a manageable toxicity profile and promising clinical activity in a number of B cell malignancies. Multiple potential mechanisms of action for lenalidomide have been identified including direct effects on tumor cells, alteration of the tumor microenvironment, anti-angiogenic properties, modification of cytokine profiles, and immune (T and NK) cell modulation. Since these effects have the potential to alter factors involved in cHL pathogenesis, we hypothesized that lenalidomide may have clinical activity in cHL. Methods N=38 rel/ref cHL patients (pts) who previously underwent (or were not candidates for) ASCT were enrolled from October 2007 to May 2009 in a prospective, multi-center, Phase II study of single agent lenalidomide. The treatment regimen was 25 mg/day of lenalidomide on days 1-21 of a 28 day cycle. Treatment continued until progressive disease or an unacceptable adverse event at the lowest allowed dose (5 mg) of lenalidomide. The primary endpoint was overall response rate [complete remission (CR) + partial remission (PR)] as defined by the 2007 revised IWG criteria, and cytostatic response defined as CR + PR + stable disease (SD) 〉 6 months. Results Median age at treatment was 34 (range 25-63) years with 23 females. Median number of prior therapies was 4 (range 2-9). 33 pts had received a prior stem cell transplant (29 ASCT, 1 syngeneic, 3 both ASCT and allogeneic). 22 pts (58%) had not responded to their last prior therapy, and the median time from last prior therapy to enrollment on this study was 3 (range 1-66) months. Median time of follow-up from enrollment on this study was 12 (range 3-22) months. Of the 38 enrolled pts, 3 were removed from the study prior to receiving 2 cycles of lenalidomide due to: elevated bilirubin and transaminases (n=1), desquamating rash (n=1), and rapid disease progression during cycle 1 (n=1). Median number of lenalidomide cycles administered per patient was 4 (range 2-22). For the 35 evaluable pts, we observed 1 CR, 5 PR, and 6 SD 〉 6 months for an overall response rate of 17% (6/35) and an overall cytostatic response rate of 34% (12/35). Median duration of CR/PR was 4.5 (range 2-10) months. The CR was observed after 2 cycles, and PRs were observed after 2 (n=2), 4 (n=1), 6 (n=1), and 18 (n=1) cycles of lenalidomide. Currently, lenalidomide treatment continues in 5 pts with SD (n=4, at ≥3, ≥7, ≥9, ≥15 cycles) or PR (n=1, ≥22 cycles). Of note, 8/12 (67%) lenalidomide responders had failed to respond to their last prior therapy before enrollment. In general, the treatment was well tolerated, and the most common grade 3-4 adverse events were neutropenia (40%), anemia (24%), leukopenia (21%), and thrombocytopenia (16%). The lenalidomide dose was reduced in 6 pts for cytopenias (n=2), fatigue (n=2), ALT/AST (n=1), and neuropathy (n=1), and discontinued in 4 pts for rash (n=2), elevated bilirubin/transaminases (n=1), and cytopenias (n=1). For pts with dose reduction, the lenalidomide doses ranged from 20 mg to 5 mg. No tumor lysis syndrome or documented tumor flare reactions were observed. Conclusions Single agent lenalidomide therapy has preliminary evidence of activity in pts with rel/ref cHL, including pts progressing after ASCT. Continuous daily dosing of lenalidomide is currently being evaluated in an effort to maximize single agent efficacy, and exploration of lenalidomide combinations with other active agents in rel/ref cHL is warranted. Disclosures: Fehniger: Celgene: research funding solely to support this clinical trial. Off Label Use: The use of Lenalidomide in relapsed or refractory Hodgkin lymphoma. Bartlett:Celgene: research funding solely to support this clinical trial.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    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: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 2146-2146
