51
|
Essential Oil Composition of Alluaudia procera and in Vitro Biological Activity on Two Drug-Resistant Models. Molecules 2019; 24:molecules24162871. [PMID: 31394879 PMCID: PMC6720003 DOI: 10.3390/molecules24162871] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/03/2019] [Accepted: 08/05/2019] [Indexed: 01/02/2023] Open
Abstract
Drug resistance is a major obstacle in antibiotic and antitumor chemotherapy. In response to the necessity to find new therapeutic strategies, plant secondary metabolites including essential oils (EOs) may represent one of the best sources. EOs in plants act as constitutive defenses against biotic and abiotic stress, and they play an important role in the pharmacology for their low toxicity, good pharmacokinetic and multitarget activity. In this context, natural products such as EOs are one of the most important sources of drugs used in pharmaceutical therapeutics. The aim of this paper was to identify the chemical composition of the essential oil of Alluaudia procera leaves, obtained by hydrodistillation and analysed by gas chromatography-mass spectrometry, and to verify its biological activities on acute myeloid leukemia cancer cell HL60 and its multidrugresistant variant HL60R and the Gram-positive Staphylococcus aureus exhibiting multi-antibiotic resistance. We speculate that cytotoxic and antibiotic effects observed in the tested resistant models may be due to the coordinate activities of forty compounds detected or to the C16 macrocyclic lactones which are the major ones (30%). Our data confirm the possibility of using EOs as therapeutic strategies in resistant models is due to the heterogeneous composition of the oils themselves.
Collapse
|
52
|
m-TOR inhibitors and risk of Pneumocystis pneumonia after solid organ transplantation: a systematic review and meta-analysis. Eur J Clin Pharmacol 2019; 75:1471-1480. [DOI: 10.1007/s00228-019-02730-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/20/2019] [Indexed: 12/22/2022]
|
53
|
Lee EJ, Kim HK, Ahn S, Lee W, Kim HS, Chun S, Min WK. Accuracy evaluation of automated electrochemiluminescence immunoassay for everolimus and sirolimus compared to liquid chromatography-tandem mass spectrometry. J Clin Lab Anal 2019; 33:e22941. [PMID: 31197901 PMCID: PMC6757180 DOI: 10.1002/jcla.22941] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/03/2019] [Accepted: 05/16/2019] [Indexed: 12/22/2022] Open
Abstract
Background We evaluated the analytical performance of a newly developed electrochemiluminescence immunoassay for everolimus and sirolimus compared to that of liquid chromatography‐tandem mass spectrometry (LC‐MS/MS). Methods According to Clinical and Laboratory Standards Institute guidelines, the analytical performance including precision, recovery, linearity, and carryover was evaluated. For correlation evaluation, the results of Elecsys® analysis of everolimus and sirolimus were compared with those of LC‐MS/MS using 120 samples from patients treated with everolimus or sirolimus. Results The within‐run and total imprecision values were as follows: 2.3%‐4.5% and 4.5%‐6.4% for the everolimus assay; 3.3%‐4.8% and 4.7%‐8.1% for the sirolimus assay, respectively. The measured concentration was linear over the range of 0.718‐27.585 ng/mL for everolimus analysis and 0.789‐26.880 ng/mL for sirolimus analysis (all R2 > 0.99). Recovery was 93.5%‐105.5% for the everolimus assay and 99.2%‐109.1% for the sirolimus assay (except lowest levels). Carryover was −1.09% for the everolimus assay and −0.12% for the sirolimus assay. The results of the two chemiluminescence immunoassays showed acceptable correlations with those of LC‐MS/MS (R = 0.9585 and R = 0.9799, respectively). The two immunoassays showed slightly proportional biases compared to LC‐MS/MS. Conclusion Elecsys® Everolimus and Sirolimus assays showed acceptable analytical performance in precision, linearity, and correlation compared to LC‐MS/MS These methods can be adopted in the clinical laboratory for rapid therapeutic drug monitoring of patients who require treatment with immunosuppressants.
Collapse
Affiliation(s)
- Eun Jin Lee
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Laboratory Medicine, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-Si, Korea
| | - Hyun-Ki Kim
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sunyoung Ahn
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woochang Lee
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyun Soo Kim
- Department of Laboratory Medicine, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-Si, Korea
| | - Sail Chun
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Won-Ki Min
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
54
|
Turunç V, Açıkgöz S, Dheir H. Early Switch to Mammalian Target of Rapamycin Inhibitors Is a Sustainable Treatment Approach in Renal Transplant Recipients: 7-Year Results. Transplant Proc 2019; 51:1070-1073. [DOI: 10.1016/j.transproceed.2019.01.098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/21/2019] [Indexed: 10/27/2022]
|
55
|
Use of Mammalian Target of Rapamycin Inhibitors for Pancreas Transplant Immunosuppression Is Associated With Improved Allograft Survival and Improved Early Patient Survival. Pancreas 2019; 48:644-651. [PMID: 31091210 DOI: 10.1097/mpa.0000000000001322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Mammalian targets of rapamycin inhibitors (mTORi) are considered second-line immunosuppression agents because of associated increases in rejection and impaired wound healing. Recent reports indicate mTORi have been linked to improved survival, decreased inflammatory response in pancreatitis, and antiproliferative and antiangiogenic activity. Mammalian targets of rapamycin inhibitors have not been extensively analyzed in pancreas transplant recipients. METHODS Adults with pancreas and kidney-pancreas transplants from 1987 to 2016 in the United Network for Organ Sharing database were analyzed (N = 25,837). Subjects were stratified into 2 groups: use of mTORi (n = 4174) and use of non-mTORi-based immunosuppression (n = 21,663). The log-rank test compared survival rates. Univariate and multivariate Cox regression analyses assessed patient and graft survival. RESULTS Mammalian targets of rapamycin inhibitors were associated with a 7% risk reduction in allograft failure (hazard ratio, 0.931; P = 0.006). Allograft survival rates were significantly different between mTORi versus non-mTORi (P < 0.0001).The mTORi group showed a significantly higher patient survival rate 1, 3, 5, and 10 years posttransplant compared. Patient survival at 15 years was not significantly different. CONCLUSIONS The use of mTORi for immunosuppression in pancreas transplant is associated with improved allograft survival and early patient survival posttransplant (up to 10 years).
Collapse
|
56
|
Stove V, Ramos PA, Wallemacq P, Vogeser M, Schuetzenmeister A, Schmiedel C, Shipkova M. Measurement of sirolimus concentrations in human blood using an automated electrochemiluminescence immunoassay (ECLIA): a multicenter evaluation. Clin Chem Lab Med 2019; 56:764-775. [PMID: 29206642 DOI: 10.1515/cclm-2017-0583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/28/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) of sirolimus is essential in transplant recipients. We evaluated the performance of a new electrochemiluminescence immunoassay (ECLIA) for measuring sirolimus concentrations in whole blood at five European laboratories. METHODS Study assessments included repeatability, intermediate precision and functional sensitivity (concentration at coefficient of variation [CV] of 20%) experiments. Method comparisons with liquid chromatography-tandem mass spectrometry (LC-MS/MS; reference method) and two immunoassays (chemiluminescent microparticle immunoassay [CMIA] and antibody-conjugated magnetic immunoassay [ACMIA]) were performed using native samples from patients with kidney transplants. RESULTS Imprecision testing CVs were ≤6.4% and ≤10.7% across the sirolimus concentration range for both repeatability and intermediate precision, respectively. The ECLIA showed excellent functional sensitivity: the CV did not reach 20%; the CV at the assay's limit of quantitation (1.5 μg/L) was 7.0%. Agreement between the ECLIA and LC-MS/MS using native kidney samples was close, with weighted Deming regression analysis yielding a slope of 1.05, an intercept of 0.154 μg/L and a Pearson's correlation coefficient (r) of 0.94, while Bland-Altman analysis showed a combined mean bias of 0.41 μg/L (±2 standard deviation [SD], -1.96 to 2.68). The ECLIA also showed good correlation with the two other immunoassays: the CMIA (slope=0.91, intercept=0.112 μg/L and r=0.89) and the ACMIA (slope=0.99, intercept=0.319 μg/L and r=0.97). CONCLUSIONS The ECLIA showed good precision, functional sensitivity and agreement with other methods of sirolimus measurement used in clinical practice, suggesting that the assay is suitable for TDM in transplant recipients and provides an alternative to LC-MS/MS.
