manual regional carlo 2010

162
MANUAL OF REGIONAL ANESTHESIA Carlo D. Franco, MD Chairman Orthopedic Anesthesia JHS Hospital of Cook County Associate Professor Anesthesiology and Anatomy Rush University Medical Center www.CookCountyRegional.com Chicago, IL Fourth Edition 2010

Upload: ivette-m-carroll-d

Post on 10-Apr-2015

409 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Manual Regional Carlo 2010

MANUAL OF REGIONAL ANESTHESIA

Carlo D. Franco, MD Chairman Orthopedic Anesthesia

JHS Hospital of Cook County

Associate Professor Anesthesiology and Anatomy Rush University Medical Center

www.CookCountyRegional.com

Chicago, IL

Fourth Edition 2010

Page 2: Manual Regional Carlo 2010

2 | P a g e

This manual is intended for anesthesiology residents, nurse anesthetists and

fellow faculty members of the Department of Anesthesiology and Pain Management,

Cook County Hospital of Chicago. The writing in these pages reflects the author’s own

views and understanding of various regional anesthesia issues, as well as his

interpretation of the pertinent literature. The author has made every effort to give proper

credit to outside sources when applicable.

Patients and models appearing in this manual provided their written permission to

the author, to have their photographs taken for the purpose of teaching. Their decision

was voluntary and did not involve compensation of any kind. The photographs of cadaver

material shown in these pages, originate from dissections performed by the author in the

Anatomy Laboratory of Rush University Medical Center in Chicago, in compliance with

Rush University’s guidelines, as well as State and Federal laws and regulations.

Care has been taken to confirm the accuracy of the information presented.

However, the author is not responsible for errors or omissions, or for any consequences

resulting from application of the information and techniques in this manual, and makes

no warranty, expressed or implied, with respect to the contents of it.

This manual is in accordance with current recommendations, as of February 2010.

However, recommendations and guidelines change, therefore the reader is urged to check

for new indications, warnings and precautions.

Page 3: Manual Regional Carlo 2010

3 | P a g e

For my residents,

who make my coming to work

intellectually challenging and pleasurable

and

to the memory of my father

Page 4: Manual Regional Carlo 2010

4 | P a g e

CONTENTS

Chapter 1: Introduction

General considerations………………………………………………………………….. 8

Patient selection and premedication……………………………………………………. 8

Monitoring……………………………………………………………………………..… 9

Outcome issues…………………………………………………....................................... 9

Airway and regional anesthesia………………………………………………………….11

References……………………………………………………………………………..…12

Chapter 2: Local Anesthetics

Historical perspective………………..…………………………………………………..14

Chemical structure ……………………………………………………………………... 15

Mechanism of action and Na+ channels……………….………………………………....17

Pregnancy and local anesthetics……………………………………………………….....17

Fiber size and pattern of blockade………………….…………………………………....17

Local anesthetics additives…………….……………………….………………………..20

Metabolism………………………….………………………….……………………......24

Dibucaine number…………………………………………………………………...….. 24

Toxicity…………………………………………………….……………………….........25

Tumescent anesthesia……………………………………………………………….........26

Lipid emulsion……………………………………………………………………....…...27

Maximum dose…………………………………………………………………………...29

Methemoglobinemia……………………………………………………………….....….29

Allergy ………………………………………………………………………………......30

References……………………………………………………………………………......34

Chapter 3: Neuraxial Anesthesia

Spinal anesthesia

Anatomy………………………………………………………….........................37

Cerebrospinal fluid………………………………………………………….........38

Site of action and indications…………………………………….........................39

Determinants of spread………………………………………………………..…39

Anesthesia duration…………………………………………………………........41

Side effects and complications……………………………………………..…….41

Postdural puncture headache………………………………………………….….43

Transient neurological symptoms……………………………………………......44

Cauda equina syndrome…………………………………………………….........45

Back pain ………………………………………………………………….…….45

Spinal in the outpatient………………………………………………………..…46

Intrathecal adjuncts………………………………………………........................46

Page 5: Manual Regional Carlo 2010

5 | P a g e

Epidural Anesthesia

Anatomy………………………………………………………….........................47

Blockade characteristics………………………………………….........................47

Spread of local anesthetics……………………………………….........................47

Type of needles and catheters……………………………………………………48

Test dose…………………………………………………………........................48

Activating an epidural………………………………………………………........48

References…………………………………………………………………………......…49

Chapter 4: Regional Anesthesia and Anticoagulation

Introduction…………………………………………………………………………........51

Strength and grade of recommendations…………………………………………………51

Venous thromboembolism……………………………………………………………….51

Risk of bleeding………………………………………………………………………….53

Thrombolytics……………………………………………………………………………54

Unfractionated heparin…………………………………………………………………..54

Low molecular weight heparin…………………………………………………………..55

Oral anticoagulants………………………………………………………………………56

Thrombin inhibitors……………………………………………………………………...57

References………………………………………………………………………………..59

Chapter 5: Peripheral Nerve Blocks

Bringing the needle close to its target………………………….....……………………...61

Nerve stimulation……………………………………………………………………...…61

Ultrasound………………………………………………………………………………..62

Nerve injury……………………………………………………………………….…......65

Use of epinephrine……………………………………………………………………….66

Persistent paresthesia…………………………………………………………………….66

Pre-existing neurological condition……………………………………………………..67

Electrophysiological testing……………………………………………………………..67

Tourniquet……………………………………………………………………………….68

References……………………………………………………………………………….70

Chapter 6: Upper Extremity Blocks

Anatomy of the brachial plexus…………………………………………………...……..73

Interscalene block…………………………………………………………………...…...79

Supraclavicular block…………………………………………………………………….86

Infraclavicular block…………………………………………………………………......96

Axillary block………………………………………………………………………......104

References………………………………………………………………………………112

Page 6: Manual Regional Carlo 2010

6 | P a g e

Chapter 7: Lower Extremity Blocks

Anatomy………………………………………………………………………………..115

Lateral femoral cutaneous nerve block………………………………………………...123

Femoral block……………………………………………………………………….….124

Obturator nerve block………………………………………………………………..... 128

Lumbar plexus block…………………………………………………………………....133

Sciatic nerve block, classic (Labat-Winnie)……………………………………….…...136

Sciatic nerve block, Franco’s…………………………………………………….……..138

Sciatic subgluteal nerve block, di Benedetto’s…………………………………….…...144

Sciatic subgluteal nerve block, Franco’s…………………………………………….....146

Popliteal nerve block, Franco’s…………………………………………………….…...148

References………………………………………………………………………….…...154

Chapter 8: Continuous Nerve Blocks

Introduction……………………………………………………………………………..156

Benefits…………………………………………………………………………………156

Stimulating versus non-stimulating catheters………………………………………….157

Catheter-related problems………………………………………………………………157

References……………………………………………………………………………....158

Chapter 9: Other Blocks

Trans Abdominal Plane (TAP) Block…………………………………………………..160

Refrences………………………………………………………………………………..162

Page 7: Manual Regional Carlo 2010

7 | P a g e

CHAPTER 1

INTRODUCTION

Page 8: Manual Regional Carlo 2010

8 | P a g e

General considerations

Regional anesthesia is a combination of techniques (i.e., peripheral or neuraxial)

that are able to render part of the human body insensate to pain by selectively interrupting

nerve transmission without having to alter the patient’s level of consciousness.

In this manual I discuss several aspects related to regional anesthesia, according

to the techniques most commonly used in the United States, although with special

emphasis on the techniques we perform at Cook County Hospital of Chicago.

Regional anesthesia has been traditionally considered an “art”. As such, it is

usually practiced by “artists”, who use their particular talents to produce results difficult

to reproduce by anesthesiologists devoid of artistic talents. I have a great respect and

admiration for all the pioneers who introduced and/or helped popularized the various

regional anesthesia techniques available to us now. The anesthesiology community owes

them a debt of gratitude for they built the foundations of our current practice. However, I

also believe that the practice of regional anesthesia in the 21st century should be more a

science than an art, taking advantage of all the various technologies available to us now.

Using technology to help our work does not demean our practice; on the contrary, it

makes it more rational, reproducible, and potentially easier and safer. The introduction of

ultrasound in regional anesthesia is an example of that.

The nerve blocks that we perform, and which I describe in these pages, are based

on anatomical, physiological, and pharmacological facts. The endpoints chosen are

objective. We use local anesthetic solutions in volumes and concentrations considered

adequate and safe by clinical experience. Regional anesthesia practiced in this manner,

should likely lead to predictable and reproducible results.

Regional anesthesia carries the risks and complications associated with the use of

local anesthetics (i.e., local anesthetic toxicity), the risks and complications of using

needles and drugs in the proximity of nerves (e.g., neuropraxia, irreversible nerve

damage) and those risks associated with a particular technique (e.g., pneumothorax, total

spinal). As with any other anesthetic technique, choosing regional anesthesia requires a

thorough assessment that involves the patient, the surgeon, the nature of the procedure

and its estimated duration, as well as the anesthesiologist’s level of experience with

regional anesthesia and its management.

Patient selection and premedication

The type of anesthesia for any procedure must be tailored to every individual

patient. There are patients who in general are not good candidates for regional anesthesia,

especially if they remain awake (e.g., drug abusers, pediatric patients). On the other hand,

we have a large successful experience with peripheral nerve blocks on drug abusers and

some pediatric patients, confirming that each case must be individually evaluated.

Judicious use of sedation increases patient’s cooperation and acceptance. Sedation

should be used to calm anxiety, but not to turn the patient unconscious or otherwise

unresponsive. This is especially true in blocks performed close to the neuraxis, like

interscalene blocks and lumbar plexus blocks. Keeping the patient lightly sedated, but

awake and cooperative, makes the procedure easier for both the patient and the

anesthesiologist. Traditionally it has been considered that an awaken patient would

Page 9: Manual Regional Carlo 2010

9 | P a g e

contribute to the safety of the technique by being able to communicate with us (e.g., pain

at injection, early subjective symptoms indicating impending systemic toxicity, etc). This

is now controversial since there is some evidence that nerves can be penetrated and

injection can be performed intraneurally, although extrafascicular, without pain.

Improvements in ultrasound technology with better imaging resolution could potentially

improve safety.

Monitoring

Every nerve block, whether it is performed in a dedicated room, holding area, OR,

PACU or office, must be treated as potentially dangerous. Monitoring blood pressure,

heart rate and pulse oximetry, as well as the establishment of an IV access must always

be considered. Supplemental oxygen should be given especially when sedation is being

used. Resuscitation equipment, including oxygen, ambu bag, airways of different sizes,

intubation equipment and tubes, along with appropriate resuscitation drugs and suction

capabilities, must always be readily available.

A clear strategy to deal with and treat complications must be in place. It is always

advisable, before starting a technique, to leave room at the head of the bed for the

anesthesiologist to manage the patient’s airway, should that become necessary.

Familiarity with the surroundings helps when dealing with emergencies.

Outcome

Is regional anesthesia safer than general anesthesia?

Every discussion on regional anesthesia must address the issue of its relative

safety compared to general anesthesia. Despite several studies suggesting it and an

intuitive feeling that regional anesthesia seems “safer’ than general anesthesia, no definite

and general answer can be given. The inability to give a clear answer comes from paucity

of evidence in the literature. Most of the outcome studies available to us have compared

the relative benefits of neuraxial anesthesia (spinal or epidural) versus general anesthesia

in intra abdominal surgery. Most of the studies lack the power (number of cases) to be

able to see a true difference, if it existed, and most of them are retrospective. Lack of

randomization raises the possibility of technique bias selection (i.e., regional anesthesia

may have been preferred in sicker patients obscuring its potential benefits).

Other problems have to do with the parameter chosen for comparison. To

compare mortality for example, the sample would have to be extremely large in order to

find a statistically significant difference, since mortality under any type of anesthesia is

extremely low. Other parameters like DVT, myocardial infarction, pneumonia seem more

adequate for comparison, but their rates vary according to the procedure and not just type

of anesthesia.

The physiological response to the stress of surgery or “surgical stress response”

involves release of local and central mediators leading to increased levels of, among

others, cathecolamines, cortisol, aldosterone and renin. It is also frequently associated

with hypercoagulability, immune response depression and protein wasting. The release of

local tissue inflammatory factors like cytokines and interleukins can be partially blocked

by non-steroidal anti-inflammatory drugs and peripheral nerve blocks using local

Page 10: Manual Regional Carlo 2010

10 | P a g e

anesthetics. The central response, responsible for the release of cathecolamines and

cortisol, can only be blocked by neuraxial blocks using local anesthetics. Determination

of hormonal markers for stress can be demonstrated after general anesthesia and after

certain regional anesthesia techniques. However, its impact on morbidity has not been

clearly established. If physiological parameters are measured (e.g., PO2, O2 sat) the

values obtained are frequently better (at least in the short term) after regional than general

anesthesia. However, the real impact that better postoperative physiological parameters

have on morbidity is not clear.

Nonetheless, there seems to be some agreement that regional anesthesia improves

the outcome of selective surgical procedures in a number of different ways, including

decreased rates of DVT, PE and blood loss.

Surgeries most associated with improved outcome after regional anesthesia include:

1. Hip surgery (hip fracture surgery and total hip arthroplasty): rates of DVT, PE and

blood loss are reduced after neuraxial anesthesia. The mechanism is unknown, but

may involve better peripheral circulation and less stasis.

Mortality rates also have been shown to be significantly lower with epidural

anesthesia as compared to general anesthesia.

2. Total knee arthroplasty: rates of DVT and PE are lower with neuraxial anesthesia.

3. Prostatectomy: similar reduction rates in DVT and PE and may also involve better

peripheral circulation and decreased venous stasis.

4. Peripheral vascular surgery: epidural anesthesia and postoperative epidural

analgesia have shown to improve graft patency after peripheral vascular surgery,

but does not seem to improve outcome after intra-abdominal vascular surgery.

Mechanism is not clear. Improve runoff due to vasodilatation or preservation of

normal coagulation has been mentioned.

5. Colon surgery: postoperative thoracic epidural analgesia with local anesthetics

has shown to enhance colonic activity after colon resection. If narcotics are used

in conjunction with local anesthetics this beneficial effect is lost.

Procedures where regional anesthesia has not shown benefits as compared to

general anesthesia include:

1. Upper abdominal and thoracic surgery, this is despite the fact that better pain

scores and times to extubation after regional anesthesia can be demonstrated.

2. Upper and lower extremity surgery, even though the patients receiving regional

anesthesia may have a higher degree of satisfaction, better pain control and fewer

side effects like nausea and vomiting, especially immediately after surgery. This

difference rapidly disappears at 24 h.

An interesting meta-analysis on the subject of comparative outcome was

published in December 2000 in the British Medical Journal, by Rodgers et al from New

Zealand. The authors reviewed the literature looking for randomized trials with or

without use of neuraxial anesthesia (spinal or epidural) before 1997. A total of 141 trials

Page 11: Manual Regional Carlo 2010

11 | P a g e

including 9,559 patients were included in this meta-analysis. The following are the main

findings:

1. Overall mortality was about one third less in the neuraxial group (103 deaths/4871

patients versus 144/4688 patients, P=0.006). This decrease was observed

regardless as to whether neuraxial was used alone or in combination with general

anesthesia.

2. DVT decreased by 44%

3. PE decreased by 55%

4. Transfusion requirement decreased by 50%

5. Pneumonia decreased by 39%

6. There were also reductions in myocardial infarction and renal failure.

The authors concluded that neuraxial blocks “reduce postoperative mortality and

other serious complications”. It was not clear whether these effects were due “solely to

benefits of neuraxial blockade or partly to avoidance of general anaesthesia”.

Meta-analysis has the advantage of pooling large number of patients making it

possible to study infrequent clinical events. However, it also means putting together trials

from different institutions, frequently from different countries and cultures. It remains to

be seen whether theseencouraging results can be duplicated, and whether they could

apply to other regional anesthesia techniques (i.e., peripheral nerve blocks).

Other authors, like Christopher Wu from Johns Hopkins, have shown the benefits

of regional over general anesthesia, when non-traditional outcomes are measured. These

outcome parameters include patient satisfaction (including analgesia, prevention of

nausea and vomiting and discharge readiness), ability to undergo physical rehabilitation,

and cost. These so-called “soft” parameters are very important in today’s cost-conscious

practice.

Airway and regional anesthesia

For some anesthesiologists managing a difficult airway almost always means

securing it. This approach negates the benefits that regional anesthesia can provide when

judiciously used. Evidence is lacking to support the superiority of neither approach.

We believe, that regional anesthesia, with its capacity to produce safe and dense

surgical anesthesia with minimal physiological derangements, should be carefully

contemplated, on a case by case basis, in all kind of patients, including those with

potential difficult airway. This does not mean that the anesthesiologist should not be

prepared at all times to manage the airway, and have at his/her immediate disposal all

necessary equipment and personnel to do it. It is important to emphasize also, that

attempting to secure the airway in all patients is not completely devoid of risks and could

lead to severe morbidity in some cases.

In our practice we routinely provide regional anesthesia to patients with

challenging airways. These patients include the obese, as well as trauma patients wearing

halos and cervical collars. These patients are assessed individually. The discussion needs

to involve the patient and the surgeon and must take into account the anesthesiologist’s

expertise and familiarity with regional anesthesia. If a regional anesthesia option is

selected, a backup plan, that can be readily implemented, must be available at all times.

Page 12: Manual Regional Carlo 2010

12 | P a g e

References

1. Liu SS, Carpenter RL, Neal JM. Epidural anesthesia and analgesia. Their role in

postoperative outcome. Anesthesiology 1995; 82:1474-1506

2. Sharrock NE: Risk-Benefit Comparisons for Regional and General Anesthesia, In:

Finucane BT (ed), Complications of Regional Anesthesia. New York, Churchill

Livingstone, 1999, pp 31-38

3. Neal JM, McDonald SB. Regional Anesthesia and Analgesia: Outcome and Cost

Effectiveness. In: Neal JM, Mulroy MF, Liu SS (eds), Problems in Anesthesia,

Philadelphia, Lippincott, Williams & Wilkins, 2000, pp 188-198

4. Neal JM: Regional anesthesia and Outcome. In: Rathmell JP, Neal JM, Viscomi

CM (eds), Regional Anesthesia, The Requisites in Anesthesiology, Philadelphia,

Elsevier Mosby, 2004, pp 164-170

5. Rodgers A, Walker N, Schung S et al. Reduction of postoperative mortality and

morbidity with epidural or spinal anaesthesia: results from overview of

randomized trials. Br Med J, 2000; 321: 1493-504

6. Wu CL, Fleisher LA. Outcomes research in regional anesthesia and analgesia,

Anesth Analg 2000; 91: 1232-1242

7. Urban MK: Is Regional Anesthesia Superior to General Anesthesia for Hip

Surgery?, In: Fleisher LA (ed), Evidence-Based Practice of Anesthesiology.

Philadelphia, Saunders, 2004, pp267-269

Page 13: Manual Regional Carlo 2010

13 | P a g e

CHAPTER 2

LOCAL ANESTHETICS

Page 14: Manual Regional Carlo 2010

14 | P a g e

LOCAL ANESTHETICS

The cell membrane’s resting potential is negative and close to the potential

determined by potassium alone (-70 mV). During the transmission of an action potential,

Na+ moves into the cell through open Na

+ channels depolarizing the membrane and

bringing its potential to -20 mV or more.

Local anesthetics are compounds that have the ability to interrupt the transmission

of the action potential in excitable membranes. They bind to specific receptors in the Na+

channels and their action, at clinically recommended doses, is reversible. Conduction can

still continue, although at a slower pace, with up to 90% of receptors blocked.

All local anesthetics are potentially neurotoxic if injected intraneurally, especially

if that injection is intrafascicular. The neuronal damage may be directly related to the

degree of hydrostatic pressure reached inside the axoplasma. Local anesthetics injected

around nerves could also be toxic as result of the concentration of the agent and the

duration of the exposure (e.g., cauda equina after intrathecal local anesthetics).

The local anesthetics available in clinical practice are usually racemic mixtures,

that is a mixture of both R and S enantiomers. Exceptions are lidocaine, levo-bupivacaine

and ropivacaine. The S isomer appears to have similar efficacy than the R isomer, but

lesser cardiac toxicity.

Historical perspective

Anesthesia by compression was common in the antiquity. Cold as an anesthetic

was widely used until the 1800s, and then it came cocaine. The native Indians of Peru

chewed coca leaves and knew about their cerebral-stimulating effects. The leaves of

erythroxylon coca were taken to Europe where Niemann in Germany isolated cocaine in

1860. Carl Koller, a contemporary and friend of Sigmund Freud, is credited with the

introduction of cocaine as a topical ophthalmic local anesthetic in Austria in 1884. In

1888 Koller came to the US and established a successful ophthalmology practice at

Mount Sinai Hospital in New York until the year of his death in 1944.

Recognition of cocaine’s cardiovascular side effects, as well as its potential for

dependency and abuse, led to a search for better local anesthetic drugs. Cocaine is a good

topical local anesthetic that also produces vasoconstriction and for this reason it is still

used by some, for topical anesthesia of the nose and other mucous membranes. Cocaine

blocks the reuptake of cathecolamines from nerve endings. Total dose should not exceed

100 mg (2.5 mL of a 4% solution), to avoid systemic effects like hypertension,

tachycardia and cardiac arrhythmias.

Ropivacaine is the only other local anesthetic able to produce some

vasoconstriction, and that effect is weak.

Highlights on local anesthesia and related issues

1850s Invention of the syringe and hypodermic hollow needle.

1884 Halsted, an American surgeon, blocks the brachial plexus with a solution of cocaine

under direct surgical exposure.

Page 15: Manual Regional Carlo 2010

15 | P a g e

1885 Wood, in the United Kingdom, is credited with the introduction of conduction

anesthesia through hypodermic injection.

1897 Epinephrine is isolated by John Abel at Johns Hopkins Medical School.

1897 Braun in Germany relates cocaine toxicity with systemic absorption and advocates

the use of epinephrine.

1898 Bier is set to receive the first planned spinal anesthesia from his assistant

Hildebrandt. After CSF is obtained, the syringe is found not fit the needle and therefore

no injection could be performed. Bier then performs the first spinal anesthesia on

Hildebrandt using cocaine. They both experience the first spinal headaches.

1908 Bier introduces the intravenous peripheral nerve block (Bier block) with procaine.

1911 Hirschel performs the first percutaneous axillary block.

1911 Kulenkampff performs the first percutaneous supraclavicular block.

1922 Gaston Labat of France, a disciple of Pauchet, introduces in the US his book

“Regional Anesthesia Its Technic and Clinical Application”, the first manual of regional

anesthesia published in America.

1923 Labat establishes the first American Society of Regional Anesthesia.

1953 Daniel Moore, practicing at Virginia Mason Clinic in Seattle, publishes his

influential book “Regional Block”.

1975 Alon Winnie, L. Donald Bridenbaugh, Harold Carron, Jordan Katz, and P. Prithvi

Raj establish the current American Society of Regional Anesthesia (ASRA) in Chicago.

1976 The first ASRA meeting is held in Phoenix, Arizona.

1976 Regional Anesthesia Journal, volume 1, number 1 is published.

1983 Winnie introduces his book, Plexus Anesthesia, Perivascular Techniques of

Brachial Plexus Block.

Date of introduction in clinical practice of some local anesthetics:

1905 procaine; 1932 tetracaine; 1947 lidocaine; 1955 chloroprocaine (last ester

type that is still in clinical use); 1957 mepivacaine; 1963 bupivacaine; 1997 ropivacaine;

1999 levobupivacaine.

Chemical structure of local anesthetics

Local anesthetics are weak bases with a pka above 7.4 and poorly soluble in

water. They are commercially available as acidic solutions (pH 4-7) of hydrochloride

salts, which are hydrosoluble. A typical local anesthetic molecule is composed of two

parts, a benzene ring (lipid soluble, hydrophobic) and an ionizable amine group (water

soluble, hydrophilic). These two parts are linked by a chemical chain, which can be either

an ester (-CO-) or an amide (-HNC-). This is the basis for the classification of local

anesthetics as either esters or amides.

Injecting local anesthetics in the proximity of a nerve(s) triggers a sequential set

of events, which eventually culminates with the interaction of some of their molecules

with receptors located in the Na+ channels of nerve membranes. The injected local

anesthetic volume spreads initially by mass movement, moving across “points of least

resistance”, which unfortunately do not necessarily lead into the desired nerve(s). This

fact emphasizes the importance of injecting in close proximity to the target nerve(s). The

Page 16: Manual Regional Carlo 2010

16 | P a g e

local anesthetic solution then diffuses through tissues; each layer acting as a physical

barrier. In the process part of the solution gets absorbed into the circulation. Finally a

small percentage of the anesthetic reaches the target nerve membrane, at which point the

different physicochemical properties of the individual anesthetic will dictate the speed,

duration and nature of the interaction with the receptors.

