Spinal cord blood flow and ischemic injury after experimental sacrifice of thoracic and abdominal segmental arteries

Similar documents
Experimental Two-Stage Simulated Repair of Extensive Thoracoabdominal Aneurysms Reduces Paraplegia Risk

Daniela Branzan MD, Department of Vascular Surgery and Department of Interventional Angiology University Hospital Leipzig

Paraplegia in endovascular repair of TAA and in TEVAR: Incidence, prevention and therapy. Johannes Lammer Medical University Vienna, Austria

Neuromonitor-guided repair of thoracoabdominal aortic aneurysms

Replacement of the thoracoabdominal aorta for extensive

Neurological Complications of TEVAR. Frank J Criado, MD. Union Memorial-MedStar Health Baltimore, MD USA

Accepted Manuscript. Perioperative renal function and thoracoabdominal aneurysm repair: Where do we go from here? Leonard N. Girardi, M.D.

Thoracoabdominal Aorta: Advances and Novel Therapies

Combination of Myogenic and Neurogenic Motor Evoked Potential Monitoring During Thoracoabdominal Aortic Surgery

Assessment of Spinal Cord Circulation and Function in Endovascular Treatment of Thoracic Aortic Aneurysms

Aortic Arch/ Thoracoabdominal Aortic Replacement

Spinal cord protection segmental artery embolization. Christian D. Etz, MD, PhD Heisenberg Professor for Aortic Surgery

How to manage the left subclavian and left vertebral artery during TEVAR

Spinal cord ischemia in thoracoabdominal aneurysm surgery: monitoring and conditioning the spinal cord de Haan, P.

Selective Visceral Perfusion during Thoracoabdominal Aortic Aneurysm Repair

Dr Brigitta Brandner UCLH

Current strategies to prevent spinal cord ischemia in TAAA repair

ORIGINAL ARTICLE. Systemic Temperature and Paralysis After Thoracoabdominal and Descending Aortic Operations

Kopp R, Puippe G, Rancic Z, Hofmann M, Pecoraro F, Pfammatter T, Lachat M.. University Hospital Zurich, Switzerland

Thoracic and Thoracoabdominal Aneurysm Repair: Is Reimplantation of Spinal Cord Arteries a Waste of Time?

What is the benefit. of MEP s in BEVAR for TAAA. in preventing paraplegia?

Intra-operative neurologic injuries: Avoidance and prompt response

Lumbar Drain Management Thoracic Aortic Aneurysm Surgery

Open surgical repair of thoracoabdominal aneurysms - the Massachusetts General Hospital experience

Thoracoabdominal aortic aneurysms by definition traverse

Management of Bone and Spinal Cord in Spinal Surgery.

Lumbar CSF Drains for Thoracic Aortic Surgery

Postoperative risk factors for delayed neurologic deficit after thoracic and thoracoabdominal aortic aneurysm repair: A case-control study

SPINAL CORD ISCHEMIA AFTER THORACIC ANEURYSM REPAIR: RISK STRATIFICATION & PREVENTION DISCLOSURES. INDIVIDUAL None

Evolving Strategy and Results of Spinal Cord Protection in Type I and II Thoracoabdominal Aortic Aneurysm Repair

Open fenestration for complicated acute aortic B dissection

Intraoperative spinal cord monitoring (IOM) during surgery

Early- and medium-term results after aortic arch replacement with frozen elephant trunk techniques a single center study

Influence of Perioperative Hemodynamics on Spinal Cord Ischemia in Thoracoabdominal Aortic Repair

SELECTIVE ANTEGRADE TECHNIQUE OF CHOICE

Preoperative and operative predictors of delayed neurologic deficit following repair of thoracoabdominal aortic aneurysm

2012 What is New in Aortic Surgery: Monitoring and Preventing Spinal Cord Injuries - Teamwork

Combined use of an epidural cooling catheter and systemic moderate hypothermia enhances spinal cord protection against ischemic injury in rabbits

Hybrid Repair of a Complex Thoracoabdominal Aortic Aneurysm

The SPIDER-Graft for Thoracoabdominal Aortic Repair a feasability study in pigs

Descending aorta replacement through median sternotomy

Mortality and Paraplegia After Thoracoabdominal Aortic Aneurysm Repair: A Risk Factor Analysis

Gelweave TM. Thoracic and Thoracoabdominal Graft Geometries. Ante-Flo TM 4 Branch Plexus. Siena Valsalva TM Trifurcate Arch Graft. Coselli.

Measurement of spinal cord blood flow by an inhalation method and intraarterial injection of hydrogen gas

Paraplegia has been a serious and recalcitrant problem. Cooling catheter for spinal cord preservation in thoracic aortic surgery

Thoracoabdominal aortic replacement for Crawford extent II aneurysm after thoracic endovascular aortic repair

Neurologic improvement after thoracic, thoracolumbar, and lumbar spinal cord (conus medullaris) injuries

Anatomical Study of Blood Supply to the Spinal Cord

SEP Monitoring. Andres A Gonzalez, MD Director, Surgical Neurophysiology Keck Medical Center of USC University of Southern California

Table I. Associated diseases

Extent of Aortic Coverage and Incidence of Spinal Cord Ischemia After Thoracic Endovascular Aneurysm Repair

Pulmonary Complications After Descending Thoracic and Thoracoabdominal Aortic Aneurysm Repair: Predictors, Prevention, and Treatment

Acute dissections of the descending thoracic aorta (Debakey

Total arch replacement with separated graft technique and selective antegrade cerebral perfusion

SEP Monitoring. Outline. Outline 1/22/2015. Development of SEPs Stimulation and recording techniques Predictive value of SEP Uses of SEP monitoring

NIH Public Access Author Manuscript J Vasc Surg. Author manuscript; available in PMC 2011 January 1.

Protecting the brain and spinal cord in aortic arch surgery

Thoracic aortic aneurysms are life threatening and

Cold blood spinoplegia under motor-evoked potential monitoring during thoracic aortic surgery

Challenges. 1. Sizing. 2. Proximal landing zone 3. Distal landing zone 4. Access vessels 5. Spinal cord ischemia 6. Endoleak

Treatment of Thoracoabdominal Aneurysms Is there a need for custom-made devices?

Spinal injury. Structure of the spine

Thoracoabdominal aortic aneurysm

Combined Endovascular and Surgical Repair of Thoracoabdominal Aortic Pathology: Hybrid TEVAR

Open repair of Abdominal Aortic Aneurysms (AAA)

Audit and Compliance Department 1

Epidural Versus Subdural Spinal Cord Cooling: Cerebrospinal Fluid Temperature and Pressure Changes

Influence of segmental arteries, extent, and atriofemoral bypass on postoperative paraplegia after thoracoabdominal aortic operations

Deliberate Renal Ischemia

Aggressive Resection/Reconstruction of the Aortic Arch in Type A Dissection

NVM5 NERVE MONITORING SYSTEM AN INTRODUCTION TO

Airway Management in a Patient with Klippel-Feil Syndrome Using Extracorporeal Membrane Oxygenator

Reinhard Kopp, Karin Pfister, Beatrix Cucuruz, Konstantinos Gallis, Piotr M Kasprzak

TAAA / Spinal Cord Protection

CORONARY ARTERY BYPASS GRAFTING (CABG) (Part 1) Mark Shikhman, MD, Ph.D., CSA Andrea Scott, CST

Intended Learning Outcomes

Major Aortic Reconstruction; Cerebral protection and Monitoring

THE EFFECT OF RETROGRADE CEREBRAL PERFUSION AFTER PARTICULATE EMBOLIZATION TO THE BRAIN

IOM at University of. Training for physicians. art of IOM. neurologic. injury during surgery. surgery on by IOM. that rate is.