Collapse
Affiliation(s)
- Veronique Stove
- Clinical Biologist, Department of Laboratory Medicine, Ghent University and Ghent University Hospital, De Pintelaan 185, Ghent, Belgium
| | - Pedro Alía Ramos
- IDIBELL - Bellvitge Biomedical Research Institute, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Michael Vogeser
- Institute of Laboratory Medicine, Hospital of the University of Munich, Munich, Germany
| | | | | | - Maria Shipkova
- Central Institute for Laboratory Medicine and Clinical Chemistry, Klinikum-Stuttgart, Stuttgart, Germany
| |
Collapse
|
57
|
Litou C, Effinger A, Kostewicz ES, Box KJ, Fotaki N, Dressman JB. Effects of medicines used to treat gastrointestinal diseases on the pharmacokinetics of coadministered drugs: a PEARRL Review. J Pharm Pharmacol 2019; 71:643-673. [PMID: 30062750 DOI: 10.1111/jphp.12983] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/27/2018] [Indexed: 01/06/2025]
Abstract
OBJECTIVES Drugs used to treat gastrointestinal diseases (GI drugs) are widely used either as prescription or over-the-counter (OTC) medications and belong to both the 10 most prescribed and 10 most sold OTC medications worldwide. The objective of this review article is to discuss the most frequent interactions between GI and other drugs, including identification of the mechanisms behind these interactions, where possible. KEY FINDINGS Current clinical practice shows that in many cases, these drugs are administered concomitantly with other drug products. Due to their metabolic properties and mechanisms of action, the drugs used to treat gastrointestinal diseases can change the pharmacokinetics of some coadministered drugs. In certain cases, these interactions can lead to failure of treatment or to the occurrence of serious adverse events. The mechanism of interaction depends highly on drug properties and differs among therapeutic categories. Understanding these interactions is essential to providing recommendations for optimal drug therapy. SUMMARY Interactions with GI drugs are numerous and can be highly significant clinically in some cases. While alterations in bioavailability due to changes in solubility, dissolution rate, GI transit and metabolic interactions can be (for the most part) easily identified, interactions that are mediated through other mechanisms, such as permeability or microbiota, are less well-understood. Future work should focus on characterising these aspects.
Collapse
Affiliation(s)
- Chara Litou
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Angela Effinger
- Department of Pharmacy and Pharmacology, Faculty of Science, University of Bath, Bath, UK
| | - Edmund S Kostewicz
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, Faculty of Science, University of Bath, Bath, UK
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
58
|
Gurău F, Baldoni S, Prattichizzo F, Espinosa E, Amenta F, Procopio AD, Albertini MC, Bonafè M, Olivieri F. Anti-senescence compounds: A potential nutraceutical approach to healthy aging. Ageing Res Rev 2018; 46:14-31. [PMID: 29742452 DOI: 10.1016/j.arr.2018.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/02/2018] [Accepted: 05/03/2018] [Indexed: 01/10/2023]
Abstract
The desire of eternal youth seems to be as old as mankind. However, the increasing life expectancy experienced by populations in developed countries also involves a significantly increased incidence of the most common age-related diseases (ARDs). Senescent cells (SCs) have been identified as culprits of organismal aging. Their number rises with age and their senescence-associated secretory phenotype fuels the chronic, pro-inflammatory systemic state (inflammaging) that characterizes aging, impairing the regenerative ability of stem cells and increasing the risk of developing ARDs. A variegated class of molecules, including synthetic senolytic compounds and natural compounds contained in food, have been suggested to possess anti-senescence activity. Senolytics are attracting growing interest, and their safety and reliability as anti-senescence drugs are being assessed in human clinical trials. Notably, since SCs spread inflammation at the systemic level through pro-oxidant and pro-inflammatory signals, foods rich in polyphenols, which exert antioxidant and anti-inflammatory actions, have the potential to be harnessed as "anti-senescence foods" in a nutraceutical approach to healthier aging. We discuss the beneficial effects of polyphenol-rich foods in relation to the Mediterranean diet and the dietary habits of long-lived individuals, and examine their ability to modulate bacterial genera in the gut.
Collapse
Affiliation(s)
- Felicia Gurău
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Simone Baldoni
- School of Medicinal Sciences and Health Products, University of Camerino, Camerino, Italy
| | | | - Emma Espinosa
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Amenta
- School of Medicinal Sciences and Health Products, University of Camerino, Camerino, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | | | - Massimiliano Bonafè
- DIMES- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy; Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Forlì, Italy.
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy.
| |
Collapse
|
59
|
Brandt C, Hillmann P, Noack A, Römermann K, Öhler LA, Rageot D, Beaufils F, Melone A, Sele AM, Wymann MP, Fabbro D, Löscher W. The novel, catalytic mTORC1/2 inhibitor PQR620 and the PI3K/mTORC1/2 inhibitor PQR530 effectively cross the blood-brain barrier and increase seizure threshold in a mouse model of chronic epilepsy. Neuropharmacology 2018; 140:107-120. [PMID: 30081001 DOI: 10.1016/j.neuropharm.2018.08.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
The mTOR signaling pathway has emerged as a possible therapeutic target for epilepsy. Clinical trials have shown that mTOR inhibitors such as everolimus reduce seizures in tuberous sclerosis complex patients with intractable epilepsy. Furthermore, accumulating preclinical data suggest that mTOR inhibitors may have anti-seizure or anti-epileptogenic actions in other types of epilepsy. However, the chronic use of rapalogs such as everolimus is limited by poor tolerability, particularly by immunosuppression, poor brain penetration and induction of feedback loops which might contribute to their limited therapeutic efficacy. Here we describe two novel, brain-permeable and well tolerated small molecule 1,3,5-triazine derivatives, the catalytic mTORC1/C2 inhibitor PQR620 and the dual pan-PI3K/mTOR inhibitor PQR530. These derivatives were compared with the mTORC1 inhibitors rapamycin and everolimus as well as the anti-seizure drugs phenobarbital and levetiracetam. The anti-seizure potential of these compounds was determined by evaluating the electroconvulsive seizure threshold in normal and epileptic mice. Rapamycin and everolimus only poorly penetrated into the brain (brain:plasma ratio 0.0057 for rapamycin and 0.016 for everolimus). In contrast, the novel compounds rapidly entered the brain, reaching brain:plasma ratios of ∼1.6. Furthermore, they significantly decreased phosphorylation of S6 ribosomal protein in the hippocampus of normal and epileptic mice, demonstrating effective mTOR inhibition. PQR620 and PQR530 significantly increased seizure threshold at tolerable doses. The effect of PQR620 was more marked in epileptic vs. nonepileptic mice, matching the efficacy of levetiracetam. Overall, the novel compounds described here have the potential to overcome the disadvantages of rapalogs for treatment of epilepsy and mTORopathies directly connected to mutations in the mTOR signaling cascade.
Collapse
Affiliation(s)
- Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | | | - Andreas Noack
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Leon A Öhler
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Denise Rageot
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Anna Melone
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alexander M Sele
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
60
|
Tarasewicz A, Dębska-Ślizień A, Rutkowska B, Szurowska E, Matuszewski M. Efficacy and Safety of Mammalian Target of Rapamycin Inhibitor Use-Long-term Follow-up of First Tuberous Sclerosis Complex Patient Treated De Novo With Sirolimus After Kidney Transplantation: A Case Report. Transplant Proc 2018; 50:1904-1909. [PMID: 30056926 DOI: 10.1016/j.transproceed.2018.03.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/25/2018] [Accepted: 03/06/2018] [Indexed: 01/09/2023]
Abstract
Mammalian target of rapamycin inhibitors (mTORI) are increasingly used in the treatment of tuberous sclerosis complex (TSC) and as immunosuppressants after organ transplantation. In TSC patients, mTORI are the treatment of choice after kidney transplantation. It is still under debate if benefits from long-term mTORI use will not be limited by side effects. MATERIALS AND METHODS We report long-term follow-up data of the first TSC patient after kidney transplantation treated with sirolimus de novo. In 2005, a female patient was transplanted with a kidney graft after bilateral nephrectomy due to angiomyolipoma. Initial immunosuppressive treatment consisted of antithymocyte globulin, methylprednisolone, tacrolimus, and, due to TSC diagnosis, sirolimus. Creatinine level at discharge was 1.2 mg/dL. RESULTS Long-term mTORI use resulted in skin lesion regression (angiofibromas, "confetti" skin lesions, shagreen patch) and disease stabilization in brain, abdominal, and chest magnetic resonance imaging/computed tomography scans. Pulmonary function tests showed improvement in restriction and slow deterioration in obstruction and diffusion parameters. Sirolimus related adverse reactions were hyperlipidemia and hypertriglyceridemia and respiratory and urinary tract infections. No gastrointestinal or hematologic symptoms occurred. Sirolimus concentrations ranged between 1.7 and 8.2 ng/mL (mean 4.01 ± 2.09 ng/mL). Since 2009 proteinuria and slow increase in creatinine level have been observed. No biopsy was performed to establish etiology and potential association with mTORI. In 2017 creatinine level was 2.2 mg/dL. CONCLUSION The case of the patient confirms clinical effectiveness and acceptable safety of long-term mTORI treatment. Long-term mTORI use requires meticulous patient observation to optimize dosage, achieve immunosuppressive effect, and improve TSC manifestations with minimal side effects.
Collapse
Affiliation(s)
- A Tarasewicz
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - A Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - B Rutkowska
- Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
| | - E Szurowska
- Department of Radiology, Medical University of Gdańsk, Gdańsk, Poland
| | - M Matuszewski
- Department of Urology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
61
|
Stavrou M, Philip B, Traynor-White C, Davis CG, Onuoha S, Cordoba S, Thomas S, Pule M. A Rapamycin-Activated Caspase 9-Based Suicide Gene. Mol Ther 2018; 26:1266-1276. [PMID: 29661681 PMCID: PMC5993966 DOI: 10.1016/j.ymthe.2018.03.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 02/26/2018] [Accepted: 03/03/2018] [Indexed: 01/20/2023] Open
Abstract
Engineered T cell therapies show considerable promise in the treatment of refractory malignancies. Given the ability of engineered T cells to engraft and persist for prolonged periods along with unpredicted toxicities, incorporation of a suicide gene to allow selective depletion after administration is desirable. Rapamycin is a safe and widely available immunosuppressive pharmaceutical that acts by heterodimerization of FKBP12 with the FRB fragment of mTOR. The apical caspase caspase 9 is activated by homodimerization through its CARD domain. We developed a rapamycin-induced caspase 9 suicide gene. First, we showed that caspase 9 could be activated by a two-protein format with replacement of the CARD domain with both FRB and FKBP12. We next identified an optimal compact single-protein rapamycin caspase 9 (rapaCasp9) by fusing both FRB and FKBP12 with the catalytic domain of caspase 9. Functionality of rapaCasp9 when co-expressed with a CD19 CAR was demonstrated in vitro and in vivo.
Collapse
Affiliation(s)
| | - Brian Philip
- Cancer Institute, University College London, London, UK
| | | | | | | | | | - Simon Thomas
- Autolus Ltd., Forest House, White City, London, UK
| | - Martin Pule
- Autolus Ltd., Forest House, White City, London, UK; Cancer Institute, University College London, London, UK.
| |
Collapse
|
62
|
Ozturk N, Ozturk D, Pala-Kara Z, Kaptan E, Sancar-Bas S, Ozsoy N, Cinar S, Deniz G, Li XM, Giacchetti S, Lévi F, Okyar A. The immune system as a chronotoxicity target of the anticancer mTOR inhibitor everolimus. Chronobiol Int 2018; 35:705-718. [PMID: 29400578 DOI: 10.1080/07420528.2018.1432632] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The circadian timing system controls many biological functions in mammals including xenobiotic metabolism, detoxification, cell proliferation, apoptosis and immune functions. Everolimus is a mammalian target of rapamycin inhibitor, whose immunosuppressant properties are both desired in transplant patients and unwanted in cancer patients, where it is indicated for its antiproliferative efficacy. Here we sought whether everolimus circadian timing would predictably modify its immunosuppressive effects so as to optimize this drug through timing. C57BL/6J mice were synchronized with light-dark 12h:12h, with L onset at Zeitgeber Time (ZT) 0. Everolimus was administered orally to male (5 mg/kg/day) and female mice (15 mg/kg/day) at ZT1, during early rest span or at ZT13, during early activity span for 4 weeks. Body weight loss, as well as hematological, immunological and biochemical toxicities, were determined. Spleen and thymus were examined histologically. Everolimus toxicity was less severe following dosing at ZT13, as compared to ZT1, as shown with least body weight inhibition in both genders; least reductions in thymus weight both in males (p < 0.01) and females (p < 0.001), least reduction in female spleen weight (p < 0.05), and less severe thymic medullar atrophy both in males (p < 0.001) and females (p < 0.001). The mean circulating counts in total leukocytes, total lymphocytes, T-helper and B lymphocytes displayed minor and non-significant changes following dosing at ZT13, while they were decreased by 56.9% (p < 0.01), 45.5% (p < 0.01), 43.1% (p < 0.05) and 48.7% (p < 0.01) after everolimus at ZT1, respectively, in only male mice. Chronotherapy of everolimus is an effective way to increase the general tolerability and decrease toxicity on the immune system.
Collapse
Affiliation(s)
- Narin Ozturk
- a Department of Pharmacology, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| | - Dilek Ozturk
- b Department of Pharmacology, Faculty of Pharmacy , Bezmialem Vakif University , Fatih-İstanbul , Turkey
| | - Zeliha Pala-Kara
- a Department of Pharmacology, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| | - Engin Kaptan
- c Department of Biology, Faculty of Science , Istanbul University , Vezneciler-Istanbul , Turkey
| | - Serap Sancar-Bas
- c Department of Biology, Faculty of Science , Istanbul University , Vezneciler-Istanbul , Turkey
| | - Nurten Ozsoy
- d Department of Biochemistry, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| | - Suzan Cinar
- e Department of Immunology, Aziz Sancar Institute of Experimental Medicine , Istanbul University , Sehremini- Istanbul , Turkey
| | - Gunnur Deniz
- e Department of Immunology, Aziz Sancar Institute of Experimental Medicine , Istanbul University , Sehremini- Istanbul , Turkey
| | - Xiao-Mei Li
- f INSERM UMRS 935 "Modèles de cellules souches malignes et thérapeutiques" , Campus CNRS , Villejuif-Cedex , France
| | - Sylvie Giacchetti
- g AP-HP, Hôpital Saint-Louis, Breast Disease Unit , University Paris Diderot , Paris , France
| | - Francis Lévi
- f INSERM UMRS 935 "Modèles de cellules souches malignes et thérapeutiques" , Campus CNRS , Villejuif-Cedex , France.,h Warwick Systems Biology Centre, Warwick Medical School , Cancer Chronotherapy Unit , Coventry , UK
| | - Alper Okyar
- a Department of Pharmacology, Faculty of Pharmacy , Istanbul University , Beyazit-Istanbul , Turkey
| |
Collapse
|
63
|
Kraig E, Linehan LA, Liang H, Romo TQ, Liu Q, Wu Y, Benavides AD, Curiel TJ, Javors MA, Musi N, Chiodo L, Koek W, Gelfond JAL, Kellogg DL. A randomized control trial to establish the feasibility and safety of rapamycin treatment in an older human cohort: Immunological, physical performance, and cognitive effects. Exp Gerontol 2018; 105:53-69. [PMID: 29408453 DOI: 10.1016/j.exger.2017.12.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 12/31/2017] [Indexed: 12/21/2022]
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) pathway by rapamycin (RAPA), an FDA-approved immunosuppressive drug used as a clinical therapy to prevent solid organ allograft rejection, enhances longevity in mice. Importantly, RAPA was efficacious even when initiated in relatively old animals, suggesting that mTOR inhibition could potentially slow the progression of aging-associated pathologies in older humans (Harrison et al., 2009; Miller et al., 2011). However, the safety and tolerability of RAPA in older human subjects have not yet been demonstrated. Towards this end, we undertook a placebo-controlled pilot study in 25 generally healthy older adults (aged 70-95 years); subjects were randomized to receive either 1 mg RAPA or placebo daily. Although three subjects withdrew, 11 RAPA and 14 controls completed at least 8 weeks of treatment and were included in the analysis. We monitored for changes that would indicate detrimental effects of RAPA treatment on metabolism, including both standard clinical laboratory assays (CBC, CMP, HbA1c) and oral glucose tolerance tests (OGTTs). We also monitored parameters typically associated with aging that could potentially be modified by RAPA; these included cognitive function which was assessed by three different tools: Executive Interview-25 (EXIT25); Saint Louis University Mental Status Exam (SLUMS); and Texas Assessment of Processing Speed (TAPS). In addition, physical performance was measured by handgrip strength and 40-foot timed walks. Lastly, changes in general parameters of healthy immune aging, including serum pro-inflammatory cytokine levels and blood cell subsets, were assessed. Five subjects reported potential adverse side effects; in the RAPA group, these were limited to facial rash (1 subject), stomatitis (1 subject) and gastrointestinal issues (2 subjects) whereas placebo treated subjects only reported stomatitis (1 subject). Although no other adverse events were reported, statistically significant decrements in several erythrocyte parameters including hemoglobin (HgB) and hematocrit (Hct) as well as in red blood cell count (RBC), red blood cell distribution width (RDW), mean corpuscular volume (MCV), and mean corpuscular hemoglobin (MCH) were observed in the RAPA-treatment group. None of these changes manifested clinically significant effects during the short duration of this study. Similarly, no changes were noted in any other clinical laboratory, cognitive, physical performance, or self-perceived health status measure over the study period. Immune parameters were largely unchanged as well, possibly due to the advanced ages of the cohort (70-93 years; mean age 80.5). RAPA-associated increases in a myeloid cell subset and in TREGS were detected, but changes in most other PBMC cell subsets were not statistically significant. Importantly, the OGTTs revealed no RAPA-induced change in blood glucose concentration, insulin secretion, and insulin sensitivity. Thus, based on the results of our pilot study, it appears that short-term RAPA treatment can be used safely in older persons who are otherwise healthy; a trial with a larger sample size and longer treatment duration is warranted.
Collapse
Affiliation(s)
- Ellen Kraig
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; Department of Cell Systems and Anatomy, The University of Texas Health Science Center, San Antonio, USA.
| | - Leslie A Linehan
- Department of Cell Systems and Anatomy, The University of Texas Health Science Center, San Antonio, USA
| | - Hanyu Liang
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA
| | - Terry Q Romo
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; GRECC, South Texas Veterans Health Care System, The University of Texas Health Science Center, San Antonio, USA
| | - Qianqian Liu
- Department of Epidemiology and Biostatistics, The University of Texas Health Science Center, San Antonio, USA
| | - Yubo Wu
- Department of Medicine, The University of Texas Health Science Center, San Antonio, USA
| | - Adriana D Benavides
- Department of Microbiology, Immunology, and Molecular Genetics, The University of Texas Health Science Center, San Antonio, USA
| | - Tyler J Curiel
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; Department of Medicine, The University of Texas Health Science Center, San Antonio, USA
| | - Martin A Javors
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; Department of Psychiatry, The University of Texas Health Science Center, San Antonio, USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; Department of Medicine, The University of Texas Health Science Center, San Antonio, USA; GRECC, South Texas Veterans Health Care System, The University of Texas Health Science Center, San Antonio, USA
| | - Laura Chiodo
- GRECC, South Texas Veterans Health Care System, The University of Texas Health Science Center, San Antonio, USA
| | - Wouter Koek
- Department of Cell Systems and Anatomy, The University of Texas Health Science Center, San Antonio, USA; Department of Psychiatry, The University of Texas Health Science Center, San Antonio, USA
| | - Jonathan A L Gelfond
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; Department of Epidemiology and Biostatistics, The University of Texas Health Science Center, San Antonio, USA
| | - Dean L Kellogg
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, USA; Department of Medicine, The University of Texas Health Science Center, San Antonio, USA; GRECC, South Texas Veterans Health Care System, The University of Texas Health Science Center, San Antonio, USA
| |
Collapse
|
64
|
Hui DS, Lee N, Chan PK, Beigel JH. The role of adjuvant immunomodulatory agents for treatment of severe influenza. Antiviral Res 2018; 150:202-216. [PMID: 29325970 PMCID: PMC5801167 DOI: 10.1016/j.antiviral.2018.01.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 12/31/2017] [Accepted: 01/05/2018] [Indexed: 12/18/2022]
Abstract
A severe inflammatory immune response with hypercytokinemia occurs in patients hospitalized with severe influenza, such as avian influenza A(H5N1), A(H7N9), and seasonal A(H1N1)pdm09 virus infections. The role of immunomodulatory therapy is unclear as there have been limited published data based on randomized controlled trials (RCTs). Passive immunotherapy such as convalescent plasma and hyperimmune globulin have some studies demonstrating benefit when administered as an adjunctive therapy for severe influenza. Triple combination of oseltamivir, clarithromycin, and naproxen for severe influenza has one study supporting its use, and confirmatory studies would be of great interest. Likewise, confirmatory studies of sirolimus without concomitant corticosteroid therapy should be explored as a research priority. Other agents with potential immunomodulating effects, including non-immune intravenous immunoglobulin, N-acetylcysteine, acute use of statins, macrolides, pamidronate, nitazoxanide, chloroquine, antiC5a antibody, interferons, human mesenchymal stromal cells, mycophenolic acid, peroxisome proliferator-activated receptors agonists, non-steroidal anti-inflammatory agents, mesalazine, herbal medicine, and the role of plasmapheresis and hemoperfusion as rescue therapy have supportive preclinical or observational clinical data, and deserve more investigation preferably by RCTs. Systemic corticosteroids administered in high dose may increase the risk of mortality and morbidity in patients with severe influenza and should not be used, while the clinical utility of low dose systemic corticosteroids requires further investigation.
