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: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 4, No. 1 ( 2016-12)
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2016
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3613-3613
    Abstract: Background. Aplastic anemia (AA), the prototypical bone marrow (BM) failure syndrome, is caused by immune-mediated destruction of hematopoietic stem/progenitor cells (HSPCs). CD8+ cytotoxic T cells with restricted TCR diversity (oligoclonal T cells) are expanded in AA, leading to production of proinflammatory cytokines, such as IFN-γ, which induce apoptosis of HSPCs. Recent studies have identified a new subset of memory T cells with stem cell-like properties, TSCM, which are the least differentiated cells of all distinct memory populations. Functionally, TSCM possess an enhanced capacity for self-renewal and can generate multiple memory T cell populations, and they likely have an important role in controlling immunity. In autoimmune diseases, there is abnormal CD4+ and CD8+ T cell activation. We evaluated TSCM frequency in AA and its association with severity, treatment response, relapse, and changes after immunosuppressive therapy (IST). Further, to evaluate the TSCM in other autoimmune diseases, we examined CD4+ and CD8+ TSCM frequencies in uveitis, systemic lupus erythematosus (SLE), and sickle cell disease (SCD), as compared with healthy controls. Method. We retrospectively analyzed CD4+ and CD8+ TSCM populations by flow cytometry. PB specimens were collected from 55 AA samples and 41 age-matched healthy donor samples. Among 55 AA samples, 21 samples were analyzed at diagnosis and 34 after IST. For comparison, blood samples were obtained from 34 uveitis patients (27 inactive or 7 active cases), 43 SLE patients who met the American College of rheumatology (ACR) criteria for the disease [19 inactive SLE (SLE disease activity index-2K (SLEDAI-2K) score 〈 3; and 24 active SLE (SLEDAI-2K score 〉 3)], and 5 SCD patients who were receiving frequent transfusions. TSCM was defined as CD3+ CD4 (CD8)+ CD45RO- CD45RA+ CCR7+ CD27+ CD95+ population. Results and Discussion. In healthy controls, TSCM represented a relatively small percentage of circulating CD4+ or CD8+ T cells (median 2.4% CD4+ TSCM and 2.1% CD8+ TSCM, Fig. 1A). A significantly higher CD8+ TSCM frequency was detected in AA patients (4.2% vs. 2.1%, p 〈 0.05) while there was no difference in the CD4+ TSCM frequency (p 〉 0.05), compared to controls (Fig. 1B-C). In AA, high CD8+ TSCM frequency at diagnosis correlated with complete (CR) or partial response (PR) to IST [5.0 % in CR and PR vs 2.8 % in non-responders (NR), p 〈 0.05). In AA patients prior to IST (n=21), CD8+ TSCM frequency was not correlated with age, sex, absolute neutrophil count, platelet count, time from diagnosis to therapy, and serum ferritin levels. CD8+ TSCM were significantly increased in the two AA cohorts (with or without IST), relative to controls (p 〈 0.05, respectively). Higher CD8+ TSCM frequency after IST associated with treatment-failure (3.5 % in responders vs 5.5 % in NR or relapse, p 〈 0.05). Stimulation with anti-CD3/CD28 beads successfully induced cytokine production in CD4+ and CD8+ T cells from AA and healthy controls. Elevated IFN- γ and IL-2 levels were seen in CD4+ and CD8+ TSCM in AA compared to healthy controls. We next compared CD4+ or CD8+ TSCM frequency between each patient group (AA, uveitis, SLE, or SCD) and a healthy control group. Among the four patient groups, the uveitis group alone displayed a reduction in CD4+ TSCM frequency (1.8%) relative to the healthy controls (2.4 %; p 〈 0.05). An elevated CD8+ TSCM frequency was observed in AA (4.2 %), uveitis (3.6 %), and SCD (4.3 %), but not in SLE, compared to controls (2.1%; p 〈 0.05) (Fig. 2A). Positive correlation between CD4+ and CD8+ TSCM frequencies was found in AA, autoimmune uveitis, and SLE (Fig. 2B). Evaluation of PD-1 expression revealed that TSCM were the least exhausted T cell compartment, as compared to other types of memory T cells. Immune therapies appeared to have negative effects on the TSCM population both in uveitis and SLE patient cohorts, as well as in AA. Conclusion. We provide evidence for increased circulating CD8+ TSCM in AA, underscoring the importance of this novel subset in regulation of immune responses and pathogenesis of autoimmunity. Our work described previously unknown potential roles of TSCM in AA, such as cytokine secretion correlated with effector functions. Understanding the CD8+ TSCM population may offer new therapeutic strategies and novel mechanistic insight into the various autoimmune diseases. Figure 1. Figure 1. Figure 2. Figure 2. Disclosures Townsley: Novartis: Research Funding; GSK: Research Funding. Dumitriu:Novartis: Research Funding; GSK: Research Funding. Young:Novartis: Research Funding; GSK: 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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 380-380