Physicochemical properties-activity relationship

1. Lipid solubility: determines both the potency and the duration of action of local

anesthetics, by facilitating their transfer through membranes and by keeping the

drug close to the site of action and away from metabolism. In addition, the local

anesthetic receptor site in Na+ channels is thought to be hydrophobic, so its

affinity for hydrophobic drugs is greater. Hydrophobicity also increases toxicity,

so the therapeutic index of more lipid soluble drugs is decreased.

2. Protein binding: local anesthetics are bound in large part to plasma and tissue

proteins. The bound portion is not pharmacologically active. The plasmatic

unbound fraction is responsible for systemic toxicity. The most important binding

proteins in plasma are albumins and alpha-1-acid glycoprotein (AAG). Although

albumin has a greater binding capacity than AAG, the latter has a greater affinity

for drugs with pka higher than 8, the case for most local anesthetics. Newborn

infants have very low concentration of AAG, only reaching adult values by 10

months of age. The elderly and debilitated also frequently have decreased levels

of albumin and other plasma proteins. These patient populations could be at

increased risk for toxicity.

On the other hand, AAG levels increase during stress and for several days

after the postoperative period. Higher levels of AAG lead to decreased levels of

unbound fraction of local anesthetics and a decreased potential for local anesthetic

toxicity. However, changes in protein binding are only clinically important for

drugs highly protein-bound, such as bupivacaine, which is 96% bound, and

sufentanil and alfentanil, which are both 92% bound (Booker et al, Br J Anaesth

1996; 76:365-8).

The fraction of drug bound to protein in plasma correlates with the

duration of action of local anesthetics: bupivacaine (95%) = ropivacaine (94%)>

tetracaine (85%) > mepivacaine (75%) > lidocaine 65%) > procaine (5%) and 2-

chloroprocaine (negligible). This suggests that the binding site for the local

anesthetic molecule in the sodium channel receptor protein, may share a similar

sequence of amino acids with the plasma protein binding site.

Drugs as lidocaine, tetracaine, bupivacaine and morphine (e.g., DepoDur)

have been incorporated into liposomes to prolong their duration of action.

Liposomes are vesicles with two layers of phospholipids, which slow down the

release of the drug.

3. Pka: determines the ratio between the ionized (cationic) and the uncharged (base)

forms of the drug. The pka of local anesthetics ranges from 7.6 to 9.2. By

definition the pka is the pH at which 50% of the drug is ionized and 50% is

Page 17: Manual Regional Carlo 2010

17 | P a g e

present as a base. The pka generally correlates with the speed of onset of most

local anesthetics. The closer the pka is to physiologic pH, the faster the onset. For

example, lidocaine with a pka of 7.7 is 25% non-ionized at pH 7.4. Its onset is

therefore faster than bupivacaine, whose pka of 8.1 makes it only 15% non-

ionized at that pH. One important exception is 2-chloroprocaine that, despite its

pka of 9.1, has a very rapid onset. This is usually attributed to the relatively high

concentrations (3%) used in clinical practice that are possible thanks to its low

toxicity. It has also been claimed that 2-chloroprocaine has better “tissue

penetrability”.

Mechanism of action and sodium channels

The non-charged hydrophobic fraction (B), which exists in equilibrium with the

hydrophilic charged portion (BH+), crosses the lipidic nerve membrane and initiates the

events that lead to Na+ channel blockade. Once inside the cell, the pka of the drug and the

intracellular pH dictate a new equilibrium between the two fractions. Because of the

relative more acidic intracellular environment, the relative proportion of charged fraction

(BH+) increases. This hydrophilic, charged fraction is the active form on the Na

+ channel.

The Na+

channel is a protein structure that communicates the extracellular of the

nerve with its axoplasm. It consists of four repeating alpha subunits and two beta

subunits, beta-1 and beta-2. The alpha subunits are involved in ion movement and local

anesthetic activity. It is generally accepted that the main action of local anesthetics

involves interaction with specific binding sites within the Na+

channel. Local anesthetics

may also block to some degree calcium and potassium channels as well as N-methyl-

D-aspartate (NMDA) receptors. Local anesthetics do not ordinarily affect the membrane

resting potential.

The Na+ channels seem to exist in three different states, closed (resting), open and

inactivated. Under adequate stimulation, the protein molecules of the channel undergo

conformational changes, from the resting state to the ion-permeable state or open state,

allowing the inflow of extracellular Na+, which depolarizes the membrane. After a few

milliseconds the channel goes then through a transitional inactivated state, where the

proteins leave the channel closed and ion-impermeable. With repolarization the proteins

revert to their resting configuration.

Other drugs, like tricyclic antidepressants (amitriptyline), meperidine, volatile

anesthetics and ketamine, also exhibit Na+ channel-blocking properties. Tetrodotoxin and

other biotoxins also interact with the Na+

channels, although

their actions are exerted on

the extracellular side of the channel.

Frequency-dependent blockade

Local anesthetics show more affinity for open Na+ channels. When a nerve is

experiencing a high frequency of depolarization, like during spontaneous pain or

voluntary muscle contractions, it becomes more sensitive to blockade, because the

chances of interaction, between local anesthetics molecules and Na+

channels, increase.

The concept of frequency-dependent blockade also explains the greater

susceptibility to blockade exhibited by small sensory fibers, as they generate long action

Page 18: Manual Regional Carlo 2010

18 | P a g e

potential (5 ms) at high frequency. Motor fibers on the other hand generate short action

potentials (0.5 ms) at lower frequency making them more difficult to block.

Pregnancy and local anesthetics

Increased sensitivity to local anesthetics, demonstrated as faster onset and more

profound block, may be present during pregnancy.

Alterations in protein binding of bupivacaine, may result in increased

concentrations of active unbound drug in the pregnant patient, increasing its potential for

toxicity.

Placental transfer is also more active for lipid soluble local anesthetics. In any

case, agents with a pka closer to physiologic pH have a higher placental transfer. For

example the umbilical vein/maternal vein ratio for mepivacaine is 0.8 (pka 7.6) while for

bupivacaine is 0.3 (pka 8.1).

In the presence of fetal acidosis, local anesthetics cross the placenta and become

ionized in higher proportion than at normal pH. The ionized fraction cannot cross back to

the maternal circulation, originating what is called “ion trapping”. Therefore, 2-

chloroprocaine, with its very short maternal and fetal half-lives, is an ideal local

anesthetic in the presence of fetal acidosis.

Fiber size and pattern of blockade

As a general rule small nerve fibers are more susceptible to local anesthetics than

large fibers. However, other factors like myelinization and relative position of the fibers

within a nerve (mantle versus core) may also play a role. The depolarization in

myelinated fibers is saltatory. About three nodes of Ranvier need to be blocked in order

to block the transmission of the action potential.

The smallest nerve fibers are nonmyelinated and are blocked more readily than

larger myelinated fibers. However at similar size, myelinated fibers are blocked before

nonmyelinated fibers. In general autonomic fibers, small nonmyelinated C fibers

(mediating pain, temperature and touch), and small myelinated A delta fibers (mediating

pain and cold temperature) are blocked before A alpha, A beta and A gamma fibers

(motor, propioception, touch, and pressure).

It has been speculated that in large nerve trunks, motor fibers would be usually

located in the outer portion (mantle) of the nerve bundle, therefore more “accessible” to

local anesthetics. This would help explain why motor fibers tend to be blocked before

sensory fibers in large mixed nerves. In contrast, the frequency-dependence of local

anesthetic action would favor block of small sensory fibers, as they generate long action

potentials at high frequency, whereas motor fibers generate short action potentials at

lower frequency.

Page 19: Manual Regional Carlo 2010

19 | P a g e

(Figure from Morgan’s Clinical Anesthesiology, 3

rd edition, 2006, reproduced with permission)

Modulating local anesthetic action

pH adjustment

The ionized fraction of local anesthetics is the active form in the Na+

channel,

although the rate-limiting step in this cascade is membrane penetration of local

anesthetics in its non-ionized form. Unfortunately, only a small proportion of local

anesthetic in solution exists in the non-ionized state. Changes in pH can theoretically

reduce the onset time by increasing its proportion. At a pH of 5.0 to 5.5 the cation/base

ratio is 1000:1, at a pH of 7.4 the same ratio becomes 60:40. The limiting factor for pH

adjustment is the solubility of the base form before reaching precipitation. The most lipid

soluble agents, like bupivacaine and ropivacaine, cannot be alkalinized above a pH of 6.5

because they precipitate.

DiFazio et al (Anesth Analg 1986:65; 760-64) demonstrated a more than 50%

decrease in onset of epidural anesthesia, when the pH of commercially available

lidocaine with epinephrine was raised from 4.5 to 7.2, by the addition of bicarbonate.

Capogna et al (Reg Anesth 1995; 20: 369-377) randomized 180 patients to study the

effects of alkalinizing lidocaine, mepivacaine and bupivacaine for nerve blocks. They

concluded that alkalinization of lidocaine and bupivacaine shortens the onset of epidural;

alkalinization of lidocaine shortens the onset of axillary block and alkalinization of

mepivacaine shortens the onset of sciatic/femoral blocks. However, when only small

changes in pH can be achieved, because of the limited solubility of the base, only small

decreases in onset time will occur, as when plain bupivacaine is alkalinized.

It is generally accepted that adding bicarbonate to local anesthetics, may speed the

onset of local anesthetics solutions that have epinephrine added by the manufacturer

Page 20: Manual Regional Carlo 2010

20 | P a g e

(vials have a lower pH), while the effect would be negligible when fresh epinephrine is

added to a plain solution.

Chloroprocaine plus 1 mL of sodium bicarbonate for 30 mL of solution raises the

pH to 6.8. Adding 1 mL of sodium bicarbonate per 10 mL of lidocaine or mepivacaine

raises the pH of the solution to 7.2 and adding 0.1 mL of bicarbonate per 10 mL of

bupivacaine raises the pH of the solution to 6.4 (from Mulroy’s Regional Anesthesia, 3rd

edition, 2002).

Carbonation

Another approach to shortening onset time has been the use of carbonated local

anesthetic solutions. These solutions contain large amounts of carbon dioxide, which

readily diffuses into the axoplasm of the nerve, lowering the pH and favoring the

formation of the cationic active form of the local anesthetic inside the cell. Carbonated

solutions are not available in the United States.

LOCAL ANESTHETICS ADDITIVES

Vasoconstrictors

Epinephrine is the most common vasoconstrictor added to local anesthetics to

prolong the anesthetic effect and to decrease absorption. Epinephrine is also used to

detect intravascular injection. Without beta-blockers on board, 15 mcg of epinephrine

should produce a 30% increase in heart rate within 30 seconds.

Vasoconstrictors may also improve the quality and density of the block, especially

with spinal and epidural anesthesia. This has been demonstrated with tetracaine, lidocaine

and bupivacaine. The mechanism is unclear. Epinephrine may simply increase the

amount of local anesthetic available by reducing absorption. It could also have some local

anesthetic effect by means of its α2-agonist actions. Subarachnoid epinephrine also

delays voiding and discharge readiness. The prolongation of effect in peripheral nerve blocks can be 30-60%, depending

on site of injection and type of local anesthetics (more vascular sites like intercostal see

more effect, and intermediate agents like lidocaine benefit more). Peripherally

epinephrine does not have any significant alpha-2 effect.

In general, epinephrine added to spinal anesthesia prolongs the effect of the less

lipid soluble agents like lidocaine and mepivacaine (20-30%). The exception to this rule

is tetracaine, a highly lipid soluble agent, that gets the largest prolongation of all spinal

local anesthetics (up to 60% in lumbar dermatomes).

The usual dose of intrathecal epinephrine is 200 mcg, but doses as small as 50

mcg can be sufficient. In the epidural space the usual dose is 5 mcg/mL. Epinephrine,

other than intrathecal, is absorbed systemically and may produce adverse cardiovascular

effects. In small doses the beta-adrenergic effects predominate, with increased cardiac

output and heart rate. Dose larger than 0.25 mg (250 mcg) may be associated with

arrhythmias or other undesirable cardiac effects.

The potential risk for peripheral nerve ischemia, as a result of epinephrine acting

on epineural vessels and vaso nervorum has to be balanced against the lower risk of

systemic toxicity, the ability to detect intravascular injection and the prolongation of

Page 21: Manual Regional Carlo 2010

21 | P a g e

action. According to Neal (Reg Anesth Pain Med 2003;28:124-134) adding 5 mcg/mL

(1:200,000 dilution) prolongs the duration of lidocaine for peripheral nerve blocks from

186 min to 264 min. Adding only 2.5 mcg/mL (1:400,000 dilution) prolongs the block to

240 min (almost the same prolongation), without apparent effect on nerve blood flow.

Patients with micro angiopathy (e.g., diabetics), who could be at increase risk for neural

ischemia secondary to vasoconstriction, potentially could benefit from the use of more

diluted epinephrine. Adding only 1:400,000 epinephrine to local anesthetic solutions for

nerve blocks has become the standard in our practice, in both diabetics and non-diabetics

patients. In 2006 Bigeleisen reported in Anesthesiology a study in which he demonstrated

intraneural injection by ultrasound in 72 out of 104 nerves studied in the axilla. The local

anesthetic used was a combination of bupivacaine plus lidocaine and contained 3 ucg/mL

of epinephrine. The author did not find any evidence of nerve injury in up to 6 month

follow up.

Intrathecal epinephrine does not lead to cord ischemia, because it does not

decrease spinal cord blood flow, although it decreases epidural blood flow (Kosody R, et

al. Can Anaesth Soc J; 31: 503-8, 1984). In fact spinal cord ischemia due to epinephrine

is “improbable because the cord vessels are autoregulated and show very minimal

response to endogenous or exogenous vasoactive agents” (Neal JM In: Regional

Anesthesia, The Requisites. Elsevier Mosby, Philadelphia 2004, pp 25-31)

Although epinephrine-containing local anesthetics are usually contraindicated in

areas of terminal circulation (e.g., digits) this recommendation is not based on hard

evidence. Anecdotal use of epinephrine-containing solutions in digits is cited in the

literature. Lalonde et al published a multicenter study including 3,110 consecutive cases

of use of epinephrine in the fingers and hand from 2002-2004. The authors (surgeons)

defined “low dose” epinephrine as 1:100,000 and they reported no instance “of digital

tissue loss” (J Hand Surg 2005; 30:1061-67). At this time we do not recommend this

practice.

Dilution/concentration issues

By definition a 1:1,000 dilution means 1 g solute in 1,000 mL of solution. That is

the same than to say 1 mg/mL or 1,000 mcg/mL

Thus, if 1:1,000 equals 1,000 mcg/mL, then:

1:10,000 equals 100 mcg/mL

1:100,000 equals 10 mcg/mL

1:200,000 equals 5 mcg/mL 1:400,000 equals 2.5 mcg/mL

Opioids

1. Neuraxial use: The addition of opioids to local anesthetics has a synergistic

effect, both in anesthesia and postoperative analgesia (especially visceral pain).

They block pain pathways without significantly affecting motor or sympathetic

fibers.

Page 22: Manual Regional Carlo 2010

22 | P a g e

The hydrophilic opioid morphine can be used in doses of 0.1-0.3 mg spinal and 1-

3 mg epidural. It has a slow onset of 45 min, providing an analgesic action that

lasts 12-24 h. Morphine reaches the brainstem and 4th

ventricle slowly. Delayed

respiratory depression (8-10 h) is a risk with all neuraxial opioids, but it is more

frequently seen with hydrophilic drugs like morphine, and in susceptible

populations like the elderly and debilitated. Neuraxial morphine is also associated

with higher incidence (40-50%) of nausea and vomiting than systemic opioids,

more pruritus (60-80%, 20% of it severe), and delayed voiding. It is not suitable

for outpatients.

Short-acting opioids, such as fentanyl and sufentanil, when added to spinal

anesthetics can also intensify the block, and prolong the duration of anesthesia,

beyond the duration of local anesthetics. Respiratory depression with these agents

is rare and usually early (within 4 h). Sufentanil spinal can be used in doses of

2.5-10 mcg. Fentanyl spinal is used in doses of 10-25 mcg and 25-150 mcg

epidural. Onset occurs at 5-15 min, peak effect at 10-20 min and duration of 1-3

h. Hypotension, pruritus, nausea and vomiting are some common side effects.

Extended-release epidural morphine (DepoDur): is a new liposomal

formulation designed for epidural use, providing 48 h of pain relief. DepoDur was

approved in 2004. It is supplied in a 2 mL vial containing 10 mg/mL dose in

sterile saline. It is only approved as a single lumbar epidural dose prior to

surgery or after clamping of the umbilical cord during C-section. The

recommended dose is 10 mg for C-section, 10-15 mg for lower abdominal surgery

and 15 mg for major orthopedic surgery of the lower extremities. Respiratory

depression is dose-related. The most common adverse events reported during

clinical trials were decreased oxygen saturation, hypotension, urinary retention,

nausea and vomiting, constipation and pruritus.

2. Peripheral nerve blocks: The usefulness of opioids in peripheral nerve blocks is

mostly unsupported by the evidence. Opioids have been shown useful when

injected in the intra-articular space.

Clonidine

Alpha-2 agonists have central (sedation, analgesia, bradycardia) and peripheral

effects (vasoconstriction/vasodilation with net hypotension, anti shivering, diuresis). The

site for sedative action is the locus ceruleus of the brain stem, while the principal site for

analgesia seems to be the spinal cord.

The main alpha-2 effect on the heart is decreased tachycardia by blocking

cardioacelerator fibers, and bradycardia through a vagomimetic effect. In the periphery

clonidine produces both vasodilation via sympatholysis and vasoconstriction through

receptors on smooth muscle. The cause for its anti shivering and diuretic effects are yet to

be established.

Side effects, including sedation, hypotension and bradycardia, limit alpha-2

agonists use. Small doses of clonidine (50-75 mcg) have shown to significantly prolong

analgesia in spinal, epidural, IV regional, and peripheral nerve blocks, both when injected

Page 23: Manual Regional Carlo 2010

23 | P a g e

along local anesthetics and when given orally. Injected intrathecally, they also can delay

voiding and can produce orthostasis. Side effects do not occur often at clonidine doses

below 1.5 mcg/kg or a total dose less than 150 mcg.

Iskandar et al in France in 2001, showed that adding 50 mcg of clonidine to

selected nerves (median and musculocutaneous) prolonged mepivacaine sensory

anesthesia by 50%, compared to placebo, after a mid-humeral block, without prolonging

motor effect. Because the prolongation was observed only in the nerves that received

clonidine they postulated that the effect must be peripheral and not central through

absorption.

Dexmedetomidine It is a more selective alpha-2 agonist agent with an alpha-2:alpha-1 receptor ratio

of 1,600:1, seven times greater than that of clonidine. Its elimination half-life is only 2 h

compared to more than 8 h for clonidine. Dexmedetomidine may offer extended

analgesia with lesser side effects. This drug is gaining popularity as a sedative both in the

ICU and the OR.

Neostigmine It is an acetylcholinesterase inhibitor that prevents the breakdown of acetylcholine

promoting its accumulation. Acetylcholine is an endogenous spinal neurotransmitter that

induces analgesia. Neostigmine does not cause neural blockade nor have any action on

opioid receptors.

Spencer Liu et al in 1999 (Anesthesiology 1999; 90:710-717) studied the effects

of different doses of neostigmine added to bupivacaine spinal. They reported that 50 mcg

of neostigmine increased sensory and motor anesthesia, but also delayed discharge time

and was accompanied by 67% nausea and up to 50% vomiting. Lower doses did not show

analgesic effect, but still had significant rates of side effects (nausea and vomiting).

N-methyl-D-aspartate (NMDA) receptor antagonists

Activation of NMDA receptors makes the neurons of the spinal cord more

responsive to all types of input including pain stimuli (central sensitization). NMDA

receptor antagonists, like ketamine, have shown analgesic activity. In fact in IV regional

0.1 mg/kg of ketamine is superior to clonidine (1 mcg/kg) in preventing tourniquet pain.

Errando in Spain showed that commercially available ketamine containing benzethonium

chloride is toxic in swine (Reg Anesth Pain Med 1999; 24:146-52). Preservative-free

solutions of ketamine have proven safe.

Hyaluronidase

It breaks down collagen bonds potentially facilitating the spread of local

anesthetic through tissue planes. However, the evidence shows that at least in the epidural

space it can decrease the quality of anesthesia. Its use seems limited to retrobulbar blocks.

Dextran

Dextran and other high-molecular-weight compounds have been advocated to

increase the duration of local anesthetics. The evidence is lacking.

Page 24: Manual Regional Carlo 2010

24 | P a g e

METABOLISM OF LOCAL ANESTHETICS

Ester local anesthetics

They are rapidly hydrolyzed at the ester linkage by plasma pseudocholinesterase,

the same enzyme that hydrolyses acetylcholine and succinylcholine. The hydrolysis of 2-

chloroprocaine is about four times faster than procaine, which in turn is hydrolyzed about

four times faster than tetracaine. However, even tetracaine has a metabolic half-life of

only 2.5-3.0 min (Tetzlatt JE. In: Ambulatory Anesthesia Perioperative Analgesia. New

York, McGraw-Hill, 2005, p 193).

In individuals with atypical plasma pseudocholinesterase the half-life of these

drugs is prolonged and potentially could lead to plasma accumulation. Cerebrospinal

fluid does not contain esterase enzymes, so if an ester is used for spinal anesthesia (e.g.,

tetracaine) its termination of action depends on blood absorption.

The hydrolysis of all ester local anesthetics leads to the formation of para-

aminobenzoic acid (PABA), which is associated with a low potential for allergic

reactions. Allergic reactions may also develop from the use of multiple dose vials of

amide local anesthetics that contain methylparaben (PABA derivative) as a preservative.

As opposed to other ester type anesthetics, cocaine is partially metabolized in the

liver and partially excreted unchanged in the urine.

Amide local anesthetics

They are transported into the liver before their biotransformation. The two major

factors controlling the clearance of amide local anesthetics by the liver are hepatic blood

flow and hepatic function. The metabolism of local anesthetics as well as that of many

other drugs occurs in the liver by the cytochrome P-450 enzyme system. Because of the

liver large metabolic capacity it is unlikely that drug interaction would affect the

metabolism of local anesthetics. The rate of metabolism is agent specific (prilocaine >

lidocaine > mepivacaine > ropivacaine > bupivacaine).

The metabolism of amide local anesthetics is relatively fast, although slower than

esters. Elimination half-life for lidocaine is 1.5-2 h. Drugs such as general anesthetics,

norepinephrine, cimetidine, propranolol and calcium channel blockers can decrease

hepatic blood flow and potentially increase the elimination half-life of amides. Similarly,

decreases in hepatic function caused by a lowering of body temperature, immaturity of

the hepatic enzyme system in the fetus, or liver damage (e.g., cirrhosis) can lead to

decreased rate of hepatic metabolism of the amides. Renal clearance of unchanged local

anesthetic is a minor route of elimination (e.g., lidocaine is only 3% to 5% recovered

unchanged in the urine of adults, while bupivacaine is 10% to 16%).

The primary metabolic pathway for mepivacaine is oxidation to 3-hydroxy and 4-

hydroxymepivacaine. This pathway is less developed in neonates resulting in slower

metabolism of mepivacaine in newborns than in adults (Raj’s Regional Anesthesia).

The dibucaine number

People with atypical plasma pseudocholinesterase exhibit prolonged recovery

after a dose of succinylcholine or mivacurium. Dibucaine is an amide local anesthetic that

Page 25: Manual Regional Carlo 2010

25 | P a g e

helps to identify those patients. Dibucaine binds strongly to normal plasma

pseudocholinesterase inhibiting its action. This inhibition is reported as a number from 1-

100 representing the percentage of normal enzyme inhibition, the larger the number the

larger the proportion of normal enzyme. A number of 80 or higher means that dibucaine

is able to inhibit at least 80% of the enzyme and that the patient is a normal homozygous.

A dose of succinylcholine will last 4-6 min. A dibucaine number of 50 means that the

patient is heterozygous and that the effect of succinylcholine will be prolonged to up to

30 min. A number of 20 is related to the homozygous atypical enzyme and the effect of

succinylcholine could be expected to last up to 6 h (incidence 1:3,300).