Influence of segmental spinal cord perfusion on intrathecal oxygen tension during experimental thoracic aortic crossclamping

THORACOABDOMINAL AORTIC ANEURYSMS HYBRID REPAIR

3/6/2017. Endovascular Selective Cerebral Hypothermia First-in-Human Experience

Dissection: The Fetal Pig

Management of Acute Aortic Syndromes. M. Grabenwoger, MD Dept. of Cardiovascular Surgery Hospital Hietzing, Vienna, Austria

A Patient s Guide to Intraoperative Monitoring

Leo Happel, PhD Professor, Neurology, Neurosurgery, Physiology, and Neuroscience LSU Health Science Center

Tripler Army Medical Center Obstetric Anesthesia Service - FAQs

Toward Total Endovascular Therapy of the Aorta. Adam W. Beck, MD. Associate Professor of Surgery Division of Vascular Surgery and Endovascular Therapy

Percutaneous Approaches to Aortic Disease in 2018

Descending Thoracic Aortic Aneurysm: Surgical Approach and Treatment Using the Adjuncts Cerebrospinal Fluid Drainage and Distal Aortic Perfusion

I-Hui Wu, M.D. Ph.D. Clinical Assistant Professor Cardiovascular Surgical Department National Taiwan University Hospital

University of Florida Department of Surgery. CardioThoracic Surgery VA Learning Objectives

Femoral Versus Aortic Cannulation for Surgery of Chronic Ascending Aortic Aneurysm

POSITION STATEMENT. Abstract BACKGROUND

Cardiopulmonary Bypass for Thoracic Aortic Aneurysm: A Report on 488 Cases

Disease of the aortic valve is frequently associated with

Index. Note: Page numbers of article titles are in boldface type.

AORTIC DISSECTIONS Current Management. TOMAS D. MARTIN, MD, LAT Professor, TCV Surgery Director UF Health Aortic Disease Center University of Florida

Hypothermic cardiopulmonary bypass with intervals

Free Esophageal Perforation Following Hybrid Visceral Debranching and Distal Endograft Extension to Repair a Ruptured Thoracoabdominal Aortic

Transcription:

European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 www.elsevier.com/locate/ejcts Spinal cord blood flow and ischemic injury after experimental sacrifice of thoracic and abdominal segmental arteries Abstract Christian D. Etz a, *, Tobias M. Homann a, Maximilian Luehr a, Fabian A. Kari a, Donald J. Weisz b,c, George Kleinman c, Konstadinos A. Plestis a, Randall B. Griepp a a Department of Cardiothoracic Surgery, Mount Sinai School of Medicine, New York, NY, USA b Department of Neurophysiology, Mount Sinai School of Medicine, New York, NY, USA c Department of Neuropathology, Mount Sinai School of Medicine, New York, NY, USA Received 4 September 2007; received in revised form 23 January 2008; accepted 28 January 2008; Available online 11 April 2008 Objective: Spinal cord blood flow (SCBF) after sacrifice of thoracoabdominal aortic segmental arteries (TAASA) during thoracoabdominal aortic aneurysm (TAAA) repair remains poorly understood. This study explored SCBF for 72 h after sacrifice of all TAASA. Methods: Fourteen juvenile Yorkshire pigs underwent complete serial TAASA sacrifice (T4 L5). Six control pigs underwent anesthesia and cooling to 32 8C with no TAASA sacrifice. In the experimental animals, spinal cord function was continuously monitored using motor evoked potentials (MEPs) until 1 h after clamping the last TAASA. Fluorescent microspheres enabled segmental measurement of SCBF along the entire spinal cord before, and 5 min, 1 h, 5 h, 24 h and 72 h after complete TAASA sacrifice. A modified Tarlov score was obtained for 3 days after surgery. Results: All the pigs with complete TAASA sacrifice retained normal cord function (MEP) until 1 h after TAASA ligation. Seven pigs (50%) with complete TAASA sacrifice recovered after 72 h; seven pigs suffered paraparesis or paraplegia. Intraoperatively, and until 1 h postoperatively, SCBF was similar among the three groups along the entire cord. Postoperatively, SCBF did not decrease in any group, but significant hyperemia occurred at 5 h in controls and recovery animals, but did not occur in pigs that developed paraparesis or paraplegia in the T8 L2 segments ( p = 0.0002) and L3 S segments ( p = 0.0007). At 24 h, SCBF remained marginally lower from T8 caudally; at 72 h, SCBF was similar among all groups along the entire cord. SCBF in the segments T8 L2 at 5 h predicted functional recovery ( p = 0.003). Conclusions: This study suggests that critical spinal cord ischemia after complete TAASA sacrifice does not occur immediately (intraoperatively), but is delayed 1 5 h or longer after clamping, and represents failure to mount a hyperemic response to rewarming and awakening. The short duration of low SCBF associated with spinal cord injury suggests that hemodynamic and metabolic manipulation lasting only 24 72 h may allow routine preservation of normal cord function despite sacrifice of all TAASA secondary to surgical or endovascular repair of large TAAA. # 2008 European Association for Cardio-Thoracic Surgery. Published by Elsevier B.V. All rights reserved. Keywords: Spinal cord perfusion/protection; Paraparesis/paraplegia; Segmental artery sacrifice; Thoracoabdominal aortic aneurysm repair (TAA/A) 1. Introduction The mortality and morbidity of even extensive thoracoabdominal replacement has improved markedly in recent years [1]. However, postoperative paraplegia remains a devastating albeit rare complication often associated with significantly reduced long-term survival and its occurrence is still somewhat unpredictable [2 6]. Most often, neurologic injury becomes apparent immediately postoperatively and is therefore assumed to be a consequence of ischemic injury during intraoperative aortic cross-clamping, or sacrifice of vessels critical to spinal cord Presented at the 21st Annual Meeting of the European Association for Cardio-thoracic Surgery, Geneva, Switzerland, September 16 19, 2007. * Corresponding author. Address: Mount Sinai School of Medicine, Department of Cardiothoracic Surgery, One Gustave L. Levy Place, PO Box 1028, New York, NY 10029, USA. Tel.: +1 212 659 6800; fax: +1 212 659 6818. E-mail address: christian.etz@mountsinai.org (C.D. Etz). function during operative repair. In recent years, various strategies to support the spinal cord circulation during aneurysm resection, often accompanied by intraoperative motor evoked potential (MEP) and somatosensory evoked potential (SSEP) monitoring, have become important features of aortic aneurysm surgery and have contributed to a reduction of permanent postoperative spinal cord injury to below 3% in experienced centers [7]. The studies described in this report were undertaken to try to gain a better understanding of the impact of extensive sacrifice of segmental arteries (SA) arising from the thoracic and abdominal aorta (TAASA) on spinal cord blood flow (SCBF) intraoperatively and in the early postoperative period under experimental conditions which approximate the circumstances prevailing during clinical thoracoabdominal aortic surgery. Previous studies with this model have established the feasibility of routine extensive SA sacrifice without loss of function, revealing the presence of a dense and complex 1010-7940/$ see front matter # 2008 European Association for Cardio-Thoracic Surgery. Published by Elsevier B.V. All rights reserved. doi:10.1016/j.ejcts.2008.01.069

C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 1031 collateral arterial network feeding the spinal cord [8 10]. Recent studies have revealed that spinal cord perfusion pressure (SCPP) is significantly but transiently reduced for at least several hours after SA sacrifice in this model, potentially causing a critical, albeit temporary, reduction in spinal cord blood flow (SCBF) [11]. Further investigation of the physiological and functional response of the collateral spinal cord perfusion network to the sacrifice of important contributors to its blood supply, and especially of the time course of this response, should help to elucidate how best to prevent even the rare occurrence of paraplegia after extensive SA sacrifice. Avoiding spinal cord injury despite occlusion of most SAs is critical not only for surgical repair but for the eventual successful endovascular treatment of large thoracoabdominal aneurysms. 2. Materials and methods 2.1. Study design Twenty female juvenile Yorkshire pigs (Animal Biotech Industries, Allentown, NJ, USA), 4 5 months of age, weighing 28 32 kg, were used for this experiment. Fourteen pigs underwent complete serial TAASA sacrifice. In all animals, the descending thoracic and abdominal aorta were exposed, and all TAASA (T4 13, L1 4/5) carefully dissected. Thereafter, all thoracic and abdominal segmental arteries were sequentially clamped along the craniocaudal axis during mild hypothermia (32 8C), allowing a 3-min interval between clamping of successive arteries. Spinal cord function was continuously monitored using MEPs until 1 h after clamping the last TAASA. Six control pigs underwent anesthesia and cooling to 32 8C with exposure of the segmental vessels but no TAASA sacrifice. Fluorescent microspheres enabled segmental measurement of SCBF along the entire spinal cord before, and 5 min, 1 h, 5 h, 24 h and 72 h after complete aortic TAASA sacrifice. Functional recovery was evaluated for 5 days using a modification of the Tarlov score, and histopathological examination was carried out after sacrifice. Pigs that died before completion of the protocol were replaced. This experimental model closely simulates the procedure used for resection of descending thoracic and thoracoabdominal aneurysms clinically at our institution. However, it should be noted that the anatomy of the pig differs from that of humans in having 13 thoracic (and 5 lumbar) segmental arteries that arise together from the descending (abdominal) aorta and subsequently divide. Recent studies suggest the subclavian arteries and the median sacral arteries play a major role in the perfusion of the paraspinous collateral vascular network in both species, although the iliac arteries may provide a greater proportion of direct blood supply in humans than in pigs. This experimental model is simplified to allow an uncomplicated focus on the input to the collateral network in the wake of segmental artery sacrifice. Previous experiments with this model have demonstrated that the spinal cord perfusion pressure and the collateral flow in the pig behave in ways very similar to what is observed under comparable circumstances clinically in humans. 2.2. Perioperative management and anesthesia All animals received humane care in compliance with the guidelines of Principles of Laboratory Animal Care formulated by the National Society for Medical Research and the Guide for the Care and Use of Laboratory Animals published by the National Institute of Health (NIH Publication No. 88-23, revised 1996). The Mount Sinai Institutional Animal Care and Use Committee approved the protocols for all experiments. After pretreatment with intramuscular ketamine (15 mg/ kg) and atropine (0.03 mg/kg), an endotracheal tube is placed. The animals are then transferred to the operating room and are mechanically ventilated with a FiO 2 of 0.5, and a minute volume adequate for maintenance of a normal pco 2 (35 40 mmhg). Anesthesia is induced via the bolus intravenous administration of propofol (1 mg/kg) and fentanyl (50 mg/kg) and is maintained with infusions of ketamine (15 mg/kg/h), propofol (7 mg/kg/h) and fentanyl (5 mg/kg/h). This anesthetic regimen has no major effect on MEP responses, and has been described previously [12]. Paralysis for intubation is achieved with intravenous pancuronium (0.1 mg/kg), but no further doses are administered subsequently to avoid interfering with measurement of MEPs. The ventilator rate and the tidal volume are adjusted to maintain the arterial carbon dioxide tension at 35 40 mmhg. End-expiratory carbon dioxide (PPG Biomedical Systems, Model 2010-200 R, Lenexa, KS, USA) is monitored continuously. Arterial oxygen tension is maintained >90 mmhg. A bladder catheter (Foley 8 10 F) is inserted for online measurement of urine output. Electrocardiographic measurements are recorded continuously. An arterial line is placed in the right brachial artery for pressure monitoring and blood sampling (ph, oxygen tension, carbon dioxide tension, oxygen saturation, base excess, hematocrit, hemoglobin and glucose, lactate, Blood Gas Analyzer, Ciba Corning 865, Chiron Diagnostics, Norwood, MA, USA). 2.3. Body temperature management After inducing anesthesia, the pigs are cooled to 32 8C rectal temperature by covering them with packs of artificial refrigerants for a period of 30 min. In addition, a cooling blanket is used even after the target temperature has been reached to maintain hypothermia and prevent an upward temperature drift during the procedure. The operating room temperature is reduced to 14 8C. No local cooling of the vertebral column is undertaken. The animals are subsequently warmed using a heating blanket and a heating lamp, usually for 90 100 min, and by raising the operating room temperature to 24 8C. To prevent any intraoperative temperature drift, the small left thoracotomy in the fourth intercostal space is temporarily closed after clamping the thoracic spinal arteries. 2.4. Monitoring of intra- and postoperative systemic perfusion pressure (MAP) Two arterial pressure lines are placed: one in the right brachial artery to monitor intraoperatively and another in the descending aorta, for direct postoperative aortic pressure

1032 C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 monitoring. These lines enable systemic perfusion pressure monitoring, blood sampling (ph, oxygen tension, carbon dioxide tension, oxygen saturation, base excess, hematocrit, hemoglobin and glucose, lactate; blood gas analyzer, Ciba Corning 865, Chiron Diagnostics, Norwood, MA, USA) as well as reference sampling for microsphere measurements prior to, during, and after complete TAASA sacrifice. 2.5. Spinal cord blood flow (SCBF) measurements microsphere injections In all animals, the left pulmonary vein is surgically catheterized distally to the hilus of the left lung allowing for direct microsphere injection into the left atrium while the descending aortic catheter is used for reference sampling. Fluorescent microspheres were injected to measure SCBF (immediately) prior to TAASA sacrifice (BASELINE, color 1), 5 min after complete TAASA sacrifice (ENDCLAMPING, color 2), at 1 h after complete TAASA sacrifice (1 h, color 3), 5 h complete TAASA sacrifice (5 h, color 4), at 24 h after complete TAASA sacrifice (24 h, color 5) and finally at 72 h after complete TAASA sacrifice (72 h, color 6). 2.6. Monitoring technique for motor evoked potentials (MEPs) A 5 cm longitudinal incision is made in the scalp overlying the skull, and the periosteum is removed to expose the sagittal and coronal sutures of the calvarium. Four stainless steel screw electrodes with attached wire leads are screwed into the skull 10 mm lateral to the sagittal suture. Two screws are placed on the left side (8 mm anterior and 8 mm posterior to the coronal suture), and two equally placed on the right. The wire leads are connected to an electrical stimulator (Digitimer Stimulator Model D 180A, Welwyn Garden City, United Kingdom). Electromyographic recordings are made from sterile stainless steel needle electrodes placed through the skin over the tibialis muscle in the hind leg and the muscles in the foreleg. A stimulation train (three pulses, 200 300 V, 100 ms pulse duration, and 2 ms interstimulus interval) delivered to the skull electrodes is used to elicit MEPs. MEPs are amplified (gain 2000), bandpass filtered (10 1000 Hz), digitized, and stored on an optical disk for subsequent analysis by a Spectrum 32 neurophysiological recording system (Cadwell Laboratories Inc., Kennewick, WA, USA). MEPs were recorded before clamping, during the 3-min interval after clamping of each segmental pair, and after clamping of all thoracic and abdominal segmental arteries for a period of 60 ( 90) min. The baseline value was determined just prior to the start of SA clamping. A lack of response to the stimulus is considered evidence of acute spinal cord ischemia. Data acquisition and analysis were performed on a computer with an AD converter and software (LabVIEW, National Instruments, Austin, TX) as previously published. 2.7. Neurobehavioral assessment All animals were videotaped at the same time daily, and a neuroscientist, blinded to the intraoperative course of events, utilized the coded videotapes to carry out neurological scoring using a modification of the Tarlov score. The modified Tarlov score is scaled as follows: no voluntary movements (0); perceptible movements at joints (1); good movements at joints but inability to stand (2); ability to get up and stand with assistance <1 min (3); ability to get up with assistance and stand unassisted <1 min (4); ability to get up with assistance and stand unassisted >1 min (5); ability to get up and stand unassisted >1 min (6); ability to walk <1 min (7); ability to walk >1 min (8); complete recovery (9). 2.8. Regional blood flow assessment (microspheres) Our regional blood flow assessment strategy involves the use of microspheres (15 mm polystyrene beads) that fluoresce under ultraviolet light. Use of different colors (yellow, pink, purple, and coral, each of which is available in low, medium, and high varieties), enables multiple measurements in the same tissue; sampling technicalities limit this to about 8 per tissue. The microsphere bolus consists of 2.5 million spheres which are administered centrally into a left heart chamber. At the same time as the injection is made, a reference sample of blood is withdrawn from an artery downstream in the arterial tree from the injection site. This is withdrawn using a specialized pump at a precise rate, in our experiments 2.91 ml/min. This basically allows a ratio calculation: the ratio of microspheres in a given piece of tissue to those in the blood reference sample is the same as the ratio of their blood flows. The reference samples are placed in tubes with EDTA anticoagulant. Multiple injections of different colors are made at relevant stages of the surgical protocol. At the end of the experiment, the pig is sacrificed by exsanguination under anesthesia and the tissues are harvested for blood flow determination, histological analysis, or both. The spinal cord is removed from the animal throughamidlinedorsalincisionwhichrunsfromtheskullto thesacrum.theparaspinalmusclesaredissectedoffthe vertebral column and the spinal canal is then entered via bilateral laminectomies down the length of the back exposing the spinal cord, which is then removed. The anatomical levels can be identified through the origins of the spinal nerves and the appropriate divisions made for analysis. The blood and tissue samples are then analyzed at Interactive Medical technologies (IMT) Ltd., Irvine, California. The samples are analyzed in a flow cytometer (fluorescent spectrophotometer) which measures the fluorescence at the various wavelengths. Spinal cord blood flow (SCBF) is determined from the fluorescent intensities (counts) of the tissue and blood reference samples using the formula: SCBF ðml=min=gþ ¼ R It I br Wt where R = blood reference withdrawal rate (2.91 mls/min), I t and I br are the tissue and blood reference samples fluorescent intensities or counts, and Wt is the weight of the tissue sample (g).

C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 1033 2.9. Histopathological evaluation Portions of the spinal cord not used for microsphere analysis were fixed in 10% formalin solution, embedded in paraffin, and then sectioned transverse to the craniocaudal axis, with samples at 0.5 cm intervals. Sections 6 mm in thickness were stained with hematoxylin and eosin, examined and scored blindly by an experienced neuropathologist according to a schematic grading system which was developed to classify the ischemic spinal cord damage (ISCD) at each segmental level: 1 = necrosis of single (motor) neurons only; 2 = necrosis of one posterior horn only; 3 = necrosis of both posterior horns only; 4 = necrosis of both posterior horns + surrounding white matter; 5 = necrosis of both anterior horns only; 6 = central necrosis involving posterior and anterior horns + parts of white matter; 7 = complete necrosis of gray matter only; 8 = complete necrosis of the whole section (Fig. 3). 2.10. Data analysis For the purposes of data analysis, pigs were classified into one of three groups: controls that had no segmental arteries resected, pigs that had all possible TAASA resected and Table 1 Intra- and postoperative data subsequently recovered spinal cord function, and pigs that had all TAASA resected but were left with either paraplegia or paraparesis. All analyses were implemented with SAS software for the PC version 9.1.3, SAS Institute Inc., Cary, NC. Physiologic data are described by means and standard deviations in each of these groups. Hierarchical linear models, using Proc Mixed, were used to analyze the association of flows with groups and segments. In separate analyses of the three segment regions at the 5 h and 24 h time points, flows were modeled as a function of group, segments, and interaction, with repeated observations (over segments) and an unstructured or autoregressive covariance matrix. Pair wise comparisons between groups were obtained as solutions to the fixed effects. The chi-square test for trend was used for predicting the binary outcome, recovery of spinal cord function, on the basis of the 5 h flows. 3. Results Variable/group Baseline prior to ligation After TAASA ligation 3.1. Comparability of experimental groups The results of this experiment were analyzed by comparing three groups: pigs which were operated on to expose the 5 min 1 h 5 h 24 h 72 h MAP (mmhg) Control group 90 2 90 1 96 8 93 7 91 2 90 1 Paraparetic/paraplegic group 90 8 93 15 90 9 96 15 78 15 92 16 Recovery group 91 9 91 8 89 7 96 16 99 21 108 17 ph Control group 7.52 0.04 7.51 0.05 7.44 0.05 7.38 0.03 7.53 0.01 7.49 0.02 Paraparetic/paraplegic group 7.48 0.06 7.44 0.05 7.40 0.05 7.39 0.07 7.45 0.05 7.47 0.04 Recovery group 7.50 0.04 7.48 0.05 7.50 0.04 7.36 0.08 7.47 0.04 7.47 0.03 PCO 2 Control group 33.0 3.8 33.2 5.5 39.7 8.0 43.7 3.9 35.8 2.0 38.0 2.8 Paraparetic/paraplegic group 40.0 7.9 41.6 6.3 44.0 7.4 40.4 5.1 33.3 2.0 33.9 4.1 Recovery group 37.7 3.6 40.2 7.6 37.4 3.5 37.5 4.7 32.4 2.9 33.6 1.9 PO 2 Control group 364 59 362 54 367 23 76 18 84 5 85 8 Paraparetic/paraplegic group 390 54 388 47 382 54 74 8 78 18 73 18 Recovery group 381 46 357 36 364 58 72 18 87 14 76 8 Hb Control group 10.9 0.6 10.1 0.6 9.9 0.8 10.3 0.8 9.8 1.2 9.9 1.3 Paraparetic/paraplegic group 10.4 1.4 10.8 2.1 9.7 1.3 10.5 1.7 10.8 2.6 9.6 2.2 Recovery group 11.4 0.7 11.9 0.9 11.3 1.5 13.7 1.1 11.1 1.6 9.3 1.5 O 2 saturation (%) Control group 99.8 0.06 99.8 0.0 99.8 0.0 94.6 4.7 97.3 0.5 97.0 0.8 Paraparetic/paraplegic group 99.8 0.05 99.8 0.04 99.8 0.1 94.7 0.7 94.9 2.5 94.7 3.1 Recovery group 99.8 0.05 99.8 0.06 99.8 0.1 92.3 5.5 93.8 8.0 93.4 6.6 Glucose Control group 89 42 78 35 83 21 119 18 94 12 94 12 Paraparetic/paraplegic group 115 38 133 49 118 33 159 102 91 19 72 12 Recovery group 97 32 92 42 80 39 103 32 110 36 94 12 Lactate Control group 1.6 0.9 1.6 1.1 1.3 0.8 2.3 2.1 0.6 0.2 0.7 0.5 Paraparetic/paraplegic group 1.4 0.7 1.5 1.1 1.4 0.6 2.7 1.3 3.0 1.3 1.9 1.8 Recovery group 2.5 1.7 2.0 1.3 1.6 0.9 5.7 2.4 4.1 5.0 2.2 1.9 Mean aortic pressures (MAP) and blood gas analyses for control (N = 6), paraparetic/paraplegic (N = 7) and recovered (N = 7) animals.

1034 C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 entire aorta but had no interruption of any segmental arteries (n = 6; weight: 30.2 2.4 kg); pigs that had all possible TAASA resected and subsequently recovered spinal cord function (n = 7; weight: 26.1 2.0 kg), and pigs that had all TAASA resected but were left with either paraparesis or paraplegia (n = 7; weight: 27.2 2.8 kg). The physiological data with regard to each of the groups are shown in Table 1. Target aortic mean aortic pressure was 90 mmhg intraoperatively. Only volume infusions but no pharmaceuticals were used to maintain aortic pressures. The mean aortic pressures intraoperatively were similar between the spinal cord injury group and the pigs that recovered. No clinically significant differences in intraoperative physiological parameters were evident. 3.2. Relationship between functional recovery and spinal cord blood flow (SCBF) All the pigs retained normal cord function (MEP) until 1 h after complete TAASA ligation. Since our main interest was in the relationship between flow parameters and spinal cord integrity, the pigs with TAASA ligation were divided for subsequent analysis into two groups depending upon the degree of functional recovery postoperatively. Those animals that regained normal function within the five days of postoperative observation, with a modified Tarlov score 4, indicating that they were able to stand without assistance, were considered to have recovered. Those with a score <4 by the fifth day postoperatively were considered to have sustained spinal cord injury (paraparesis/paraplegia; Fig. 1). Seven pigs (50%) with complete TAASA sacrifice recovered after 72 h, but seven suffered paraparesis/paraplegia. SCBF was similar in the two groups with TAASA sacrifice and in the controls intraoperatively, and until 1 h postoperatively, as shown in Fig. 2. Postoperatively, however, SCBF in pigs with ischemic cord injury was significantly lower at 5 h in the T8 L2 segments Fig. 1. Postoperative neurobehavioral recovery after complete TAASA ligation (N = 14 animals) using a modified Tarlov score, as detailed in the text. Recovery was defined as a score 4, which requires the pig to get up with assistance and stand unassisted <1 min. ( p = 0.0002) and L3 S segments ( p = 0.0007) than in contemporaneous controls without TAASA ligation. At 24 h, SCBF remained marginally lower from T8 caudally in the pigs that subsequently developed paraparesis or paraplegia. By 72 h postoperatively, SCBF was similar among all groups at all levels of the spinal cord. Outcome was predictable on the basis of SCBF at 5 h post TAASA ligation (Table 2). SCBF 5 h after complete TAASA ligation, averaged over these four central segments (T8 L2) showed a statistically significant association ( p = 0.003, chisquare test for trend) with outcome (paraparetic/paraplegic vs recovery). In examining the results, the pattern of SCBF in the control group is notable in several respects. As indicated by the dotted lines in Fig. 2, the blood flow in the cool, anesthetized pigs was much lower than in the postoperative state, awake at 37 8C. More surprisingly, there was a marked hyperemia at 5 h in the control pigs at all levels of the spinal cord. This hyperemia was also present in the pigs with TAASA ligation in the cervical region, but was not seen to the same extent in the pigs with TAASA ligation in the central and, to a lesser extent, in the lumbosacral region of the cord, and was significantly absent, as noted above, in those pigs that did not recover spinal cord function. 3.3. Statistical analyses of regional SCBF at 5 h and 24 h SCBF was similar among the three groups prior to TAASA clamping along the entire cord, but differed significantly at 5 h in the T8 L2 segments ( p = 0.0002) and the L3 S segments ( p = 0.0007). Later differences (at 24 h and 72 h) did not reach statistical significance. Lower mean SCBF values in T8 L2 at 5 h were associated with higher proportions of pigs with paraparesis/paraplegia, as shown in Table 2. Looking at the different regions of the spinal cord at 5 h, in the upper spinal cord (segments C1 T7) there was no significant overall difference in SCBF between recovered, paraparetic/paraplegic and control animals ( p = 0.46). In the middle portion of the cord (segments T8 L2) regional SCBF at 5 h was significantly different among the three groups ( p = 0.0002). However, between paraparetic/paraplegic and recovered animals there was only a trend ( p = 0.1) whereas paraparetic/paraplegic and control animals were highly statistically significantly different ( p < 0.0001) in this area. In the caudal spinal cord (segments L3 S) regional SCBF was statistically significantly different among the three groups ( p = 0.0007). However, again paraparetic/paraplegic and recovered animals were similar ( p = 0.40), whereas paraparetic/paraplegic and control animals were significantly different ( p = 0.02). By 24 h after complete TAASA sacrifice, there were no longer any statistically significant differences in SCBF among the groups: segments C1 T7, p = 0.62; segments T8 L2, p = 0.17, and segments L3 S, p = 0.08. 3.4. MEP monitoring MEP monitoring was carried out to assess the impact on function as the TAASAs were being serially sacrificed, as

C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 1035 Fig. 2. Regional spinal cord blood flow (SCBF) was determined using fluorescent microspheres (as detailed in the text) at various time points following serial ligation of all segmental arteries (TAASA). No segmental artery sacrifice was carried out in the controls. Pigs in the recovery group subsequently regained hind limb function, but those in the paraparetic/paraplegia group suffered permanent spinal cord injury. The dashed lines represent baseline flows at 32 8C during anesthesia, and after awakening, at 37 8C. The three graphs represent cervical and upper thoracic, lower thoracic and upper lumbar, and lumbosacral regions of the spinal cord, as indicated. The first baseline was recorded at 32 8C under anesthesia, and the second at 72 h. It is notable that there were no significant differences among the groups in the cervical region of the cord. Below this level, however, there was a significant difference between the paraparetic animals and the controls at 5 h, with the controls exhibiting a marked hyperemia. There were no significant differences among the groups at the later time points. described in detail previously, and until 1 h after the last of them had been sacrificed: later monitoring is not possible without prolonging the duration of anesthesia. As was seen in earlier studies, MEPs after extensive SA sacrifice were intact at the end of the procedure even in those pigs that subsequently suffered a loss of spinal cord function. This suggests that critical ischemia occurred in these pigs after the last measurement of MEPs had been carried out (1 h after the last of the TAASAs had been occluded), at a time when spinal cord perfusion pressure is still very low, but MEPs can no longer be recorded. 3.5. Histopathological findings Despite complete TAASA sacrifice, there was no evidence of necrosis in the cervical and higher thoracic segments in either paraparetic/paraplegic or recovered animals to the level of T7 in all animals (and T9 in recovered animals; Fig. 4). In the lower thoracic and lumbar segments, however, Table 2 Predicting outcome on the basis of SCBF in T8 L2 @ 5 h after TAASA ligation SCBF (ml/min/g) in T8 L2 @ 5 h after complete TAASA ligation No. of animals Nos. with paraplegia <0.08 5 4 80% 0.08 0.125 5 3 60% 0.125 0.24 5 0 0 0.24 4 0 0 %With paraplegia The table shows the segmental flows @ 5 h after complete TAASA ligation, averaged over the four segments (T8 L2) divided into equal size groups, and their relation to the chance of paraplegia. This association is statistically significant ( p = 0.003, chi-square test for trend). paraparetic/paraplegic animals showed distinctive necrosis of the gray matter involving the posterior and anterior horns and surrounding white matter, sparing a thin peripheral rim of white matter below the pia (Figs. 3 and 4) with the center of ischemic damage between T8 and L2. Although the necrosis had an abrupt onset between segments T8 and T9 in most of the paraparetic/paraplegic animals, in other respects the areas in which necrosis was found correspond to the vascular territory of the anterior spinal artery. In pigs that regained function, all segments showed a normal population of neurons and intact gray matter at all levels at lower magnification. There was no evidence of comprehensive segmental necrosis, although higher power evaluation showed rare minute areas with necrotic cells in sections from the lower thoracic (T10 11) and lumbar levels (L1 S; see Fig. 4). 4. Discussion The blood flow studies described in this report have led to several new observations which provide valuable insight into the response of the spinal cord circulation to extensive segmental artery sacrifice (TAASA). This has led to a better understanding of the gradual adaptation of the spinal cord to loss of much of its usual vascular input, and an increase in the likelihood that elimination of postoperative spinal cord injury following surgery for extensive thoracic and thoracoabdominal aneurysms is an achievable goal. To begin with, SCBF under mild hypothermia and anesthesia, as seen in Fig. 2, is considerably less than the flow when the pig is warm and awake. This relatively reduced flow is not surprising, assuming that autoregulation is

1036 C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 Fig. 3. A schematic grading system was developed to classify the ischemic damage at each segmental level of the spinal cord: 1 = necrosis of single (motor) neurons only; 2 = necrosis of one posterior horn only; 3 = necrosis of both posterior horns only; 4 = necrosis of both posterior horns + surrounding white matter; 5 = necrosis of both anterior horns only; 6 = central necrosis involving posterior and anterior horns + parts of white matter; 7 = complete necrosis of gray matter only; 8 = complete necrosis of the whole section. present, given the expectation of a reduced metabolic rate in the spinal cord under conditions of hypothermia and muscle paralysis. But it is surprising that SCBF does not diminish below the intraoperative baseline value after all TAASA have been sacrificed, and even 1 h later. This implies that complete TAASA sacrifice does not itself cause spinal cord Fig. 4. Regional ischemic damage of the spinal cord (ISCD) after complete TAASA sacrifice paraparetic/paraplegic versus recovery group. The Y-axis shows the grade of the ischemic damage and a corresponding low power (2) transverse section of the spinal cord, stained with hematoxylin and eosin, from pigs 3 days after extensive sacrifice of segmental arteries as examples. The cord segments from which the sections are taken are indicated at the X-axis. injury, an observation which is confirmed by the presence of intact MEPs in all pigs including those who subsequently develop paraparesis or paraplegia at the 1 h time point. The ischemia which leads to spinal cord injury seems to occur between one and 5 h after complete TAASA sacrifice. But even at 5 h, SCBF does not fall below intraoperative baseline values, even though we know from previous studies in the same model that spinal cord perfusion pressures are very low [12]. Rather, as seen from SCBF measurements in the control animals who did not undergo any TAASA ligation, there seems to be a requirement for increased blood flow during rewarming and recovery from anesthesia: the control levels are above those seen after the pigs are completely recovered at normothermia. At the 5-h time point, pigs that subsequently develop spinal cord injury have levels of SCBF that are higher than the anesthetized, hypothermic baseline, but below the baseline level for awake normothermic pigs. In contrast, the pigs that recover function after TAASA ligation have levels of SCBF at 5 h in the same range as the normothermic awake baseline, but except in the cervical regions of the cord they do not exhibit the hyperemia seen in the controls. Why should there be a requirement for spinal cord hyperemia during recovery from anesthesia and return to normothermia? Again assuming some degree of autoregulation, spinal cord hyperemia implies the presence of an increase in demand and thus a higher level of metabolism. This could be a consequence of enhanced neural activity associated with recovery from anesthesia, including spontaneous movement and shivering. Beginning at about 24 h, SCBF returns to normal baseline levels, as defined by the controls in whom no TAASA were ligated, within 72 h, even in pigs that subsequently show evidence of spinal cord injury. It is intriguing to speculate on how this is achieved after TAASA ligation whether by an increase in diameter and change in structure of pre-existing collateral vessels (which has been termed arteriogenesis), or by formation of entirely new vessels by endothelial sprouting (angiogenesis). The implications arising from these observations are heartening. First of all, whatever strategies are required to prevent spinal cord injury need to be put in place for only 72 h. The most direct approach is maintenance of a high perfusion pressure not only intraoperatively, but also for the entire vulnerable postoperative recovery period. Another possibility, however, is to prevent the putative increase in spinal cord metabolic rate from occurring during the period before cord flow can return to normothermic awake baseline levels. A temporary reduction in metabolic rate could possibly be achieved by prolonging hypothermia, or by transient pharmacological paralysis of muscles usually activated upon awakening from anesthesia. The optimal approach may turn out to be a combination of techniques to enhance blood flow and to decrease metabolic rate in the spinal cord for at least 24 h, and possibly as long as 72 h. The histological evidence of minute areas of necrosis even in the pigs that recover function after extensive TAASA sacrifice, and the failure of the levels of SCBF in recovered animals to reach the same levels as the controls at 5 and 24 h both suggest that the margin of safety (in terms of SCBF) for preservation of spinal cord integrity after TAASA sacrifice is currently very small.

C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 1037 5. Conclusion This study of spinal cord perfusion has documented that extensive ligation of TAASA does not diminish SCBF below intraoperative baseline values. What distinguishes pigs that develop spinal cord injury from those who recover normal function is a failure to mount an adequate hyperemic response during awakening from anesthesia and rewarming. But even in animals with spinal cord injury, flow returns to normal within 72 h, suggesting that a combination of strategies to optimize SCBF and decrease spinal cord metabolism for 72 h postoperatively could reliably prevent paraplegia following extensive TAASA sacrifice. This would enable endovascular treatment of TAA/A, offering therapy to many patients too old and frail to be considered for open surgical treatment. References [1] Svensson LG. Paralysis after aortic surgery: in search of lost cord function. Surgeon 2005;3:396 405. [2] Coselli JS, LeMaire SA, Miller 3rd CC, Schmittling ZC, Koksoy C, Pagan J, Curling PE. Mortality and paraplegia after thoracoabdominal aortic aneurysm repair: a risk factor analysis. Ann Thorac Surg 2000;69:409 14. [3] Bachet J, Guilmet D, Rosier J, Cron C, Dreyfus G, Goudot B, Piquois A, Brodaty D, Dubois C, de Lentdecker P. Protection of the spinal cord during surgery of thoraco-abdominal aortic aneurysms. Eur J Cardiothorac Surg 1996;10:817 25. [4] Kouchoukos NT, Rokkas CK. Hypothermic cardiopulmonary bypass for spinal cord protection: rationale and clinical results. Ann Thorac Surg 1999;67:1940 2. discussion 1953 48. [5] Svensson LG, Patel V, Robinson MF, Ueda T, Roehm Jr JO, Crawford ES. Influence of preservation or perfusion of intraoperatively identified spinal cord blood supply on spinal motor evoked potentials and paraplegia after aortic surgery. J Vasc Surg 1991;13:355 65. [6] Yamauchi T, Takano H, Nishimura M, Matsumiya G, Sawa Y. Paraplegia and paraparesis after descending thoracic aortic aneurysm repair: a risk factor analysis. Ann Thorac Cardiovasc Surg 2006;12:179 83. [7] Etz CD, Halstead JC, Spielvogel D, Shahani R, Lazala R, Homann TM, Weisz DJ, Plestis K, Griepp RB. Thoracic and thoracoabdominal aneurysm repair: is reimplantation of spinal cord arteries a waste of time? Ann Thorac Surg 2006;82:1670 7. [8] Strauch JT, Spielvogel D, Lauten A, Zhang N, Shiang H, Weisz D, Bodian CA, Griepp RB. Importance of extrasegmental vessels for spinal cord blood supply in a chronic porcine model. Eur J Cardiothorac Surg 2003;24:817 24. [9] Strauch JT, Spielvogel D, Lauten A, Zhang N, Shiang H, Weisz D, Bodian CA, Griepp RB. Importance of extrasegmental vessels for spinal cord blood supply in a chronic porcine model. Rev Port Cir Cardiothorac Vasc 2003;10:185 91. [10] Strauch JT, Lauten A, Spielvogel D, Rinke S, Zhang N, Weisz D, Bodian CA, Griepp RB. Mild hypothermia protects the spinal cord from ischemic injury in a chronic porcine model. Eur J Cardiothorac Surg 2004;25:708 15. [11] Etz CD, Homann TM, Plestis KA, Zhang N, Luehr M, Weisz DJ, Kleinman G, Griepp RB. Spinal cord perfusion after extensive segmental artery sacrifice: can paraplegia be prevented? Eur J Cardiothorac Surg 2007;31: 643 8. [12] Etz CD, Luehr M, Kari F, Bodian CA, Smego S, Plestis KA, Griepp RB. Paraplegia after extensive thoracic and thoracoabdominal aortic aneurysm repair: does critical spinal cord ischemia occur postoperatively? J Thorac Cardiovasc Surg 2008;135:324 30. Appendix A. Conference discussion Dr J. Bachet (Paris, France): Thank you for sharing with us this very important experimental work from the laboratory of Dr Randall Griepp at Mount Sinai Hospital in New York. It is in perfect continuity with several other published experimental studies from your group aimed at elucidating the patho-physiology of the spinal cord vascularization and the occurrence of spinal cord injury during surgery of the thoracic or thoracoabdominal aorta. It is, in particular, the obvious and logical continuation of the study that you have presented last year in this very Association and that was published recently in the EJCTS. It represents another piece of evidence that the Collateral Network Concept as developed by Dr Griepp s group in New York and Dr Biglioli also in Milan and published as early as 1996 is a reality which might in the near future drastically modify our surgical techniques and certainly dramatically reduce, if not totally suppress, the dreadful perspective of postoperative paraplegia. The animal model used in the present study was exactly the same as the one used in your previously published work, right? Dr Etz: That s correct. Dr Bachet: All the animals were identical and underwent exactly the same protocol. However, seven, that is 50%, were paraplegic and seven were not. And apparently, the occurrence of spinal cord injury could not be predicted intraoperatively or before the fifth postoperative hour. But, and this will be my first question, do you think that all animals were actually identical? That is, could the spinal cord vascularization and, in particular, the spinal anterior arterial axis or the collateral network coming from the subclavian and medial lumbar arteries have variations which could explain why some animals are more sensitive to the segmental arteries exclusion than others? In other words, do you think that any anatomical or radiological study of this vascularization with any kind of imaging technique could determine preoperatively which animals are at higher risk? It is quite interesting to note that 5 h after exclusion of the last segmental arteries an important hyperhemic rebound of the spinal cord blood flow could be observed in the normal or recovering animals but not in the paraplegic animals. My second question is, then: is the spinal cord injury the consequence of this absence of hyperhemia as you infer, or on the contrary, is the absence of hyperhemia the result of an already reduced metabolic demand due to the already pathological status of the neurons? In other words, could the low flow during and immediately after the segmental arteries exclusion trigger some delayed phenomena such as excitotoxicity and/or apoptosis? In this regard, could the dosages of glutamate or TUNEL histological studies, for example, give any information about those processes? Another obvious point of interrogation lies, as it is often the case, in the translation of those experimental results to the clinical setting, of course. You state indeed in your paper that this experimental model closely simulates the procedure for resection of descending thoracic and thoracoabdominal aneurysms clinically in your institution. Is it so? The pig and human anatomies are not exactly comparable. In particular, the pig has more segmental arteries and only one at each level which then divides in two, right? Dr Etz: Correct. Dr Bachet: In addition, your animals were perfectly healthy. They had no voluminous aneurysms, and dissection and exclusion of the segmental arteries was certainly easy. This is not the case in many patients in whom the segmental arteries are often very difficult to find and to exclude before opening the aneurysm. And for my personal information and teaching, how do you do that in patients? More importantly, most patients are operated on for huge atheromatous lesions or chronic dissections. Many segmental arteries are already occluded and the collateral network supplied by the subclavian and hypogastric systems may be jeopardized. Clamping the already occluded arteries can be a loss of time, whereas clamping the few remaining might suppress an important element of an already fragile blood supply. Again, do you think that in some selected patients with heavy atheroma or huge extended lesions or previous aortic surgery a radiological assessment of the cord vascularization is worthless? Similarly, it is noteworthy that in your experiments the animals were not submitted to CSF drainage which is presently considered by most groups as part of the routine armamentarium in thoracic and thoracoabdominal aortic surgery. I am aware that CSF drainage in pigs is difficult and could be troublesome in the postoperative aftermath. Nevertheless, don t you think that absence of CSF drainage in your pigs could have impaired or worsened the spinal cord blood flow and precipitated the cord ischemia?

1038 C.D. Etz et al. / European Journal of Cardio-thoracic Surgery 33 (2008) 1030 1038 Those questions might be irrelevant. Indeed, your group has published in 2006 a clinical experience using the Collateral Network Concept in 100 consecutive patients operated between 2002 and 2004 with a paraplegia/ paraparesis rate of 2%, which, to my knowledge, is among the lowest ever published. If I dare to use an old surgical joke, I would say that now that you have proved that it works in human beings, you are trying to prove that it works in pigs as well. Dr Etz: Let me first start with the anatomy in pig as compared to humans. That is the most obvious question we have to answer, and since we were curious as to how it actually compared, we did cast studies prior to these experiments. What you see on the left is a cast of the human vascular system focusing on the abdominal lower thoracic aorta and showing the segmental arteries, and on the right side, a cast we produced in the pig. You are absolutely right: the pig has a T13 segmental artery one more than in humans and the majority of segmental arteries divide about 5 mm after they take origin from the aorta. We believe that the anatomy, which we investigated with about 20 of these casts in pigs, has very little variability in the pigs we are using, and that the anatomy is very similar to what we see in humans. Here we can see the 13 segmental arteries. Not all of them have one single origin from the aorta. In the pig thoracic aorta, there are frequently some segmental arteries very similar to human arteries, with two origins. We do verify in all animals at post-mortem that all segmental arteries have been sacrificed. We also do not think that there is a huge variability in the anatomy of the collateral inflow, and it is also not very different in pigs and humans. The hypogastric arteries are a little greater in caliber in pigs, but they are pretty close to what one sees in the humans. You are also right that we do not have access to atherosclerotic pigs, much as we would love that. In humans, segmental arteries frequently are occluded prior to surgery. We are currently trying to investigate the remodeling activities of the paraspinal and segmental artery network, and we can see that changes set in after experimental segmental artery clamping which may be similar to what occurs after atherosclerotic occlusion of the segmental arteries. We believe that when occlusion of the segmental arteries occurs, it may provoke development of the collateral network, making it not more dangerous, but possibly even safer to clamp the segmental arteries which remain. As to the predictability of spinal cord blood flow recovery: this is the table that shows the predictability of outcome based on spinal cord blood flow. We can see that if the flow is below a certain threshold, 80% of animals develop paraplegia, and that none have spinal cord injury if the flow is only about 50% higher. So it is critical to have an adequate blood flow from T8 to about L2. We do not think that reduced flow is secondary to earlier ischemia-induced necrosis, since all animals have intact motor-evoked potentials until just before warming and awakening. We think that variability of collateral flow, which is probably dependent upon minor anatomical variations, explains differences in the ability of spinal cord hyperemia to occur. With regard to clinical comparisons, one must bear in mind that although the paraplegia rate was only 2%, the average number of sacrificed segmental vessels in our patients is much smaller than in our experimental model. Our ultimate aim, however, is to learn how to manipulate the collateral network in such a way that, in the clinical situation, cord viability can be assured routinely with sacrifice of all segmental vessels. This is an absolute prerequisite for safe endovascular treatment of the entire thoracoabdominal aorta. With regard to cerebrospinal fluid drainage, you are absolutely correct that we would expect to reach even lower paraplegia rates with its use: during the year in question, we had 50% paraplegia without CSF drainage. In the last year we have had a recovery rate of 60 70%. We are convinced that if we could do CSF drainage, we would lower the paraplegia rate even further. CSF drainage is not impossible in the pig; we have done it in the past. But it is very complicated to have the pigs survive for 5 days with yet another incision, and that is why we decided not to do it in these experiments. Dr F. Beyersdorf (Freiburg, Germany): One of the most frequently asked question in the field of endovascular aortic surgery is why there is such a low paraplegia rate in endovascular treatment? You and other members of Dr Griepp s group have shown that open conventional surgery may also result in low paraplegia rates, if you ligate all segmental branches before opening the lumen of the aorta. Now, in your present study you sacrificed the thoracic and lumbar segmental arteries and wouldn t you expect a low paraplegia rate and a high percentage of recovery? Dr Etz: Yes. Dr Beyersdorf: So my question is, if the hypothesis is correct that paraplegia results from a steal phenomenon (bleeding from these branches during open surgery), why is in your model the paraplegia rate still 30%, 40%, 50%? Is it due to the fact, that you have not done extra neuroprotective measures what Dr Bachet asked for, or is this related to the model you have used? Dr Etz: You have to consider one very important point here. We take all segmental arteries arising from the aorta, so there is not a single one left. That is not the usual case if you do stenting, and it only occurs in perhaps 10% even of open thoracoabdominal aortic aneurysmal repairs. We really take every single artery that is there, from T4 to L5: most of the pigs lose 15 segmental arteries. That is 80% of the inflow. We created these extreme conditions deliberately, to better demonstrate differences between experimental groups. We have no doubt that we could reach recovery rates of 90 100% if we left the segmental arteries in this critical area in the lumbar part of the aorta intact.