    Abstract: Introduction: The technique of selectively depleting alloreacting T cells from the stem cell transplant (SCT) to prevent graft-versus-host disease (GVHD) while sparing T cells with useful reactivity against viruses and leukemia is commonly termed selective depletion (SD). This approach involves the ex vivo stimulation of donor T cells with host antigen presenting cells (APCs) followed by targeted elimination of the activated alloreactive donor T cell repertoire. SD has been achieved through immunotoxin or magnetic bead depletion targeting T cell activation markers such as CD25 or photodepletion with a rhodamine-like dye. These strategies, while having some success in clinical trials, are limited by poor depletion efficacy or off-target depletion. Here, we present an optimized, novel SD method employing ex vivo treatment of alloactivated T cells with pharmacological concentrations of the purine nucleotide adenosine that preserves quiescent lymphocytes, regulatory T cells (Treg), and T cells with antiviral and antileukemic specificities. Methods: Mature dendritic cell (DC) or irradiated peripheral blood mononuclear cell (PBMCs) "recipient" stimulators were co-cultured with HLA mismatched "donor" PBMC for up to 7 days. Pharmacologic grade adenosine was added at various time points in culture at doses ranging from 100uM-2mM. Depletion efficacy was analyzed by proliferation of the depleted product when challenged separately against stimulator, responder, and 3rd party APCs in 5 day CFSE-dilution assay to measure residual alloreactivity, background proliferation, and response to de novo antigens. Activity against viral antigens was assessed through 6 hour peptide challenge followed by flow-cytometry based intracellular cytokine staining. Leukemia-specific cells were generated from the allodepleted product through two weekly stimulations with autologous DCs transduced to express leukemia associated antigens WT1 and PRAME. Results: Adenosine reduced alloreactive T lymphocytes in HLA-mismatched allogeneic co-cultures to background control frequencies or lower (Figure 1). Optimum allodepletion was achieved with addition of 2mM adenosine on days 1, 2, and 5 after establishing co-culture. Allodepleted lymphocyte products comprised 37±3% of initial cell numbers with 90±1% viability (n=18). Similarly, this method reproducibly achieved SD in four haploidentical donor-recipient pairs, reducing CFSE-tracked proliferation in challenge against haplo APCs below the background of proliferation in challenge against autologous APCs. Adenosine depletion equally affected CD4 and CD8 T cell subsets while sparing NK and B cell populations. Analysis of naïve, effector memory (EM), central memory (CM), and terminal effector (EMRA) T cell compartments revealed no significant differences in depletion of a particular subset in the allodepleted T cell products (n=6). Critically, CD4+CD25+FOXP3+ Treg populations (n=6) and activity against viral antigens (cytomegalovirus, Epstein-Barr virus, and adenovirus) were maintained and in some cases enriched (n=6). Significant activity against leukemia associated antigens (PRAME and WT1) was achieved in allodepleted products from 3 donors. Conclusion: This novel SD technique employing adenosine as a pharmacological agent satisfies the requirements of allodepletion with preservation of viral and leukemia-specific immune responses and thus presents a potentially economical method to deplete alloreacting T cells in SCT products for clinical applications. 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: 2015
    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. 124, No. 21 ( 2014-12-06), p. 667-667