LOCAL ANESTHETIC TOXICITY

The capacity of a local anesthetic to produce systemic toxicity is directly related

to plasma level of unbound drug. This plasma level depends on:

1. Total dose

2. Net absorption, which depends on: vasoactivity of the drug, site vascularity and

use of a vasoconstrictor

3. Metabolism and elimination of the drug from the circulation

Brown et al reported a 1.2 in 10,000 incidence of systemic toxicity after epidural

anesthesia and 19 in 10,000 after peripheral nerve blocks. CNS signs of toxicity usually

precedes CV manifestations. According to Mather et al, central nervous system (CNS)

and cardiovascular (CV) effects are “poorly correlated with arterial drug concentrations”

and better correlated with the “respective regional venous drainage”. According to them,

lung uptake reduces the drug concentration by 40% and slower injection (3 min

compared to 1 min) achieves similar decreases (Reg Anesth Pain Med 2005; 30: 553-66).

Peak local anesthetic blood levels are directly related to the dose administered at

any given site. However the vascularity of the site at similar doses is very important in

determining different plasma levels. The absorption of local anesthetics from different

sites is from highest to lowest: endotracheal > intercostal > caudal > epidural > plexus

blocks > sciatic/femoral > subcutaneous infiltration. Generally the administration of a 100-mg dose of lidocaine in the epidural or

caudal space results in approximately a 1 mcg/mL peak blood level in an average adult.

The same dose injected into less vascular areas (e.g., brachial plexus axillary approach or

subcutaneous infiltration) produces a peak blood level of app 0.5 mcg/mL. The same

dose injected in the intercostal space produces a 1.5 mcg/mL plasma level.

Peak blood levels may also be affected by the rate of biotransformation and

elimination. In general this is the case only for very actively metabolized drugs such as 2-

chloroprocaine, which has a plasma half-life of about 45- 60 seconds.

For amide local anesthetics like lidocaine peak plasma levels after regional

anesthesia primarily result from absorption and usually occur within 1 h (please see

difference with tumescent anesthesia).

Rodriguez et al studied 10 end-stage renal disease patients coming for A-V fistula

(Eur J Anaesthesiol 2001; 18: 171-6). The patients received an axillary block with a total

of 650 mg of plain mepivacaine. Plasma levels were studied during 150 min. Peak levels

Page 26: Manual Regional Carlo 2010

26 | P a g e

of 8.28 mcg/mL (range 3.83-11.21) were obtained (normal 5 mcg/mL) within 60 min and

decreased steadily thereafter. Patients did not exhibit signs of toxicity despite these high

plasma levels.

This is in contrast with a case report by Tanoubi et al (Ann Fr Anesthe Reanim

2006; 25: 33-5), where an end-stage renal patient for A-V fistula received an axillary

block with 375 mg (25 mL) of 1.5% mepivacaine and the patient presented with

dysarthria, mental confusion and loss of consciousness without convulsions or

arrhythmia. Mepivacaine plasma level at the time of symptoms was 5.1 mcg/mL

Tumescent (diluted) anesthesia for liposuction

The use of highly diluted concentrations of lidocaine (0.1% or less) plus

epinephrine (usually 1 mg per liter or 1:1,000,000) allows for painless and bloodless

liposuction procedures. Lidocaine bounds to tissue proteins in this subdermal drug

reservoir from where it is subsequently slowly released into the systemic circulation.

Diluted lidocaine, along with epinephrine-induced vasoconstriction, makes

systemic uptake so slow as to match the liver maximum lidocaine clearance capacity of

250 mg/h. Therefore, according to de Jong, “the blood level remains below 5 mcg/mL

toxic threshold, despite the administration of many times (e.g., 35 mg/kg) the

conventional upper dose limit of undiluted full strength lidocaine” (Int J Cosmetic Surg

2002; 4: 3-7).

Peak plasma levels of lidocaine using tumescent technique occur between 5-17

hours compared to less than 1 h for common infiltration.

Central nervous system toxicity

Toxic plasma levels are usually produced by inadvertent intravascular injection.

This is the basis for fractionating of the dose and the use of vasoconstrictors. Toxic

plasma levels could rarely result from the slow absorption from the injection site. A

sequence of symptoms may include numbness of the tongue, lightheadedness, tinnitus,

restlessness, tachycardia, convulsions and respiratory arrest. There is no evidence that

patients suffering from seizure disorders are at any increased risk for CNS local

anesthetic toxicity, including seizures. The site of action for local anesthetic-induced

seizures seems to be the amigdala, part of the limbic system, in the base of the brain.

Cardiovascular system toxicity

The cardiovascular manifestations usually follow the CNS effects (therapeutic

index). The exception is bupivacaine, which can produce cardiac toxicity at sub

convulsant concentrations.

Rhythm and conduction are rarely affected by lidocaine, mepivacaine and

tetracaine, but bupivacaine and etidocaine can produce ventricular arrhythmias.

EKG shows a prolongation of PR and widening of the QRS

The incidence of CV toxicity with local anesthetics is higher in pregnancy due to

higher proportion of unbound fraction.

CV toxicity is increased under conditions of hypoxia and acidosis.

Page 27: Manual Regional Carlo 2010

27 | P a g e

Toxic plasma concentration thresholds

The following are accepted plasma levels of selected local anesthetics, above

which systemic effects are expected in humans:

Lidocaine 5 mcg/mL; mepivacaine 5 mcg/mL; bupivacaine 1.5 mcg/mL;

ropivacaine 4 mcg/mL

Management of systemic toxicity

The best treatment for toxic reactions is prevention. When local anesthetic-

induced seizures occur, hypoxia, hypercarbia and acidosis develop rapidly. ABC

(Airway, Breathing and Circulation) is the mainstay of treatment. Administration of O2

by mask, or ventilation support by bag and mask, is often all that is necessary to treat

seizures. If seizures interfere with ventilation, benzodiazepines, propofol or thiopental

can be used. The use of succinylcholine effectively facilitates ventilation and, by

abolishing muscular activity, decreases the severity of acidosis. However neuronal

seizure activity is not inhibited and therefore, cerebral metabolism and oxygen

requirements remain increased.

In an interesting study by Mayr et al, out of Innsbruck, Austria (Anesth Analg

2004; 98: 1426-3), the authors induced cardiac arrest in 28 pigs by administering 5 mg/kg

of 0.5% bupivacaine and stopping ventilation until asystole occurred. CPR was initiated

after 1 min of cardiac arrest. After 2 min the animals received every 5 min either

epinephrine alone; vasopressin alone; epinephrine plus vasopressin or placebo IV. In the

vasopressin/epinephrine group all pigs survived and in the placebo group all pigs died. In

the vasopressin alone 5 of 7 survived and in the epinephrine group 4 of 7 survived. This

is in line with current ACLS recommendations of using one single dose of 40U of

vasopressin IV before using epinephrine.

Little information is available regarding the treatment of local anesthetic

cardiovascular toxicity in humans. Animal data suggest:

1. Vasopressin 40 U, IV, single dose, one time only followed by, if needed,

high doses of epinephrine (1 mg IV every 3-5 minutes) to support heart

rate and blood pressure.

2. Atropine may be useful for bradycardia.

3. DC cardioversion is often successful.

4. Ventricular arrhythmias are probably better treated with amiodarone than

with lidocaine. Amiodarone is used as for ACLS, 150 mg over 10 min,

followed by 1 mg/min for 6 hrs then 0.5 mg/min. Supplementary infusion

of 150 mg as necessary up to 2 g. For pulseless VT or VF, initial

administration is 300 mg rapid infusion in 20-30 mL of saline or dextrose

in water.

Bupivacaine toxicity and use of lipid emulsion to treat it

Bupivacaine cardiac toxicity was highlighted in an editorial report by Albright in

1979, in which he described several cases of refractory cardiac arrests in association with

the use of bupivacaine (Anesthesiology 1979; 51:285-7). In 2003, Weinberg and

Page 28: Manual Regional Carlo 2010

28 | P a g e

colleagues from the University of Illinois, published an interesting paper describing the

use of a 20% lipid emulsion in combination with cardiac massage to successfully return

normal hemodynamics to 9 out of 9 dogs, after asystole brought by a bolus injection of

10 mg/kg of bupivacaine (Reg Anesth Pain Med 2003; 28:198-202).

The results of this study led them to recommend treating bupivacaine-associated

cardiac arrest with a 20% lipid emulsion IV. The treatment protocol includes a 1 mL/kg

bolus of 20% lipid emulsion (such as intralipid), followed by an infusion of 0.25

mL/kg/min for 10 min, and the continuation of basic life support. The bolus can be

repeated every 5 min, up to three times as needed. The maximum dose of 20% lipid

emulsion is not known, but the authors suggest that more than 8 mL/kg would not likely

be needed, nor successful, if lower doses do not work. This protocol will deliver a

significant volume load to the patient. The paper was accompanied by an editorial by

Groban and Butterworth from Wake Forest, in Winston-Salem, North Carolina. They

believe that the most likely mechanism of action of lipid emulsion is that “in some way

the lipid is serving to more rapidly remove LA molecules from whatever binding site

serves to produce the cardiovascular depression that has come to be known as

bupivacaine toxicity”.

ACLS protocols must be followed with prompt defibrillation and use of pressors

like vasopressin followed by epinephrine, to support coronary perfusion if necessary.

Amiodarone should be favored over lidocaine to treat arrhythmias and initiate the lipid

emulsion at the “earliest sign of severe local anesthetic-induced cardiac toxicity.

In 2006 Rosenblatt et al (Anesthesiology 2006; 105: 217-8) published a case

report of successful use of 20% lipid emulsion (Intralipid, Baxter Pharmaceuticals) on a

58-year old male who developed a cardiac arrest, presumably linked to bupivacaine after

an interscalene block. They described that after 20 min of cardiac compressions and with

the patient in asystole, 100 mL of intralipid IV was given resulting in an apparent

“immediate” return of patient’s rhythm. This dose is higher than the recommended 1

mL/kg. A continuous infusion of intralipid was given at 0.5 mL/kg/min for 2 h. The

patient was extubated 2.5 after hours after the episode, without any apparent neurological

sequelae. In an accompanying editorial, Weinberg suggested having 20% lipid emulsion

available in all sites where local anesthetics are used.

Also in 2006, soon after Rosenblatt’s report, Litz et al reported a case of

successful use of intralipid after ropivacaine-induced asystole. More recently, in March

2008, McCutchen and Gerancher reported a case of ventricular tachycardia treated with

150 mg of amiodarone, 10 mL of 20% intralipid and a synchronized countershock of 120

J, after which there was a prompt return to normal sinus rhythm. The authors speculate

that the use of intralipid might have prevented the patient from going into cardiac arrest.

In summary:

1. Evidence is accumulating on the beneficial effect of a 20% lipid emulsion to treat

bupivacaine-related cardiac toxicity.

2. Propofol has the same vehicle than intralipid, but only half the concentration

(10%). Giving propofol probably will not provide enough lipids, but instead it

will produce a negative inotropic effect due to the presence of the active

ingredient di-isopropylphenol (anesthetic action), exacerbating cardiac

Page 29: Manual Regional Carlo 2010

29 | P a g e

depression. Therefore, propofol is not indicated to treat local anesthetic-induced

cardiac toxicity.

Maximum dose

Regional anesthesiologists perform peripheral nerve blocks with an amount of

local anesthetics that usually exceeds what traditionally have been considered “maximum

recommended doses”. However the traditional recommendations are based on

extrapolation from animal data that do not necessarily apply to clinical practice.

According to Rosenberg et al, the common recommendations for maximum doses, as

suggested by the literature, “are not evidence based” (Reg Anesth Pain Med 2004; 29:

564-575), and according to Milroy have proven to be “poor approximation of safety”

(Reg Anesth Pain Med 2005; 30: 513-515).

It is known that peak plasma levels do not correlate with patient size or body

weight. Many practitioners have called to review these guidelines to better reflect the

reality of clinical practice. The American Society of Regional Anesthesia convened a

“Conference in Local Anesthetic Toxicity” with a panel of experts in 2001, to discuss the

subject. Many papers related to that conference have been published. In a review article

by Rosenberg et al (just cited) the authors argue that the safe doses instead should be

block specific and related to patient’s age (e.g., epidural), organ dysfunction (especially

for repeated doses) and whether the patient is pregnant. They suggest also adding

epinephrine 2.5 to 5 mcg/mL, when not contraindicated.

The fact is that most of the systemic toxicity occurs with unintentional direct

intravascular injection (Mather et al, Reg Anesth Pain Med 2005; 30: 553-566).

Methemoglobinemia

Normal hemoglobin contains an iron molecule in the reduced or ferrous form

(Fe2+

), the only form suitable for oxygen transport by hemoglobin. When hemoglobin is

oxidized, the iron molecule is converted into the ferric state (Fe3+

) or methemoglobin.

Methemoglobin lacks the electron that is needed to form a bond with oxygen and

therefore it is incapable of oxygen transport. Because red blood cells are continuously

exposed to various oxidant stresses, blood normally contains approximately 1%

methemoglobin levels. Prilocaine and benzocaine can oxidize the ferrous form of the

hemoglobin to the ferric form, creating methemoglobin. It is more frequently seen with

nitrates like nitroglycerin. When MetHb exceeds 4 g/dL cyanosis can occur.

Prilocaine doses of more than 600 mg are needed to produce clinically significant

methemoglobinemia. Depending on the degree, methemoglobinemia can lead to tissue

hypoxia. The oxyHb curve shifts to the left (P50 < 27 mmHg). MetHb has a larger

absorbance than Hb and 02Hb at 940 nm, but simulates Hb at 660 nm. In the presence of

high MetHb concentrations the SaO2 falsely approaches 85%, independent of the actual

arterial oxygenation. Diagnosis needs clinical suspicion and confirmation by blood

analysis.

Methemoglobinemia is easily treated by the administration of methylene blue (1-

2mg/kg of a 1% solution over 5 min) or less successfully with ascorbic acid (2 mg/kg).

Page 30: Manual Regional Carlo 2010

30 | P a g e

Allergy

True allergy (type I or IgE mediated) to local anesthetics is rare and presents

within minutes after the exposure. It is relatively more frequent with esters, which are

metabolized to para-amino-benzoic acid (PABA). PABA is frequently used in the

pharmaceutical and cosmetic industries. Allergy to amide local anesthetics is exceedingly

rare. There is no cross allergy between esters and amides. However use of methylparaben

as a preservative in multidose vials can elicit allergy in patients allergic to PABA.

Delayed hypersensitivity reactions (type IV) are T-cell mediated and present 24 to

48 h after exposure. There are few cases in the literature of delayed hypersensitivity to

lidocaine, but recent reports suggest it may be more frequent than previously reported.

The North American Contact Dermatitis Group found that 0.7 % of patients who were

patch tested in 2001-02 demonstrated delayed allergy to lidocaine (ASRA News,

February 2006).

Eutectic mixture of local anesthetics (EMLA)

EMLA cream is a 1:1 mixture of 5% lidocaine and 5% prilocaine. One gram of

EMLA contains 25 mg of lidocaine, 25 mg of prilocaine, an emulsifier, a thickener and

distilled water. EMLA is a liquid at room temperature, containing up to 80%

concentration of the uncharged base form of local anesthetic, which confers better dermal

penetration. Anesthesia onset takes between 45 to 60 minutes. Its main use is in children.

One or 2 grams of EMLA cream are applied per 10 cm2 of skin and covered with an

occlusive dressing (maximum application area 2000 cm2

or 100 cm2 in children less than

10 kg).

Drug interactions

Local anesthetics potentiate the effects of non-depolarizing muscle relaxants.

Simultaneous administration of succinylcholine and ester local anesthetics, both

metabolized by pseudocholinesterases, may potentiate the effect of each other.

Cimetidine and propranolol decrease hepatic blood flow and amide local anesthetic

clearance increasing the potential for systemic toxicity. Opioids and alpha-2 adrenergic

agonists potentiate the effects of local anesthetics and vice versa.

Profile summary of selected agents

1. Procaine: Type: ester

Pka: 8.9

Protein biding: 5%

Characteristics: intermediate onset, low potency, short duration. Very short half-

life (20 sec).

Other: it provides a short-duration spinal (potential benefit on outpatients).

Page 31: Manual Regional Carlo 2010

31 | P a g e

2. 2-Chloroprocaine:

Type: ester

Pka: 9.3

Protein binding: negligible

Characteristics: very fast onset, despite high pka (ability to use higher

concentrations could be the reason). Short duration (it has 30 minutes 2-segment

regression in epidural). Very short half-life (30 sec).

Other: The original preparation contained sodium metabisulfite as a preservative.

It was associated with serious neurological deficits when a large injection,

planned for epidural, ended intrathecally. A second preservative, ethylenediamine

tetra-acetic acid (EDTA) was associated with severe muscle spasm after epidural

in ambulatory patients. EDTA chelates ionized calcium and this side effect may

be secondary to action on paraspinal muscles.

The present solution is prepared without preservatives, and no back spasms have

been reported.

3. Tetracaine:

Type: ester

Pka: 8.6

Protein binding: 85%

Characteristics: slow onset, high potency, long duration. Short plasma half-life

(2.5 to 4 min).

Other: early experience with this product at high doses resulted in CNS toxicity,

giving it a bad reputation, mostly undeserved. We still use it occasionally in our

practice, as lyophilized crystals dissolved in liquid mepivacaine for a final

concentration of 0.2% tetracaine. It prolongs duration of surgical anesthesia in

peripheral nerve blocks to 4-6 h. Tetracaine also is the drug that gets the longest

prolongation from adding epinephrine to spinal anesthesia (up to 60% in the

lumbar dermatomes).

4. Cocaine:

Type: ester

Pka: 8.6

Protein binding: ?

Characteristics: slow onset, short duration. Elimination half life 60-90 min.

Urinary excretion of unchanged cocaine is usually less than 1%, but it can be up

to 9% especially in acid urine. At the end of 4 hours, most of the drug is

eliminated from the plasma. Cocaine metabolites (benzoylecgonine and ecgonine)

may be present in the urine for 24-36 hours, but some metabolites may be

identified for up to 144 h after administration (Ellenhom and Barceloux, 1988).

Other: It produces vasoconstriction, while most of the LA with the exception of

ropivacaine, produce some degree of vasodilation. It interferes with the reuptake

of cathecolamines, resulting in hypertension, tachycardia, arrhythmias and

myocardial ischemia. It is used mainly for topical anesthesia of the nose. Doses

below 100 mg (2.5 mL) are usually safe.

Page 32: Manual Regional Carlo 2010

32 | P a g e

Cocaine can potentiate cathecolamine-induced arrhythmias by halothane,

theophylline or antidepressants. Cocaine can induce coronary vasospasm and

potential myocardial ischemia, without the need for coronary artery disease.

Mixtures of lidocaine and phenylephrine are safer alternatives.

5. Benzocaine:

Type: ester

Pka 3.5

Characteristics: slow onset, short duration and the only LA with a secondary

amine structure that limits its ability to pass through membranes (topical use

only).

Other: Doses higher than 300 mg can induce methemoglobinemia.

6. Lidocaine:

Type: amide

Pka: 7.8

Protein binding: 65%

Characteristics: intermediate onset and duration, elimination half-life 45-60 min.

Other: it is versatile (topical, infiltration, IV regional, neuraxial, antiarrhythmic)

and widely used. Spinal use is associated with around 30% of TNS, especially

with lithotomy position, knee arthroscopy and obesity. Lowering the

concentration does not eliminate the problem with doses larger than 40 mg. Doses

of 25-40 mg highly reduce the incidence of TNS.

7. Mepivacaine:

Type: amide

Pka: 7.6

Protein binding: 75%

Characteristics: intermediate onset and duration. Elimination half-life is 2-3 h in

adults and 8-9 h in neonates.

Other: It produces less vasodilation than lidocaine. It has been used in spinal

anesthesia. It has lower (but not zero) incidence of TNS.

It is the agent we most commonly use for peripheral nerve blocks. A 1.5% of

plain solution provides a short onset and dense surgical anesthesia lasting 2-3 h

(3-4 h with 1:400,000 epinephrine). Prolonged postoperative analgesia, as with all

other LA, is negligible after single-shot blocks.

The primary oxidative metabolic pathway for mepivacaine is less developed in

neonates resulting in slower metabolism of mepivacaine in newborns than in

adults (Raj’s Textbook of Regional Anesthesia).

8. Bupivacaine:

Type: amide

Pka: 8.1

Protein binding: 95%

Characteristics: high potency, slow onset, long duration. Elimination half-life 3-

3.5 h in adults and around 8 h in neonates.

Page 33: Manual Regional Carlo 2010

33 | P a g e

Other: lower concentrations (0.25% and less) produce analgesia with increased

motor sparing (desirable in outpatients and obstetrics). Commercial bupivacaine is

a 50:50 racemic mixture of the R and S enantiomers. Cardiac arrest associated

with bupivacaine is difficult to treat possibly due to its high protein binding and

high lipid solubility (please see toxicity).

9. Ropivacaine:

Type: amide

Pka: 8.2

Protein binding: 94%

Characteristics: onset and duration similar to bupivacaine, with slight lesser

potency. Elimination half-life 1-3 h in adults.

Like bupivacaine, it is chemically related to mepivacaine, but as opposed to most

local anesthetics, it is supplied as the pure S enantiomer of the drug. The S

enantiomer is associated with less cardiac toxicity, intermediate between that of

lidocaine and bupivacaine.

Other: It is a weak vasoconstrictor (only one other than cocaine). At lower

concentrations (less than 0.5%) it may show a greater selectivity for sensory than

motor blockade than bupivacaine.

10. Levobupivacaine: Type: amide

Pka: 8.1

Protein binding: 97%

Characteristics: S enantiomer of bupivacaine, very similar to ropivacaine.

Not available at this time in the US.

Page 34: Manual Regional Carlo 2010

34 | P a g e

References

1. Lou L, Sabar R, Kaye A: Local Anesthetics, In: Raj P (ed), Textbook of Regional

Anesthesia. New York, Churchill Livingstone, 2002, pp 177-213

2. Brown DL, Fink R: The History of Neural Blockade and Pain Management, In:

Cousins MJ, Bridenbaugh PO (eds), Neural Blockade, 3rd

edition. Philadelphia,

Lippincott-Raven, 1998, pp 3-32

3. Strichartz GR: Neural Physiology and Local Anesthetic Action, In: Cousins MJ,

Bridenbaugh PO (eds), Neural Blockade, 3rd

edition. Philadelphia, Lippincott-

Raven, 1998, pp 35-54

4. Tucker GT, Mather LE: Properties, Absorpton, and Disposition of Local

Anesthetic Agents, In: Cousins MJ, Bridenbaugh PO (eds), Neural Blockade, 3rd

edition. Philadelphia, Lippincott-Raven, 1998, pp 55-95

5. Covino BG, Wildsmith JAW: Clinical Pharmacology of Local Anesthetic Agents,

In: Cousins MJ, Bridenbaugh PO (eds), Neural Blockade, 3rd

edition.

Philadelphia, Lippincott-Raven, 1998, pp 97-128

6. Morgan GE, Mikhail MS, Murray MJ: Clinical Anesthesiology, 4th

edition. New

York, McGraw-Hill, 2006, pp 263-288

7. Mulroy MF: Regional Anesthesia, 3rd

edition. Philadelphia, Lippincott Williams

& Wilkins, 2002, pp 1-63

8. Liu SS, Joseph RS: Local Anesthetics, In: Barash PG, Cullen BF, Stoelting RK

(eds), Clinical Anesthesia. Philadelphia, Lippincott Williams & Wilkins, 2006, pp

453-471

9. DiFazio CA, Carron H, Grosslight KR, et al. Comparison of ph-adjusted lidocaine

solutions for epidural anesthesia. Anesth Analg 1986; 65: 760-64

10. Hilgier M. Alkalinization of bupivacaine for brachial plexus block. Reg Anesth

1985;10: 59-61

11. Booker PD, Taylor C, Saba G. Perioperative changes in α1-acid glycoprotein

concentrations in infants undergoing major surgery. Br J Anaesth 1996; 76: 365-

368

12. Stoelting RK: Pharmacology and Physiology in Anesthetic Practice, 3rd

edition.

Philadelphia, Lippincott-Raven, 1999, pp158-181

13. Tetzlaff JE: Local anesthetics and adjuvants for ambulatory anesthesia. In: Steele

SM, Nielsen KC, Klein SM (eds), Ambulatory Anesthesia Perioperative

Analgesia. New York, McGraw-Hill, 2005, pp 193-205

14. Weinberg GL et al. Lipid emulsion infusion rescues dogs from bupivacaine-

induced cardiac toxicity. Reg Anesth Pain Med 2003; 28:198-202

15. Mayr VD, Raedler C, Wenzel V, et al. A comparison of epinephrine and

vasopressin in a porcine model of cardiac arrest after rapid intravenous injection

of bupivacaine. Anesth Analg 2004; 98:1426-3

16. Neal JM. Effects of epinephrine in local anesthetics on the central and peripheral

nervous systems: Neurotoxicity and neural blood flow. Reg Anesth Pain Med

2003; 28:124-134

17. Rosenberg PH, Veering VT, Urmey WF. Maximum recommended doses of local

anesthetics: A multifactorial concept. Reg Anesth Pain Med 2004; 29: 564-575.

