52 results on '"Yinyan Xu"'
Search Results
2. HIV-Specific T Cell Responses Are Highly Stable on Antiretroviral Therapy
- Author
-
Yinyan Xu, Ilana M. Trumble, Joanna A. Warren, Genevieve Clutton, Maria Abad-Fernandez, Jennifer Kirchnerr, Adaora A. Adimora, Steven G. Deeks, David M. Margolis, JoAnn D. Kuruc, Cynthia L. Gay, Nancie M. Archin, Katie R. Mollan, Michael Hudgens, and Nilu Goonetilleke
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,medicine.medical_treatment ,T cell ,Human immunodeficiency virus (HIV) ,Viremia ,medicine.disease_cause ,Peripheral blood mononuclear cell ,Article ,Vaccine Related ,03 medical and health sciences ,0302 clinical medicine ,Clinical Research ,Genetics ,medicine ,lcsh:QH573-671 ,Molecular Biology ,lcsh:Cytology ,business.industry ,ELISPOT ,virus diseases ,Evaluation of treatments and therapeutic interventions ,HIV ,Immunotherapy ,CD8 ,medicine.disease ,3. Good health ,Clinical trial ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,Infectious Diseases ,Good Health and Well Being ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Immunology ,Molecular Medicine ,HIV/AIDS ,Immunization ,immunotherapy ,business ,Infection - Abstract
HIV infection induces a robust Tcell response that is sustained by high viremia, but falls following the onset of antiretroviral therapy (ART). Relatively little has been reported on the subsequent stability of the HIV-specific Tcell response in individuals on durable therapy. Such data are critical for powering clinical trials testing Tcell-based immunotherapies. In a cross-sectional study, HIV-specific Tcell responses were detectable by exvivo interferon (IFN)-γ ELISpot (average ∼1,100 spot-forming units [SFUs]/106 peripheral blood mononuclear cells) in persons living with HIV (PLWH; n= 34), despite median durableART suppression of 5.0 years. No substantial association was detected between the summed HIV-specific Tcell response and the size of the replication-competent HIV reservoir. Tcell responses were next measured in participants sampled weekly, monthly, or yearly. HIV-specific Tcell responses were highly stable over the time periods examined; within-individual variation ranged from 16% coefficient of variation (CV) for weekly to 27% CV for yearly sampling. These data were used to generate power calculations for future immunotherapy studies. The stability of the HIV-specific Tcell response in suppressed PLWH will enable powered studies of small sizes (e.g., n= 6-12), facilitating rapid and iterative testing for Tcell-based immunotherapies against HIV.
- Published
- 2019
3. Reliable Estimation of CD8 T Cell Inhibition of
- Author
-
Yinyan, Xu, Ann Marie, Weideman, Maria, Abad-Fernandez, Katie R, Mollan, Sallay, Kallon, Shahryar, Samir, Joanna A, Warren, Genevieve, Clutton, Nadia, Roan, Adaora A, Adimora, Nancie, Archin, JoAnn, Kuruc, Cindy, Gay, Michael G, Hudgens, and Nilu, Goonetilleke
- Subjects
CD4-Positive T-Lymphocytes ,VIA ,JRCSF ,Host Microbial Interactions ,Immunology ,HIV Core Protein p24 ,virus diseases ,HIV ,CD8 ,CD8-Positive T-Lymphocytes ,Virus Replication ,p24 ,Antiviral Agents ,Coculture Techniques ,CD4 ,Cross-Sectional Studies ,Treatment Outcome ,T-cell ,Case-Control Studies ,HIV Seropositivity ,HIV-1 ,Methods ,Humans ,ROC ,Cells, Cultured - Abstract
The HIV-1 viral inhibition assay (VIA) measures CD8 T cell-mediated inhibition of HIV replication in CD4 T cells and is increasingly used for clinical testing of HIV vaccines and immunotherapies. The VIA has multiple sources of variability arising from in vitro HIV infection and co-culture of two T cell populations. Here, we describe multiple modifications to a 7-day VIA protocol, the most impactful being the introduction of independent replicate cultures for both HIV infected-CD4 (HIV-CD4) and HIV-CD4:CD8 T cell cultures. Virus inhibition was quantified using a ratio of weighted averages of p24+ cells in replicate cultures and the corresponding 95% confidence interval. An Excel template is provided to facilitate calculations. Virus inhibition was higher in people living with HIV suppressed on antiretroviral therapy (n=14, mean: 40.0%, median: 43.8%, range: 8.2 to 73.3%; p < 0.0001, two-tailed, exact Mann-Whitney test) compared to HIV-seronegative donors (n = 21, mean: -13.7%, median: -14.4%, range: -49.9 to 20.9%) and was stable over time (n = 6, mean %COV 9.4%, range 0.9 to 17.3%). Cross-sectional data were used to define 8% inhibition as the threshold to confidently detect specific CD8 T cell activity and determine the minimum number of culture replicates and p24+ cells needed to have 90% statistical power to detect this threshold. Last, we note that, in HIV seronegative donors, the addition of CD8 T cells to HIV infected CD4 T cells consistently increased HIV replication, though the level of increase varied markedly between donors. This co-culture effect may contribute to the weak correlations observed between CD8 T cell VIA and other measures of HIV-specific CD8 T cell function.
- Published
- 2021
4. Identification of CPAF as the immunoprevalent antigen of Chlamydia trachomatis
- Author
-
Yanli Li, Joanna Warren, Taylor Poston, Fiona Shaw, Shayla Conrad, Yinyan Xu, Xiaojing Zheng, Catherine M O’Connell, Sharon L Hillier, Harold C Wiesenfeld, Toni Darville, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
Chlamydia trachomatis (CT) is a common sexually transmitted bacterial infection, that in women can cause pelvic inflammatory disease and infertility. No preventative vaccine has been developed against CT. Immunity to CT is primarily mediated by Th1 CD4+ T cells. We are defining immunoprevalent CT proteins in a well-defined cohort of CT seropositive women with the goal of defining novel vaccine immunogens. We screened 30 women one month after a CT-positive test by cultured IFN-γ ELISpot. Ten-day short-term cell lines (STCL) were generated against overlapping peptides spanning 21 CT antigens. The threshold for a positive CT-specific T cell response (≥ 300 spot-forming cells, SFU, per 106 cells) was defined following a screening of 12 CT seronegative donors. CT− specific T cell responses were detected in 27/30 CT-seropositive women. On average, women harbored T cell responses to two CT proteins (range 0–6). Strikingly, CT858 (CPAF) elicited a T cell response in 16/30 women with an average of 966 IFN-g SFU/106 cells (range 300–3,633 SFU/106 cells). Data to date suggest CPAF-specific T cell responses are predominantly CD4-restricted. In preliminary studies, we have also detected CT-specific T cell responses in men with documented CT infection, including a T cell response to CPAF. We are currently mapping CPAF T cell epitopes and expanding our screen to other CT secreted proteins. In summary, CT858 (CPAF) is an immunoprevalent antigen in women and a promising vaccine immunogen. Supported by UNC Chlamydia Vaccine Initiative (U19AI144181)
- Published
- 2022
- Full Text
- View/download PDF
5. Conserved-region MVA vaccines can shift HIV T cell immunodominance in PWH on ART - the M&M Study
- Author
-
Yinyan Xu, shahryat Samir, Ann Marie K. Weideman, Sallay Kallon, Shayla Conrad, Fiona Shaw, Joanna Warren, Maria Abad Fernandez, Lawrence Fox, David M. Margolis, Michael G. Hudgens, Tomas Hanke, JoAnn Kuruc, Cindy Gay, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
CD8+ T cell immunity is essential to the control of HIV viremia. We examined the safety and immunogenicity of MVA-vectored vaccines expressing highly conserved HIV regions in a first-in-man Phase I study in people living with HIV on ART. Participants received a single intramuscular dose of MVA.tHIVconsv3 (M3), MVA.tHIVconsv4 (M4), combined M3+M4 or saline in a 7:7:7:3 ratio. M3 and M4 span the same 6 HIV regions but differ by approximately 10% of amino acids; a design to increase vaccine coverage of circulating HIV variants. We employed ex vivo IFN-g ELISpot assays to measure changes in HIV-specific T cell magnitude and breadth to M3 and/or M4 immunogens following vaccination. We also examined whether M3, M4 or M3+M4 vaccination increased the ability of CD8+ T cells inhibit HIV in vitro replication. The M&M study is fully enrolled but presently is blinded. Analysis of blinded data show that vaccination was safe and well tolerated. Vaccination induced strong increases in the T cell response to M3, M4 vaccine immunogens producing a 2- to 18-fold increase in magnitude in 16/20 participants tested to date. M3/M4-specific T cell breadth also increased across participants. Vaccine-associated T cell responses mostly remained elevated (>2-fold increase) for at least 70 days post-vaccination visit. Vaccination was also associated with clear and sustained increases in in vitro virus inhibition. The percentage of the total HIV T cell response targeting conserved HIV regions in participants increased on average from 40 to 60% post-vaccination, suggesting M3/M4/M3+4 vaccination successfully produced a sustained shift in T cell immunodominance. Unblinded data will be presented at this meeting. Supported by U01AI131310
- Published
- 2022
- Full Text
- View/download PDF
6. The HIV-1 latent reservoir is largely sensitive to circulating T cells
- Author
-
Joanna A Warren, Shuntai Zhou, Yinyan Xu, Matthew J Moeser, Daniel R MacMillan, Olivia Council, Jennifer Kirchherr, Julia M Sung, Nadia R Roan, Adaora A Adimora, Sarah Joseph, JoAnn D Kuruc, Cynthia L Gay, David M Margolis, Nancie Archin, Zabrina L Brumme, Ronald Swanstrom, and Nilu Goonetilleke
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Male ,Human immunodeficiency virus (HIV) ,HIV Infections ,medicine.disease_cause ,Virus Replication ,Cohort Studies ,immunology ,T-Cell Epitopes ,Epitopes ,0302 clinical medicine ,Immunology and Inflammation ,Biology (General) ,Phylogeny ,Pediatric ,General Neuroscience ,General Medicine ,Viral Load ,Middle Aged ,3. Good health ,Virus Latency ,medicine.anatomical_structure ,Infectious Diseases ,030220 oncology & carcinogenesis ,Proteome ,HIV/AIDS ,Medicine ,Female ,medicine.symptom ,Infection ,hiv cure ,Research Article ,Human ,Adult ,Anti-HIV Agents ,QH301-705.5 ,T cell ,Science ,Inflammation ,Biology ,T cell response ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Clinical Research ,medicine ,Humans ,human ,Aged ,General Immunology and Microbiology ,HIV ,CD8 ,Virology ,Antiretroviral therapy ,030104 developmental biology ,Good Health and Well Being ,inflammation ,Mutation ,HIV-1 ,hiv reservoir ,Biochemistry and Cell Biology - Abstract
HIV-1-specific CD8+ T cells are an important component of HIV-1 curative strategies. Viral variants in the HIV-1 reservoir may limit the capacity of T cells to detect and clear virus-infected cells. We investigated the patterns of T cell escape variants in the replication-competent reservoir of 25 persons living with HIV-1 (PLWH) durably suppressed on antiretroviral therapy (ART). We identified all reactive T cell epitopes in the HIV-1 proteome for each participant and sequenced HIV-1 outgrowth viruses from resting CD4+ T cells. All non-synonymous mutations in reactive T cell epitopes were tested for their effect on the size of the T cell response, with a≥50% loss defined as an escape mutation. The majority (68%) of T cell epitopes harbored no detectable escape mutations. These findings suggest that circulating T cells in PLWH on ART could contribute to control of rebound and could be targeted for boosting in curative strategies.
- Published
- 2020
7. CD8 co-receptor links T cell avidity and metabolism
- Author
-
Genevieve Tyndale Clutton, Ann Marie Weideman, Sallay Kallon, Yinyan Xu, Joanna Warren, Erik Lenarcic, Lin Lin, Olivia Council, Michael Muehlbauer, Adam Mincey, Demitrius Hill, Nathaniel Moorman, null RolTisch, Christopher Newgard, James Bain, Paul Armistead, Michael Hudgens, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
This study investigated the mechanistic basis behind the reported superior efficacy of high-avidity CD8 T cells. CMV and HIV are both chronic viral infections, but while CMV-specific CD8 T cells can mediate lifelong viral control, in untreated HIV infection HIV-specific CD8 T cells progressively lose function. Using in vitro studies of human cells, we show that avidity-dependent downregulation of the CD8 co-receptor directly programs metabolism, due to a novel association between CD8 and the glucose transporter GLUT1. We used flow cytometry to profile ex vivo and virus-specific CD8 T cells from HIV-infected individuals on antiretroviral therapy. Ex vivo, cells expressing low levels of CD8 (CD8dim) expressed more CD69 but less cell surface GLUT1, and took up less glucose (2-NBDG) than CD8bright T cells. Following antigen stimulation, CD3, CD8, and GLUT1 were downregulated from the cell surface in an avidity-dependent manner. CMV-specific CD8 T cells, which were of higher avidity, downregulated these proteins to a greater extent than lower-avidity HIV-specific CD8 T cells. GLUT1 downregulation strongly correlated with CD8 but not CD3 downregulation. Antibody-mediated downregulation of CD8 from the cell surface resulted in reduced glucose uptake and increased fatty acid (Bodipy) uptake, independent of CD3. Finally, CD3, CD8, and GLUT1 downregulation by HIV-specific CD8 T cells was impaired following viral escape mutations that reduced CD8 T cell avidity. We confirmed this finding in a transduction setting with a single clonal TCR. Our data reveal a novel function of the CD8 co-receptor, linking the avidity and metabolism of CD8 T cells.
- Published
- 2021
- Full Text
- View/download PDF
8. CD3 and CD8 coreceptor down-modulation are inversely associated with CD8 T cell functional avidity in humans
- Author
-
Genevieve Tyndale Clutton, Sallay Kallon, Ann Weideman, Yinyan Xu, Joanna Warren, Olivia Council, Damir Alzhanov, Hannah Thaxton, Michael Hudgens, Joann Kuruc, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
This study investigated the function of memory CD8 T cells in HIV-infected people durably suppressed on antiretroviral therapy (HIV+ cART). We assessed bulk and virus-specific memory CD8 T cells in HIV+ cART and HIV-seronegative individuals (HIV−) by flow cytometry. We observed a population of CD3+ CD8dim CD14− CD16− (CD8dim) T cells that was expanded as a percentage of total CD8 T cells in both HIV− and CMV-seropositive individuals. Bulk memory CD8dim T cells expressed significantly higher CD69 and less MHC Class I and CD127 ex vivo than CD8bright T cells, suggesting recent activation. CD8dim T cells expressed less GLUT1 and PGC-1α and took up less glucose (2-NBDG) and lipid (Bodipy) than CD8bright T cells, indicating relatively lower metabolic activity. Multimer reactivity was used to examine CMV-, EBV- and HIV-specific CD8 T cells ex vivo. Virus-specific populations were consistently CD8high. However, after peptide stimulation, antigen-specific CD8 T cells down-regulated CD3 and CD8. CMV-specific CD8 T cells down-regulated CD3 and CD8 more than HIV-specific cells. CD3 and CD8 downregulation were strongly correlated with the functional avidity of the response. A strong correlation between GLUT1 down-regulation and CD8 down-regulation was also observed, suggesting an association between CD8 expression and metabolic activity. These results suggest that the expanded CD8dim population in HIV+ cART individuals, who are >90% CMV-seropositive, may be driven by ongoing activation of high-avidity CMV-specific CD8 T cells. They also suggest that different virus-specific CD8 T cell populations differentially downregulate components of the TCR complex and metabolism after antigen stimulation, possibly to avoid excessive activation.
- Published
- 2020
- Full Text
- View/download PDF
9. Markedly High Plasma Thrombopoietin (TPO) Level is a Predictor of Poor Response to Immunosuppressive Therapy in Children With Acquired Severe Aplastic Anemia
- Author
-
Shaimaa Elmahdi, N Nishio, Atsushi Narita, Yinyan Xu, Yusuke Okuno, Hideki Muramatsu, Olfat Ismael, Yoshiyuki Takahashi, Xinan Wang, Seiji Kojima, and Asahito Hama
- Subjects
Response rate (survey) ,medicine.medical_specialty ,Multivariate analysis ,Globulin ,biology ,business.industry ,Proportional hazards model ,Hematology ,Odds ratio ,medicine.disease ,Gastroenterology ,Confidence interval ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Immunology ,medicine ,biology.protein ,Aplastic anemia ,business ,Thrombopoietin ,030215 immunology - Abstract
Background Immunosuppressive therapy (IST) is commonly used for patients with acquired severe aplastic anemia (SAA). Because the clinical response rate and therapeutic outcome for individual patients to IST varies, an in vitro test that identifies potential responders would be desirable. Methods We evaluated the relationship between thrombopoietin (TPO) levels at the time of diagnosis and the response to IST at 6 months in 85 children (median age, 9.0 years; range, 1.0–15.5 years) with acquired SAA using enzyme-linked immunosorbent assay. Thirty-one age-matched healthy individuals were used as controls. All patients received antithymocyte globulin and cyclosporine. Results Overall, 39 patients (45.9%) responded to IST at 6 months. TPO plasma levels were significantly higher in nonresponders than in responders (1,555.8 vs. 1,284.7 pg/ml, respectively; P = 0.031). Multivariate analysis identified the TPO levels of >1,796.7 pg/ml (TPO-high group, 20 patients; odds ratio (OR), 8.285; 95% confidence interval (CI), 2.114–32.904; P = 0.002) as independent poor predictors of IST response at 6 months. Moreover, the TPO-high group was associated with lower 5-year failure-free survival rates (30% vs. 68%, P = 0.012) compared with the TPO-low group. Conclusion The measurement of TPO levels at diagnosis is useful for predicting the response to IST in children with SAA and may help in decision making.