    Abstract: Background: Acute myeloid leukemia (AML) is a diagnosis encompassing a diverse group of myeloid malignancies. Heterogeneous genetic etiology, together with the potential for oligoclonality within the individual patient, has made the identification of a single high sensitivity marker of disease burden challenging. PCR based assessment of WT1 expression has been extensively tested as a marker of submicroscopic AML disease burden but is not universally overexpressed in all cases. Our hypothesis was that the addition of other individually suboptimal gene expression assays to the backbone of WT1 PCR based testing in a multi-gene array format would improve prediction of relapse and survival when tested prior to allogeneic stem cell transplantation (allo-SCT). Patients and Methods: We identified pre-transplantation peripheral blood samples from 74 AML patients who had received allo-SCT as part of NHLBI clinical protocols performed at the NIH clinical center in Bethesda, Maryland between 1994 and 2012. All had morphological evaluation of disease status within the two months prior to transplantation and at least twelve months of documented post-SCT clinical outcomes data or until date of death if this occurred before twelve months. Transplantswere predominately myeloablative, T cell depleted, peripheral blood stem cell sibling matched allogeneic transplants with cyclosporine-based graft versus host disease prophylaxis. Peripheral blood samples from fifty healthy adults donors were used as baseline controls. All clinical protocols and permission to use blood for research were conducted in accordance with Declaration of Helsinki principles and were approved by the Institutional Review Board with written informed consent obtained from all subjects. One microgram of total RNA isolated from cryostored peripheral blood samples was reverse-transcribed into cDNA and used for quantitative real time PCR (qRT-PCR) reactions using custom multi-gene PCR array plates (MG-MRD) and SYBR green, with normalization to c-abl. Arrays included primers for detection of WT1, CCNA1, PRTN3, PRAME, MSLN and ABL transcripts. Thresholds for positivity were established based on expression levels seen in healthy normal donors. Statistical analysis was performed using GraphPad Prism (La Jolla, CA, USA). Comparison between survival or relapse curves was performed using the Log-rank (Mantel-Cox) test. Results: In this cohort of 74 patients the overall survival observed at three years after transplant (3yr OS) was calculated to be 36%. Traditional response criteria from examination of bone marrow risk stratified these 74 patients into two groups, the morphological CR group (n=48) with a 3yr OS of 48% and a group of 26 patents with active (refractory/refractory) disease at the time of allo-SCT with a 3yr OS of only 15%. Peripheral blood testing for WT1 overexpression identified 25 patients as positive (3 of whom had no clinically evident disease by morphology on bone marrow examination, ie; minimal residual disease, MRD). Testing using the MG-MRD assay identified an additional 9 patients from the WT1 negative group as also having residual disease (2 had morphologically evident disease, 7 with MRD). The mortality in this group of patients following transplantation was 100%. The 34 (of the total 74) patients with positive MG-MRD prior to transplantation had a three year overall survival of just 9%. This MG-MRD positive group also experienced 22 out of a total of 24 relapses observed in the first year following transplantation. Pre-SCT MG-MRD testing correctly predicted all cases of early relapse (ie: within 100 days after allo-SCT) compared to just 57% (8/14) using WT1 alone. For the 37 patients who entered transplant in a pathological CR, and who did not suffer non-relapse mortality in the first year, the 1yr relapse rate was 24%. Pre-SCT WT1 testing had 100% positive predictive value (PPV) but only 33% sensitivity in identifying relapses in this subgroup, compared to 100% PPV and 89% sensitivity for the MG-MRD array. Conclusion: PCR based pre-SCT MRD testing using a multi-gene panel outperformed WT1 alone, correctly identifying 36% more patients (n=9, all of whom died) as belonging to a group at high-risk for post-SCT relapse and death. Collaborations are now being sought to validate these findings in additional cohorts. 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: 2014
    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. 132, No. Supplement 1 ( 2018-11-29), p. 4595-4595