Collapse
Affiliation(s)
- David S Hui
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Nelson Lee
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong; Division of Infectious Diseases, University of Alberta, Edmonton, Canada
| | - Paul K Chan
- Department of Microbiology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - John H Beigel
- Leidos Biomedical Research Inc, Support to National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
65
|
Pallet N, Fernández-Ramos AA, Loriot MA. Impact of Immunosuppressive Drugs on the Metabolism of T Cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:169-200. [DOI: 10.1016/bs.ircmb.2018.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
66
|
Zhang Q, Chen J, Gao H, Zhang S, Zhao C, Zhou C, Wang C, Li Y, Cai Z, Mou L. Drug repurposing: Ibrutinib exhibits immunosuppressive potential in organ transplantation. Int J Med Sci 2018; 15:1118-1128. [PMID: 30123049 PMCID: PMC6097265 DOI: 10.7150/ijms.24460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/12/2018] [Indexed: 11/05/2022] Open
Abstract
Long-term administration of classic immunosuppressants can induce severe adverse effects. The development of novel immunosuppressants confronts great challenges and opportunities. Ibrutinib, an approved drug for B-cell lineages and chronic graft versus host disease (cGVHD), exhibits immunosuppressive efficacy in autoimmune diseases. Ibrutinib's potential as an immunosuppressant in organ transplantation has not been investigated to date. In a xeno-artery patch model ex vivo, ibrutinib inhibited the proliferation of PBMCs (POD 14-42), mainly CD3+CD4+ and CD3+CD8+ T cells ex vivo. The secretion of cytokines (IL-6, IL-2 and IFN-γ) was suppressed in response to ibrutinib. In allo-skin transplantation models, ibrutinib delayed the rejection of grafted skins. Ibrutinib decreased the amount of T/B cells and lymphocyte infiltration. Altogether, ibrutinib exhibited immunosuppressive potential through cytokine regulation and T cell inhibition ex vivo and in vitro. Repositioning of ibrutinib as an immunosuppressant will greatly facilitate novel immunosuppressant development.
Collapse
Affiliation(s)
- Qing Zhang
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| | - Jicheng Chen
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| | - Hanchao Gao
- Shenzhen Longhua District Central Hospital, Shenzhen, China.,Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| | - Song Zhang
- The Department of Anesthesiology, Weifang Medical University, Weifang, China
| | - Chengjiang Zhao
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China.,Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Cuibing Zhou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| | - Chengjun Wang
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| | - Yang Li
- School of Information Science and Engineering, Shandong Agricultural University, Tai'an, China
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
67
|
Haeri A, Osouli M, Bayat F, Alavi S, Dadashzadeh S. Nanomedicine approaches for sirolimus delivery: a review of pharmaceutical properties and preclinical studies. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1-14. [DOI: 10.1080/21691401.2017.1408123] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Azadeh Haeri
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahraz Osouli
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Bayat
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sonia Alavi
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
68
|
Shihab F, Qazi Y, Mulgaonkar S, McCague K, Patel D, Peddi VR, Shaffer D. Association of Clinical Events With Everolimus Exposure in Kidney Transplant Patients Receiving Low Doses of Tacrolimus. Am J Transplant 2017; 17:2363-2371. [PMID: 28141897 PMCID: PMC5600116 DOI: 10.1111/ajt.14215] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/04/2017] [Accepted: 01/24/2017] [Indexed: 01/25/2023]
Abstract
A key objective in the use of immunosuppression after kidney transplantation is to attain the optimal balance between efficacy and safety. In a phase 3b, multicenter, randomized, open-label, noninferiority study, the incidences of clinical events, renal dysfunction, and adverse events (AEs) were analyzed at 12 months in 309 de novo renal transplant recipients receiving everolimus (EVR), low-dose tacrolimus (LTac), and prednisone. Cox proportional hazard regression modeling was used to estimate the probability of clinical events at specified combinations of time-normalized EVR and Tac trough concentrations. At 12 months, the highest incidence of treated biopsy-proven acute rejection (tBPAR) and graft loss occurred most often in patients with EVR trough concentration <3 ng/mL (64.7% and 10.5%, respectively). At 1 month and 12 months, increasing EVR levels were associated with fewer tBPAR events (both p < 0.0001). Low estimated glomerular filtration rate (eGFR) and decreased eGFR occurred more often in patients with lower EVR and higher Tac levels. AEs were most often observed in patients with EVR levels <3 ng/mL. This study supports maintaining an EVR trough concentration of 3-8 ng/mL, when combined with LTac, to achieve balanced efficacy and safety in renal transplant recipients. TRIAL REGISTRATION NCT01025817.
Collapse
Affiliation(s)
| | - Y. Qazi
- University of Southern CaliforniaLos AngelesCA
| | | | - K. McCague
- Novartis Pharmaceuticals CorporationEast HanoverNJ
| | - D. Patel
- Novartis Pharmaceuticals CorporationEast HanoverNJ
| | - V. R. Peddi
- California Pacific Medical CenterSan FranciscoCA
| | - D. Shaffer
- Vanderbilt University Medical CenterNashvilleTN
| |
Collapse
|
69
|
Daniel JM, Dutzmann J, Brunsch H, Bauersachs J, Braun-Dullaeus R, Sedding DG. Systemic application of sirolimus prevents neointima formation not via a direct anti-proliferative effect but via its anti-inflammatory properties. Int J Cardiol 2017; 238:79-91. [DOI: 10.1016/j.ijcard.2017.03.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/06/2017] [Accepted: 03/12/2017] [Indexed: 01/15/2023]
|
70
|
van Gelder T, Fischer L, Shihab F, Shipkova M. Optimizing everolimus exposure when combined with calcineurin inhibitors in solid organ transplantation. Transplant Rev (Orlando) 2017; 31:151-157. [DOI: 10.1016/j.trre.2017.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/13/2017] [Accepted: 02/21/2017] [Indexed: 01/05/2023]
|
71
|
Mišľanová C, Príbojová J, Valachovičová M, Žilinská Z. Determination of Immunosuppressive Pharmaceuticals in Whole Blood Following Kidney Transplantation by High-performance Liquid Chromatography–Tandem Mass Spectrometry. ANAL LETT 2017. [DOI: 10.1080/00032719.2017.1297452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Csilla Mišľanová
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Jana Príbojová
- Faculty of Nursing and Professional Health Studies, Slovak Medical University, Bratislava, Slovakia
| | - Martina Valachovičová
- Faculty of Nursing and Professional Health Studies, Slovak Medical University, Bratislava, Slovakia
| | - Zuzana Žilinská
- Department of Urology with Kidney Transplant Centre, University Hospital, Bratislava, Slovakia
| |
Collapse
|
72
|
Mizuno T, Emoto C, Fukuda T, Hammill AM, Adams DM, Vinks AA. Model-based precision dosing of sirolimus in pediatric patients with vascular anomalies. Eur J Pharm Sci 2017; 109S:S124-S131. [PMID: 28526601 DOI: 10.1016/j.ejps.2017.05.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
Abstract
Sirolimus is the first drug to show efficacy in the treatment of patients with complicated vascular anomalies. The current study expands on the evolution of a PK model-based strategy for the precision dosing of sirolimus as part of prospective concentration controlled clinical trials in pediatric patients with vascular anomalies. Twelve month follow up data collected from 52 pediatric patients participating in the Phase 2 clinical trial were analyzed. Target attainment across the age range of 3weeks to 18years after 2-3months of therapy was 94% (49 out of 52 patients). The mean sirolimus dose to achieve the target of ~10ng/mL for patients older than 2years was 1.8mg/m2 twice daily (range 0.8-2.9), while it was 0.7 to 1.6mg/m2 twice daily for patients 3weeks of age to 2years. A total of 676 blood concentration data were used for the population PK analysis by nonlinear mixed effect modeling using NONMEM. The final model included a maturation function for sirolimus clearance and allometrically scaled body weight to account for size differences. The mean allometrically scaled sirolimus clearance estimates increased from 3.9 to 17.0L/h per 70kg with age from shortly after the birth to 2years of age while the mean estimate for patients older than 2years was 18.5L/h per 70kg. The developed model based dosing strategy provides a foundation for ongoing efforts to define the sirolimus exposure-response and clinical outcome relationships across the pediatric age spectrum from birth to adolescence.
Collapse
Affiliation(s)
- Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chie Emoto
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Tsuyoshi Fukuda
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Adrienne M Hammill
- Division of Hematology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Denise M Adams
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
73
|
Rodríguez-Jiménez C, García-Saiz M, Pérez-Tamajón L, Salido E, Torres A. Influence of genetic polymorphisms of CYP3A5 and ABCB1 on sirolimus pharmacokinetics, patient and graft survival and other clinical outcomes in renal transplant. Drug Metab Pers Ther 2017; 32:49-58. [PMID: 28245187 DOI: 10.1515/dmpt-2016-0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/08/2017] [Indexed: 06/06/2023]
Abstract
BACKGROUND In transplant patients receiving de novo anticalcineurin-free sirolimus (SRL)-based immunosuppression, we determined the influence of cytochrome P450 3A5 (CYP3A5) and ATP-binding cassette, sub-family B (MDR/TAP), member (ABCB1) genotypes on SRL blood levels and medium-term relevant clinical outcomes, in order to improve effectiveness of immunosuppression strategies when anti-mammalian target of rapamycin (anti-mTOR) inhibitor is indicated for clinical reasons. METHODS Forty-eight renal transplant recipients (suffered 48% diabetes mellitus, 91% hypertension, and 47% dyslipidemia) were genotyped for CYP3A5 (6986A>G) and ABCB1 (3435C>T) polymorphisms by polymerase chain reaction-restriction fragment length polymorphism. Sirolimus blood levels were determined using microparticle enzyme immunoassay technique. Relationships between genotypes and pharmacokinetics, graft function, and patient-graft survival were determined by univariate analysis. RESULTS CYP3A5*1/*3 showed lower SRL levels than CYP3A5*3/*3 (4.13±1.54 vs. 8.49±4.18 ng/mL; p=0.003) and level/dose ratio (LDR) (92.74±37.47 vs. 178.62±116.45; p=0.019) in early post-transplant period. In ABCB1 polymorphisms, CT genotypes showed higher SRL levels than CC and TT (8.93±2.22 vs. 7.28±2.47 vs. 7.35±1.15 ng/mL; p=0.038) in the late period; LDR in CC and CT were 171.29±36.24 vs. 335.66±138.71 (p=0.003), despite receiving lower doses (p=0.018). Acute rejection rate was 14% vs. 42% for *3/*3 and 14% (TT), 48% (CT), and 31% (CC). Median patient survival was 45 months, significantly lower than that of *3/*3 patients (69 months). Death-censored graft survival during 5-year follow-up was similar for both CYP3A5 genotypes and significantly lower in TT than CT and CC groups, without survival differences. CONCLUSIONS CYP3A5 and ABCB1 polymorphisms influenced SRL levels; preliminary data suggest this may affect patient and graft survival. Genotyping renal transplant patients could help select candidates for SRL (genotype*3/*3 for CYP3A5 and CT for ABCB1), when anti-mTOR immunosuppression is indicated.
Collapse
|
74
|
Girdlestone J. Mesenchymal stromal cells with enhanced therapeutic properties. Immunotherapy 2016; 8:1405-1416. [DOI: 10.2217/imt-2016-0098] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
75
|
Lutz M, Mielke S. New perspectives on the use of mTOR inhibitors in allogeneic haematopoietic stem cell transplantation and graft-versus-host disease. Br J Clin Pharmacol 2016; 82:1171-1179. [PMID: 27245261 PMCID: PMC5061796 DOI: 10.1111/bcp.13022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022] Open
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) has been exploited largely both in solid tumour oncology and solid organ transplantation. More recently mTOR inhibitors such as sirolimus and everolimus have been introduced to the field of allogeneic haematopoietic stem cell transplantation where their unique combination of immunosuppressive purposes offering reduced nephrotoxicity and potential antimalignant effects reflect a unique drug profile that has led to their widespread use in both prophylaxis and therapy of graft-versus-host disease (GVHD). On the other hand haematological insufficiency, infectious complications as well as vasculopathies, have been frequently reported as limiting toxicities. Here, we review both the retrospective and prospective experience available to date and stress the need for prospective registration trials to reduce off label use and improve patient safety by optimizing dosing and enhancing pharmacovigilance. Furthermore, we speculate on the future role of mTOR inhibitors in allogeneic haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Mathias Lutz
- Department of Medicine A, Münster University Medical Center, Münster, Germany
| | - Stephan Mielke
- Department of Internal Medicine II, Würzburg University Medical Center, Würzburg, Germany.
| |
Collapse
|
76
|
Malvezzi P, Jouve T, Rostaing L. Costimulation Blockade in Kidney Transplantation: An Update. Transplantation 2016; 100:2315-2323. [PMID: 27472094 PMCID: PMC5084636 DOI: 10.1097/tp.0000000000001344] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022]
Abstract
In the setting of solid-organ transplantation, calcineurin inhibitor (CNI)-based therapy remains the cornerstone of immunosuppression. However, long-term use of CNIs is associated with some degree of nephrotoxicity. This has led to exploring the blockade of some costimulation pathways as an efficient immunosuppressive tool instead of using CNIs. The only agent already in clinical use and approved by the health authorities for kidney transplant patients is belatacept (Nulojix), a fusion protein that interferes with cytotoxic T lymphocyte-associated protein 4. Belatacept has been demonstrated to be as efficient as cyclosporine-based immunosuppression and is associated with significantly better renal function, that is, no nephrotoxicity. However, in the immediate posttransplant period, significantly more mild/moderate episodes of acute rejection have been reported, favored by the fact that cytotoxic T lymphocyte-associated protein pathway has an inhibitory effect on the alloimmune response; thereby its inhibition is detrimental in this regard. This has led to the development of antibodies that target CD28. The most advanced is FR104, it has shown promise in nonhuman primate models of autoimmune diseases and allotransplantation. In addition, research into blocking the CD40-CD154 pathway is underway. A phase II study testing ASK1240, that is, anti-CD40 antibody has been completed, and the results are pending.
Collapse
Affiliation(s)
- Paolo Malvezzi
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
| | - Thomas Jouve
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
- Université Joseph Fourier, Grenoble, France
| | - Lionel Rostaing
- Clinique Universitaire de Néphrologie, Unité de Transplantation Rénale, CHU Grenoble, France
- UniversitéToulouse III Paul Sabatier, Toulouse, France
- INSERM U563, IFR-BMT, CHU Purpan, Toulouse, France
| |
Collapse
|
77
|
Fernandes-Silva G, Ivani de Paula M, Rangel ÉB. mTOR inhibitors in pancreas transplant: adverse effects and drug-drug interactions. Expert Opin Drug Metab Toxicol 2016; 13:367-385. [DOI: 10.1080/17425255.2017.1239708] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Gabriel Fernandes-Silva
- Universidade Federal de São Paulo/Hospital do Rim e Hipertensão, Nephrology Department, São Paulo, SP, Brazil
| | - Mayara Ivani de Paula
- Universidade Federal de São Paulo/Hospital do Rim e Hipertensão, Nephrology Department, São Paulo, SP, Brazil
| | - Érika B. Rangel
- Universidade Federal de São Paulo/Hospital do Rim e Hipertensão, Nephrology Department, São Paulo, SP, Brazil
- Hospital Israelita Albert Einstein, Instituto Israelita de Ensino e Pesquisa, São Paulo, SP, Brazil
| |
Collapse
|
78
|
Vanhove T, Remijsen Q, Kuypers D, Gillard P. Drug-drug interactions between immunosuppressants and antidiabetic drugs in the treatment of post-transplant diabetes mellitus. Transplant Rev (Orlando) 2016; 31:69-77. [PMID: 27665059 DOI: 10.1016/j.trre.2016.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 02/06/2023]
Abstract
Post-transplant diabetes mellitus is a frequent complication of solid organ transplantation that generally requires treatment with lifestyle interventions and antidiabetic medication. A number of demonstrated and potential pharmacokinetic drug-drug interactions (DDIs) exist between commonly used immunosuppressants and antidiabetic drugs, which are comprehensively summarized in this review. Cyclosporine (CsA) itself inhibits the cytochrome P450 (CYP) 3A4 enzyme and a variety of drug transporters. As a result, it increases exposure to repaglinide and sitagliptin, will likely increase the exposure to nateglinide, glyburide, saxagliptin, vildagliptin and alogliptin, and could theoretically increase the exposure to gliquidone and several sodium-glucose transporter (SGLT)-2 inhibitors. Currently available data, although limited, suggest that these increases are modest and, particularly with regard to gliptins and SGLT-2 inhibitors, unlikely to result in hypoglycemia. The interaction with repaglinide is more pronounced but does not preclude concomitant use if repaglinide dose is gradually titrated. Mycophenolate mofetil and azathioprine do not engage in DDIs with any antidiabetic drug. Although calcineurin inhibitors (CNIs) and mammalian target of rapamycin inhibitors (mTORi) are intrinsically prone to DDIs, their disposition is not influenced by metformin, pioglitazone, sulfonylureas (except possibly glyburide) or insulin. An effect of gliptins on the disposition of CNIs and mTORi is unlikely, but has not been definitively ruled out. Based on their disposition profiles, glyburide and canagliflozin could affect CNI and mTORi disposition although this requires further study. Finally, delayed gastric emptying as a result of glucagon-like peptide-1 agonists seems to have a limited, but not necessarily negligible effect on CNI disposition.
Collapse
Affiliation(s)
- Thomas Vanhove
- Department of Microbiology and Immunology, KU Leuven - University of Leuven, and Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| | - Quinten Remijsen
- Department of Medical Affairs, AstraZeneca BeLux, Uccle, Belgium
| | - Dirk Kuypers
- Department of Microbiology and Immunology, KU Leuven - University of Leuven, and Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Gillard
- Laboratory and Clinic of Experimental Medicine and Endocrinology, KU Leuven - University of Leuven, and Department of Endocrinology, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
79
|
The role of regulatory B cells (Bregs) in the Tregs-amplifying effect of Sirolimus. Int Immunopharmacol 2016; 38:90-6. [DOI: 10.1016/j.intimp.2016.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/03/2023]
|
80
|
Abstract
This chapter reviews the neurologic complications of medications administered in the hospital setting, by class, introducing both common and less common side effects. Detail is devoted to the interaction between pain, analgesia, sedation, and their residual consequences. Antimicrobials are given in nearly every hospital setting, and we review their capacity to produce neurologic sequelae with special devotion to cefepime and the antiviral treatment of human immunodeficiency virus. The management of hemorrhagic stroke has become more complex with the introduction of novel oral anticoagulants, and we provide an update on what is known about reversal of the new oral anticoagulants. Both central and peripheral nervous system complications of immunosuppressants and chemotherapies are reviewed. Because diagnosis is generally based on clinical acumen, alone, neurotoxic syndromes resulting from psychotropic medications may be easily overlooked until severe dysautonomia develops. We include a practical approach to the diagnosis of serotonin syndrome and neuroleptic malignant syndrome.
Collapse
Affiliation(s)
- Elliot T Dawson
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Sara E Hocker
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neurology, Division of Critical Care Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
81
|
Gualdoni GA, Mayer KA, Göschl L, Boucheron N, Ellmeier W, Zlabinger GJ. The AMP analog AICAR modulates the Treg/Th17 axis through enhancement of fatty acid oxidation. FASEB J 2016; 30:3800-3809. [PMID: 27492924 DOI: 10.1096/fj.201600522r] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/27/2016] [Indexed: 02/06/2023]
Abstract
T cells must tightly regulate their metabolic processes to cope with varying bioenergetic demands depending on their state of differentiation. The metabolic sensor AMPK is activated in states of low energy supply and modulates cellular metabolism toward a catabolic state. Although this enzyme is known to be particularly active in regulatory T (Treg) cells, its impact on T helper (Th)-cell differentiation is poorly understood. We investigated the impact of several AMPK activators on Treg-cell differentiation and found that the direct activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide), but not the indirect activators metformin and 2-deoxyglucose, strongly enhanced Treg-cell induction by specifically enhancing Treg-cell expansion. Conversely, Th17 generation was impaired by the agent. Further investigation of the metabolic background of our observations revealed that AICAR enhanced both cellular mitochondrogenesis and fatty acid uptake. Consistently, increased Treg induction was entirely reversible on inhibition of fatty acid oxidation, thus confirming the dependence of AICAR's effects on metabolic pathways alterations. Translating our findings to an in vivo model, we found that the substance enhanced Treg cell generation on IL-2 complex-induced immune stimulation. We provide a previously unrecognized insight into the delicate interplay between immune cell function and metabolism and delineate a potential novel strategy for metabolism-targeting immunotherapy.-Gualdoni, G. A., Mayer, K. A., Göschl, L., Boucheron, N., Ellmeier, W., Zlabinger, G. J. The AMP analog AICAR modulates the Treg/Th17 axis through enhancement of fatty acid oxidation.
Collapse
Affiliation(s)
- Guido A Gualdoni
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Austria; and
| | - Katharina A Mayer
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Austria; and
| | - Lisa Göschl
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Austria; and.,Division of Rheumatology, Department of Internal Medicine 3, Medical University of Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Austria; and
| | - Wilfried Ellmeier
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Austria; and
| | - Gerhard J Zlabinger
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Austria; and
| |
Collapse
|
82
|
Capron A, Haufroid V, Wallemacq P. Intra-cellular immunosuppressive drugs monitoring: A step forward towards better therapeutic efficacy after organ transplantation? Pharmacol Res 2016; 111:610-618. [PMID: 27468645 DOI: 10.1016/j.phrs.2016.07.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 12/15/2022]
Abstract
Immunosuppressive drugs (IS) used in solid organ transplantation are critical dose drugs with high intra- and inter-subject variability. Therefore, IS therapeutic drug monitoring (TDM), mainly as trough levels analysis, is a major support to patient management, mandatory to optimize clinical outcome. Even though transplant patients undoubtedly benefited by this pre-dose (C0) monitoring, the relationship between these C0 concentrations and the incidence of graft rejections remains hardly predictable. Identification and validation of additional biomarkers of efficacy are therefore very much needed. As the main IS effects are mediated through the inhibition of lymphocyte proliferation pathways, direct drug quantification within this target compartment would appear meaningful, providing hopefully more consistent information on drug efficacy. Due to the analytical performances improvement, these intracellular concentrations became accessible for comprehensive studies regarding clinical benefit of intracellular IS TDM after solid organ transplantation. Over the last ten years, number of studies investigated the potential relationship between IS blood and intracellular pharmacokinetics, genetic variability, and clinical efficacy after solid organ transplantation. A recent literature review suggests that calcineurin inhibitors (tacrolimus and cyclosporine) intracellular concentrations appear more closely related to drug efficacy than blood levels. This closer association has however not been described for the m-TOR inhibitors (sirolimus, everolimus) and the antimetabolite (mycophenolic acid). Additional larger and multicenter clinical trials are needed to confirm these observations.
Collapse
Affiliation(s)
- A Capron
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium; Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium.
| | - V Haufroid
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium; Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - P Wallemacq
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium; Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
83
|
Li D, Wang C, Yao Y, Chen L, Liu G, Zhang R, Liu Q, Shi FD, Hao J. mTORC1 pathway disruption ameliorates brain inflammation following stroke via a shift in microglia phenotype from M1 type to M2 type. FASEB J 2016; 30:3388-3399. [PMID: 27342766 DOI: 10.1096/fj.201600495r] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/14/2016] [Indexed: 11/11/2022]
Abstract
Inflammatory factors secreted by microglia play an important role in focal ischemic stroke. The mammalian target of rapamycin (mTOR) pathway is a known regulator of immune responses, but the role that mTORC1 signaling plays in poststroke neuroinflammation is not clear. To explore the relationship between microglial action in the mTORC1 pathway and the impact on stroke, we administered the mTORC1 inhibitors sirolimus and everolimus to mice. Presumably, disrupting the mTORC1 pathway after focal ischemic stroke should clarify the subsequent activity of microglia. For that purpose, we generated mice deficient in the regulatory associated protein of mTOR (Raptor) in microglia, whose mTORC1 signaling was blocked, by crossing Raptor loxed (Raptorflox/flox) mice with CX3CR1CreER mice, which express Cre recombinase under the control of the CX3C chemokine receptor 1 promoter. mTORC1 blockade reduced lesion size, improved motor function, dramatically decreased production of proinflammatory cytokines and chemokines, and reduced the number of M1 type microglia. Thus, mTORC1 blockade apparently attenuated behavioral deficits and poststroke inflammation after middle cerebral artery occlusion by preventing microglia polarization toward the M1 type.-Li, D., Wang, C., Yao, Y., Chen, L., Liu, G., Zhang, R., Liu, Q., Shi, F.-D., Hao, J. mTORC1 pathway disruption ameliorates brain inflammation following stroke via a shift in microglia phenotype from M1 type to M2 type.
Collapse
Affiliation(s)
- Daojing Li
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunjiong Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yang Yao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Chen
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Guiyou Liu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Rongxin Zhang
- Department of Immunology, Tianjin Medical University, Tianjin, China; and
| | - Qiang Liu
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Fu-Dong Shi
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Junwei Hao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China;
| |
Collapse
|
84
|
Kajiwara M, Masuda S. Role of mTOR Inhibitors in Kidney Disease. Int J Mol Sci 2016; 17:ijms17060975. [PMID: 27338360 PMCID: PMC4926507 DOI: 10.3390/ijms17060975] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/13/2016] [Accepted: 06/16/2016] [Indexed: 02/06/2023] Open
Abstract
The first compound that inhibited the mammalian target of rapamycin (mTOR), sirolimus (rapamycin) was discovered in the 1970s as a soil bacterium metabolite collected on Easter Island (Rapa Nui). Because sirolimus showed antiproliferative activity, researchers investigated its molecular target and identified the TOR1 and TOR2. The mTOR consists of mTOR complex 1 (mTORC1) and mTORC2. Rapalogues including sirolimus, everolimus, and temsirolimus exert their effect mainly on mTORC1, whereas their inhibitory effect on mTORC2 is mild. To obtain compounds with more potent antiproliferative effects, ATP-competitive inhibitors of mTOR targeting both mTORC1 and mTORC2 have been developed and tested in clinical trials as anticancer drugs. Currently, mTOR inhibitors are used as anticancer drugs against several solid tumors, and immunosuppressive agents for transplantation of various organs. This review discusses the role of mTOR inhibitors in renal disease with a particular focus on renal cancer, diabetic nephropathy, and kidney transplantation.
Collapse
Affiliation(s)
- Moto Kajiwara
- Department of Pharmacy, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Satohiro Masuda
- Department of Pharmacy, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
85
|
Pharmacogenetic Biomarkers Predictive of the Pharmacokinetics and Pharmacodynamics of Immunosuppressive Drugs. Ther Drug Monit 2016; 38 Suppl 1:S57-69. [DOI: 10.1097/ftd.0000000000000255] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
86
|
Waldner M, Fantus D, Solari M, Thomson AW. New perspectives on mTOR inhibitors (rapamycin, rapalogs and TORKinibs) in transplantation. Br J Clin Pharmacol 2016; 82:1158-1170. [PMID: 26810941 DOI: 10.1111/bcp.12893] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 02/06/2023] Open
Abstract
The macrolide rapamycin and its analogues (rapalogs) constitute the first generation of mammalian target of rapamycin (mTOR) inhibitors. Since the introduction of rapamycin as an immunosuppressant, there has been extensive progress in understanding its complex mechanisms of action. New insights into the function of mTOR in different immune cell types, vascular endothelial cells and neoplastic cells have opened new opportunities and challenges regarding mTOR as a pharmacological target. Currently, the two known mTOR complexes, mTOR complex (mTORC) 1 and mTORC2, are the subject of intense investigation, and the introduction of second-generation dual mTORC kinase inhibitors (TORKinibs) and gene knockout mice is helping to uncover the distinct roles of these complexes in different cell types. While the pharmacological profiling of rapalogs is advanced, much less is known about the properties of TORKinibs. A potential benefit of mTOR inhibition in transplantation is improved protection against transplant-associated viral infections compared with standard calcineurin inhibitor-based immunosuppression. Preclinical and clinical data also underscore the potentially favourable antitumour effects of mTOR inhibitors in regard to transplant-associated malignancies and as a novel treatment option for various other cancers. Many aspects of the mechanisms of action of mTOR inhibitors and their clinical implications remain unknown. In this brief review we discuss new findings and perspectives of mTOR inhibitors in transplantation.
Collapse
Affiliation(s)
- Matthias Waldner
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Plastic Surgery, University of Zurich, Zurich, Switzerland
| | - Daniel Fantus
- Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mario Solari
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Angus W Thomson
- Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
87
|
Drion CM, Borm LE, Kooijman L, Aronica E, Wadman WJ, Hartog AF, van Vliet EA, Gorter JA. Effects of rapamycin and curcumin treatment on the development of epilepsy after electrically induced status epilepticus in rats. Epilepsia 2016; 57:688-97. [DOI: 10.1111/epi.13345] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2016] [Indexed: 01/13/2023]
Affiliation(s)
- Cato M. Drion
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Lars E. Borm
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Lieneke Kooijman
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Eleonora Aronica
- Department (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Epilepsy Institute in the Netherlands; Heemstede The Netherlands
| | - Wytse J. Wadman
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Aloysius F. Hartog
- Van‘t Hoff Institute for Molecular Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Erwin A. van Vliet
- Department (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jan A. Gorter
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
88
|
Matz M, Fabritius K, Liu J, Lorkowski C, Brakemeier S, Unterwalder N, Dürr M, Mashreghi MF, Neumayer HH, Budde K. Conversion to Belatacept based regimen does not change T-cell phenotype and function in renal transplantation. Transpl Immunol 2015; 33:176-84. [PMID: 26478531 DOI: 10.1016/j.trim.2015.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/08/2015] [Accepted: 10/13/2015] [Indexed: 11/19/2022]
Abstract
Belatacept offers a new option for renal allograft recipients who are suffering from side effects of calcineurin inhibitors or mTOR inhibitors,which may result in renal and extrarenal benefits.We prospectively performed flow cytometric immunophenotyping with a T-cell panel. In total we were able to fully investigate the immunophenotypic change in 8 patients before and after conversion from calcineurin inhibitor (n = 5) or mTOR inhibitor (n=2) to Belatacept or additional administration (n=1). Cells were analysed pre conversion, 1 month, 3 months, 6 months and 12 months after first Belatacept administration. The percentage of central memory, naïve, effector memory and terminally differentiated effector memory CD4+ and CD4− T-cells was determined. CD28, CD25 and CD69 expression on CD4+ and CD4− T-cells was measured ex vivo and also after 3 days of mitogen stimulation. Intracellular cytokines IFNgamma and IL-2 were measured after polyclonal cellular stimulation. The expression of activation markers and intracellular cytokines as well as the percentage of T-cell subsets did not change significantly during the observation period compared to the time-point pre conversion. Therefore the conversion of calcineurin inhibitor or mTOR inhibitor to Belatacept seems to have no obvious impact on the immunophenotype of T-cells in patients after kidney transplantation.
Collapse
Affiliation(s)
- Mareen Matz
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany.
| | - Katharina Fabritius
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Juan Liu
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Christine Lorkowski
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Brakemeier
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Nadine Unterwalder
- Labor Berlin GmbH, Immunology Department, Universitätsmedizin Charité Campus Virchow, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Michael Dürr
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | | | - Hans-H Neumayer
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Klemens Budde
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|