Page 35: Manual Regional Carlo 2010

35 | P a g e

18. Mulroy MF. Local anesthetics: Helpful science, but don’t forget the basic clinical

steps (editorial). Reg Anesth Pain Med 2005; 30: 513-515.

19. Mather LE, Copeland SE, Ladd LA. Acute toxicity of local anesthetics:

Underlying pharmacokinetic and pharmacodynamic concepts (A review article).

Reg Anesth Pain Med 2005; 30: 553-566

20. Horlocker TT. One hundred years later, I can still make your heart stop and your

legs weak: the relationship between regional anesthesia and local anesthetic

toxicity. Reg Anesth Pain Med 2002; 27(6): 543-4

21. Mulroy MF. Systemic toxicity and cardiotoxicity from local anesthetics:

Incidence and preventative measures (editorial). Reg Anesth Pain Med 2002;

27(6): 556-61

22. Horlocker TT, Wedel DJ. Local, anesthetic toxicity-Does product labeling reflect

actual risk? Reg Anesth Pain Med 2002; 27(6): 562-567

23. Weinberg GL. Current concepts in resuscitation of patients with local anesthetic

cardiac toxicity. Reg Anesth Pain Med 2002; 27(6): 568-575

24. Myer Leonard. Carl Koller: Mankind’s greatest benefactor? The story of local

anesthesia. J Dent Res 1998; 77:535-8

25. De Jong R. Tumescent anesthesia: lidocaine dosing dichotomy. Int J Cosmetic

Surg 2002; 4: 3-7

26. Nordstrom H, Stange K. Plasma lidocaine levels and risks after liposuction with

tumescent anaesthesia. Acta Anaesthesiol Scand 2005; 49: 1487-1490

27. Rosenblatt MA, Abel M, Fischer GW, et al. Successful use of a 20% lipid

emulsion to resuscitate a patient after a presumed bupivacaine-related cardiac

arrest. Anesthesiology 2006; 105:217-8

28. Litz RJ, Popp M, Stehr SN, et al. Succesful resuscitation of a patient with

ropivacaine-induced asystole after axillary plexus block using lipid infusion.

Anaesthesia 2006; 61: 800-801

29. Rodriguez J, Quintela O, Lopez-Rivadulla M, et al. High doses of mepivacaine

for brachial plexus block in patients with end-stage chronic renal failure. A pilot

study. Eur J Anaesthesiol 2001; 18: 171-176

30. Kamibayashi T, Maze M. Clinical uses of 2-adrenergic agonists. Anesthesiology

2000; 93:1345-9

31. Tanoubi I, Vialles N, Cuvillon P, et al. Systemic toxicity with mepivacaine

following axillary block in a patient with terminal kidney failure. Ann Fr Anesth

Reanim 2006; 25:33-5

32. McCutchen T, Gerancher JC. Early Intralipid Therapy May Have Prevented

Bupivacaine-Associated Cardiac Arrest. Reg Anesth Pain Med 2008; 33: 178-180

33. Bigeleisen PE. Nerve puncture and apparent intraneural injection during

ultrasound-guided axillary block does not invariably result in neurological injury.

Anesthesiology 2006; 105: 779-783

Page 36: Manual Regional Carlo 2010

36 | P a g e

CHAPTER 3

NEURAXIAL ANESTHESIA

Page 37: Manual Regional Carlo 2010

37 | P a g e

SPINAL ANESTHESIA

It is one of the easiest and most reliable techniques of regional anesthesia. The

very small doses of local anesthetics used to produce spinal anesthesia are devoid of

direct systemic effects.

In 1885 James Corning, an American neurologist, was the first person to use

cocaine intrathecally to treat some neurological conditions. Augustus Bier, a German

surgeon, was the first person to use intrathecal cocaine to produce surgical anesthesia. In

a classic paper published in 1899, he described the failed attempt, by his assistant

Hildebrandt, to perform a spinal anesthesia on him, and his successful spinal on

Hildebrandt. Both of them became the first patients suffering from post dural puncture

headaches.

Anatomy

The spinal canal has a protective sheath composed of three layers. From the

outside to the inside they are: duramater, arachnoid and piamater. The potential space

between the dura and arachnoid is called subdural space. The cerebrospinal fluid (CSF)

flows between the arachnoid and piamater in the space called subarachnoid space.

The spinal cord begins cranially at the foramen magnum, as a continuation of the

medulla oblongata. It terminates caudally at the conus medullaris, which in the adult

corresponds to the level of the lower border of L1, and in the young child to the upper

border of L3. From this end, a prolongation of the piamater called the filum terminale

attaches the spinal cord to the coccyx. The dural sac itself ends at the level of the second

sacral vertebra.

The spinal cord is composed of a core of gray matter surrounded by white matter.

The gray matter on cross section has an H shape, with ventral (motor) and dorsal

(sensory) horns. The white matter is described as having anterior, lateral and posterior

white columns.

There are 31 pairs of spinal nerves; each one being formed by two roots, a ventral

or motor root and a dorsal or sensory root. The dorsal root has the dorsal root ganglion.

Because the spinal cord of an adult is shorter than the vertebral column, the spinal nerves

descend a variable distance in the spinal canal before exiting through the intervertebral

foramen. The most distal lumbar and sacral nerves travel the longest distance inside the

spinal canal, forming what is known as the cauda equina. As the spinal nerve pierces the

dural sac, it draws with it a dural sleeve. The spinal nerves exit through the intervebral

foramen, formed between two vertebrae. There are 8 cervical nerves. The first cervical

nerve exits through the occipital bone and C1, the 8th

cervical nerve exits between C7 and

T1. Distal to T1 each spinal nerve exits below the corresponding vertebra.

The vertebral column has a series of curvatures in the anteroposterior plane. The

cervical and lumbar curvatures have an anterior convexity (lordosis) and the thoracic and

sacral have posterior convexity (xiphosis). These curvatures play a role in the spread of

the local anesthetic solution, as we will review later.

The blood supply to the spinal cord comes from one anterior spinal artery and two

posterior spinal arteries. These arteries anastomose to form longitudinal vessels,

reinforced by segmental arteries that enter the vertebral canal trough the intervertebral

Page 38: Manual Regional Carlo 2010

38 | P a g e

foramina. The anterior two thirds of the spinal cord are supplied by the anterior spinal

artery reinforced in the neck by branches of the vertebral artery.

In the thoracic region the anterior spinal artery receives only a few radicular

arteries from the aorta. In the lumbar region a large branch called radicularis magna or

artery of Adamkiewicz, reinforces the anterior spinal artery. It arises 78% of the times on

the left side, and typically enters the spinal canal through a single intervertebral foramen

between T8 and L3. This important branch is at risk of damage during retroperitoneal

dissections (e.g., surgery on the distal aorta), which could lead to ischemia of the spinal

cord. A case of transient paraplegia after neurolytic celiac plexus block on a pancreatic

cancer patient was reported in 1995 by Wong and Brown. The proposed mechanism was

reversible arterial spasm post injection of ethanol solution.

Planes between the surface of the skin and subarachnoid space

The needle used to perform a diagnostic spinal tap or a spinal anesthesia needs to

cross the skin, subcutaneous tissue, supraspinous ligament, interspinous ligament,

ligamentum flavum, duramater and arachnoid, before reaching the subarachnoid space

and CSF. The space between the ligamentum flavum and duramater is the epidural space.

Cerebrospinal fluid

It is primarily formed in the choroids plexus of the cerebral ventricles. The CSF

flows from the lateral ventricles to the third and fourth ventricles, and from there to the

cisterna magna. It flows then around the brain and spinal cord, within the subarachnoid

space. The CSF is absorbed into the venous system of the brain by the villi in the

arachnoid membrane. CSF is formed and reabsorved at a rate of 0.3-0.4 mL/min.

The CSF volume in the brain is between 100-150 mL. The volume of CSF below

T12, where most of the spinal anesthetics are performed is, according to Hogan and

collaborators, widely variable among individuals, ranging from 28-80 mL. CSF volume is

decreased with increased abdominal pressure, like the one accompanying pregnancy and

obesity. Therefore, increased abdominal pressure could potentially lead to higher spread

of a neuraxial blockade.

Composition of cerebrospinal fluid and serum in humans

CSF Serum

Sodium (mEq/L) 141 140

Potassium (mEq/L) 2.9 4.6

Calcium (mEq/L) 2.5 5.0

Magnesium (mEq/L) 2.4 1.7

Chloride (mEq/L) 124 101

Bicarbonate (mEq/L) 21 23

Glucose mg/100mL) 61 92

Protein (mg/100mL) 28 7000

pH 7.31 7.41

Osmolality (mOsm/kg H2O) 289 289

Page 39: Manual Regional Carlo 2010

39 | P a g e

Site of action

The nerve root is the main site of action for both spinal and epidural anesthesia. In

spinal anesthesia the concentration of local anesthetic in CSF is thought to have minimal

effect on the spinal cord itself.

Indications

Abdominal and lower extremity procedures are the most common. It has been

used for lumbar spine surgery. Saddle blocks are frequently used for rectal surgery.

Baricity

It is the result of dividing density of the local anesthetic solution by that of the

CSF. The density of CSF has a mean value of 1.0003. If the baricity is 1.0 it is by

definition isobaric; if greater than 1 it is hyperbaric and if less than 1 it is hypobaric.

1. Hypobaric solutions

Tetracaine is the local anesthetic most frequently used for hypobaric spinal

anesthesia. Solutions of 0.1% to 0.33% tetracaine in water are reliably hypobaric

in all patients. The most common uses of hypobaric solutions are for rectal

procedures in jackknife position and for hip surgery injecting in lateral position

with the surgical side up.

2. Isobaric solutions

Tetracaine and plain bupivacaine diluted with CSF make good isobaric solutions.

These solutions stay very close to the point of injection.

3. Hyperbaric solutions

The easiest, safest and most widely used way of providing spinal anesthesia. The

solution is rendered hyperbaric by adding glucose. Gravity and patient’s position

determines the spread. In supine position L3 and T6 are the highest points of the

spine and subsequently they become the limits for spread.

Determinants of local anesthetics spread in the spinal fluid

1. Major factors

Baricity acting together with gravity

Position of patient (except isobaric solutions)

Dosage, rather than volume or concentration

Baricity is the main factor that determines local anesthetic spread in the

subarachnoid space. It obviously works in conjunction with gravity and patient position.

When plain local anesthetics are used, total dose is more important than injected volume

or concentration. Van Zundert et al reported in 1996, that a 70 mg dose of plain

subarachnoid lidocaine produced the same quality of spinal block over a wide range of

concentrations and volumes. Sheskey et al in 1983 demonstrated similar sensory levels

with 10 mg of plain bupivacaine, at different concentrations and volumes. However,

doses of 15-20 mg of plain bupivacaine produced higher sensory levels of spinal (T2-T4

Page 40: Manual Regional Carlo 2010

40 | P a g e

level) than 10 mg (T5-T8 level). When hyperbaric bupivacaine or tetracaine solutions are

used, similar levels of spinal blocks are obtained at different doses, when the

concentration is maintained constant. In the case of hyperbaric bupivacaine, it seems that

this applies as long as the dose is higher than 7.5 mg. Above this dose the level is

determined by baricity acting along with the curvatures of the spine, patient position, and

gravity. In general, the higher the spread the shorter the duration of the sensory blockade,

because the concentration of the drug decreases from the point of injection.

2. Minor factors

Level of injection

Increased abdominal pressure (obesity and pregnancy)

Patient height (only at extremes)

Coughing

Direction of needle bevel can affect spread of isobaric preparations. The bevel

should be directed toward the desired region.

3. No effect

Addition of vasoconstrictors

Barbotage (aspirating and injecting technique to produce CSF turbulence)

Age

Gender

Techniques

Sitting, midline approach

Sitting position is commonly used for neuraxial blocks. It may be the preferred

position in patients whose midline may be difficult to determine, like obese patients. The

position of the iliac crest is frequently used to determine the L4-L5 interspace. However,

accumulation of adipose tissue around the patient mid section, could lead to a higher-

than-desired level for needle placement.

The Closed Claims Project shows cases of spinal cord injury by the spinal needle,

in which the level of needle placement was grossly underestimated. I suggest instead

using the upper end of the intergluteal sulcus to determine the position of the sacral

hiatus. In adults the L5-S1 interspace is around 10 cm (4 inches) cephalad to this point

(height of the sacrum). This measurement in adults should always be distal to the

termination of the spinal cord at L1.

Using a hyperbaric solution in the sitting position, and leaving the patient in that

position for at least 5 minutes, produces a saddle block. However, up to 20 minutes is

necessary to wait, in the desired position, to achieve any appreciable “saddle” or

“lateralized” distribution blockade.

Lateral position

It is the position of choice in many institutions. The patient lies on his/her side. It

is more comfortable for the patient and decreases the risk for accidental fall and

vasovagal problems. The technique otherwise is similar to sitting position

Page 41: Manual Regional Carlo 2010

41 | P a g e

Paramedian approach

In some elderly patients, with calcified ligaments, it is difficult to advance the thin

spinal needle through the midline. The lateral approach is a good alternative in those

cases. The spinous process is identified and the point of entrance is marked about 2 cm

paramedian. The needle is directed slightly medial and cephalad.

Taylor Approach

Usually the L5-S1 interspace is the larger. A spinal technique through it is known

as Taylor approach. The entrance point is 1 cm medial and 1 cm caudal to the posterior

superior iliac spine directing the needle cephalad and toward the midline.

Anesthesia duration

The local anesthetic used and the rate at which it is removed from the

subarachnoid space determines duration. Elimination is entirely dependent on vascular

absorption and does not involve metabolism of local anesthetics within the subarachnoid

space. Absorption occurs in the subarachnoid space itself and in the epidural space (local

anesthetics cross the dura both ways).

Side effects and Complications

1. Hypotension It is the most frequent seen side effect. It is mainly the result of venous pooling

with decreased cardiac output secondary to sympathetic blockade. There is also a

small component of arteriolar dilation. However systemic blood pressure does not

decrease proportionally because of compensatory vasoconstriction, especially in

the upper extremities with intact sympathetic innervation. Even with total

sympathetic blockade after spinal anesthesia the decrease in systemic vascular

resistance is less than 15%. This is because arterioles retain intrinsic tone and do

not dilate maximally.

The magnitude of the blood pressure decrease depends on the extent of

sympathetic blockade, intravascular volume, and cardiovascular status. Preloading

the patient with 250-500 mL, while frequently used, is unsupported by the

evidence.

A mild vasopressor like ephedrine in 5-10 mg increments and fluid are all that is

usually necessary to treat hypotension. Ephedrine is usually the drug of choice

because it produces vasoconstriction and increases cardiac output.

Phenylephrine is a good second choice especially if tachycardia is present. It

causes vasoconstriction, and it could decrease the cardiac output.

Trendelenburg position can alleviate the venous pooling, but may produce an

even higher spinal level. Elevating the legs with the patient sitting at 30-45

degrees is a good compromise.

Page 42: Manual Regional Carlo 2010

42 | P a g e

2. Bradycardia When the sympathetic block reaches T2 level, the cardioacelerator fibers are

blocked and the vagus action is unopposed. The extent to which heart rate

decreases in response to total sympathetic block during spinal usually is moderate

(10-15%). However severe bradycardia and asystole have been reported in

normal patients during otherwise uneventful spinal anesthesia. It can occur even

in the absence of hypotension and can occur after 30-45 minutes of spinal.

The Bezold-Jarisch reflex has been implicated. This reflex would be triggered by

decreased venous return to the heart producing a paradoxical hypervagal

response. Early recognition and treatment is essential. Ephedrine, atropine and

in some cases epinephrine are indicated along with fluid replacement. |

3. Total spinal Spinal anesthetic that involves the cervical region. It is manifested by respiratory

arrest, bradycardia, hypotension and unconsciousness. The respiratory arrest most

likely is a manifestation of ischemia of the medullary respiratory center secondary

to intense hypotension and drop in cardiac output (complete sympathetic

blockade) severe enough to compromise cerebral circulation.

Block of phrenic nerve is not a likely cause. Management involves ABC with

control of the airway, ventilator support, use of vasopressors, and atropine and

fluid replacement as needed.

Miscellaneous physiologic effects

1. Respiratory Arterial gases are usually unaffected in patients breathing room air.

Tidal volume, maximum inspiratory volumes and negative intrapleural pressure

during inspiration are unaffected, despite intercostals muscle paralysis with high

thoracic levels. This is because diaphragmatic activity remains intact.

Expiratory volumes and total vital capacity are significantly diminished in high

thoracic spinal, as are maximum intrapleural pressures during forced exhalation,

and coughing. This is mainly due to paralysis of abdominal muscles.

2. Hepatic Hepatic blood flow decreases to the extent of hypotension to a degree similar than

after general anesthesia. Spinal anesthesia has not proven to be an advantage or

disadvantage in patients with liver disease. For intraabdominal surgery the

decrease in hepatic perfusion is mainly due to surgical manipulation.

3. Renal Renal blood flow as cerebral blood flow is autoregulated through a wide range of

arterial pressure. In the absence of renal vasoconstriction renal blood flow does

not decrease until mean arterial pressure decreases below 50 mm Hg. Thus, in the

absence of severe hypotension, renal blood flow and urinary output remain

unaffected during spinal anesthesia. Loss of autonomic bladder control results in

urinary retention. This is more frequent in males.

Page 43: Manual Regional Carlo 2010

43 | P a g e

4. Endocrine and metabolic Spinal anesthesia, but not general anesthesia, blocks the hormonal and metabolic

stress response associated with surgery. This response involves increases in

ACTH, cortisol, epinephrine, norepinephrine and vasopressin as well as activation

of the rennin-angiotensin-aldosterone system. However this effect seems to wear

off along with the spinal anesthesia, producing metabolic and hormonal responses

similar than after general anesthesia for the same operation.

5. Gastrointestinal The small intestine contracts during spinal and sphincters relax due to unopposed

vagus nerve activity. The combination of contracted gut and complete relaxation

of abdominal muscle provide good surgical conditions.

Other effects and complications

1. Nausea Frequent side effect due to imbalance of sympathetic and parasympathetic

visceral tone. Hypotension, bradycardia or hypoxia must be rule out.

Antiemetics like ondansetron or droperidol are usually effective.

2. Post dural puncture headache (PDPH) PDPH is due to CSF leak through the dural puncture site. The subsequent loss of

CSF pressure produces stretching of the meningeal coverings of intracranial

nerves whenever the upright position is assumed.

The pain characteristics, involving exacerbation in the upright position and relief

in the recumbent position, remain the main diagnostic tool. It is more frequent in

females, in younger patients and during pregnancy. The size and type of needle

are proven factors. Pencil point needles significantly reduce the risk.

Spinal needles are either cut-bevel (Quincke-type) or pencil-point (Whitacre-

type). It has been usually accepted that the collagen fibers of the duramater are

oriented longitudinally and that the bevel of a cutting needle should be oriented

vertically to reduce trauma to the dural fibers. This concept has been challenged

by Reina and collaborators. They found that dural fibers are arranged in laminas

with fibers in all different directions and not necessarily longitudinal. They also

showed that pencil point needles produce a more traumatic lesion in the dura than

cutting-point needles. They hypothesized that a more traumatic lesion may

stimulate more inflammation than a cleaner cut does. The inflammatory response

and edema would then limit the leakage of CSF. This observation agrees with the

surprisingly low incidence of PDPH after continuous spinal anesthesia with an

18-gauge epidural needle and a 20-gauge epidural catheter. The catheter might act

as foreign object producing an inflammatory reaction.

This low incidence can also, at least in part, reflect the fact that continuous spinal

are more frequently performed in older patients. Older age is accompanied by a

decreased risk of PDPH.

Page 44: Manual Regional Carlo 2010

44 | P a g e

In the issue of PDPH:

Pencil point needles less than or equal to 22 gauge and cut-bevel needles

less than or equal to 27 gauge produce an incidence of PDPH of

approximately 1%.

Continuous spinal with 20 gauge catheters is not likely to produce PDPH

in an older patient population.

Obstetric patients undergoing spinal anesthesia with small pencil point

needles show a 3-4% rate of post dural puncture headache. Conservative

treatment involves bed rest, IV or oral fluids, acetaminophen and NSAIDs.

Hydration and caffeine stimulates production of CSF.

Epidural blood patch with 15-20 mL of autologous blood, injected at the

same original puncture level or one space below, is a very effective

treatment. The effect can be immediate or be delayed by a few hours. A

single blood patch is about 90% effective.

3. Transient neurological symptoms (TNS) Usually appears 12- 24 hrs after surgery and consist of mild to moderate pain or

sensory abnormalities in the lower back, buttocks or lower extremities. It resolves

between 6 hrs and 4 days. No patient with TNS has ever been reported to develop

neurological deficits or motor weakness. If present, other more serious diagnosis

must be ruled out: epidural hematoma, nerve root damage, cauda equine

syndrome. The first report appeared in the literature in 1993 when Schneider et al

published a series of 4 patients with buttocks pain after spinal.

Prospective, randomized studies have shown:

A higher (but variable) incidence after lidocaine spinal. Decreasing the

concentration of lidocaine to 0.5% does not appear to change this

incidence.

Its incidence seems related to other factors like: lithotomy (30-36%), knee

arthroscopy (18-22%), whereas the risk after supine position appears to be

relatively low (5 to 8%).

The cause for TNS is not well understood and could represent a mild and

reversible form of neuropathy. Many possible causes have been postulated: local

anesthetic toxicity, needle trauma, neural ischemia secondary to sciatic nerve

stretching, patient positioning, small gauge, pencil-point needles promoting local

anesthetic pooling, muscle spasm, early mobilization, etc. Because of the low

incidence of TNS after bupivacaine spinal, we could be reasonably sure than TNS

is not the result of the subarachnoid block per se, the needle or the position for it.

Even though neurotoxicity is frequently mentioned as possible cause for TNS,

a case can be made against it. Cauda equina syndrome (CES) is known to result

from local anesthetic toxicity; however the factors that increase CES (e.g., higher

doses/concentration of local anesthetics and the addition of vasoconstrictors), do

not have an effect on TNS.

Page 45: Manual Regional Carlo 2010

45 | P a g e

We know that TNS is mostly associated with lidocaine spinal, lithotomy

position, knee arthroscopy and ambulatory surgical status (obesity could be a

contributing factor) and that it is very rarely associated with bupivacaine spinal.

We also know that decreasing the concentration of lidocaine from 5% to 0.5%

does not decrease the incidence of TNS and that hyperosmolarity, hyperbaricity

and addition of glucose ARE NOT contributing factors.

First line of treatment is reassurance, NSAIDs, comfortable positioning and

heating pad. A second line of treatment can include narcotics and muscle

relaxants like cyclobenzaprine. Trigger point injections have been used with

reported success.

Eliminating lidocaine from subarachnoid block probably is not warranted at

this point. However do not use it for ambulatory surgery in lithotomy position or

knee arthroscopy (high risk). On the other hand, the incidence of TNS after

inguinal hernia with lidocaine spinal is only 8%, after C-section is 0-8% and after

tubal ligation is 3%, similar to non-pregnant patients undergoing surgery in the

supine position. Bupivacaine, even in small doses, increases discharge time.

Perhaps the combination of small doses of bupivacaine plus narcotics is the best

possible approach.

4. Cauda equina syndrome It is a rare but devastating complication resulting in perineal anesthesia and

possible loss of bowel and bladder control. Most of the reported cases have been

associated with the use of continuous spinal with microcatheters (30-gauge and

smaller) along with use of 5% hyperbaric lidocaine. Low flow rates promoting

pooling of concentrated drug around the sacral roots have been postulated as the

reason for this condition. In 1992 the FDA issued a safety alert that resulted in the

withdrawal of these catheters from the US market.

The incidence of CES increases with increased concentration of local anesthetics

as well as the addition of vasoconstrictors. There have been reports of cauda

equina syndrome after epidural anesthesia.

5. Back pain

As many as 40% of patients may complain of this annoying side effect. It is

postulated to be the result of stretching of the ligaments following the relaxation

of back muscles. This is similar to what is seen in up to 25-30% of patients

receiving general anesthesia in the supine position. It can also be the result of

localized inflammatory response with muscle spasm. Rest, local heat and

NSAIDs are the treatment of choice.