    Abstract: INTRODUCTION: The potent graft versus leukemia (GVL) effect of allogeneic stem cell transplantation (allo-SCT) is considered as a blueprint for cellular immunotherapy. However, failure of GVL leads to relapse of underlying leukemia, the major cause of death after allo-SCT. In solid tumors, higher tumor mutation burdens are associated with better response to check point inhibitors which implies the importance of neoantigen specific T-cell functions in cancer immunity. In contrast, the frequencies of somatic mutations in acute leukemia are generally low, therefore the role of neoantigens in GVL remains undetermined. Here, we developed a platform to screen for potential neoantigens by performing whole exome sequencing (WES) and RNA sequencing (RNAseq) in matched samples: leukemic blasts at relapse after allo-SCT, recipient T cell controls, and donor cells. METHODS: Leukemic blasts from patients in relapse were enriched by flow sorting from bone marrow aspirate or peripheral blood samples. Recipient T cells were isolated from pre-transplant peripheral blood as germline controls, and donor monocytes or CD34-positive cells were used as hematopoietic-lineage cell controls. WES was performed to 100X coverage, paired with RNAseq 40M reads per sample. Somatic mutations were detected with mutect and mpileup, followed by annotation with SnpEff. High confidence somatic mutations were subjected to pVAC-seq for neoantigen predictions. RESULTS: Six patients with relapsed acute leukemia (AML 5, ALL 1) after allo-SCT and their transplant donors (matched sibling 3, haplo-identical 3) had suitable samples available for analysis. On average, somatic mutations were identified in 297 genes (range 108- 609) by comparing leukemic blasts and germline control T cells. Among those mutations, potential candidates of neoantigen were identified in five out of six subjects. Allele frequencies of mutant genes varied. Most of neoantigens were predicted to bind HLA of both class I (median 5, range 0-15) and class II (median 6, range 0-12). One subject had only HLA class II restricted peptides as predicted neoantigens. Of interest majority of antigens were derived from molecules known to play important roles in leukemia or tumor biology which include ETV6, CCNY, IDH2, PTPN11, SF3B1, and TP53. Evolution analysis of neoantigen showed an emergence of new antigens in relapsed leukemia while a few driver gene mutations persisted after allo-SCT (Figure). CONCLUSION: Our in-silico analysis demonstrated the possibility that somatic mutation in acute leukemia could serve as putative neoantigens applicable for novel immunotherapy after allo-SCT. The binding capacity of mutant peptides to class I and II HLA implies the importance of both CD4 and CD8 contributions to anti-neoantigen immunity. Next, we will search for neoantigen specific T cells exerting an anti-leukemia effect to validate the GVL potential of these mutations in allo-SCT. Figure. Figure. 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: 2018
    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. 132, No. Supplement 1 ( 2018-11-29), p. 812-812
    Abstract: Background: Reactivation of latent viruses post-allogeneic stem cell transplant (SCT) negatively affects outcomes and increases non-relapse mortality. We have reported over 80% early CMV reactivation rate and significant additional costs in recipients of T cell depleted (TCD) SCT. Ex vivo generated multi-virus specific T cells (MVSTs) are effective as a therapy of active infection, but have not been evaluated as prophylaxis early after transplant. In a Phase I study (NIH 14-H-0182) we transferred MVSTs targeting immunodominant viral proteins of CMV, Epstein-Barr virus (EBV), BK and adenovirus (Ad) immediately post SCT as a novel approach to prevent viral reactivation post-SCT. Methods: All subjects were enrolled in HLA-matched T cell depleted (TCD) transplant protocol (NIH 13-H-0144). MVSTs cells were manufactured from SCT sibling donors by stimulation of elutriated lymphocytes for 14 days with seven overlapping peptide libraries (pepmixes) pulsed onto autologous dendritic cells (DCs) in presence of IL-7, IL-15 and IL-2. MVSTs were infused as early as possible (day 0 to +60) post SCT. A Phase I 3+3 dose escalation design was used as follows: Cohort 1 - 1x10e5 total nucleated cells (TNC)/kg, Cohort 2 - 5x10e5 TNC/kg, Cohort 