- Published
- 2015
- Full Text
- View/download PDF
10. A Cytokine-Based Diagnostic Program in Pediatric Aplastic Anemia and Hypocellular Refractory Cytopenia of Childhood
- Author
-
Xinan Wang, Seiji Kojima, N Nishio, Masafumi Ito, Yusuke Okuno, Daisuke Hasegawa, Yoshiyuki Takahashi, Olfat Ismael, Hideki Muramatsu, Shaimaa Elmahdi, Asahito Hama, Nozomu Kawashima, Yinyan Xu, Atsushi Manabe, and Atsushi Narita
- Subjects
medicine.medical_specialty ,Cytopenia ,business.industry ,Myelodysplastic syndromes ,Hematology ,Odds ratio ,medicine.disease ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Refractory ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Immunology ,medicine ,Aplastic anemia ,Differential diagnosis ,Prospective cohort study ,business ,Thrombopoietin ,030215 immunology - Abstract
Background Distinguishing hypocellular refractory cytopenia of childhood (RCC) from aplastic anemia (AA) is challenging. Thus far, no studies have compared the cytokine profiles in patients with AA to those with hypocellular RCC. In the present study, we addressed whether thrombopoietin (TPO) and interleukin 17 (IL-17) plasma levels are useful for differentiating between the two diseases. Methods We measured the endogenous plasma concentrations of TPO and IL-17 in 29 patients with AA, 34 patients with hypocellular RCC, and 31 healthy controls using sensitive enzyme-linked immunosorbent assays. Results The TPO and IL-17 plasma levels were significantly lower in patients with hypocellular RCC than in patients with AA (P < 0.001 and P = 0.007, respectively). The multivariate logistic regression analysis identified moderate disease severity, TPO levels of
- Published
- 2015
- Full Text
- View/download PDF
11. Integrated molecular profiling of juvenile myelomonocytic leukemia
- Author
-
Xinan Wang, Asahito Hama, Nozomu Kawashima, Kyogo Suzuki, Masashi Sanada, Atsushi Narita, Hiroyuki Aburatani, Yuichi Shiraishi, Arata Watanabe, Hideki Muramatsu, Seishi Ogawa, Genta Nagae, Seiji Kojima, Kenichi Yoshida, Masashi Hirayama, Hiroko Tanaka, Toshihide Ueno, Yoshiyuki Takahashi, Satoru Miyano, Hirotoshi Sakaguchi, Yusuke Okuno, Hiroyuki Mano, Norihiro Murakami, Masafumi Ito, Kenichi Chiba, and Yinyan Xu
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Male ,Adolescent ,Oncogene Proteins, Fusion ,Immunology ,Gene mutation ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Crizotinib ,hemic and lymphatic diseases ,ROS1 ,Medicine ,Humans ,Child ,Protein Kinase Inhibitors ,Myeloproliferative neoplasm ,Cell Proliferation ,Juvenile myelomonocytic leukemia ,business.industry ,Gene Expression Profiling ,Myeloid leukemia ,Infant ,Cell Biology ,Hematology ,DNA, Neoplasm ,DNA Methylation ,medicine.disease ,Leukemia ,030104 developmental biology ,Leukemia, Myelomonocytic, Juvenile ,030220 oncology & carcinogenesis ,Child, Preschool ,Mutation ,Cancer research ,Female ,business ,medicine.drug ,Genome-Wide Association Study - Abstract
Juvenile myelomonocytic leukemia (JMML), a rare and aggressive myelodysplastic/myeloproliferative neoplasm that occurs in infants and during early childhood, is characterized by excessive myelomonocytic cell proliferation. More than 80% of patients harbor germ line and somatic mutations in RAS pathway genes (eg, PTPN11, NF1, NRAS, KRAS, and CBL), and previous studies have identified several biomarkers associated with poor prognosis. However, the molecular pathogenesis of 10% to 20% of patients and the relationships among these biomarkers have not been well defined. To address these issues, we performed an integrated molecular analysis of samples from 150 JMML patients. RNA-sequencing identified ALK/ROS1 tyrosine kinase fusions (DCTN1-ALK, RANBP2-ALK, and TBL1XR1-ROS1) in 3 of 16 patients (18%) who lacked canonical RAS pathway mutations. Crizotinib, an ALK/ROS1 inhibitor, markedly suppressed ALK/ROS1 fusion-positive JMML cell proliferation in vitro. Therefore, we administered crizotinib to a chemotherapy-resistant patient with the RANBP2-ALK fusion who subsequently achieved complete molecular remission. In addition, crizotinib also suppressed proliferation of JMML cells with canonical RAS pathway mutations. Genome-wide methylation analysis identified a hypermethylation profile resembling that of acute myeloid leukemia (AML), which correlated significantly with genetic markers with poor outcomes such as PTPN11/NF1 gene mutations, 2 or more genetic mutations, an AML-type expression profile, and LIN28B expression. In summary, we identified recurrent activated ALK/ROS1 fusions in JMML patients without canonical RAS pathway gene mutations and revealed the relationships among biomarkers for JMML. Crizotinib is a promising candidate drug for the treatment of JMML, particularly in patients with ALK/ROS1 fusions.
- Published
- 2017
12. Aldehyde dehydrogenase-2 polymorphism contributes to the progression of bone marrow failure in children with idiopathic aplastic anaemia
- Author
-
Xinan Wang, Hideki Muramatsu, Seiji Kojima, Atsushi Narita, Koji Nakanishi, Asahito Hama, Nozomu Kawashima, Yoshiyuki Takahashi, Yinyan Xu, Sayoko Doisaki, and Hirotoshi Sakaguchi
- Subjects
Male ,Idiopathic aplastic anaemia ,Adolescent ,Pancytopenia ,Aldehyde dehydrogenase ,Bone Marrow ,medicine ,Humans ,Child ,ALDH2 ,Polymorphism, Genetic ,biology ,business.industry ,Aldehyde Dehydrogenase, Mitochondrial ,Bone marrow failure ,Anemia, Aplastic ,Infant ,Hematology ,Aldehyde Dehydrogenase ,medicine.disease ,Child, Preschool ,Immunology ,Disease Progression ,biology.protein ,Female ,business - Published
- 2014
- Full Text
- View/download PDF
13. High Natural Killer Cell Count at Diagnosis Predicts a Good Response to Immunosuppressive Therapy in Aplastic Anemia
- Author
-
Yusuke Okuno, Seiji Kojima, Eri Nishikawa, Nobuhiro Nishio, Hirohito Yamazaki, Shinji Nakao, Yinyan Xu, Hideki Muramatsu, Atsushi Narita, Motoharu Hamada, Nozomu Kawashima, Daisuke Ichikawa, and Yoshiyuki Takahashi
- Subjects
biology ,business.industry ,Immunology ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Acquired immune system ,Biochemistry ,CD19 ,Transplantation ,Immune system ,medicine ,biology.protein ,IL-2 receptor ,Aplastic anemia ,business ,Interleukin-7 receptor ,CD8 - Abstract
Introduction: Aplastic anemia (AA) is an autoimmune-mediated disease with a complex mechanism. Innate as well as acquired immune system have been considered to participate in its pathogenesis. Although several studies have demonstrated the role of T and B cells in AA, there is limited research focusing on natural killer (NK) cells, which are important in innate and adaptive immune system. NK cells interact with T cells and alter their immune responses in a variety of diseases. Recent investigation suggests that NK cells serve immunoregulatory roles by attenuating autologous CD8+ T-cell response in AA (Chen et al. Cell Immunol 335:6-14, 2019). Here we assessed the kinetics of NK cells before and after immunosuppressive therapy (IST) in patients with AA to determine the association between NK cells and clinical courses. Methods: Patients with severe AA, in Japan, who required IST as first-line therapy were prospectively enrolled between May 2012 and October 2017. The IST regimen comprised rabbit anti-thymocyte globulin (ATG, thymoglobulin®, 2.5 or 3.5 mg/kg/day for 5 days), cyclosporine A (6 mg/kg/day for minimum 6 months), and methylprednisolone (2 mg/kg/day for 5 days) with subsequent tapering of the dose for 28 days. Flow cytometry was used to assess lymphocyte subsets, consisting of CD3+, CD4+, and CD8+ T cells, CD19+ B cells, CD56+ NK cells, and CD4+CD25+CD127+ regulatory T cells (Tregs), before and after ATG administration on days 0, 14, 28, 60, 90, 120, and 180. Plasma rabbit ATG levels were measured using a rabbit IgG ELISA kit on days 14 and 28 (Bethyl Laboratories, Montgomery, TX, USA). Receiver-operator characteristic (ROC) curves were generated to differentiate between response and no response to IST. Results: A total of 81 patients (aged 1.7-67.9 years) were randomized; 43 and 38 patients received 2.5 and 3.5 mg/kg of r-ATG, respectively. Median follow-up duration was 445 days (range; 183-2165 days) from first ATG infusion. After 6 months, 50 patients (58%) responded to the therapy, including 4 (5%) who achieved a complete response and 45 (56%) a partial response. The response rates did not differ significantly between the two dosages in ATG groups at 6 months (2.5 mg/kg, 63% vs. 3.5 mg/kg, 58%, P = 0.820). The median absolute number of NK cells before IST was 92/µL (0-1,085/µL). To judge patients' responses to IST, we defined 138/µL as the absolute number of NK cells count before IST as the cut-off value using ROC curves. The response rate of patients with higher NK cells (≥ 138 /mL) to IST at 6 months was significantly higher than those with lower NK cells (Figure 1, < 138 /mL, 78% vs. 52%, P = 0.031). Multivariate logistic regression analysis identified the absolute number of NK cells before IST and reticulocyte at diagnosis (odds ratio [OR] = 0.16; 95% confidence interval [CI], 0.04-0.62; P = 0.008 and OR = 0.20; 95% CI, 0.06-0.67; P = 0.009, respectively) and the r-ATG plasma concentration on day 28 (OR = 0.11; 95% CI, 0.03-0.39; P = 0.001) as independent predictors of response to IST. The median absolute number of NK cells on days 90, 120, and 180 after IST was higher in the responders (113/mL, 118/mL, and 159/mL, respectively) than in the non-responders (57/mL, 53/mL, and 64/mL, P = 0.001, P Conclusions: The current study indicates that NK cells play a role in the pathogenesis of AA and may be the predictors of response to IST. Recovery of NK cell count correlated with hematopoietic recovery following IST, indicating that they may be used as markers to assess disease activity, treatment response and relapse. Disclosures Yamazaki: Novartis Pharma K.K.: Honoraria; Sanofi K.K.: Honoraria; Nippon Shinyaku Co., Ltd.: Honoraria. Nakao:Ono Pharmaceutical: Honoraria; Kyowa Kirin: Honoraria; Alaxion Pharmaceuticals: Honoraria; Ohtsuka Pharmaceutical: Honoraria; Celgene: Honoraria; Daiichi-Sankyo Company, Limited: Honoraria; Janssen Pharmaceutical K.K.: Honoraria; SynBio Pharmaceuticals: Consultancy; Novartis Pharma K.K: Honoraria; Bristol-Myers Squibb: Honoraria; Takeda Pharmaceutical Company Limited: Honoraria; Chugai Pharmaceutical Co.,Ltd: Honoraria.
- Published
- 2019
- Full Text
- View/download PDF
14. Quantifying Virus Escape from T Cells in the Latent HIV Reservoir
- Author
-
Joanna A Warren, Shuntai Zhou, Yinyan Xu, Matthew Moeser, Jenn Kirchherr, Julia Sung, Nadia Roan, Adaora Adimora, JoAnn Kuruc, Cindy Gay, David Margolis, Nancie Archin, Ronald Swanstrom, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
There is no cure for HIV, largely because HIV establishes a small but sustained pool of latently infected cells that are not cleared by antiretroviral therapy (ART). Current strategies include firstly reactivating the latent HIV reservoir and then using T cell immunotherapy to clear reactivated cells. However, pre-ART CD8 T cell escape variants have been reported in the HIV reservoir, which may limit CD8 T cell recognition and clearance of HIV-infected cells. The extent of virus escape from T cells in the latent reservoir is unclear. HIV-specific T cell responses were comprehensively mapped across the Clade B HIV proteome by IFN-g ELISpot in 25 ART-suppressed participants. In parallel, replication competent viruses derived from supernatants of autologous resting CD4 T cells following mitogenic reactivation were sequenced. Peptides spanning virus variants within reactive T cell epitopes were synthesized and examined by ELISpot for evidence of escape, defined as ≥50% difference in T cell magnitude between peptide variants. No correlations were observed between the size of the latent HIV reservoir and either HIV-specific T cell breadth (1–19 epitopes) or magnitude (156–2855 SFU/M PBMCs). T cell escape was assessed in 17 participants. 39% of reactive T cell epitopes (48/124) harbored ≥1 amino acid variants in the sequenced latent reservoir. Of those 48, 20 afforded T cell escape, providing an overall escape frequency in the reservoir of 16% (20/124). These data show that the majority of replication competent latent HIV viruses do not harbor CD8 T cell escape mutants, suggesting that immunotherapy approaches that boost CD8 T cell responses can successfully target the latent reservoir in HIV curative and or remission strategies.
- Published
- 2019
- Full Text
- View/download PDF
15. Integrated NY-ESO-1 antibody and CD8 + T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab
- Author
-
Gerd Ritter, Jedd D. Wolchok, Mario Sznol, Teresa S. Rasalan, Sacha Gnjatic, Lloyd J. Old, Ruth Halaban, Stephanie L. Terzulli, Humilidad F. Gallardo, Evelina Pogoriler, Deborah Kuk, Jianda Yuan, Matthew Adamow, Brian A. Ginsberg, Achim A. Jungbluth, Katherine S. Panageas, James P. Allison, Yinyan Xu, and Erika Ritter
- Subjects
Adult ,Adoptive cell transfer ,medicine.medical_treatment ,Enzyme-Linked Immunosorbent Assay ,Ipilimumab ,CD8-Positive T-Lymphocytes ,Immune system ,Antigen ,Antigens, Neoplasm ,Humans ,Cytotoxic T cell ,Medicine ,CTLA-4 Antigen ,Melanoma ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,Multidisciplinary ,biology ,business.industry ,Antibodies, Monoclonal ,Membrane Proteins ,Immunosuppression ,Biological Sciences ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Survival Analysis ,Immunology ,biology.protein ,Antibody ,business ,medicine.drug - Abstract
Ipilimumab, a monoclonal antibody against cytotoxic T lymphocyte antigen 4 (CTLA-4), has been shown to improve survival in patients with advanced metastatic melanoma. It also enhances immunity to NY-ESO-1, a cancer/testis antigen expressed in a subset of patients with melanoma. To characterize the association between immune response and clinical outcome, we first analyzed NY-ESO-1 serum antibody by ELISA in 144 ipilimumab-treated patients with melanoma and found 22 of 140 (16%) seropositive at baseline and 31 of 144 (22%) seropositive following treatment. These NY-ESO-1–seropositive patients had a greater likelihood of experiencing clinical benefit 24 wk after ipilimumab treatment than NY-ESO-1–seronegative patients ( P = 0.02, relative risk = 1.8, two-tailed Fisher test). To understand why some patients with NY-ESO-1 antibody failed to experience clinical benefit, we analyzed NY-ESO-1–specific CD4 + and CD8 + T-cell responses by intracellular multicytokine staining in 20 NY-ESO-1–seropositive patients and found a surprising dissociation between NY-ESO-1 antibody and CD8 responses in some patients. NY-ESO-1–seropositive patients with associated CD8 + T cells experienced more frequent clinical benefit (10 of 13; 77%) than those with undetectable CD8 + T-cell response (one of seven; 14%; P = 0.02; relative risk = 5.4, two-tailed Fisher test), as well as a significant survival advantage ( P = 0.01; hazard ratio = 0.2, time-dependent Cox model). Together, our data suggest that integrated NY-ESO-1 immune responses may have predictive value for ipilimumab treatment and argue for prospective studies in patients with established NY-ESO-1 immunity. The current findings provide a strong rationale for the clinical use of modulators of immunosuppression with concurrent approaches to favor tumor antigen-specific immune responses, such as vaccines or adoptive transfer, in patients with cancer.
