Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Nat Med ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992129

ABSTRACT

Adoptive cell transfer (ACT) with neoantigen-reactive T lymphocytes can mediate cancer regression. Here we isolated unique, personalized, neoantigen-reactive T cell receptors (TCRs) from tumor-infiltrating lymphocytes of patients with metastatic gastrointestinal cancers and incorporated the TCR α and ß chains into gamma retroviral vectors. We transduced autologous peripheral blood lymphocytes and adoptively transferred these cells into patients after lymphodepleting chemotherapy. In a phase 2 single-arm study, we treated seven patients with metastatic, mismatch repair-proficient colorectal cancers who had progressive disease following multiple previous therapies. The primary end point of the study was the objective response rate as measured using RECIST 1.1, and the secondary end points were safety and tolerability. There was no prespecified interim analysis defined in this study. Three patients had objective clinical responses by RECIST criteria including regressions of metastases to the liver, lungs and lymph nodes lasting 4 to 7 months. All patients received T cell populations containing ≥50% TCR-transduced cells, and all T cell populations were polyfunctional in that they secreted IFNγ, GM-CSF, IL-2 and granzyme B specifically in response to mutant peptides compared with wild-type counterparts. TCR-transduced cells were detected in the peripheral blood of five patients, including the three responders, at levels ≥10% of CD3+ cells 1 month post-ACT. In one patient who responded to therapy, ~20% of CD3+ peripheral blood lymphocytes expressed transduced TCRs more than 2 years after treatment. This study provides early results suggesting that ACT with T cells genetically modified to express personalized neoantigen-reactive TCRs can be tolerated and can mediate tumor regression in patients with metastatic colorectal cancers. ClinicalTrials.gov registration: NCT03412877 .

2.
Pharmaceuticals (Basel) ; 16(5)2023 May 02.
Article in English | MEDLINE | ID: mdl-37242468

ABSTRACT

Rivaroxaban (RIV) is one of the direct oral anticoagulants used to prevent and treat venous and arterial thromboembolic events. Considering the therapeutic indications, RIV is likely to be concomitantly administered with various other drugs. Among these is carbamazepine (CBZ), one of the recommended first-line options to control seizures and epilepsy. RIV is a strong substrate of cytochrome P450 (CYP) enzymes and Pgp/BCRP efflux transporters. Meanwhile, CBZ is well known as a strong inducer of these enzymes and transporters. Therefore, drug-drug interaction (DDI) between CBZ and RIV is expected. This study aimed to predict the DDI profile of CBZ and RIV in humans by using a population pharmacokinetics (PK) model-based approach. We previously investigated the population PK parameters of RIV administered alone or with CBZ in rats. In this study, those parameters were extrapolated from rats to humans by using simple allometry and liver blood flow scaling, and then applied to back-simulate the PK profiles of RIV in humans (20 mg RIV per day) used alone or with CBZ (900 mg CBZ per day). Results showed that CBZ significantly reduced RIV exposure. The AUCinf and Cmax of RIV decreased by 52.3% and 41.0%, respectively, following the first RIV dose, and by 68.5% and 49.8% at the steady state. Therefore, the co-administration of CBZ and RIV warrants caution. Further studies investigating the extent of DDIs between these drugs should be conducted in humans to fully understand their safety and effects.

3.
Pharmaceutics ; 15(1)2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36678810

ABSTRACT

Tegoprazan is a novel potassium-competitive acid blocker (P-CAB) developed by CJ Healthcare (Korea) for the treatment of gastroesophageal reflux disease and helicobacter pylori infections. Tegoprazan is mainly metabolized by cytochrome P450 (CYP) 3A4. Considering the therapeutic indications, tegoprazan is likely to be administered in combination with various drugs. Therefore, the investigation of drug-drug interactions (DDI) between tegoprazan and CYP3A4 perpetrators is imperative. In the present study, we first aimed to develop a physiologically based pharmacokinetic (PK) model for tegoprazan and its major metabolite, M1, using PK-Sim®. This model was applied to predict the DDI between tegoprazan and CYP3A4 perpetrators. Clarithromycin, a potent inhibitor of CYP3A4, and rifampicin, a strong inducer of CYP3A4, were selected as case studies. Our results show that clarithromycin significantly increased the exposure of tegoprazan. The area under the concentration-time curve (AUC) and Cmax of tegoprazan in the steady state increased up to 4.54- and 2.05-fold, respectively, when tegoprazan (50 mg, twice daily) was coadministered with clarithromycin (500 mg, three times daily). Rifampicin significantly reduced the exposure of tegoprazan. The AUC and Cmax of tegoprazan were reduced by 5.71- and 3.51-fold when tegoprazan was coadministered with rifampicin (600 mg, once daily). Due to the high DDI potential, the comedication of tegoprazan with CYP3A4 perpetrators should be controlled. The dosage adjustment for each individual is suggested.