3 - 1x10e6 TNC/kg. Three additional subjects received MVST cells manufactured using pepmix-pulsed mononuclear cells as stimulators (Cohort 3A; 1x10e6TNC/kg) under an amended protocol. The primary safety endpoint at day 42 post infusion was the occurrence of dose limiting toxicity (DLT) defined as Grade IV GVHD or any other severe adverse even (SAE) deemed to be at least "probably" or "definitely" related to the MVST infusion. Patients were followed to day +100 post SCT for secondary outcomes of efficacy, immune reconstitution and GVHD biomarkers (ST2, REG3). CDR3 sequencing (ImmunoSEQ) was performed on selected MVST products and peripheral blood samples post MVST and compared to control SCT recipients. GVHD biomarkers were analyzed pre- and post-treatment. Results: Twelve subjects were treated: nine received MVSTs generated using DCs and three subjects using mononuclear cells (cohort 3A). Median time from SCT to MVST administration was 13 days (range D +2 to +52 post-SCT). Median time to MVST for subjects in Cohort 3A was 3 days (range 2-7). There were no immediate infusion-related adverse events or DLT at the highest dose level. De novo grade II-III aGVHD post-MVST infusion was seen in three subjects (one in Cohort 1 and two in Cohort 3A), but GVHD biomarker elevation predated MVST infusion. CMV reactivation post-MVST occurred in 6 out of 12 subjects (50%) vs. 45 out of 52 patients (50% vs 87%) in a historical group of recipients of T cell depleted SCT. In all cases CMV reactivation occurred in the context of high dose steroids and in two subjects MVSTs were derived from CMV seronegative donors with minimal anti-CMV activity. One subject experienced rapidly rising AdV viremia (asymptomatic) and received an additional infusion of MVSTs. We saw self-limiting low level EBV replication in 8 cases and one BK viremia, but no disease. CDR3 sequencing of MVST products and serial peripheral blood from subjects revealed a robust contribution of ex vivo expanded cells to the overall repertoire, in contrast to untreated controls where the repertoire of (sham) MVST cell products generated from the transplant donors did not significantly overlap with the immune repertoire in peripheral blood of TCD-SCT recipients in the early post SCT period. Only at day +180 some convergence of repertoires became visible indicating spontaneous immune reconstitution (Figure). Detailed CDR3 analysis of cytokine-captured CMV pp65 and IE-1 specific CD4+ and CD8+ T cells was performed in a representative subject clearly demonstrating the detrimental effect of high-dose steroids on frequency of anti-viral T cells, precipitating CMV reactivation. Conclusions: This is the first report demonstrating safety and feasibility of using MVST immediately post-SCT to rapidly reconstitute anti-viral immunity and ameliorate the detrimental impact of the early viral reactivation in SCT recipient. No DLTs were seen and a minimal risk of aGVHD was observed, as there was no correlation with GVHD biomarkers. As revealed by serial CDR3 sequencing, MVSTs robustly contributed to the T cell repertoire. Our results suggest efficacy of this strategy in reducing viral reactivation. A Phase II study is warranted. 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: 2018
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5377-5377
    Abstract: Low CD4+ T cell counts early post-SCT are associated with a greater risk of developing chronic GVHD (cGVHD). However, the factors affecting post graft CD4+ T cell recovery are not known. Furthermore, the CD4 T cell subset correlating best with protection from cGvHD is not defined. We hypothesized that the donor immune repertoire determines CD4+ reconstitution after SCT. We studied 220 donor-recipient pairs undergoing a myeloablative matched-sibling T-cell depleted SCT for a variety of hematological malignancies including AML, ALL, CML, and MDS. Median donor and patient age was 36 years (range, 9 - 70). Median CD34+ cell dose was 5.56 x106/kg (range, 2.3-14.5), with a fixed T cell dose per protocol of 1-5 x104/kg. Ninety-four patients (43%) developed chronic GvHD (32 limited, 64 extensive). Prior to SCT a cryopreserved mononuclear fraction (PBMC) was obtained by leukopheresis on all donors. PBMC from 139 donors were stained with fluorescently conjugated antibodies and acquired on a custom-built LSR Fortessa (BD) flow cytometer. Regulatory T cells (Tregs) and Treg subsets were identified within the CD4+ T cell population using a combination of FOXP3 and Helios. The combination of FOXP3 with Helios has recently been shown to identify natural, thymus-derived Tregs (nTregs), whereas CD4+FOXP3+ T cells lacking Helios represent a pool of induced Tregs (Thornton et al., 2010, J Immunol. 