- Published
- 2011
- Full Text
- View/download PDF
16. Determinants of CD8+ T Cell Immunodominance in Acute HIV-1 Infection
- Author
-
Joanna A. Warren, Yinyan Xu, Margaret Constanzo, Victoria Whale, Michael Liu, Rasmi Thomas, Sodsai Tovanabutra, Gustavo H. Kijak, Leigh A. Eller, Morgane Rolland, Nelson L. Michael, Merlin L. Robb, Michael A. Eller, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
CD8+T cells detect and clear virus-infected cells. During human immunodeficiency virus-1 (HIV-1) infection, different HIV-specific CD8+ T cell populations are expanded, each targeting a discreet epitope. These different HIV-specific CD8+ T cell populations do not arise equally, and instead establish a hierarchy (immunodominance) based on strength of T cell response to a given viral epitope. Our previous work has shown that immunodominance is a major determinant of the rate at which HIV can mutate to escape T cell immune pressure. The precise mechanism of how the first CD8+ T cell response to HIV is generated, and which factors determine T cell dominance is unclear. To investigate determinants of T cell immunodominance in acute HIV-1 infection (~45 days after infection), we determined all primary HIV-1 specific CD8+ T cell responses in 10 individuals from the RV217 early acute HIV-1 cohort. Participants are high-risk volunteers from Kenya and Thailand, and acute HIV-1 infection was determined from twice-weekly blood draws using a nucleic acid test. T cell responses were mapped against the unique infecting HIV-1 virus/es in each individual using IFN-γ ELISpot. Across participants, the immunodominant T cell response mostly targeted the Env, Gag and Pol proteins of the HIV-1 genome. We detected a wide (4–14 epitopes) breadth of T cell response across our cohort, with all HIV-1 proteins targeted and different rates of virus escape observed. The summed magnitude of response ranged from 1,388–16,044 SFU/106 cells. Ongoing studies are focusing on better understanding how factors, such as T cell receptor repertoire, avidity, antigen presentation and function contribute to T cell immunodominance, which in turn will inform HIV-1 vaccine design.
- Published
- 2018
- Full Text
- View/download PDF
17. CD8+ T cell-mediated inhibition of HIV-1
- Author
-
Yinyan Xu, Maria Abad, Joanna A. Warren, Genevieve Clutton, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
CD8+ T cells contribute to the control of viral infections, including HIV-1. Previous studies have reported that CD8+-mediated virus inhibition is predictive of subsequent HIV plasma viremia and CD4+T cell decline in HIV-infected, antiretroviral naïve individuals; higher virus inhibition is associated with a better clinical outcomes. Ex vivo CD8+ T cell-mediated inhibition of HIV replication is measured after co-culture of CD8+ T cells with HIV-1 super-infected, autologous CD4+ T cells, at different effector to target ratios. We adapted and standardized the CD8+ T cell virus inhibition assay (CD8-VIA) for use as an endpoint assay in therapeutic testing of HIV-1 T cell vaccines. CD8+ T cells from HIV infected, durably (>2 years) antiretroviral suppressed participants (n=11) were co-cultured with JR-CSF infected autologous CD4+ T cells at E:T ratios ranging from 1/2:1 to 1/160:1, generating CD8+ T cell virus inhibition curves for each participant. IC50 values varied by 100-fold across participants suggesting that CD8+ T cells harbor a broad range of virus inhibitory capacity even when viremia in participants is durably suppressed. Ongoing studies are comparing IC50 values to other measurements (magnitude, specificity, avidity, proliferation and production of cytokines and lytic molecules) of HIV-specific T cell immunity to understand the critical determinants of CD8+ T cell inhibitory capacity.
- Published
- 2018
- Full Text
- View/download PDF
18. Development of an in vitro cell culture model to investigate HCMV priming of CD8+ T cells
- Author
-
Maria Abad, Erik Lenarcic, Yinyan Xu, Jason Wong, Genevieve Clutton, Joanna A. Warren, Barbara Savoldo, Nathaniel J. Moorman, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
Human cytomegalovirus (HCMV)-specific CD8+ T cells are characterized by an unique, non-exhausting, effector memory phenotype. How HCMV induces this phenotype is poorly characterized. We hypothesized that HCMV may skew T cell priming producing functionally distinct CD8+ T cells. To investigate this question, we have established an autologous 3-cell culture system using human umbilical tissue in which naïve CD8+ T cells are isolated from cord blood, myeloid DCs (mDCs) are generated from cord blood CD34+ precursor cells and fibroblasts, which are a primary target of HCMV infection, are generated from matching umbilical cord (UC-F). Previous studies have reported IFN-γ treatment of fibroblasts induces MHC-II expression, leading to suggestions that fibroblasts can acquire APC-like activity. To investigate this, we used flow cytometry to examine the effects of HCMV infection +/− IFN-γ on MHC-II, CD40, CD80 and CD86 surface expression on fibroblasts. HCMV alone did not induce MHC-II or costimulatory molecules. HCMV infection +/− IFN-γ induced MHC-II but costimulatory molecules remained undetectable. This suggests HCMV-infected fibroblasts do not acquire APC-like phenotype and that fibroblasts mediate T cell priming through cross-presentation. Fibroblasts release extracellular vesicles (EVs), which are important mediators of cell signaling, including antigen cross-presentation. We used electron microscopy to confirm that fibroblasts release EVs. Following AD169-GFP infection, 5.5–8% of EV collected were GFP-positive by ImageStream. Future studies will investigate whether UC-F modulate mDC priming of CD8+ T cells through direct cell-cell contact and/or indirectly, including through the release of EVs.
- Published
- 2018
- Full Text
- View/download PDF
19. Downregulation of GATA-2 and overexpression of adipogenic gene-PPARγ in mesenchymal stem cells from patients with aplastic anemia
- Author
-
Nobuhiro Nishio, Hideki Muramatsu, Makito Tanaka, Yinyan Xu, Seiji Kojima, Nao Yoshida, Yoshiyuki Takahashi, Hiroshi Yagasaki, Asahito Hama, Itzel Bustos Villalobos, and Yue Wang
- Subjects
Adult ,Male ,Cancer Research ,Adolescent ,Blotting, Western ,Down-Regulation ,Gene Expression ,Peroxisome proliferator-activated receptor ,Bone Marrow Cells ,Biology ,Young Adult ,Downregulation and upregulation ,Genetics ,medicine ,Humans ,Aplastic anemia ,Child ,Molecular Biology ,Cells, Cultured ,chemistry.chemical_classification ,Adipogenesis ,Reverse Transcriptase Polymerase Chain Reaction ,Mesenchymal stem cell ,Anemia, Aplastic ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,GATA2 Transcription Factor ,PPAR gamma ,Haematopoiesis ,medicine.anatomical_structure ,chemistry ,Child, Preschool ,Immunology ,Cancer research ,Female ,Bone marrow ,Stem cell - Abstract
Aplastic anemia (AA) is characterized by a reduced number of hematopoietic stem cells and fatty replacement in the bone marrow. Transcriptional factor GATA-2 plays several important roles in both hematopoiesis and adipogenesis. Decreased levels of GATA-2 compromise the proliferation and survival of hematopoietic stem cells. GATA-2 suppresses adipocyte differentiation through direct inhibition of adipogenic factors, including peroxisome proliferator-activated receptor-gamma (PPARgamma). Previous studies have shown that expression of GATA-2 is decreased in marrow CD34-positive cells in AA. To elucidate the mechanisms of fatty marrow replacement, we evaluated the mRNA expression for GATA-2 and PPARgamma in mesenchymal stem cells (MSCs) from patients with AA by quantitative real-time polymerase chain reaction. GATA-2 expression by MSCs from AA patients was significantly lower than in normal subjects. Conversely, expression of PPARgamma was significantly higher in AA patients. Western blot analysis demonstrated that protein levels of GATA-2 were lower in AA patients than those in normal subjects. Moreover, incubation with interferon-gamma induced downregulation of GATA-2 levels in MSCs from normal subjects. These findings indicate that fatty marrow replacement in AA patients can be explained by downregulation of GATA-2 and overexpression of PPARgamma in MSCs. Decreased expression of GATA-2 might be responsible for the pathogenesis and development of the clinical features of the disease.
- Published
- 2009
- Full Text
- View/download PDF
20. Optimization and validation of a robust human T-cell culture method for monitoring phenotypic and polyfunctional antigen-specific CD4 and CD8 T-cell responses
- Author
-
Yun Lin, Humilidad Gallardo, Geoffrey Ku, Hao Li, Gregor Manukian, Teresa Rasalan, Yinyan Xu, Stephanie Terzulli, Lloyd Old, James Allison, Alan Houghton, Jedd Wolchok, and Jianda Yuan
- Subjects
Cancer Research ,Transplantation ,Oncology ,Immunology ,Immunology and Allergy ,Cell Biology ,Genetics (clinical) - Published
- 2009
- Full Text
- View/download PDF
21. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit
- Author
-
Achim A. Jungbluth, Lloyd J. Old, Gerd Ritter, Geoffrey Y. Ku, Yinyan Xu, Teresa S. Rasalan, Erika Ritter, Ruth Ann Roman, James P. Allison, Humilidad F. Gallardo, Melanie Heywood, Jedd D. Wolchok, Hao Li, Neil H. Segal, Evelina Pogoriler, Sacha Gnjatic, Sarah Powel, Gregor Manukian, Jianda Yuan, and Stephanie L. Terzulli
- Subjects
Adult ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunoglobulins ,T-Cell Antigen Receptor Specificity ,Enzyme-Linked Immunosorbent Assay ,Ipilimumab ,Article ,Immune system ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,Humans ,Medicine ,Cytotoxic T cell ,CTLA-4 Antigen ,Neoplasm Metastasis ,Antibodies, Blocking ,Melanoma ,Aged ,Aged, 80 and over ,B-Lymphocytes ,Clinical Trials as Topic ,Membrane Glycoproteins ,Multidisciplinary ,business.industry ,Immunity ,Antibodies, Monoclonal ,Membrane Proteins ,Immunotherapy ,Middle Aged ,Biological Sciences ,Peptide Fragments ,Treatment Outcome ,medicine.anatomical_structure ,CTLA-4 ,Antibody Formation ,Immunology ,Cytokines ,business ,CD8 ,gp100 Melanoma Antigen ,medicine.drug - Abstract
Blockade of inhibitory signals mediated by cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) has been shown to enhance T cell responses and induce durable clinical responses in patients with metastatic melanoma. The functional impact of anti-CTLA-4 therapy on human immune responses is still unclear. To explore this, we analyzed immune-related adverse events and immune responses in metastatic melanoma patients treated with ipilimumab, a fully human anti-CTLA-4 monoclonal antibody. Fifteen patients were selected on the basis of availability of suitable specimens for immunologic monitoring, and eight of these showed evidence of clinical benefit. Five of the eight patients with evidence of clinical benefit had NY-ESO-1 antibody, whereas none of seven clinical non-responders was seropositive for NY-ESO-1. All five NY-ESO-1 seropositive patients had clearly detectable CD4+and CD8+T cells against NY-ESO-1 following treatment with ipilimumab. One NY-ESO-1 seronegative clinical responder also had a NY-ESO-1 CD4+and CD8+T cell response, possibly related to prior vaccination with NY-ESO-1. Among five clinical non-responders analyzed, only one had a NY-ESO-1 CD4+T cell response and this patient did not have detectable anti-NY-ESO-1 antibody. Overall, NY-ESO-1-specific T cell responses increased in frequency and functionality during anti-CTLA-4 treatment, revealing a polyfunctional response pattern of IFN-γ, MIP-1β and TNF-α. We therefore suggest that CTLA-4 blockade enhanced NY-ESO-1 antigen-specific B cell and T cell immune responses in patients with durable objective clinical responses and stable disease. These data provide an immunologic rationale for the efficacy of anti-CTLA-4 therapy and call for immunotherapeutic designs that combine NY-ESO-1 vaccination with CTLA-4 blockade.
- Published
- 2008
- Full Text
- View/download PDF
22. Paroxysmal nocturnal hemoglobinuria and telomere length predicts response to immunosuppressive therapy in pediatric aplastic anemia
- Author
-
Asahito Hama, Nozomu Kawashima, Etsuro Ito, Shouichi Ohga, Seiji Kojima, Hiroshi Moritake, Kazuko Kudo, Ryoji Kobayashi, Akira Ohara, Nobuhiro Nishio, Yinyan Xu, Hirotoshi Sakaguchi, Atsushi Narita, Yusuke Okuno, Yoshiyuki Takahashi, Hiromasa Yabe, Hideki Muramatsu, Xinan Wang, Yuko Sekiya, Nao Yoshida, Masao Kobayashi, and Sayoko Doisaki
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Anemia ,medicine.medical_treatment ,Population ,Hemoglobinuria, Paroxysmal ,Hematopoietic stem cell transplantation ,Gastroenterology ,Telomere Homeostasis ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Aplastic anemia ,education ,Child ,Immunosuppression Therapy ,education.field_of_study ,business.industry ,Anemia, Aplastic ,Infant ,Immunosuppression ,Hematology ,Articles ,Telomere ,medicine.disease ,Prognosis ,Child, Preschool ,Immunology ,Paroxysmal nocturnal hemoglobinuria ,Hemoglobinuria ,Female ,business - Abstract
Acquired aplastic anemia is an immune-mediated disease characterized by severe defects in stem cell number resulting in hypocellular marrow and peripheral blood cytopenias. Minor paroxysmal nocturnal hemoglobinuria populations and a short telomere length were identified as predictive biomarkers of immunosuppressive therapy responsiveness in aplastic anemia. We enrolled 113 aplastic anemia patients (63 boys and 50 girls) in this study to evaluate their response to immunosuppressive therapy. The paroxysmal nocturnal hemoglobinuria populations and telomere length were detected by flow cytometry. Forty-seven patients (42%) carried a minor paroxysmal nocturnal hemoglobinuria population. The median telomere length of aplastic anemia patients was -0.99 standard deviation (SD) (range -4.01-+3.01 SD). Overall, 60 patients (53%) responded to immunosuppressive therapy after six months. Multivariate logistic regression analysis identified the absence of a paroxysmal nocturnal hemoglobinuria population and a shorter telomere length as independent unfavorable predictors of immunosuppressive therapy response at six months. The cohort was stratified into a group of poor prognosis (paroxysmal nocturnal hemoglobinuria negative and shorter telomere length; 37 patients) and good prognosis (paroxysmal nocturnal hemoglobinuria positive and/or longer telomere length; 76 patients), respectively. The response rates of the poor prognosis and good prognosis groups at six months were 19% and 70%, respectively (P
- Published
- 2015
23. Correlation of rabbit antithymocyte globulin serum levels and clinical outcomes in children who received hematopoietic stem cell transplantation from an alternative donor
- Author
-
Xinan Wang, Yinyan Xu, Seiji Kojima, Yuko Sekiya, Yoshinori Ito, Atsushi Narita, Yusuke Okuno, Olfat Ismael, Yuka Torii, Yoshiyuki Takahashi, Shaimaa Elmahdi, Hideki Muramatsu, Asahito Hama, and Nozomu Kawashima
- Subjects
Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,Gastroenterology ,Correlation ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Recurrence ,Risk Factors ,Internal medicine ,Neoplasms ,medicine ,Animals ,Humans ,Transplantation, Homologous ,Cumulative incidence ,Risk factor ,Young adult ,Child ,Antilymphocyte Serum ,Retrospective Studies ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Infant ,Retrospective cohort study ,medicine.disease ,Tissue Donors ,surgical procedures, operative ,Graft-versus-host disease ,Treatment Outcome ,030220 oncology & carcinogenesis ,Child, Preschool ,Hematologic Neoplasms ,Pediatrics, Perinatology and Child Health ,Immunology ,Multivariate Analysis ,Female ,Rabbits ,Neoplasm Recurrence, Local ,business ,030215 immunology - Abstract
We analyzed the correlation between rabbit ATG (rATG) serum levels and clinical outcomes in 37 children who received rATG at a total dose of 10 or 15 mg/kg during HSCT conditioning from an alternative donor. Fourteen patients had advanced malignant diseases, 13 had severe AA, and 10 had inherited disorders. Complete engraftment was achieved in all patients, and no rejection occurred. The cumulative incidence of grades II-IV acute GVHD and extensive chronic GVHD was 27% (95% CI, 12.5-39.6%) and 8.1% (95% CI, 0-23.1%), respectively. Multivariate analysis identified lower rATG levels at week 4 as an independent risk factor in the development of grades II-IV acute GVHD (p = 0.037). Serious infections were not observed in any patient following HSCT. No correlation was found between EBV reactivation and rATG levels at week 2 and week 4 after HSCT. Furthermore, no correlation was found between relapse and rATG levels two and four wk post-transplantation. The probability of five-yr OS among patients was 70.3% (95% CI, 59.8-79.2%). Our results suggest that targeted rATG administration may protect patients from severe acute GVHD without increasing the risk of EBV reactivation or relapse.