6. Hearing loss

Transient minor hearing loss has been described after spinal anesthesia. The risk

seems larger with larger-gauge needles and it might be the result of temporary

decrease in CSF pressure with traction of intracranial nerves.

The problem is mild but well documented with audiometry. It resolves on its own.

Page 46: Manual Regional Carlo 2010

46 | P a g e

7. Infection

Abscess or meningitis is rare. The development of meningitis after lumbar

puncture in bacteremic patients is a concern. Animal models suggest that

perioperative use of antibiotics eliminates this risk. Lumbar puncture in patients

infected with HIV is controversial. Neuraxial techniques including blood patch

have been performed on these patients without apparent problems. The risk has to

be evaluated on an individual basis.

Spinal anesthesia in the outpatient setting

A few years ago spinal anesthesia was favored for same day surgery patients.

However, widely available, poorly-soluble general anesthetic agents and LMA have

decreased its use. Home readiness involves short duration and in many institutions,

ability to void. Duration is a function of the agent and dose used. The spread of the agent

dictates the duration at a given dermatome. It is likely that the more segments blocked by

a given dose (more spread) the shorter the duration at any given segment. Hyperbaric

solutions and isobaric solutions injected rapidly with the bevel turned caudad concentrate

around the sacral roots and can delay sensory motor recovery and the ability to void. On a

milligram basis, isobaric preparations injected rapidly with the bevel facing cephalad are

more likely to improve home readiness and voiding. Procaine and very small doses of

bupivacaine plus narcotics have been used in the outpatient setting with variable success.

Intrathecal adjuncts

1. Epinephrine It prolongs duration, but also prolongs the recovery time and voiding time. Thus it

should not be used in the ambulatory setting.

2. Fentanyl The lipophilic synthetic opioids appear to improve the quality of the block

without prolonging recovery. Ben-David et al in 1997 showed that 5 mg of

hyperbaric bupivacaine was inadequate in 27% of cases of spinal for knee

arthroscopy. Adding 10 ucg of fentanyl reduced the failure rate to zero.

Fentanyl produces pruritus in about 50% of the patients. Serotonin inhibitors (like

ondansetron) are being used to treat this side effect too. Respiratory effects are

unlikely with doses below 25 mcg.

3. Morphine The use of hydrophilic intrathecal narcotics is accompanied by a longer lasting

analgesia, but also by a higher rate of complications. Among them are: delay

respiratory depression (4-6 hrs after the injected dose), increased nausea and

vomiting, pruritus and delayed voiding.

4. Clonidine and neostigmine They potentiate spinal local anesthetics and produce postoperative analgesia, but

they produce unacceptably high rates of hypotension and sedation (clonidine) and

protracted vomiting (neostigmine).

Page 47: Manual Regional Carlo 2010

47 | P a g e

EPIDURAL ANESTHESIA

It is technically more difficult to perform than spinal and because larger doses of

local anesthetics are used it has the potential for systemic toxicity. On the other hand, it

offers a greater degree of flexibility in the extent and duration of anesthesia.

Anatomy

The spinal epidural space extends from the foramen magnum to the end of the

dural sac at the level of S2. It is bounded anteriorly by the vertebral bodies and

posteriorly by the laminae and ligamentum flavum. The epidural space outlines the spinal

canal immediately superficial to the dura. In the cervical region the epidural space is

smaller and it is wider in the lumbar area. A volume of local anesthetic about 10 times

larger is required to produce lumbar epidural anesthesia than for equivalent subarachnoid

blockade. Smaller volumes are sufficient for the thoracic space. The epidural space is

filled with connective tissue, fat and veins, which can become enlarged during

pregnancy. The spinal nerves travel through this space surrounded by a sheath of dura.

Characteristic of an epidural blockade

Epidural anesthesia produces a band of segmental anesthesia spreading cephalad

and caudad from the site of injection. Epidural anesthesia has a slower onset and usually

it is not as dense as spinal. This characteristic can be used as an advantage to obtain a

more pronounced differential blockade. Dilute concentrations can spare the motor fibers

while still able to produce sensory analgesia. This is commonly employed in labor

epidural analgesia.

Factors affecting the spread of local anesthetics in the epidural space

In general 1-2 mL of local anesthetic is needed per every segment to be blocked.

Thus, to achieve a T4 level from an L4-5 injection 12-24 mL of local anesthetic is

needed.

1. Dose and volume: The total dose and the volume affect the height of the block.

The effect of volume is linear but it plateaus at about 20 mL, after which there is

a greater loss through intervertebral foramina, especially in younger patients.

2. Age: As opposed to spinal, age is a major factor in the spread of epidural

anesthesia with smaller volumes producing a higher spread in older patients.

This may be due to the narrowing of the intervertebral foramina with age.

3. The site of injection influences the spread. Volumes as small as 6-8 mL of

solution injected at the thoracic level can produce anesthesia due to smaller

volume of the epidural space.

4. Body weight: heavier patients have smaller volume requirements.

5. Height: Plays a small role with taller patients requiring higher volumes.

6. Gravity: is not a very important factor, as sitting position does not appear to

enhance sacral spread.

Page 48: Manual Regional Carlo 2010

48 | P a g e

Techniques

Lumbar epidural

It is the most common site for epidural anesthesia. The midline or paramedian

approach can be used. A block below the termination of the spinal cord at L1 should be

safer. An accidental dural puncture (“wet tap”) could result in spinal cord damage at

higher levels.

Thoracic epidural

It is technically more challenging and has a greater risk for spinal cord injury. It is

rarely used as the primary anesthetic. Many people prefer the paramedian approach in the

thoracic level, because of the extreme obliquity of the thoracic spinous processes.

Epidural needles

The Tuohy needle is the most commonly used. A typical needle is 17-18 gauge,

3.5 inches long. It has a blunt bevel with a gentle curve of 15-30° at the tip. The blunt tip

helps push the dura away, “tenting” it, after the ligamentum flavum has been pierced.

Epidural catheters

They provide the means for continuous infusion. Usually they are 19-20 gauge in

size. The needle bevel is directed in the desired direction (not a guarantee for catheter

final location) and the catheter is advanced 2-6 cm. A short insertion increases the chance

for accidental dislodgement. The farther in, the greater the chance of unilateral epidural

and other complications (bloody tap, catheter knotting). Four to five cm is a good

compromise.

Test dose

It is important because of the large doses of LA injected into the epidural space.

The classic test dose is 3 mL of 1.5% lidocaine (45 mg) with 1:200,000 of epinephrine

(15 mcg). The 45 mg of lidocaine, if intrathecal, should produce spinal anesthesia. The 15

mcg of epinephrine, if intravascular, should produce at least a 20% increase in heart rate

within 30 sec or 30 beats between 20-40 sec (Barash’s, 5th

edition, 2006). In patients who

are beta blocked the heart rate increase may not happen and an increase in systolic

pressure of 20 mmHg or more may be more reliable (Barash’s 5th

edition, 2006). The use

of epinephrine as a test dose in obstetrics is controversial. Some suggest instead the use

of only 30 mg of lidocaine or 5 mg of bupivacaine.

Activating an epidural, Incremental dosing After a negative test dose most of practitioners will inject incremental doses of 5

mL at a time. This technique helps decrease the risk of systemic toxicity in case of

catheter migration (intravascular or intrathecal).

Termination of action

It is related to type of drug and degree of spread. It is commonly described as the

time it takes to a two-segment regression of sensory blockade. The approximate time for

two-segment regression (sensory) for chloroprocaine is 50-70 minutes, for lidocaine is

90-150 minutes and for bupivacaine is 200-260 minutes.

Page 49: Manual Regional Carlo 2010

49 | P a g e

References

1. Snell R: Clinical Anatomy for Medical Students, 5th

edition. Boston, Little,

Brown and Company, 1995

2. Bernards CM: Epidural and Spinal Anesthesia, In: Barash PJ, Cullen BF,

Stoelting RK, Clinical Anesthesia, 5th

edition. Philadelphia, Lippincott Williams

& Wilkins, 2006, pp 691-717

3. Mulroy MF: Regional Anesthesia, 3rd

edition. Philadelphia, Lippincott Williams

& Wilkins, 2002, pp 65-118

4. Wong G, Brown D. Transient paraplegia following alcohol celiac plexus block.

Reg Anesth 1995; 20: 352-355

5. Hogan QH. Magnetic resonance imaging of cerebrospinal fluid volume and the

influence of body habitus and abdominal pressure. Anesthesiology 1996; 84;

1341-1349

6. Bridenbaugh PO, Greene NM, Brull SJ, Spinal (Subarachnoid) Neural

Blockade, In: Cousins MJ, Bridenbaugh PO (eds), Neural Blockade, 3rd

edition.

Philadelphia, Lippincott-Raven, 1998, pp 203-241

7. Cousins MJ, Veering BT: Epidural Neural Blockade, In: Cousins MJ,

Bridenbaugh PO (eds), Neural Blockade, 3rd

edition. Philadelphia, Lippincott-

Raven, 1998, pp 243-320

8. Salinas FV: Pharmacology of Drugs Used for Spinal and Epidural Anesthesia

and Analgesia, In: Wong CA (ed), Spinal and Epidural Anesthesia. New York,

McGraw-Hill, 2007, pp 75-109

9. Sheskey MC, Rocco AG, Bizzarri-Schmid M, et al. A dose-response study of

bupivacaine for spinal anesthesia. Anesth Analg 1983; 62: 931-935

10. Van Zundert AAJ, Grouls RJE, Korsten HHM, et al. Spinal anesthesia: Volume

or concentration-What matters? Reg Anesth 1996; 21: 112-118

11. Giroux CL, Wescott DJ. Stature estimation based on dimensions of the bony

pelvis and proximal femur. J Forensic Sci, 2008; 53: 65-68

12. Reina MA, de Leon-Casasola OA, Lopez A, et al. An in vitro study of dural

lesions produced by 25-gauge Quincke and Whitacre needles evaluated by

scanning electron microscope. Reg Anesth Pain Med 2000; 25: 393-402

13. Swisher JL. Spinal Anesthesia: Past and Present. In Problems in Anesthesia,

2000:12; 141-147

14. Ben-David B, Solomon E, Levin H, et al. Intrathecal fentanyl with small-dose

dilute bupivacaine: Better anesthesia without prolonging recovery. Anesth

Analg 1997: 85; 560-565

15. Pollock JE. Transient neurological symptoms: etiology, risk factors, and

management. Reg Anesth Pain Med 2002:27; 581-86

16. Morgan GE, Mikhail MS, Murray MJ: Clinical Anesthesiology, 4th

edition. New

York, McGraw-Hill, 2006, pp 289-323

Page 50: Manual Regional Carlo 2010

50 | P a g e

CHAPTER 4

REGIONAL ANESTHESIA AND ANTICOAGULATION

Page 51: Manual Regional Carlo 2010

51 | P a g e

Regional anesthesia in the patient receiving antithrombotic or thrombolytic therapy

The American Society of Regional Anesthesia (ASRA) convened its Third

Consensus Conference on Regional Anesthesia and Anticoagulation. The new guidelines

were published in the January-February 2010 issue of the ASRA Journal. This chapter is

mainly based on these guidelines.

Epidural hematoma is defined as a rare, but potentially catastrophic complication

of spinal or epidural anesthesia. The introduction of low molecular weight heparin

(LMWH) in the United States coincided with an increased number of reported cases of

epidural hematoma. Although, it can happen spontaneously, its incidence increases with

age, associated abnormalities of the spinal cord or vertebral column, underlying

coagulopathy, difficult needle placement and an indwelling catheter in the presence of

anticoagulation. The actual incidence of hemorrhagic complications in association with

neuraxial anesthesia is unknown, but has been estimated at less than 1 in 150,000 for

epidural and less than 1 in 220,000 for spinal anesthesia. Recent studies suggest that this

incidence may be higher, some say as high as 1 in 3,000 in selected populations.

At the moment there is no laboratory model to study this problem and its rarity

precludes a prospective randomized study. As a result the ASRA consensus represents

the opinions of experts based on case reports, clinical series, pharmacology, hematology

and risks factors for surgical bleeding.

Strength and grade of recommendations

A cornerstone in evidence-based medicine is the quality of the available evidence.

The validity of the recommendation improves with the quality of the evidence. There are

three levels of strength of recommendation according to the quality of the available

evidence:

A: Highest level of evidence. These are randomized clinical trials and meta-

analysis. Because neuraxial bleeding is rare this type of evidence is mostly not

available.

B: Inconsistent or limited quality patient-oriented evidence. These are

observational and epidemiological series. Depending on the quality of these

studies and the degree of risk reductions showed, recommendation from these

sources may be categorized as level of evidence A or B.

C: recommendations derived from case reports or expert opinion.

The recommendations are also graded to indicate the strength of the guideline and the

degree of consensus:

Grade 1: represents general agreement on the efficacy.

Grade 2: Notes conflicting evidence or opinion.

Grade 3: Suggests that the procedure may not be useful and possibly harmful

(e.g., epidural procedure in a patient receiving twice-daily LMWH).

Venous thromboembolism VTE

This is an important health care problem. Neuraxial anesthesia has been

associated with improved patient outcomes, including mortality and major morbidity.

This probably results from the “attenuation of the hypercoagulable response” and

Page 52: Manual Regional Carlo 2010

52 | P a g e

decreased venous thrombosis after these techniques. However the beneficial effect of

neuraxial techniques on coagulation is insufficient as the sole method of

thromboprophylaxis. As a result, anticoagulants, antiplatelets and thrombolytic

medications are commonly used in the prevention and treatment of thromboembolism.

Nearly all hospitalized patients have at least one risk factor and 40% of patients

have 3 or more risk factors (according to Geerts et al, as cited by the 2010 ASRA

statement). The following is a table for risk factors for VTE taken from the 2010 ASRA

statement:

Accordingly, most hospitalized patients benefit from some type of

thromboprophylaxis. The following table, also taken from the ASRA 2010 statement,

lists the recommended prophylaxis according to risk:

Because of concerns with surgical bleeding associated with thromboprophylaxis,

the American Academy of Orthopaedic Surgeons (AAOS) published its own guidelines

Page 53: Manual Regional Carlo 2010

53 | P a g e

in 2007 for the prevention of symptomatic PE in patients undergoing total joint

replacement. The following table taken from the 2010 ASRA guidelines shows the

AAOS recommendations:

Administration of thromboprophylaxis

In terms of agents and doses, the 2010 ASRA statement recommends to follow

the American College of Chest Physicians ACCP guidelines advising the clinicians to

follow the manufacturer-suggested dosing guidelines (Evidence Grade 1C).

Risk of bleeding Bleeding, especially intracranial, intraspinal, intraocular, mediastinal or

retroperitoneal, is the most feared complication of anticoagulant and thrombolytic

therapy. Risks factors include increased age, female sex, history of gastrointestinal

bleeding, concomitant aspirin use and length of therapy.

During warfarin therapy an INR of 2.0 to 3.0 is associated with a 3% low risk of

bleeding during a 3-month treatment period. Stronger regimens (INR >4) increase the

risk of bleeding significantly to 7%.

The incidence of hemorrhagic complications during therapeutic anticoagulation

with IV or subcutaneous heparin is less than 3% and even lower with LMWH.

Thrombolytic therapy is associated with the highest risk of bleeding, with major

bleeding occurring in 6% to 30% of patients treated with thrombolytic therapy for DVT,

ischemic stroke, or ST elevation myocardial infarction. There is no significant difference

in the risk of bleeding among thrombolytic agents.

The addition of potent anticoagulants (LMWH, hirudin) or antiplatelets

(glycoprotein IIb/IIIa agents) therapy increases even more the risk of major bleeding.

“Therefore, although thromboembolism remains a source of significant

perioperative morbidity and mortality, its prevention and treatment are also

associated with risk” (2010 ASRA statement, page 67).

Page 54: Manual Regional Carlo 2010

54 | P a g e

Anesthetic management of the patient receiving thrombolytic therapy

These patients are at risk of serious bleeding. We will discuss several situations:

1. Patients scheduled to receive thrombolytic therapy: Avoid performing lumbar

punctures and neuraxial anesthesia and avoid thrombolytic therapy for 10 days

if these procedures have been performed (evidence Grade 1A).

2. Patients who have received thrombolytic therapy: Do not perform spinal or

epidural procedures (Evidence Grade 1A). Data not available as to how long

we need to wait.

3. Patients who have received neuraxial blocks at or near the time of fibrinolytic

and thrombolytic therapy: Neurological monitoring every 2 hours or less “for

an appropriate interval”. If epidural catheter present avoid drugs producing

sensory and motor block to facilitate neurological assessment (Evidence

Grade 1C).

4. Patient with an epidural catheter who unexpectedly received thrombolytic

therapy: There is no definite recommendation as to when to remove it. They

suggest to measure fibrinogen levels (one of the last clotting factors to

recover) for appropriate timing of catheter removal (Evidence Grade 2C).

Anesthetic management of the patient receiving unfractionated heparin (UFH)

There is a long experience in the management of these patients. However recent

guidelines suggesting a three-time dose (thrice daily) of subcutaneous heparin for some

patients and its potential for increased bleeding have prompted a modification to the

ASRA guidelines as follows:

1. Daily review of patient’s medical records to identify the concomitant use of

other drugs affecting coagulation like antiplatelets, LMWH and oral

anticoagulants (Grade 1B).

2. Patients receiving 5000 U of UFH twice daily do not have contraindication for

neuraxial techniques. The risk of bleeding may be reduced by delay of the

heparin dose until after the block. The risk may be increased in debilitated

patients after prolonged therapy (Grade 1C).

3. The safety of neuraxial blocks on patients receiving more than twice daily

dose or doses greater than 10000 U of UFH daily has not been established.

Suggest frequent neurological exam if neuraxial has been done (Grade 2C).

4. Patients receiving heparin for more than 4 days (heparin-induced

thrombocytopenia) should have a platelet count before neuraxial block and

catheter removal.

5. Combining neuraxial techniques with intraoperative anticoagulation with

heparin during vascular surgery is acceptable with the following

recommendations (Grade 1A):

a. Avoid the technique in patients with other coagulopathies.

b. Delay heparin administration for 1 hr after needle placement.

c. Remove catheter 2-4 hr after the last heparin dose; re-heparin 1 hr after

catheter removal.

Page 55: Manual Regional Carlo 2010

55 | P a g e

d. Monitor the patient postoperatively to provide early detection of motor

blockade. Avoid local anesthetics through catheter.

e. The occurrence of bloody or difficult neuraxial technique may increase

risk, but data does not support mandatory cancellation. Risk-benefit

discussion with surgeon about proceeding.

f. Insufficient data exist about risk of bleeding when neuraxial

techniques are combined with the full anticoagulation of cardiac

surgery. They suggest neurological monitoring and avoidance of local

anesthetics (Grade 2C).

Anesthetic management of the patient receiving LMWH

The extensive European experience is useful to us. The 2010 ASRA consensus

respects the labeled dosing regimens of LMWH as established by the Food and Drug

Administration. Although it is impossible to eliminate the risk of neuraxial hematoma

previous recommendations have been deemed useful.

1. The anti-Xa level is not predictive of the risk of bleeding. Recommend against

the routine use of it (Grade 1A).

2. Antiplatelets and other anticoagulants administered in conjunction with

LMWH increase the risk of spinal hematoma. Avoid concomitant use of

antiplatelets drugs, unfractionated heparin, or dextran regardless of LMWH

dosing regimen (Grade 1A).

3. The presence of blood during neuraxial technique does not necessitate

postponement of surgery. Recommendation to delay initiation of LMWH for

24 hr in discussion with the surgeon (Grade 2C).

4. Preoperative use of LMWH:

a. Patients receiving LMWH can be assumed to have altered coagulation.

Recommend needle placement at least 12 hr after the LMWH last dose

(Grade 1C).

b. Patients receiving higher doses of LMWH, such as enoxaparin 1

mg/kg every 12 hrs, enoxaparin 1.5 mg/kg daily, dalteparin 120 U/kg

every 12 hrs, dalteparin 200 U/kg daily, or tinzaparin 175 U/kg daily,

the recommendation is to delay neuraxial technique for at least 24 hrs

(Grade 1C).

c. Patients given a dose of LMWH 2 hrs preoperatively (general surgery

patients) the recommendation is to avoid neuraxial techniques because

of peak anticoagulant activity (Grade 1A).

5. Postoperative use of LMWH: Patients to undergo postoperative LMWH

prophylaxis may safely undergo single-injection and continuous catheter

techniques. Management is based on total daily dose, timing of the first

postoperative dose and dosing schedule (Grade 1C):

a. Twice-daily dosing. This dosing is associated with increased risk of

spinal hematoma. The first dose of LMWH should be administered no

earlier than 24 hrs postoperatively. Indwelling catheters may be left in

Page 56: Manual Regional Carlo 2010

56 | P a g e

place overnight but must be removed before initiation of LMWH, and

the first dose should be delayed for 2 hrs after catheter removal.

b. Single-daily dosing. The first postoperative LMWH dose should be

administered 6-8 hrs postoperatively and the second no sooner than 24

hrs later. Indwelling catheters may be safely maintained although it

should be removed a minimum of 10-12 hrs after the last dose of

LMWH. Subsequent dosing should not be given for at least 2 hrs after

catheter removal. No other drugs with effect in coagulation should be

given because of risk of additive effects.

Regional anesthetic management of the patient on oral anticoagulants

The management of patients receiving perioperative warfarin remains

controversial.

1. In the first 1-3 days after warfarin discontinuation the coagulation status

(reflected primarily by factors II and X levels) may not be adequate despite a

decrease in the INR (indicating a return of factor VII activity). Adequate

levels of II, VII, IX and X may not be present until the INR is normal. The

recommendation is that warfarin must be stopped 4-5 days prior to the

procedure and the INR measured before a neuraxial block is attempted (Grade

1B).

2. Avoid using other drugs with anticoagulation effect like aspirin and other

NSAIDs, ticlopidine, and clopidogrel, UFH, and LMWH (Grade 1A).

3. In patients who are likely to have an enhanced response to the drug, it is

recommended to use the available algorithms to guide in the dosing based on

desired indication, patient factors, and surgical factors (Grade 1B).

4. In patients receiving an initial dose of warfarin before surgery, the

recommendation is to check the INR prior to neuraxial block if the first dose

of warfarin was administered more than 24 hrs earlier or if a second dose has

been administered (Grade 2C).

5. In patients receiving low-dose warfarin therapy during epidural analgesia, the

suggestion is to monitor the INR daily (Grade 2C).

6. For patients on warfarin therapy receiving epidural analgesia neurologic

testing of motor and sensory function should be performed routinely. To

facilitate the neurologic evaluation keep the local anesthetics to a minimum

(Grade 1C).

7. As warfarin therapy is initiated it is suggested that neuraxial catheters should

be removed with an INR of less than 1.5. This value correlates hemostasis

with clotting factor activity levels greater than 40%. The suggestion is to keep

neurologic testing after catheter removal for at least 24 hrs (Grade 2C).

8. In patients with INR more than 1.5 but less than 3 the suggestion is to remove

catheters with caution after reviewing medication records for other

medications affecting coagulation that may not affect the INR (e.g., NSAIDs,

clopidogrel, ticlopidine, UFH, LMWH (Grade 2C). It is also recommended to

check neurological status before catheter removal and continued until the INR

has stabilized at the desired prophylaxis level (Grade 1C).

Page 57: Manual Regional Carlo 2010

57 | P a g e

9. In patients with INR greater than 3 and an indwelling catheter, the

recommendation to hold or reduce the warfarin dose (Grade 1A). No

definitive recommendation can be made for removal of catheters in patients

with therapeutic levels of anticoagulation (Grade 2C).

Anesthetic management of the patient receiving antiplatelet medications

Antiplatelet medications exert diverse effects on platelet function. These drugs

include NSAIDs, thienopyridine derivatives (ticlopidine and clopidogrel) and platelet

glycoprotein (GP) IIb/IIIa antagonists (abciximab, eptifibatide, tirofiban). There is no

wholly accepted test, including the bleeding time, to guide antiplatelet therapy.

1. NSAIDs seem to present no added significant risk of spinal bleeding related to

neuraxial techniques. No specific concerns exist at this time about this drugs

and the timing of single-shot or catheter insertion or removal (Grade 1A).

2. In patients receiving NSAIDs, the recommendation is not to perform neuraxial

techniques if other drugs like oral anticoagulants, UFH, and LMWH are being

used concurrently. Cyclooxygenase-2 (cox-2) inhibitors have minimal effect

on platelet function and should be considered in patients requiring anti-

inflammatory therapy in the presence of anticoagulation (Grade 2C).

