Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amy Beck is active.

Publication


Featured researches published by Amy Beck.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Tumor-infiltrating NY-ESO-1–specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer

Junko Matsuzaki; Sacha Gnjatic; Paulette Mhawech-Fauceglia; Amy Beck; Austin Miller; Takemasa Tsuji; Cheryl Eppolito; Feng Qian; Shashikant Lele; Protul Shrikant; Lloyd J. Old; Kunle Odunsi

NY-ESO-1 is a “cancer-testis” antigen frequently expressed in epithelial ovarian cancer (EOC) and is among the most immunogenic tumor antigens defined to date. In an effort to understand in vivo tolerance mechanisms, we assessed the phenotype and function of NY-ESO-1–specific CD8+ T cells derived from peripheral blood lymphocytes (PBLs), tumor-infiltrating lymphocytes (TILs), and tumor-associated lymphocytes (TALs) of EOC patients with NY-ESO-1-expressing tumors, with or without humoral immunity to NY-ESO-1. Whereas NY-ESO-1–specific CD8+ T cells were readily detectable ex vivo with tetramers in TILs and TALs of seropositive patients, they were only detectable in PBLs following in vitro stimulation. Compared with PBLs, tumor-derived NY-ESO-1–specific CD8+ T cells demonstrated impaired effector function, preferential usage of dominant T-cell receptor, and enriched coexpression of inhibitory molecules LAG-3 and PD-1. Expression of LAG-3 and PD-1 on CD8+ T cells was up-regulated by IL-10, IL-6 (cytokines found in tumor ascites), and tumor-derived antigen-presenting cells. Functionally, CD8+LAG-3+PD-1+ T cells were more impaired in IFN-γ/TNF-α production compared with LAG-3+PD-1− or LAG-3−PD-1− subsets. Dual blockade of LAG-3 and PD-1 during T-cell priming efficiently augmented proliferation and cytokine production by NY-ESO-1–specific CD8+ T cells, indicating that antitumor function of NY-ESO-1-specific CD8+ T cells could potentially be improved by therapeutic targeting of these inhibitory receptors.


PLOS ONE | 2014

Ovarian Cancer Spheroid Cells with Stem Cell-Like Properties Contribute to Tumor Generation, Metastasis and Chemotherapy Resistance through Hypoxia-Resistant Metabolism

Jianqun Liao; Feng Qian; Nana Tchabo; Paulette Mhawech-Fauceglia; Amy Beck; Zikun Qian; Xinhui Wang; Wendy J. Huss; Shashikant Lele; Carl Morrison; Kunle Odunsi

Cells with sphere forming capacity, spheroid cells, are present in the malignant ascites of patients with epithelial ovarian cancer (EOC) and represent a significant impediment to efficacious treatment due to their putative role in progression, metastasis and chemotherapy resistance. The exact mechanisms that underlie EOC metastasis and drug resistance are not clear. Understanding the biology of sphere forming cells may contribute to the identification of novel therapeutic opportunities for metastatic EOC. Here we generated spheroid cells from human ovarian cancer cell lines and primary ovarian cancer. Xenoengraftment of as few as 2000 dissociated spheroid cells into immune-deficient mice allowed full recapitulation of the original tumor, whereas >105 parent tumor cells remained non-tumorigenic. The spheroid cells were found to be enriched for cells with cancer stem cell-like characteristics such as upregulation of stem cell genes, self-renewal, high proliferative and differentiation potential, and high aldehyde dehydrogenase (ALDH) activity. Furthermore, spheroid cells were more aggressive in growth, migration, invasion, scratch recovery, clonogenic survival, anchorage-independent growth, and more resistant to chemotherapy in vitro. 13C-glucose metabolic studies revealed that spheroid cells route glucose predominantly to anaerobic glycolysis and pentose cycle to the detriment of re-routing glucose for anabolic purposes. These metabolic properties of sphere forming cells appear to confer increased resistance to apoptosis and contribute to more aggressive tumor growth. Collectively, we demonstrated that spheroid cells with cancer stem cell-like characteristics contributed to tumor generation, progression and chemotherapy resistance. This study provides insight into the relationship between tumor dissemination and metabolic attributes of human cancer stem cells and has clinical implications for cancer therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients

Kunle Odunsi; Junko Matsuzaki; Julia Karbach; Antje Neumann; Paulette Mhawech-Fauceglia; Austin Miller; Amy Beck; Carl Morrison; Gerd Ritter; Heidi Godoy; Shashikant Lele; Nefertiti C. duPont; Robert P. Edwards; Protul Shrikant; Lloyd J. Old; Sacha Gnjatic; Elke Jäger

Recombinant poxviruses (vaccinia and fowlpox) expressing tumor-associated antigens are currently being evaluated in clinical trials as cancer vaccines to induce tumor-specific immune responses that will improve clinical outcome. To test whether a diversified prime and boost regimen targeting NY-ESO-1 will result in clinical benefit, we conducted two parallel phase II clinical trials of recombinant vaccinia-NY-ESO-1 (rV-NY-ESO-1), followed by booster vaccinations with recombinant fowlpox-NY-ESO-1 (rF-NY-ESO-1) in 25 melanoma and 22 epithelial ovarian cancer (EOC) patients with advanced disease who were at high risk for recurrence/progression. Integrated NY-ESO-1-specific antibody and CD4+ and CD8+ T cells were induced in a high proportion of melanoma and EOC patients. In melanoma patients, objective response rate [complete and partial response (CR+PR)] was 14%, mixed response was 5%, and disease stabilization was 52%, amounting to a clinical benefit rate (CBR) of 72% in melanoma patients. The median PFS in the melanoma patients was 9 mo (range, 0–84 mo) and the median OS was 48 mo (range, 3–106 mo). In EOC patients, the median PFS was 21 mo (95% CI, 16–29 mo), and median OS was 48 mo (CI, not estimable). CD8+ T cells derived from vaccinated patients were shown to lyse NY-ESO-1-expressing tumor targets. These data provide preliminary evidence of clinically meaningful benefit for diversified prime and boost recombinant pox-viral-based vaccines in melanoma and ovarian cancer and support further evaluation of this approach in these patient populations.


Journal of Clinical Oncology | 2001

MCM2 Is an Independent Predictor of Survival in Patients With Non–Small-Cell Lung Cancer

Nithya Ramnath; Francisco J. Hernandez; Dongfeng Tan; Joel A. Huberman; Nachimuthu Natarajan; Amy Beck; Andrew Hyland; Ivan T. Todorov; John J. Brooks; Gerold Bepler

PURPOSE Minichromosome maintenance protein 2 (MCM2) is a component of the prereplicative complex. It is essential for eukaryotic DNA replication and is only expressed in proliferating cells. The prognostic utility of MCM2 compared with Ki-67, another marker of proliferating cells, on survival of patients with non-small-cell lung cancer (NSCLC) was studied. PATIENTS AND METHODS We examined the immunohistochemical expression of MCM2 and Ki-67 in primary pathologic tumor specimens from 221 NSCLC patients. For each marker, the fraction of tumor cells with positive staining was assessed as a percentage and categorized into four groups: 0% to 24%, 25% to 49%, 50% to 74%, and > or = 75%. MCM2 and Ki-67 immunoreactivities were compared with each other, and associations with pathologic and clinical parameters predictive of survival were analyzed with the chi(2) test. Cox regression models were used to assess associations between MCM2 and Ki-67 and survival while controlling for confounders. RESULTS Independent variables significantly associated with survival were tumor stage, performance status, and staining category. Patients with less than 25% MCM2 immunoreactivity had a longer median survival time than patients with > or = 25% MCM2 immunoreactivity (46 v 31 months; P =.039) and a lower relative risk (RR) of death (RR, 0.55, 95% confidence interval, 0.34 to 0.88). There was no significant association between survival and Ki-67 expression. CONCLUSION Immunostaining of tumor cells for MCM2 is an independent prognostic parameter of survival for patients with NSCLC. Interpretable results can be obtained on more than 96% of paraffin-embedded specimens, and approximately 35% will be in the favorable subgroup, with less than 25% positively stained tumor cells. Whether MCM2 is predictive of response to therapy needs to be studied.


