scholarly journals CD 80 and CD 86 Expression, Clinical Implications Are Cancer Dependent as Revealed Through Pan-cancer Analysis

Author(s):  
Ayobami Matthew Olajuyin ◽  
Sharon Ibialate Georgewill ◽  
Adefunke Kafayat Olajuyin ◽  
Jamiyu Ayodeji Saliu ◽  
Malachy I. Okeke ◽  
...  

Abstract BackgroundCluster of Differentiation 80 and CD 86 can also be called B7-1 and B7-2 respectively. They are proteins fundamentally expressed on antigen-presenting cells (APCs), including induced dendritic cells (IDCs), langerhans cells, germinal center dendritic cells (GCDCs), activated monocytes, macrophages and B-cells. They are considered to be a possible therapeutic target and biomarker of great significance. However, there are still inconsistent pieces of information and their clinical importance is yet to be established. MethodsHere we investigated CD 80 and 86 as biomarkers by utilizing several large genomic data collections. (The Cancer Genome Atlas, Cancer Cell Line Encyclopedia, Quantitative proteomics Cancer cell line Encyclopedia Genotype-Tissue Expression,) and analyzed CD 80 and CD 86 expression in thousands of normal and cancer samples and cell lines along with their clinical survival analysis.ResultsThis study presented that CD 86 was expressed more in post-treatment blood cancer in the blood and post-treatment blood cancer in the bone marrow while it was expressed least in normal tissues and cell lines. The Hodgkin lymphoma cell line L428 cell lysate illustrated that there was a high relative protein expression of 6.6 for the CD 86 gene. it indicated that cancer in the esophagus had the highest copy number value and indicated a medium level amplification of the CD 86 gene and prostate cancer had a hemizygous deletion of the CD 86 gene with the least copy number value. Furthermore, on the non-Hodgkin lymphoma cell line REC1, illustrated the highest relative protein expression of the CD 86 gene among the other types of cancer cell line, its protein expression value was 8.19. Also, for cancer type leukemia, the subtype acute myeloid leukemia showed a significant relative protein expression. The acute myeloid leukemia cell line EOL1 indicated that there was a high relative protein expression of 6.5. However, the protein expression for CD 80 is yet to be elucidated.ConclusionsTaken together, CD 80 ad 86 may be potential biomarkers of great clinical significance. The Kaplan Meier plots unveiled that CD 86 and CD 80 were significantly associated with overall survival analysis in the Large B-cell lymphoma, and the different tumor types.

2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Julia Stomper ◽  
Ruth Meier ◽  
Tobias Ma ◽  
Dietmar Pfeifer ◽  
Gabriele Ihorst ◽  
...  

Abstract Background Mutations in the EZH2 gene are recurrently found in patients with myeloid neoplasms and are associated with a poor prognosis. We aimed to characterize genetic and epigenetic alterations of EZH2 in 58 patients (51 with acute myeloid leukemia and 7 with myelodysplastic or myeloproliferative neoplasms) by integrating data on EZH2 mutational status, co-occurring mutations, and EZH2 copy number status with EZH2 protein expression, histone H3K27 trimethylation, and EZH2 promoter methylation. Results EZH2 was mutated in 6/51 acute myeloid leukemia patients (12%) and 7/7 patients with other myeloid neoplasms. EZH2 mutations were not overrepresented in patients with chromosome 7q deletions or losses. In acute myeloid leukemia patients, EZH2 mutations frequently co-occurred with CEBPA (67%), ASXL1 (50%), TET2 and RAD21 mutations (33% each). In EZH2-mutated patients with myelodysplastic or myeloproliferative neoplasms, the most common co-mutations were in ASXL1 (100%), NRAS, RUNX1, and STAG2 (29% each). EZH2 mutations were associated with a significant decrease in EZH2 expression (p = 0.0002), which was similar in patients with chromosome 7 aberrations and patients with intact chromosome 7. An association between EZH2 protein expression and H3K27 trimethylation was observed in EZH2-unmutated patients (R2 = 0.2, p = 0.01). The monoallelic state of EZH2 was not associated with EZH2 promoter hypermethylation. In multivariable analyses, EZH2 mutations were associated with a trend towards an increased risk of death (hazard ratio 2.51 [95% confidence interval 0.87–7.25], p = 0.09); similarly, low EZH2 expression was associated with elevated risk (hazard ratio 2.54 [95% confidence interval 1.07–6.04], p = 0.04). Conclusions Perturbations of EZH2 activity in AML/MDS occur on different, genetic and non-genetic levels. Both low EZH2 protein expression and, by trend, EZH2 gene mutations predicted inferior overall survival of AML patients receiving standard chemotherapy.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1446-1446 ◽  
Author(s):  
Nianxi Zhao ◽  
Sung-nan Pei ◽  
Jianjun Qi ◽  
Pei Lin ◽  
Youli Zu

Abstract The current treatment paradigm for acute myeloid leukemia (AML) is remission induction chemotherapy, followed by either consolidation chemotherapy or allogeneic stem cell transplantation. As most patients diagnosed with AML are in their sixth or seventh decade of life, many are not candidates for standard remission induction chemotherapy because of the risk of toxicities, such as profound myelosuppression, life-threatening infections, and cardiotoxicity. The development of new effective and safe treatments for AML is therefore needed. The “ideal” therapy should specifically target AML tumor cells with no side-effect on normal cells. Since CD117 (c-Kit) is a transmembrane receptor on tumor cells surface and expresses in 70% cases of AML, it is a potential molecule for developing new targeted therapy. Aptamers are single-stranded oligonucleotides (DNA or RNA), which have the ability to specifically bind to their targets with high affinity. As a “chemical antibody”, aptamers can be chemically synthesized, easily conjugated with therapeutic drugs, and more importantly, less or not immunogenic. In this study, we developed single strand DNA (ssDNA) aptamer specific for CD117 by using a unique hybrid SELEX approach with both cell-selection and protein-enrichment. When sequencing the enriched ssDNA library with million reads, one dominant sequence had over 80% copies of total sequence reads. Cell binding analysis of the aptamer by flow cytometry and fluorescent microscopy (figure 1) demonstrated that, the aptamer molecule specifically bound to CD117-expressing AML cells, but did not react to CD117-negative control cells including Histiocytic lymphoma Cell line: U937, Burkitt's lymphoma cell line: CA46, Breast adenocarcinoma cell line: 468 and human prostate carcinoma cell line: LNCap. In addition, the presence of aptamers inhibited cell binding anti-CD117 antibody, indicating that aptamer targeted CD117 receptor on leukemic cells. Notably, this aptamer specifically targeted CD117-positve AML cells of clinical specimens with an identical staining pattern to that observed with anti-CD117 antibody (figure 2), indicating potential for in clinical use. For targeted therapy, an aptamer -drug-conjugate was formulated by chemical conjugation of ssDNA CD117 aptamer (Apt) to chemotherapeutic drug, Methotrexate (MTX). For treatment study, cell mixture of leukemic cells HEL (CD117+) and U937 (CD117-), which showed the same sensitivity to free MTX, were used (figure 3A). Exposure of cells to the Apt-MTX revealed that the formed aptamer-drug-conjugate specifically killed CD117 positive cells, but had no effect on the growth of the off-target cells in the same cultures. For therapeutic effect, Apt-MTX killed 60% of positive cells at as low as 10nM final concentration (figure 3B). In contrast, under the same condition, free MTX had no effect on cell growth (figure 3A). Our study demonstrated that the aptamer-drug-conjugate could be a new targeted therapeutics in addition to current antibody-drug-conjugate. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1427-1427 ◽  
Author(s):  
Tristan Knight ◽  
Xinan Qiao ◽  
Holly Edwards ◽  
Hai Lin ◽  
Jeffrey W. Taub ◽  
...  

Abstract Introduction: FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase, and is mutated in approximately one third of acute myeloid leukemia (AML) patients; this mutation confers a poor prognosis. Two FLT3 mutations are commonly seen in AML: internal tandem duplications (ITD) in the juxtamembrane domain (~25% of AML), and point mutations in the receptor tyrosine kinase at codon 835 (D835) (~7% of AML). Both mutations result in constitutive FLT3 activation, causing downstream activation of multiple pathways, in particular, those involved in cell survival including the RAS-RAF-MEK-ERK, JAK-STAT5, and PI3K/AKT pathways. PI3K-AKT may also be activated by AXL, also a tyrosine kinase, via its targets PLC, Grb2, and PI3K. Logically, then, inhibition of FLT3 is a promising pharmacological approach for treating this subtype of AML. Gilteritinib (ASP-2215) is a novel dual inhibitor of FLT3 and AXL, exposure to which results in upregulation of FLT3 as a resistance mechanism. Previously, we found that the novel dual PI3K/histone deacetylase (HDAC) inhibitor CUDC-907 downregulates FLT3 expression in AML cells (Figure 1A). Additionally, inhibition of FLT3 and AXL by gilteritinib may not result in robust inactivation of both the PI3K-Akt and MEK/ERK pathways due to crosstalk between the two pathways. Thus, our hypothesis was that CUDC-907 would sensitize AML cells to gilteritinib, resulting in concurrent inhibition of all the downstream signaling pathways of FLT3 and AXL, leading to synergistic antileukemic activities again FLT3-mutated AML (Figure 1B). Methods: FLT3-ITD AML cell lines (MV4-11 and MOLM-13) and primary patient samples were treated with CUDC-907, gilteritinib, both, or neither for 24 hours, at clinically achievable concentrations. Annexin V/Propidium Iodide (PI) staining and flow cytometry analyses was performed, and combination indexes (CI) calculated; CI<1, CI=1, and CI>1 indicating synergistic, additive, or antagonistic effects, respectively. Western blots were performed after treatment for 0-24 hours to determine protein expression of relevant targets. Results: CUDC-907 and gilteritinib demonstrated potent synergistic antileukemic effects in FLT3-ITD AML cell lines and FLT3-ITD patient samples (AML#171, AML#180), the combination exceeding either in isolation (Figure 1C). These findings were confirmed via western blot, which showed accentuated upregulation of cleaved caspase3 with combination therapy, in both cell lines and one patient sample, demonstrating drug-induced apoptosis. We confirmed that CUDC-907 abolishes gilteritinib-induced expression of FLT3 in a time-dependent fashion in cell lines MV4-11 and MOLM-13 (Figure 1D). Gilteritinib treatment decreased p-AKT, p-S6, and p-STAT5, while inhibition of the ERK pathway, as assessed by p-ERK expression, varied amongst the samples (Figure 1E). CUDC-907 treatment decreased both p-AKT and p-ERK. MOLM-13 cells showed increased p-ERK following gilteritinib treatment and increased p-STAT5 after CUDC-907 treatment. In all samples, combination of gilteritinib with CUDC-907 resulted in decrease of p-STAT5 and p-S6, similar to gilteritinib treatment alone, and further reduction of p-AKT and p-ERK compared to single drug treatments. Gilteritinib treatment also reduced expression of anti-apoptotic protein Mcl-1, which was further decreased in combination treated cells. Subsequently, time-course analysis was performed in both cell lines; findings were consistent with prior observations, and confirmed that protein expression changed over time, in relation to gilteritinib/CUDC-907/combined treatment exposure. Conclusion: We confirmed that CUDC-907 and Gilteritinib synergistically induce apoptosis in both cell lines and primary patient samples derived from patients with FLT3-ITD AML, and that CUDC-907 abolishes Gilteritinib-induced FLT3 expression. Additionally, the combination cooperatively inhibits the PI3K-AKT, JAK-STAT, and RAS-RAF pathways, while preventing escape via alternative pathways. Our results provide a strong foundation for subsequent in vivo murine studies, and eventual clinical evaluation of the combination of gilteritinib and CUDC-907 for the treatment of AML. Figure 1. Figure 1. Disclosures Ge: MEI Pharma: Research Funding.


Leukemia ◽  
2012 ◽  
Vol 26 (8) ◽  
pp. 1842-1849 ◽  
Author(s):  
A Zebisch ◽  
A Wölfler ◽  
I Fried ◽  
O Wolf ◽  
K Lind ◽  
...  

2011 ◽  
Author(s):  
Michael Roberts ◽  
David Bittner ◽  
Sarah Brnich ◽  
Bryan Conner ◽  
Carla Cox ◽  
...  

2015 ◽  
Vol 38 (2) ◽  
pp. e23-e26
Author(s):  
L. Li ◽  
X.L. Qi ◽  
X.H. Chen ◽  
F.G. Ren ◽  
Z.F. Xu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document