3. The actual risk of spinal hematoma with ticlopidine and clopidogrel and the

GP IIb/IIIa antagonists is unknown. Recommendations are based on labeling

precautions and the clinical experience (Grade 1C).

a. On the basis of labeling and surgical experience the waiting period

between discontinuation of a drug and neuraxial block is:

i. ticlopidine: 14 days

ii. clopidogrel: 7 days. If a neuraxial block is indicated between 5-

7 days after its discontinuation, normalization of platelet

function should be documented.

b. Platelet GP IIb/IIIa inhibitors have a profound effect on platelet

aggregation. Neuraxial techniques should be avoided until platelet

function has recovered. This time is:

i. Abciximab: 24-48 hrs

ii. Eptifibatide and tirofiban: 4-8 hrs.

Anesthetic management of the patient receiving herbal therapy

Herbal drugs by themselves do not interfere with the performance of neuraxial

techniques. The recommendation is against mandatory discontinuation of herbs or

avoidance of regional techniques in these patients (Grade 1C).

Anesthetic management of patients receiving thrombin inhibitors (desirudin,

lepirudin, bivalirudin, and argatroban)

In these patients the recommendation is not to perform neuraxial techniques

(Grade 2C).

Page 58: Manual Regional Carlo 2010

58 | P a g e

Anesthetic management of the patient receiving fondaparinux

The actual risk is unknown. Until further experience is available, performance of

neuraxial techniques should be avoided.

Anesthetic management of the anticoagulated parturient

In the absence of large series of neuraxial technique in pregnant women receiving

anticoagulation the recommendation is to follow the ASRA guidelines for the rest of

surgical patients (Grade 2C).

Anesthetic management of the patient undergoing plexus or peripheral block

The recommendation is to apply the ASRA guidelines for neuraxial techniques

(Grade 1C).

Page 59: Manual Regional Carlo 2010

59 | P a g e

References

1. Neal JM: Neural Blockade and Anticoagulation. In: Regional Anesthesia, The

Requisites in Anesthesiology. Rathmell J, Neal J, Viscomi C (eds). Philadelphia,

Elsevier Mosby, 2004, pp 151-156

2. Bergqvist D, Wu CL, Neal JM. Anticoagulation and Neuraxial Regional

Anesthesia: Perspectives. [Editorial] Reg Anesth Pain Med 2003; 28: 163-166

3. Horlocker tt, Wedel DJ, Benzon H, et al. Regional anesthesia in the

anticoagulated patient: Defining the risks (The Second ASRA Consensus

Conference on Neuraxial Anesthesia and Anticoagulation). Reg Anesth Pain Med

2003; 28: 172-197

4. Horlocker TT, Wedel DJ, Rowlingson JC, Enneking FK, Kopp SL, Benzon HT,

Brown DL, Heit JA, Mulroy MF, Rosenquist RW, Tryba M, Yuan CS. Regional

Anesthesia in the Patient Receiving Antithrombotic or Thrombolytic Therapy.

American Society of Regional Anesthesia and Pain Medicine Evidence-Based

Guidelines (Third Edition). Reg Anesth Pain Med 2010; 35: 64-101

Page 60: Manual Regional Carlo 2010

60 | P a g e

CHAPTER 5

PERIPHERAL NERVE BLOCKS

Page 61: Manual Regional Carlo 2010

61 | P a g e

Peripheral Nerve Blocks

A successful peripheral nerve block results from injecting an adequate volume of

an adequate concentration of local anesthetic in the proximity of the target nerve(s).

Intraneural injection (especially intrafascicular) might be harmful to the nerve and can

lead to permanent damage. Therefore, a balance must be achieved between the need to

get close to a nerve and safety.

Bringing the needle close to the nerve(s)

There are many ways to ascertain the correct placement of a needle with respect to a

nerve. A good knowledge of the anatomy makes things easier and safer. The methods are:

1. Purely anatomical: the practitioner bases his/her technique solely on anatomical

facts to bring the needle in proximity to the nerve. For example, the pulse of the

femoral artery can be used to locate the femoral nerve located lateral to it, and the

pulse of the axillary artery can guide the injection of the terminal branches of the

brachial plexus in the axilla.

This anatomical method is extremely operator-dependant with good

success in the hands of the few and limited success in the hands of the majority.

This method does not take into account anatomical variations, lacks depth

perception and can not gauge proximity to a nerve with any degree of certainty.

Therefore, the needle might end up too far from the nerve (failed block) or too

close to it (intraneural).

2. Paresthesia: this technique requires a combination of anatomical knowledge and

patient collaboration. The needle is brought to the point of physical contact with

the target nerve. The patient is instructed to acknowledge the electrical sensation

elicited (paresthesia). The location of the paresthesia, as referred by the patient,

helps the practitioner locate the position of the needle. At this time the needle is

withdrawn a few mm, before the injection is started, to decrease the risk of

intraneural injection.

For the longest time, Moore’s dictum “no paresthesia no anesthesia”, was

the “law of the land” in regional anesthesia. Works by Selander and others,

starting in the 1970s, have questioned the safety of this practice. Although, there

is not enough evidence to believe that paresthesias lead to nerve damage, there

seems to be enough circumstantial evidence to be cautious, especially if repeated

paresthesias are elicited.

3. Nerve stimulation: the idea of locating mixed nerves by electrical stimulation

was developed in Germany in 1912 by Perthes. However, it was not until 1962

when Greenblatt and Denson introduced a portable, transistorized nerve

stimulator that was suitable for the clinical setting.

The nerve stimulator is connected to a needle, usually insulated, that

delivers a current to its tip. The A alpha fibers (motor) are readily depolarized by

the small currents used, but not the sensory fibers. As the needle approaches a

Page 62: Manual Regional Carlo 2010

62 | P a g e

mixed nerve, a painless muscle twitch is produced. The intensity of the response

is inversely proportional to the needle tip-nerve distance (actually to the square

root of it). A visible response at lower currents (less than 0.5 mA), suggests close

proximity between the needle tip and the target nerve. There is a good amount of

clinical evidence to suggest that a current of 0.5 mA or less, capable of eliciting a

visible response, is a reliable indicator of critical proximity. However, evidence is

lacking as to what exactly that distance is, and as to whether the distance is

different for different nerves. In general it is thought that 1 mA of current will

produce depolarization of a motor nerve at a distance of about 1 cm (10 mm).

Nowadays nerve stimulator techniques are widely practiced around the

world. With modern nerve stimulators the practitioner can adjust the pulse

intensity (magnitude of the current) in mA; the pulse frequency (amount of

pulses per second) in Hz (1 or 2) and the pulse width (duration of the pulse) in

milliseconds (ms). The pulse duration most suitable for stimulating motor fibers

in a mixed nerve is 0.1 ms (100 microsec).

4. Ultrasound: It is the latest and most sophisticated piece of technology introduced

to the practice of regional anesthesia and has already caused a revolution. It is the

only method that can provide real time assessment of the position of the needle

with respect to the nerve, as well as an image of the surrounding structures. An

added advantage is that the practitioner is able to see the spread of the local

anesthetic, giving him/her the chance to more accurately predict the success of the

technique as well as the need for supplementation.

Ultrasound could theoretically produce warming of tissues or gas

formation. This technology is still expensive, and requires competency on

interpretation of cross-section anatomy from “grainy” images. However, it has

been rapidly progressing and in many centers, including ours, is the main method

used to perform regional blocks of every kind.

Characteristics of ultrasound

The human ear can hear sounds between 20 and 20,000 Hz (cycles per

second) or 20 KHz. Ultrasounds waves travel at a higher frequency than the

highest frequency detectable by the human ear. Ultrasound waves used in

medicine usually are in the 1 to 20 MHz range (1 MHz = 1 million Hz).

Ultrasound waves travel easily through fluids and soft tissue, but have

problems traveling through bone and air. Ultrasound is better reflected at the

transition between two different types of tissues like soft tissue-air, bone-air and

soft tissue-bone. This transition plane is seen as a hyperechoic line on the screen.

The ultrasound is delivered from a small probe that contains piezoelectric

crystals that under the influence of an electric current are made to vibrate

producing a wave of ultrasonic sound. The ultrasound waves in the form of a

narrow beam travel through tissues at a speed that depends on the nature of the

human tissues, but for calculations and image production is assumed to be an

average value of 1,540 m/sec. This value closely approximates the speed of

ultrasound through soft tissue (1,540 m/sec), muscle (1,580 m/sec), blood (1,560

Page 63: Manual Regional Carlo 2010

63 | P a g e

m/sec), but differs to the speed through bone (4,000 m/sec), lung (500 m/sec) or

air (330 m/sec).

Part of the ultrasound waves are reflected back to the transducer,

especially at tissue interfaces, where the mechanical energy is converted back to

electrical energy. The information is then processed by the software of the

ultrasound machine to generate an image. Therefore, the transducer delivers

ultrasound for part of the time and for part of the time it “listens” for the returned

waves. The distance is calculated as a function of the time it takes for the waves

to return. Tissues with high density like bone reflect most of the waves and

produce a bright image, known as hyperechoic. A tissue like blood that permits

easy passage of the ultrasound waves through it appears dark or anechoic. The

rest of tissues present intermediate characteristics between anechoic o hypoechoic

to hyperechoic.

Better images are obtained when the probe is perpendicular to the

structure being searched (e.g., nerve, needle). This is because more bouncing

sound waves can be detected by the transducer. Changes as small as 10 degrees

from the perpendicular can distort the echogenicity of a nerve, reducing the

amount of waves returning to the transducer and decreasing the quality of the

image. This is known as anisotropy, the change of the quality of the echo image

as a result of change in the angle of incidence of the probe with respect to the

target structure. Tendons characteristically have higher anisotropy than peripheral

nerves.

Short versus long axis views

The most common way to identify a peripheral nerve is through a

transverse scan of it, also called “short axis view”. This provides a cross section

image of the nerve(s) and surrounding structures. A “long axis view” of a nerve is

also possible, although sometimes more challenging, because the nerves

trajectories are not necessarily linear. In addition, in a long axis view the operator

looses the ability to readily recognize lateral and medial sides of the nerve on the

2-dimensional image obtained.

In plane versus out of plane techniques

The needle can be advanced “in plane” or “out of plane” with respect to

the main axis of the probe. In the in plane approach the needle is advanced in

coincidence with the long axis of the probe, in the same plane of the ultrasound

beam. This makes possible the visualization of the needle as it advances toward

the target nerve(s). Good needle visualization depends on its angle of insertion,

with the best visualization obtained when the needle trajectory is parallel to the

probe. As the angle of penetration increases (deeper targets) the difficult to

visualize the needle also increases. When the insertion angle is more than 45

degrees with respect to the plane of the probe the needle cannot be visualized

anymore. At this point tissue movement and injection of small amounts of local

anesthetics can help determine the location of the needle tip.

Page 64: Manual Regional Carlo 2010

64 | P a g e

With the out of plane approach the needle is advanced perpendicular to the

main axis of the probe, so only the tip of the needle can be visualized at the point

where it crosses under the ultrasound beam. The tip is seen as a very hyperechoic

bright point on the screen. The main advantage of an out of plane technique, from

my perspective, is that the trajectory of the needle to the target is shorter.

Regardless of the approach the goal is to bring the tip of the needle into the

proximity of the nerve(s) for injection.

High versus low frequency probes

High frequency probes (8-15 MHz) are usually linear probes that provide

good resolution, but limited penetration (3-4 cm). These probes are used at

different levels of the brachial plexus, abdominal wall and at different locations in

the lower extremity. For deeper structures, lower frequency (4-7 MHz) curved

probes are needed providing a wider field and deeper penetration at the expense

of resolution. Deep scanning of intra abdominal organs requires frequencies of 3-

5 MHz The quality of the image is also affected by other factors like compound

imaging (the capture of different views of structures before producing an image)

and color Doppler.

We will refer again to ultrasound when we describe individual techniques.

Insulated versus non insulated needles

Insulated needles are the needles most commonly used in conjunction with nerve

stimulation and ultrasound techniques nowadays in the United States, Europe and other

parts of the world. The current applied to this needle concentrates at its tip, making the

localization of nerves more accurate. Several brands of these needles exist in the market

and they come ready with a connection that only fits the negative electrode. Connecting

the negative electrode to the exploring needle lowers the amount of current necessary to

depolarize a nerve.

Non-insulated needles transmit the current preferentially to the tip, but also along

the shaft of the needle making the localization of nerves less accurate. Insulated needles

are more expensive than non-insulated needles.

Short versus long-bevel needles

Standard needles have a tip angle of around 14 degrees and are known as “sharp’

needles. It is frequently recommended to perform regional block with short-bevel needles

with an angle of 30 to 45 degrees. This recommendation comes from studies by Selander

et al who demonstrated more neural damage in isolated sciatic nerves when sharp needles

were used. The damage with sharp needles was also more extensive when the orientation

of the sharp bevel was perpendicular to the fibers. With short bevel needles, the damage

was less frequent as the fibers were pushed away by the advancing needle.

This concept has been challenged by Rice et al. According to these authors it may

be more difficult to penetrate a nerve fascicle with a short-bevel needle than with a sharp

needle, but should it occur, the lesions may be more severe. Recently in 2009 Sala-

Page 65: Manual Regional Carlo 2010

65 | P a g e

Blanch and collaborators published in Regional Anesthesia and Pain Medicine a study in

which sharp long beveled versus blunt short beveled (30 degrees) needles were

introduced into a sciatic nerve of a human cadaver. After the punctures the specimen was

investigated under the microscope for evidence of fascicular damage. They demonstrated

that with either needle was very difficult to penetrate the fascicles. In fact they found no

histological evidence of fascicular damage with short beveled needles and only 3.2% of

fascicular damage (4 fascicles) with sharp needles.

Nerve injury

Persistent paresthesias can occur after regional anesthesia, although severe

neurologic injury is extremely rare. Neal estimates the incidence of persistent neuropathy

after regional anesthesia to be less than 0.4%.

A large survey by Auroy et al in France in 1997, involving 71,053 neuraxial

blocks and 21,278 peripheral nerve blocks, showed a low incidence (0.03%) of nerve

complications after regional anesthesia. The survey showed that neurological deficits

although low, were relatively more frequent after spinal (70%) than either epidural (18%)

or peripheral nerve block (12%). In two thirds of the cases of neuropathy after spinal, and

100% of the cases after epidural, a paresthesia was elicited either by the needle or during

the injection. Among the neurological deficits that developed after non-traumatic spinals,

75% of them were in association with the use of 5% hyperbaric lidocaine.

Cheney et al in 1999 reviewed the American Society of Anesthesiologist closed-

claims database and found that out of 4,183 claims, 670 (16%) were considered

“anesthesia-related nerve injury”. Injury to the ulnar nerve represented 28% of the total,

and in 85% of the cases it was associated to general anesthesia. Other nerve injuries were

brachial plexus in 20%, lumbosacral trunk in 16% and spinal cord 13% and these were

more related to regional anesthesia. In 31% of the brachial plexus injuries the patient had

experienced a paresthesia with the needle or during injection. They concluded that

prevention strategies are difficult because the mechanism for nerve injury, especially of

the ulnar nerve, is not apparent.

Lee et al in 2004 conducted a new review of the Closed Claims Data for the 1980

to 1999 period focusing in regional anesthesia. A total of 1,005 regional anesthesia-

related claims were reviewed. These claims were 37% obstetric related and 63% non-

obstetric. All regional anesthesia, obstetric claims were related to neuraxial

anesthesia/analgesia. In 21% of the non-obstetric claims, peripheral nerve blocks were

involved. The most common block was axillary block (44%). Upper extremity blocks

were more involved in claims than lower extremity blocks. Nerve injury temporary or

permanent was claimed in 59% of the peripheral nerve injury claims.

Death or brain damage was usually the result of cardiac arrest associated with

neuraxial block. Pneumothorax accounted for 10% of the claims and “emotional distress”

was claimed in 2% of the cases. Eye blocks accounted for 5% of the claims.

Regional anesthesia could result in nerve damage directly from a needle or

catheter or be the result of ischemia or other unknown mechanism. Ischemia could be the

potential result of vasoconstrictor use or by an intraneural injection that produces an

increase of the intraneural pressure leading to nerve ischemia. Local anesthetic toxicity

could play a role in cauda equina syndrome and transient neurological symptoms.

Page 66: Manual Regional Carlo 2010

66 | P a g e

Another mechanism of nerve injury could be hematoma and infection leading to scar

formation.

It has been a common belief in regional anesthesia that nerve puncture and

intraneural injection lead to nerve damage. In 2006 Bigeleisen published in

Anesthesiology a study that seems to discredit this notion. In his study conducted under

ultrasound guidance 21 of 26 patients had nerve punctures of at least one nerve, and 72

out of 104 nerves had intraneural injection (2-3 mL). A 6 month follow up failed to

demonstrate nerve injury. Incidentally it is important to notice that the local anesthetic

mixture injected (bupivacaine plus lidocaine) contained 3 microgr/mL of epinephrine.

Since peripheral nerves are formed by neural tissue (fascicles) and connective

tissue, it is possible to penetrate the nerve (intraneural), but still be extrafascicular. In

2004 Sala-Blanch et al reported in Anesthesiology two cases of inadvertent intraneural,

extrafascicular injection after anterior approach of the sciatic nerve block with nerve

stimulation performed in two diabetic patients, as evidenced by CT scan. These two cases

also demonstrate that painless nerve punctures and even intraneural (although

extrafascicular) injections are possible without apparent sequelae.

A preexisting neurological injury should always be documented. It is important to

realize that nerve damage can occur perioperatively for a reason other than regional

anesthesia. Nerves can be injured during surgery by direct trauma, use of retractors and

tourniquets and by improper positioning. Nerves can also be damaged postoperatively by

a tight cast or splint, wound hematoma or surgical edema.

Use of epinephrine

Epinephrine-containing local anesthetic solutions may theoretically produce nerve

ischemia by vasoconstriction of the epineural and perineural blood vessels. Patients at

increased risk would be those with previous impaired microcirculation (e.g., diabetics).

There is no evidence at this time to suggest a detrimental effect of epinephrine in regional

anesthesia, as used in clinical practice. Epinephrine has been used extensively and

presumably safely in regional anesthesia. As mentioned earlier in reference to nerve

injury, in 2006 Bigeleisen reported intraneural injection in the axilla with local anesthetic

without apparent problems. It is interesting to mention that the author injected local

anesthetic containing 3 microgr/mL of epinephrine. We use epinephrine 1:400,000 (2.5

microgr/mL) extensively, in all kind of patients, and we appreciate its role as indicator of

inadvertent intravascular injection (please see further discussion on epinephrine in local

anesthetic chapter).

Persistent paresthesia, Clinical presentation

The symptoms can appear within 24 h after the injury, but sometimes they do not

present until days or weeks after the offending procedure took place. The degree of

symptoms is usually related to the severity of the injury. The cases are usually mild with

symptoms like tingling and numbness that usually disappear within weeks, or more rarely

they can progress to severe cases of neuropathic pain and motor involvement that can last

months and even years.

Page 67: Manual Regional Carlo 2010

67 | P a g e

Pre-existing neurologic condition and regional anesthesia

A pre existing neurologic condition per se is not a contraindication to regional

anesthesia. However a careful preoperative assessment must be performed, and any

neurological deficit must be documented in the patient’s chart. A thorough discussion

with the patient and the surgeon is always important.

Certain progressive neurologic conditions like multiple sclerosis, acute

poliomyelitis, amyotrophic lateral sclerosis and Guillian Barre syndrome are relative

contraindications to regional anesthesia, because the development of new symptoms

postoperatively may be confused with complications from the nerve block. In these cases

the risks and benefits must be carefully evaluated before proceeding with regional

anesthesia. In 2006 Koff et al published in Anesthesiology a case of severe plexopathy

after an ultrasound-guided interscalene block in a patient with multiple sclerosis.

There are other stable neurologic conditions like a preexisting peripheral

neuropathy, inactive lumbosacral radiculopathy and neurologic sequelae of stroke that

can be adequately managed with regional anesthesia, provided that all preexisting

neurological deficits are well documented in the chart.

Persistent paresthesia prevention and management

In order to minimize the risk of neurologic injury after regional anesthesia the

anesthesiologist needs to consider several factors, including procedure, patient and

surgeon. A meticulous nerve block technique, avoiding direct trauma to the nerve and

appropriate selection of local anesthetic volume and concentration are important. The role

of vasoconstrictors, especially low dose (1:400,000), on clinical development of neural

ischemia, has not been elucidated.

When a neuropathy develops in the postoperative period, a prompt evaluation is

necessary and a multidisciplinary approach, with participation of neurology, radiology,

and surgery, is recommended. A detailed history must be obtained including the timing

and nature of symptoms. A physical exam should look for any signs of hematoma or

infection. A neurological exam by a neurologist is also crucial.

Electrophysiological testing

Although electrophysiological studies remain normal for 14 to 21 days after the

injury, ordering them early could help establish a baseline and rule out any preexisting

condition. These tests have limitations, as they only assess large motor and sensory fibers

and not small unmyelinated fibers. They usually include nerve conduction velocity

studies and electromyography and sometimes may include evoked potentials.

1. Sensory Nerve Conduction Studies

They assess functional integrity of sensory nerves by measuring amplitude and

velocity of peripheral nerve conduction. Injuries involving fascicular damage

primarily show a decrease in the amplitude of the action potential, a sign that the

impulses are being transmitted by a reduced amount of fibers. Conduction

velocity in these cases may be minimally affected. When the lesion is

Page 68: Manual Regional Carlo 2010

68 | P a g e

demyelinating, like the ones seen after tourniquet compression, nerve conduction

velocity is greatly affected while the amplitude remains normal.

2. Electromyography It records electrical activity in the muscles helping to locate the denervated

muscles in reference to the level at which the nerve damage has occurred. Within

2-3 weeks post injury, spontaneous activity can be recorded from the muscle, in

the form of sharp waves and muscle fibrillation. After 3 months the pattern may

change, as nerve regeneration by “sprouting” takes place. In permanent injuries,

electromyography remains abnormal.

Use of tourniquet

Use of crude compression devices to control surgical bleeding from the

extremities, can be, according to Bailey, traced back to ancient Rome. The term

“tourniquet” was apparently first used by Petit in France in 1718 to describe a mechanical

screw-like contraption that he introduced to provide surgical hemostasis. Lister in 1864

was the first surgeon who used a tourniquet to produce a bloodless surgical field. Modern

tourniquet devices have a microprocessor, use an air pump and are able to accurately and

safely maintain the desired pressure. A fail-safe mechanism protects from pressure ever

exceeding 500 mmHg.

Tourniquet time: Recommended tourniquet time varies, but the most commonly

accepted limit is 2 hours. This recommendation is based on a work by Wilgis, published

in 1971 in which he demonstrated more acidosis after 2 hours of ischemia. Surgeons

should be made aware when the 2-hour limit has been reached and the tourniquet should

be deflated at that time, unless the procedure is at a crucial time. This communication

with the surgical team needs to be documented in the chart.

Despite the widely accepted 2-hour limit, Klenerman, as cited by Bailey, showed

minimal muscle damage with tourniquet times not exceeding 3 hours, using electron

microscopy.

Some people advocate deflating the tourniquet at 1.5 h for 5-15 minutes followed by an

additional 1.5 h of inflation time.

Tourniquet inflation pressure: It is believed that inflation pressure is more important of

a factor than time in influencing injury. It is recommended to use the minimum inflation

pressure that accomplishes ischemia. In general 100 mmHg above the systolic pressure is

a common setting. Roekel and Thurston in 1985 showed that 200 mm Hg for the upper

extremity and 250 mm Hg for the lower extremity were adequate parameters. Adding

layers of padding is important. Wrinkles in the padding should be avoided, since they

may become pressure points.

Tourniquet associated problems: The exsanguination with an Esmarch bandage prior to

tourniquet inflation causes an increase in preload, which can be significant when bilateral

tourniquets are used in the lower extremities. Eliminating circulation in part of one

extremity also can lead to an increase in afterload. This may cause problems in patients

with cardiac problems and decreased cardiac output. Exsanguination of lower extremities

has also been associated with pulmonary embolism and cardiovascular collapse.

Page 69: Manual Regional Carlo 2010

69 | P a g e

Some patients may develop post-tourniquet nerve palsy, affecting more frequently larger

motor fibers than sensory fibers. These lesions are usually reversible. The magnitude and

duration of the compression dictate the severity of the injury.