Cancer Research | 2009

Efficacy of Levo-1-Methyl Tryptophan and Dextro-1-Methyl Tryptophan in Reversing Indoleamine-2,3-Dioxygenase-Mediated Arrest of T-Cell Proliferation in Human Epithelial Ovarian Cancer

Feng Qian; Jeannine Villella; Paul K. Wallace; Paulette Mhawech-Fauceglia; Joseph D. Tario; Chris Andrews; Junko Matsuzaki; Danila Valmori; Maha Ayyoub; P.J. Frederick; Amy Beck; Jianqun Liao; Richard T. Cheney; Kirsten B. Moysich; Shashikant Lele; Protul Shrikant; Lloyd J. Old; Kunle Odunsi

It has been reported that levo-1-methyl tryptophan (L-1MT) can block indoleamine-2,3-dioxygenase (IDO) expressed by human dendritic cells (DC), whereas dextro-1-methyl tryptophan (D-1MT) is inefficient. However, whether L-1MT or D-1MT can efficiently reverse IDO-induced arrest of human T-cell proliferation has not been clarified. Here, we show a marked immunosuppressive effect of IDO derived from INDO-transfected 293 cell, IDO+ ovarian cancer cells, and monocyte-derived DCs on CD4+ Th1 cells, CD8+ T cells, and natural killer cells derived from peripheral blood, ascites, and tumors of ovarian cancer patients. We found that, whereas L-1MT and D/L-1MT can restore proliferation of tumor-derived and peripheral blood T-cell subsets, D-1MT does not effectively restore IDO-induced arrest of T-cell proliferation. Although D-1MT inhibited kynurenine production at high concentrations, L-1MT was more effective in abrogating kynurenine generation and tryptophan depletion, whereas tryptophan was completely depleted by IDO even in the presence of high amounts of D-1MT. Together, the results indicate that, whereas the generation of tryptophan metabolites (kynurenines) by IDO is important in mediating suppression of T-cell proliferation, the degree to which tryptophan depletion is restored by 1MT is also critical in overcoming IDO-induced arrest of T-cell proliferation.


Cancer immunology research | 2014

Epigenetic Potentiation of NY-ESO-1 Vaccine Therapy in Human Ovarian Cancer

Kunle Odunsi; Junko Matsuzaki; Smitha R. James; Paulette Mhawech-Fauceglia; Takemasa Tsuji; Austin Miller; Wa Zhang; S.N. Akers; Elizabeth A. Griffiths; Anthony Miliotto; Amy Beck; Carl A. Batt; Gerd Ritter; Shashikant Lele; Sacha Gnjatic; Adam R. Karpf

Odunsi and colleagues show that the DNA methyltransferase inhibitor decitabine augmented the efficacy of the NY-ESO-1 vaccine and doxorubicin treatment of patients with refractory epithelial ovarian cancer, demonstrating the potential of the combined chemo-immunotherapy regimen. The cancer–testis/cancer germline antigen, NY-ESO-1, is a vaccine target in epithelial ovarian cancer (EOC), but its limited expression is a barrier to vaccine efficacy. As NY-ESO-1 is regulated by DNA methylation, we hypothesized that DNA methyltransferase inhibitors may augment NY-ESO-1 vaccine therapy. In agreement, global DNA hypomethylation in EOC was associated with the presence of circulating antibodies to NY-ESO-1. Preclinical studies using EOC cell lines showed that decitabine treatment enhanced both NY-ESO-1 expression and NY-ESO-1–specific CTL-mediated responses. On the basis of these observations, we performed a phase I dose-escalation trial of decitabine, as an addition to NY-ESO-1 vaccine and doxorubicin liposome chemotherapy, in 12 patients with relapsed EOC. The regimen was safe, with limited and clinically manageable toxicities. Both global and promoter-specific DNA hypomethylation occurred in blood and circulating DNAs, the latter of which may reflect tumor cell responses. Increased NY-ESO-1 serum antibodies and T-cell responses were observed in the majority of patients, and antibody spreading to additional tumor antigens was also observed. Finally, disease stabilization or partial clinical response occurred in six of ten evaluable patients. On the basis of these encouraging results, evaluation of similar combinatorial chemo-immunotherapy regimens in EOC and other tumor types is warranted. Cancer Immunol Res; 2(1); 37–49. ©2014 AACR.