    Abstract: Multiple myeloma is the second most common hematologic malignancy in the US. The current standard of care for transplant eligible patients is therapy with high-dose melphalan (HDM) followed by autologous stem cell transplant (ASCT). Although ASCT improves progression-free (PFS) and overall survival (OS), it is not curative and virtually all patients will relapse. Attempts to improve upon HDM by adding other agents to transplant protocols have largely resulted in unacceptable increases in toxicity. Preclinical studies performed by our group suggest that granulocyte-stimulating factor (G-CSF) disrupts the bone marrow microenvironment, resulting in a striking loss of plasmablasts, plasma cells, and decreased expression of chemokine/cytokines contributing to plasma cell maintenance. Tbo-filgrastim (Granix, Teva Pharmaceuticals) is a recombinant methionyl human granulocyte colony-stimulating factor. We hypothesized that tbo-filgrastim treatment may provide a potent and well-tolerated method to disrupt the 'myeloma cell niche', rendering patients more sensitive to HDM. Methods: Here, we report results from an open label, single center, phase II randomized study to test the efficacy and safety of tbo-filgrastim plus HDM (tbo-filgrastim arm) versus HDM alone (SOC arm) prior to ASCT (NCT02112045). Patients were randomized 1:1 to tbo-filgrastim on Day -7 through Day -2 (480 or 960 mcg/day, based on weight) and melphalan on Day -2 prior to ASCT (140 or 200 mg/m2, based on age) or melphalan alone. The primary objective was to compare CR rate at day 100. Secondary objectives included comparison of the toxicity, overall response rate, PFS, OS, and rate of neutrophil and platelet engraftment between the two arms. Eligible patients were ≥18 years with symptomatic multiple myeloma enrolled within 12 months of receiving at least 2 cycles of any systemic therapy, were undergoing their first ASCT, and had an adequate ASCT collection product (at least 2 million CD34+ cells/kg). Target enrollment was 176 patients, with an interim analysis of efficacy and futility planned after 88 patients reached Day 100 post-ASCT. Early stopping rules for unacceptable toxicity were in place. Responses were evaluated by IMWG criteria. Results: Ninety patients were enrolled (median age 59.5, range 33 to 77) and 89 were evaluable for response. The early stopping rules for toxicity were not met. The planned interim analysis showed that the proportion of patients in CR at Day 100 was similar between the arms and the study was halted for futility (39.5% on the tbo-filgrastim arm vs. 37.8% on SOC arm). The overall response rate (CR + VGPR + PR) between the tbo-filgrastim and the SOC arm was 95% vs 93%, respectively. At the interim analysis, with median follow-up time for the study of 21.7 months, (range 8.8 to 25.8), the median PFS and OS had not been reached for either arm. There was no difference in PFS between the tbo-filgrastim and the SOC arm (84% vs 80%, respectively, p=0.60). There was no difference in OS between the tbo-filgrastim and the SOC arm (90.9% vs 95.6%, respectively, p=0.43). All patients in the study achieved neutrophil (ANC 〉 0.5 K/cumm) and platelet ( 〉 20 K/cumm) engraftment. The median time to neutrophil engraftment for the tbo-filgrastim arm was 5 days (range, 3-9) vs 4 days (range, 3-7) in the SOC arm, p 〈 0.001. There was no difference in the median time to platelet engraftment between the arms (11 days in the tbo-filgrastim arm, range 2-23, and 10 days in the SOC arm, range 1-24, p=0.67). Adverse events for both arms were typical of those observed in the ASCT population. Conclusions: The administration of tbo-filgrastim in the setting of HDM prior to ASCT is feasible, without excess toxicity or loss of engraftment. There was no difference in Day +100 CR or ORR rates, PFS, or OS in patients treated with tbo-filgrastim plus HDM versus HDM alone with a median follow-up of 21.7 months. The lack of efficacy may be secondary to the high pre-ASCT response rates seen with modern agents. Disclosures Jacoby: Celgene: Speakers Bureau; NovoNordisk: Consultancy. Schroeder:Amgen Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees. Vij:Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Jazz Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees; Jansson: Honoraria, Membership on an entity's Board of Directors or advisory committees; Karyopharma: 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, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 109, No. 8 ( 2007-04-15), p. 3424-3431
    Abstract: Inhibition of osteoclast (OC) activity has been associated with decreased tumor growth in bone in animal models. Increased recognition of factors that promote osteoclastic bone resorption in cancer patients led us to investigate whether increased OC activation could enhance tumor growth in bone. Granulocyte colony-stimulating factor (G-CSF) is used to treat chemotherapy-induced neutropenia, but is also associated with increased markers of OC activity and decreased bone mineral density (BMD). We used G-CSF as a tool to investigate the impact of increased OC activity on tumor growth in 2 murine osteolytic tumor models. An 8-day course of G-CSF alone (without chemotherapy) significantly decreased BMD and increased OC perimeter along bone in mice. Mice administered G-CSF alone demonstrated significantly increased tumor growth in bone as quantitated by in vivo bioluminescence imaging and histologic bone marrow tumor analysis. Short-term administration of AMD3100, a CXCR4 inhibitor that mobilizes neutrophils with little effect on bone resorption, did not lead to increased tumor burden. However, OC-defective osteoprotegerin transgenic (OPGTg) mice and bisphosphonate-treated mice were resistant to the effects of G-CSF administration upon bone tumor growth. These data demonstrate a G-CSF–induced stimulation of tumor growth in bone that is OC dependent.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3419-3419
    Abstract: Background:Allogeneic hematopoietic cell transplantation is a cornerstone of therapy for hematologic malignancies and often a patient's only curative intent treatment. Following development of post-transplant cyclophosphamide (PTCy) regimens, the use of haploidentical hematopoietic cell transplantation (haplo-HCT) has expanded. While overall outcomes for haploidentical transplantation appear to be excellent, its novel approach brings toxicities that are particular to its biological and clinical milieu. We recently described occurrence of severe cytokine release syndrome (CRS) after haplo-HCT. We further reported that severe CRS was associated with poor clinical outcomes, including transplant related mortality (TRM), overall survival (OS), and neutrophil engraftment (Abboud et al, BBMT, 2016). However, the factors predicting the occurrence of and long-term outcomes of patients who develop severe CRS after haplo-HCT is currently not known. Objective: To describe our clinical experience with CRS in an expanded cohort of haplo-HCT patients, its implication on clinical outcomes and elucidation of possible risk factors for the development of severe CRS. Patients and Methods: We performed a retrospective review of patients who had undergone peripheral blood T-Cell replete haplo-HCT with PTCy from July 2009 through March 2016 at our institution. A total of 137 patients were identified, 51% (74) were male, with a median age at transplant of 52 (19-73), a total of 40% (57) had active disease at the time of transplant. The most common diagnosis was AML (93 pts), followed by ALL (16 pts) and MDS (15 pts). Thirty-one percent (44 pts) had undergone prior transplant. In grading CRS, we used our approach modified from by Lee et al (Blood, 2014). Twenty-two patient, donor and disease characteristics were examined to identify predictors for the development of severe CRS. Results:One hundred and twenty-four (90%) of patients met criteria for CRS, and 26 (19%) suffered from severe (grade 3-4) CRS. Virtually all patients (99%) with CRS suffered from fevers. Patients with severe CRS had a significant delay in neutrophil (p 〈 0.0001) and platelet (p 〈 0.0001) engraftment compared to the patients who developed mild or no CRS (Figure 1A and 1B). Severe CRS was also associated with a high early transplant related mortality; the rate of death before post-transplant day 28 was 6.9 times higher for patients with grade 3-4 CRS compared with those with mild CRS (p 〈 0.0001, Figure 1C). Consistent with these findings, the development of severe CRS was associated with extremely poor survival. Median survival was 3 months for grade 3-4 CRS, 15 months for grade 1-2 CRS, and 13 months for no CRS. One-year OS was 4% for grade 3-4 CRS, 55% for grade 1-2 CRS, and 50% for no CRS (Figure 1D). There was no difference in the cumulative incidence of relapse, acute graft versus host disease, and chronic graft versus host disease (data not shown). A total of nine patients received Anti-IL-6 Therapy with tociluzimab (4 mg/kg of actual body weight), 4 of which suffered from severe CRS. In terms of predictive factors, the development of severe CRS was associated with disease risk index (p=0.037), HCT-CI score (p=0.005) and presence of a previous transplant (p=0.026) by univariate analysis. Risk and severity of CRS did not differ by age, ABO mismatch, age, CMV status of donor, donor sex, T-cell or CD34 cell dose. There was no difference in rates of CRS among patients in remission or with active disease at the time of transplant. Conclusions: Severe CRS after peripheral blood haplo-HCT is associated with high early TRM, poor OS and delayed neutrophil and platelet engraftment. Furthermore, patients with high DRI, high HCT-CI and prior HCT are at a higher risk for the development of severe CRS after haplo-HCT. We have previously shown the safety and potential efficacy of using anti-IL-6 therapy in these patients. Our current results suggest potential benefit to targeting this pathway prophylactically in patients at high risk for the development of severe CRS. Table Patient Characteristics Table. Patient Characteristics Figure CRS impacts neutrophil (A) and platelet (B) engraftment and is associated with high TRM (C) and poor OS (D). Figure. CRS impacts neutrophil (A) and platelet (B) engraftment and is associated with high TRM (C) and poor OS (D). Disclosures DiPersio: Incyte Corporation: Research Funding. Abboud:Gerson and Lehman Group: Consultancy; Merck: Research Funding; Teva: Research Funding, Speakers Bureau; Novartis: Research Funding; Pfizer: Research Funding; Seattle Genetics: Research Funding; Alexion: Honoraria; Baxalta: Honoraria; Pharmacyclics: Honoraria; Takeda: Honoraria; Cardinal: Honoraria. Fehniger:Affimed: Consultancy; Celgene: Research Funding; Fortress Biotech: Consultancy.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4698-4698
    Abstract: Background: Hematopoietic cell transplantation (HCT) is the only curative therapy for many patients with hematologic malignancies. In matched related donor (MRD) and matched unrelated donor (MUD) transplants, absolute lymphocyte count (ALC) recovery has been used as a surrogate marker for immune reconstitution, and a faster rate of ALC recovery is associated with improved overall survival (OS), relapse-free survival (RFS), and transplant-related mortality (TRM). Higher ALC counts have also been associated with higher rates of graft-versus-host disease (GVHD). For patients that do not have an HLA-matched donor available, haploidentical transplant (haplo-HCT) is now a viable option. Less is known about the impact of ALC recovery on outcome in the setting of haploidentical transplant with post-transplant cyclophosphamide (PTCy). Methods: In this study we retrospectively evaluated all patients who underwent haplo-HCT with post-transplant cyclophosphamide (PTCy) at our institution between June 2009 and January 2016. ALC and total white blood cell (WBC) count at days 30, 100, 180, and 365 post-transplant were collected along with demographics, treatment details, and outcome measures (OS, RFS, NRM, acute GVHD, and severe chronic GHVD). Since ALC is a component of total WBC used to assess for immune reconstitution, we performed additional analysis to assess if a high ALC relative to total WBC Ð or conversely, a low ALC relative to total WBC Ð had an effect on outcomes. At each time point, patients were divided into subgroups based on percentile of total WBC. Individual survival curves were generated for each subgroup for comparison. Cox proportional hazard models were used to measure the association between ALC and each outcome. The proportional hazards assumptions and functional assessment of ALC values were carried out using the K-S test and simulated martingale residuals. ALC was analyzed on a log scale to improve the fit of these models. Results: Of 141 evaluable patients, 128 survived to day 30 and were included in analysis. Clinical characteristics are summarized in table 1. At day 30, 100, 180 and 365 there were128, 99, 78 and 48 patients were alive, respectively. A high ALC at day 180 following transplant was associated with improved OS (mean ALC 915, HR 0.61, 95% CI 0.39-0.953; p = 0.030) and RFS (mean ALC 912, HR 0.593, 95% CI 0.422-0.834, p = 0.003). Furthermore, a higher ALC at day 100 was found to be associated with improved TRM (mean ALC 528, HR 0.54, 95% CI 0.35-0.84, p = 0.006). ALC was not associated with acute GVHD at any of the four time points. Severe chronic GVHD only occurred in 4 patients and therefore was not modeled. While a high ALC remained a significant predictor of improved OS, RFS, and TRM, there was no statistically significant difference between subgroups adjusted for percentile of WBC at each time point. Conclusions: ALC recovery is associated with improved OS, RFS, and TRM. Our results suggest that ALC is a useful prognostic marker for patients undergoing haploidentical HCT with post-transplant cyclophosphamide. Further studies examining immune reconstitution in this setting are planned, including the recovery of various T cell and NK cell subsets and their potential impact on outcomes after haplo-HCT using PTCy. Table 1 Clinical and demographic characteristics of patients undergoing haplo-HCT with PTCy. Table 1. Clinical and demographic characteristics of patients undergoing haplo-HCT with PTCy. Figure 1 Months to outcome or last follow-up by ALC, adjusted for percentile of total WBC. Figure 1. Months to outcome or last follow-up by ALC, adjusted for percentile of total WBC. 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 ...
  • 8
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2011
    In:  Annals of Surgical Oncology Vol. 18, No. 4 ( 2011-4), p. 946-951
    In: Annals of Surgical Oncology, Springer Science and Business Media LLC, Vol. 18, No. 4 ( 2011-4), p. 946-951
    Type of Medium: Online Resource
    ISSN: 1068-9265 , 1534-4681
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 2074021-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 29, No. 7 ( 2011-03-01), p. 875-883
    Abstract: Downstaging (DS) of rectal cancers is achieved in approximately 45% of patients with neoadjuvant fluorouracil (FU) -based chemoradiotherapy (CRT). Polymorphisms in the thymidylate synthase gene (TYMS) had previously defined two risk groups associated with disparate tumor DS rates (60% v 22%). We conducted a prospective single-institution phase II study using TYMS genotyping to direct neoadjuvant CRT for patients with rectal cancer. Patients and Methods Patients with T3/T4, N0-2, M0-1 rectal adenocarcinoma were evaluated for germline TYMS genotyping. Patients with TYMS *2/*2, *2/*3, or *2/*4 (good risk) were treated with standard chemoradiotherapy using infusional FU at 225 mg/m 2 /d. Patients with TYMS *3/*3 or *3/*4 (poor risk) were treated with FU/RT plus weekly intravenous irinotecan at 50 mg/m 2 . The primary end point was pathologic DS. Secondary end points included complete tumor response (ypT0), toxicity, recurrence rates, and overall survival. Results Overall, 135 patients were enrolled, of whom 27.4% (37 of 135) were considered poor risk. The prespecified statistical goals were achieved, with DS and ypT0 rates reaching 64.4% and 20% for good-risk and 64.5% and 42% for poor-risk patients, respectively. Conclusion To our knowledge, this is the first study to prospectively use TYMS genotyping to direct neoadjuvant CRT in patients with rectal cancer. High rates of DS and ypT0 were achieved among both risk groups when personalized treatment was based on TYMS genotype. These results are encouraging, and further evaluation of this genotype-based strategy using a randomized study design for locally advanced rectal cancer is warranted.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2011
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2015
    In:  Journal of Clinical Oncology Vol. 33, No. 15_suppl ( 2015-05-20), p. 6043-6043
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 33, No. 15_suppl ( 2015-05-20), p. 6043-6043
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2015
    detail.hit.zdb_id: 2005181-5
    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...