- Published
- 2015
24. Enhanced expression of urocortin in lung tissues of rats with allergic asthma
- Author
-
Shengnan Li, Yuqing Wu, Yinyan Xu, and Hong Zhou
- Subjects
Male ,Time Factors ,Corticotropin-Releasing Hormone ,Bronchoalveolar Lavage ,Biochemistry ,Dexamethasone ,Epithelium ,Rats, Sprague-Dawley ,Medicine ,Mast Cells ,Lung ,Urocortins ,Urocortin ,medicine.diagnostic_test ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Degranulation ,respiratory system ,Immunohistochemistry ,Up-Regulation ,medicine.symptom ,Glucocorticoid ,medicine.drug ,endocrine system ,medicine.medical_specialty ,Ovalbumin ,Blotting, Western ,Biophysics ,Bronchi ,Inflammation ,Proinflammatory cytokine ,Internal medicine ,Hypersensitivity ,Animals ,RNA, Messenger ,Glucocorticoids ,Molecular Biology ,Models, Statistical ,business.industry ,Cell Biology ,Actins ,Asthma ,Rats ,respiratory tract diseases ,Disease Models, Animal ,Endocrinology ,Bronchoalveolar lavage ,Gene Expression Regulation ,Immunology ,biology.protein ,Peptides ,business - Abstract
Bronchial asthma is defined as a chronic airway inflammatory disease characterized by sustained activation of many inflammatory cells including mast cells. Urocortin (UCN) is synthesized and secreted by human mast cells and activated mast cells release more UCN. On the other hand, UCN can induce mast cell degranulation and generation of many proinflammatory factors. The purpose of this study was to examine the expression profile of UCN in rat lung with allergic asthma. Twenty-four male Sprague-Dawley rats were allocated to normal control, asthma model, and dexamethasone group, respectively. Animals were actively sensitized by subcutaneous injection of ovalbumin (OVA) and challenged by an aerosol of 1% OVA 2 weeks after sensitization. Both UCN mRNA and peptide were expressed in normal rat lungs. Rats in asthma model group developed severe infiltration of inflammatory cells and inflammation in airway, together with a significantly up-regulated expression of urocortin mRNA detected by semi-quantitative reverse transcriptase-polymerase chain reaction and peptide measured both by immunohistochemistry and Western blot analysis. In contrast, treatment with dexamethasone resulted in markedly ameliorated airway inflammation and alleviated airway inflammatory cell infiltration, coupled with a significantly decreased urocortin expression. Regression analysis revealed a positive correlation between urocortin expression and the number of inflammatory cells in bronchoalveolar lavage fluid (P
- Published
- 2006
- Full Text
- View/download PDF
25. Understanding the determinants of CD8+ T cell immunodominance in acute HIV infection
- Author
-
Joanna A Warren, Yinyan Xu, Margaret Costanzo, Victoria Whale, Michael P.K. Liu, Rasmi Thomas, Sodsai Tovanabutra, Gustavo Kijak, Leigh Anne Eller, Morgane Rolland, Nelson L. Michael, Merlin L. Robb, Michael A. Eller, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
CD8+ T cells detect and clear virus-infected cells. During human immunodeficiency virus-1 (HIV-1) infection, different HIV-specific CD8+ T cell populations are expanded, each targeting a discreet epitope. These different HIV-specific CD8+ T cell populations do not arise equally, and instead establish a hierarchy (immunodominance) based on strength of T cell response to a given viral epitope. Our previous work has shown that relative immunodominance is a major determinant of the rate at which HIV can mutate to escape T cell immune pressure. The precise mechanism of how the first CD8+ T cell response to HIV is generated, and which factors determine T cell dominance is unclear. To investigate determinants of T cell immunodominance in acute HIV-1 infection (~45 days following infection), we determined all primary HIV-1 specific CD8+ T cell responses in 10 individuals in the RV217 early HIV cohort (ECHO). T cell responses were mapped against the unique infecting HIV virus/es in each individual using IFN-γ ELISpot. Across participants, the dominant T cell response mostly targeted variable regions of the HIV-1 genome. We detected a wide (4–13 epitopes) breadth of T cell response across our cohort that was independent of HLA haplotype, with all HIV-1 proteins targeted and different rates of virus escape observed. Ongoing studies are focusing on better understanding how factors, such as T cell receptor repertoire, avidity, antigen presentation and function contribute to T cell immunodominance, which in turn will inform HIV-1 vaccine design.
- Published
- 2017
- Full Text
- View/download PDF
26. Do T cells from HIV-infected individuals have normal metabolic function?
- Author
-
Genevieve Tyndale Clutton, Olivia Council, Yinyan Xu, Joanna Warren, De’Ashia Lee, Maria Abad Fernandez, Nancie Archin, JoAnn Kuruc, Joseph Eron, Cindy Gay, David Margolis, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
Untreated HIV infection is characterized by generalized immune activation, T cell dysfunction, and ultimately the exhaustion of HIV-specific CD8 T cell responses. Durable virus suppression resulting from anti-retroviral therapy (ART) is associated with greatly improved immune function, but some phenotypic and functional abnormalities persist even after years of suppressive ART. We have observed that, relative to HIV-seronegative individuals, CD8 T cells from HIV+ participants on ART are skewed toward a more effector-like (T-bethigh Eomeslow) phenotype and have significantly reduced proliferative capacity in response to antigenic stimulation. Metabolic pathways play a key role in shaping T cell maturation, phenotype, and function, and the ability to produce ATP via both glycolytic and oxidative phosphorylation (mitochondrial) pathways is critical to T cell function and longevity. However, whether underlying metabolic abnormalities contribute to residual CD8 T cell dysfunction in HIV+ individuals on ART has not been fully explored. Here we compare the metabolic phenotype of T cells from HIV+ participants on ART with those from HIV-seronegative participants. We are using MitoTracker® dyes to assess mitochondrial mass and polarization in T cell memory subsets, ex vivo and in response to antigen, and measuring expression of the glucose transporter GLUT1 and the mitochondrial biogenesis master regulator PGC-1α in different CD8 T cell effector subsets. Our findings will inform whether phenotypic and functional abnormalities in CD8 T cells from HIV+ individuals on ART reflect underlying metabolic dysfunction and if metabolic interventions could improve T cell function.
- Published
- 2017
- Full Text
- View/download PDF
27. Peripheral blood lymphocyte telomere length as a predictor of response to immunosuppressive therapy in childhood aplastic anemia
- Author
-
Xinan Wang, Etsuro Ito, Seiji Kojima, Hideki Muramatsu, Nobuhiro Nishio, Nao Yoshida, Kazuko Kudo, Hiromasa Yabe, Hirotoshi Sakaguchi, Akira Ohara, Atsushi Narita, Sayoko Doisaki, Yoshiyuki Takahashi, Hiroshi Moritake, Kazuhiro Nakamura, Asahito Hama, Nozomu Kawashima, Ryoji Kobayashi, Shouichi Ohga, and Yinyan Xu
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Anemia ,Gastroenterology ,Telomere Homeostasis ,Predictive Value of Tests ,Internal medicine ,medicine ,Humans ,Lymphocytes ,Aplastic anemia ,Child ,medicine.diagnostic_test ,business.industry ,Hazard ratio ,Anemia, Aplastic ,Infant ,Hematology ,Articles ,Telomere ,medicine.disease ,Confidence interval ,Treatment Outcome ,Predictive value of tests ,Peripheral blood lymphocyte ,Child, Preschool ,Immunology ,Female ,business ,Immunosuppressive Agents ,Fluorescence in situ hybridization - Abstract
Predicting the response to immunosuppressive therapy could provide useful information to help the clinician define treatment strategies for patients with aplastic anemia. In our current study, we evaluated the relationship between telomere length of lymphocytes at diagnosis and the response to immunosuppressive therapy in 64 children with aplastic anemia, using flow fluorescence in situ hybridization. Median age of patients was ten years (range 1.5-16.2 years). Severity of the disease was classified as very severe in 23, severe in 21, and moderate in 20 patients. All patients were enrolled in multicenter studies using antithymocyte globulin and cyclosporine. The response rate to immunosuppressive therapy at six months was 52% (33 of 64). The probability of 5-year failure-free survival and overall survival were 56% (95% confidence interval (CI): 41-69%) and 97% (95%CI: 87-99%), respectively. Median telomere length in responders was -0.4 standard deviation (SD) (-2.7 to +3.0 SD) and -1.5 SD (-4.0 to +1.6 (SD)) in non-responders (P
- Published
- 2014
28. Clinical course of juvenile myelomonocytic leukemia in the blast crisis phase treated by acute myeloid leukemia-oriented chemotherapy and allogeneic hematopoietic stem cell transplantation
- Author
-
Naoki Sakata, Xinan Wang, Seiji Kojima, Hideki Muramatsu, Satoshi Ueda, Yinyan Xu, Takeshi Higa, Hirotoshi Sakaguchi, Toshihiro Yamaguchi, and Tsukasa Takemura
- Subjects
Male ,medicine.medical_specialty ,Myeloid ,medicine.medical_treatment ,Hepatosplenomegaly ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Hematopoietic stem cell transplantation ,Bone Marrow ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Transplantation, Homologous ,Hematology ,Juvenile myelomonocytic leukemia ,business.industry ,Remission Induction ,Acute erythroid leukemia ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,medicine.disease ,PTPN11 ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,Leukemia, Myelomonocytic, Juvenile ,Child, Preschool ,Immunology ,Mutation ,medicine.symptom ,business ,Blast Crisis - Abstract
Juvenile myelomonocytic leukemia (JMML) is a mixed myeloproliferative and myelodysplastic disorder that occurs in early childhood. The clinical course of JMML is highly variable. A third of patients follow a relatively indolent course, although approximately 15 % cases are thought to develop acute myeloid leukemia, referred to as blast crisis. The etiology and clinical characteristics of blast crisis remain unclear. We document the case of a 27-month-old boy who presented with hepatosplenomegaly, skin rash, and lymphadenopathy. An initial diagnosis of acute erythroid leukemia was made according to the French–American–British classification. Following estimation of hypersensitivity to GM-CSF and genetic analysis of PTPN11, he was diagnosed with JMML in the blast crisis phase. Although he had several poor prognostic factors, including monosomy 7 and high HbF percentage, he achieved partial remission after treatment with acute myeloid leukemia-oriented chemotherapy followed by allogeneic hematopoietic stem cell transplantation. He has been in complete remission for over 6 years.
- Published
- 2013
29. Effects of different treatment regimens on T cell function in acute HIV infection
- Author
-
Genevieve Tyndale Clutton, Yinyan Xu, Maria Abad Fernandez, Amanda Crooks, Blair Turner, Nicole Maponga, Anna Cope, Susan Fiscus, Kara McGee, Mehri McKellar, JoAnn Kuruc, Guido Ferrari, David Margolis, Charles Hicks, Joseph Eron, Cindy Gay, and Nilu Goonetilleke
- Subjects
Immunology ,Immunology and Allergy - Abstract
In HIV-infected individuals, anti-retroviral therapy (ART) is effective in maintaining low viral loads, restoring CD4+ T cell counts, and prolonging life. However, chronic inflammation and immune dysfunction persist even after years of successful ART. Previous studies have suggested that early initiation of ART may result in improved immune reconstitution, but whether more rapid suppression of viral load during early treatment also improves immunological outcomes is unknown. In this study we compare the effects of two regimens initiated during acute infection on T cell phenotype and function. Regimen 1 (n=30) is a non-nucleoside reverse transcriptase inhibitor (NNRTI)-based regimen and Regimen 2 (n=30) combines an integrase strand transfer inhibitor (INSTI) with nucleoside reverse transcriptase inhibitors (NRTIs). NRTIs and NNRTIs inhibit reverse transcription of viral RNA to DNA in the infected cell, while INSTIs block integration of viral DNA into the host genome. Since Regimen 2 targets multiple stages of the viral life cycle, we hypothesize that more rapid viral suppression may be achieved with this treatment schedule. In a cross-sectional study performed 96 weeks after treatment initiation, we are comparing the effects of the two regimens on T cell activation (CD25, CD69, CD38, HLA-DR), exhaustion (PD-1, Tim-3, CD160, T-bet, Eomes), and proliferation using standardized flow cytometry panels. This study will provide valuable insights into the relationship between viral decay kinetics and immune preservation during acute infection, informing the design of future ART regimens.
- Published
- 2016
- Full Text
- View/download PDF
30. Lack of CD4⁺CD25⁺FOXP3⁺ regulatory T cells is associated with resistance to intravenous immunoglobulin therapy in patients with Kawasaki disease
- Author
-
Asahito Hama, Seiji Kojima, Itzel Bustos Villalobos, Taichi Kato, Kazuyuki Akane, Yinyan Xu, Yusuke Okuno, Hideki Muramatsu, Yu Hirabayashi, Shinji Hasegawa, and Yoshiyuki Takahashi
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Drug Resistance ,chemical and pharmacologic phenomena ,Coronary Artery Disease ,Mucocutaneous Lymph Node Syndrome ,Peripheral blood mononuclear cell ,Gastroenterology ,T-Lymphocytes, Regulatory ,Flow cytometry ,Young Adult ,Intravenous Immunoglobulin Therapy ,Internal medicine ,medicine ,Humans ,Immunologic Factors ,IL-2 receptor ,Child ,biology ,medicine.diagnostic_test ,business.industry ,Case-control study ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Immunoglobulins, Intravenous ,Infant ,hemic and immune systems ,Forkhead Transcription Factors ,Middle Aged ,medicine.disease ,Flow Cytometry ,CD4 Lymphocyte Count ,Case-Control Studies ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Immunology ,CD4 Antigens ,biology.protein ,Kawasaki disease ,Female ,Antibody ,business ,Biomarkers - Abstract
The aim of this study was to investigate changes in CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) throughout the clinical course of Kawasaki disease (KD) and correlations with response to intravenous immunoglobulin (IVIg) therapy. Participants comprised 18 patients who fulfilled the diagnostic criteria for KD and 20 healthy subjects. Expressions of CD25 and FOXP3 among all CD4(+) T cells in peripheral blood mononuclear cells were analyzed by flow cytometry before and 7 and 30 days after IVIg therapy. Before treatment, percentages of CD4(+)CD25(+)FOXP3(+) Tregs among total CD4(+) Tregs were significantly lower among KD patients (4.19 %; range, 0.16-8.11 %) than among healthy subjects (7.32 %; 4.18-13.42 %; P = 0.0001). Both percentages and absolute numbers of CD4(+)CD25(+)FOXP3(+) Tregs on day 7 after IVIg therapy were significantly increased compared with values before treatment (8.02 % (range, 0.51-12.6 %) vs. 4.19 % (range, 0.16-8.11 %), P = 0.0005; 93.25/ μL (range, 6.67-258.05) vs. 41.85/ μL (range, 0.44-160.62), P 0.0001, respectively). Moreover, percentages and absolute numbers of CD4(+)CD25(+)FOXP3(+) Tregs before treatment were significantly lower in the IVIg-resistant group than in the IVIg-sensitive group (0.18 % (range, 0.16-3.34 %) vs. 4.52 % (range, 2.8-8.11 %), P = 0.0022; 0.68/μL (range, 0.44-53.81) vs. 51.66/μL (range, 2.88-160.62), P = 0.0098, respectively). The frequency of CD4(+)CD25(+)FOXP3(+) Tregs in four of the five IVIg-resistant patients at diagnosis was more than 3 standard deviations below that in healthy subjects. Two of these four patients displayed coronary abnormalities, and one of these two patients developed coronary aneurysm.Lack of CD4(+)CD25(+)FOXP3(+) Tregs before treatment may predict resistance to IVIg therapy in patients with KD.
- Published
- 2012
31. Somatic mosaicism for oncogenic NRAS mutations in juvenile myelomonocytic leukemia
- Author
-
Xinan Wang, Masanori Sato, Yasuhide Hayashi, Akira Shimada, Hideki Muramatsu, Nao Yoshida, Makito Tanaka, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, Hitoshi Kiyoi, Seiji Kojima, Hiroyuki Kawaguchi, Kiyofumi Yamada, Atsushi Narita, Sayoko Doisaki, Makiko Mori-Ezaki, Manabu Sotomatsu, Hideaki Hoshino, Akitoshi Kinoshita, Kenichi Koike, Yoko Furukawa-Hibi, Hirotoshi Sakaguchi, Yinyan Xu, and Olfat Ismael
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Male ,Somatic cell ,medicine.medical_treatment ,Immunology ,DNA Mutational Analysis ,Molecular Sequence Data ,Hematopoietic stem cell transplantation ,Biology ,medicine.disease_cause ,Biochemistry ,Myeloid Neoplasm ,medicine ,Humans ,Child ,Juvenile myelomonocytic leukemia ,Base Sequence ,Mosaicism ,Infant ,Cell Biology ,Hematology ,Oncogenes ,medicine.disease ,PTPN11 ,Leukemia ,Leukemia, Myelomonocytic, Juvenile ,Mutation ,ras Proteins ,KRAS - Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myeloid neoplasm characterized by excessive proliferation of myelomonocytic cells. Somatic mutations in genes involved in GM-CSF signal transduction, such as NRAS, KRAS, PTPN11, NF1, and CBL, have been identified in more than 70% of children with JMML. In the present study, we report 2 patients with somatic mosaicism for oncogenic NRAS mutations (G12D and G12S) associated with the development of JMML. The mutated allele frequencies quantified by pyrosequencing were various and ranged from 3%-50% in BM and other somatic cells (ie, buccal smear cells, hair bulbs, or nails). Both patients experienced spontaneous improvement of clinical symptoms and leukocytosis due to JMML without hematopoietic stem cell transplantation. These patients are the first reported to have somatic mosaicism for oncogenic NRAS mutations. The clinical course of these patients suggests that NRAS mosaicism may be associated with a mild disease phenotype in JMML.
- Published
- 2012
32. CTLA-4 blockade increases antigen-specific CD8(+) T cells in prevaccinated patients with melanoma: three cases
- Author
-
David B. Page, Achim A. Jungbluth, Sylvia Adams, Yinyan Xu, Lloyd J. Old, Humilidad F. Gallardo, Klaus J. Busam, James P. Allison, Yanyun Li, Nina Bhardwaj, Jedd D. Wolchok, Jianda Yuan, Brian A. Ginsberg, and Teresa S. Rasalan
- Subjects
Adult ,Male ,Cancer Research ,Skin Neoplasms ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Ipilimumab ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,complex mixtures ,Cancer Vaccines ,Article ,Immune system ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,CTLA-4 Antigen ,Neoplasm Metastasis ,Melanoma ,Cells, Cultured ,Aged ,Immunogenicity ,Antibodies, Monoclonal ,Immunotherapy ,Middle Aged ,medicine.disease ,Peptide Fragments ,Oncology ,CTLA-4 ,Cytokines ,Female ,Tumor Escape ,medicine.drug - Abstract
Anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4) antibodies, such as ipilimumab, have generated measurable immune responses to Melan-A, NY-ESO-1, and gp100 antigens in metastatic melanoma. Vaccination against such targets has potential for immunogenicity and may produce an effector-memory T-cell response.To determine the effect of CTLA-4 blockade on antigen-specific responses following vaccination, in-depth immune monitoring was performed on three ipilimumab-treated patients prevaccinated with gp100 DNA (IMF-24), gp100(209-217) and tyrosinase peptides plus GM-CSF DNA (IMF-32), or NY-ESO-1 protein plus imiquimod (IMF-11); peripheral blood mononuclear cells were analyzed by tetramer and/or intracellular cytokine staining following 10-day culture with HLA-A*0201-restricted gp100(209-217) (ITDQVPFSV), tyrosinase(369-377) (YMDGTMSQV), or 20-mer NY-ESO-1 overlapping peptides, respectively. Tumors from IMF-32 were analyzed by immunohistochemistry to help elucidate mechanism(s) underlying tumor escape.Following vaccination, patients generated weak to no CD4(+) or CD8(+) T-cell response specific to the vaccine antigen but demonstrated increases in effector-memory (CCR7(lo)CD45RA(lo)) tetramer(+)CD8(+) T cells. After ipilimumab induction, patients experienced a robust, although sometimes transient, antigen-specific response for gp100 (IMF-32 and IMF-24) or NY-ESO-1 (IMF-11) and produced polyfunctional intracellular cytokines. Primary and metastatic tumors expressed tyrosinase but not gp100 or class I/II MHC molecules.Vaccination induced a measurable antigen-specific T-cell response that increased following CTLA-4 blockade, potentially "boosting" the vaccine-primed response. Tumor escape may be related to antigen loss or lack of MHC expression necessary for immune activity. These results in a limited number of patients support the need for further research into combining vaccination with ipilimumab and provide insight into mechanisms underlying tumor escape.
- Published
- 2011
33. Aberrant phosphorylation of STAT5 by granulocyte-macrophage colony-stimulating factor in infant cytomegalovirus infection mimicking juvenile myelomonocytic leukemia
- Author
-
Makito Tanaka, Yinyan Xu, Seiji Kojima, Nao Yoshida, Nobuhiro Nishio, Asahito Hama, Akira Shimada, Sayoko Doisaki, Yoshiyuki Takahashi, Hirotoshi Sakaguchi, and Hideki Muramatsu
- Subjects
Male ,Cancer Research ,Congenital cytomegalovirus infection ,Efficiency ,Biology ,Peripheral blood mononuclear cell ,Sensitivity and Specificity ,Flow cytometry ,Diagnosis, Differential ,medicine ,STAT5 Transcription Factor ,Humans ,Progenitor cell ,Phosphorylation ,medicine.diagnostic_test ,Juvenile myelomonocytic leukemia ,Granulocyte-Macrophage Colony-Stimulating Factor ,Infant ,Hematology ,medicine.disease ,Flow Cytometry ,Leukemia ,Granulocyte macrophage colony-stimulating factor ,Oncology ,Leukemia, Myelomonocytic, Juvenile ,Child, Preschool ,Immunology ,Cytomegalovirus Infections ,biology.protein ,Female ,Antibody ,medicine.drug - Abstract
Juvenile myelomonocytic leukemia (JMML) progenitor cells exhibit in vitro hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF). Phospho-specific flow cytometry using anti-phosphorylated STAT5 antibody is a new method recently reported to detect GM-CSF hypersensitivity of cells. However, colony assays using methylcellulose medium to measure GM-CSF-hypersensitivity remain as the current gold standard. Interestingly, cytomegalovirus (CMV) infection in infancy often presents with a variety of clinical symptoms that mimic JMML, with CMV giving a positive result by colony assay. We wanted to determine whether aberrant STAT5 activation occurs in CMV infection by using phospho-specific flow cytometry, and to ascertain whether this method is effective at discriminating CMV infection from JMML. Peripheral blood mononuclear cells from patients with JMML and CMV infection displayed an elevated proportion of p-STAT5 cells after low-dose GM-CSF stimulation when compared with cells from normal individuals. However, we found no significant differences in the percentage of p-STAT5 positive cells from patients with CMV infection and JMML at any doses of the GM-CSF doses used. We conclude that patients with CMV infection cannot be discriminated from patients with JMML by this new diagnostic method.
- Published
- 2010
34. Optimization and validation of a robust human T-cell culture method for monitoring phenotypic and polyfunctional antigen-specific CD4 and CD8 T-cell responses
- Author
-
Jedd D. Wolchok, Lloyd J. Old, Yun Lin, Yinyan Xu, Teresa S. Rasalan, Humilidad F. Gallardo, Stephanie L. Terzulli, Jianda Yuan, James P. Allison, Gregor Manukian, Alan N. Houghton, Hao Li, and Geoffrey Y. Ku
- Subjects
CD4-Positive T-Lymphocytes ,Cancer Research ,T cell ,Immunology ,Cell Culture Techniques ,Computational biology ,Biology ,CD8-Positive T-Lymphocytes ,Cancer Vaccines ,Article ,Immune system ,Antigen ,Antigen specific ,Antigens, CD ,Neoplasms ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,Antigens ,Genetics (clinical) ,Cells, Cultured ,Transplantation ,Cancer ,Cell Biology ,medicine.disease ,Phenotype ,Antigens, Differentiation ,medicine.anatomical_structure ,Oncology ,Cell culture ,Cytokines ,Feasibility Studies - Abstract
Monitoring cellular immune responses is one prerequisite for the rational development of cancer vaccines.We describe an extensive effort to optimize and validate quantitatively an in vitro T-cell culture method by determining the phenotype and function of both CD4(+) and CD8(+) T cells, including measurement of the phenotype markers CCR7, CD45RA, CD28 and CD27 and the functional markers interferon (IFN)-gamma, interleukin (IL)-2, macrophage inflammatory protein (MIP)-1beta, tumor necrosis factor (TNF)-alpha and CD107a.Autologous peripheral blood mononuclear cells (PBMC) were potent stimulators that expanded antigen (Ag)-specific CD8(+) T cells during short-term culture with the addition of IL-2 and IL-15 cytokines. Polyfunctional Ag-specific CD4(+) and CD8(+) T cells were detectable using this method.Our culture system represents a robust human T-cell culture protocol that permits phenotypic, quantitative and qualitative evaluation of vaccine-induced CD4(+) and CD8(+) T-cell responses.
- Published
- 2009
35. Aberrant DNA Methylation Is Associated with a Poor Outcome in Juvenile Myelomonocytic Leukemia
- Author
-
Hideki Muramatsu, Nao Yoshida, Seishi Ogawa, Xinan Wang, Yinyan Xu, Kenichi Yoshida, Sayoko Doisaki, Asahito Hama, Kiyofumi Yamada, Hideki Makishima, Yoshiyuki Takahashi, Atsushi Narita, Yuichi Shiraishi, Jaroslaw P. Maciejewski, Hirotoshi Sakaguchi, Yusuke Okuno, Seiji Kojima, Yoko Furukawa-Hibi, and Satoru Miyano
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Adolescent ,Bisulfite sequencing ,lcsh:Medicine ,Biology ,Cohort Studies ,CDKN2A ,Internal medicine ,medicine ,Humans ,Epigenetics ,Child ,lcsh:Science ,Genetic Association Studies ,Myeloproliferative neoplasm ,Survival analysis ,Multidisciplinary ,Juvenile myelomonocytic leukemia ,lcsh:R ,Infant ,DNA Methylation ,medicine.disease ,Survival Analysis ,Leukemia ,Treatment Outcome ,Leukemia, Myelomonocytic, Juvenile ,Child, Preschool ,Multivariate Analysis ,Mutation ,DNA methylation ,Immunology ,Female ,lcsh:Q ,Research Article - Abstract
Juvenile myelomonocytic leukemia (JMML), an overlap of myelodysplastic / myeloproliferative neoplasm, is an intractable pediatric myeloid neoplasm. Epigenetic regulation of transcription, particularly by CpG methylation, plays an important role in tumor progression, mainly by repressing tumor-suppressor genes. To clarify the clinical importance of aberrant DNA methylation, we studied the hypermethylation status of 16 target genes in the genomes of 92 patients with JMML by bisulfite conversion and the pryosequencing technique. Among 16 candidate genes, BMP4, CALCA, CDKN2A, and RARB exhibited significant hypermethylation in 72% (67/92) of patients. Based on the number of hypermethylated genes, patients were stratified into three cohorts based on an aberrant methylation score (AMS) of 0, 1–2, or 3–4. In the AMS 0 cohort, the 5-year overall survival (OS) and transplantation-free survival (TFS) were good (69% and 76%, respectively). In the AMS 1–2 cohort, the 5-year OS was comparable to that in the AMS 0 cohort (68%), whereas TFS was poor (6%). In the AMS 3–4 cohort, 5-year OS and TFS were markedly low (8% and 0%, respectively). Epigenetic analysis provides helpful information for clinicians to select treatment strategies for patients with JMML. For patients with AMS 3–4 in whom hematopoietic stem cell transplantation does not improve the prognosis, alternative therapies, including DNA methyltransferase inhibitors and new molecular-targeting agents, should be established as treatment options.
- Published
- 2015
- Full Text
- View/download PDF
36. P-145 Sequential gain of SETBP1 mutations in severe aplastic anemia evolving into acute myeloid leukemia with monosomy 7
- Author
-
Kenichi Yoshida, Seiji Kojima, Yinyan Xu, Yoshiyuki Takahashi, Akira Shimada, Hideki Muramatsu, Hirotoshi Sakaguchi, Yusuke Okuno, Seishi Ogawa, and Asahito Hama
- Subjects
Chromosome 7 (human) ,Cancer Research ,Oncology ,business.industry ,Immunology ,Myeloid leukemia ,Medicine ,Hematology ,business ,Severe Aplastic Anemia - Published
- 2013
- Full Text
- View/download PDF
37. Predicting Response to Immunosuppressive Therapy By the Combination of Minor Paroxysmal Nocturnal Hemoglobinuria Clones and Lymphocyte Telomere Length in Children with Aplastic Anemia
- Author
-
Xinan Wang, Hideki Muramatsu, Michi Kamei, Yuko Sekiya, Masahiro Irie, Yoshiyuki Takahashi, Asahito Hama, Nozomu Kawashima, Atsushi Narita, Seiji Kojima, Nobuhiro Nishio, Hirotoshi Sakaguchi, Yusuke Okuno, Yinyan Xu, Nao Yoshida, and Sayoko Doisaki
- Subjects
medicine.medical_specialty ,education.field_of_study ,Univariate analysis ,Predictive marker ,Thymoglobulin ,business.industry ,Lymphocyte ,Immunology ,Population ,Salvage therapy ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,medicine.anatomical_structure ,Internal medicine ,Paroxysmal nocturnal hemoglobinuria ,medicine ,Aplastic anemia ,education ,business - Abstract
Introduction: Acquired aplastic anemia (AA) is an immune mediated disease characterized by severe quantitative defects in stem cell number resulting in a hypocellular marrow and peripheral blood cytopenias. Immunosuppressive therapy (IST) using anti-thymocyte globulin (ATG) and cyclosporine A (CyA) is the standard therapy for patients with AA who lack a human leukocyte antigen (HLA)-matched sibling donor. Previous studies identified minor paroxysmal nocturnal hemoglobinuria (PNH) clones and short telomere length (TL) as predictive markers of the response to IST. However, the likelihood ratio of these markers was insufficient to influence clinical decisions. Here we assessed whether the combination of these markers could more accurately predict the response to IST. Methods: Between July 2001 and November 2013, 116 patients (65 boys and 51 girls) diagnosed with AA were examined to identify a minor population of PNH-type cells and TL of peripheral blood lymphocytes. Fifty-three patients received horse ATG (Lymphoglobulin, 15mg/kg/day for 5 days) and 63 received rabbit ATG (Thymoglobulin, 2.5−3.75 mg/kg/day for 5 days). CyA (6mg/kg/day) was started on day 1 and continued to at least day 180. The dose of CyA was adjusted to maintain trough levels between 100 and 200 ng/ml. Written informed consent was obtained from all patients’ parents. The ethics committee of each participating hospital approved the study. We used a flow cytometric assay to detect PNH-type granulocytes and red blood cells (RBCs). Presence of >0.007% CD13+ CD55− CD59− granulocytes and/or >0.009% glycophorin A+ CD55− CD59−RBCs was defined as PNH positive (PNH+), based on the results from healthy individuals. The average relative TL of peripheral lymphocytes was measured by flow-fluorescence in situ hybridization (flow-FISH) using a Telomere PNA Kit (Dako Cytomation). Based on our previous study, we defined −1.0 SD of age-adjusted controls as the cut-off value. Results: The median age at IST was 9.8 years (0.9−16.0). Disease severity of patients was assessed as moderate in 39, severe in 37, and very severe in 40, respectively. The causes of AA were idiopathic in 104 patients and hepatitis-associated in 12 patients. The median follow-up period from the time of IST was 36 months (range 1−134). Thirty-nine patients (34%) carried a minor PNH clone at diagnosis. Compared with healthy individuals, their median of TL was −1.08 SD (−4.01 to +3.01 SD). Overall, 60 of 116 patients (51.7%) responded to IST at 6 months after administration of ATG. In univariate analysis, the response rate at 6 months was higher in PNH+ patients than in PNH- patients (74.4% vs. 40.3%, respectively, p = 0.001). Longer TLs (≥−1.0 SD of age-adjusted control) were also associated with a favorable response to IST at 6 months (73.2% vs. 31.7%, p Conclusion: Combination of the absence of a minor PNH clone and a short TL was an efficient predictor of poor response to IST. These patients had low chances to respond to IST, which challenges the current standard of treatment. Prospective clinical trials are required to confirm these findings. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
38. Whole-Exome Sequencing Reveals a Paucity of Somatic Gene Mutations in Aplastic Anemia and Refractory Cytopenia of Childhood
- Author
-
Yoshiyuki Takahashi, Atsushi Narita, Hirotoshi Sakaguchi, Yusuke Okuno, Hideki Muramatsu, Seishi Ogawa, Hiroko Tanaka, Xinan Wang, Asahito Hama, Nozomu Kawashima, Kenichi Yoshida, Seiji Kojima, Kenichi Chiba, Sayoko Doisaki, Yuichi Shiraishi, Satoru Miyano, and Yinyan Xu
- Subjects
Oncology ,Cytopenia ,medicine.medical_specialty ,Pathology ,Myelodysplastic syndromes ,Immunology ,Cell Biology ,Hematology ,Biology ,Gene mutation ,medicine.disease ,Biochemistry ,Somatic evolution in cancer ,Germline mutation ,Internal medicine ,medicine ,Refractory cytopenia with multilineage dysplasia ,Exome ,Exome sequencing - Abstract
