Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell ; 171(2): 385-397.e11, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28919076

ABSTRACT

T cell receptor (TCR) signaling without CD28 can elicit primary effector T cells, but memory T cells generated during this process are anergic, failing to respond to secondary antigen exposure. We show that, upon T cell activation, CD28 transiently promotes expression of carnitine palmitoyltransferase 1a (Cpt1a), an enzyme that facilitates mitochondrial fatty acid oxidation (FAO), before the first cell division, coinciding with mitochondrial elongation and enhanced spare respiratory capacity (SRC). microRNA-33 (miR33), a target of thioredoxin-interacting protein (TXNIP), attenuates Cpt1a expression in the absence of CD28, resulting in cells that thereafter are metabolically compromised during reactivation or periods of increased bioenergetic demand. Early CD28-dependent mitochondrial engagement is needed for T cells to remodel cristae, develop SRC, and rapidly produce cytokines upon restimulation-cardinal features of protective memory T cells. Our data show that initial CD28 signals during T cell activation prime mitochondria with latent metabolic capacity that is essential for future T cell responses.


Subject(s)
CD28 Antigens/metabolism , Lymphocyte Activation , Mitochondria/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Carnitine O-Palmitoyltransferase , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Humans , Interleukin-15/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Stress, Physiological , T-Lymphocytes/metabolism
2.
Acta Biochim Biophys Sin (Shanghai) ; 53(10): 1367-1376, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34357372

ABSTRACT

Many patients with cancers have low levels of CD4+ in their peripheral blood. However, the molecular mechanism is still unclear. Here, we found that the blood levels of miR-221 and miR-222 were dramatically increased in patients with colorectal cancer (CRC), and both circulating miR-211 and miR-222 served as sensitive diagnostic markers with an area under the curve of 0.8790 and 0.9148, respectively. Transfection of either miR-221 or miR-222 resulted in the reduction of the surface CD4 antigen level but not the surface CD8 antigen level. The luciferase reporter assay showed that miR-221/222 directly regulated CD4 expression in human primary T cells. These data showed that miR-221/222 levels were upregulated in the blood of patients with CRC and that the expression of CD4 in human primary T cells was inhibited by miR-221/222. These findings provide a novel strategy for modulating the number of CD4+ T cells in the blood and further adjusting the microenvironment suitable for immunotherapy.


Subject(s)
CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/genetics , MicroRNAs/blood , MicroRNAs/immunology , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/blood , Colorectal Neoplasms/immunology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kaplan-Meier Estimate , Male , MicroRNAs/metabolism , Middle Aged , ROC Curve , Up-Regulation
3.
Proc Natl Acad Sci U S A ; 111(6): E692-701, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469819

ABSTRACT

A technology that visualizes tumor stem cells with clinically relevant tracers could have a broad impact on cancer diagnosis and treatment. The AC133 epitope of CD133 currently is one of the best-characterized tumor stem cell markers for many intra- and extracranial tumor entities. Here we demonstrate the successful noninvasive detection of AC133(+) tumor stem cells by PET and near-infrared fluorescence molecular tomography in subcutaneous and orthotopic glioma xenografts using antibody-based tracers. Particularly, microPET with (64)Cu-NOTA-AC133 mAb yielded high-quality images with outstanding tumor-to-background contrast, clearly delineating subcutaneous tumor stem cell-derived xenografts from surrounding tissues. Intracerebral tumors as small as 2-3 mm also were clearly discernible, and the microPET images reflected the invasive growth pattern of orthotopic cancer stem cell-derived tumors with low density of AC133(+) cells. These data provide a basis for further preclinical and clinical use of the developed tracers for high-sensitivity and high-resolution monitoring of AC133(+) tumor stem cells.


Subject(s)
Antigens, CD/immunology , Glycoproteins/immunology , Neoplastic Stem Cells/immunology , Peptides/immunology , Positron-Emission Tomography/methods , AC133 Antigen , Animals , Brain Neoplasms/diagnostic imaging , Fluorescence , Glioblastoma/diagnostic imaging , Heterografts , Mice , Multimodal Imaging , Tomography, X-Ray Computed
4.
Comput Biol Chem ; 110: 108058, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593480

ABSTRACT

Exploring the relationship between proteins and drugs plays a significant role in discovering new synthetic drugs. The Drug-Target Interaction (DTI) prediction is a fundamental task in the relationship between proteins and drugs. Unlike encoding proteins by amino acids, we use amino acid subsequence to encode proteins, which simulates the biological process of DTI better. For this research purpose, we proposed a novel deep learning framework based on Bidirectional Encoder Representation from Transformers (BERT), which integrates high-frequency subsequence embedding and transfer learning methods to complete the DTI prediction task. As the first key module, subsequence embedding allows to explore the functional interaction units from drug and protein sequences and then contribute to finding DTI modules. As the second key module, transfer learning promotes the model learn the common DTI features from protein and drug sequences in a large dataset. Overall, the BERT-based model can learn two kinds features through the multi-head self-attention mechanism: internal features of sequence and interaction features of both proteins and drugs, respectively. Compared with other methods, BERT-based methods enable more DTI-related features to be discovered by means of attention scores which associated with tokenized protein/drug subsequences. We conducted extensive experiments for the DTI prediction task on three different benchmark datasets. The experimental results show that the model achieves an average prediction metrics higher than most baseline methods. In order to verify the importance of transfer learning, we conducted an ablation study on datasets, and the results show the superiority of transfer learning. In addition, we test the scalability of the model on the dataset in unseen drugs and proteins, and the results of the experiments show that it is acceptable in scalability.


Subject(s)
Deep Learning , Proteins , Proteins/chemistry , Proteins/metabolism , Pharmaceutical Preparations/chemistry , Computational Biology
5.
Immunol Cell Biol ; 91(10): 615-24, 2013.
Article in English | MEDLINE | ID: mdl-24100387

ABSTRACT

The efficacy of immunotherapy based on natural killer (NK) cells is hampered by intrinsic non-specific cytotoxicity and insufficient activation of NK cells. Here, we confer the T-cell receptor-like (TCR-like) specificity on NK cells, taking advantage of both the innate and adaptive immune arms of the immune response to generate enhanced anti-melanoma activity. The TCR-like antibody (Ab) GPA7 was selected against melanoma-associated gp100/human leukocyte antigen (HLA)-A2 complex and then fused to intracellular domain of CD3-ζ chain. This fusion construct was incorporated into NK-92MI cell line and expressed as a chimeric antigen receptor on the surface of the cell. The anti-tumour activity of the transgenic NK-92MI-GPA7-ζ cell line was assessed against melanoma in vitro and in vivo. The engineered NK-92MI-GPA7-ζ cells could recognize melanoma cells in the context of HLA-A2 and showed enhanced killing of both melanoma cell lines and primary melanoma. Furthermore, adoptively transferred NK-92MI-GPA7-ζ cells significantly suppressed the growth of human melanoma in a xenograft model in mice. Collectively, these results demonstrate that the TCR-like Ab, GPA7, could redirect NK cells to target the intracellular antigen gp100 and enhance anti-melanoma activity, providing a promising immunotherapeutic strategy to prevent and treat melanoma.


Subject(s)
Melanoma/immunology , Receptors, Antigen, T-Cell/immunology , Single-Domain Antibodies/immunology , Animals , Antibody Specificity/immunology , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic , Humans , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Protein Multimerization , Xenograft Model Antitumor Assays
6.
Front Comput Sci ; 17(5): 175903, 2023.
Article in English | MEDLINE | ID: mdl-36532946

ABSTRACT

Prediction of drug-protein binding is critical for virtual drug screening. Many deep learning methods have been proposed to predict the drug-protein binding based on protein sequences and drug representation sequences. However, most existing methods extract features from protein and drug sequences separately. As a result, they can not learn the features characterizing the drug-protein interactions. In addition, the existing methods encode the protein (drug) sequence usually based on the assumption that each amino acid (atom) has the same contribution to the binding, ignoring different impacts of different amino acids (atoms) on the binding. However, the event of drug-protein binding usually occurs between conserved residue fragments in the protein sequence and atom fragments of the drug molecule. Therefore, a more comprehensive encoding strategy is required to extract information from the conserved fragments. In this paper, we propose a novel model, named FragDPI, to predict the drug-protein binding affinity. Unlike other methods, we encode the sequences based on the conserved fragments and encode the protein and drug into a unified vector. Moreover, we adopt a novel two-step training strategy to train FragDPI. The pre-training step is to learn the interactions between different fragments using unsupervised learning. The fine-tuning step is for predicting the binding affinities using supervised learning. The experiment results have illustrated the superiority of FragDPI. Electronic Supplementary Material: Supplementary material is available for this article at 10.1007/s11704-022-2163-9 and is accessible for authorized users.

7.
J Immunol ; 184(12): 6910-9, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483727

ABSTRACT

Bone remodeling involves bone resorption by osteoclasts and synthesis by osteoblasts and is tightly regulated by the receptor activator of the NF-kappaB ligand (RANKL)/receptor activator of the NF-kappaB (RANK)/osteoprotegerin molecular triad. RANKL, a member of the TNF superfamily, induces osteoclast differentiation, activation and survival upon interaction with its receptor RANK. The decoy receptor osteoprotegerin inhibits osteoclast formation by binding to RANKL. Imbalance in this molecular triad can result in diseases, including osteoporosis and rheumatoid arthritis. In this study, we report the crystal structures of unliganded RANK and its complex with RANKL and elucidation of critical residues for the function of the receptor pair. RANK represents the longest TNFR with four full cysteine-rich domains (CRDs) in which the CRD4 is stabilized by a sodium ion and a rigid linkage with CRD3. On association, RANK moves via a hinge region between the CRD2 and CRD3 to make close contact with RANKL; a significant structural change previously unseen in the engagement of TNFR superfamily 1A with its ligand. The high-affinity interaction between RANK and RANKL, maintained by continuous contact between the pair rather than the patched interaction commonly observed, is necessary for the function because a slightly reduced affinity induced by mutation produces significant disruption of osteoclast formation. The structures of RANK and RANKL-RANK complex and the biological data presented in the paper are essential for not only our understanding of the specific nature of the signaling mechanism and of disease-related mutations found in patients but also structure based drug design.


Subject(s)
RANK Ligand/chemistry , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/chemistry , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction/immunology , Amino Acid Sequence , Animals , Crystallization , Humans , Mice , Molecular Sequence Data , Protein Structure, Quaternary , Sequence Homology, Amino Acid , X-Ray Diffraction
8.
Cancer Lett ; 524: 121-130, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34687790

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has been successful in treating hematological malignancy, but solid tumors remain refractory. Here, we demonstrated that knocking out transcription factor IKZF3 in HER2-specific CAR T cells targeting breast cancer cells did not affect CAR expression or CAR T cell differentiation, but markedly enhanced killing of the cancer cells in vitro and in a xenograft model, which was associated with increased T cell activation and proliferation. Furthermore, IKZF3 KO had similar effects on the CD133-specific CAR T cells targeting glioblastoma cells. AlphaLISA and RNA-seq analyses indicate that IKZF3 KO increased the expression of genes involved in cytokine signaling, chemotaxis and cytotoxicity. Our results suggest a general strategy for enhancing CAR T efficacy on solid tumors.


Subject(s)
Breast Neoplasms/therapy , Ikaros Transcription Factor/genetics , Receptor, ErbB-2/genetics , Receptors, Chimeric Antigen/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , Heterografts , Humans , Ikaros Transcription Factor/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Mice , RNA-Seq , Receptors, Chimeric Antigen/antagonists & inhibitors , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology
9.
Cancer Lett ; 537: 215680, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35461758

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. Following the publication of the above article, the Editor was notified that an error occurred in which all images were published with incorrect versions. The Editor has taken the decision that the manuscript is no longer acceptable in its current form, nor with a corrigendum, as the extensive changes to the figures and publication would lead to ambiguity for our readers. We have therefore made the decision to retract this manuscript from Cancer Letters with the possibility of resubmission and republication of the manuscript in its corrected form after peer review.

10.
Cancer Lett ; 538: 215697, 2022 07 10.
Article in English | MEDLINE | ID: mdl-35487310

ABSTRACT

Metastatic small cell lung cancer (SCLC) is not curable. While SCLC is initially sensitive to chemotherapy, remissions are short-lived. The relapse is induced by chemotherapy-selected tumor stem cells, which express the AC133 epitope of the CD133 stem cell marker. We studied the effectiveness of AC133-specific CAR T cells post-chemotherapy using human primary SCLC and an orthotopic xenograft mouse model. AC133-specific CAR T cells migrated to SCLC tumor lesions, reduced the tumor burden, and prolonged survival in a humanized orthotopic SCLC model, but were not able to entirely eliminate tumors. We identified CD73 and PD-L1 as immune-escape mechanisms and combined PD-1-inhibition and CD73-inhibition with CAR T cell treatment. This triple-immunotherapy induced cures in 25% of the mice, without signs of graft-versus-host disease or bone marrow failure. AC133+ cancer stem cells and PD-L1+CD73+ myeloid cells were detectable in primary human SCLC tissues, suggesting that patients may benefit from the triple-immunotherapy. We conclude that the combination of AC133-specific CAR T cells, anti-PD-1-antibody and CD73-inhibitor specifically eliminates chemo-resistant tumor stem cells, overcomes SCLC-mediated T cell inhibition, and might induce long-term complete remission in an otherwise incurable disease.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , B7-H1 Antigen , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Lung Neoplasms/pathology , Mice , Neoplasm Recurrence, Local , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/therapy
11.
Front Pharmacol ; 12: 619800, 2021.
Article in English | MEDLINE | ID: mdl-33776765

ABSTRACT

Chimeric antigen receptor (CAR) T cells are powerful in eradicating hematological malignancies, but their efficacy is limited in treating solid tumors. One of the barriers is the immunosuppressive response induced by immunomodulatory signaling pathways. Pharmacological targeting of these immunosuppressive pathways may be a simple way to improve the efficacy of CAR T cells. In this study, anti-CD133 and anti-HER2 CAR T cells were generated from healthy donors, and combination therapy using CAR T cells and small molecules targeting adenosine receptors was performed in vitro and in vivo with the goal of probing for potential synergistic antitumor activities. The adenosine A2b receptor agonist, BAY 60-6583, was found to significantly increase cytokine secretion of CD133-or HER2-specific CAR T cells when co-cultured with the respective target tumor cells. The in vitro cytotoxicity and proliferation of CAR T cells were also enhanced when supplied with BAY 60-6583. Furthermore, the combination with this small molecule facilitated the anti-HER2 CAR T cell-mediated elimination of tumor cells in a xenograft mouse model. However, the enhanced antitumor activities could not be suppressed by knockout of the adenosine A2b receptor in CAR T cells. Furthermore, mass spectrometry and computational methods were used to predict several potential alternative targets. Four potential targets (pyruvate kinase M (PKM), Talin-1, Plastin-2, and lamina-associated polypeptide 2) were captured by a photo-affinity probe, of which PKM and Talin-1 were predicted to interact with BAY 60-6583. Overall, our data suggest that BAY 60-6583 upregulates T cell functions through a mechanism independent of the adenosine A2b receptor.

12.
Adv Sci (Weinh) ; 8(6): 2003091, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33747727

ABSTRACT

Using T-cell chimeric antigen receptors (CAR-T) to activate and redirect T cells to tumors expressing the cognate antigen represents a powerful approach in cancer therapy. However, normal tissues with low expression of tumor-associated antigens (TAAs) can be mistargeted, resulting in severe side effects. An approach using a collection of T cells expressing a diverse, 106-member combinatorial cellular library of CARs, in which members can be specifically enriched based on avidity for cell membrane antigens, is reported. Using CD38 as the target antigen, an efficient and effective selection of CARs specifically recognizing CD38+ tumor cells is demonstrated. These selected CAR-T's produce cytokines known to be associated with T cell activation in a CD38 expression-dependent manner. This avidity-based selection endows the engineered T cells with minimal off-tumor effects, while retaining robust antitumor efficacy both in vitro and in vivo. The described method may facilitate the application of CAR-T therapy to TAAs previously considered undruggable.

13.
Cancer Lett ; 520: 385-399, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34407431

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. Following the publication of the above article, the Editor was notified that an error occurred in which all images were published with incorrect versions. The Editor has taken the decision that the manuscript is no longer acceptable in its current form, nor with a corrigendum, as the extensive changes to the figures and publication would lead to ambiguity for our readers. We have therefore made the decision to retract this manuscript from Cancer Letters with the possibility of resubmission and republication of the manuscript in its corrected form after peer review.


Subject(s)
5'-Nucleotidase/genetics , AC133 Antigen/genetics , B7-H1 Antigen/genetics , Small Cell Lung Carcinoma/therapy , 5'-Nucleotidase/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Antibodies, Anti-Idiotypic/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Female , Heterografts , Humans , Immunotherapy, Adoptive/trends , Male , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/pathology , T-Lymphocytes/immunology , Tumor Burden
14.
Immunol Cell Biol ; 88(6): 667-75, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20212511

ABSTRACT

Antibodies (Abs) have been engineered into small antigen-binding fragments and rebuilt into multivalent high-avidity molecules for improving in vivo pharmacokinetics and efficacy in clinical use. To increase the avidity of a T-cell receptor-like single-domain Ab (sdAb) specific for HLA-A2 complex, we fused the sdAb to a coiled-coil peptide derived from human cartilage oligomeric matrix protein (COMP48) to make an sdAb multimer, termed combody. The combody improved the binding avidity of sdAb significantly, whereas the specificity for the targeted cells was retained. The strategy was also expanded to create a bispecific combody by fusing an sdAb to the N-terminal and an anti-CD3 single-chain variable fragment to the C-terminal of COMP48. The dual-specific combody was able to efficiently mediate cytotoxicity against the target cells in vitro. Taken together, the strategy to make combody could be widely adopted to increase the avidity of Ab fragment for further application.


Subject(s)
Antibodies, Bispecific/metabolism , Antibody Affinity/genetics , Carcinoembryonic Antigen/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , MART-1 Antigen/metabolism , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibody Affinity/immunology , Binding Sites, Antibody/genetics , CD3 Complex/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Cartilage Oligomeric Matrix Protein , Extracellular Matrix Proteins/genetics , Feasibility Studies , Glycoproteins/genetics , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , MART-1 Antigen/genetics , MART-1 Antigen/immunology , Matrilin Proteins , Protein Engineering , Protein Multimerization , Protein Structure, Tertiary/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics
15.
Cell Transplant ; 29: 963689720920825, 2020.
Article in English | MEDLINE | ID: mdl-32967454

ABSTRACT

Chimeric antigen receptor (CAR) T-cell immunotherapy still faces many challenges in the treatment of solid tumors, one of which is T-cell dysfunction or exhaustion. Immunomodulator lenalidomide may improve CAR T-cell function. In this study, the effects of lenalidomide on CAR T-cell functions (cytotoxicity, cytokine secretion, and cell proliferation) were investigated. Two different CAR T cells (CD133-specific CAR and HER2-specific CAR) were prepared, and the corresponding target cells including human glioma cell line U251 CD133-OE that overexpress CD133 and human breast cancer cell line MDA-MB-453 were used for functional assay. We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide also enhanced the cytotoxicity against MDA-MB-453 and the cytokine secretion of HER2-CAR T cells but did not affect their proliferation significantly. Furthermore, lenalidomide may regulate the function of CAR T cells by inducing the degradation of transcription factors Ikaros and Aiolos.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Immunotherapy/methods , Lenalidomide/therapeutic use , Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Female , Humans , Lenalidomide/pharmacology , Male , Receptors, Chimeric Antigen/metabolism
16.
Eur J Histochem ; 64(s2)2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32705856

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy still faces the challenge of immunosuppression when treating solid tumors. TGF-ß is one of the critical factors in the tumor microenvironment to help tumors escape surveillance by the immune system. Here we tried using the combination of a small molecule inhibitor of TGF-ß receptor I, Galunisertib, and CAR T cells to explore whether Galunisertib could enhance CAR T cell function against solid tumor cells. In vitro experiments showed Galunisertib could significantly enhance the specific cytotoxicity of both CD133- and HER2-specific CAR T cells. However, Galunisertib had no direct killing effect on target cells. Galunisertib significantly increased the cytokine secretion of CAR T cells and T cells that do not express CAR (Nontransfected T cells). Galunisertib did not affect the proliferation of T cells, the antigen expression on target cells and CD69 on CAR T cells. We found that TGF-ß was secreted by T cells themselves upon activation, and Galunisertib could reduce TGF-ß signaling in CAR T cells. Our findings can provide the basis for further preclinical and clinical studies of the combination of Galunisertib and CAR T cells in the treatment of solid tumors.


Subject(s)
Neoplasms/therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/drug effects , Cell Line, Tumor , Cytokines/metabolism , Humans , Immunotherapy, Adoptive , Neoplasms/drug therapy , T-Lymphocytes/metabolism
17.
Brief Funct Genomics ; 18(2): 119-128, 2019 03 22.
Article in English | MEDLINE | ID: mdl-29982317

ABSTRACT

Next-generation sequencing has produced a large quantity of DNA or RNA sequences related to the processes occurring within tumors and their microenvironment in a reasonable time and cost. These data have been used to guide the identification of neoantigens and to determine their specific T-cell receptors. Furthermore, adoptive T-cell therapy targeting neoantigens is under development for cancer treatment. In this review, we first provide an overview of sequencing technologies and the updated findings concerning neoantigens related to adoptive T-cell therapy and then summarize the methods and principles underlying the development of next-generation sequencing-based neoantigen-reactive T-cell therapy for cancer.


Subject(s)
Antigens, Neoplasm/immunology , Cell- and Tissue-Based Therapy/methods , High-Throughput Nucleotide Sequencing/methods , Immunotherapy , Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/genetics , Humans , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes/immunology
18.
Hum Gene Ther ; 30(4): 446-458, 2019 04.
Article in English | MEDLINE | ID: mdl-29706119

ABSTRACT

CRISPR/Cas9-mediated programmed cell death protein 1 (PD-1) disruption in chimeric antigen receptor (CAR) T cells could be an appealing choice to improve the therapeutic efficacy of CAR T cells in an immunosuppressive tumor microenvironment. In most of the reported cases, Cas9 was delivered into T cells by way of electroporation with RNA or protein. However, transient expression of Cas9 by transfection with a plasmid encoding its gene is apparently simpler, as it avoids the steps of in vitro transcription of DNA or protein production. This study tried nucleofection into human primary T cells of plasmids encoding both CRISPR/Cas9 for disrupting the PD-1 gene and the piggyBac transposon system for expressing CD133-specific CAR in one reaction. Based on drug selection, CD133-specific CAR T cells were obtained in which, on average, 91.5% of the PD-1 gene sites were disrupted, but almost no Cas9 gene expression was found in the final engineered CAR T cells. The PD-1-deficient CD133-specific CAR T cells showed similar levels of cytokine secretion and improved proliferation and cytotoxicity in vitro, and enhanced inhibition of tumor growth in an orthotopic mouse model of glioma, compared to conventional CD133-CAR T cells. The described method could be useful for the production of PD-1-deficient CAR T cells for cancer immunotherapy.


Subject(s)
AC133 Antigen/immunology , CRISPR-Cas Systems , Gene Silencing , Plasmids/genetics , Programmed Cell Death 1 Receptor/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , AC133 Antigen/antagonists & inhibitors , Animals , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Gene Editing , Gene Knockdown Techniques , Genetic Engineering , Humans , Immunophenotyping , Immunotherapy, Adoptive , Lymphocyte Activation , Mice , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/metabolism , Receptors, Chimeric Antigen/genetics , T-Cell Antigen Receptor Specificity , Transfection/methods , Xenograft Model Antitumor Assays
19.
Theranostics ; 6(6): 862-74, 2016.
Article in English | MEDLINE | ID: mdl-27162556

ABSTRACT

Near-infrared photoimmunotherapy (NIR-PIT), which employs monoclonal antibody (mAb)-phototoxic phthalocyanine dye IR700 conjugates, permits the specific, image-guided and spatiotemporally controlled elimination of tumor cells. Here, we report the highly efficient NIR-PIT of human tumor xenografts initiated from patient-derived cancer stem cells (CSCs). Using glioblastoma stem cells (GBM-SCs) expressing the prototypic CSC marker AC133/CD133, we also demonstrate here for the first time that NIR-PIT is highly effective against brain tumors. The intravenously injected theranostic AC133 mAb conjugate enabled the non-invasive detection of orthotopic gliomas by NIR fluorescence imaging, and reached AC133+ GBM-SCs at the invasive tumor front. AC133-targeted NIR-PIT induced the rapid cell death of AC133+ GBM-SCs and thereby strong shrinkage of both subcutaneous and invasively growing brain tumors. A single round of NIR-PIT extended the overall survival of mice with established orthotopic gliomas by more than a factor of two, even though the harmless NIR light was applied through the intact skull. Humanised versions of this theranostic agent may facilitate intraoperative imaging and histopathological evaluation of tumor borders and enable the highly specific and efficient eradication of CSCs.


Subject(s)
AC133 Antigen/immunology , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Immunotherapy/methods , Phototherapy/methods , Theranostic Nanomedicine/methods , Animals , Antibodies/administration & dosage , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Disease Models, Animal , Heterografts , Humans , Indoles/administration & dosage , Isoindoles , Mice , Stem Cells/immunology , Survival Analysis , Treatment Outcome
20.
Oncoscience ; 2(5): 476-82, 2015.
Article in English | MEDLINE | ID: mdl-26097880

ABSTRACT

Adoptive transfer of T cells expressing chimeric antigen receptors (CARs) holds great promise for cancer treatment. We recently developed CAR T cells targeting the prototypic cancer stem cell marker AC133 and showed that these CAR T cells killed AC133+ glioblastoma stem cells (GBM-SCs) in vitro and inhibited the growth of brain tumors initiated from GBM-SCs in xenograft mouse models in vivo . Upon coincubation with GBM-SCs, we observed strong upregulation of the T cell aging marker CD57, but other phenotypical or functional changes usually associated with terminal T cell differentiation could not immediately be detected. Here, we provide evidence suggesting that CD57 is rapidly and efficiently transferred from CD57+ GBM-SCs to preactivated T cells and that the transfer is greatly enhanced by specific CAR/ligand interaction. After separation from CD57+ tumor cells, CD57 epitope expression on T cells decreased only slowly over several days. We conclude that CD57 transfer from tumor cells to T cells may occur in patients with CD57+ tumors and that it may have to be considered in the interpretation of phenotyping results for tumor-infiltrating lymphocytes and perhaps also in the characterization of tumor-specific T cells from tumor or lymph node homogenates or peripheral blood mononuclear cells.

SELECTION OF CITATIONS
SEARCH DETAIL