184:3433-3441). Combined expression of CD31 and CD45RA in the CD4+ and nTreg populations was used to identify recent thymic emigrants (RTE). Absolute lymphocyte counts in 220 donors were 2.11/µl (range 0.89 - 4.12). A low absolute lymphocyte count was significantly correlated with a higher cumulative probability for time to incidence of extensive cGVHD (HR 2.38, p = 0.0008). This relationship between lymphocytes and cGVHD was retained in CD4+ T cell subsets: CD3+CD4+ (HR 2.48, p = 0.005); CD3+CD4+CD25+ & lt; 30 cells/μL (HR 2.1, p = 0.02); nTregs & lt; 8 cells/μL (HR 1.97, p = 0.043). RTE of total CD4 did not correlate with RTE nTregs, indicating that nTreg production varies among donors with similar levels of thymic activity. Furthermore recipients of donors with low RTE nTregs relative to overall thymic output had a significantly higher likelihood of developing extensive chronic GvHD (HR 2.78, p=0.0014). In multivariate analysis including patient age, acute GvHD (grade II - IV), graft direction (female donor to male recipient vs. other combinations), disease and status at transplant, and administration of post SCT donor lymphocyte infusion, only thymic output of nTregs (HR = 0.8, p = 0.037) and graft direction (HR = 2.1, p = 0.023) remained significant risk factors for extensive cGvHD. These data show that the thymic output of nTreg varies inherently among donors. Donors with low thymic output of nTreg may recreate a less tolerant immune system in the recipient leading to cGVHD of greater severity. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5377. doi:1538-7445.AM2012-5377
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Society of Hematology ; 2015
    In:  Blood Vol. 126, No. 23 ( 2015-12-03), p. 4294-4294
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 4294-4294
    Abstract: Introduction: Polyomaviruses (PyV) are a family of DNA-viruses responsible for serious morbidity and mortality in immunocompromised individuals including those with HIV or receiving immunosuppressive treatments for transplant or other conditions. BK polyomavirus (BKV) causes hemorrhagic cystitis and nephropathy in stem cell and organ transplant recipients. JC polyomavirus (JCV) causes progressive multifocal encephalopathy (PML) - an often fatal complication in immunosuppressed transplant recipients and in patients treated with natalizumab for multiple sclerosis. Merkel Cell Carcinoma (MCC) is an aggressive skin cancer of elderly or immunocompromised patients caused by Merkel Cell polyomavirus (MCV). There are no established antiviral drugs targeting PyVs and therefore current strategy is limited to reversing immunosuppression where possible. There is strong evidence that PyV can be controlled through T-cell based immunity and adoptive T cell transfer can cure or prevent BKV and JCV viral diseases after stem cell transplants (SCT). Similarly,evidence exists for immune susceptibility of MCC to T cell-mediated immune attack. JCV, BKV, and MCV are closely related and display a high degree of homology in the large T (LT) and VP1 antigens at the protein sequence level. Therefore, generation of a single T cell product containing a polyclonal cross-reactive donor T cell repertoire recognizing antigens from multiple PyV is an attractive strategy with potential clinical application for adoptive immunotherapy to treat patients suffering from HC, nephropathy, PML, MCC, and possibly other PyV-related diseases. Methods: T cells recognizing LT and VP1 antigens from BKV, MCV, and JCV were generated from healthy donor peripheral blood lymphocytes in two weekly stimulations against autologous, mature dendritic cells (DCs) pulsed with relevant peptide libraries or lentivirally transduced to express full length proteins. The cultures were supplemented with cytokines IL-7 and IL-15 to promote memory formation, and IL-2 was added after the second stimulation to promote expansion. These ex vivo generated T cells were tested against autologous DC targets pulsed with the relevant or irrelevant peptide libraries. In