Expression of Leukemia-Associated Antigens (LAA) in Patients with Acute Myelogenous Leukemia (AML).

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4497-4497
Author(s):  
Bjoern Schoettker ◽  
Peter Reimer ◽  
Volker Kunzmann ◽  
Hermann Einsele ◽  
Florian Weissinger

Abstract Introduction: Immunotherapeutic approaches may be effective additional options to conventional chemotherapy and stem cell transplantation regimens in the treatment of patients with acute myelogenous leukaemia (AML). Without eliciting significant side effects they may help to prolong disease-free survival or at least may delay the time to progression. Immuniotherapeutic strategies comprise vaccination with professional antigen-presenting dendritic cells or transfusion of T-cells with specific anti-leukemic activity. Leukemia-associated antigens are useful targets as they are expressed predominantly in the malignant cells and constitutively not detectable in normal tissue, or as they are at least significantly overexpressed in the leukemic blasts. To determine potential targets for specific immunotherapy in our patients with AML, we started to analyze the antigen profiles of these patients’ leukemia cells using the previously characterized leukemia-associated antigens survivin, WT1, and proteinase-3 (with the HLA-A2-specific nonameric peptide PR-1), and c-Ski, an antigen recently found to be predominantly but not exclusively overexpressed in AML with deletion or monosomy of chromosome 7, indicating a poor prognosis. Methods: The mRNA expression of the above mentioned antigens was examined by conventional RT-PCR. Blasts from patients with AML were collected at diagnosis or at first relapse from peripheral blood and analyzed. Results: The antigen expression pattern in comparison to the AML cell lines HL-60 and K-562 is shown in Table 1. Conclusions: Although treatment of AML patients became more effective during the last decades, high relapse rates contribute to the poor prognosis of the disease. Additional therapeutic strategies are needed to help and prevent disease relapse. Immunotherapy directed to LAA might elicit specific CTL responses effectively eliminating minimal residual disease. Using the LAA profile shown above, we should be able to determine at least one antigen in each patient with AML to serve as a target for individual specific immunotherapy with antigen-loaded autologous dendritic cells or /and specific autologous T-cells. Table 1: Expression pattern of the LAA survivin, WT1, proteinase-3 und c-Ski in AML cell lines HL-60 and K-562, and in seven patients with AML. +/++/+++ positive, - negative, n.d. not determined yet -actinβ survivin WT-1 PRO3 c-ski HL-60 +++ +++ ++ + +++ K-562 +++ ++ ++ − − B.R. +++ +++ ++ +++ +++ B.G. +++ − ++ + − B.C. +++ − n.d. n.d. n.d. E.B. +++ − − n.d. n.d. E.E. +++ +++ +++ ++ + S.T. +++ +++ +++ ++ + V.B. +++ +++ n.d. n.d. n.d.

2021 ◽  
Vol 12 ◽  
Author(s):  
Darina Ocadlikova ◽  
Mariangela Lecciso ◽  
Javier Martin Broto ◽  
Katia Scotlandi ◽  
Michele Cavo ◽  
...  

BackgroundHigh-grade sarcomas are a heterogeneous group of aggressive tumors arising in bone and soft tissues. After relapse, treatment options are limited. The multi-targeted receptor tyrosine kinase inhibitors (TKIs) sunitinib and inhibitor of PD-1 (anti-PD-1) nivolumab have shown antitumor activity in selected subtypes. In this study, we examine the role of TKIs and PD-1 based therapy in in vitro cocultures of sarcoma.MethodsThe human osteosarcoma (SaOS-2) and synovial sarcoma (SYO-1) cell lines were treated with sunitinib. After cell death and proliferation assessment, expression of PD-L1 was analyzed by flow cytometry. Sunitinib-treated sarcoma cells were cocultured with dendritic cells (DCs), and the phenotype of mature DCs was determined by flow cytometry. Mature DCs were cultured with autologous T cells. PD-1 expression on T cells, their proliferation, T regulatory cell (Tregs) induction and IFN-γ production, before and after nivolumab exposure, were analyzed.ResultsAlong with its anti-proliferative and direct pro-apoptotic effect on sarcoma cell lines, sunitinib prompted PD-L1 upregulation on sarcoma cells. Interestingly, sunitinib-treated sarcoma cells drive DCs to full maturation and increase their capacity to induce sarcoma-reactive T cells to produce IFN-γ. Conversely, no effect on T cell proliferation and T cell subpopulation composition was observed. Moreover, both bone and synovial sarcoma cell lines induced Tregs through DCs but sunitinib treatment completely abrogated Treg induction. Finally, sarcoma cell lines induced PD-1 upregulation on both effector T cells and Tregs when loaded into DCs, providing a rationale for using PD-1 blockade. Indeed, PD-1 blockade by nivolumab synergized with sunitinib in inducing IFN-γ-producing effector T cells.ConclusionsTaken together, our in vitro data indicate that the treatment of sarcoma cells with sunitinib can exert significant changes on immune cell subsets toward immune activation, leading to DC-based cross-priming of IFN-γ-producing effector T cells and reduced Treg induction. PD-1 blockade with nivolumab has a synergistic effect with sunitinib, supporting the use of TKI and anti-PD-1 approach in sarcomas, and perhaps in other cancers. DC-targeted drugs, including toll-like receptor 3 inhibitors and CD47 inhibitors, are under development and our preclinical model might help to better design their clinical application.


2021 ◽  
Vol 2021 ◽  
pp. 1-11
Author(s):  
Yue Luo ◽  
Junhao Lin ◽  
Jiakang Zhang ◽  
Zhenghui Song ◽  
Dayong Zheng ◽  
...  

Long noncoding RNAs (lncRNAs) have been substantially reported to have critical roles in regulating tumorigenesis in recent years. However, the expression pattern and biological function of SNHG17 in hepatocellular carcinoma (HCC) remain unclear. Bioinformatics analysis and qRT-PCR were performed to detect the expression pattern of SNHG17 in HCC tissues, adjacent nontumorous tissues, and cell lines. The effect of SNHG17 on proliferation, migration, and apoptosis of HCC was investigated by knockdown and overexpressing SNHG17 in HCC cell lines. RNA sequencing was utilized to explore the underlying mechanism. Utilizing publicly available TCGA-LIHC, GSE102079 HCC datasets, and qRT-PCR, we found SNHG17 was significantly upregulated in HCC tissues and cell lines and was notably associated with larger tumor size, poorly differentiation, presence of vascular invasion, and advanced TNM stage. Furthermore, gain- and loss-of-function studies demonstrated that SNHG17 promoted cell proliferation and migration and inhibited apoptosis of HCC. By employing RNA sequencing, we found knockdown of SNHG17 caused 1037 differentially expressed genes, highly enriched in several pathways, including metabolic, PI3K-Akt, cell adhesion, regulation of cell proliferation, and apoptotic pathway; among them, 92 were overlapped with SNHG17-related genes in the TCGA-LIHC dataset. Furthermore, ERH, TBCA, TDO2, and PDK4 were successfully validated and found significantly dysregulated in HCC tissues. Moreover, HCC patients with higher SNHG17 expression had a relatively poor overall survival and disease-free survival, and ERH and PDK4 also played a marked role in the prognosis of HCC. Broadly, our findings illustrate that SNHG17 acts as a noncoding oncogene in HCC progression, suggesting its potential value as a novel target for HCC therapy.


2020 ◽  
Vol 2020 ◽  
pp. 1-8
Author(s):  
Mirela Cioplea ◽  
Luciana Nichita ◽  
Daniela Georgescu ◽  
Liana Sticlaru ◽  
Alexandra Cioroianu ◽  
...  

Cutaneous melanoma is a significant immunogenic tumoral model, the most frequently described immune phenomenon being tumor regression, as a result of the interaction of tumoral antigens and stromal microenvironment. We present a retrospective cohort study including 52 cases of melanoma with regression. There were evaluated correlations of the most important prognostic factors (Breslow depth and mitotic index) with FOXP3 expression in tumor cells and with the presence of regulatory T cells and dendritic cells in the tumoral stroma. FOXP3 expression in tumor cells seems an independent factor of poor prognosis in melanoma, while regression areas are characterized by a high number of dendritic cells and a low number of regulatory T cells. FOXP3 is probably a useful therapeutical target in melanoma, since inhibition of FOXP3-positive tumor clones and of regulatory T cells could eliminate the ability of tumor cells to escape the immune defense of the host.


Blood ◽  
1999 ◽  
Vol 93 (3) ◽  
pp. 780-786 ◽  
Author(s):  
A. Choudhury ◽  
J.C. Liang ◽  
E.K. Thomas ◽  
L. Flores-Romo ◽  
Q.S. Xie ◽  
...  

Abstract We have previously reported that leukemic dendritic cells (DC) can be generated ex vivo from myelomonocytic precursors in chronic myelogenous leukemia. In this study we report the generation of DC from acute myelogenous leukemia (AML) cells and their potent ability to stimulate leukemia-specific cytolytic activity in autologous lymphocytes. DC were generated in vitro using granulocyte-macrophage colony-stimulating factor +interleukin-4 in combination with either tumor necrosis factor- or CD40 ligand (CD40L). Cells from 19 AML patients with a variety of chromosomal abnormalities were studied for their ability to generate DC. In all but 1 case, cells with the morphology, phenotypic characteristics, and T-cell stimulatory properties of DC could be generated. These cells expressed high levels of major histocompatibility complex class I and class II antigens as well as the costimulatory molecules B7-2 and ICAM-1. In three cases these cells were determined to be of leukemic origin by fluorescence in situ hybridization for chromosomal abnormalities or Western blotting for the inv(16) fusion gene product. Autologous lymphocytes cocultured with AML-derived DC (DC-AL) were able to lyse autologous leukemia targets, whereas little cytotoxicity was noted against autologous, normal cells obtained from the patients during remission. We conclude that leukemia derived DC may be useful for immunotherapy of many AML patients.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1131-1131
Author(s):  
Fernando J. Suarez Saiz ◽  
Serban San-Marina ◽  
Mark D. Minden