4.
CPT Pharmacometrics Syst Pharmacol ; 11(11): 1430-1442, 2022 11.
Article in English | MEDLINE | ID: mdl-36193622

ABSTRACT

Rivaroxaban (RIV; Xarelto; Janssen Pharmaceuticals, Beerse, Belgium) is one of the direct oral anticoagulants. The drug is a strong substrate of cytochrome P450 (CYP) enzymes and efflux transporters. This study aimed to develop a physiologically-based pharmacokinetic (PBPK) model for RIV. It contained three hepatic metabolizing enzyme reactions (CYP3A4, CYP2J2, and CYP-independent) and two active transporter-mediated transfers (P-gp and BCRP transporters). To illustrate the performance of the developed RIV PBPK model on the prediction of drug-drug interactions (DDIs), carbamazepine (CBZ) was selected as a case study due to the high DDI potential. Our study results showed that CBZ significantly reduces the exposure of RIV. The area under the concentration-time curve from zero to infinity (AUCinf ) of RIV was reduced by 35.2% (from 2221.3 to 1438.7 ng*h/ml) and by 25.5% (from 2467.3 to 1838.4 ng*h/ml) after the first dose and at the steady-state, respectively, whereas the maximum plasma concentration (Cmax ) of RIV was reduced by 37.7% (from 266.3 to 166.1 ng/ml) and 36.4% (from 282.3 to 179.5 ng/ml), respectively. The developed PBPK model of RIV could be paired with PBPK models of other interested perpetrators to predict DDI profiles. Further studies investigating the extent of DDI between CBZ and RIV should be conducted in humans to gain a full understanding of their safety and effects.


Subject(s)
Models, Biological , Rivaroxaban , Humans , Rivaroxaban/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Neoplasm Proteins , Drug Interactions , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Carbamazepine
5.
Cancer Immunol Res ; 10(8): 932-946, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35749374

ABSTRACT

Adoptive cellular therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from patient-specific mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. We performed whole-exome sequencing on 163 patients with metastatic solid cancers, identified 78 who had TP53 missense mutations, and through immunologic screening, identified 21 unique T-cell reactivities. Here, we report a library of 39 T-cell receptors (TCR) targeting TP53 mutations shared among 7.3% of patients with solid tumors. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner in vitro and in vivo. Twelve patients with chemorefractory epithelial cancers were treated with ex vivo-expanded autologous tumor-infiltrating lymphocytes (TIL) that were naturally reactive against TP53 mutations. However, limited clinical responses (2 partial responses among 12 patients) were seen. These infusions contained low frequencies of mutant p53-reactive TILs that had exhausted phenotypes and showed poor persistence. We also treated one patient who had chemorefractory breast cancer with ACT comprising autologous peripheral blood lymphocytes transduced with an allogeneic HLA-A*02-restricted TCR specific for p53R175H. The infused cells exhibited an improved immunophenotype and prolonged persistence compared with TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these proof-of-concept data suggest that the library of TCRs targeting shared p53 neoantigens should be further evaluated for the treatment of patients with advanced human cancers. See related Spotlight by Klebanoff, p. 919.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neoplasms , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Genes, T-Cell Receptor , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
6.
Science ; 375(6583): 877-884, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35113651

ABSTRACT

The accurate identification of antitumor T cell receptors (TCRs) represents a major challenge for the engineering of cell-based cancer immunotherapies. By mapping 55 neoantigen-specific TCR clonotypes (NeoTCRs) from 10 metastatic human tumors to their single-cell transcriptomes, we identified signatures of CD8+ and CD4+ neoantigen-reactive tumor-infiltrating lymphocytes (TILs). Neoantigen-specific TILs exhibited tumor-specific expansion with dysfunctional phenotypes, distinct from blood-emigrant bystanders and regulatory TILs. Prospective prediction and testing of 73 NeoTCR signature-derived clonotypes demonstrated that half of the tested TCRs recognized tumor antigens or autologous tumors. NeoTCR signatures identified TCRs that target driver neoantigens and nonmutated viral or tumor-associated antigens, suggesting a common metastatic TIL exhaustion program. NeoTCR signatures delineate the landscape of TILs across metastatic tumors, enabling successful TCR prediction based purely on TIL transcriptomic states for use in cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Metastasis , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transcriptome , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Regulatory Networks , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/genetics , Neoplasms/metabolism , RNA-Seq , Single-Cell Analysis
7.
Clin Cancer Res ; 27(18): 5084-5095, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34168045

ABSTRACT

PURPOSE: Immunotherapies mediate the regression of human tumors through recognition of tumor antigens by immune cells that trigger an immune response. Mutations in the RAS oncogenes occur in about 30% of all patients with cancer. These mutations play an important role in both tumor establishment and survival and are commonly found in hotspots. Discovering T-cell receptors (TCR) that recognize shared mutated RAS antigens presented on MHC class I and class II molecules are thus promising reagents for "off-the-shelf" adoptive cell therapies (ACT) following insertion of the TCRs into lymphocytes. EXPERIMENTAL DESIGN: In this ongoing work, we screened for RAS antigen recognition in tumor-infiltrating lymphocytes (TIL) or by in vitro stimulation of peripheral blood lymphocytes (PBL). TCRs recognizing mutated RAS were identified from the reactive T cells. The TCRs were then reconstructed and virally transduced into PBLs and tested. RESULTS: Here, we detect and report multiple novel TCR sequences that recognize nonsynonymous mutant RAS hotspot mutations with high avidity and specificity and identify the specific class-I and -II MHC restriction elements involved in the recognition of mutant RAS. CONCLUSIONS: The TCR library directed against RAS hotspot mutations described here recognize RAS mutations found in about 45% of the Caucasian population and about 60% of the Asian population and represent promising reagents for "off-the-shelf" ACTs.


Subject(s)
Immunotherapy, Adoptive , Mutation , Neoplasms/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/therapeutic use , ras Proteins/genetics , Humans
8.
J Immunother ; 44(1): 1-8, 2021 01.
Article in English | MEDLINE | ID: mdl-33086340

ABSTRACT

Engineered T cells expressing tumor-specific T-cell receptors (TCRs) are emerging as a mode of personalized cancer immunotherapy that requires identification of TCRs against the products of known driver mutations and novel mutations in a timely fashion. We present a nonviral and non-next-generation sequencing platform for rapid, and efficient neoantigen-specific TCR identification and evaluation that does not require the use of recombinant cloning techniques. The platform includes an innovative method of TCRα detection using Sanger sequencing, TCR pairings and the use of TCRα/ß gene fragments for putative TCR evaluation. Using patients' samples, we validated and compared our new methods head-to-head with conventional approaches used for TCR discovery. Development of a unique demultiplexing method for identification of TCRα, adaptation of synthetic TCRs for gene transfer, and a reliable reporter system significantly shortens TCR discovery time over conventional methods and increases throughput to facilitate testing prospective personalized TCRs for adoptive cell therapy.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/genetics , Immunotherapy, Adoptive/methods , Sequence Analysis, DNA/methods , T-Lymphocytes/metabolism , Antigens, Neoplasm/immunology , Cells, Cultured , Coculture Techniques , Genes, T-Cell Receptor alpha , Humans , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
9.
Pharmaceutics ; 12(12)2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33291732

ABSTRACT

Galgeuntang (GGT), a traditional herbal medicine, is widely co-administered with acetaminophen (AAP) for treatment of the common cold, but this combination has not been the subject of investigation. Therefore, we investigated the herb-drug interaction between GGT and AAP by population pharmacokinetics (PKs) modeling and simulation studies. To quantify PK parameters and identify drug interactions, an open label, three-treatment, three-period, one-sequence (AAP alone, GGT alone, and AAP and GGT in combination) clinical trial involving 12 male healthy volunteers was conducted. Ephedrine (EPD), the only GGT component detected, was identified using a one-compartment model. The PKs of AAP were described well by a one-compartment model and exhibited two-phase absorption (rapid followed by slow) and first-order elimination. The model showed that EPD significantly influenced the PKs of AAP. The simulation results showed that at an AAP dose of 1000 mg × 4 times daily, the area under the concentration versus time curve of AAP increased by 16.4% in the presence of GGT compared to AAP only. In conclusion, the PKs of AAP were affected by co-administration of GGT. Therefore, when AAP is combined with GGT, adverse effects related to overdose of AAP could be induced possibly.

10.
Pharmaceutics ; 12(11)2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33143037

ABSTRACT

Rivaroxaban (RIV) is commonly prescribed with carbamazepine or phenytoin (CBZ/PHT) in post-stroke seizure or post-stroke epilepsy patients. Although adverse events have been reported in several previous studies when they are coadministered, there are no studies of the interactions between these drugs. Therefore, our study was conducted to solve this lack of information. The potential effects of CBZ/PHT were investigated by comparing the pharmacokinetic (PK) and pharmacodynamic (PD) parameters of RIV between the control group (RIV alone) and the test groups (RIV administered with CBZ/PHT) in rats using the noncompartmental analysis (NCA) and the compartmental model approach. The NCA results indicate that AUCt of RIV decreased by 57.9% or 89.7% and Cmax of RIV decreased by 43.3% or 70.0% after administration of CBZ/PHT, respectively. In addition, both CBZ and PHT generally reduced the effects of RIV on the prothrombin times of the blood samples. PK profiles of RIV were most properly described by a two-compartment disposition model with a mixed first- and zero-order absorption kinetics and a first-order elimination kinetics. The compartmental model approach showed that a 211% or 1030% increase in CL/F of RIV and a 33.9% or 43.4% increase in D2 of RIV were observed in the test groups by the effects of CBZ/PHT, respectively. In conclusion, CBZ and PHT significantly reduced RIV exposure and therefore reduced the therapeutic effects of RIV. Consequently, this might result in adverse events due to insufficient RIV concentration to attain its therapeutic effects. Further studies are needed to validate this finding.

11.
CPT Pharmacometrics Syst Pharmacol ; 9(10): 584-595, 2020 10.
Article in English | MEDLINE | ID: mdl-32945613

ABSTRACT

HL2351 (hIL-1Ra-hyFc) is a novel recombinant protein formed by the fusion of two human interleukin-1 receptor antagonist components into one antibody-derived fragment crystallizable portion. Although HL2351 has a pharmacological mechanism of action similar to that of anakinra as a commercialized biopharmaceutical drug, HL2351 has been desired to reduce the dose frequency and improve therapeutic efficacy due to its long circulation half-life. In this study, we aimed to develop a population pharmacokinetic (PK) model for HL2351 using a neonatal Fc receptor (FcRn)-mediated recycling model based on a quasi-steady-state approximation of target-mediated drug disposition (TMDD) for the description of interactions between the drug and FcRn. FcRn recycling was expected in the case of HL2351 because of PK related to the antibody portion. A TMDD model was also applied to describe interactions of IL1R with HL2351 or anakinra. PK data were collected from a phase I study conducted in six groups (1, 2, 4, 8, 12 mg/kg HL2351 and 100 mg anakinra single subcutaneous administration; n = 8 per group). In consequence, the PK of anakinra and HL2351 following administration of multiple doses at different dosages were simulated. Optimized doses were considered based on average concentrations of IL1R bound to anakinra and HL2351. HL2351 at doses of 326 mg or 4.267, 4.982, 5.288, 5.458, or 5.748 mg/kg once weekly or HL2351 at 1726 mg or 21.92, 26.86, 29.10, 30.36, or 32.53 mg/kg once biweekly would have similar therapeutic effects with anakinra at a dose of 100 mg or 1, 2, 3, 4, or 8 mg/kg administered once daily, respectively.


Subject(s)
Antirheumatic Agents/pharmacokinetics , Histocompatibility Antigens Class I/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacokinetics , Receptors, Fc/metabolism , Recombinant Proteins/pharmacology , Adult , Antibodies, Monoclonal/pharmacokinetics , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/pharmacology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Biological Products , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Half-Life , Humans , Injections, Subcutaneous , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin 1 Receptor Antagonist Protein/pharmacology , Male , Middle Aged , Pharmacokinetics , Republic of Korea/epidemiology
12.
Environ Pollut ; 259: 113815, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31884210

ABSTRACT

Increasing soil contamination of arsenic (As) and antimony (Sb) is posing a serious concern to human health. Due to insufficient studies on Sb, the biogeochemical behaviour and plant uptake of Sb are assumed to be similar to that of As. As part of extensive research unravelling As and Sb biogeochemistry and plant uptake, the diffusive gradients in thin films (DGT) technique and sequential extraction procedure (SEP) were applied to evaluate As and Sb uptake by the white icicle radish (Raphanus sativus) cultivated in diluted cattle dip soils contaminated with As only and diluted mining soils contaminated with both As and Sb under agricultural conditions. Labile As and Sb in these soils measured by DGT (CDGT), soil solution (Csol), and SEP (CSEP-labile), were compared with As and Sb bioaccumulation in R. sativus tissues. Regardless of contamination sources and measurement techniques, the results showed that As was consistently more labile than Sb although total As concentrations in two soil types were lower than total Sb. Labile As in cattle dip soils was higher than that in mining soils, although there were no significant differences in soil As concentrations. The analysis of R. sativus tissues revealed that the overall As bioaccumulation was 4.5-fold higher than for Sb, and that As translocation to shoots was limited. In contrast, considerable Sb translocation to shoots was observed. The As and Sb bioaccumulation were strongly correlated with their CSEP-labile, CDGT, and Csol (R2 = 0.87-0.99), demonstrating the effectiveness of these techniques in predicting As and Sb in the white icicle radish. Compared with the cherry bell radish previously studied, the white icicle radish exhibited higher bioaccumulation factors (BAF) for Sb, but lower BAF for As, and lower translocation of As and Sb to shoots, providing understanding of how As and Sb are accumulated by radish cultivars.


Subject(s)
Antimony/metabolism , Arsenic/metabolism , Environmental Monitoring , Raphanus/metabolism , Soil Pollutants/metabolism , Antimony/chemistry , Arsenic/chemistry , Humans , Soil , Soil Pollutants/chemistry
13.
J Clin Invest ; 129(11): 4992-5004, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31609250

ABSTRACT

Tumor-resident lymphocytes can mount a response against neoantigens expressed in microsatellite-stable gastrointestinal (GI) cancers, and adoptive transfer of neoantigen-specific lymphocytes has demonstrated antitumor activity in selected patients. However, whether peripheral blood could be used as an alternative minimally invasive source to identify lymphocytes targeting neoantigens in patients with GI cancer with relatively low mutation burden is unclear. We used a personalized high-throughput screening strategy to investigate whether PD-1 expression in peripheral blood could be used to identify CD8+ or CD4+ lymphocytes recognizing neoantigens identified by whole-exome sequencing in 7 patients with GI cancer. We found that neoantigen-specific lymphocytes were preferentially enriched in the CD8+PD-1+/hi or CD4+PD-1+/hi subsets, but not in the corresponding bulk or PD-1- fractions. In 6 of 7 individuals analyzed we identified circulating CD8+ and CD4+ lymphocytes targeting 6 and 4 neoantigens, respectively. Moreover, neoantigen-reactive T cells and a T cell receptor (TCR) isolated from the CD8+PD-1+ subsets recognized autologous tumor, albeit at reduced levels, in 2 patients with available cell lines. These data demonstrate the existence of circulating T cells targeting neoantigens in GI cancer patients and provide an approach to generate enriched populations of personalized neoantigen-specific lymphocytes and isolate TCRs that could be exploited therapeutically to treat cancer.


Subject(s)
Antigens, Neoplasm/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Neoplasms/immunology , Neoplasm Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Female , Humans , Male
14.
Cancer Discov ; 9(8): 1022-1035, 2019 08.
Article in English | MEDLINE | ID: mdl-31164343

ABSTRACT

Immunotherapies can mediate regression of human tumors with high mutation rates, but responses are rarely observed in patients with common epithelial cancers. This raises the question of whether patients with these common cancers harbor T lymphocytes that recognize mutant proteins expressed by autologous tumors that may represent ideal targets for immunotherapy. Using high-throughput immunologic screening of mutant gene products identified via whole-exome sequencing, we identified neoantigen-reactive tumor-infiltrating lymphocytes (TIL) from 62 of 75 (83%) patients with common gastrointestinal cancers. In total, 124 neoantigen-reactive TIL populations were identified, and all but one of the neoantigenic determinants were unique. The results of in vitro T-cell recognition assays demonstrated that 1.6% of the gene products encoded by somatic nonsynonymous mutations were immunogenic. These findings demonstrate that the majority of common epithelial cancers elicit immune recognition and open possibilities for cell-based immunotherapies for patients bearing these cancers. SIGNIFICANCE: TILs cultured from 62 of 75 (83%) patients with gastrointestinal cancers recognized neoantigens encoded by 1.6% of somatic mutations expressed by autologous tumor cells, and 99% of the neoantigenic determinants appeared to be unique and not shared between patients.This article is highlighted in the In This Issue feature, p. 983.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Disease Susceptibility , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/metabolism , Mutation , Biomarkers, Tumor , Gastrointestinal Neoplasms/pathology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
15.
Nat Commun ; 10(1): 449, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30683863

ABSTRACT

T cells targeting shared oncogenic mutations can induce durable tumor regression in epithelial cancer patients. Such T cells can be detected in tumor infiltrating lymphocytes, but whether such cells can be detected in the peripheral blood of patients with the common metastatic epithelial cancer patients is unknown. Using a highly sensitive in vitro stimulation and cell enrichment of peripheral memory T cells from six metastatic cancer patients, we identified and isolated CD4+, and CD8+ memory T cells targeting the mutated KRASG12D and KRASG12V variants, respectively, in three patients. In an additional two metastatic colon cancer patients, we detected CD8+ neoantigen-specific cells targeting the mutated SMAD5 and MUC4 proteins. Therefore, memory T cells targeting unique as well as shared somatic mutations can be detected in the peripheral blood of epithelial cancer patients and can potentially be used for the development of effective personalized T cell-based cancer immunotherapy across multiple patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Mucin-4/immunology , Proto-Oncogene Proteins p21(ras)/immunology , Smad5 Protein/immunology , Antigen Presentation , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Separation/methods , Coculture Techniques , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Immunologic Memory , Lymphatic Metastasis , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Molecular Targeted Therapy , Mucin-4/genetics , Mutation , Neoplastic Cells, Circulating/immunology , Neoplastic Cells, Circulating/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Smad5 Protein/genetics , Transduction, Genetic
16.
Eur J Pharm Sci ; 118: 154-164, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29604332

ABSTRACT

Fixed dose combination (FDC) of valsartan (VAL) and hydrochlorothiazide (HCT) or VAL and amlodipine (AML) has been available in many countries for the treatment of hypertension. Due to drug-drug interaction potentials, in the current study we aimed to evaluate potent effects of HCT and AML on pharmacokinetics (PKs) of VAL when they are orally co-administered as FDC (VAL/HCT at 80/12.5 mg or 160/12.5 mg; and VAL/AML at 160/5 mg or 160/10 mg) products in healthy Korean subjects. Population pharmacokinetic (PK) modeling and analysis were performed by the nonlinear mixed-effects modeling software. PKs of VAL was described by two-compartment disposition model, first-order elimination, four-sequential first-order absorption model, correlation between apparent clearances and volumes of distribution, and lag time. For all FDCs, there were no statistically significant differences in both maximum concentration and areas under the concentration-time curves (AUCs) of VAL in comparison to those when administered VAL alone, except the combination of VAL/AML at 160/10 mg, where AUC0-∞ increased by 11.8% in mean and 6.86% in median. In addition, there was an increasing trend in time to reach peak (Tmax) of VAL in FDCs, where it was increased by 0.22-0.34 h in mean and 0.40-0.44 h in median, except the combination of VAL/HCT at 160/12.5 mg. However, these differences in AUC0-∞ and Tmax might not be considered as clinically important. In conclusion, HCT or AML has no potent effect on PKs of VAL when they are co-administered as FDC products. No dose adjustment for VAL is recommended when co-administered with HCT or AML.


Subject(s)
Amlodipine/pharmacology , Antihypertensive Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Hydrochlorothiazide/pharmacology , Models, Biological , Valsartan/pharmacokinetics , Administration, Oral , Antihypertensive Agents/blood , Asian People , Drug Interactions , Healthy Volunteers , Humans , Male , Valsartan/blood
17.
Environ Sci Process Impacts ; 20(5): 833-844, 2018 May 23.
Article in English | MEDLINE | ID: mdl-29693094

ABSTRACT

Elevated concentrations of As and Sb impact environmental quality and human health. In this study total and bioavailable As and Sb were measured from recently and historically contaminated soils and the phytotoxicity of these soils was evaluated with Ipomoea aquatica (35-d exposure from germination) using biomass, length of plant tissues and photosynthetic efficiency. As and Sb were both present within the soil (co-contaminated). The bioavailable As and Sb in soils were determined by a Sequential Extraction Procedure (SEP) and compared to total soil concentrations and bioaccumulation in the edible parts of I. aquatica. For both As and Sb, bioavailable concentrations increased proportionally with the total soil concentrations and greater bioavailability in recently contaminated soil was observed. Tissue dry mass and length drastically reduced with increasing total and SEP-bioavailable As and Sb soil concentrations. The total soil concentration was a less sensitive measure of the phytotoxicity of As and Sb than the bioavailable fraction. Shoot length was inhibited by 50% (EC50) at bioavailable As concentrations of 80-96 mg kg-1 in both recently and historically contaminated soils; however, bioavailable Sb EC50 for shoot length was achieved at lower bioavailable concentrations, 96 (42-219) and 12 (7-19) mg kg-1 in recently contaminated soils and historically contaminated soils, respectively. Shoot biomass was inhibited by 50% (EC50) at bioavailable As concentrations of 11 (4-30) and 49 (37-65) mg kg-1 in recently and historically contaminated soils, respectively whereas this occurred at much lower bioavailable Sb concentrations, 2-5 mg kg-1 in both recently and historically contaminated soils. Aging is important in contaminated soils, it decreases the lability of As and Sb and hence their bioavailability to agricultural plants, thus posing a lower risk of exposure of these metalloids to humans through agricultural plants grown in contaminated soils.


Subject(s)
Antimony/toxicity , Arsenic/toxicity , Environmental Monitoring/methods , Ipomoea/drug effects , Soil Pollutants/toxicity , Antimony/analysis , Antimony/pharmacokinetics , Arsenic/analysis , Arsenic/pharmacokinetics , Biological Availability , Crops, Agricultural , Humans , Ipomoea/growth & development , Ipomoea/metabolism , Soil/chemistry , Soil Pollutants/analysis , Soil Pollutants/pharmacokinetics
18.
Int J Clin Pharmacol Ther ; 56(6): 292-300, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29648532

ABSTRACT

OBJECTIVE: This study was conducted to determine whether a fixed-dose combination (FDC) tablet of repaglinide/metformin (2/500 mg) is equivalent to coadministration of equivalent doses of individual (EDI) tablets of repaglinide (2 mg) and metformin (500 mg) in healthy Korean male subjects. MATERIALS AND METHODS: This study was conducted as an open-label, randomized, single-dose, two-period, two-sequence crossover design in 50 healthy Korean male subjects who received an FDC tablet or EDI tablets. Plasma concentrations of repaglinide and metformin were determined for up to 24 hours using a validated UPLC-MS/MS method. Bioequivalence was assessed according to current guidelines issued by the U.S. Food and Drug Administration (FDA) and Korean legislation. Tolerability was also evaluated throughout the study via subject interview, vital signs, and blood sampling. RESULTS: Point estimates (90% CIs) for AUC0-t, AUC0-∞, and Cmax based on EDI tablets were 110.07 (102.25 - 118.49), 109.90 (101.70 - 118.39), and 112.60 (101.49 - 124.85), respectively, for repaglinide. They were 95.18 (89.62 - 101.05), 95.00 (89.74 - 100.65), and 98.44 (92.72 - 104.50), respectively, for metformin. These results satisfied the bioequivalence criteria of 80.00 - 125.00% proposed by the FDA and Korean legislation. CONCLUSION: Results of pharmacokinetic analysis suggested that repaglinide and metformin in FDC tablets were bioequivalent to EDI tablets of repaglinide (2 mg) and metformin (500 mg) in healthy Korean male subjects. Both formulations appeared to be well tolerated.
.


Subject(s)
Carbamates/administration & dosage , Carbamates/pharmacokinetics , Metformin/administration & dosage , Metformin/pharmacokinetics , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Adult , Cross-Over Studies , Drug Combinations , Drug Therapy, Combination , Humans , Male , Tablets , Therapeutic Equivalency
19.
J Pharmacokinet Pharmacodyn ; 44(6): 567-579, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29018999

ABSTRACT

The objective of this study was to perform population pharmacokinetic (PK) analysis of gabapentin in healthy Korean subjects and to investigate the possible effect of genetic polymorphisms (1236C > T, 2677G > T/A, and 3435C > T) of ABCB1 gene on PK parameters of gabapentin. Data were collected from bioequivalence studies, in which 173 subjects orally received three different doses of gabapentin (300, 400, and 800 mg). Only data from reference formulation were used. Population pharmacokinetics (PKs) of gabapentin was estimated using a nonlinear mixed-effects model (NONMEM). Gabapentin showed considerable inter-individual variability (from 5.2- to 8.7-fold) in PK parameters. Serum concentration of gabapentin was well fitted by a one-compartment model with first-order absorption and lag time. An inhibitory Emax model was applied to describe the effect of dose on bioavailability. The oral clearance was estimated to be 11.1 L/h. The volume of distribution was characterized as 81.0 L. The absorption rate constant was estimated at 0.860 h-1, and the lag time was predicted at 0.311 h. Oral bioavailability was estimated to be 68.8% at dose of 300 mg, 62.7% at dose of 400 mg, and 47.1% at dose of 800 mg. The creatinine clearance significantly influenced on the oral clearance (P < 0.005) and ABCB1 2677G > T/A genotypes significantly influenced on the absorption rate constant (P < 0.05) of gabapentin. However, ABCB1 1236C > T and 3435C > T genotypes showed no significant effect on gabapentin PK parameters. The results of the present study indicate that the oral bioavailability of gabapentin is decreased when its dosage is increased. In addition, ABCB1 2677G > T/A polymorphism can explain the substantial inter-individual variability in the absorption of gabapentin.


Subject(s)
Amines/pharmacokinetics , Analgesics/pharmacokinetics , Asian People/genetics , Cyclohexanecarboxylic Acids/pharmacokinetics , Pharmacogenomic Variants/genetics , Polymorphism, Genetic/genetics , gamma-Aminobutyric Acid/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/blood , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Dose-Response Relationship, Drug , Gabapentin , Humans , Male , Republic of Korea/epidemiology , Young Adult
20.
J Pharmacokinet Pharmacodyn ; 44(4): 291-303, 2017 08.
Article in English | MEDLINE | ID: mdl-28316019

ABSTRACT

In this study, the population pharmacokinetic (PK) analysis of rebamipide (Reba) in healthy male Korean subjects was analyzed using the nonlinear mixed effects modeling method. The possible effects of physiological covariates and the multidrug resistance (MDR1) gene 3435C>T polymorphism on PK parameters were also investigated. Data were collected from a bioequivalence study, in which 26 subjects who participated in the study were administered a single oral dose of 100 mg Reba; only data from the reference formulation were used. Reba showed a relatively large inter-individual variability (from 2.6- to 3.3-fold) in the PK parameters with double peaks or the concentration plateau after the peak concentration in its serum concentration-time profiles. The population PKs of Reba was best described by a one-compartment model with three fraction absorption processes followed a single Weibull-type function and two first-order kinetics, and lag times. The study suggests that the efflux transporter MDR1 3435C>T allele affects the substantial inter-individual variability in the absorption of Reba according to genetic polymorphism. A significant difference was found in the absorption rate ka 1 among the MDR1 3435C>T genotype groups (P < 0.05) (CT group, 79.8% increase; and TT group, 115% increase). The use of combined MDR1 3435C>T and body mass index as covariates for ka 1 exerted a more significant effect (P < 0.05). In addition, body surface area significantly affected the apparent total clearance (P < 0.05).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Alanine/analogs & derivatives , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/pharmacokinetics , Gastrointestinal Absorption/physiology , Quinolones/administration & dosage , Quinolones/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , Administration, Oral , Adult , Alanine/administration & dosage , Alanine/blood , Alanine/pharmacokinetics , Anti-Ulcer Agents/blood , Cross-Over Studies , Gastrointestinal Absorption/drug effects , Healthy Volunteers , Humans , Male , Quinolones/blood , Republic of Korea/epidemiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...