addition, to more accurately assess their potential activity in vivo, the T cells were tested against autologous DC targets lentivirally transduced to express naturally-processed epitopes of BKV and MCV antigens and analyzed for their ability to secrete polyfunctional cytokines by flow cytometry. Results: Flow cytometry revealed polyfunctional activity of BKV-primed cells against cognate BK peptide libraries and antigen-expressing transduced DCs as well as JC peptide libraries and MCV-expressing DCs in the production of TNFa, IFNg, and IL2 in both CD4 and CD8 subsets upon stimulation. The lentivirally engineered antigen-expressing DCs proved to be effective stimulators in generating BKV and MCV-recognizing T cells. While a high degree of cross-reactivity was observed among LT antigens of each virus, cross-reactivity among VP1 antigens was lower and largely donor dependent. T cells separately generated against VP1 antigens from Ia and IV serotypes of BKV showed no significant difference in activity between targets, indicating high overlap in TCR recognition of both serotypes. The antigen-primed T cultures could be highly enriched for antigen-specific T cells by CD137 (4-1BB) sorting 24 hours following simulation. T cells generated against JCV peptide libraries and MCV antigen expressing DCs displayed similar polyfunctional cross-reactivity among antigens from the three polyomaviruses, thus establishing the feasibility of generating a single human polyomavirus-targeted T cell product capable of recognizing multiple viruses within the polyomaviradae family. Conclusions: We have demonstrated successful generation of polyclonal cross reactive T cell repertoires targeted against the LT or VP1 antigens from BKV, JCV or MCV. The LT antigen-specific T cells were highly cross reactive among these PyV. There is a high degree of cross reactivity among the VP1-specific T cells from different serotypes of BKV. These universal PyV-specific T cells could have potential clinical application in the treatment and prevention of PyV infections and related diseases. 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: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1956-1956
    Abstract: Background: High rates of cervical HPV disease in women after allogeneic stem cell transplantation (SCT) have been reported, but risk factors related to severe, multifocal, including vaginal and vulvar, HPV disease are not defined. Objective: To determine rates and risk factors for multifocal and severe HPV disease in post-transplant women. Methods: In a prospective long-term study after SCT, gynecologic history and assessment, cervical cytology and HPV testing were obtained with follow-up colposcopy and surgery as indicated for abnormal results. Prior HPV disease, genital graft-versus-host-disease (gGVHD), chronic GVHD (cGVHD) and immunosuppression treatment (IST) 〉 3years were assessed for their association with extent and severity of genital HPV disease. Logistic regressions were used for multivariate analysis. Results: Sixty five long term ( 〉 3 year) SCT survivors were studied prospectively on protocol. Patients received allogeneic transplantation from HLA-identical sibling donors with most undergoing myeloablative total body irradiation (94%) and T lymphocyte-depleted peripheral blood stem cells in 91% Of 65 women, 62 had gynecologic assessment with 8 (13%) having prior history of HPV disease; 16 (26%) had gGVHD. 20 women (32%) had acute GVHD, 46 (74%) had cGVHD; extent was limited in 23(37%) and extensive in 23(37%). 26(42%) had cGVHD requiring IST 〉 3years. Of 21(34%) women with HPV disease after transplant, 12 required surgery and 7 had multifocal disease. Extensive chronic GVHD (but not acute GVHD) was found to significantly impact occurrence (OR=3.5, p=0.038), high-grade severity (OR=7.1, p=0.024) or multifocal HPV disease (OR=14.6, p=0.017). Conclusion: Women who have undergone SCT have an increased risk of genital HPV disease, with highest rates in women with extensive cGVHD. Likely as a result of chronic immune dysregulation and the temporal nature of HPV, these women are at high risk of severe, multifocal disease, which if untreated may progress to genital cancer. Thus, gynecologic assessment as well as possible post-transplant HPV vaccination are critical aspects of care for women with significant GVHD post-transplant. Support: Intramural programs of NHLBI, Clinical Center and NICHD, NCT00106925 Disclosures Stratton: Allergan: 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2013-2013