Journal of Clinical Oncology | 2002

Prognostic Significance of Molecular Genetic Aberrations on Chromosome Segment 11p15.5 in Non–Small-Cell Lung Cancer

Gerold Bepler; Ashish Gautam; Lauren M. McIntyre; Amy Beck; David S. Chervinsky; Young-Chul Kim; Diana M. Pitterle; Andrew Hyland

PURPOSE The assessment of prognosis and decisions on treatment for patients with non-small-cell lung cancer (NSCLC) are determined on the basis of disease stage and performance status. NSCLC frequently manifests loss of heterozygosity (LOH) at chromosome segment 11p15.5. Whether LOH at 11p15.5 is an independent prognostic variable has yet to be determined. PATIENTS AND METHODS We developed five novel markers, which can be assessed by polymerase chain reaction and restriction enzyme digestion. LOH at 11p15.5 was assessed in 193 patients who underwent surgical resection for pathologic stage I and II of the disease. RESULTS LOH at 11p15.5 was associated with poor survival (P =.021). Multivariate logistic regression analysis showed that after disease stage, performance status, weight loss, sex, age at diagnosis, and smoking history were controlled for, patients with LOH were two times more likely to die than those without LOH (relative risk [RR] = 2.01, P =.021). Cox regression analysis with disease stage and LOH revealed that the survival of patients with stage I disease and LOH was similar to the survival of patients with stage II disease, and it was significantly worse than the survival of stage I patients without LOH (RR = 2.38, P =.038). CONCLUSION LOH in a 310-kb region on chromosome segment 11p15.5 that includes the gene for the regulatory subunit of the enzyme ribonucleotide reductase is highly predictive of poor survival from NSCLC. The future utility of analysis of the allelic status of this region may involve treatment decisions, such as the use of neoadjuvant and adjuvant chemotherapy for patients with stage I disease.


Diagnostic Molecular Pathology | 2003

HER-2/neu Protein Expression and Gene Alteration in Stage I-IIIA Non-Small-Cell Lung Cancer: A Study of 140 Cases Using a Combination of High Throughput Tissue Microarray, Immunohistochemistry, and Fluorescent In Situ Hybridization

Dongfeng Tan; George Deeb; Jianmin Wang; Harry K. Slocum; Janet Winston; Sam M. Wiseman; Amy Beck; Sheila N.J. Sait; Timothy M. Anderson; Chukwumere Nwogu; Nithya Ramnath; Gregory M. Loewen

Regarding HER-2/ neu expression (gene or protein level) in lung cancer, several studies with inconsistent results have been recently reported, partially due to variable techniques used and/or heterogeneous populations examined. The objective of this study was to examine HER-2/ neu expression in a well-defined cohort of non–small-cell lung cancers (NSCLC) and in nonneoplastic lung tissue utilizing a combination of high-density tissue microarray, immunohistochemistry (IHC), and fluorescent in situ hybridization (FISH) under uniform test conditions. One hundred forty stage I-IIIA primary NSCLCs and 38 non-neoplastic lung samples were examined. IHC, using an FDA-approved Hercept monoclonal antibody kit, was performed and HER-2/ neu gene alteration was assessed by FISH. The association of expression of HER-2/ neu with clinicopathologic parameters was analyzed. Ninety-four percent of tumor samples (131/140) were fully interpretable after tissue processing. Twenty-five of them (19%) overexpressed (2+, 3+) HER-2/ neu, while 106 (81%) had no or weak expression. All thirty-four interpretable non-neoplastic lung samples were negative for HER-2/ neu alteration at protein and gene level. HER-2/ neu protein overexpression correlated well with HER-2/ neu gene amplification (r =.83, P < 0.001). HER-2/ neu overexpression was significantly associated with histologic subtype: 19 adenocarcinomas (19/82, 23%) versus 4 squamous cell carcinomas (4/44, 9%) overexpressed Her-2/ neu (P = 0.04). Statistical significance was observed between HER-2/ neu expression and tumor differentiation, with strong positive (3+) expression observed more frequently in poorly differentiated tumors (P = 0.01). Patients with HER-2/ neu abnormalities, particularly HER-2/ neu gene amplification, exhibited a shorter survival (P = 0.043). The statistically significant difference (P < 0.005) between HER-2/ neu alteration in tumor samples(25/131, 19%) and in the nonneoplastic tissue (0/34, 0%) implies that HER-2/ neu may have a role in the carcinogenesis of NSCLC. The findings provide evidence supporting the hypothesis that the HER-2/ neu receptor may represent a useful molecular target in the treatment of NSCLC. The significant association of HER-2/ neu expression and gene amplification with poorly differentiated carcinoma compared with well differentiated carcinoma suggests that HER-2/ neu may be involved in NSCLC tumor evolution. Patients with HER-2/ neu gene amplification and strong positive expression of HER-2/ neu protein showed a strong tendency toward shorter survival.


BMC Cancer | 2001

MCM2 - a promising marker for premalignant lesions of the lung: a cohort study

Dong Feng Tan; Joel A. Huberman; Andrew Hyland; Gregory M. Loewen; John J. Brooks; Amy Beck; Ivan Todorov; Gerold Bepler

BackgroundBecause cells progressing to cancer must proliferate, marker proteins specific to proliferating cells may permit detection of premalignant lesions. Here we compared the sensitivities of a classic proliferation marker, Ki-67, with a new proliferation marker, MCM2, in 41 bronchial biopsy specimens representing normal mucosa, metaplasia, dysplasia, and carcinoma in situ.MethodsParallel sections were stained with antibodies against MCM2 and Ki-67, and the frequencies of staining were independently measured by two investigators. Differences were evaluated statistically using the two-sided correlated samples t-test and Wilcoxon rank sum test.ResultsFor each of the 41 specimens, the average frequency of staining by anti-MCM2 (39%) was significantly (p < 0.001) greater than by anti-Ki-67 (16%). In metaplastic lesions anti-MCM2 frequently detected cells near the epithelial surface, while anti-Ki-67 did not.ConclusionsWe conclude that MCM2 is detectable in 2-3 times more proliferating premalignant lung cells than is Ki-67. The promise of MCM2 as a sensitive marker for premalignant lung cells is enhanced by the fact that it is present in cells at the surface of metaplastic lung lesions, which are more likely to be exfoliated into sputum. Future studies will determine if use of anti-MCM2 makes possible sufficiently early detection to significantly enhance lung cancer survival rates.


Histopathology | 2006

Diagnostic utility of FLI-1 monoclonal antibody and dual-colour, break-apart probe fluorescence in situ (FISH) analysis in Ewing's sarcoma/primitive neuroectodermal tumour (EWS/PNET). A comparative study with CD99 and FLI-1 polyclonal antibodies.

Paulette Mhawech-Fauceglia; François Herrmann; Remedios Penetrante; Amy Beck; Sheila N.J. Sait; A. M. Block; Kunle Odunsi; J. Fisher; L. Balos; Richard T. Cheney

Aims:  To compare the sensitivity and specificity of the recently commercially available FLI‐1 monoclonal (FLI‐1m) antibody with the currently used antibodies [CD99 and FLI‐1 polyclonal (FLI‐1p)] in the diagnosis of Ewings sarcoma/primitive neuroectodermal tumour (EWS/PNET) and to determine the diagnostic value of the EWSR1 (22q12) dual‐colour, break‐apart rearrangement probe fluorescence in situ hybridization (FISH) technique.

Collaboration


Dive into the Amy Beck's collaboration.

Top Co-Authors

Avatar

Paulette Mhawech-Fauceglia

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Kunle Odunsi

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Shashikant Lele

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Austin Miller

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Junko Matsuzaki

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Sacha Gnjatic

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Anthony Miliotto

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Feng Qian

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Lloyd J. Old

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Protul Shrikant

Roswell Park Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge