1
|
Wang P, Okada-Rising S, Scultetus AH, Bailey ZS. The Relevance and Implications of Monoclonal Antibody Therapies on Traumatic Brain Injury Pathologies. Biomedicines 2024; 12:2698. [PMID: 39767605 PMCID: PMC11672875 DOI: 10.3390/biomedicines12122698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Traumatic brain injury (TBI) is a global public health concern. It remains one of the leading causes of morbidity and mortality. TBI pathology involves complex secondary injury cascades that are associated with cellular and molecular dysfunction, including oxidative stress, coagulopathy, neuroinflammation, neurodegeneration, neurotoxicity, and blood-brain barrier (BBB) dysfunction, among others. These pathological processes manifest as a diverse array of clinical impairments. They serve as targets for potential therapeutic intervention not only in TBI but also in other diseases. Monoclonal antibodies (mAbs) have been used as key therapeutic agents targeting these mechanisms for the treatment of diverse diseases, including neurological diseases such as Alzheimer's disease (AD). MAb therapies provide a tool to block disease pathways with target specificity that may be capable of mitigating the secondary injury cascades following TBI. This article reviews the pathophysiology of TBI and the molecular mechanisms of action of mAbs that target these shared pathological pathways in a wide range of diseases. Publicly available databases for various applications of mAb therapy were searched and further classified to assess relevance to TBI pathology and evaluate current stages of development. The authors intend for this review to highlight the potential impact of current mAb technology within pathological TBI processes.
Collapse
Affiliation(s)
- Ping Wang
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (S.O.-R.); (A.H.S.); (Z.S.B.)
| | | | | | | |
Collapse
|
2
|
Wang P, Yang X, Yang F, Cardiff K, Houchins M, Carballo N, Shear DA, Scultetus AH, Bailey ZS. Intravenous Administration of Anti-CD47 Antibody Augments Hematoma Clearance, Mitigates Acute Neuropathology, and Improves Cognitive Function in a Rat Model of Penetrating Traumatic Brain Injury. J Neurotrauma 2024; 41:2413-2427. [PMID: 38874230 DOI: 10.1089/neu.2024.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Abstract
Traumatic brain injury (TBI)-induced intracerebral hematoma is a major driver of secondary injury pathology such as neuroinflammation, cerebral edema, neurotoxicity, and blood-brain barrier dysfunction, which contribute to neuronal loss, motor deficits, and cognitive impairment. Cluster of differentiation 47 (CD47) is an antiphagocytic cell surface protein inhibiting hematoma clearance. This study was designed to evaluate the safety and efficacy of blockade of CD47 via intravenous (i.v.) administration of anti-CD47 antibodies following penetrating ballistic-like brain injury (PBBI) with significant traumatic intracerebral hemorrhage (tICH). The pharmacokinetic (PK) profile of the anti-CD47 antibody elicited that antibody concentration decayed over 7 days post-administration. Blood tests and necropsy analysis indicated no severe adverse events following treatment. Cerebral hemoglobin levels were significantly increased after injury, however, anti-CD47 antibody administration at 0.1 mg/kg resulted in a significant reduction in cerebral hemoglobin levels at 72 h post-administration, indicating augmentation of hematoma clearance. Immunohistochemistry assessment of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (IBA1) demonstrated a significant reduction of GFAP levels in the lesion core and peri-lesional area. Based on these analyses, the optimal dose was identified as 0.1 mg/kg. Lesion volume showed a reduction following treatment. Rotarod testing revealed significant motor deficits in all injured groups but no significant therapeutic benefits. Spatial learning performance revealed significant deficits in all injured groups, which were significantly improved by the last testing day. Anti-CD47 antibody treated rats showed significantly improved attention deficits, but not retention scores. These results provide preliminary evidence that blockade of CD47 using i.v. administration of anti-CD47 antibodies may serve as a potential therapeutic for TBI with ICH.
Collapse
Affiliation(s)
- Ping Wang
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Xiaofang Yang
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Fangzhou Yang
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Katherine Cardiff
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Melonie Houchins
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Noemy Carballo
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Deborah A Shear
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Anke H Scultetus
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Zachary S Bailey
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| |
Collapse
|
3
|
Banderwal R, Kadian M, Garg S, Kumar A. 'Comprehensive review of emerging drug targets in traumatic brain injury (TBI): challenges and future scope. Inflammopharmacology 2024:10.1007/s10787-024-01524-w. [PMID: 39023681 DOI: 10.1007/s10787-024-01524-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Traumatic brain injury (TBI) is a complex brain problem that causes significant morbidity and mortality among people of all age groups. The complex pathophysiology, varied symptoms, and inadequate treatment further precipitate the problem. Further, TBI produces several psychiatric problems and other related complications in post-TBI survival patients, which are often treated symptomatically or inadequately. Several approaches, including neuroprotective agents targeting several pathways of oxidative stress, neuroinflammation, cytokines, immune system GABA, glutamatergic, microglia, and astrocytes, are being tried by researchers to develop effective treatments or magic bullets to manage the condition effectively. The problem of TBI is therefore treated as a challenge among pharmaceutical scientists or researchers to develop drugs for the effective management of this problem. The goal of the present comprehensive review is to provide an overview of the several pharmacological targets, processes, and cellular pathways that researchers are focusing on, along with an update on their current state.
Collapse
Affiliation(s)
- Rittu Banderwal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Sukant Garg
- Department of General Pathology, Dr HS Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
4
|
Kanji S, Williamson D, Hartwick M. Potential pharmacological confounders in the setting of death determined by neurologic criteria: a narrative review. Can J Anaesth 2023; 70:713-723. [PMID: 37131030 PMCID: PMC10202973 DOI: 10.1007/s12630-023-02415-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 05/04/2023] Open
Abstract
Guidelines for the determination of death by neurologic criteria (DNC) require an absence of confounding factors if clinical examination alone is to be used. Drugs that depress the central nervous system suppress neurologic responses and spontaneous breathing and must be excluded or reversed prior to proceeding. If these confounding factors cannot be eliminated, ancillary testing is required. These drugs may be present after being administered as part of the treatment of critically ill patients. While measurement of serum drug concentrations can help guide the timing of assessments for DNC, they are not always available or feasible. In this article, we review sedative and opioid drugs that may confound DNC, along with pharmacokinetic factors that govern the duration of drug action. Pharmacokinetic parameters including a context-sensitive half-life of sedatives and opioids are highly variable in critically ill patients because of the multitude of clinical variables and conditions that can affect drug distribution and clearance. Patient-, disease-, and treatment-related factors that influence the distribution and clearance of these drugs are discussed including end organ function, age, obesity, hyperdynamic states, augmented renal clearance, fluid balance, hypothermia, and the role of prolonged drug infusions in critically ill patients. In these contexts, it is often difficult to predict how long after drug discontinuation the confounding effects will take to dissipate. We propose a conservative framework for evaluating when or if DNC can be determined by clinical criteria alone. When pharmacologic confounders cannot be reversed, or doing so is not feasible, ancillary testing to confirm the absence of brain blood flow should be obtained.
Collapse
Affiliation(s)
- Salmaan Kanji
- Department of Pharmacy, The Ottawa Hospital, 501 Smyth Rd, Ottawa, ON, K1H 8L6, Canada.
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | - David Williamson
- Faculté de pharmacie, Université de Montréal, Montreal, QC, Canada
- Pharmacy Department, Hôpital du Sacré-Cœur de Montréal and CIUSSS-Nord-de-l'ile-de-Montreal Research Center, Montreal, QC, Canada
| | - Michael Hartwick
- Department of Pharmacy, The Ottawa Hospital, 501 Smyth Rd, Ottawa, ON, K1H 8L6, Canada
- Department of Critical Care, The Ottawa Hospital, Ottawa, Canada
| |
Collapse
|
5
|
Irzan H, Pozzi M, Chikhladze N, Cebanu S, Tadevosyan A, Calcii C, Tsiskaridze A, Melbourne A, Strazzer S, Modat M, Molteni E. Emerging Treatments for Disorders of Consciousness in Paediatric Age. Brain Sci 2022; 12:198. [PMID: 35203961 PMCID: PMC8870410 DOI: 10.3390/brainsci12020198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
The number of paediatric patients living with a prolonged Disorder of Consciousness (DoC) is growing in high-income countries, thanks to substantial improvement in intensive care. Life expectancy is extending due to the clinical and nursing management achievements of chronic phase needs, including infections. However, long-known pharmacological therapies such as amantadine and zolpidem, as well as novel instrumental approaches using direct current stimulation and, more recently, stem cell transplantation, are applied in the absence of large paediatric clinical trials and rigorous age-balanced and dose-escalated validations. With evidence building up mainly through case reports and observational studies, there is a need for well-designed paediatric clinical trials and specific research on 0-4-year-old children. At such an early age, assessing residual and recovered abilities is most challenging due to the early developmental stage, incompletely learnt motor and cognitive skills, and unreliable communication; treatment options are also less explored in early age. In middle-income countries, the lack of rehabilitation services and professionals focusing on paediatric age hampers the overall good assistance provision. Young and fast-evolving health insurance systems prevent universal access to chronic care in some countries. In low-income countries, rescue networks are often inadequate, and there is a lack of specialised and intensive care, difficulty in providing specific pharmaceuticals, and lower compliance to intensive care hygiene standards. Despite this, paediatric cases with DoC are reported, albeit in fewer numbers than in countries with better-resourced healthcare systems. For patients with a poor prospect of recovery, withdrawal of care is inhomogeneous across countries and still heavily conditioned by treatment costs as well as ethical and cultural factors, rather than reliant on protocols for assessment and standardised treatments. In summary, there is a strong call for multicentric, international, and global health initiatives on DoC to devote resources to the paediatric age, as there is now scope for funders to invest in themes specific to DoC affecting the early years of the life course.
Collapse
Affiliation(s)
- Hassna Irzan
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (H.I.); (A.M.); (M.M.)
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 7JE, UK
| | - Marco Pozzi
- Scientific Institute IRCCS E. Medea, Acquired Brain Injury Unit, 22040 Bosisio Parini, Italy; (M.P.); (S.S.)
| | - Nino Chikhladze
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University, Tbilisi 0179, Georgia; (N.C.); (A.T.)
| | - Serghei Cebanu
- Faculty of Medicine, Nicolae Testemitanu State University of Medicine and Pharmacy, MD-2004 Chišināu, Moldova; (S.C.); (C.C.)
| | - Artashes Tadevosyan
- Department of Public Health and Healthcare Organization, Yerevan State Medical University, Yerevan 0025, Armenia;
| | - Cornelia Calcii
- Faculty of Medicine, Nicolae Testemitanu State University of Medicine and Pharmacy, MD-2004 Chišināu, Moldova; (S.C.); (C.C.)
| | - Alexander Tsiskaridze
- Faculty of Medicine, Ivane Javakhishvili Tbilisi State University, Tbilisi 0179, Georgia; (N.C.); (A.T.)
| | - Andrew Melbourne
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (H.I.); (A.M.); (M.M.)
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 7JE, UK
| | - Sandra Strazzer
- Scientific Institute IRCCS E. Medea, Acquired Brain Injury Unit, 22040 Bosisio Parini, Italy; (M.P.); (S.S.)
- Rehabilitation Service, “Usratuna” Health and Rehabilitation Centre, Juba, South Sudan
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (H.I.); (A.M.); (M.M.)
| | - Erika Molteni
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (H.I.); (A.M.); (M.M.)
| |
Collapse
|
6
|
Janzen RWC, Lambeck J, Niesen WD, Erbguth F. [Irreversible loss of brain function-Part 1: pitfalls in clinical diagnosis]. DER NERVENARZT 2020; 91:743-757. [PMID: 32705299 DOI: 10.1007/s00115-020-00952-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A significant change in the fourth update of the German guidelines on determining brain death is that it includes an explicit profile of requirements on physicians involved in ILBF diagnosis. These requisite qualification criteria have also been formulated due to the fact that, in many hospitals, ILBF diagnosis is only rarely carried out and, as a result, uncertainty frequently arises. Typical difficulties emerge at all stages of ILBF diagnosis, and numerous relevant pitfalls arise that need to be taken into consideration and which might also be relevant in the selection of the method(s) to detect irreversibility. The approaches presented here are suited to achieving a valid result in the evaluation of equivocal ILBF.
Collapse
Affiliation(s)
| | - J Lambeck
- Klinik für Neurologie und Neurophysiologie, Universitätsklinikum Freiburg, Breisacher Str. 64, 79106, Freiburg, Deutschland
| | - W-D Niesen
- Klinik für Neurologie und Neurophysiologie, Universitätsklinikum Freiburg, Breisacher Str. 64, 79106, Freiburg, Deutschland.
| | - F Erbguth
- Klinik für Neurologie, Universitätsklinik der Paracelsus Medizinischen Privatuniversität, Klinikum Nürnberg, Nürnberg, Deutschland
| |
Collapse
|
7
|
Chandrasekar A, Olde Heuvel F, Wepler M, Rehman R, Palmer A, Catanese A, Linkus B, Ludolph A, Boeckers T, Huber-Lang M, Radermacher P, Roselli F. The Neuroprotective Effect of Ethanol Intoxication in Traumatic Brain Injury Is Associated with the Suppression of ErbB Signaling in Parvalbumin-Positive Interneurons. J Neurotrauma 2018; 35:2718-2735. [PMID: 29774782 DOI: 10.1089/neu.2017.5270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ethanol intoxication (EI) is a frequent comorbidity of traumatic brain injury (TBI), but the impact of EI on TBI pathogenic cascades and prognosis is unclear. Although clinical evidence suggests that EI may have neuroprotective effects, experimental support is, to date, inconclusive. We aimed at elucidating the impact of EI on TBI-associated neurological deficits, signaling pathways, and pathogenic cascades in order to identify new modifiers of TBI pathophysiology. We have shown that ethanol administration (5 g/kg) before trauma enhances behavioral recovery in a weight-drop TBI model. Neuronal survival in the injured somatosensory cortex was also enhanced by EI. We have used phospho-receptor tyrosine kinase (RTK) arrays to screen the impact of ethanol on TBI-induced activation of RTK in somatosensory cortex, identifying ErbB2/ErbB3 among the RTKs activated by TBI and suppressed by ethanol. Phosphorylation of ErbB2/3/4 RTKs were upregulated in vGlut2+ excitatory synapses in the injured cortex, including excitatory synapses located on parvalbumin (PV)-positive interneurons. Administration of selective ErbB inhibitors was able to recapitulate, to a significant extent, the neuroprotective effects of ethanol both in sensorimotor performance and structural integrity. Further, suppression of PV interneurons in somatosensory cortex before TBI, by engineered receptors with orthogonal pharmacology, could mimic the beneficial effects of ErbB inhibitors. Thus, we have shown that EI interferes with TBI-induced pathogenic cascades at multiple levels, with one prominent pathway, involving ErbB-dependent modulation of PV interneurons.
Collapse
Affiliation(s)
| | | | - Martin Wepler
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Rida Rehman
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Annette Palmer
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Alberto Catanese
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Birgit Linkus
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Albert Ludolph
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Tobias Boeckers
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Markus Huber-Lang
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Peter Radermacher
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Francesco Roselli
- 1 Department of Neurology, Ulm University , Ulm, Germany .,4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| |
Collapse
|
8
|
Farrokh S, Tahsili-Fahadan P, Ritzl EK, Lewin JJ, Mirski MA. Antiepileptic drugs in critically ill patients. Crit Care 2018; 22:153. [PMID: 29880020 PMCID: PMC5992651 DOI: 10.1186/s13054-018-2066-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The incidence of seizures in intensive care units ranges from 3.3% to 34%. It is therefore often necessary to initiate or continue anticonvulsant drugs in this setting. When a new anticonvulsant is initiated, drug factors, such as onset of action and side effects, and patient factors, such as age, renal, and hepatic function, should be taken into account. It is important to note that the altered physiology of critically ill patients as well as pharmacological and nonpharmacological interventions such as renal replacement therapy, extracorporeal membrane oxygenation, and target temperature management may lead to therapeutic failure or toxicity. This may be even more challenging with the availability of newer antiepileptics where the evidence for their use in critically ill patients is limited. MAIN BODY This article reviews the pharmacokinetics and pharmacodynamics of antiepileptics as well as application of these principles when dosing antiepileptics and monitoring serum levels in critically ill patients. The selection of the most appropriate anticonvulsant to treat seizure and status epileptics as well as the prophylactic use of these agents in this setting are also discussed. Drug-drug interactions and the effect of nonpharmacological interventions such as renal replacement therapy, plasma exchange, and extracorporeal membrane oxygenation on anticonvulsant removal are also included. CONCLUSION Optimal management of antiepileptic drugs in the intensive care unit is challenging given altered physiology, polypharmacy, and nonpharmacological interventions, and requires a multidisciplinary approach where appropriate and timely assessment, diagnosis, treatment, and monitoring plans are in place.
Collapse
Affiliation(s)
- Salia Farrokh
- Department of Pharmacy, The Johns Hopkins Hospital, 600 N. Wolfe Street, Carnegie 180, Baltimore, MD 21287 USA
| | - Pouya Tahsili-Fahadan
- Department of Neurology, The Johns Hopkins Hospital, Baltimore, MD USA
- Department of Medicine, Virginia Commonwealth University School of Medicine, INOVA Campus, Falls Church, VA USA
| | - Eva K. Ritzl
- Department of Neurology, The Johns Hopkins Hospital, Baltimore, MD USA
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Hospital, Baltimore, MD USA
| | - John J. Lewin
- Department of Pharmacy, The Johns Hopkins Hospital, 600 N. Wolfe Street, Carnegie 180, Baltimore, MD 21287 USA
| | - Marek A. Mirski
- Department of Pharmacy, The Johns Hopkins Hospital, 600 N. Wolfe Street, Carnegie 180, Baltimore, MD 21287 USA
| |
Collapse
|
9
|
Alimardani S, Sadrai S, Masoumi HT, Salari P, Najafi A, Eftekhar B, Mojtahedzadeh M. Pharmacokinetic Behavior of Phenytoin in Head Trauma and Cerebrovascular Accident Patients in an Iranian Population. J Res Pharm Pract 2018; 6:217-222. [PMID: 29417081 PMCID: PMC5787907 DOI: 10.4103/jrpp.jrpp_17_58] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Objective: Acute brain injury is one of the leading causes of morbidity and mortality worldwide. Phenytoin has been commonly used as an anticonvulsant agent for the treatment or prophylaxis of seizures following acute brain injury. After a severe head injury, several pharmacokinetic changes occur. The aim of this study is the comparative evaluation of phenytoin serum concentration in patients with traumatic and nontraumatic brain injury (TBI). Methods: This prospective observational study was performed on twenty adult brain injury patients who were admitted to an Intensive Care Unit and required phenytoin for the treatment or prophylaxis of postinjury seizures. For all the patients, phenytoin serum concentration was determined in three scheduled time points. Phenytoin serum concentration and pharmacokinetic parameters were compared between patients with TBI and cerebrovascular accident (CVA). Findings: The Vmaxand Kmwere significantly higher in head trauma (HT) patients than the CVA group. The phenytoin concentration (Cp) and the Cp/dose ratio were significantly higher in the CVA group patients during the first sampling (P < 0.05). The Acute Physiology and Chronic Health Evaluation П (APACHE П) score was significantly lower than the baseline at the end of the study in each group of patients (P < 0.05). In addition, no significant correlation was observed between Vmax, Km, Cp, Cp/dose ratio, and APACHE II scores at the time of sampling. Conclusion: Due to significant differences in phenytoin plasma concentration and pharmacokinetic parameters between HT and CVA patients, close attention must be paid to the pharmacokinetic behavior of phenytoin in the efforts to improve the patient's outcome after a severe HT.
Collapse
Affiliation(s)
- Shahnaz Alimardani
- Department of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sima Sadrai
- Department of Pharmaceutics, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Pooneh Salari
- Medical Ethics and History of Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Atabak Najafi
- Department of Anesthesiology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Behzad Eftekhar
- Department of Neurosurgery, Nepean Hospital, the University of Sydney, Sydney, Australia
| | - Mojtaba Mojtahedzadeh
- Department of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Pandey S, Singh K, Sharma V, Pandey D, Jha RP, Rai SK, Chauhan RS, Singh R. A prospective pilot study on serum cleaved tau protein as a neurological marker in severe traumatic brain injury. Br J Neurosurg 2017; 31:356-363. [PMID: 28293977 DOI: 10.1080/02688697.2017.1297378] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Neurotrauma has been labelled as a "silent epidemic" affecting both the developed and the developing nations. To date, no single brain-specific biomarker has been unanimously accepted for routine clinical use in TBI. Our study aims to determine the correlation of "cleaved-tau protein" in severe traumatic brain injury (TBI) with Glasgow Coma Scale (GCS) at the time of admission, mode of injury, CT findings and outcome at discharge. METHODS The study has been approved by the institutional ethical committee. 40 cases with severe TBI and 40 randomly selected healthy controls were included in this prospective study. Venous blood samples were collected and serum cleaved tau protein levels were measured and correlated with gender, mode of injury, CT findings GCS score and GOS score at discharge. RESULTS In the severe TBI group, the mean serum cleaved tau protein levels in males were 91.65 ± 41.34 pg/ml (mean ± S.D.), and females were 104.43 ± 53.08 pg/ml (mean ± S.D.), (p = 0.27). Mean serum C-tau level in study group was 95.48 ± 44.87 pg/ml (range 36.44-192.34), 95% C.I. (81.13-109.83) and in controls was 33.82 ± 13.65 pg/ml (range 2.48-66.54), 95% C.I. (29.46-38.19) (p < 0.001). The distribution of serum C-tau was in severe TBI group varied in all categories of GCS at 0th day (p < 0.001). Serum cleaved tau protein levels in the good outcome group were 74.26 ± 25.43 pg/ml (mean ± S.D.), range 36.44-144.54, 95% C.I. (63.52-85.00) and the poor-outcome group were 127.32 ± 49.40 pg/ml, range 66.65-192.34, 95% C.I. (100.99-153.64) (p = 0.001). CONCLUSION In severe TBI, serum cleaved tau protein levels were significantly higher as compared to the controls in this prospective study. However, results of this study are preliminary in nature and there is a need to undertake larger prospective studies to reach a definitive conclusion.
Collapse
Affiliation(s)
- Sharad Pandey
- a Department of Neuro Surgery , Sir Sunder Lal Hospital, IMS, BHU , Varanasi , Uttar Pradesh , India
| | - Kulwant Singh
- a Department of Neuro Surgery , Sir Sunder Lal Hospital, IMS, BHU , Varanasi , Uttar Pradesh , India
| | - Vivek Sharma
- a Department of Neuro Surgery , Sir Sunder Lal Hospital, IMS, BHU , Varanasi , Uttar Pradesh , India
| | - Deepa Pandey
- b Department of Clinical Microbiology , Central Hospital DLW , Varanasi , Uttar Pradesh , India
| | - Ravi Prakash Jha
- c Department of Community Medicine, Division of Biostatics , Sir Sunder Lal Hospital, IMS BHU , Varanasi , Uttar Pradesh , India
| | - Sunil Kumar Rai
- d Department of Anatomy , Sir Sunder Lal Hospital, IMS BHU , Varanasi , Uttar Pradesh , India
| | - Richa Singh Chauhan
- e Department of Radio diagnosis , Sir Sunder Lal Hospital, IMS BHU , Varanasi , Uttar Pradesh , India
| | - Royana Singh
- d Department of Anatomy , Sir Sunder Lal Hospital, IMS BHU , Varanasi , Uttar Pradesh , India
| |
Collapse
|
11
|
Peptide Pharmacological Approaches to Treating Traumatic Brain Injury: a Case for Arginine-Rich Peptides. Mol Neurobiol 2016; 54:7838-7857. [PMID: 27844291 DOI: 10.1007/s12035-016-0287-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/02/2016] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury (TBI) has a devastating effect on victims and their families, and has profound negative societal and economic impacts, a situation that is further compounded by the lack of effective treatments to minimise injury after TBI. The current strategy for managing TBI is partly through preventative measures and partly through surgical and rehabilitative interventions. Secondary brain damage remains the principal focus for the development of a neuroprotective therapeutic. However, the complexity of TBI pathophysiology has meant that single-action pharmacological agents have been largely unsuccessful in combatting the associated brain injury cascades, while combination therapies to date have proved equally ineffective. Peptides have recently emerged as promising lead agents for the treatment of TBI, especially those rich in the cationic amino acid, arginine. Having been shown to lessen the impact of ischaemic stroke in animal models, there are reasonable grounds to believe that arginine-rich peptides may have neuroprotective therapeutic potential in TBI. Here, we review a range of peptides previously examined as therapeutic agents for TBI. In particular, we focus on cationic arginine-rich peptides -- a new class of agents that growing evidence suggests acts through multiple neuroprotective mechanisms.
Collapse
|
12
|
Anderson GD, Peterson TC, Vonder Haar C, Farin FM, Bammler TK, MacDonald JW, Kantor ED, Hoane MR. Effect of Traumatic Brain Injury, Erythropoietin, and Anakinra on Hepatic Metabolizing Enzymes and Transporters in an Experimental Rat Model. AAPS JOURNAL 2015; 17:1255-67. [PMID: 26068867 DOI: 10.1208/s12248-015-9792-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/26/2015] [Indexed: 12/18/2022]
Abstract
In contrast to considerable data demonstrating a decrease in cytochrome P450 (CYP) activity in inflammation and infection, clinically, traumatic brain injury (TBI) results in an increase in CYP and UDP glucuronosyltransferase (UGT) activity. The objective of this study was to determine the effects of TBI alone and with treatment with erythropoietin (EPO) or anakinra on the gene expression of hepatic inflammatory proteins, drug-metabolizing enzymes, and transporters in a cortical contusion impact (CCI) injury model. Microarray-based transcriptional profiling was used to determine the effect on gene expression at 24 h, 72 h, and 7 days post-CCI. Plasma cytokine and liver protein concentrations of CYP2D4, CYP3A1, EPHX1, and UGT2B7 were determined. There was no effect of TBI, TBI + EPO, or TBI + anakinra on gene expression of the inflammatory factors shown to be associated with decreased expression of hepatic metabolic enzymes in models of infection and inflammation. IL-6 plasma concentrations were increased in TBI animals and decreased with EPO and anakinra treatment. There was no significant effect of TBI and/or anakinra on gene expression of enzymes or transporters known to be involved in drug disposition. TBI + EPO treatment decreased the gene expression of Cyp2d4 at 72 h with a corresponding decrease in CYP2D4 protein at 72 h and 7 days. CYP3A1 protein was decreased at 24 h. In conclusion, EPO treatment may result in a significant decrease in the metabolism of Cyp-metabolized drugs. In contrast to clinical TBI, there was not a significant effect of experimental TBI on CYP or UGT metabolic enzymes.