    Abstract: Introduction: T cell depletion of the stem cell allotransplant (SCT) has the advantage of reducing incidence and severity of GVHD but can be complicated by relapse and infection. An optimum residual T cell dose in T depleted SCT is not known. To optimize T cell depletion we delivered a series of fixed T cell transplant doses with scheduled T cell add back post-SCT in a series of protocols to determine the T cell doses that secured immune reconstitution, and minimized relapse and infection. Here we retrospectively examined large series of patients (pts) transplanted in a single institution where T cell dose at SCT and add back were quantitated. Unique to these protocols was the precise measurement of CD3 dose/kg at transplant for every patient which enabled us to relate transplant T cell doses to outcome. Patients and methods: 205 pts (106 males, 99 females) underwent HLA identical sibling allogeneic T cell depleted SCT between 1994 and 2014. Diagnosis at SCT included AML (48%), MDS (17%), ALL (27%), CLL (5%), MM (3%). Disease risk at SCT was classified as high (57%) or standard (43%). The median age at SCT was 37 (range: 10-75) yrs. TBI based myeloablative conditioning was used. The graft source was marrow in 20 pts and peripheral blood in 195 pts. GVHD prophylaxis was low dose cyclosporine. Different T cell depleted methods were used consecutively: elutriation, Isolex ®, Cellpro ® CD34, and Miltenyl © CD34 selection. A defined T cell dose was allocated at SCT by protocol ranging from 2 - 50 × 104 CD3+ cells/kg. Various schedules were used to add back T lymphocytes between day 30 to 90 with doses ranging 5 - 60 ×106 CD3+ cells/kg by protocol and no T cell add-back was given in 28 pts in recent protocols. Overall survival (OS) was estimated by the Kaplan-Meier method, and cumulative incidence of relapse and nonrelapse mortality (NRM) was estimated by Gray's method to account for competing risks. Cox proportional hazard regression models were used to assess the association of factors at baseline, day 100 and GVHD with the post-SCT outcomes. Results: At a median follow-up of 8.6 yrs (range: 0.7- 19.8) for surviving pts, 112 pts died (52 from NRM) and 68 pts relapsed. OS was 47%, 43% and 41%, NRM was 24%, 27% and 27%, relapse was 32%, 34% and 38% at 5 yr, 10 yr and 15 yr post SCT, respectively. Grade II-IV and III-IV acute GVHD were 41% and 13%, and chronic GVHD was 42% (25% limited, 17% extensive). In the multivariate models of baseline risk factors that adjusted for age at SCT and disease risk, T-cell doses at SCT did not affect OS, NRM or relapse. A higher dose of CD34+ cells at SCT was significantly associated with better OS and lower NRM. Disease risk was an independent predictor, with high-risk pts having more relapse and worse OS, compared to pts with standard risk. A landmark analysis of 156 pts surviving and relapse-free beyond day 100 was carried out to examine the effects of add back T cell schedules by day 100 and aGVHD. The total T-cell dose at add back by day 100 and different add-back T-cell schedules from day 30-90 had no impact on any outcome, controlling for T-cell dose at SCT. In the models controlling for age, risk, CD34+ and T-cell dose at SCT, pts with grade III-IV aGVHD by day 100 had an increased risk for overall mortality and NRM beyond day 100 (HR= 3 and 3.6, both P 〈 0.001), but did not affect relapse. An analysis of pts surviving and relapse-free beyond 1 yr showed pts with extensive cGVHD had higher NRM rate (39%) and lower relapse rate (0%) after 1 yr post-HSCT compared to pts with no cGVHD ( NRM, 9%, P = 0.020; relapse 24%, P=0.026). Conclusion: These findings indicate that for myeloablative matched sibling SCT there is no ideal prescription of T cell dose at transplant within a range of 104 - 105 or for scheduled add back of lymphocytes within a range of T depletion 5 - 60 × 106. Instead, factors other than T cell dose either at transplant or when add-back was delayed determined GVHD incidence, relapse, and survival. These findings set a limit on the efficacy of any T cell depletion procedure to optimize transplant outcome. 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: 2015
    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...