Introduction: The appropriate classification of bone marrow (BM) failure syndromes in children is challenging, particularly in relation to histological distinction between aplastic anemia (AA), refractory cytopenia of childhood (RCC), and refractory cytopenia with multilineage dysplasia (RCMD). The goal of this study is to characterize the molecular pathogenesis of these conditions by identifying the full spectrum of gene mutations in 29 children with three diseases using whole-exome sequencing. Patients and Methods: Wediagnosed AA, RCC, or RCMD on the basis of morphology and histological findings of bone marrow (BM) according to the 2008 World Health Organization (WHO) classification criteria. Patients with AA exhibited hypocellular BM and no morphologically dysplastic changes in any of three hematopoietic cell lineages, while patients with RCC had 10% in one cell lineage. Patients with RCMD exhibited >10% dysplastic changes in two or more cell lineages. We obtained peripheral blood and BM samples from 29 children (16 boys and 13 girls) with AA (n = 8), RCC (n = 11), or RCMD (n = 10). The median age at diagnosis was 11 years (range, 2–15 years). We performed exome capture from paired DNA (non-T cells/CD3+ lymphocytes) using SureSelect® Human All Exon V4 kit (Agilent Technologies, Santa Clara, CA), which covered all part of the coding exons, followed by massively-parallel sequencing using HiSeq 2000 (Illumina, San Diego, CA) according to the manufacturer’s protocol. Candidate somatic mutations and germline variants were detected through our pipeline for whole-exome sequencing (Genomon-exome). All candidate somatic nucleotide changes were validated by Sanger sequencing. The ethics committee of Nagoya University Graduate School of Medicine approved this study. Results: Whole-exome sequencing pipeline identified a total of 14 non-synonymous somatic (one nonsense, 11 missense, and two frameshift) changes among the 29 patients, which resulted in only 0.48 mutations per patient. The average numbers of somatic mutations per sample were not significantly different among these groups (0.50 in AA, 0.36 in RCC, and 0.60 in RCMD). As a whole, childhood AA, RCC, and RCMD were characterized by a paucity of somatic mutations compared with adult myelodysplastic syndromes (MDS) in which 10 or more mutations per exome were detected on average. Among the mutated genes, BCOR-inactivating mutations in two patients (p.S158fs in AA and p.E1286X in RCMD) were considered significant genetic events based on previous reports that it is a driver gene in MDS. With regard to germline events, we did not detect any germline mutations of inherited BM failure syndromes. Moreover, we did not identifiy significantly frequent germline events in the entire cohort or any genetic hallmarks to be able to discriminate between these three diseases. When comparing the clinical outcomes of patients with somatic mutations (n = 7) versus those without somatic mutations (n = 22), response rate to immunosuppressive therapy at 6 months (50% vs. 50%), 5-year clonal evolution rate (95% confidential interval) [0% (0%) vs. 6% (0%–26%)], and the 5-year overall survival rate (95% confidential interval) [100% (100%–100%) vs. 95% (70%–99%)] were not significantly different. Conclusion: We usedwhole-exome sequencing analysis for gene mutational profiling of children with AA, RCC, and RCMD. Idiopathic bone marrow failure syndromes in children are characterized by a paucity of somatic gene mutations, irrespective of histological diagnosis. These findings suggest that histological diagnosis based on the WHO classification system does not discriminate the mutational profile of idiopathic BM failure syndromes in children. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
39. Generation of Cell Lines Harboring SETBP1 Mutations By the Crispr/Cas9 System
- Author
-
Hideki Muramatsu, Michi Kamei, Yusuke Okuno, Yinyan Xu, Yoshiyuki Takahashi, Sayoko Doisaki, Xinan Wang, Seiji Kojima, Yuko Sekiya, Atsushi Narita, Masahiro Irie, Asahito Hama, and Nozomu Kawashima
- Subjects
Silent mutation ,Cell growth ,Somatic cell ,Point mutation ,Immunology ,Mutant ,HEK 293 cells ,Cell Biology ,Hematology ,Transfection ,Cell sorting ,Biology ,Biochemistry ,Molecular biology - Abstract
Introduction Recent advances in cancer genetics have led to the identification of somatic mutations in SET-binding protein 1 (SETBP1) in myeloid malignancies categorized as myeloproliferative neoplasm (MPN) and myelodysplastic syndromes (MDS). Heterozygous point mutations in SETBP1 are essentially found at a genomic level in myeloid malignancies, and the frequency of the mutated allele in cDNA suggests somatic heterozygosity without substantial imbalance in allelic expression. Thus, mutant SETBP1 presumably has a dominant altered biological activity. Most mutations in SETBP1 are located in the SKI homologous region. This region is suggested to include regions critical for ubiquitin binding and SETBP1 degradation. SETBP1 binds to SET, which protects against protease cleavage, and thus may result in PP2A inhibition and cell proliferation. Overexpression of SETBP1 resulting from a p.G870S alteration showed higher levels of the protein compared with wild-type (WT), indicating a prolonged halftime of SETBP1, which led to reduced PP2A activity and a higher cell proliferation rate. To date, however, our molecular biological understanding of SETBP1 mutations has been obtained only through observations of exogenous overexpression in cell lines. This may result in bias, considering the predicted dominant-negative function of SETBP1 mutations. Therefore, we used an RNA-guided endonuclease (RGEN), the CRISPR/Cas9 system, to generate a cell line harboring point mutations resulting in only relevant single amino acid substitutions in SETBP1. We analyzed cell signaling using the cell line thus established. Methods pSpCas9(BB) (PX330) was used to express humanized S. pyogenes Cas9 and gRNAs of interest. The gRNAs were designed by searching for NGG protospacer adjacent motif (PAM) sequences near the point mutation target sites. The candidate gRNAs were gRNA#1, 5′-TAGGGAGCCAATCTCGCAC-3′; gRNA#2, 5′-TGTCCCAATGCCGCTGTCGC-3′; gRNA#4, 5′-GTCCCAATGCCGCTGTCGCT-3′; and gRNA#7, 5′-GAGACGATCCCCAGCGACAG-3′. pCAG-EGxxFP harboring the 500 bp target region of WT SETBP1 was constructed for gRNA selection. For homology-dependent repair (HDR), we synthesized 70 mer single-stranded oligonucleotides (ssODN) having both the SETBP1 c.2602G>A, p.D868N mutation and synonymous mutation in the PAM. HEK293T cells were cultured in DMEM with 10% FBS. For cell signaling analysis, the cells were serum-depleted for 16 h prior to western blotting. Anti-SETBP1 antibody (ab98222), anti-phospho-Y307 PP2A antibody (E155), and anti phospho-p44/42 MAPK antibody (CST#4370) were used for cell signaling analysis. Results To validate an efficient sgRNA for DNA scission, we cotransfected pCAG-EGxxFP-SETBP1 and pSpCas9(BB)-SETBP1-gRNA plasmids into HEK293T cells. EGFP fluorescence, whose intensity is correlated with the efficacy of HDR, was observed 48 h later, and we determined that gRNA#2 was the most efficient. Next we cotransfected 293T cells with pCAG-EGxxFP-SETBP1, pSpCas9(BB)-SETBP1-gRNA#2, and ssODN for mutagenesis. Five days after transfection, single EGFP-positive clones were isolated using the FACSAria cell sorting system. Sanger sequencing confirmed that 293T cells harboring the SETBP1 p.D868N homozygous mutation were established. A clone with WT SETBP1 was also maintained as a control. To elucidate the effects of the SETBP1 mutation in 293T cells, we performed cell signaling analysis by western blotting. 293T-SETBP1 p.D868N cells showed higher levels of SETBP1 protein with lower molecular weight compared with WT, indicating a prolonged halftime, possibly due to loss of ubiquitination. In addition, 293T-SETBP1 p.D868N cells showed a higher phosphorylation level of PP2A (Y307, C subunit), a marker of PP2A inactivation. Finally, the phosphorylation level of p44/42 MAPK (ERK1/2) was increased in 293T-SETBP1 p.D868N cells. Conclusions We confirmed that the SETBP1 p.D868N mutation caused a prolonged halftime, resulting in PP2A inactivation and p44/42 MAPK activation in 293T cell lines. Our data suggest a potential therapy target for malignancies harboring SETBP1 mutations. More generally, this work illustrates the utility of RGEN technology for studying hematological malignancies. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
40. Diagnostic Efficacy of Whole-Exome Sequencing in 250 Patients with Congenital Bone Marrow Failure
- Author
-
Satoru Miyano, Yinyan Xu, Kenichi Yoshida, Kenichi Chiba, Xinan Wang, Seiji Kojima, Sayoko Doisaki, Hirotoshi Sakaguchi, Yusuke Okuno, Masashi Sanada, Hideki Muramatsu, Yoshiyuki Takahashi, Seishi Ogawa, Atsushi Narita, Asahito Hama, Nozomu Kawashima, Yuichi Shiraishi, and Hiroko Tanaka
- Subjects
Sanger sequencing ,Massive parallel sequencing ,medicine.diagnostic_test ,business.industry ,Immunology ,Cell Biology ,Hematology ,Gene mutation ,medicine.disease ,Bioinformatics ,Biochemistry ,FANCB ,symbols.namesake ,Fanconi anemia ,medicine ,symbols ,business ,Exome ,Exome sequencing ,Genetic testing - Abstract
Introduction: Congenital bone marrow failure syndromes (CBMFSs) are a heterogeneous class of diseases with overlapping phenotypes. Therefore, a precise and comprehensive genetic diagnostic system is strongly warranted to arrive at appropriate clinical decisions to avoid ineffective therapies and/or lethal complications of allogeneic hematopoietic stem cell transplantation. However, a large panel of newly identified causative genes of CBMFSs have been identified in recent years; therefore, it is virtually impossible to establish a routine genetic diagnostic test using conventional Sanger sequencing. Whole-exome sequencing (WES) is a promising solution for the diagnosis of inherited diseases because it tests virtually all genes simultaneously. For the introduction of WES into clinical practice, it is necessary to clarify whether this technique has superior diagnostic efficacy to conventional clinical genetic tests. Methods: We performed WES in 250 patients with CBMFSs lacking genetic diagnoses. Exome capture was performed using the SureSelect® Human All Exon V3–5 kit (Agilent Technologies, Santa Clara, CA, USA), which covers all known coding exons, followed by massively parallel sequencing using the HiSeq 2000 Sequencing System (Illumina, San Diego, CA, USA). Our established pipeline for WES (genomon: http://genomon.hgc.jp/exome/) detected >20,000 candidate variants per patient. Diagnoses were based on variants of 130 genes with pathogenicities confirmed by published studies. Results: Genetic diagnoses were possible in 68 patients (27%). The best efficacy was achieved in patients with Fanconi anemia [35/73, 48%; FANCG (n = 17), FANCA (n = 14), FANCB (n = 1), FANCF (n = 1), SLX4 (n = 1), and BRCA2 (n = 1)], although Sanger sequencing was not applied because of the large sizes of its causative genes. Encouraging results were obtained in patients with Diamond–Blackfan anemia [11/ 61, 18%; RPS26 (n = 3), RPS7 (n = 2), RPS19 (n = 2), RPL5 (n = 2), RPL35A (n = 1), and RPL11 (n = 1)] and dyskeratosis congenita [7/29, 24%; TERT (n = 3), TINF2 (n = 2), and DKC1 (n = 2)]. Five genetic diagnoses (7%) were inconsistent with clinical diagnoses, possibly because of overlapping disease phenotypes. Conclusion: Relative to conventional genetic testing, WES was found to be effective for the diagnoses of CBMFSs. Furthermore, the efficacy of WES will increase as our knowledge of gene mutations expands. In conclusion, the use of WES in clinical practice is warranted. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
41. Aberrant DNA Methylation Is Associated With Poor Outcomes In Juvenile Myelomonocytic Leukemia
- Author
-
Yinyan Xu, Kiyofumi Yamada, Nao Yoshida, Xinan Wang, Yoko Hibi, Atsushi Narita, Jaroslaw P. Maciejewski, Sayoko Doisaki, Asahito Hama, Nozomu Kawashima, Seiji Kojima, Hideki Makishima, Hideki Muramatsu, Hirotoshi Sakaguchi, and Yoshiyuki Takahashi
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Juvenile myelomonocytic leukemia ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,medicine.anatomical_structure ,CDKN2A ,Internal medicine ,DNA methylation ,medicine ,Hemoglobin F ,business ,Myeloproliferative neoplasm - Abstract
Recent studies suggest that aberrant methylation plays a fundamental role in the development of a variety of cancers, including myeloid malignancies. Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloid neoplasm of early childhood that is characterized by both excessive proliferation of myelomonocytic cells and hypersensitivity to granulocyte-macrophage colony-stimulating factor. It is categorized as an overlap myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) according to the World Health Organization classification. We recently reported that somatic mutations in SETBP1 and JAK3 were identified in JMML patients and were associated with poor outcomes (Nat Genet 2013;45:937–41). The goal of this study was to clarify the clinical significance of aberrant DNA methylation in JMML. We studied 92 children (61 boys and 31 girls) who were diagnosed with JMML in institutions throughout Japan. A diagnosis of JMML was made based on internationally accepted criteria. We quantitatively evaluated the CpG methylation pattern in the promoter regions of 16 candidate genes (APC, BMP4, CALCA, CDH13, CDKN2A, CDKN2B, CHFR, DAPK, DMR-H19, ER, IGF2, MGMT, MLH1, RARB, RASSF1, TP73) from genomic DNA derived from bone marrow specimens at the time of diagnosis. This was accomplished by bisulfite conversion and the pryosequencing technique. We defined aberrant methylation as >3 standard deviations from the mean methylation level derived from 8 healthy individuals. The median age at diagnosis was 16 months (range, 0.3–160). By genetic analysis, PTPN11, NF1, NRAS, KRAS, and CBL mutations were found in 39 (42%), 7 (8%), 12 (13%), 13 (14%), and 11 (12%) patients, respectively. In addition, 16 patients had SETBP1 or JAK3 mutations. Karyotypic abnormalities were detected in 15 patients, including 8 with monosomy 7. The median monocyte count, percentage of hemoglobin F, and platelet count at the time of diagnosis were 4.6x109/L (range, 0.2–31.6), 21% (range, 0–68), and 61.0x109/L (range, 1.4–483), respectively. The median observation period was 18 months (range, 1–287). During observation, 56 of the 92 patients received allogeneic hematopoietic stem cell transplantation (HSCT), and 30 of 92 patients died. Outcomes were assessed according to transplantation-free survival (TFS), in which HSCT and death were censored, and overall survival (OS) by the Kaplan-Meier method. Aberrant methylation of BMP4, CALCA, CDKN2A, CDKN2B, DMR-H19 and RARB were detected, of which hypermethylation of BMP4, CALCA, CDKN2A, and RARB were associated with poor TFS according to univariate analyses (P The present study shows that aberrant methylation of four genes (BMP4, CALCA, CDKN2A, and RARB) is associated with poor outcomes in JMML patients. Patients with SETBP1/JAK3 mutations frequently show the hypermethylation of these genes. Further, allogeneic HSCT is associated with improved outcomes for patients with AMS = 1 and 2. Therefore, these results suggest that examination of the methylation pattern of these four genes may help guide clinical decisions for the management of patients with JMML. Disclosures: Makishima: AA & MDS international foundation: Research Funding; Scott Hamilton CARES grant: Research Funding. Maciejewski:NIH: Research Funding; Aplastic anemia&MDS International Foundation: Research Funding.
- Published
- 2013
- Full Text
- View/download PDF
42. Whole Exome Sequencing Shows a Paucity Of Somatic Gene Mutations In Pediatric Idiopathic Bone Marrow Failure Syndrome
- Author
-
Hideki Muramatsu, Seishi Ogawa, Hiroko Tanaka, Xinan Wang, Sayoko Doisaki, Asahito Hama, Nozomu Kawashima, Hirotoshi Sakaguchi, Yusuke Okuno, Satoru Miyano, Yuichi Shiraishi, Yinyan Xu, Atsushi Narita, Yoshiyuki Takahashi, Seiji Kojima, Kenichi Yoshida, and Kenichi Chiba
- Subjects
Oncology ,Sanger sequencing ,medicine.medical_specialty ,Massive parallel sequencing ,business.industry ,Immunology ,Bone marrow failure ,Cell Biology ,Hematology ,Gene mutation ,medicine.disease ,Biochemistry ,Somatic evolution in cancer ,symbols.namesake ,Internal medicine ,medicine ,symbols ,business ,Refractory cytopenia with multilineage dysplasia ,Exome ,Exome sequencing - Abstract
Introduction Pancytopenia accompanied by a severe decrease in bone marrow (BM) cellularity in children can be due to a broad variety of underlying disorders. Appropriate classification of bone marrow failure syndrome in children is challenging, particularly in relation to the morphological distinction between aplastic anemia (AA), refractory cytopenia of childhood (RCC), and refractory cytopenia with multilineage dysplasia (RCMD). The goal of this study was to characterize the molecular pathogenesis of these conditions by identifying the full spectrum of gene mutations in 29 patients with these disorders through the use of exome sequencing. Patient and Methods Diagnosis of AA, RCC, or RCMD was made on basis of the 2008 World Health Organization (WHO) classification criteria. AA patients exhibited no morphologically dysplastic changes in any of their hematopoietic cell lineages, while RCC patients had10% in one cell lineage. Patients classified as RCMD exhibited >10% of the dysplastic changes in two or more cell lineages. Blood and BM samples were obtained from 29 children (16 boys and 13 girls) with AA (n = 8), RCC (n = 11), or RCMD (n = 10). The median age at diagnosis was 11 years (range, 2–15 years). Exome capture from paired DNA (non-T cells/CD3+ lymphocyte) was performed using SureSelect® Human All Exon V3 (Agilent Technologies, Santa Clara, CA) covering 50 Mb of the coding exons, followed by massive parallel sequencing using HiSeq 2000 (Illumina, San Diego, CA) according to the manufacturer’s protocol. Candidate somatic mutations were detected through our pipeline for whole exome sequencing (genomon: http://genomon.hgc.jp/exome/index.html). All candidate somatic nucleotide changes were validated by Sanger sequencing. Results Exome sequencing pipeline identified a total of 193 non-synonymous somatic mutations or indels candidates among the 29 patients (range, 2–15 per patient). After validation by Sanger sequencing, one nonsense, 11 missense, and two frame-shift mutations were confirmed as non-silent somatic mutations. The average numbers of mutations per sample were not significantly different when comparing morphological diagnostic groups (0.50 in AA, 0.36 in RCC, 0.60 in RCMD). Of these validated genes, BCOR (n = 2) and CSK (n = 2) mutations were recurrent genetic events. BCOR is a frequent mutational target in myelodysplastic syndrome, whereas CSK somatic mutations were not reported in human cancers. BCOR mutations were found both in AA (c.472delA:p.S158fs; patient 13) and in RCMD (c.G3856T:p.E1286X; patient 39). Both patients with CSK mutations were classified as RCC (c.G994A:p.D332N; patient 23 and 27). When comparing the clinical outcomes of patients with somatic mutations (n = 7) versus those without somatic mutations (n = 22), response rate to immunosuppressive therapy at 6 months (50% vs. 50%), 5-year clonal evolution rate (95% confidential interval) [0% (0% - 0%) vs. 6% (0% - 26%)], and the 5-year overall survival rate (95% confidential interval) [100% (100% - 100%) vs. 95% (70% - 99%)] were not significantly different. Conclusion Whole exome sequencing analysis was used for gene mutational profiling of patients with idiopathic bone marrow failure syndromes; i.e., AA, RCC, and RCMD. Although BCOR and CSK somatic mutations were recurrently identified, idiopathic bone marrow failure syndromes in children are characterized by a paucity of gene mutations, irrespective of morphological diagnosis. These findings suggest that morphological diagnosis based on WHO classification system does not discriminate the mutational profile and pathogenesis of bone marrow failure in children. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
- Full Text
- View/download PDF
43. Whole Exome Analysis Reveals Spectrum of Gene Mutations in Juvenile Myelomonocytic Leukemia
- Author
-
Xinan Wang, Hideki Makishima, Nao Yoshida, Kenichi Yoshida, Yinyan Xu, Hiroko Tanaka, Satoru Miyano, Asahito Hama, Kenichi Chiba, Sayoko Doisaki, Yuichi Shiraishi, Hideki Muramatsu, Jaroslaw P. Maciejewski, Seiji Kojima, Seishi Ogawa, Masashi Sanada, Koji Nakanishi, Hirotoshi Sakaguchi, Yusuke Okuno, and Yoshiyuki Takahashi
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Genetics ,Juvenile myelomonocytic leukemia ,Immunology ,Cell Biology ,Hematology ,Biology ,Gene mutation ,Compound heterozygosity ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Germline mutation ,medicine ,KRAS ,Exome ,Exome sequencing - Abstract
Abstract 170 Introduction: Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myeloid neoplasm clinically characterized by excessive proliferation of myelomonocytic cells and hypersensitivity to granulocyte–macrophage colony-stimulating factor (GM-CSF). A cardinal genetic feature of JMML is frequent somatic and/or germline mutations of RAS pathway genes involved in GM-CSF signal transduction, such as NRAS, KRAS, PTPN11, NF1, and c-CBL, which are found in >70% affected children in a mutually exclusive manner. To define the molecular pathogenesis of JMML, we identified the full spectrum of gene mutations in 13 cases of JMML using whole exome sequencing of paired tumor-normal DNA. We also performed target-deep sequencing of relevant mutational targets in 92 cases of JMML. Patient and Methods: We evaluated 92 children (61 boys and 31 girls) with JMML, including 7 with Noonan syndrome-associated myeloproliferative disorder, who were diagnosed at institutions throughout Japan. The median age at diagnosis was 19 months (range, 1–160 months). Karyotypic abnormalities were detected in 15 cases, including 8 with monosomy 7. Fifty-six of the 92 (61%) cases received allogeneic hematopoietic stem cell transplantation. Exome capture from paired tumor-normal (CD3-positive T cell) DNA obtained from 13 cases of JMML was performed using SureSelect® Human All Exon V3 (Agilent Technologies, Santa Clara, CA, USA) covering 50 Mb of the coding exons, followed by massive parallel sequencing using HiSeq 2000 (Illumina, San Diego, CA, USA) according to the manufacturers' protocol. Candidate somatic mutations were detected through our pipeline for whole exome sequencing (genomon: http://genomon.hgc.jp/exome/index.html). All candidate germline and somatic nucleotide changes were validated by Sanger/deep sequencing. A total of 92 JMML tumor specimens were screened for mutations in RAS pathway genes (PTPN11, NRAS, KRAS, c-CBL, and NF1) and 3 newly identified genes using deep sequencing. Results: For the current exome study, 10 missense and 1 nonsense single nucleotide variations were confirmed as nonsilent somatic mutations. The average number of mutations per sample (0.79; range, 0–4) was surprisingly low compared with those reported in other human cancers. Among the 11 somatic mutations, 6 involved the known RAS pathway genes (1 NF1, 1 NRAS, 2 KRAS, and 2 PTNP11 mutations) and included 5 mutations/deletions of either NF1 (n = 2), c-CBL (n = 1), or PTPN11 (n = 2) as detected in the germline samples. Nonoverlapping RAS pathway mutations were confirmed in 11 of the 13 discovered cases of JMML (85%). Five of the 11 somatic mutations were observed in 3 non-RAS pathway genes that have never been reported in JMML cases. Deep sequencing revealed RAS pathway mutations in 80 of 92 cases (87%) in a mutually exclusive manner; PTPN11 mutations were predominant (39/92 or 42%), followed by N/KRAS (24/92 or 26%), c-CBL (11/92 or 12%), and NF1 (6/92 or 6.5%) mutations. In agreement with previous reports, the majority of c-CBL (7/11) and NF1 (5/6) mutations were bi-allelic in the affected cases, showing compound heterozygous mutations or uniparental disomy of the mutant alleles, whereas most of the PTPN11 and N/KRAS mutations were heterozygous. In contrast, the remaining 12 (13%) cases were negative for known RAS pathway mutations. In addition, the 3 newly identified genes were recurrently in 18 cases (20%). Many of these mutations had lower allele frequencies compared to the accompanying RAS pathway mutations, indicating that they were responsible for disease progression rather than the establishment of JMML. The probability of 5-year transplantation-free survival (95% confidence interval) for the latter patients was significantly inferior to that of other cases (0% vs. 19% (8–34%), p < 0.001). Conclusion: Whole exome sequencing revealed the spectrum of gene mutations in cases of JMML. Together with a very high frequency of RAS pathway mutations, the paucity of non-RAS pathway mutations is a prominent feature of JMML. Somatic mutations of 3 newly identified genes were common among recurrent secondary events presumed to be involved in tumor progression and associated with poor clinical outcomes. Our findings provide an important clue that aids in understanding the pathogenesis of JMML and will help in the development of novel diagnostics and therapeutics for this type of leukemia. Disclosures: Maciejewski: NIH: Research Funding; Aplastic Anemia & MDS International Foundation: Research Funding.
- Published
- 2012
- Full Text
- View/download PDF
44. Combination Immunotherapy After ASCT for Multiple Myeloma (MM) Using MAGE-A3/Poly-ICLC Immunizations Followed by Vaccine-Primed and Activated Autologous T-Cells
- Author
-
Dan T. Vogl, Bruce L. Levine, Ling Cai, Elizabeth Veloso, YinYan Xu, Edward A. Stadtmauer, Zhaohui Zheng, Holly McConville, Sandra Westphal, Aaron P. Rapoport, Brendan M. Weiss, Ashraf Badros, Sunita Philip, Nicole A. Aqui, Andres M. Salazar, Saul Yanovich, Kelly-Marie Betts, Hong-Bin Fang, Scott Strome, Gorgun Akpek, Kathleen Ruehle, Anne Chew, and Carl H. June
- Subjects
Oncology ,medicine.medical_specialty ,Reactogenicity ,business.industry ,Immunology ,Tumor antigen vaccine ,Cell Biology ,Hematology ,Leukapheresis ,medicine.disease ,Biochemistry ,Vaccination ,Transplantation ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,business ,Multiple myeloma ,Lenalidomide ,medicine.drug - Abstract
Abstract 352 Background: Autologous stem cell transplantation (ASCT) for MM leads to complete responses in ∼20–40% of patients but rare cures. Patients with rapid recovery of T cells post ASCT may have improved outcomes suggesting possible immune mediated tumor control. We have shown that adoptive T-cell transfers after ASCT for MM using vaccine-primed and ex-vivo costimulated autologous T-cells in combination with pre- and post-transplant immunizations using a tumor antigen vaccine (+ GM-CSF and montanide) led to vaccine-directed T-cell responses in about 1/3 of patients and enhanced recovery of polyclonal T and B cell counts and function. We hypothesized that addition of Poly-ICLC (Hiltonol®) – a TLR-3 agonistic vaccine adjuvant could increase tumor antigen vaccine responses through better priming and boosting. Methods: We report interim results of a Phase II clinical trial (NCT01245673) evaluating safety and activity of autologous T cells primed in-vivo with a MAGE-A3 multipeptide vaccine (Orphan Drug Designation: GL-0817) mixed with GM-CSF and Poly-ICLC (Hiltonol®) +/− montanide. Inclusion criteria included measurable disease or high-risk cytogenetics. MAGE-A3 is expressed in ∼50% MM cases and more frequently in relapsed/extramedullary/proliferative disease. The MAGE-A3 vaccine has 2 HLA-A2-restricted class I peptides and 1 promiscuous class II peptide linked to an HIV-1-TAT membrane translocation sequence (Trojan peptide) to enhance peptide presentation. Vaccine-primed T cells were collected by leukapheresis, costimulated and expanded ex-vivo using anti-CD3/anti-CD28 mAb conjugated beads. T-cells were infused at day +2 after ASCT followed by booster immunizations at days 14, 42, 90, 120 and 150 post-transplant. Lenalidomide maintenance was started at day +100. Patients were evaluated for MM responses in accordance with IMWG criteria at days 60, 100, 180 post-transplant and Q3 months thereafter. T-cell and B-cell responses to the vaccine were evaluated by IFN-g or IL-2 cytokine production (all patients), dextramer binding to CD8 cells (HLA-A2 positive patients) and ELISA antibody assays at days 14, 60, 100 and 180 post-transplant. Results: 25 patients were transplanted on study. At a median followup of 6 months (range 2–18 months), 24 patients are surviving while 1 patient relapsed at about day +60 and died. Four additional patients have relapsed at 7,9,18 and 18 months post-transplant, yielding a 1-year Kaplan-Meier EFS of 77%. Of the 16 patients evaluable for response at day 100, 7/16 (44%) had CR/nCR using the study enrollment (post-induction) myeloma markers as a baseline while at day 180, 7/13 (53%) had CR/nCR. T-cell infusions were well-tolerated with no probable/definite grade 3 or higher toxicities. Vaccinations were associated with > 50 mm injection site reactions (redness, induration or both) after 1 or more immunizations in the majority of patients. Two patients developed large and prolonged inflammatory reactions which evolved into sterile abscesses. These resolved over 2–3 months with conservative management but as a result montanide was eliminated from the vaccine formulation for patients 11–25. Thereafter vaccine reactogenicity was decreased with no additional sterile abscesses. Of 16 patients tested for immune responses to date, 2 patients were unevaluable due to poor sample viability. Dextramer staining demonstrated MAGE-A3-specific CD8 T-cells in 4/4 (100%) of evaluable HLA-A2+ patients. Cytokine production in response to MAGE-A3 stimulation was seen in 11/14 (79%) patients; responses usually peaked at day 100 or day 180. Robust MAGE-A3 antibody responses were detected in 7/9 patients who received montanide in the vaccine formulation but in 0/7 patients who did not. Conclusions: Combination immunotherapy using a MAGE-A3 multipeptide tumor antigen vaccine plus vaccine-primed and costimulated autologous T-cells after ASCT for MM is well-tolerated and associated with encouraging early clinical responses. The addition of Poly-ICLC (Hiltonol®) to the vaccine formulation was associated with a high frequency of post-ASCT T-cell functional responses. The combination of Poly-ICLC +GM-CSF + Montanide led to robust injection site reactions that were occasionally severe and prolonged. Elimination of the montanide reduced injection site reactogenicity but may also have compromised the B-cell responses to the tumor antigen vaccine. Disclosures: Rapoport: Gliknik, Inc: Research Funding. Stadtmauer:Celgene: Consultancy, Speakers Bureau; Millenium: Consultancy, Speakers Bureau. Vogl:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Millennium/Takeda: Consultancy, Research Funding; Otsuka: Consultancy; Acetylon: Research Funding. Strome:Gliknik, Inc: Equity Ownership, Patents & Royalties, Research Funding. Salazar:Oncovir, Inc: Employment. Levine:TxCell: Consultancy, Membership on an entity's Board of Directors or advisory committees; University of Pennsylvania: financial interest due to intellectual property and patents in the field of cell and gene therapy. Conflict of interest is managed in accordance with University of Pennsylvania policy and oversight, financial interest due to intellectual property and patents in the field of cell and gene therapy. Conflict of interest is managed in accordance with University of Pennsylvania policy and oversight Patents & Royalties. June:Novartis: Research Funding, entitled to receive royalties from patents licensed to Novartis, entitled to receive royalties from patents licensed to Novartis Patents & Royalties.
- Published
- 2012
- Full Text
- View/download PDF
45. Phase II Trial of Lenalidomide - Rituximab +/− Dexamethasone in Relapsed or Refractory Indolent B-Cell or Mantle Cell Lymphomas Resistant to Rituximab
- Author
-
Tahamtan Ahmadi, Sunita D. Nasta, Jakub Svoboda, Amanda Gordon, Nicole A. Aqui, Elise A. Chong, Stephen J. Schuster, and YinYan Xu
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Rash ,Lymphoma ,Surgery ,Internal medicine ,Cohort ,medicine ,Rituximab ,Mantle cell lymphoma ,medicine.symptom ,business ,Dexamethasone ,Lenalidomide ,medicine.drug - Abstract
Abstract 266 Introduction: Lenalidomide is an immunomodulatory drug with effects on the innate immune system that may enhance antibody-dependent cell mediated cytotoxicity as well as the development of specific anti-tumor immune responses. These immunologic effects may synergize with the action of rituximab. To test the efficacy of lenalidomide combined with rituximab, we are conducting a single center, open label phase II clinical trial in patients (pts) with indolent B-cell or mantle cell lymphomas previously resistant to rituximab. Patients and Methods: Eligible pts must have relapsed/refractory indolent B-cell or mantle cell lymphoma with measurable disease that has failed to respond or has progressed within six months of a standard course of rituximab monotherapy (375 mg/m2 weekly for at least four weeks) or a prior rituximab-containing chemotherapy regimen. Thus, all pts enrolled are considered rituximab refractory. Patients were enrolled sequentially into two treatment cohorts. In Cohort 1, pts received two 28-day treatment cycles of lenalidomide 10 mg every day and dexamethasone 8 mg once weekly (Part I: lenalidomide + dexamethasone). After assessment of response to Part I, all pts received a single course of rituximab 375 mg/m2, consisting of four weekly doses during cycle 3 (Part II: lenalidomide + dexamethasone + rituximab). Treatment with lenalidomide + dexamethasone continued during and subsequent to rituximab; stable and responding pts continued on lenalidomide + dexamethasone until disease progression or development of clinically unacceptable toxicity. Response assessment after Part II was performed three months after the first dose of rituximab. In Cohort 2, dexamethasone was eliminated; otherwise, eligibility and treatment were the same. Results: As of August 9, 2011, 45 pts have started therapy (Cohort 1, n = 27; Cohort 2, n = 18); diagnoses include: follicular (n = 28), mantle cell (n = 11), small lymphocytic (n = 4), and marginal zone (n = 2) lymphomas; median age is 58 years (range: 35–85); male: female ratio is 31:14; median number of prior therapies is 3 (range: 1 – 7); LDH is increased in 18%. For Cohort 1, 24 pts completed Part II and are evaluable for response; 3 pts are not evaluable, 1 due to death (myocarditis) and 2 due to removal from study (1 thrombocytopenia attributed to myelodysplasia; 1 rash attributed to lenalidomide). For Cohort 2, 11 pts are evaluable for response assessment after Part II; 2 pts are not evaluable due to discontinuation of protocol therapy (1 rash attributed to lenalidomide; 1 with early symptomatic progression of lymphoma); 5 pts have not completed Part II. For 35 pts completing Parts I and II, at a median follow-up of 11.8 months, PFS is 73% (95%CI: 53 – 86%). Overall response rate (ORR) after Part I is 37% (6 CR; 7 PR); ORR after Part II is 60% (12 CR; 9 PR). Overall response rates after Part II do not differ between Cohort 1 (with dexamethasone) and Cohort 2 (without dexamethasone) [58% vs. 64%, respectively; p = 0.5]. There were fewer dose interruptions during Part I in Cohort 1 (15%) versus Cohort 2 (50%) [p = 0.02], reflecting reduced episodes of tumor flare and rash in Cohort 1. Conclusions: For rituximab-resistant pts with indolent B-cell or mantle cell lymphomas, the combination of continuous daily lenalidomide and a single four week course of rituximab, with or without low-dose weekly dexamethasone, achieves a high overall response rate with relatively durable responses. The addition of low-dose weekly dexamethasone may improve tolerance without decreasing the efficacy of the lenalidomide-rituximab combination. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2011
- Full Text
- View/download PDF
46. Differences in regulatory T cells (Tregs) in responding and non-responding patients with indolent B-cell or mantle cell lymphoma during treatment with lenalidomide and rituximab ± dexamethasone
- Author
-
Amanda Gordon, L. Leinbach, Jakub Svoboda, Elise A. Chong, Stephen J. Schuster, S. Nasta, Tahamtan Ahmadi, YinYan Xu, and Nicole A. Aqui
- Subjects
Cancer Research ,business.industry ,chemical and pharmacologic phenomena ,medicine.disease ,medicine.anatomical_structure ,Immune system ,Oncology ,Immunology ,medicine ,Rituximab ,Mantle cell lymphoma ,Cytotoxicity ,business ,Dexamethasone ,B cell ,medicine.drug ,Lenalidomide - Abstract
8060 Background: Lenalidomide may enhance anti-tumor immune responses by inhibiting Tregs and by augmenting NK-cell mediated antibody-dependent cytotoxicity. These immunologic effects may synergize...