Collapse
Affiliation(s)
- Gail D Anderson
- Department of Pharmacy, University of Washington, Seattle, Washington, 98195, USA,
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Franconi F, Campesi I. Sex and gender influences on pharmacological response: an overview. Expert Rev Clin Pharmacol 2014; 7:469-85. [DOI: 10.1586/17512433.2014.922866] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
14
|
Schmidt D, Friedman D, Dichter MA. Anti-epileptogenic clinical trial designs in epilepsy: issues and options. Neurotherapeutics 2014; 11:401-11. [PMID: 24420312 PMCID: PMC3996121 DOI: 10.1007/s13311-013-0252-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although trials with anti-seizure drugs have not shown anti-epileptogenic or disease-modifying activity in humans, new compounds are on the horizon that may require novel trial designs. We briefly discuss the unique challenges and the available options to identify innovative clinical trial designs that differentiate novel anti-epileptogenic and disease-modifying compounds, preferably early in phase II, from current anti-seizure drugs. The most important challenges of clinical testing of agents for epilepsy prevention include having sufficient preclinical evidence for a suitable agent to proceed with a human trial of an anti-epileptogenic drug, and to demonstrate the feasibility of doing such a trial. Major challenges in trial design to assess agents for disease modification include the choice of suitable study parameters, the identification of a high-risk study population, the type of control, the time and duration of treatment, and a feasible follow-up period.
Collapse
Affiliation(s)
- Dieter Schmidt
- Epilepsy Research Group, Goethestrasse 5, 14163, Berlin, Germany,
| | | | | |
Collapse
|
15
|
Abstract
Traumatic brain injury (TBI) is the most important cause of disability in individuals under the age of 45 years and thus represents a significant social and economic burden. Evidence strongly suggests that oxidative stress is a cornerstone event leading to and propagating secondary injury mechanisms such as excitotoxicity, mitochondrial dysfunction, apoptosis, autophagy, brain edema, and inflammation. TBI has defied conventional approaches to diagnosis and therapy development because of its heterogeneity and complexity. Therefore, it is necessary to explore alternative approaches to therapy development for TBI. The aim of this review is to present a therapeutic approach for TBI, taking into account the evidence supporting the role for oxidative stress in the pathophysiological processes of secondary brain injury. The role of agents such as mitochondria-targeted antioxidants (melatonin and new mitochondria-targeted antioxidants), nicotinamide adenine dinucleotide phosphate (NADPH) inhibitors (antioxidant vitamins and apocynin), and other compounds having mainly antioxidant properties (hydrogen-rich saline, sulforaphane, U-83836E, omega-3, and polyphenols) is covered. The rationale for innovative antioxidant therapies based on current knowledge and particularly the most recent studies regarding this field is discussed. Particular considerations and translational potential of new TBI treatments are examined and a novel therapeutic proposal for TBI is presented.
Collapse
|
16
|
Roberts DJ, Hall RI. Drug absorption, distribution, metabolism and excretion considerations in critically ill adults. Expert Opin Drug Metab Toxicol 2013; 9:1067-84. [PMID: 23682923 DOI: 10.1517/17425255.2013.799137] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION All critically ill patients require medication to treat organ dysfunction. However, the pharmacokinetics of drugs used to treat these patients is complex due to frequent alterations in drug absorption, distribution, metabolism, and excretion (ADME). AREAS COVERED This review examines pharmacokinetic aspects of drug administration for adult intensive care unit (ICU) patients. Specifically, the authors examine the ADME changes that occur and which should be considered by clinicians when delivering drug therapy to critically ill patients. EXPERT OPINION Dosage pharmacokinetics determined from single-dose or limited-duration administration studies in healthy volunteers may not apply to critically ill patients. Organ dysfunction among these patients may be due to pre-existing disease or the effects of a systemic or locoregional inflammatory response precipitated by their illness. Alterations in pharmacokinetics observed among the critically ill include altered bioavailability after enteral administration, increased volume of distribution and blood-brain barrier permeability and changes in P-glycoprotein and cytochrome P450 enzyme function. However, the effect of these changes on clinically important outcomes remains uncertain and poorly studied. Future investigations should examine not only pharmacokinetic changes among the critically ill, but also whether recognition of these changes and alterations in drug therapy directed as a consequence of their observation alters patient outcomes.
Collapse
Affiliation(s)
- Derek J Roberts
- University of Calgary, Departments of Surgery and Community Health Sciences, Calgary, Alberta T2N 5A1, Canada
| | | |
Collapse
|
17
|
|
18
|
McConeghy KW, Hatton J, Hughes L, Cook AM. A review of neuroprotection pharmacology and therapies in patients with acute traumatic brain injury. CNS Drugs 2012; 26:613-36. [PMID: 22668124 DOI: 10.2165/11634020-000000000-00000] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traumatic brain injury (TBI) affects 1.6 million Americans annually. The injury severity impacts the overall outcome and likelihood for survival. Current treatment of acute TBI includes surgical intervention and supportive care therapies. Treatment of elevated intracranial pressure and optimizing cerebral perfusion are cornerstones of current therapy. These approaches do not directly address the secondary neurological sequelae that lead to continued brain injury after TBI. Depending on injury severity, a complex cascade of processes are activated and generate continued endogenous changes affecting cellular systems and overall outcome from the initial insult to the brain. Homeostatic cellular processes governing calcium influx, mitochondrial function, membrane stability, redox balance, blood flow and cytoskeletal structure often become dysfunctional after TBI. Interruption of this cascade has been the target of numerous pharmacotherapeutic agents investigated over the last two decades. Many agents such as selfotel, pegorgotein (PEG-SOD), magnesium, deltibant and dexanabinol were ineffective in clinical trials. While progesterone and ciclosporin have shown promise in phase II studies, success in larger phase III, randomized, multicentre, clinical trials is pending. Consequently, no neuroprotective treatment options currently exist that improve neurological outcome after TBI. Investigations to date have extended understanding of the injury mechanisms and sites for intervention. Examination of novel strategies addressing both pathological and pharmacological factors affecting outcome, employing novel trial design methods and utilizing biomarkers validated to be reflective of the prognosis for TBI will facilitate progress in overcoming the obstacles identified from previous clinical trials.
Collapse
|
19
|
Marklund N, Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here? Br J Pharmacol 2011; 164:1207-29. [PMID: 21175576 PMCID: PMC3229758 DOI: 10.1111/j.1476-5381.2010.01163.x] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability in young adults. Survivors of TBI frequently suffer from long-term personality changes and deficits in cognitive and motor performance, urgently calling for novel pharmacological treatment options. To date, all clinical trials evaluating neuroprotective compounds have failed in demonstrating clinical efficacy in cohorts of severely injured TBI patients. The purpose of the present review is to describe the utility of animal models of TBI for preclinical evaluation of pharmacological compounds. No single animal model can adequately mimic all aspects of human TBI owing to the heterogeneity of clinical TBI. To successfully develop compounds for clinical TBI, a thorough evaluation in several TBI models and injury severities is crucial. Additionally, brain pharmacokinetics and the time window must be carefully evaluated. Although the search for a single-compound, 'silver bullet' therapy is ongoing, a combination of drugs targeting various aspects of neuroprotection, neuroinflammation and regeneration may be needed. In summary, finding drugs and prove clinical efficacy in TBI is a major challenge ahead for the research community and the drug industry. For a successful translation of basic science knowledge to the clinic to occur we believe that a further refinement of animal models and functional outcome methods is important. In the clinical setting, improved patient classification, more homogenous patient cohorts in clinical trials, standardized treatment strategies, improved central nervous system drug delivery systems and monitoring of target drug levels and drug effects is warranted.
Collapse
Affiliation(s)
- Niklas Marklund
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala University Hospital, Uppsala, Sweden.
| | | |
Collapse
|
20
|
Pharmacokinetics of intravenous levofloxacin administered at 750 milligrams in obese adults. Antimicrob Agents Chemother 2011; 55:3240-3. [PMID: 21576432 DOI: 10.1128/aac.01680-10] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The physiochemical properties of levofloxacin suggest that it is an agent which may exhibit altered pharmacokinetics in obese individuals. The purpose of this study was to describe the pharmacokinetics of a single 750-mg intravenous dose of levofloxacin in both hospitalized and ambulatory obese individuals. The hypothesis was that a standard dose of levofloxacin in obese individuals would achieve serum concentrations likely to be therapeutic. A single levofloxacin dose of 750 mg was infused over 90 min, and seven serial serum samples were subsequently obtained to evaluate the pharmacokinetics after the first dose. The peak concentrations of levofloxacin were comparable to those seen with normal-weight individuals. However, the area under the concentration-time curve and clearance were quite variable. Accelerated clearance was evident in the ambulatory obese individuals. Further investigation of the effects of obesity on the pharmacokinetics of levofloxacin is necessary to ensure optimal dosing.
Collapse
|
21
|
Gross AK, Norman J, Cook AM. Contemporary pharmacologic issues in the management of traumatic brain injury. J Pharm Pract 2010; 23:425-40. [PMID: 21507847 DOI: 10.1177/0897190010372322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in the United States. While there are no pharmacotherapeutic options currently available for attenuating the neurologic injury cascade after TBI, numerous pharmacologic issues are encountered in these critically ill patients. Adequate fluid resuscitation, reversal of coagulopathy, maintenance of cerebral perfusion, and treatment of intracranial hypertension are common interventions early in the treatment of TBI. Other deleterious complications such as venous thromboembolism, extremes in glucose concentrations, and stress-related mucosal disease should be anticipated and avoided. Early provision of nutrition and prevention of drug or alcohol withdrawal are also cornerstones of routine care in TBI patients. Prevention of infections and seizures may also be helpful. Clinicians caring for TBI patients should be familiar with the pharmacologic issues typical of this vulnerable population in order to develop optimal strategies of care to anticipate and prevent common complications.
Collapse
|
22
|
Cook AM, Whitlow J, Hatton J, Young B. Cyclosporine A for neuroprotection: establishing dosing guidelines for safe and effective use. Expert Opin Drug Saf 2009; 8:411-9. [DOI: 10.1517/14740330903066742] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
A critical review: does thiopental continuous infusion warrant therapeutic drug monitoring in the critical care population? Ther Drug Monit 2009; 31:153-69. [PMID: 19177032 DOI: 10.1097/ftd.0b013e318196fb9f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Thiopental is a barbiturate used in traumatic brain injuries (TBIs) to reduce intracranial pressure (ICP) and to manage cerebral ischemia. As thiopental follows Michaelis-Menten kinetics, therapeutic drug monitoring (TDM) has been used in practice to improve efficacy and reduce adverse effects. However, its role is still debatable, and TDM is not widely practiced. Current evidence suggests that thiopental therapy may improve mortality and functional outcome in a subpopulation of patients with severe TBI with elevated ICP refractory to conventional medical therapy. Several analytical methods are available to quantify thiopental concentrations. This review uses a previously published 9-step decision-making algorithm to determine whether TDM of thiopental in TBI is warranted. There seems to be poor correlation between thiopental concentration and pharmacological response in terms of neurological response, ICP, electroencephalography, and drug toxicity. There is no established therapeutic range for thiopental continuous infusion due to a wide range of plasma concentrations corresponding to efficacy (25-50 mg/L) and toxicity (30-70 mg/L) and the resulting overlap between the 2. Thiopental exhibits intrapatient and interpatient variability due to age, obesity, renal and hepatic dysfunction, Michaelis-Menten kinetics, and hepatic enzyme autoinduction. Available evidence suggests that TDM of thiopental continuous infusion is not beneficial in improving efficacy or avoiding toxicity. There are however 2 possible scenarios in which TDM may provide additional information to sound clinical judgment. The first is providing patient-specific plasma target concentration to guide titration of therapy. The second scenario is differentiating between brain death and barbiturate-induced coma.
Collapse
|
24
|
Moderate hypothermia prevents cardiac arrest-mediated suppression of drug metabolism and induction of interleukin-6 in rats. Crit Care Med 2009; 37:263-9. [PMID: 19050605 DOI: 10.1097/ccm.0b013e3181931ed3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Therapeutic hypothermia is being clinically used to reduce neurologic deficits after cardiac arrest (CA). Patients receiving hypothermia after CA receive a wide-array of medications. During hypothermia, drug metabolism is markedly reduced. Little, however, is known about the impact of hypothermia on drug metabolism after rewarming. The objective of this study was to examine the effect of CA and hypothermia on the functional regulation of two major drug metabolizing cytochrome P450 (CYP) isoforms. DESIGN Laboratory investigation. SETTING University pharmacy school and animal research facility. SUBJECTS Thirty-six male Sprague-Dawley rats. INTERVENTIONS Hypothermia was induced via surface cooling in a rat CA model and maintained for 3 hrs. Animals were killed at 5 or 24 hrs and liver was analyzed for hepatic activity and mRNA expression of CYP3A2 and CYP2E1. Plasma interleukin-6 (IL-6) concentrations were determined. The effect of IL-6 on pregnane X receptor-mediated transcription of the rat CYP3A2 promoter was evaluated via luciferase reporter in HepG2 cells. MEASUREMENTS AND MAIN RESULTS At 24 hrs after CA a decrease in CYP3A2 and CYP2E1 activity was observed, 55.7% +/- 12.8% and 46.8% +/- 29.7% of control, respectively (p < 0.01). CA decreased CYP3A2 mRNA (p < 0.05), but not CYP2E1 mRNA. Expression of other pregnane X receptor target enzymes and transporter genes were similarly down-regulated. CA also produced an approximately ten-fold increase in plasma IL-6. CA-mediated inhibition of CYP3A2 and CYP2E1 was attenuated by hypothermia, as was the increase in IL-6. Furthermore, IL-6 attenuated pregnane X receptor-mediated transcription of the CYP3A2 promoter. CONCLUSIONS CA produces CYP3A2 down-regulation at 24 hrs, potentially via IL-6 effects on pregnane X receptor-mediated transcription. Also, hypothermia attenuates the CA-mediated down-regulation, thereby normalizing drug metabolism after rewarming.
Collapse
|
25
|
Zagli G, Tarantini F, Bonizzoli M, Di Filippo A, Peris A, De Gaudio AR, Geppetti P. Altered pharmacology in the Intensive Care Unit patient. Fundam Clin Pharmacol 2008; 22:493-501. [PMID: 18684127 DOI: 10.1111/j.1472-8206.2008.00623.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Critically ill patients, not infrequently present alterations of physiological parameters that determine the success/failure of therapeutic interventions as well as the final outcome. Sepsis and polytrauma are two of the most common and complex syndromes occurring in Intensive Care Unit (ICU) and affect drug absorption, disposition, metabolism and elimination. Pharmacological management of ICU patients requires consideration of the unique pharmacokinetics associated with these clinical conditions and the likely occurrence of drug interaction. Rational adjustment in drug choice and dosing contributes to the appropriateness of treatment of those patients.
Collapse
Affiliation(s)
- Giovanni Zagli
- Department of Critical Care Medicine and Surgery, University of Florence and Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| | | | | | | | | | | | | |
Collapse
|
26
|
Dosing and therapeutic monitoring of phenytoin in young adults after neurotrauma: are current practices relevant? Clin Neuropharmacol 2008; 30:362-9. [PMID: 18090462 DOI: 10.1097/wnf.0b013e318059ae1c] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Anticonvulsant drugs are commonly used to treat and prevent seizures after neurotrauma. However, many physiological changes occur in the neurotrauma patient, which alter the pharmacokinetics of drugs such as phenytoin. This raises concerns relating to the dosage and monitoring of phenytoin in these patients compared with its routine use in epileptic patients. Examples of pharmacokinetic alterations within the neurotrauma patient include changes in hepatic metabolism, protein binding alterations, and disruption of the blood-brain barrier. Drug interactions and genetic factors may also contribute to pharmacokinetic variations. Many studies have reported that neurotrauma patients often present with either subtherapeutic or highly variable phenytoin serum concentrations. When phenytoin doses recommended for the epileptic patient are used in the neurotrauma patient, efficacy is limited to early posttraumatic seizures, with no effect on morbidity, mortality, or the onset of late posttraumatic seizures. This review examines the effect of neurotrauma on the pharmacokinetics of phenytoin alongside clinical outcomes and questions the current dosing and therapeutic monitoring practices within this area.
Collapse
|
27
|
Anderson GD, Temkin NR, Awan AB, Winn HR, Winn RH. Effect of time, injury, age and ethanol on interpatient variability in valproic acid pharmacokinetics after traumatic brain injury. Clin Pharmacokinet 2007; 46:307-18. [PMID: 17375982 DOI: 10.2165/00003088-200746040-00004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) results in an increase in hepatic metabolism. The increased metabolism is in significant contrast to a large body of in vitro and in vivo data demonstrating that activation of the host-defence response downregulates hepatic metabolism. Theoretically, this occurs because of activation of the pro-inflammatory cytokines tumour necrosis factor-alpha, interferon-gamma, interleukin (IL)-1 and IL-6. As part of a large double-blind, placebo-controlled clinical trial evaluating the use of valproic acid for prophylaxis of post-traumatic seizures, we obtained extensive valproic acid concentration-time data. Valproic acid is a hepatically metabolised, low extraction-ratio drug. Therefore, unbound clearance (CL(u)) is equal to intrinsic or metabolic clearance. OBJECTIVE The objective of this study was to evaluate the time-dependent effects of TBI on the pharmacokinetics of total and unbound valproic acid with the goal of identifying patient factors that may predict changes in total clearance (CL) and CL(u). In addition, by determining the factors that influence the magnitude and time course of induction of hepatic metabolism and understanding their interaction with the host-defence mediators, we can further our insight into the mechanism(s) responsible for the changes in CL and CL(u). STUDY DESIGN Valproic acid plasma concentration data were obtained from 158 TBI patients. Unbound valproic acid plasma concentrations were estimated using total valproic acid plasma and albumin concentrations following a Scatchard equation binding model previously developed in a subset of TBI patients. The effect of 13 patient factors on CL and CL(u) was evaluated initially in a univariate analysis. The significant factors were then included in a multiple linear regression analysis by use of step-wise selection and forward selection procedures. RESULTS CL and CL(u) were significantly increased after TBI in a time-dependent manner. The average increase was >75% by weeks 2 and 3 post-injury. The magnitude of the induction of CL was increased with decreased albumin concentrations, in addition to the presence of ethanol on admission, increased severity of head injury, tube feeding and total parenteral nutrition (TPN). The magnitude of induction of CL(u) was increased by older age, presence of ethanol on admission, increased severity of head injury, tube feeding, TPN, and if the patient had a post-injury neurosurgical procedure. The time to normalisation of CL(u) was significantly longer in patients with head injury plus other injuries compared with those with head injury alone. CONCLUSIONS As has been reported with other drugs, TBI results in a significant increase in the metabolism of valproic acid. The patient factors identified in this study that resulted in an increase in the magnitude and time course of the induction of CL(u) (ethanol, older age, presence of a neurosurgical procedure, severity of TBI and presence of multiple non-TBI injuries) have all been reported to cause a shift to the anti-inflammatory mediators IL-4 and IL-10. This suggests that the increase in hepatic metabolism after TBI may be due to the increased presence of anti-inflammatory mediators in contrast to the inhibition effect of the pro-inflammatory mediators in non-TBI inflammation and infection.
Collapse
Affiliation(s)
- Gail D Anderson
- Departments of Pharmacy and Neurological Surgery, Schools of Pharmacy and Medicine [corrected] University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Physiologic alterations in critically ill patients can significantly affect the pharmacokinetics of drugs used in the critically ill patient population. Understanding these pharmacokinetic changes is essential relative to optimizing drug therapy. This article outlines the major differences seen in the absorption, distribution, metabolism, and excretion of drugs in critically ill patients. Important strategies for drug therapy dosing and monitoring in these patients are also addressed.
Collapse
Affiliation(s)
- Bradley A Boucher
- Department of Pharmacy, University of Tennessee Health Science Center, 26 South Dunlap, Room 210, Memphis, TN 38163, USA.
| | | | | |
Collapse
|
29
|
Empey PE, McNamara PJ, Young B, Rosbolt MB, Hatton J. Cyclosporin A disposition following acute traumatic brain injury. J Neurotrauma 2006; 23:109-16. [PMID: 16430377 DOI: 10.1089/neu.2006.23.109] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Although the precise mechanism of action remains to be defined, Cyclosporin A (CsA) has demonstrated potential for neuroprotection in animal models. Predictive dosing strategies for CsA in acute traumatic brain injured (TBI) patients must account for the influence of the acute phase response on drug disposition. To characterize CsA pharmacokinetic parameters early following acute TBI, serial blood samples from patients enrolled into a Phase II dose-escalation trial were analyzed. Within eight hours of injury, thirty patients admitted with acute severe TBI were prospectively randomized into three cohorts (n = 8 CsA; n = 2 placebo per cohort) in this dose-escalation trial. Patients received one of three doses (I = 0.625 mg/kg/dose; II = 1.25 mg/kg/dose; III = 2.5 mg/kg/dose) or placebo intravenously every 12 h for 72 h. Serial blood collection began prior to dose 1 and continued for 72 h following the completion of six doses. Whole blood concentrations were determined by high-performance liquid chromatography (HPLC) with ultraviolet (UV) detection. Pharmacokinetic parameters were determined for each patient by fitting the concentration-time profile to a two-compartmental model with first order elimination. Mean area under the curve and predicted maximal blood concentration increased with each dosing cohort (I = 9840 h*microg/L, 398 microg/L; II = 18300 h*microg/L, 645 microg/L; III = 32500 h*microg/L, 1300 microg/L). Whole blood clearance, steady state volume of distribution, and beta half-life were independent of dose and higher than published reports from other populations: 0.420 L/h/kg, 5.91 L/kg, and 17.3 h, respectively. These data show patients with acute severe TBI demonstrate a more rapid clearance and a larger distribution volume of CsA. Pharmacokinetic parameters derived from this study will guide dosing strategies for future prospective clinical trials evaluating CsA therapy following acute TBI.
Collapse
Affiliation(s)
- Philip E Empey
- Department of Pharmaceutical Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0509, USA
| | | | | | | | | |
Collapse
|
30
|
Smith KM, Jeske CS, Young B, Hatton J. Prevalence and Characteristics of Adverse Drug Reactions in Neurosurgical Intensive Care Patients. Neurosurgery 2006; 58:426-33; discussion 426-33. [PMID: 16528181 DOI: 10.1227/01.neu.0000197321.57853.83] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract
OBJECTIVE:
To evaluate the prevalence and characteristics of adverse drug reactions (ADRs) in neurosurgical intensive care patients.
METHODS:
Retrospective analysis of ADR data obtained from a spontaneous reporting system in a tertiary care university hospital. Reports of suspected ADRs in adult patients admitted emergently or electively to the neurosurgical service were included.
RESULTS:
Over the 3 year period, 3496 neurosurgical intensive care unit (ICU) patient admissions accounted for 5% of all hospital admissions. A total of 10% of all neurosurgical patients developed a suspected ADR, with three patients experiencing multiple reactions. Other adult ICU patients developed ADRs at a comparable rate (9%, P>0.05). Overall, neurosurgery patients accounted for 12% of all spontaneously reported ADRs. Preventable reactions were observed in 43 (13%) cases, and treatment was required for 76%. The majority (96%) of ADRs resolved or improved at the time of the ADR report. Nausea, pruritus, thrombocytopenia, and vomiting were most frequently noted. Therapies most often associated with reported events were analgesics, antipyretics, antibiotics, anticonvulsants, and histamine H2 antagonists. The relationship between central nervous system disease and adverse event occurrence is not clear.
CONCLUSION:
Despite the narrow scope of drug regimens in neurosurgical ICU patients, ADRs can complicate therapy in this critically ill population. Neurosurgical ICU patients seem to experience ADRs no more frequently than their adult ICU counterparts.
Collapse
Affiliation(s)
- Kelly M Smith
- University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0293, USA
| | | | | | | |
Collapse
|
31
|
Wright DW, Ritchie JC, Mullins RE, Kellermann AL, Denson DD. Steady-state serum concentrations of progesterone following continuous intravenous infusion in patients with acute moderate to severe traumatic brain injury. J Clin Pharmacol 2006; 45:640-8. [PMID: 15901745 DOI: 10.1177/0091270005276201] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Progesterone (PG) has been shown to provide substantial neuroprotection after traumatic brain injury (TBI) in multiple animal models. As a first step in assessing applicability to humans, the authors examined the effects of acute TBI and extracranial trauma on the pharmacokinetics of PG given by intravenous infusion. Multiple blood samples were obtained from 11 female and 21 male trauma patients receiving PG and 1 female and 3 male patients receiving placebo infusions for 72 hours. Values for C(SS), CL, t(1/2), and Vd were obtained using AUC((0-72)) and postinfusion blood samples. C(SS) values were 337 +/- 135 ng/mL, which were significantly lower than the target concentration of 450 +/- 100 ng/mL. The lower C(SS) is attributed to the CL, which was higher than anticipated. In addition, t(1/2) was longer and V(d) was higher than anticipated. These results demonstrate that stable PG concentrations can be rapidly achieved following TBI.
Collapse
Affiliation(s)
- David W Wright
- Department of Emergency Medicine, Emory University School of Medicine, Emergency Medicine Research Center, 49 Jessie Hill Jr Drive, Atlanta, GA 30303, USA
| | | | | | | | | |
Collapse
|
32
|
De Paepe P, Belpaire FM, Van Hoey G, Boon PA, Buylaert WA. The influence of endotoxemia on the pharmacokinetics and the electroencephalographic effect of propofol in the rat. J Pharm Sci 2003; 92:104-14. [PMID: 12486687 DOI: 10.1002/jps.10275] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Endotoxemia decreases the dose requirement for anesthetics but no data are available for propofol. A rat model was used in which the influence of endotoxin administration on the pharmacokinetics and pharmacodynamics of propofol was investigated. Chronically instrumented rats were randomly allocated to either a control (n = 9) or an endotoxin (n = 9) group. Six hours after pretreatment with either endotoxin or its solvent, propofol was infused (150 mg x kg(-1) x h(-1)) until isoelectric periods of 5 s or longer were observed in the electroencephalogram. The changes observed in the electroencephalogram were quantified using aperiodic analysis and used as a surrogate measure of hypnosis. The righting reflex served as a clinical measure of hypnosis. The propofol dose needed to reach the electroencephalographic end point in the endotoxin-treated rats was reduced by almost 50% (p < 0.01). This could be attributed to a decrease in propofol clearance and in distribution volume related to the degree of physiologic and metabolic disturbances induced by endotoxin. To investigate changes in end organ sensitivity, the biphasic electroencephalographic effect versus effect-site concentration relationship was studied. This relationship was characterized by descriptors that showed an increased intrinsic efficacy of propofol in the endotoxin group. The effect-site concentration at the return of righting reflex was lower in the endotoxin group. Our study demonstrates that endotoxin-treated animals need a lower dose of propofol to reach the same degree of anesthetic effect which can mainly be attributed to changes in pharmacokinetics.
Collapse
Affiliation(s)
- Peter De Paepe
- Heymans Institute of Pharmacology, Ghent University, De Pintelaan 185, B-9000 Ghent, Belgium.
| | | | | | | | | |
Collapse
|
33
|
Mamiya K, Yukawa E, Matsumoto T, Aita C, Goto S. Synergistic effect of valproate coadministration and hypoalbuminemia on the serum-free phenytoin concentration in patients with severe motor and intellectual disabilities. Clin Neuropharmacol 2002; 25:230-3. [PMID: 12151911 DOI: 10.1097/00002826-200207000-00007] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We investigated whether a combination of risk factors affects the free phenytoin (PHT) fraction by multiple regression analyses in 30 patients with severe motor and intellectual disabilities (SMID) with epilepsy. The risk factors analyzed were gender, age, total PHT concentration, albumin concentration, aspartate aminotransferase, alanin aminotransferase, serum creatinine, blood urea nitrogen, and antiepileptic drug concentrations. Serum levels of total and free PHT were measured by fluorescence polarization immunoassay. Free PHT fractions were between 7.2% and 17.3% (average 10.9%). Two factors, hypoalbuminemia and valproate (VPA) coadministratation with PHT, increased free PHT fraction, and a combination of these two markedly increased free PHT fraction. Patients with these double risk factors have a high risk of exceeding the therapeutic range of serum-free PHT concentration even if their total PHT concentration does not. Therefore, we should monitor free PHT concentration, especially in SMID patients with epilepsy, because they may have hypoalbuminemia and are treated with antiepileptic drug polytherapy and, moreover, cannot report adverse effects of the drugs.
Collapse
|
34
|
Davey AK. Clinical Relevance of Cytokine-Induced Changes in Drug Metabolism. JOURNAL OF PHARMACY PRACTICE AND RESEARCH 2002. [DOI: 10.1002/jppr2002322147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
35
|
Mesiwala AH, Farrell L, Wenzel HJ, Silbergeld DL, Crum LA, Winn HR, Mourad PD. High-intensity focused ultrasound selectively disrupts the blood-brain barrier in vivo. ULTRASOUND IN MEDICINE & BIOLOGY 2002; 28:389-400. [PMID: 11978420 DOI: 10.1016/s0301-5629(01)00521-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
High-intensity focused ultrasound (HIFU) has been shown to generate lesions that destroy brain tissue while disrupting the blood-brain barrier (BBB) in the periphery of the lesion. BBB opening, however, has not been shown without damage, and the mechanisms by which HIFU induces BBB disruption remain unknown. We show that HIFU is capable of reversible, nondestructive, BBB disruption in a targeted region-of-interest (ROI) (29 of 55 applications; 26 of 55 applications showed no effect); this opening reverses after 72 h. Light microscopy demonstrates that HIFU either entirely preserves brain architecture while opening the BBB (18 of 29 applications), or generates tissue damage in a small volume within the region of BBB opening (11 of 29 applications). Electron microscopy supports these observations and suggests that HIFU disrupts the BBB by opening capillary endothelial cell tight junctions, an isolated ultrastructural effect that is different from the mechanisms through which other (untargeted) modalities, such as hyperosmotic solutions, hyperthermia and percussive injury disrupt the BBB.
Collapse
Affiliation(s)
- Ali H Mesiwala
- Department of Neurological Surgery, University of Washington, Seattle, WA 98105, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Sahuquillo J, de Nadal M, Biestro A. Fármacos en el traumatismo craneoencefálico grave. Neurocirugia (Astur) 2002. [DOI: 10.1016/s1130-1473(02)70613-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Marklund N, Lewander T, Clausen F, Hillered L. Effects of the nitrone radical scavengers PBN and S-PBN on in vivo trapping of reactive oxygen species after traumatic brain injury in rats. J Cereb Blood Flow Metab 2001; 21:1259-67. [PMID: 11702041 DOI: 10.1097/00004647-200111000-00002] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In previous studies, the authors showed that the nitrone radical scavenger alpha-phenyl-N- tert -butyl nitrone (PBN) and its sulfo-derivative, 2-sulfo-phenyl-N- tert -butyl nitrone (S-PBN), attenuated cognitive disturbance and reduced tissue damage after traumatic brain injury (TBI) in rats. In the current study, the production of reactive oxygen species (ROS) after TBI was monitored with microdialysis and the 4-hydroxybenzoic acid (4-HBA) trapping method. A single dose of PBN (30 mg/kg) or an equimolar dose of S-PBN (47 mg/kg) was administered intravenously 30 minutes before a controlled cortical contusion injury in rats. Plasma and brain tissue drug concentrations were analyzed at the end of the microdialysis experiment (3 hours after injury) and, in a separate experiment with S-PBN, at 30 and 60 minutes after injury. Traumatic brain injury caused a significant increase in ROS formation that lasted for 60 minutes after the injury as evidenced by increased 3,4-dihydroxybenzoic acid (3,4-DHBA) concentrations in the dialysate. PBN and S-PBN equally and significantly attenuated the posttraumatic increase in 3,4-DHBA formation. High PBN concentrations were found bilaterally in brain tissue up to 3 hours after injury. In contrast, S-PBN was rapidly cleared from the circulation and was not detectable in brain at 30 minutes after injury or at any later time point. The results suggest that scavenging of ROS after TBI may contribute to the neuroprotective properties observed with nitrone spin-trapping agents. S-PBN, which remained undetectable even in traumatized brain tissue, reduced ROS production to the same extent as PBN that readily crossed the blood-brain barrier. This finding supports an important role for ROS production at the blood-endothelial interface in TBI.
Collapse
Affiliation(s)
- N Marklund
- Department of Neuroscience, Neurosurgery, Uppsala University Hospital, Sweden
| | | | | | | |
Collapse
|
38
|
Mizukami Y, Yamada S, Kokudo N, Takashima M, Yokoyama T. Dietary iron reduces the anti-convulsion activity of phenytoin in electroconvulsion via inhibition of brain penetration. Brain Res 2001; 915:112-7. [PMID: 11578628 DOI: 10.1016/s0006-8993(01)02836-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We determined the anti-convulsion activity of phenytoin (PHT) using the maximum electron shock method in mice fed diets containing various concentrations of iron for 18 weeks. Dietary iron reduces the anti-convulsion activity of PHT in a dose-dependent manner (0-6100 ppm). High concentrations of PHT are detected in the plasma of mice fed a high iron diet compared with those fed normal and low iron diets, in contrast to the pharmacological effect. However, the concentration of PHT in the brains of mice fed high amounts of dietary iron decreased significantly 3 h after treatment with PHT, consistent with the anti-convulsion effect of PHT. The relationship between brain and plasma-unbound concentrations of PHT indicates that the penetration of PHT into brain is significantly inhibited by dietary iron.
Collapse
Affiliation(s)
- Y Mizukami
- Department of Hospital Pharmacy, Faculty of Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
39
|
Irazuzta JE, de Courten-Myers G, Zemlan FP, Bekkedal MY, Rossi J. Serum cleaved Tau protein and neurobehavioral battery of tests as markers of brain injury in experimental bacterial meningitis. Brain Res 2001; 913:95-105. [PMID: 11532253 DOI: 10.1016/s0006-8993(01)02764-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brain injury due to bacterial meningitis affects multiple areas of the brain with a heterogeneous distribution generating a challenge to assess severity. Tau proteins are microtubular binding proteins localized in the axonal compartment of neurons. Brain injury releases cleaved Tau proteins (C-tau) into the extracellular space where they are transported to the cerebral spinal fluid. We hypothesized that C-tau crosses the blood-brain barrier during inflammation and that it can be detected in serum. The correlation between serum C-tau levels and the extent of the meningitic insult was examined. Furthermore, we studied whether the use of a subset of neurobehavioral tasks can assess the extent of brain injury after meningitis. The tests were chosen primarily for their ability to detect deficits in the acoustic system, low brain, reflexive responding, as well as for impaired motor coordination and the higher brain functions of learning and memory. A rat model of group B streptococcal meningitis with variable severity was utilized. At five days after bacterial inoculation followed by antibiotic therapy neurobehavioral tests were performed and serum C-tau and histologic samples of the brain were obtained. Our study shows that during meningitis C-tau appears in serum and reflects the extent of neurologic damage. Neurobehavioral performance was altered after bacterial meningitis and could be correlated with histologic and biochemical markers of neurologic sequelae. We conclude that serum C-tau and a composite of neurobehavioral tests could become useful markers for assessing the severity of neurological damage in experimental bacterial meningitis.
Collapse
Affiliation(s)
- J E Irazuzta
- Division of Critical Care, Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | | | | | | |
Collapse
|
40
|
Abstract
The authors examined the effect of mild therapeutic hypothermia on phenytoin pharmacokinetics in 14 patients with brain damage. Each patient was given phenytoin during and after mild therapeutic hypothermia. Plasma concentrations of total phenytoin, unbound phenytoin, and 5-(p-hydroxyphenyl)-5-phenylhydantoin (5-p-HPPH), the major metabolite of phenytoin, were measured. Pharmacokinetic parameters during and after mild therapeutic hypothermia were compared. Plasma concentrations of total and unbound phenytoin were significantly higher during hypothermia than after hypothermia. The area under the plasma concentration-time curve (zero to infinity) was increased by 180% and mean residence time was prolonged by 180% during hypothermia compared with the corresponding values after hypothermia. Moreover, the elimination constant (ke) was decreased by 50% and elimination clearance of phenytoin was decreased by 67% during hypothermia compared with the corresponding values after hypothermia. The plasma concentration of 5-p-HPPH was significantly lower during hypothermia than after hypothermia. These findings suggest that phenytoin metabolism is inhibited by mild therapeutic hypothermia.
Collapse
Affiliation(s)
- Y Iida
- Department of Anesthesiology & Intensive Care Medicine, Osaka City University Medical School, Osaka, Japan
| | | | | |
Collapse
|
41
|
Barcelona SL, Coté CJ. Pediatric resuscitation in the operating room. ANESTHESIOLOGY CLINICS OF NORTH AMERICA 2001; 19:339-65. [PMID: 11469068 DOI: 10.1016/s0889-8537(05)70232-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The resuscitation of pediatric patients undergoing anesthesia involves appropriate administration of fluid and blood products and stabilization of vital signs. Crystalloid is first-line therapy for fluid resuscitation, and should be given with awareness of its potential dilution of the child's hematocrit. Many alternatives to homologous blood transfusions now exist, however, when necessary for increasing oxygen-carrying capacity or treating coagulopathy benefits likely outweight the risks. The risks for such transfusion include infectious, hemolytic, metabolic, and immunologic effects. When fluid and blood administration does not stabilize the patient, the differential diagnosis of hypotension, arrest, or arrhythmias must include medication errors, anesthetic overdose, electrolyte disturbances, hypoxemia, ventilatory problems, and surgical insults, including medications given in the operative field. Resuscitation should include treatment of hypocalcemia and hyperkalemia, chest compressions, and the administration of epinephrine when necessary.
Collapse
Affiliation(s)
- S L Barcelona
- Department of Pediatric Anesthesiology, Children's Memorial Medical Center, Northwestern University Medical School, Chicago, Illinois, USA
| | | |
Collapse
|
42
|
Glauser TA, Pippenger CE. Controversies in blood-level monitoring: reexamining its role in the treatment of epilepsy. Epilepsia 2001; 41 Suppl 8:S6-15. [PMID: 11092608 DOI: 10.1111/j.1528-1157.2000.tb02950.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This article reexamines the role of blood-level monitoring (therapeutic drug monitoring, TDM) of antiepileptic drugs (AEDs) in the current treatment of epilepsy and identifies situations in which TDM can be useful. Basic pharmacokinetic and pharmacodynamic principles are reviewed, with specific emphasis on kinetics of absorption/distribution/metabolism, elimination half-life, time to steady state, and plasma drug concentrations. The relationship between AED intensity of effect (pharmacodynamics) and plasma concentration (pharmacokinetics) is expressed mathematically, examined in the context of the major old and new AEDs, and integrated with a historical look at the role of TDM. Situations in which TDM can be useful in the modern treatment of epilepsy are presented and discussed. For both older and newer AEDs, TDM is useful in six clinical situations: establishing "baseline" effective concentrations, evaluating potential causes for lack of efficacy, evaluating potential causes for toxicity, evaluating potential causes for loss of efficacy, judging "room to move" or when to change AEDs, and minimizing predictable problems. TDM remains a valuable tool in the modern treatment of epilepsy. It can be selectively and appropriately utilized to help maximize seizure control and minimize side effects if levels are obtained in response to a patient-specific pharmacokinetic or pharmacodynamic issue or problem.
Collapse
Affiliation(s)
- T A Glauser
- Department of Neurology, Children's Hospital Medical Center, Cincinnati, Ohio 45229-3039, USA
| | | |
Collapse
|
43
|
Abstract
Preservation or restoration of optimal neurologic function following traumatic brain injury (TBI) requires timely and aggressive therapeutic interventions. Effective diagnostic tools, together with an armamentarium of treatment modalities, have augmented the treatment strategies utilized today. In addition, the Guidelinesfor the Management of Severe Head Injury have established a standardized approach for the TBI patient. This article will provide current information regarding the resuscitation priorities, appropriate interventions, and pharmacological agents used in the treatment required by the complex nature of TBI. Also, a review of the occurrences associated with TBI will be discussed.
Collapse
Affiliation(s)
- J R Yanko
- Allegheny General Hospital, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
44
|
Burt M, Anderson DC, Kloss J, Apple FS. Evidence-based Implementation of Free Phenytoin Therapeutic Drug Monitoring. Clin Chem 2000. [DOI: 10.1093/clinchem/46.8.1132] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AbstractBackground: The majority of laboratories measure total phenytoin concentration for therapeutic drug monitoring. However, there are substantial interindividual variations in free phenytoin concentrations, the pharmacologically active component.Methods: We describe the process and data used to implement monitoring of free phenytoin only in an urban medical center. Over a 6-week period, total and free phenytoin concentrations were measured, clinical charts reviewed, and indications for alterations in the percentage of free phenytoin fraction were determined.Results: Of the 189 phenytoin requests from 139 patients, 136 data points were analyzed. Free phenytoin concentrations were 6.8–35.3%, with 50% outside the expected range of 8–12%. Clinical indications likely responsible for variations were hypoalbuminemia, drug interactions, uremia, pregnancy, and age. Overall, 30% of patients demonstrated a discrepancy between therapeutic, subtherapeutic, or supratherapeutic concentrations between free and total phenytoin concentrations. The largest discordance (53%) occurred in the patient group with free phenytoin <8% or >12%.Conclusions: This study supports previous clinical findings that monitoring total phenytoin is not as reliable as free phenytoin as a clinical indicator for therapeutic and nontherapeutic concentrations. Thus, we recommend that therapeutic monitoring should use free phenytoin concentrations only.
Collapse
Affiliation(s)
- Martha Burt
- Departments of Laboratory Medicine and Pathology and Neurology, Hennepin County Medical Center, University of Minnesota School of Medicine, Minneapolis, MN 55415
| | - David C Anderson
- Departments of Laboratory Medicine and Pathology and Neurology, Hennepin County Medical Center, University of Minnesota School of Medicine, Minneapolis, MN 55415
| | - Julie Kloss
- Departments of Laboratory Medicine and Pathology and Neurology, Hennepin County Medical Center, University of Minnesota School of Medicine, Minneapolis, MN 55415
| | - Fred S Apple
- Departments of Laboratory Medicine and Pathology and Neurology, Hennepin County Medical Center, University of Minnesota School of Medicine, Minneapolis, MN 55415
| |
Collapse
|