Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
3
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found

      Association of Piriform Cortex Resection With Surgical Outcomes in Patients With Temporal Lobe Epilepsy

      1 , 2 , 3 , 1 , 1 , 1 , 4 , 5 , 1 , 1 , 2 , 6 , 1 , 1 , 7 , 1 , 1 , 8 , 9 , 2 , 8 , 9 , 1 , 2 , 8 , 9 , 10 , 8 , 11 , 1 , 1 , 1 , 1 , 2 , 12 , 13 , 12 , 13 , 14 , 12 , 15 , 15 , 16 , 15 , 16 , 17 , 8 , 9 , 10 , 18 , 19 , 20 , 21 , 1 , 2 , 22 , 23 , 1 , 2 , 1 , 2
      JAMA Neurology
      American Medical Association (AMA)

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Does resecting the piriform cortex improve surgical outcome in temporal lobe epilepsy? In this multicenter study that included 107 adults with temporal lobe epilepsy in the derivation cohort and 31 in the validation cohort, resecting a larger proportion of the piriform cortex (78% in seizure-free vs 46% in non–seizure-free cases in pooled data) was significantly associated with a favorable outcome. Removal of at least half the piriform cortex significantly increased the odds of becoming seizure free by a factor of 16. These findings support including the piriform cortex in standard anterior temporal lobe resections to achieve seizure freedom. This multicenter cohort study assesses outcomes of piriform cortex resection among patients with temporal lobe epilepsy. A functional area associated with the piriform cortex, termed area tempestas , has been implicated in animal studies as having a crucial role in modulating seizures, but similar evidence is limited in humans. To assess whether removal of the piriform cortex is associated with postoperative seizure freedom in patients with temporal lobe epilepsy (TLE) as a proof-of-concept for the relevance of this area in human TLE. This cohort study used voxel-based morphometry and volumetry to assess differences in structural magnetic resonance imaging (MRI) scans in consecutive patients with TLE who underwent epilepsy surgery in a single center from January 1, 2005, through December 31, 2013. Participants underwent presurgical and postsurgical structural MRI and had at least 2 years of postoperative follow-up (median, 5 years; range, 2-11 years). Patients with MRI of insufficient quality were excluded. Findings were validated in 2 independent cohorts from tertiary epilepsy surgery centers. Study follow-up was completed on September 23, 2016, and data were analyzed from September 24, 2016, through April 24, 2018. Standard anterior temporal lobe resection. Long-term postoperative seizure freedom. In total, 107 patients with unilateral TLE (left-sided in 68; 63.6% women; median age, 37 years [interquartile range {IQR}, 30-45 years]) were included in the derivation cohort. Reduced postsurgical gray matter volumes were found in the ipsilateral piriform cortex in the postoperative seizure-free group (n = 46) compared with the non–seizure-free group (n = 61). A larger proportion of the piriform cortex was resected in the seizure-free compared with the non–seizure-free groups (median, 83% [IQR, 64%-91%] vs 52% [IQR, 32%-70%]; P  < .001). The results were seen in left- and right-sided TLE and after adjusting for clinical variables, presurgical gray matter alterations, presurgical hippocampal volumes, and the proportion of white matter tract disconnection. Findings were externally validated in 2 independent cohorts (31 patients; left-sided TLE in 14; 54.8% women; median age, 41 years [IQR, 31-46 years]). The resected proportion of the piriform cortex was individually associated with seizure outcome after surgery (derivation cohort area under the curve, 0.80 [ P  < .001]; external validation cohorts area under the curve, 0.89 [ P  < .001]). Removal of at least half of the piriform cortex increased the odds of becoming seizure free by a factor of 16 (95% CI, 5-47; P  < .001). Other mesiotemporal structures (ie, hippocampus, amygdala, and entorhinal cortex) and the overall resection volume were not associated with outcomes. These results support the importance of resecting the piriform cortex in neurosurgical treatment of TLE and suggest that this area has a key role in seizure generation.

          Related collections

          Most cited references22

          • Record: found
          • Abstract: found
          • Article: not found

          Central olfactory connections in the macaque monkey.

          The connections between the olfactory bulb, primary olfactory cortex, and olfactory related areas of the orbital cortex were defined in macaque monkeys with a combination of anterograde and retrograde axonal tracers and electrophysiological recording. Anterograde tracers placed into the olfactory bulb labeled axons in eight primary olfactory cortical areas: the anterior olfactory nucleus, piriform cortex, ventral tenia tecta, olfactory tubercle, anterior cortical nucleus of the amygdala, periamygdaloid cortex, and olfactory division of the entorhinal cortex. The bulbar axons terminate in the outer part of layer I throughout these areas and are most dense in areas that are close to the lateral olfactory tract. Labeled axons also were found in the superficial part of nucleus of the horizontal diagonal band. Retrograde tracers injected into the olfactory bulb labeled cells in the nucleus of the diagonal band and in all of the primary olfactory cortical areas except the olfactory tubercle. Electrical stimulation of the olfactory bulb evoked short-latency unit responses and a characteristic field wave in the primary olfactory cortex. Multiunit activity in layer II tended to be of shorter latency than that in layer III and the endopiriform nucleus. Associational connections within the primary olfactory cortex were demonstrated with anterograde tracer injections into the piriform cortex and the entorhinal cortex. Injections into the piriform cortex near the lateral olfactory tract labeled axons in the deep part of layer I of many primary olfactory areas, but especially in areas near the tract. An injection into the rostral entorhinal cortex, distant to the lateral olfactory tract, labeled a complementary distribution of axons in deep layer I of olfactory areas medial and caudoventral to the tract. This organization resembles that reported in the primary olfactory cortex of the rat [Luskin and Price (1983) J. Comp. Neurol. 216:264-291]. The anterograde tracer injections into the piriform cortex and retrograde tracer injections into the orbital and medial prefrontal cortex and rostral insula label connections from the primary olfactory cortex to nine areas in the caudal orbital cortex, including the agranular insula areas Iam, Iai, Ial, Iapm, and Iapl and areas 14c, 25, 13a, and 13m. The piriform cortex projects most heavily to layer I of these areas. Only Iam, Iapm, and 13a receive a substantial projection to the deeper layers. Areas Iam, Iapm, and 13a were also the only areas that responded with multiunit action potentials to olfactory bulb stimulation in anesthetized animals.(ABSTRACT TRUNCATED AT 400 WORDS)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Temporal patterns and mechanisms of epilepsy surgery failure.

            Epilepsy surgery is an accepted treatment option in patients with medically refractory focal epilepsy. Despite various advances in recording and localization noninvasive and invasive techniques (including electroencephalography (EEG), magnetic resonance imaging (MRI), positron emission tomography (PET), single photon emission computed tomography (SPECT), magnetoencephalography (MEG), subdural grids, depth electrodes, and so on), the seizure outcome following surgical resection remains suboptimal in a significant number of patients. The availability of long-term outcome data on an increasing number of patients suggests two major temporal patterns of seizure recurrence (early vs. late) that implicate the following two different mechanisms for seizure recurrence: (1) a failure to either define/resect the epileptogenic zone, and (2) the nonstatic nature of epilepsy as a disease through the persistence of proepileptic cortical pathology. We describe the temporal patterns of epilepsy surgery failures and discuss their potential clinical, histopathologic, genetic, and molecular mechanisms. In addition, we review predictors of successful surgical interventions and analyze the natural history of epilepsy following surgical intervention. We hypothesize that the acute/early postoperative failures are due to errors in localizing and/or resecting the epileptic focus, whereas late recurrences are likely due to development/maturation of a new and active epileptic focus (de novo epileptogenesis). Wiley Periodicals, Inc. © 2013 International League Against Epilepsy.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The role of the piriform cortex in kindling.

              In epilepsy research, there is growing interest in the role of the piriform cortex (PC) in the development and maintenance of limbic kindling and other types of limbic epileptogenesis leading to complex partial seizures, i.e. the most common type of seizures in human epilepsy. The PC ("primary olfactory cortex") is the largest area of the mammalian olfactory cortex and receives direct projections from the olfactory bulb via the lateral olfactory tract (LOT). Beside the obvious involvement in olfactory perception and discrimination, the PC, because of its unique intrinsic associative fiber system and its various connections to and from other limbic nuclei, has been implicated in the study of memory processing, spread of excitatory waves, and in the study of brain disorders such as epilepsy with particular emphasis on the kindling model of temporal lobe epilepsy with complex partial seizures. The interest in the kindling model is based primarily on the following observations. (1) The PC contains the most susceptible neural circuits of all forebrain regions for electrical (or chemical) induction of limbic seizures. (2) During electrical stimulation of other limbic brain regions, broad and large afterdischarges can be observed in the ipsilateral PC, indicating that the PC is activated early during the kindling process. (3) The interictal discharge, which many consider to be the hallmark of epilepsy, originates in the PC, independent of which structure serves as the kindled focus. (4) Autoradiographic studies of cerebral metabolism in rat amygdala kindling show that, during focal seizures, the area which exhibits the most consistent increase in glucose utilization is the ipsilateral paleocortex, particularly the PC. (5) During the commonly short initial afterdischarges induced by stimulation of the amygdala at the early stages of kindling, the PC is the first region that exhibits induction of immediate-early genes, such as c-fos. (6) The PC is the most sensitive brain structure to brain damage by continuous or frequent stimulation of the amygdala or hippocampus. (7) Amygdala kindling leads to a circumscribed loss of GABAergic neurons in the ipsilateral PC, which is likely to explain the increase in excitability of PC pyramidal neurons during kindling. (8) Kindling of the amygdala or hippocampus induces astrogliosis in the PC, indicating neuronal death in this brain region. Furthermore, activation of microglia is seen in the PC after amygdala kindling. (9) Complete bilateral lesions of the PC block the generalization of seizures upon kindling from the hippocampus or olfactory bulb. Incomplete or unilateral lesions are less effective in this regard, but large unilateral lesions of the PC and adjacent endopiriform nucleus markedly increase the threshold for induction of focal seizures from stimulation of the basolateral amygdala (BLA) prior to and after kindling, indicating that the PC critically contributes to regulation of excitability in the amygdala. (10) Potentiation of GABAergic neurotransmission in the PC markedly increases the threshold for induction of kindled seizures via stimulation of the BLA, again indicating a critical role of the PC in regulation of seizure susceptibility of the amygdala. Microinjections of NMDA antagonists or sodium channel blockers into the PC block seizure generalization during kindling development. (11) Neurophysiological studies on the amygdala-PC slice preparation from kindled rats showed that kindling of the amygdala induces long-lasting changes in synaptic efficacy in the ipsilateral PC, including spontaneous discharges and enhanced susceptibility to evoked burst responses. The epileptiform potentials in PC slice preparations from kindled rats seem to originate in neuron at the deep boundary of PC. Spontaneous firing and enhanced excitability of PC neurons in response to kindling from other sites is also seen in vivo, substantiating the fact that kindling induces long-lasting changes in the PC c
                Bookmark

                Author and article information

                Journal
                JAMA Neurology
                JAMA Neurol
                American Medical Association (AMA)
                2168-6149
                June 01 2019
                June 01 2019
                : 76
                : 6
                : 690
                Affiliations
                [1 ]UK National Institute for Health Research, University College London (UCL) Hospitals Biomedical Research Centre, Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
                [2 ]Epilepsy Society MRI Unit, Epilepsy Society, Chalfont St Peter, United Kingdom
                [3 ]Department of Neurology, Kantonsspital St Gallen, St Gallen, Switzerland
                [4 ]Department of Imaging, Hospital de Especialidades Eugenio Espejo, Quito, Ecuador
                [5 ]Division of Neuroanatomy, Facultad de Medicina, Universidad Internacional del Ecuador, Quito
                [6 ]Magnetic Resonance Imaging Unit, Klinik Mara, Bethel Epilepsy Centre, Bielefeld, Germany
                [7 ]Institute of Cognitive Neuroscience, UCL, London, United Kingdom
                [8 ]Translational Imaging Group, Centre for Medical Image Computing, Department of Medical Physics and Bioengineering, UCL, London, United Kingdom
                [9 ]Wellcome EPSRC Centre for Interventional and Surgical Sciences, UCL, London, United Kingdom
                [10 ]School of Biomedical Engineering and Image Sciences, Kings College London, London, United Kingdom
                [11 ]Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, London, United Kingdom
                [12 ]Department of Neurosurgery, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania
                [13 ]Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
                [14 ]medical student at Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
                [15 ]Center for Neuroengineering and Therapeutics, Department of Bioengineering, University of Pennsylvania, Philadelphia
                [16 ]Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia
                [17 ]Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
                [18 ]Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
                [19 ]Laboratory of Developmental Epilepsy, Saul R. Korey Department of Neurology, Montefiore/Einstein Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
                [20 ]Dominick P. Purpura Department of Neuroscience, Montefiore/Einstein Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
                [21 ]Department of Pediatrics, Montefiore/Einstein Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
                [22 ]Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
                [23 ]Center for Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
                Article
                10.1001/jamaneurol.2019.0204
                6490233
                30855662
                d4df91da-0b15-4081-9594-7c3b2883ce5c
                © 2019
                History

                Comments

                Comment on this article