- Published
- 2011
- Full Text
- View/download PDF
47. Adoptive immunotherapy with autologous CD3/CD28-costimulated T cells after fludarabine-based chemotherapy in patients with chronic lymphocytic leukemia
- Author
-
Andrea L. Brennan, L. Leinbach, Bruce L. Levine, Stephen J. Schuster, Elizabeth Veloso, Elise A. Chong, Julio Cotte, William K. Decker, Elizabeth J. Shpall, Carl H. June, Zhaohui Zheng, John McMannis, Amanda Gordon, Michael J. Keating, YinYan Xu, Doyle Bosque, Chitra Hosing, Nicole A. Aqui, Catherine M. Bollard, and Jakub Svoboda
- Subjects
Oncology ,medicine.medical_specialty ,Cancer Research ,Cyclophosphamide ,medicine.medical_treatment ,CD3 ,Chronic lymphocytic leukemia ,Immunology ,Adoptive immunotherapy ,Biochemistry ,Internal medicine ,hemic and lymphatic diseases ,medicine ,In patient ,Chemotherapy ,biology ,business.industry ,CD28 ,Immunosuppression ,Cell Biology ,Hematology ,medicine.disease ,Minimal residual disease ,Fludarabine ,biology.protein ,Alemtuzumab ,Rituximab ,business ,Progressive disease ,medicine.drug - Abstract
Background Fludarabine-based therapies prolong survival of patients (pts) with chronic lymphocytic leukemia (CLL), but deplete T-cells causing immunosuppression which increases the risk of infection and may limit control of minimal residual disease by the immune system. Adoptive immunotherapy using autologous CD3/CD28-costimulated T-cells expanded ex vivo (ACTC) enhances immune reconstitution after fludarabine-based therapy of lymphoma (Schuster Blood abstract 2007). Methods We conducted a multicenter phase I/II trial of ACTC following fludarabine- or alemtuzumab-based therapy of CLL. Eligibility required IWCLL 2008 indications for treatment of CLL. After leukapheresis, pts received fludarabine- or alemtuzumab-based therapy. Eight - 12 weeks after last immuno-chemotherapy, responding pts (CR, PR) received a single dose of ACTC prepared from autologous T-cells collected before therapy. Primary study endpoints were feasibility, defined as the ability to generate a T-cell dose of 1.0 E+10+/- 20%, and safety, defined as Results Thirty-four pts with CLL were enrolled (median age: 62). Eighteen patients were previously untreated and 16 patients had relapsed or refractory CLL. Four pts (12%) had del(17p), 11 pts (33%) del(11q), and 3 (9%) both del(17p) and del(11q). For previously untreated patients, 17 received fludarabine-cyclophosphamide-rituximab (FCR) and 1 pt received fludarabine-rituximab (FR); for relapsed patients, 10 received FCR, 3 FCR-bevacizumab, 2 alemtuzumab, and 1 oxaliplatin-fludarabine-cytarabine-rituximab (OFAR). Ten patients did not receive ACTC infusion for the following reasons: 6 pts (including 1 receiving alemtuzumab) had progressive disease prior to ACTC, 1 pt developed autoimmune hemolyic anemia during FCR, 1 pt developed ITP during FCR, 1 pt developed pure red cell aplasia during FCR, and 1 pt receiving alemtuzumab was removed from study per physician preference. Median ACTC dose was 1.0 E+10 CD3 cells (range: 3.94 E+08 -1.03 E+10). Two pts had progressive disease less than 90 days after ACTC and 1 pt was lost to follow-up. Twenty-one pts (14 CR; 2 CRi; and 5 PR at the time of ACTC infusion) were evaluable for studies of immune reconstitution at ≥90 days after ACTC. After chemotherapy prior to ACTC (D0), median CD4 count was 120 (range: 19-573); 90 days after ACTC (D+90), median CD4 count was 399 (range 62-1818) (p = 0.0003; median fold CD4 increase 2.3, range -0.5–20.9) [figure 1]. Median CD8 count also increased significantly (p = 0.004; median D0 = 86, range: 4-682; D+90 = 267, range: 18-2876; median fold CD8 increase 1.1, range: -0.8-18.4) [figure 2]. The increases in CD4 and CD8 counts between D0 and D+90 were not dependent on ACTC dose. There were no SAEs or grade 3 or higher non-hematologic toxicities related to T-cell infusion. For all 21 patients evaluable for immune reconstitution at D+90, median follow-up is 33 months with median PFS 30 months (not shown). PFS for previously untreated patients is 42 months, whereas PFS for relapsed patients is 12 months (figure 3). There was no correlation between magnitude of increase in post-infusion CD4 and CD8 counts and PFS at 1 year. Conclusions For CLL patients, ACTC production is feasible. ACTC infusion is well tolerated and results in a dose-independent acceleration of CD4+ and CD8+ cell recovery after fludarabine-based chemotherapy compared to historical controls. The application of adoptive immunotherapy with ACTC to accelerate immune reconstitution after immunochemotherapy could potentially provide an optimal immunologic milieu for early application of additional immunotherapeutic interventions. Disclosures: Levine: Novartis: cell and gene therapy IP, cell and gene therapy IP Patents & Royalties. June: Novartis: Patents & Royalties, Research Funding.
- Published
- 2011
- Full Text
- View/download PDF
48. Recombinant Human Interleukin-7 (CYT107) Enhances CD4 and CD8 T Cell Recovery Following T-Cell Depleted Allogeneic Hematopoietic Stem Cell Transplant In Patients with Myeloid Malignancies
- Author
-
Molly Maloy, Miguel-Angel Perales, Bushra Zaidi, Teresa S. Rasalan, Jenna D. Goldberg, Marcel R.M. van den Brink, James W. Young, Yinyan Xu, Michel Morre, Guenther Koehne, Jianda Yuan, Glenn Heller, Leuren Lechner, Cailian Liu, Esperanza B. Papadopoulos, Ann A. Jakubowski, Jedd D. Wolchok, Ryan Kendle, Humilidad F. Gallardo, and Thérèse Croughs
- Subjects
business.industry ,medicine.medical_treatment ,T cell ,Immunology ,FOXP3 ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Graft-versus-host disease ,Immune system ,medicine.anatomical_structure ,medicine ,Cytotoxic T cell ,IL-2 receptor ,business ,CD8 - Abstract
Abstract 674 Immune recovery is an important determinant in multiple outcomes following allogeneic hematopoietic stem cell transplant (allo-HSCT). Delays in B and T cell reconstitution are associated with an increased risk of infection, relapse and secondary malignancy. Strategies to enhance post-transplant T-cell reconstitution could therefore improve morbidity and mortality after allo-HSCT. The cytokine Interleukin-7 (IL-7) is a unique therapeutic candidate to promote immune reconstitution because it has a central role in T cell development and survival. Murine models of allo-HSCT have demonstrated that IL-7 can enhance thymopoiesis as well as promote peripheral T cell survival and expansion. Initial clinical trials performed with recombinant human IL-7 (rhIL-7) have demonstrated a dose-dependent expansion of CD4+ and CD8+ T cells with an acceptable toxicity profile in patients with solid tumors or HIV infection. Hence we are conducting a phase I trial of post-transplant administration of rhIL-7 (CYT107, Cytheris Inc) in recipients of a T cell depleted (TCD) allo-HSCT to determine the safety, toxicity and biological activity on T cell reconstitution. To date, 9 patients (AML=7, MDS=2), with a median age of 59.3 years (range 27–67 years) have been treated with escalating doses of rhIL-7 (3 at 10 mcg/kg, 6 at 20 mcg/kg) administered subcutaneously weekly for 3 weeks following TCD allo-HSCT from an HLA compatible donor. Accrual is ongoing in the final cohort (30 mcg/kg). Recombinant hIL-7 was started at a median of 96 days post allo-HSCT (range 61–244 days). Most patients experienced transient minor injection site reactions. One patient (20 mcg/kg) developed a biopsy proven hypersensitivity drug rash a week after the first injection and was removed from the study (evaluable for toxicity but not immune recovery endpoints). No other significant injection-related toxicities have occurred, and no patients have developed GVHD. No anti-IL-7 antibodies or neutralizing antibodies have developed following rhIL-7 injection. Two of 9 patients with high-risk AML have relapsed (4 and 9 months post rhIL-7), an incidence consistent with published data in patients undergoing allo-HSCT for AML in CR, irrespective of T-cell depletion. Eight patients remain alive with a median follow-up of 14.5 months post rhIL-7 administration. At baseline, the median T cell counts were 91/mm3 (range 5 – 219 /mm3), 43/mm3 (range 9 – 299 /mm3) and 0 (range 0 – 17 /mm3) for CD4+, CD8+ and CD45RA+ T cells, respectively. Preliminary assessment of the immunological effects of rhIL-7 in 8 evaluable patients has demonstrated an increase in CD4+ T cells exhibiting a naïve or central memory phenotype (69% median increase over baseline at day 21 – range 8% to 35-fold increase), and CD8+ T cells exhibiting a naïve or effector memory phenotype (94% median increase over baseline at day 28 – range 0 to 11-fold increase). There was no observed effect on the frequency of CD4+CD25+FoxP3+ T cells or CD19+ B cells. TCR excision circles (TREC) analysis performed on CD4+ and CD8+ subsets in the first 6 patients, using absolute quantification real-time PCR, demonstrated increases in TRECs in 5/6 patients indicating enhanced T cell production. Finally, all 3 CMV-seropositive patients developed CD8+ T cell CMV-specific responses detected by intracellular IFNγ production to overlapping CMV-pp65 pentadecapeptides peptide pools after administration of rhIL-7. In one patient, we also analyzed CMV-specific T-cell frequency using HLA-A*0201 restricted MHC-tetramers. The highest CMV-specific response levels were noted in this patient with a history of CMV viremia and low-level CMV-specific CD8+ T cells prior to rhIL-7 (5.3-fold increase to the A0201-restricted immunodominant NLV peptide by tetramer assay after rhIL-7). Our pre-clinical data and early clinical results suggest that administration of rhIL-7 in recipients of a TCD allo-HSCT has minimal toxicity and can enhance post-transplant immune recovery without causing GVHD. Disclosures: Perales: Cytheris: Research Funding. Croughs:Cytheris: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Morre:Cytheris: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. van den Brink:Cytheris: Research Funding.
- Published
- 2010
- Full Text
- View/download PDF
49. C-Cbl but Not TET2 Mutations Are Present in Patients with Juvenile Myelomonocytic Leukemia
- Author
-
Nao Yoshida, Hiroshi Yagasaki, Heather Cazzolli, Koji Kato, Jaroslaw P. Maciejewski, Anna M. Jankowska, Hideki Makishima, Asahito Hama, Seiji Kojima, Atsushi Manabe, Christine L. O'Keefe, Yoshiyuki Takahashi, Yinyan Xu, Nobuhiro Nishio, and Hideki Muramatsu
- Subjects
Genetics ,Neuroblastoma RAS viral oncogene homolog ,Chromosome 7 (human) ,Juvenile myelomonocytic leukemia ,Immunology ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,PTPN11 ,hemic and lymphatic diseases ,medicine ,Cancer research ,Hemoglobin F ,KRAS ,SNP array - Abstract
Abstract 420 Juvenile myelomonocytic leukemia (JMML) is a distinct subtype of myelodysplastic syndrome/myeloproliferative disorder (MDS/MPD) which, in analogy to chronic myelomonocytic leukemia (CMML), is characterized by excessive proliferation of myelomonocytic cells, but unlike CMML it occurs in young children and shows characteristic hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF). Chromosomal defect are present in 22% of patients in particular involving del7/7q. Mutations of genes involved in GM-CSF signal transduction, including RAS and PTPN11, can be identified in a majority of children with JMML, constitutional mutations of NF1 can be found in another 10% of patients with JMML, but in significant proportion of patients no molecular lesions were identified. To further clarify the molecular pathogenesis of JMML we have applied a high density SNP array-based karyptyping and screened for associated mutations including established defected in RAS, PTPN11 and NF1 but also c-Cbl and TET genes, recently identified in patients with CMML and MDS/MPD. We studied 49 children with JMML diagnosed between 1988 and 2008 in 28 institutions throughout Japan. The median age at diagnosis was 28 months (range, 1-75 months). Karyotypic abnormalities were detected in 11 patients, including 7 patients with monosomy 7. Two children had clinical evidence of NF1 mutations. Out of 49 patients, 32 received hematopoietic stem cell transplantation (HSCT). We performed mutational analysis of the genes known to be affected by mutations in JMML. PTPN11 mutations were found in 26/49 (53%) while NRAS and KRAS mutations were found in 2/49 (4%) and 1/49 (2%), respectively. None of the patients screened showed the presence of TET2 mutations, previously shown to be present in a significant proportion of patients with MDS/MPD, including CMML. High-density Affymetrix 250K single nucleotide polymorphism array (SNP-A) were applied as a karyotyping platform to identify LOH and submicroscopic copy number changes. Signal intensity was analyzed and SNP calls determined using Gene Chip Genotyping Analysis Software Version 4.0 (GTYPE). Copy number and areas of UPD were investigated using Hidden Markov Model and CNAG v3.0 software. Compared to the results of conventional metaphase cytogenetics (MC), SNP-A identified significantly more genetic abnormalities (25% vs 49%; p=.02). In 1 patient UPD17q was present ivolving NF1 locus. In 4 patients UPD11q involving c-cbl locus (11q23.1) was found. Sequencing of c-Cbl gene family revealed mutations of c-Cbl in 5/49 (10%), and no Cbl-b mutations. All but 1 mutations were homozygous and were located in the RFD (exon 8 and intron 8). C-Cbl mutations were mutually exclusive with PTPN11, NRAS, and KRAS mutations or had clinical diagnosis of NF1. Unlike in CMML, no UPD4q24 or homo- or heterozygous TET2 mutations were found. Histomorphologic analysis did not reveal any distinct c-Cbl mutation-associated features or differences in count between patients grouped based on the presence of specific mutations. Similarly, there were no differences in gender, or the presence of cytogenetic abnormalities and the probability of 2 year overall survival of c-Cbl mutant cases between patients grouped according to mutational status. All patients with c-Cbl mutations displayed GM-CSF hypersensitivity at initial presentation but did not differ in this feature from the remaining JMML patients. However, mutant c-Cbl cases showed earlier presentation (median age 12 months vs. 29 months, p = .037) and lower median hemoglobin F fraction (3.5 % vs. 25%, p=.02). In sum, c-Cbl mutations constitute a novel important pathogenic lesion in JMML. While their presence suggests functional similarity to CMML, absence of TET2 mutation in JMML and rarity of PTPN11 mutations in CMML constitute important distinctive features of both diseases. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2009
- Full Text
- View/download PDF
50. Correlation of Clinical Features with Mutational Status in Children with Juvenile Myelomonocytic Leukemia
- Author
-
Hiroshi Yagasaki, Seiji Kojima, Makito Tanaka, Yinyan Xu, Yoshiyuki Takahashi, Nao Yoshida, Hirokazu Hidaka, Atsushi Manabe, Nobuhiro Nishio, and Asahito Hama
- Subjects
Chromosome 7 (human) ,Mutation ,Juvenile myelomonocytic leukemia ,business.industry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease_cause ,medicine.disease ,Biochemistry ,PTPN11 ,Pathogenesis ,Leukemia ,hemic and lymphatic diseases ,Fetal hemoglobin ,Cancer research ,Medicine ,business - Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare clonal myeloproliferative disorder that affects young children. It is characterized by specific hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) in vitro. Mutations in RAS, NF1, or PTPN11 positioned in the GM-CSF signal transduction pathway, which contribute to myeloid proliferation, have been recognized in the pathogenesis of JMML. Recently a multi-step model for leukemogenesis has been proposed. In this model, the pathogenesis of leukemia requires at least two classes of mutations:primary mutations of genes implicated in cell differentiation such as AML1 and PU.1 andadditional mutations of genes contributing to myeloid proliferation such as FLT3, RAS, and PTPN11. We hypothesized that in patients with JMML, in addition to known mutations of genes in the GM-CSF pathway involved in myeloid proliferation, potential causative mutations of other classes might be acquired. AML1 encodes a transcription factor that is essential for definitive hematopoiesis, and its mutations have recently been found in adults with acute myeloid leukemia and myelodysplastic syndrome. However, no information is available on the profiles of mutations in these genes and the relationship between these mutations and clinical features of JMML in children. We analyzed mutations of N-RAS, K-RAS, and PTPN11 in 50, and of AML1 in 30 Japanese children with JMML by direct sequencing. Correlation between the mutational status and clinical and laboratory findings, including age at diagnosis, sex, fetal hemoglobin (HbF), platelets count, cytogenetic abnormalities, and hepatomegaly, all which are suggested prognostic factors for JMML, were also assessed. Of the 50 patients with JMML, cytogenetic abnormalities were detected in 14, including 8 with monosomy 7. PTPN11 and N-/K-RAS mutations were found in 24 (48%) and 7 (14%) patients, respectively, and a novel mutation in AML1 was identified in one patient. No simultaneous mutations in these genes were found. Age at diagnosis was older (median 36 vs 11 months, p=0.0005) and HbF level was higher (31.0% vs 5.1%, p=0.033) in patients with the PTPN11 mutation than those with the RAS mutation. No difference was observed between patients with PTPN11 and RAS mutations in sex ratio, white blood cell count, platelets count, and the incidence of cytogenetic abnormalities and hepatomegaly. Our results suggest that the clinical outcome of patients with PTPN11 might differ from those with RAS mutations because a higher level of HbF and older age have been reported to be poor prognostic factors. In one patient with JMML, we identified a novel mutation in the AML1 gene that belongs to the class of genes contributing to cell differentiation instead of the class of genes in the GM-CSF pathway involved in myeloid proliferation.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.