Abstract Acute myelogenous leukemia (AML) arises due to changes in gene expression that block or alter the normal differentiation program of hematopoietic stem cells. A variety of mutations in protein-encoding genes have been shown to contribute to the development of leukemia. Recently a new class of genes called microRNAs (miRNAs) have been identified. miRNAs are a subgroup of highly conserved, non-coding RNAs found only in eukaryotes. They do not encode proteins, and appear to have a significant effect on the proteome of a cell. Their conservation between species suggests their involvement in important biological functions, and in fact been shown to be involved in hematopoietic differentiation. While the function of most miRNAs is still unknown, it is believed that they regulate expression of target mRNAs by using the siRNA machinery either to promote degradation of the mRNA or to block its translation. To begin to understand the role of miRNAs in AML, we used Quantitative Polymerase Chain Reaction (QPCR) to measure the expression level of 20 miRNA precursors in the pro erythroid cell line K562, the pro-myelocytic cell line NB4, the myelomococytic cell line OCI/AML2, AML patients’ blasts and in normal bone marrow (NBM). The investigated miRNAs included some that are known to be specific for hematopoietic tissues or involved in hematopoietic differentiation, as well as all the miRNAs in chromosome 7, a hot spot for gene deletion in AML. Our findings indicate that miRNAs are differentially expressed in patients and cell lines when compared among themselves and against normal bone marrow. For example pre-miR-142 was expressed in NBM and K562 but was found to be elevated in OCI/AML2, NB4 and in all patient samples. Pre-miR-20 was found to be overexpressed in only a subset of patients. Other miRNAs like pre-miR-335 and pre-miR-148a were expressed in NBM and in some patients and not in the cell lines. In an effort to identify possible regulators of miRNA expression, we analyzed the upstream region of pre-miR-142 and found an LMO2 binding site. In AML, the LMO2 gene can be overexpressed relative to normal bone marrow and healthy lymphocytes. This transcription factor is involved in the regulation of genes important in the development of blood cells. To investigate if LMO2 could be involved in the regulation of miR-142 expression, we performed chromatin immunoprecipitation (ChIP) from K562 using an anti-LMO2 antibody. Only the LMO2 immunoprecipitation, and not those from the pre-immune control, were enriched in promoter DNA for pre-miR-142. This is consistent with the observation that miRNAs and coding RNAs can be regulated by the same environmental signals. Based on this observation we propose that oncogenes regulate in part the phenotype and biological behaviour of leukemia by affecting the expression of miRNAs. This further suggests that different forms of leukemia may be recognized based upon the spectrum of miRNAs they express.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4555-4555
Author(s):  
Naibai Chang ◽  
Jianping Wei ◽  
Shengming Zhao ◽  
Hui Liu ◽  
Yun Fan ◽  
...  

Abstract Objective: To evaluate response of homoharringtonine in patients with high risk AML or as a second-line therapy in patients with AML refractory to anthracycline based chemotherapy. Patients and methods: From Jan 1998-Jan 2006, there were 66 patients enrolled in this regimen. Male:female=40:26. Median age was 47 (17–69). There were 48 newly diagnosed AML, 5 relapsed AML and 13 secondary AML (secondary to MDS) in this group. Among these patients, there were 40 patients untreated previously — 20 patients had unfavourable chromasomal abmormalities, 10 patients had pgp positive, 10 patients had normal chromasome but high CD34 expression on leukemic cells. The remaining 26 patients were previously treated with daunorubincin or mitoxantrane combined with cytarabine at least two cycles and failed to achieve response. Homoharringtonine was given at dose of 4mg/m2/d(2.5mg/m2/d for age≥60 years) intraveniously for 7 days. Cytarabine was given at dose of 100mg/m2/d at same time. The therapy was repeated every 21 days.Post remission therapy was divided into two cohot —same regimen maintainance in 20 cases and cytarabine 1g/m2/d q12h for 4 days at least 4 cycles in 20 cases. Results: 40/66 patients achieved complete remission. 3/66 achieved partial remission. In patients refactory to anthracycline based regimen, the CR rate was 57.7%(15/26). In previously untreated high risk AML patients,the CR rate was 62.5%(25/40). Median disease free survival were 4.25 months (2–30) in cohot 1 and 18 months (12–47) in cohot 2 (P<0.05). CD95(APO-1/Fas) was tested by flow cytometry during treatment. APO-1/Fas (CD95) increased from (9.56±5.58)% to (25.64±0.70)% after induction chemotherapy. Main toxicities were marrow suppression and infection. There were 7 early deaths, 5 patients died of cerebral hemorrage and 2 cardiovascular events. Conclusions: Homoharringtonine is effective in patients with high risk AML. It is also a choice of second line therapy in patients refractory to anthracycline based chemotherapy. Intensive post remission therapy is superior to conventional dose maintainance therapy in high risk AML. One of the ways for homoharringtonine to induce leukemic cell apoptosis is probably through Fas pathway.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4905-4905
Author(s):  
Timothy J Brown ◽  
Brian Barth ◽  
David F. Claxton

Abstract Abstract 4905 Background: While understanding of Acute Myelogenous Leukemia (AML) pathogenesis has advanced greatly in recent years, drug discovery and development have added little to therapy. Ceramide and other sphingolipids are of therapeutic interest for many neoplasms (Jiang Y, DiVittore NA, Kaiser JM, et al. Combinatorial therapies improve the therapeutic efficacy of nanoliposomal ceramide for pancreatic cancer. Cancer Biol Ther. Oct 1 2011;12(7):574–585). Cellular ceramide accumulation favors a pro-apoptotic state, while accumulation of sphingosine-1-phosphate promotes survival. We have targeted the ceramide balance of AML cells in vitro with varying concentrations of liposomal formulations of C6 ceramide (Lip-C6), the sphingosine kinase-1 inhibitor safingol (Lip-Saf), and tamoxifen (Lip-Tam) to determine potential synergistic anti-leukemic efficacy. Safingol is a sphingosine kinase inhibitor currently in phase 1 trials. In addition, Tamoxifen can reverse drug resistance of many cancer cell types (Chapman JV, Gouaze-Andersson V, Messner MC, et al. Metabolism of short-chain ceramide by human cancer cells–implications for therapeutic approaches. Biochem Pharmacol. Aug 1 2010;80(3):308–315.). This has been shown to be due to the ability of Tamoxifen to block the activities of glucosylceramide synthase (GCS) and p-glycoprotein (P-GP), which coordinate to detoxify ceramide at the Golgi membrane. In the present study, liposomal drug formulations were chosen to enhance drug delivery and prevent premature drug metabolism. Methods: The cell lines C1498, HL-60, HL-60/VCR, GFPp210, Wehi-3B, K562, U937, and KG-1 were used in this study. Drugs were synthesized into liposomal formulations by the Penn State Hershey Drug Discovery and Delivery Core laboratory. Cellular viability was measured after treatment with Lip-C6, Lip-Saf, Lip-Tam, or a combination for 48 hours. Synergy and dose-response curves were modeled using CalcuSyn software. Apoptosis and cell proliferation were assessed using flow cytometry after treatment of drug for 24 hours at the calculated IC50 from MTS assays. Autophagy was also measured in C1498 cells to confirm an established safingol cell-death mechanism. Primary human AML collected by our lab from consenting, patients was assessed in methylcellulose for blast clonogenicity. Results and Conclusions: Several cell lines showed a favorable change in the IC50 of the drugs when used in combination, indicating a possible synergistic anti-leukemic mechanism of action. When Lip-C6 was combined with Lip-Saf in a varying ratios, synergistic growth inhibition was observed in the human AML cell lines HL-60, HL-60/VCR, and KG-1 (Figure 1). Interestingly, Lip-Tam caused complete cell population death at concentrations less than 15 μM in the Wehi-3B, K562, GFPp210, and C1498 lines. When cells were treated with Lip-C6 and Lip-Tam in a 1:1 combination, complete cell population killing was noted at concentrations of less than 10 μM in every cell line tested. Additionally, flow cytometric data confirmed findings of other investigators suggesting that Lip-Saf caused enhanced autophagy. Therefore, the observed synergistic leukemia cell death is likely due in part to the novel combination of an autophagy-inducer with an apoptosis-inducer. Clonogenic data has shown that combination of Lip-C6and Lip-Saf cooperate to inhibit formation of blast colonies from human AML, indicating a potential use in lessening leukemia burden (Figure 1). In conclusion, novel ceramide-centered drug combinations promote improved cell death of leukemia cell lines via accumulation of ceramide and inhibition of ceramide metabolic pathways. This study acts as a persuasive proof-of-concept of the effect of inhibiting a single ceramide degradation pathway within cells. By inhibiting sphingosine kinase-1 or the GCS activity of P-GP in these cell lines, it becomes apparent that the cellular ceramide balance shifts to favor a pro-apoptotic state. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document