Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Sci Rep ; 13(1): 12365, 2023 07 31.
Article in English | MEDLINE | ID: mdl-37524755

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has emerged as a promising modality for anti-cancer treatment. Its efficacy is quite remarkable in hematological tumors. Owing to their excellent clinical results, gene- modified cell therapies, including T cells, natural killer (NK) cells, and macrophages, are being actively studied in both academia and industry. However, the protocol to make CAR immune cells is too complicated, so it is still unclear how to efficiently produce the potent CAR immune cells. To manufacture effective CAR immune cells, we need to be aware of not only how to obtain highly infective viral particles, but also how to transduce CAR genes into immune cells. In this paper, we provide detailed information on spinoculation, which is one of the best known protocols to transduce genes into immune cells, in a methodological view. Our data indicate that gene transduction is significantly dependent on speed and duration of centrifugation, concentration and number of viral particles, the concentration of polybrene, and number of infected immune cells. In addition, we investigated on the optimal polyethylene glycol (PEG) solution to concentrate the viral supernatant and the optimized DNA ratios transfected into 293T cells to produce high titer of viral particles. This study provides useful information for practical production of the gene-modified immune cells using viral vectors.


Subject(s)
Genetic Vectors , Neoplasms , Humans , Transduction, Genetic , Genetic Vectors/genetics , Killer Cells, Natural , T-Lymphocytes , Immunotherapy, Adoptive/methods
2.
Cancers (Basel) ; 13(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830894

ABSTRACT

Chimeric antigen receptor (CAR) technology has been highlighted in recent years as a new therapeutic approach for cancer treatment. Although the impressive efficacy of CAR-based T cell adoptive immunotherapy has been observed in hematologic cancers, limited effect has been reported on solid tumors. Approximately 20% of gastric cancer (GC) patients exhibit a high expression of c-Met. We have generated an anti c-Met CAR construct that is composed of a single-chain variable fragment (scFv) of c-Met antibody and signaling domains consisting of CD28 and CD3ζ. To test the CAR construct, we used two cell lines: the Jurkat and KHYG-1 cell lines. These are convenient cell lines, compared to primary T cells, to culture and to test CAR constructs. We transduced CAR constructs into Jurkat cells by electroporation. c-Met CAR Jurkat cells secreted interleukin-2 (IL-2) only when incubated with c-Met positive GC cells. To confirm the lytic function of CAR, the CAR construct was transduced into KHYG-1, a NK/T cell line, using lentiviral particles. c-Met CAR KHYG-1 showed cytotoxic effect on c-Met positive GC cells, while c-Met negative GC cell lines were not eradicated by c-Met CAR KHYG-1. Based on these data, we created c-Met CAR T cells from primary T cells, which showed high IL-2 and IFN-γ secretion when incubated with the c-Met positive cancer cell line. In an in vivo xenograft assay with NSG bearing MKN-45, a c-Met positive GC cell line, c-Met CAR T cells effectively inhibited the tumor growth of MKN-45. Our results show that the c-Met CAR T cell therapy can be effective on GC.

3.
Anticancer Res ; 41(4): 1811-1819, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33813386

ABSTRACT

BACKGROUND/AIM: Glioblastoma is the most common cancer among primary brain tumors, however, its prognosis and treatment advances are very poor. Here, we investigated whether c-Met, FOLR1, and AXL proteins are promising targets for chimeric antigen receptor (CAR) T-cell therapy, for they are known to be over-expressed in a variety of solid tumors. MATERIALS AND METHODS: CAR constructs were prepared and CAR KHYG-1 cells targeting c-Met, FOLR1, or AXL were made by lentiviral transduction. The activity of CAR KHYG-1 cells against cancer cells was measured by cytokine secretion and cell lysis assays. RESULTS: c-Met and AXL were over-expressed in most glioblastoma cell lines (11/13), but not in neuroblastoma cell lines (0/8). FOLR1 was over-expressed only in one among 16 glioblastoma cell lines. Our antigen-specific CAR KHYG-1 cells eradicated target positive glioblastoma cells selectively. CONCLUSION: Anti-c-Met and anti-AXL CAR NK or T cells could be effective in glioblastoma cells.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Coculture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic , Folate Receptor 1/immunology , Folate Receptor 1/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Jurkat Cells , Killer Cells, Natural/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Axl Receptor Tyrosine Kinase
4.
Int J Mol Sci ; 21(23)2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33271901

ABSTRACT

CD19 is the most promising target for developing chimeric-antigen receptor (CAR) T cells against B-cell leukemic cancer. Currently, two CAR-T-cell products, Kymriah and Yescarta, are approved for leukemia patients, and various anti-CD19 CAR T cells are undergoing clinical trial. Most of these anti-CD19 CAR T cells use FMC63 single-chain variable fragments (scFvs) for binding CD19 expressed on the cancer cell surface. In this study, we screened several known CD19 scFvs for developing anti-CD19 CAR T cells. We used the KHYG-1 NK/T-cell line for screening of CD19 scFvs because it has advantages in terms of cell culture and gene transduction compared to primary T cells. Using our CAR construct backbone, we made anti-CD19 CAR constructs which each had CD19 scFvs including FMC63, B43, 25C1, BLY3, 4G7, HD37, HB12a, and HB12b, then made each anti-CD19 CAR KHYG-1 cells. Interestingly, only FMC63 CAR KHYG-1 and 4G7 CAR KHYG-1 efficiently lysed CD19-positive cell lines. In addition, in Jurkat cell line, only these two CAR Jurkat cell lines secreted IL-2 when co-cultured with CD19-positive cell line, NALM-6. Based on these results, we made FMC63 CAR T cells and 4G7 CAR T cells from PBMC. In in vitro lysis assay, 4G7 CAR T cells lysed CD19-positive cell line as well as FMC63 CAR T cells. In in vivo assay with NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice, 4G7 CAR T cells eradicated NALM-6 as potently as FMC63 CAR T cells. Therefore, we anticipate that 4G7 CAR T cells will show as good a result as FMC63 CAR T cells for B-cell leukemia patients.


Subject(s)
Antigens, CD19/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Gene Order , Humans , Immunotherapy, Adoptive , Leukemia/immunology , Leukemia/pathology , Leukemia/therapy , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Xenograft Model Antitumor Assays
5.
Anticancer Res ; 40(9): 4929-4935, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32878781

ABSTRACT

BACKGROUND: Mutations in the isocitrate dehydrogenase 1 (IDH1) gene are frequently found in various cancer types. IDH1 mutants produce 2-hydroxyglutarate (2-HG), an oncometabolite, from alpha-ketoglutarate (α-KG). This 2-HG plays a key role in tumorigenesis via inhibition of α-KG dependent enzymes. For this reason, IDH1 mutant could be an ideal target for the treatment of cancer. MATERIALS AND METHODS: To find a new IDH1 inhibitor, 8,364 compounds were obtained from Korea Chemical Bank. Using high-throughput screening (HTS) of a chemical library, we unveiled a compound that could inhibit the IDH1 mutant. RESULTS: According to the enzyme assay, our compound (KRC-09) effectively inhibited the activity of IDH1 R132H mutant. In addition, KRC-09 decreased the concentration of intracellular 2-HG in the U-87 MG cell line harboring IDH1 R132H. CONCLUSION: In this article, we present a novel chemical scaffold that suppresses the activity of an IDH1 mutant.


Subject(s)
Antineoplastic Agents/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Discovery , Glutarates/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Molecular Structure , Mutation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
6.
ACS Synth Biol ; 9(5): 987-992, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32352759

ABSTRACT

Despite the excellent efficacy of chimeric antigen receptor (CAR T) cell therapy, concerns about its safety have been constantly raised. The side effects of CAR T cells result from an aberrantly upregulation of CAR T cell activity. Therefore, it is crucial to control the CAR T cell activity whenever the patient is at risk. For this purpose, the iCas9 system, which induces apoptosis in CAR T cell through caspase-9 dimerization by compound, has been invented and is currently going under clinical trial. However, the iCas9 system is irreversible, as the entire CAR T cell population is removed from the patient. Thus, CAR T cells, which are very expensive, should be reinfused to the patients after they recovered from the side-effect. Here, we propose a new CAR T cell safety strategy, which targets CAR "protein", not CAR "T cell". In this system, the CAR construct is modified to bear a bromodomain (BD). The addition of a BD in the CAR protein did not interfere with the original CAR functions, such as cytokine secretion and target cell lysis. Our data showed that the use of a proteolysis-targeting chimaera (PROTAC) compound against BD successfully degraded the BD-containing CAR protein. Moreover, the CAR expression is recovered when the PROTAC compound is removed from the cell, demonstrating that our system is reversible. In a target cell lysis assay, the PROTAC compound successfully suppressed the lytic activity of CAR T cells by degrading the CAR protein. In conclusion, we developed a new safety system in which CAR T cells can be "reversibly" controlled by a compound.


Subject(s)
Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , Azepines/chemistry , Azepines/pharmacology , Cell Line , Histone Acetyltransferases/chemistry , Humans , Interleukin-2/metabolism , Protein Domains , Proteolysis/drug effects , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thalidomide/analogs & derivatives , Thalidomide/chemistry , Thalidomide/pharmacology , Transcription Factors/chemistry , Ubiquitin-Protein Ligases/metabolism
7.
Sci Rep ; 9(1): 19654, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31873151

ABSTRACT

Proteolysis targeting chimeras (PROTACs) are an emerging strategy for promoting targeted protein degradation by inducing the proximity between targeted proteins and E3 ubiquitin ligases. Although successful degradation of numerous proteins by PROTACs has been demonstrated, the elements that determine the degradability of PROTAC-targeted proteins have not yet been explored. In this study, we developed von Hippel-Lindau-Cereblon (VHL-CRBN) heterodimerizing PROTACs that induce the degradation of CRBN, but not VHL. A quantitative proteomic analysis further revealed that VHL-CRBN heterodimerizing PROTACs induced the degradation of CRBN, but not the well-known immunomodulatory drug (IMiD) neo-substrates, IKAROS family zinc finger 1 (IKZF1) and -3 (IZKF3). Moreover, truncation of disordered regions of CRBN and the androgen receptor (AR) attenuated their PROTAC-induced degradation, and attachment of the disordered region to stable CRBN or AR facilitated PROTAC-induced degradation. Thus, these results suggest that the intrinsically disordered region of targeted proteins is essential for efficient proteolysis, providing a novel criterion for choosing degradable protein targets.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Proteolysis , Recombinant Fusion Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/genetics , HEK293 Cells , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Jurkat Cells , Protein Domains , Recombinant Fusion Proteins/genetics , Ubiquitin-Protein Ligases/genetics
8.
Biochem Biophys Res Commun ; 505(2): 542-547, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30274779

ABSTRACT

Recently, proteolysis targeting chimera (PROTAC) technology is highlighted in drug discovery area as a new therapeutic approach. PROTAC as a heterobifunctional molecule is comprised of two ligands, which recruit target protein and E3 ligase, respectively. To degrade the anaplastic lymphoma kinase (ALK) fusion protein, such as NPM-ALK or EML4-ALK, we generated several ALK-PROTAC molecules consisted of ceritinib, one of the ALK inhibitors, and ligand of von Hippel-Lindau (VHL) E3 ligase. Among these molecules, TD-004 effectively induced ALK degradation and inhibited the growth of ALK fusion positive cell lines, SU-DHL-1 and H3122. We also confirmed that TD-004 significantly reduced the tumor growth in H3122 xenograft model.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Ligands , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Oncogene Proteins, Fusion/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/therapeutic use , Proteolysis , Pyrimidines/chemistry , Sulfones/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
9.
PLoS One ; 13(6): e0198347, 2018.
Article in English | MEDLINE | ID: mdl-29874279

ABSTRACT

Gastric cancer is a malignancy that has a high mortality rate. Although progress has been made in the treatment of gastric cancer, many patients experience cancer recurrence and metastasis. Folate receptor 1 (FOLR1) is overexpressed on the cell surface in over one-third of gastric cancer patients, but rarely is expressed in normal tissue. This makes FOLR1 a potential target for chimeric antigen receptor (CAR) T cell immunotherapy, although the function of FOLR1 has not been elucidated. CAR are engineered fusion receptor composed of an antigen recognition region and signaling domains. T cells expressing CAR have specific activation and cytotoxic effects against cancer cells containing the target antigen. In this study, we generated a CAR that targets FOLR1 composed of a single-chain variable fragment (scFv) of FOLR1 antibody and signaling domains consisting of CD28 and CD3ζ. Both FOLR1-CAR KHYG-1, a natural killer cell line, and FOLR1-CAR T cells recognized FOLR1-positive gastric cancer cells in a MHC-independent manner and induced secretion of various cytokines and caused cell death. Conclusively, this is the first study to demonstrate that CAR KHYG-1/T cells targeting FOLR1 are effective against FOLR1-positive gastric cancer cells.


Subject(s)
Folate Receptor 1/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Stomach Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Jurkat Cells , K562 Cells , Mice , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Stomach Neoplasms/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
10.
Eur J Med Chem ; 126: 536-549, 2017 Jan 27.
Article in English | MEDLINE | ID: mdl-27915169

ABSTRACT

The piperidine fragment in ceritinib was replaced with diverse aliphatic amines to improve inherent resistance issues of ceritinib. While most of the prepared compounds exhibit as similar in vitro activities as ceritinib, compound 10 shows encouraging activities against wild-type ALK as well as crizotinib-resistant mutants including extremely resistant G1202R mutant with an IC50 of 1.8 nM. Furthermore, pharmacokinetic profiles of 10 is apparently better than that of ceritinib. In murine xenograft studies, compound 10 turns out to be as active as ceritinib, suggesting that further optimization of 10 may lead to clinical candidates overcoming ALK mutant issues.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/chemistry , Pyrazoles , Pyridines , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/drug effects , Sulfones/pharmacology , Amines/chemistry , Anaplastic Lymphoma Kinase , Animals , Crizotinib , Drug Resistance, Neoplasm/genetics , Heterografts/drug effects , Humans , Mice , Mutation , Piperidines/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Receptor Protein-Tyrosine Kinases/genetics , Sulfones/chemistry , Sulfones/pharmacokinetics
11.
Cancer Lett ; 374(2): 272-8, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26923554

ABSTRACT

Ceritinib, an ALK inhibitor, was hurriedly approved by the US FDA last year, and demonstrates impressive results in EML4-ALK positive patients. To get a superior ALK inhibitor, we synthesized several ceritinib derivatives with minor modifications to the phenylpiperidine moiety. Biochemical and cellular assays demonstrated the improved activity of KRCA-386 over that of ceritinib. KRCA-386 has superior inhibitory activity against ALK mutants commonly found in crizotinib-resistant patients. Particularly, KRCA-386 has considerably greater activity than ceritinib against the G1202R mutant, one of the most challenging mutations to overcome. The cell cycle analysis indicates that ALK inhibitors induce G1/S arrest, resulting in apoptosis. The in vivo xenograft data also demonstrate that KRCA-386 is significantly better than ceritinib. KRCA-386 dosed at 25 mpk caused 105% tumor growth inhibition (TGI) compared to 72% TGI with ceritinib dosed at 25 mpk. (n = 8, P = 0.010) The kinase profiling assay revealed that several kinases, which are known to be critical for tumor growth, are inhibited by KRCA-386, but not by ceritinib. We anticipate that this characteristic of KRCA-386 enhances its in vivo efficacy. In addition, KRCA-386 shows excellent blood brain barrier penetration compared to ceritinib. These results suggest that KRCA-386 could be useful for crizotinib-resistant patients with brain metastases.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Oncogene Proteins, Fusion/biosynthesis , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Neoplasms/pathology , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/pharmacokinetics , Sulfones/pharmacology , Xenograft Model Antitumor Assays
12.
Biochem Biophys Res Commun ; 464(3): 762-7, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26168728

ABSTRACT

Here, we show the newly synthesized and potent ALK inhibitor having similar scaffold to KRCA-0008, which was reported previously, and its molecular mechanism against cancer cells harboring EML4-ALK fusion protein. Through ALK wild type enzyme assay, we selected two compounds, KRCA-0080 and KRCA-0087, which have trifluoromethyl instead of chloride in R2 position. We characterized these newly synthesized compounds by in vitro and in vivo assays. Enzyme assay shows that KRCA-0080 is more potent against various ALK mutants, including L1196M, G1202R, T1151_L1152insT, and C1156Y, which are seen in crizotinib-resistant patients, than KRCA-0008 is. Cell based assays demonstrate our compounds downregulate the cellular signaling, such as Akt and Erk, by suppressing ALK activity to inhibit the proliferation of the cells harboring EML4-ALK. Interestingly, our compounds induced strong G1/S arrest in H3122 cells leading to the apoptosis, which is proved by PARP-1 cleavage. In vivo H3122 xenograft assay, we found that KRCA-0080 shows significant reduction in tumor size compared to crizotinib and KRCA-0008 by 15-20%. Conclusively, we report a potent ALK inhibitor which shows significant in vivo efficacy as well as excellent inhibitory activity against various ALK mutants.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Crizotinib , Female , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL