Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Crit Rev Biomed Eng ; 52(2): 39-50, 2024.
Article in English | MEDLINE | ID: mdl-38305277

ABSTRACT

Deep learning offers a promising methodology for the registration of prostate cancer images from histopathology to MRI. We explored how to effectively leverage key information from images to achieve improved end-to-end registration. We developed an approach based on a correlation attention registration framework to register segmentation labels of histopathology onto MRI. The network was trained using paired prostate datasets of histopathology and MRI from the Cancer Imaging Archive. We introduced An L2-Pearson correlation layer to enhance feature matching. Furthermore, our model employed an enhanced attention regression network to distinguish between key and nonkey features. For data analysis, we used the Kolmogorov-Smirnov test and a one-sample t-test, with the statistical significance level for the one-sample t-test set at 0.001. Compared with two other models (ProsRegNet and CNNGeo), our model exhibited improved performance in Dice coefficient, with increases of 9.893% and 2.753%, respectively. The Hausdorff distance was reduced by approximately 50% and 50%, while the average label error (ALE) was reduced by 0.389% and 15.021%. The proposed improved multimodal prostate registration framework demonstrated high performance in statistical analysis. The results indicate that our enhanced strategy significantly improves registration performance and enables faster registration of histopathological images of patients undergoing radical prostatectomy to preoperative MRI. More accurate registration can prevent over-diagnosing low-risk cancers and frequent false positives due to observer differences.


Subject(s)
Deep Learning , Prostatic Neoplasms , Male , Humans , Prostate/diagnostic imaging , Prostate/pathology , Prostate/surgery , Magnetic Resonance Imaging/methods , Prostatic Neoplasms/diagnostic imaging , Image Processing, Computer-Assisted/methods
2.
Transfusion ; 62(11): 2334-2348, 2022 11.
Article in English | MEDLINE | ID: mdl-36239134

ABSTRACT

BACKGROUND: There are two FDA-approved anti-CD38 monoclonal antibodies for treatment of multiple myeloma: isatuximab and daratumumab. Owing to expression of CD38 on reagent red blood cells (RBCs), these antibodies interfere with indirect antiglobulin tests (IATs). We sought to understand differences in such interference by performing binding experiments. STUDY DESIGN AND METHODS: In vitro experiments to compare the binding to RBCs of isatuximab and daratumumab alone or in the presence of a mouse anti-human CD38 antibody (HB-7 or AT13/5) or a nicotinamide adenine dinucleotide-analog CD38 inhibitor were performed and quantified by flow cytometry, imaging, mass spectrometry, surface plasmon resonance, and LigandTracer technologies. Serologic testing was performed on plasma samples spiked with isatuximab or daratumumab. RESULTS: CD38 expressed on RBCs can be directly bound by daratumumab, whereas isatuximab requires a co-factor, such as HB-7, AT13/5, or a CD38 inhibitor, suggesting that the isatuximab epitope on RBCs is masked in vitro. Daratumumab samples more frequently showed interference and had stronger reactions than isatuximab samples. Dithiothreitol treatment was equally effective in mitigating the interference caused by either drug. DISCUSSION: Both isatuximab and daratumumab interfere with IATs but at different magnitudes, reflecting distinct binding to CD38 on RBCs. From the binding studies, we conclude that the isatuximab epitope on RBCs is masked in vitro and binding requires a certain CD38 conformation or co-factor. This circumstance may explain why interference is seen only in a subset of patients receiving isatuximab when compared with interference seen in most patients on daratumumab therapy.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Neuroblastoma , Mice , Animals , ADP-ribosyl Cyclase 1 , Epitope Mapping , Antibodies, Monoclonal , Multiple Myeloma/therapy , Antineoplastic Agents/therapeutic use , Neuroblastoma/drug therapy , Epitopes
4.
Nature ; 603(7900): 328-334, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35197632

ABSTRACT

Effective antitumour immunity depends on the orchestration of potent T cell responses against malignancies1. Regression of human cancers has been induced by immune checkpoint inhibitors, T cell engagers or chimeric antigen receptor T cell therapies2-4. Although CD8 T cells function as key effectors of these responses, the role of CD4 T cells beyond their helper function has not been defined. Here we demonstrate that a trispecific antibody to HER2, CD3 and CD28 stimulates regression of breast cancers in a humanized mouse model through a mechanism involving CD4-dependent inhibition of tumour cell cycle progression. Although CD8 T cells directly mediated tumour lysis in vitro, CD4 T cells exerted antiproliferative effects by blocking cancer cell cycle progression at G1/S. Furthermore, when T cell subsets were adoptively transferred into a humanized breast cancer tumour mouse model, CD4 T cells alone inhibited HER2+ breast cancer growth in vivo. RNA microarray analysis revealed that CD4 T cells markedly decreased tumour cell cycle progression and proliferation, and also increased pro-inflammatory signalling pathways. Collectively, the trispecific antibody to HER2 induced T cell-dependent tumour regression through direct antitumour and indirect pro-inflammatory/immune effects driven by CD4 T cells.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Female , Humans , Mice , Receptor, ErbB-2/genetics
5.
Mol Cancer Ther ; 20(10): 1916-1925, 2021 10.
Article in English | MEDLINE | ID: mdl-34376579

ABSTRACT

This study reports the pharmacologic effects of isatuximab, a CD38 mAb, on T- and B-cell acute lymphoblastic leukemia (ALL). We analyzed CD38 expression in 50-T-ALL and 50 B-ALL clinical samples, and 16 T-ALL and 11 B-ALL cell lines. We primarily focused on in vitro assessments of isatuximab-mediated antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). In vivo assessment of isatuximab activity was performed in several ALL xenograft models, including disseminated and subcutaneous tumor models in female C.B-17 severe combined immunodeficiency mice. Our study reveals that most patients (90%-100%) carried CD38+ blasts independent of disease burden. The median CD38 receptor density on abnormal lymphoblasts is 41,026 copies/cell on T-ALL and 28,137 copies/cell on B-ALL, respectively. In patients with T-ALL, there is a significant increase of CD38 expression in abnormal blasts compared with normal T cells. High-level CD38 receptor density (RD) is critical to trigger effective isatuximab-mediated ADCC against target ALL cells. In addition, a correlation between CD38 RD and isatuximab-mediated ADCP is demonstrated. In the disseminated CD38+, T-ALL, and B-ALL xenograft models, isatuximab is able to induce robust antitumor activity, even at low doses. This study shows that isatuximab has significant in vitro and in vivo activity against ALL cells with robust ADCC and ADCP effects that are associated with CD38 expression levels in both T-ALL and B-ALL.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Front Immunol ; 11: 1771, 2020.
Article in English | MEDLINE | ID: mdl-32922390

ABSTRACT

Isatuximab is a monoclonal antibody targeting the transmembrane receptor and ectoenzyme CD38, a protein highly expressed on hematological malignant cells, including those in multiple myeloma (MM). Upon binding to CD38-expressing MM cells, isatuximab is thought to induce tumor cell killing via fragment crystallizable (Fc)-dependent mechanisms, including antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC), as well as via direct Fc-independent mechanisms. Here, these mechanisms of action were investigated in MM and diffuse large B-cell lymphoma (DLBCL) cell lines, as well as in peripheral blood mononuclear cells derived from healthy donors, and in MM patient-derived samples. Our findings show that isatuximab-mediated cytotoxicity occurred primarily via ADCC and ADCP in MM cell lines and via ADCC and apoptosis in DLBCL cell lines expressing high levels of CD38. We identified the programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) pathway and MM cell-secreted transforming growth factor-beta (TGF-ß) as tumor cell-related features that could suppress CD38-mediated ADCC. Furthermore, we established that isatuximab can directly activate natural killer (NK) cells and promote NK cell-mediated cytotoxicity via crosslinking of CD38 and CD16. Finally, isatuximab-induced CDC was observed in cell lines with high CD38 receptor density (>250,000 molecules/cell) and limited expression of inhibitory complement regulatory proteins (CD46, CD55, and CD59; <50,000 molecules/cell). Taken together, our findings highlight mechanistic insights for isatuximab and provide support for a range of combination therapy approaches that could be tested for isatuximab in the future.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Multiple Myeloma/immunology , Apoptosis/drug effects , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects
7.
Nat Cancer ; 1(1): 86-98, 2020 01.
Article in English | MEDLINE | ID: mdl-35121834

ABSTRACT

Despite the significant therapeutic advances provided by immune-checkpoint blockade and chimeric antigen receptor T cell treatments, many malignancies remain unresponsive to immunotherapy. Bispecific antibodies targeting tumor antigens and activating T cell receptor signaling have shown some clinical efficacy; however, providing co-stimulatory signals may improve T cell responses against tumors. Here, we developed a trispecific antibody that interacts with CD38, CD3 and CD28 to enhance both T cell activation and tumor targeting. The engagement of both CD3 and CD28 affords efficient T cell stimulation, whereas the anti-CD38 domain directs T cells to myeloma cells, as well as to certain lymphomas and leukemias. In vivo administration of this antibody suppressed myeloma growth in a humanized mouse model and also stimulated memory/effector T cell proliferation and reduced regulatory T cells in non-human primates at well-tolerated doses. Collectively, trispecific antibodies represent a promising platform for cancer immunotherapy.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Animals , Antibodies, Bispecific/therapeutic use , CD28 Antigens , Mice , Multiple Myeloma/drug therapy , Receptors, Antigen, T-Cell , T-Lymphocytes
8.
PLoS One ; 6(7): e21503, 2011.
Article in English | MEDLINE | ID: mdl-21750714

ABSTRACT

Small interfering RNAs (siRNAs) are routinely used to reduce mRNA levels for a specific gene with the goal of studying its function. Several studies have demonstrated that siRNAs are not always specific and can have many off-target effects. The 3' UTRs of off-target mRNAs are often enriched in sequences that are complementary to the seed-region of the siRNA. We demonstrate that siRNA off-targets can be significantly reduced when cells are treated with a dose of siRNA that is relatively low (e.g. 1 nM), but sufficient to effectively silence the intended target. The reduction in off-targets was demonstrated for both modified and unmodified siRNAs that targeted either STAT3 or hexokinase II. Low concentrations reduced silencing of transcripts with complementarity to the seed region of the siRNA. Similarly, off-targets that were not complementary to the siRNA were reduced at lower doses, including up-regulated genes that are involved in immune response. Importantly, the unintended induction of caspase activity following treatment with a siRNA that targeted hexokinase II was also shown to be a concentration-dependent off-target effect. We conclude that off-targets and their related phenotypic effects can be reduced for certain siRNA that potently silence their intended target at low concentrations.


Subject(s)
Hexokinase/genetics , RNA Interference , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , 3' Untranslated Regions/genetics , Base Sequence , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
J Lipid Res ; 52(6): 1265-1271, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21371998

ABSTRACT

Apolipoprotein CIII (apoCIII), a major constituent of triglyceride-rich lipoprotein, has been proposed as a key contributor to hypertriglyceridemia on the basis of its inhibitory effects on lipoprotein lipase. Many immunochemical methods have been developed for human apoCIII quantification, including ELISA. However, a sensitive and quantitative assay for nonhuman primates is not commercially available. We developed a sensitive, quantitative, and highly specific sandwich ELISA to measure apoCIII in both nonhuman primate and human serum. Our assay generates a linear calibration curve from 0.01 µg/ml to 10 µg/ml using an apoCIII standard that was purified from cynomolgus monkey serum. It is highly reproducible (intra- and interplate CV < 5% and < 8%, respectively), sensitive enough to distinguish 10% difference of apoCIII present in serum, and has no interference from purified human apolipoprotein AI, AII, B, CI, CII, or E. The same assay can also be used to measure human apoCIII with a linear calibration curve from 0.005 µg/ml to 1 µg/ml using purified human apoCIII as the standard. This fast and highly sensitive ELISA could be a useful tool to investigate the role of apoCIII in lipoprotein transport and cardiovascular disease.


Subject(s)
Antibodies/metabolism , Apolipoprotein C-III/blood , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay/methods , Hypertriglyceridemia/blood , Animals , Antibodies/immunology , Blotting, Western , Humans , Hypertriglyceridemia/physiopathology , Limit of Detection , Macaca fascicularis , Male , Reference Standards , Reproducibility of Results , Sensitivity and Specificity
10.
Opt Express ; 18(2): 513-22, 2010 Jan 18.
Article in English | MEDLINE | ID: mdl-20173871

ABSTRACT

Through retrofitting the descriptor of a scale-invariant feature transform (SIFT) and developing a new similarity measure function based on trajectories generated from Lissajous curves, a new remote sensing image registration approach is constructed, which is more robust and accurate than prior approaches. In complex cases where the correct rate of feature matching is below 20%, the retrofitted SIFT descriptor improves the correct rate to nearly 100%. Mostly, the similarity measure function makes it possible to quantitatively analyze the temporary change of the same geographic position.


Subject(s)
Algorithms , Artificial Intelligence , Environmental Monitoring/methods , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Sensitivity and Specificity
11.
Zhong Xi Yi Jie He Xue Bao ; 7(4): 360-5, 2009 Apr.
Article in Chinese | MEDLINE | ID: mdl-19361367

ABSTRACT

OBJECTIVE: To investigate the effects of Caulis Sargentodoxae Granule (CSG), a compound traditional Chinese herbal medicine for treating endometriosis, on expressions of vascular endothelial growth factor (VEGF) and its receptor-2 fetal liver kinase-1 (Flk-1) in rats with endometriosis. METHODS: A rat model of endometriosis was established by autotransplant of endometria. Forty-eight endometriosis rats were randomly divided into 6 groups: untreated group, castrate group, mifepristone group (5 mg/kg) and low- (20 g/kg), medium- (40 g/kg) and high-dose (80 g/kg) CSG group. After 21-day consecutive treatment, the expressions of VEGF and its receptor Flk-1 in ectopic endometria were detected by immunohistochemical method, and the volume of the ectopic endometria was also measured by using electronic digital calipers. RESULTS: Compared with the untreated group, the endometrial volumes in the treated groups were significantly decreased (P<0.05) except for the low-dose CSG group. Histopathologic observation showed that the epithelial layer of ectopic endometrium was less and the thickness of epithelial layer was thinner than those in the untreated group (P<0.01), but the low-dose CSG group had few changes. The VEGF and its receptor Flk-1 expressed abundantly in the cytoplasm of the glandular epithelium and vascular endothelial cell in all the groups. Compared with the untreated group, the expressions of VEGF and Flk-1 of ectopic endometrium in the CSG-treated groups, the mifepristone group and the castrate group were significantly decreased (P<0.05, P<0.01). CONCLUSION: CSG can treat the endometriosis in rats, and its action may be associated with decreasing the expressions of VEGF and its receptor Flk-1 in ectopic endometrium.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Endometriosis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Drugs, Chinese Herbal/therapeutic use , Endometriosis/drug therapy , Endometriosis/pathology , Endometrium/metabolism , Female , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...