Patients can also develop “post-tourniquet syndrome”, a clinical picture characterized

by interstitial edema, arm weakness and numbness secondary to cell injury and alteration

or permeability. It usually resolves within a week.

When the tourniquet is deflated blood pressure drops (sudden drop in preload and

afterload) and heart rate increases as blood rushes into an ischemic, vasodilated bed

(reactive hyperemia).

Carbon dioxide and potassium levels increase and so does lactic acid leading to acidosis.

These effects peak at about 3 minutes post deflation. There is also a decreased in patient’s

temperature.

Tourniquet pain: It is commonly observed despite signs of otherwise good anesthesia of

the extremity. Unpremedicated volunteers refer intolerable pain by 30 minutes. Signs of

tourniquet pain, manifested as a gradual rise in blood pressure, are also observed under

neuraxial blocks and general anesthesia. Patients report this pain under the tourniquet and

distal to it.

Controversy exists as to how this pain is transmitted. De Jong and Cullen in 1963

proposed that tourniquet pain was transmitted by small non-myelinated sympathetic

fibers. However tourniquet pain can arise even when high thoracic levels of anesthesia

are present.

It seems that tourniquet pain is transmitted, as other painful sensations, by A-delta

myelinated fibers and C unmyelinated fibers. Tourniquet pain is usually described as

burning, cramping or heaviness. The burning and aching sensations, characteristics of

ischemia, are believed to be conducted by unmyelinated fibers (MacIver and Tanelian,

1992), while the sharp pain, usually a small component of tourniquet pain, is transmitted

by A-delta fibers.

MacIver and Tanelian proposed that C fiber activation by ischemia-induced alterations

are responsible for tourniquet pain. They studied in an in-vitro model the effects of

ischemic alterations (i.e., hypoxia, hypoglycemia, lactic acid, and decreased ph) on A-

delta and C pain fibers. They showed that hypoxia and hypoglycemia induced under

ischemia, increased C fiber tonic action potential activity, but did not affect A-delta

fibers. Increased lactate and decreased pH did not alter the discharge frequency of C

fibers in this model. The activation of C fibers by ischemia products seems crucial in

tourniquet pain. Whether these C fibers eventually enter the spinal cord at a level above

the somatic nerve block is debatable.

Page 70: Manual Regional Carlo 2010

70 | P a g e

References

1. Mulroy MF: Complications of Regional Anesthesia, In: Mulroy MF, Regional

Anesthesia, 3rd

edition. Philadelphia, Lippincott Williams & Wilkins, 2002, pp

29-41

2. Selander D, Dhuner KG, Lundborg G. Peripheral nerve injury due to injection

needles used for regional anesthesia. Acta Anaesth Scan 1977; 21: 182-188

3. Rice ASC, McMahon SB. Peripheral nerve injury caused by injection needles

used in regional anaesthesia: Influence of bevel configuration, studied in a rat

model. Br J Anaesth 1992; 69: 433-438

4. Selander D: Peripheral Nerve Injury After Regional Anesthesia, In: Finucane BT

(ed), Complications of Regional Anesthesia. New York, Churchill Livingstone,

1999, pp 105-115

5. Horlocker TT: Persistent paresthesia, In: Atlee JL (ed), Complications in

Anesthesia. Philadelphia, W.B. Saunders Company, 1999, pp 290-292

6. Auroy Y, Narchi P, Messiah A, et al. Serious complications related to regional

anesthesia. Results of a prospective survey in France. Anesthesiology 1997; 87:

479-486

7. Cheney FW, Domino KB, Caplan RA, et al. Nerve injury associated with

anesthesia: A closed claims analysis. Anesthesiology, 1999; 90: 1062-1069

8. Lee LA, Posner KL, Domino KB, et al. Injuries associated with regional

anesthesia in the 1980s and 1990s: A closed claims analysis. Anesthesiology,

2004; 101: 143-152

9. Morgan GE, Mikhail MS, Murray MJ: Clinical Anesthesiology, 4th

edition. New

York, McGraw-Hill, 2006, pp324-358

10. Hebl JR: Peripheral Nerve Injury, In: Neal JM, Rathmell JP, Complications In

Regional Anesthesia and Pain Management. Philadelphia, Saunders Elsevier,

2007, pp 125-140

11. Hadzic A: Textbook of Regional Anesthesia and Acute Pain Management.

McgRaw-Hill, 2007

12. Sites BD: Introduction to Ultrasound-Guided Regional Anesthesia: Seeing Is

Believing, In: Schwartz AJ (ed), ASA Refresher Courses in Anesthesiology,

2006, pp 151-163

13. Bailey MK: Use of the Tourniquet in Orthopedic Surgery, In: Conroy JM,

Dorman H (eds), Anesthesia for Orthopedic Surgery. New York, Raven Press,

Ltd, 1994, pp 79-88

14. MacIver MB, Tanelian DL. Activation of C fibers by metabolic perturbations

associated with tourniquet ischemia. Anesthesiology 1992; 76: 617-623

15. Hamid B, Zuccherelli L: Nerve Injuries, In: Boezaart AP (ed), Anesthesia and

Orthopedic Surgery. New York, McGraw-Hill, 2006, pp 405-419

16. Darmanis S, Papanikolaou A, Pavlakis D. Fatal intra-operative pulmonary

embolism following application of an Esmarch bandage. Injury 2002; 33: 761-764

17. Lu CW, Chen YS, Wang MJ. Massive pulmonary embolism after application of

an Esmarch bandage. Anesth Analg 2004; 98: 1187-1189

Page 71: Manual Regional Carlo 2010

71 | P a g e

18. Martin G, Breslin D, Stevens T: Anesthesia for Orthopedic Surgery, In:

Longnecker DE, Brown DL, Newman MF, Zapol WM (eds), Anesthesiology.

New York, McGraw Hill, 2008, pp 1541-1557

19. Bigeleisen PE. Nerve puncture and apparent intraneural injection during

ultrasound-guided axillary block does not invariably result in neurological injury.

Anesthesiology 2006; 105: 779-783

20. Sala_Blanch X, Pomes J, Matute P, Valls-Sole J, Carrera A, Tomas X Garcia-

Diez A. Intraneural injection during anterior approach for sciatic nerve block.

Anesthesiology 2004; 101: 1027-1030

21. Koff MD, Cohen JA, McIntyre JJ, Carr CF, Sites BD. Severe brachial plexopathy

after an ultrasound-guided single-injection nerve block for total shoulder

arthroplasty in a patient with multiple sclerosis. Anesthesiology 2008; 108: 325-

328

22. Sala-Blanch X, Ribalta T, Rivas E, Carrera A, Gaspa A, Reina MA, Hadzic A.

Structural injury to the human sciatic nerve after intraneural needle insertion. Reg

Anesth Pain Med 2009; 34: 201-205

Page 72: Manual Regional Carlo 2010

72 | P a g e

CHAPTER 6

UPPER EXTREMITY NERVE BLOCKS

Page 73: Manual Regional Carlo 2010

73 | P a g e

UPPER EXTREMITY BLOCKS

Anatomy of the brachial plexus

Roots

The brachial plexus is most frequently formed by five roots originating from the

ventral divisions of spinal nerves C5 through T1. After exiting through the corresponding

intervertebral foramen, the roots of the plexus are found in the cervical paravertebral

space, between the anterior and middle scalene muscles. In the cervical region the spinal

roots emerge above the corresponding cervical vertebrae, as seen in figure 6-1. Because

there are 8 cervical nerves but only 7 cervical vertebrae, starting with T1 the spinal roots

emerge below the corresponding vertebra (e.g., T1 exits between T1 and T2).

The distance from C5 to T1 roots is large and irreducible, and equal to the height

of four vertebrae. This fact in itself could help explain the frequent lack of dense

anesthesia in the C8-T1 dermatomes after an injection performed at the level of the C5-

C6 roots (interscalene block). Another important and frequently ignored reason is the

expansive wave created by the pulse of the subclavian artery and felt mostly by the distal

roots of the plexus (C8-T1), the lower trunk and its divisions. Because the local

anesthetic diffuses to points of least resistance, this expanding pulsatile force may keep

the local anesthetic from reaching the most distal elements of the plexus.

In addition to knowing the formation of the plexus and its architecture throughout

its trajectory, it is also important from my perspective to understand the plexus in terms

of its relative surface area at different locations. The five roots occupy an area that is

elongated in the frontal plane, but very narrow in the sagittal plane (anteroposterior).

When the five roots combine together to form three trunks, not only there is a 40%

Fig 6-1. Left supraclavicular area. The

sternocleidomastoid, scalene muscles, great

vessels, soft tissue and fascias have been

removed. The vertebral artery is shown in red.

The roots of the plexus are seen exiting in

between two vertebrae. The dome of the pleura is

shown in blue at the bottom of the image.

(Dissection by Dr. Franco. Copyrighted image).

Page 74: Manual Regional Carlo 2010

74 | P a g e

reduction in the number of nerve structures (from 5 to 3), but also the trunks become

physically contiguous, as shown in figure 6-2, helping reduce their combined surface

area. In fact this is the point at which the brachial plexus is reduced to its smallest surface

area. This striking convergence of innervation is unique to the brachial plexus and has no

parallel in the lower extremity and helps explain the rapid onset and high success rate of

the supraclavicular approach. The surface area of the plexus increases again when the

trunks originate six divisions although they stay together so the small increase in surface

area is compensated by a larger surface of absorption. The surface area increases the most

at the level of the axilla where the plexus gives off the terminal branches.

The scalene muscles

The anterior scalene muscle originates in the anterior tubercles of the transverse

processes of C3 to C6 and inserts on the scalene tubercle of the superior aspect of the first

rib. The middle scalene muscle originates in the posterior tubercles of the transverse

processes of C2 to C7 and inserts on a large area of superior aspect of the first rib, behind

the subclavian groove.

Brachial plexus structure: Trunks to terminal branches

The five roots converge toward each other to form three trunks -upper, middle and

lower- stacked one on top of the other, as they traverse the triangular interscalene space

formed between the anterior and middle scalene muscles. This space becomes wider in

the anteroposterior plane as the muscles approach their insertion on the first rib.

While the roots of the plexus are long, the trunks are almost as short (app 1cm) as

they are wide, soon giving rise to a total of six divisions (three anterior and three

posterior), as they reach the clavicle. The area of the trunks corresponds to the point

where the brachial plexus is confined to its smallest surface area, three nerve structures,

closely related to one another, carrying the entire sensory, motor and sympathetic

innervation of the upper extremity, with the exception of a small area in the axilla and

upper middle arm, which is innervated by the intercostobrachial nerve, a branch of the

Fig. 6-2. Left supraclavicular area. The SCM,

great vessels and fascias have been removed. The

trunks (S, M, I) of the plexus are seen emerging in

between the anterior scalene (AS) and medial

scalene (MS). Also shown are the anterior (a) and

posterior (p) divisions of the upper trunk and its

supraescapular branch (supr), the subclavian artery

(SA) and vertebral artery (VA). (Dissection by Dr.

Franco. Copyrighted image).

Page 75: Manual Regional Carlo 2010

75 | P a g e

second intercostal nerve. This great reduction in surface area allows the plexus to

negotiate the narrow passage between the clavicle and the first rib at the apex of the

axilla.

The brachial plexus, represented by its divisions, enters the apex of the axilla

lateral to the axillary artery, the latter being the continuation of the subclavian artery. In

order to offer a short profile the neurovascular bundle “spread” from medial to lateral

with the axillary vein the most medial structure, followed by the axillary artery in the

center and the divisions of the plexus most lateral, as shown in figure 6-3 and 6-4.

It is important to realize that immediately below the clavicle and before arriving

at the coracoid process, the six divisions of the plexus and the origin of the three cords

are located lateral to the artery and not around it (see figures 6-3 and 6-4). This is an

important anatomical detail while considering different infraclavicular approaches.

As the cords approach the level of the coracoid process the lateral cord remains on

the lateral side while the posterior and medial cords migrate behind the artery adopting all

of them the characteristic position around it from which they take their name. At this

level the cords are covered superficially by pectoralis minor and pectoralis major

muscles. It seems to me important to mention that the rotation of the cords behind the

artery from their original lateral position is usually arrested before the medial cord

reaches a true medial position with respect to the artery and before the posterior cord get

to be truly posterior to it. So, a cross section of the neurovascular bundle at the level of

the coracoid process reveals that the cords are not exactly located at the 3, 6 and 9

o’clock position Instead, on the right side, the lateral cord is usually in position 10

(anterolateral), the posterior cord is in position 7 (posterolateral) and the medial cord is in

position 4 (posteromedial). On the left side, the lateral cord is in position 2

(anterolateral), the posterior cord in position 5 (posterolateral) and the medial cord in

Fig 6-3. Neurovascular bundle under the

clavicle, left side. The soft tissue and fascias have

been removed for clarity. The neurovascular bundle

crosses under the clavicle with the vein most

medial, the axillary artery in the center and then the

divisions most laterally. (Dissection by Dr. Franco.

Copyrighted image).

Fig 6-4. Neurovascular bundle under the

clavicle in cross section, left side. The alignment

of the neurovascular bundle under the clavicle is

shown in cross section with some of the

connective tissue intact. The arrows point to the

6 divisions located lateral to the axillary artery.

(Dissection by Dr. Franco. Copyrighted image).

Page 76: Manual Regional Carlo 2010

76 | P a g e

position 8 (posteromedial). This means that usually there are two cords on the lateral side

of the artery (lateral and posterior cords) and only one on its medial side (medial cord),

making the approach from the lateral side more rational, especially during blind

techniques.

At about the level of the lateral border of the pectoralis minor muscle the three

cords give off their terminal branches. The posterior cord originates the axillary and

radial nerves; the medial cord originates part of the median nerve, plus the ulnar, medial

brachial and medial antebrachial cutaneous nerves. The lateral cord originates the rest of

median nerve and musculocutaneous nerve. Sometimes the musculocutaneous nerve

remains attached to the median nerve until reaching the proximal arm.

Brachial plexus sheath

For some authors the uneven spread of local anesthetic frequently observed after a

single injection in the axilla is enough evidence that the brachial plexus sheath does not

exist. This is not necessarily so. On the one hand, it is clear both in the surgical suite and

the anatomy laboratory, that connective tissue surrounds all types of neurovascular

structures in the body, especially in more exposed areas like the axilla, the neck, the

groin, etc. It is a reality that nerves and vessels are immersed in a connective tissue

matrix and are not simply “cables” lying between two muscular planes. Our own

published cadaver studies have provided macroscopic photographic evidence of its

existence. Part of that is shown in figures 6-5 A, B and C.

Fig 6-5 A and B. Axillary sheath. Left axillary region dissection showing A: the axillary sheath

intact from which the musculocutaneous nerve (MCN) is seen exiting. B: shows the same specimen

after the sheath has been open. The intra sheath portion of the MCN can be seen taking off from the

lateral cord (LC). Cadaver dissection by Dr Franco. Images are copyrighted.

(On a model with permission). Fig 6-5 C. Axillary sheath in cross section.

The axillary sheath just below the clavicle

(apex) is shown with arrows as a well defined

sturdy fascia surrounding the neurovascular

bundle. The interior is otherwise filled with

loose connective tissue. The three cords of the

plexus (c) are shown lateral to the artery.

Cadaver dissection by Dr Franco. Image is

copyrighted.

Page 77: Manual Regional Carlo 2010

77 | P a g e

Ultrasound, on the other hand, has confirmed that nerves and vessels in all regions

of the body, but especially in exposed places like the axilla, are embedded in a matrix of

soft connective tissue surrounded by a fascial sheath. This loose connective tissue within

the sheath “soaks” the local anesthetic towards areas of least resistance (i.e., along the

nerves) and by doing so this soft connective tissue becomes an obstacle to free

circumferential diffusion. In addition to this internal (within the sheath) factor, there are

other factors outside the sheath that may also play a role in the spread of local anesthesia

within it. Some of these factors have to do with the nature of the tissues through which

the neurovascular bundle travels through and the pressure they exert over the sheath. In

the axilla, for example, the sheath and its content are resting posteriorly over muscle and

bone (scapula). So its posterior aspect is subjected to more outside pressure or resistance

than its anterior aspect that is only covered by the complaint axillary fat. As a result local

anesthetic, which can only move to points of least resistance, will have more difficulty

spreading from the anterior aspect of the axillary sheath to its posterior aspect than vice

versa and this difficulty in the spread within the sheath has nothing to do with any

specific septa.

Distribution of the branches of the brachial plexus

Axillary nerve (C5-C6): gives an articular branch to the shoulder joint, motor innervation

to the deltoid and teres minor muscles and sensory innervation to part of deltoid and

scapular regions.

Radial nerve (C5-C6-C7-C8): supplies the skin of the posterior and lateral arm down to

the elbow, the posterior forearm down to the wrist, lateral part of the dorsum of the hand

and the dorsal surface of the first three and one-half fingers proximal to the nail beds. It

also provides motor innervation to the triceps, anconeus, part of the brachialis,

brachioradialis, extensor carpi radialis and all the extensor muscles of the posterior

compartment of the forearm. Its injury produces a characteristic “wrist drop”.

Median nerve (C5-C6-C7-C8-T1): gives off no cutaneous or motor branches in the axilla

or the arm. In the forearm it provides motor innervation to the anterior compartment

except the flexor carpi ulnaris and the medial half of the flexor digitorum profundus

(ulnar nerve). In the hand provides motor innervation to the thenar eminence and the first

two lumbricals. It provides the sensory innervation of the lateral half of the palm of the

hand and dorsum of first three and one-half fingers including the nail beds.

Ulnar nerve (C8-T1): like the median nerve, the ulnar nerve does not give off branches in

the axilla or the arm. Its motor component supplies the flexor carpi ulnaris and the medial

half of the flexor digitorum profundus. In the hand it provides the motor supply to all the

small muscles of the hand except the thenar eminence and first two lumbricals (median).

Its sensory branches supply the medial third of the dorsum and palmar sides of the hand

and dorsum of the 5th

finger and dorsum of the medial side of 4th

finger.

Medial brachial cutaneous nerve (T1): it is solely a sensory nerve. It supplies the skin of

the medial side of the arm. It is joined here by the intercostobrachial nerve, branch of the

second intercostal.

Page 78: Manual Regional Carlo 2010

78 | P a g e

Medial antebrachial cutaneous nerve (C8-T1): It is also a sensory nerve. It supplies the

medial side of the anterior forearm.

Musculocutaneous nerve (C5-C6-C7): gives motor innervation to the choracobrachialis,

biceps and brachialis muscles. At the elbow it becomes purely sensory innervating the

lateral anterior aspect of the forearm to the wrist.

Pearls

With the shoulder down the three trunks of the brachial plexus and the origin of

the divisions are located above the clavicle, therefore during a supraclavicular

block the needle does not need to reach below the clavicle.

For the most part the first intercostal space is located below the clavicle (with the

exception of the most posterior paravertebral part), therefore its penetration is

unlikely during a properly performed supraclavicular block.

During procedures using a needle in the supraclavicular area, the needle should

never cross medial to the parasagital plane of the anterior scalene muscle because

of risk of pneumothorax.

The pulsatile effect of the subclavian artery exerted mainly against C8-T1 roots

and the lower trunk explains why the C8-T1 dermatome can be spared during

interscalene and supraclavicular blocks. To avoid this problem during a

supraclavicular block the injection needs to be performed in the vicinity of the

lower trunk or its divisions, evidenced by fingers twitch with a nerve stimulator

or by injecting between the subclavian artery and the first rib when using

ultrasound. In the case of interscalene block this is usually not a problem since its

main indication is anesthesia/analgesia of the shoulder that does not require

anesthesia of C8-T1 dermatomes.

The SCM muscle inserts on the medial third of the clavicle, the trapezius muscle

on the lateral third of it, leaving the middle third for the neurovascular bundle.

These proportions are maintained regardless of patient’s size. Bigger muscle bulk

through exercise does not influence the size of the muscle insertion area.

Page 79: Manual Regional Carlo 2010

79 | P a g e

INTERSCALENE BLOCK

NERVE STIMULATOR TECHNIQUE

Indications

Its main indication is anesthesia or analgesia of the shoulder, including the

clavicle and proximal part of the humerus.

Point of contact of the needle with the brachial plexus

The needle approaches the plexus at the level of the roots, high in the interscalene

space, approximately at the level of C5-C6 roots (most likely C5).

Main characteristics

This block is superficial and usually easy to perform. Characteristically it misses

the C8-T1 dermatomes, which include the sensory territories of ulnar, medial

antebrachial cutaneous, and medial brachial cutaneous nerves (medial side of the upper

extremity).

Patient position and landmarks

The patient is lightly sedated. Older, obese and recent trauma patients can be

expected to be extremely sensitive to the depressant effects of benzodiazepines and/or

narcotics. Titrate to effect.

The patient is placed in a semi sitting position and the space between the cricoid

and thyroid cartilages (cricothyroid membrane) is located and marked as shown in figure

6-6. The patient is asked to lower his shoulders and to slightly rotate the head to the

opposite side. It is important to emphasize here that the patient should rotate and not

incline the head away so as to keep the midline neutral. With the midline in neutral

position the intervertebral foramen looks caudal, lateral and slightly posterior. Tilting the

head away from the operator, on the other hand, could align the intervertebral foramen

with the needle trajectory.

A horizontal plane that starts at the cricothyroid membrane medially and

intercepts the posterior border of the SCM laterally is established, as shown in figure 6-7.

Fig 6-6. Cricoid thyroid

membrane. The level of the

cricothyroid membrane is

located by palpation and

marked on the skin.

(On a model with permission).

Page 80: Manual Regional Carlo 2010

80 | P a g e

The index and middle fingers of the palpating hand are placed behind the SCM at

this level pushing it slightly forward (medially), as shown in figure 6-8. This maneuver

brings the palpating fingers under the SCM and on top (anterior) to the anterior scalene

muscle. The fingers are then rolled back until they fall into the interscalene groove, which

at this proximal point in the neck is a real structure and easy to identify. This is the point

of needle insertion.

Type of needle

A 2.5 cm or 5cm, 22-G, insulated needle can be used.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 0.8-0.9 mA, at a pulse frequency

of 1 Hz and pulse duration of 0.1 msec (100 microsec). A small skin wheal is raised with

1% lidocaine or 1% mepivacaine using a small needle (ideally 27-G).

Needle insertion

The needle is introduced between the two palpating fingers in a medial and

slightly caudal direction, but most importantly with a 20 to 30-degree posterior direction,

as shown in figure 6-9.

Fig 6-8. Point of needle insertion. The

interscalene groove is found at the

intersection of the cricoid plane with the

posterior border of the SCM.

(On a model with permission).

Fig 6-7. Cricoid-SCM plane. The

level of the cricothyroid membrane is

projected laterally to intercept the

posterior border of the SCM.

(On a model with permission).

Page 81: Manual Regional Carlo 2010

81 | P a g e

It is important to realize that this is a superficial block that should take place

within the confines of the tips of the palpating fingers and not beyond them. In no

circumstance the needle should be introduced further than the projection of the clavicular

head of the SCM.

Any distal motor twitch as well as biceps, triceps or deltoid muscles are adequate.

There is some controversy in the literature as to whether a shoulder twitch is acceptable

for an interscalene block. Besides the usual arm twitches, anatomical and clinical

evidence support accepting deltoid twitches. Motor twitches from trapezius (spinal

accessory nerve) and levator scapulae (dorsal scapular nerve) are not acceptable. For

further reading on this issue please see: Silverstein W et al. Interscalene block with a

nerve stimulator: A deltoid motor response is a satisfactory endpoint for successful block.

Reg Anesth Pain Med 2000; 25:356-359 and accompanying editorial by William Urmey,

same journal page 340-342.

A twitch of the abdomen signals phrenic nerve stimulation and it is evidence that

the needle is anterior to the anterior scalene. In this case the needle should be withdrawn

and redirected slightly posteriorly. A motor twitch of the scapulae or trapezius muscle

indicates that the position of the needle is too posterior and needs to be repositioned

anteriorly.

Local anesthetic and volume

For single shot techniques in adults, 30 mL of 1.5% mepivacaine plain provides

2-3 h of anesthesia. The addition of 1:400,000 of epinephrine prolongs the anesthesia to

about 3-4 h. The residual analgesia post anesthesia is variable in duration, although rarely

persists for more than 2 h after block resolution. The addition of lyophilized tetracaine

(20 mg per 10 mL of solution) to 1.5% mepivacaine, for a final concentration of 0.2%

tetracaine, provides around 6 h of surgical anesthesia.

Ropivacaine 0.5% can be used in the same volume to provide 12 h plus of

anesthesia. The injection of local anesthesia is performed slowly with frequent

aspirations.

Also 20 mL of 0.2% ropivacaine can be used to provide postoperative analgesia

for surgery performed under general anesthesia.

Side effects and complications

Systemic local anesthetic reaction can occur as with any block. More specific

(and frequent) side effects related to interscalene block are: Horner’s syndrome (ptosis,

miosis and anhydrosis) due to stellate ganglion block and hoarseness due to recurrent

Fig 6-9. Needle insertion. The

needle is advanced medial,

caudal and posterior.

(On a model with permission).

Page 82: Manual Regional Carlo 2010

82 | P a g e

laryngeal nerve involvement. Characteristically this block produces also 100% of

phrenic nerve block with diaphragmatic paralysis (Urmey et al, Anesth Analg, 1991).

This can produce dyspnea and reductions in respiratory volumes of up to 30%.

Pneumothorax is possible, but rare with this block.

Clinical pearls

Because of the position of the shoulder, so close to the head of the patient, the

anesthesiologist must carefully evaluate the patient and surgeon before deciding

to perform an interscalene block as the only anesthesia for the case. A nervous

patient and a rough surgeon could be indications for interscalene analgesia

combined with general anesthesia.

It must be remembered that some of these procedures are performed in positions

other than supine (e.g., beach chair, lateral), which can make the management of

the airway, if needed, a bit more challenging.

A language barrier between patient and anesthesiologist is also a relative

contraindication for interscalene block as the sole anesthetic.

This is a very superficial block that can be performed at 1-2 cm from the skin in

most patients.

Page 83: Manual Regional Carlo 2010

83 | P a g e

INTERSCALENE BLOCK

ULTRASOUND TECHNIQUE

Indications

Shoulder anesthesia and/or analgesia, including clavicle and proximal humerus.

Patient position

The patient is placed semi seated, with the shoulder down and the head slightly

turned the opposite way, as shown in figure 6-10.

Type of needle

A 2.5 cm or 5 cm, 22-G, insulated needle is what we frequently use.

Type of transducer

This is a superficial block for which a high frequency (8-15 MHz) linear probe is

well suited.

Scanning

Two are the most frequent ways to scan the neck to visualize the roots of the

plexus. One is to start a transverse scan over the sternocleidomastoid muscle (SCM) just

lateral to the cricoid cartilage, or start more distally parallel to the clavicle and then trace

the plexus proximally to the roots. In either case the probe ends in a semi transverse

position (cephalad rotation) overlapping the SCM with a slight distal orientation as

shown in figure 6-11.

Fig 6-10. Position. The patient is semi

seated, with shoulder down and head

slightly rotated to the opposite side. The

ultrasound machine is placed on the

opposite side.

(On a model with permission).

Page 84: Manual Regional Carlo 2010

84 | P a g e

The image obtained at this level is shown in figure 6-12.

Needle insertion

The needle can be advanced in plane from medial to lateral, or lateral to medial,

or out of plane usually from cephalad to caudal. It is our preference to insert the needle in

plane from lateral to medial as shown in figures 6-13 and 6-14.

Fig 6-11. Probe position. To get a good

perpendicular cut of the roots, the probe is

slightly rotated as shown. (On a model

with permission)

Fig 6-12. Interscalene image. With the

probe in the position shown in fig 6-11

it is possible to visualize part of the

SCM, ant and middle scalene muscles

and the proximal roots of the plexus.

(Author’s archive)

Fig 6-13. Needle insertion. The

needle is introduced in plane from

lateral to medial. (On a model with

permission)

Fig 6-14. US image. Vertical arrows show the

needle while the two horizontal arrows show

part of the hypoechoic spread of local

anesthetic around the plexus roots. (Author’s

archive)

alteral to medial. The

Page 85: Manual Regional Carlo 2010

85 | P a g e

This technique resembles Winnie’s classical interscalene approach. With the

needle under direct visualization the injection is performed in the proximity of C6 root.

The spread of local anesthetic should expand the interscalene space and bathe C5-C6 and

C7 roots, as shown in figure 6-14. If the spread is insufficient around a particular root the

needle is repositioned accordingly for a new injection.

Local anesthetic and volume

We use between 20-30 mL of local anesthetic of the same kind used for nerve

stimulation techniques.

Side effects and complications

The side effects and complications are essentially the same described for nerve

stimulation techniques. It is possible that ultrasound techniques, with a more targeted

injection and potentially smaller volumes, may theoretically decrease the incidence of

side effects, but this remains to be proven.

Page 86: Manual Regional Carlo 2010

86 | P a g e

SUPRACLAVICULAR BLOCK

NERVE STMULATOR TECHNIQUE

Indications

This block is indicated for any surgery on the upper extremity distal to the

shoulder or for analgesia of the entire upper extremity.

Point of contact of the needle with the brachial plexus

The needle approaches the plexus at the level of the trunks, and ideally the

injection should take place in the vicinity of the lower trunk.

Main characteristics

This block is considered by some as more difficult to learn than other upper

extremity blocks and historically it has been associated with a higher risk of

pneumothorax. The literature cites pneumothorax rates between 0.5-6.1 percent. However

with good anatomy and meticulous technique we have been able to practically eliminate

this risk.

A supraclavicular block is usually associated with a short onset, dense anesthesia

and high success rate. As we discussed it in the anatomy section, this is due to the

compact arrangement of the plexus at the level of trunks and divisions. Because of these

favorable characteristics, the supraclavicular block has been called the “spinal of the

upper extremity”.

We perform our own variation of the supraclavicular block, a very anatomical

approach that starts by determining the pleura boundaries as the first step. This allows us

to take advantage of such extraordinary block while limiting its potential drawbacks. Our

experience to late 2009 includes more than 5,000 supraclavicular blocks without ever

having demonstrated a single pneumothorax. A common question posed to us is whether

we perform routine chest X-rays after a supraclavicular block. The answer is no. In fact

we only do an X-ray when the clinical situation merits it (e.g., an unusually difficult

technique and or symptomatic patient). Traditionally our anesthesiology textbooks have

left the impression that a pneumothorax following a supraclavicular block has a late

onset, making the technique a bad choice for outpatients. Our review of the literature fails

to demonstrate this. In fact most of the cases of pneumothorax associated with

supraclavicular block published in the literature, have been diagnosed within a few hours

after the block and most of them have been investigated because of the patients’ early

symptoms. We perform this technique with great success in all kinds of patients,

including same day surgery and trauma patients.

Some history of the supraclavicular approach

The supraclavicular block was introduced into clinical practice in Germany by

Kulenkampff in 1911. A publication of his technique appeared later in the English

literature in 1928. Kulenkampff accurately described the plexus as being more compacted

in the neighborhood of the subclavian artery, where he rightly believed that a single

injection could provide adequate anesthesia of the entire upper extremity. Kulenkampff’s

technique was simple and in many ways sound. Unfortunately his recommendation to

introduce the needle toward the first rib, in the direction of the spinous process of T2 or

Page 87: Manual Regional Carlo 2010

87 | P a g e

T3, carried an inherited risk for pneumothorax that would be responsible for the

technique falling into disfavor.

Albeit with several modifications, the supraclavicular approach remained a

popular choice until the early 1960’s. Eventually, the combined effect of pneumothorax

fear and the introduction of the axillary approach by Accardo and Adriani in 1949, and

especially by Burnham in 1958, marked the beginning of the decline for one of the best

approaches in regional anesthesia.

The axillary approach introduced a good technique with its share of shortcomings

(e.g., smaller area of anesthesia than supraclavicular, tendency to produce “patchy”

blocks and lower overall success rate), but definitely devoid of pneumothorax risk. The

axillary block received a big push when in 1961 De Jong published an article in

Anesthesiology praising it. His paper was based on cadaver dissections and included the

now famous calculation of 42 mL as the volume needed to fill a cylinder 6 cm long that,

according to De Jong, “should be sufficient to completely bathe all branches of the

brachial plexus”. Coincidentally (or not) the same journal issue carried a paper by Brand

and Papper out of New York, comparing axillary and supraclavicular techniques in their

hands. This article is the source of the 6.1% pneumothorax rate frequently quoted for

supraclavicular block. The authors were determined to prove that the axillary block was

safer and better than the supraclavicular block. They succeeded by not only producing the

highest percentage of pneumothorax (6.1%), but the highest number (14 cases) for an

individual study. This study should be considered an aberration.

In retrospect these two articles could be considered the turning point at which the

axillary route became the preferred approach here in the United States and the rest of the

world. With some exceptions this is still true today. Fortunately ultrasound in regional

anesthesia has caused a renewed interest in this approach and we could not be happier.

The supraclavicular technique with its rapid onset, density, high success rate

along with large area of anesthesia are highly desirable. These good characteristics are,

according to David Brown and colleagues, “unrivaled” by other techniques. In our

practice the supraclavicular approach is the cornerstone of upper extremity regional

anesthesia.

Patient position and landmarks

The patient lies in the semi sitting position, the ipsilateral shoulder down and the

head turned to the opposite side, as shown in figure 6-15. The arm to be blocked is flexed

at the elbow and, if possible, the wrist is supinated to easily detect a twitch of the fingers.

Fig 6-15. Patient position. The patient

is semi seated with the head of the bed

elevated 30 degrees. The head of the

patient is turned away, the shoulder is

down and the arm is flexed at the elbow

and supinated at the wrist. (On a model

with permission).

Page 88: Manual Regional Carlo 2010

88 | P a g e

The point at which the clavicular head of the SCM muscle inserts in the clavicle is

then identified, as shown in fig 6-16. A parasagital (parallel to the midline) plane at this

level determines an “unsafe” zone medial to it, where the risk of pneumothorax is high

and a lateral zone that is safer.

Because the trunks are short and run in a very steep direction caudally towards the

clavicle, there is a narrow “window of opportunity” to perform the block above the

clavicle. It must be performed at enough distance from the insertion of the SCM on the

clavicle to be safely away from the pleural dome, but not too far to miss the trunks and

the plexus completely. We call this distance “the safety margin”. In adults we calculate

this distance to be about 1 inch (2.5 cm), which corresponds to the width of the author’s

thumb. This distance is marked on the skin over the clavicle for orientation, as shown in

figure 6-17.

This is only an orientation point that usually will coincide with the midpoint of

the clavicle in an adult patient. At this level the brachial plexus is usually easily palpable,

either as a groove or as some type of bump(s). This is usually called “interscalene

groove”, but the interscalene groove only exists high in the C5-C6 level. The groove is

lost more distally as the scalene muscles diverge from each other in the frontal and

sagittal planes. The palpation of the plexus is what determines the actual point of needle

entrance and not a fixed distance. The plexus can be palpated a few mm medial or

lateral to the orientation point, but never too far from it.

The palpating finger is placed parallel to the clavicle and the point of needle

entrance is located immediately cephalad to it, as shown in figure 6-18.

Fig 6-16. Lateral head of SCM. The

most lateral insertion of the SCM on

the clavicle is found and marked with

an arrow. Crossing this plane medially

increases the risk of pneumothorax.

(On a model with permission).

Fig 6-17. Safety margin. A safety

margin of 1” (2.5 cm) lateral to the

insertion of the SCM on the clavicle is

marked on the skin. (On a model with

permission).

Page 89: Manual Regional Carlo 2010

89 | P a g e

Type of needle

A 5cm, 22-G, insulated needle is used for this technique.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 0.8-0.9 mA, at a pulse frequency

of 1 Hz and pulse duration of 0.1 msec (100 microsec). A small skin wheal is raised with

1% lidocaine or 1% mepivacaine using a small needle (ideally 27-G).

Needle insertion

The needle is inserted first anteroposterior (toward the bed) with a 30 degree

caudal orientation, as shown in figure 6-19, for a distance of a few mm and up to 1.5 cm,

depending on the amount of subcutaneous tissue. After a short distance, a twitch of the

upper trunk (shoulder) is usually found as evidence that the needle is approaching the

frontal plane of the plexus.

The direction of the needle is then changed from anteroposterior to caudal,

advancing it parallel to the midline (and parallel to the most lateral pleural boundary),

with a slight (10 degrees) posterior orientation, as shown in figure 6-20.

Fig 6-18. Orientation arrows. The medial arrow

pointing up shows the lateral insertion of SCM

(pleura’s lateral boundary). The lateral arrow pointing

caudally shows the needle entrance point. The two

lateral arrows pointing at each other show the needle

trajectory (parallel to the patient’s midline). (On a

model with permission).

Fig 6-19. Needle insertion. The

needle is first introduced in a posterior

direction (toward the bed) with a 30

degree caudal orientation. (On a model

with permission).

Fig 6-20. Direction of the needle.

The needle is then advanced caudad,

parallel to the midline, with a slight

posterior orientation. (On a model

with permission).

Page 90: Manual Regional Carlo 2010

90 | P a g e

The reference to the midline is easy to ascertain and avoids confusion. The use of

other landmarks (e.g., nipple) provides lesser accuracy because of variability among

patients.

The needle is advanced caudally with a slight posterior angle to match the slight

posterior rotation of the plexus (the upper trunk is the most anterior and the inferior trunk

the most posterior). Because the trunks are physically contiguous, as the needle is

advanced, a twitch of the upper trunk (shoulder) should be followed by one from the

middle trunk (pectoralis, triceps, supination, pronation) and finally a twitch from the

lower trunk (wrist and fingers). The goal of the technique is to produce an isolated

muscle twitch of the fingers. Wrist flexion and extension are also acceptable responses,

but supination or pronation or any other more proximal motor twitches are not.

If after advancing the needle the motor twitch of the shoulder disappears and no

twitch is elicited from the middle trunk, it usually means that the angle of insertion of the

needle is not matching the orientation of the trunks, and that the tip of the needle is

wandering away from the trunks (usually anteriorly). The needle should be slowly

withdrawn until the original twitch is elicited once again, and then redirected either

posteriorly (most of the times) or anteriorly, but always parallel to the midline.

It is very important not to advance the needle more than 2 cm in the caudal

direction if no twitch is visible. In this case the situation should be reassessed starting

with the nerve stimulator and its connections and determination of landmarks. On the

other hand, when a twitch from the brachial plexus is being elicited the depth of needle

insertion is less important as such motor twitch reveals that the needle is still in close

proximity to the plexus.

Side effects and complications

Besides the common complications accompanying any block, the supraclavicular

technique can also be followed by Horner’s syndrome, hoarseness and phrenic nerve

palsy, but less frequently than after interscalene block. Neal et al in 1998 studied

diaphragmatic paralysis in 8 volunteers after supraclavicular block using ultrasound

(replicating what Urmey et al did in 1991 to demonstrate 100% of diaphragmatic

paralysis after interscalene block). They found an incidence of 50% of diaphragmatic

paralysis. No subject experienced changes in pulmonary function tests (PFTs) or

subjective symptoms of respiratory difficulty. This is our experience too.

In the issue of pneumothorax, I already mentioned that the literature cites a risk of

0.5% to 6.1%, the latter being an aberration. A careful and meticulous technique should

carry a minimal risk of pneumothorax. In our long experience including thousands of

cases in all sorts of patients we have never demonstrated a case of pneumothorax.

Clinical pearls

This is not a block for a practitioner that rarely performs peripheral nerve blocks.

The person interested in learning to perform it should first become familiar with

the anatomy of the supraclavicular area including the location of the dome of the

pleura. Using ultrasound makes the visualization of the pleura easier, but still

requires the operator to be familiar with the anatomy of the area.

Page 91: Manual Regional Carlo 2010

91 | P a g e

When using a nerve stimulator technique, the block should not be attempted

unless the insertion of the sternocleidomastoid in the clavicle is clearly

established. In fact this is a must especially for a person not experienced with the

technique. With time it becomes easier to ascertain the boundaries of the SCM.

It helps to know that the neurovascular bundle crosses the clavicle under the

midpoint of it, so this should be kept in mind as a reliable reference.

Due to the steep direction of the plexus from the neck to the axilla, the higher in

the neck (the further away from the clavicle) the more medial the plexus is. By the

same token, the further below the clavicle the more lateral to its midpoint the

plexus is.

The needle should never be inserted more than 2 cm caudal if no twitch is elicited.

This warning applies to every patient regardless of weight.

The injection should always be slow, alternated with frequent and gentle

aspirations. This technique provides time to recognize accidental intravascular

injection in those cases where blood is not aspirated. I also believe it helps to keep

the needle from moving backwards as a result of high speed flow at the tip of the

needle.

Page 92: Manual Regional Carlo 2010

92 | P a g e

SUPRACLAVICULAR BLOCK

ULTRASOUND TECHNIQUE

Indications

Anesthesia and/or analgesia for any procedure on the upper extremity distal to the

shoulder.

Patient position

The patient is placed in the semi seated position as shown in figure 6-21.

Type of needle

A 5cm, 22-G, insulated needle is used.

Type of transducer

This is also a superficial block for which a high frequency (8-15 MHz) is used.

Scanning

We usually start scanning medially, over the sternocleidomastoid muscle, right

above the clavicle, as shown in figure 6-22.

Fig 6-21. Position. The patient is

placed semi seated with the shoulder

down and the head turned the

opposite way. (On a model with

permission).

Fig 6-22. Probe position, first stage.

The probe is place over the SCM and

above and parallel to the clavicle. (On

a model with permission).

Page 93: Manual Regional Carlo 2010

93 | P a g e

At this level we get to see the dome of the pleura and above it, the subclavian vein

at that point where it is joining the internal jugular vein to form the brachiocephalic vein,

as shown in figure 6-23.

The probe is then slid laterally towards the midpoint of the clavicle, as shown in

figure 6-24,

At this level a cross section of the subclavian artery, the first rib and plexus can be

visualized, as shown in figure 6-25.

Fig 6-23. Scanning, first image.

With the probe over the SCM the

subclavian vein and pleural dome can

be visualized. (Author’s archive).

Fig 6-24. Probe position, second

stage. The probe is moved laterally

to visualize the plexus as it passes

over the 1st rib. (On a model with

permission).

Fig 6-25. Scanning the plexus above the

clavicle. The subclavian artery (SA) is seen

above the first rib, which is shown with three

arrows pointing up. A small arrow pointing

down shows the pleura while the larger single

arrow shows the position of the divisions of the

plexus. (Author’s archive).

Page 94: Manual Regional Carlo 2010

94 | P a g e

Needle insertion

The needle is advanced in plane, from lateral to medial, as shown in figure 6-26.

The entrance point is located at about 1 cm away from the probe to decrease the angle of

insertion and improve the chances of needle visualization.

The needle is then slowly advanced under direct visualization, towards the angle

formed by the first rib and the subclavian artery, as shown in figures 6-27 A and B.

Intermittent injections of small amounts of local anesthetic solution helps keep

contact with the tip of the needle as it advances and gently expands the volume of the

connective tissue surrounding the nerves, what has been called “hydro dissection”. This

contributes to clear a path for the needle decreasing the chances of inadvertent neural

puncture.

The goal of the supraclavicular technique is to see the spread of local anesthetic

reaching the angle between the first rib and the subclavian artery, as shown in figure 6-

28.

Fig 6-26. Needle insertion. The

needle is advanced in plane, from

lateral to medial. (On a model with

permission).

Fig 6-27, A and B. Needle insertion. The

needle is slowly brought behind the

subclavian artery (AA) and above the first rib.

(Author’s archive).

Page 95: Manual Regional Carlo 2010

95 | P a g e

Local anesthetic and volume

For single shot techniques in adults, 30 mL of 1.5% mepivacaine plain will

provide 2-3 h of anesthesia. The addition of 1:400,000 epinephrine prolongs the

anesthesia to about 3-4 h. The residual analgesia post anesthesia is variable in duration,

although it rarely persists for more than 2 h after block resolution. The addition of 2

mg/mL of lyophilized tetracaine to 1.5% mepivacaine, for a final concentration of 0.2%

tetracaine, prolongs the duration of surgical anesthesia to 4-6 hours.

Ropivacaine 0.5% can be used in the same volume to provide more than 12 h of

anesthesia.

Also 20-30 mL of 0.2% ropivacaine can be used to provide postoperative

analgesia for surgery performed under general anesthesia.

Fig 6-28. Injection. The local anesthetic

spread should be seen reaching the angle

formed by the 1st rib (vertical arrows

pointing up) and the subclavian artery

(SA). The local anesthetic is seen as a

hypoechoic (dark) shadow projecting from

the tip of the needle. (Author’s archive).

Page 96: Manual Regional Carlo 2010

96 | P a g e

INFRACLAVICULAR BLOCK

NERVE STIMULATOR TECHNIQUE

Indications

This block can provide anesthesia/analgesia of a large area of the upper extremity

including the elbow, especially if performed proximally near the apex of the axilla. It is

considered a good approach for continuous techniques because it offers more stability

than other more mobile locations.

Point of contact of the needle with the brachial plexus

The needle approaches the plexus at the level of the cords, or even divisions if the

block is performed proximally, closed to the clavicle.

Main characteristics The infraclavicular block could be considered an axillary block in which the

needle enters the axilla through its anterior wall (pectoralis muscles), instead of through

its base. The infraclavicular space of the anesthesiologists corresponds to part of the

axillary pyramid of the anatomists. With the arm in adduction it is represented on the skin

by a triangular area whose base is superior (clavicle), a medial wall formed by the

projection on the skin of the thoracic cage and a lateral wall formed by the medial side of

the upper arm. Depending on the patient’s amount of subcutaneous tissue and/or muscle

this block can be deep. Patients should be adequately sedated.

It is widely recommended when using a nerve stimulator to obtain a distal twitch

in the hand or wrist and to avoid either a biceps twitch (musculocutaneous nerve or

lateral cord) or pronation of the forearm (lateral cord). This recommendation is based on

clinical experience. A biceps twitch could be the result of musculocutaneous nerve

stimulation, outside the sheath, or from lateral cord stimulation inside the sheath. Because

the operator cannot accurately distinguish one from the other, this response is unreliable.

It is likely that a twitch from the posterior cord (elbow, wrist and or finger

extension) could be best, because the posterior cord is located at about the same distance

from the other two, and the spread of local anesthetic from this central location might be

more even. There could be another good reason to inject behind the artery, although it

may be more difficult to get there. Because the posterior structures (including the

posterior cord) are more closely packed, the spread of local anesthetic from anterior to

posterior may be more difficult than from posterior to anterior. Ultrasound, with

visualization of the axillary artery and the cords around it, makes this injection easier to

accomplish.

Different infraclavicular techniques have been described. A simple technique is

the coracoid approach first described by Whiffler in the British Journal of Anaesthesia in

1981 and later redefined by MRI studies performed in 40 volunteers by Wilson, Brown et

al, and published in Regional Anesthesia in 1998. This is the technique we most

frequently perform when using nerve stimulation.

Page 97: Manual Regional Carlo 2010

97 | P a g e

Patient position and landmarks

The patient is placed semi seated with the ipsilateral shoulder down. The arm is

slightly abducted 30-45 degrees, as shown in figure 6-29, to bring the neurovascular

bundle away from the thoracic cage and decrease the chance of pneumothorax.

As the neurovascular bundle follows the arm its relationship to the coracoid

process is pretty much maintained. The coracoid process is found by palpation at the

level of the deltopectoral groove (junction between the middle third with the lateral third

of the clavicle), about 2 cm below the clavicle, and marked on the skin, as shown in

figures 6-30 and 6-31.

Needle insertion point

The point of needle entrance is marked 2 cm caudal and 2 cm medial to the

coracoid process as shown in fig 6-32.

Fig 6-29. Patient position. The

patient lays semi seated with

shoulders down and the arm to be

blocked in slight abduction. (On a

model with permission).

Fig 6-30. Coracoid palpation. The

coracoid is found below the clavicle

in the deltopectoral groove. (On a

model with permission).

Fig 6-31. Coracoid marking. The

position of the coracoid is marked

on the skin. (On a model with

permission).

Page 98: Manual Regional Carlo 2010

98 | P a g e

Type of needle

It is possible to use sometimes a 5cm, 22-G insulated needle, but a 10cm, 21-G

insulated needle is usually necessary.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 0.8-0.9 mA, pulse frequency of

1 Hz and pulse duration of 0.1 msec (100 microsec).

Needle insertion

The needle attached to the nerve stimulator is advanced in the anteroposterior

direction, towards the bed, as shown in figure 6-33.

Before entering in contact with the plexus the needle passes through pectoralis

major and pectoralis minor muscles producing a visible local twitch. The brachial plexus

is found deep to them. If not response from the plexus is obtained, the needle is redirected

caudal (most of the times) or cephalad, but maintaining the same parasagital plane

without medial or lateral deviation.

Local anesthetic and volume

The nerve stimulator-guided infraclavicular technique usually requires a relatively

high volume of local anesthetic for better results. Usually 40 mL of 1.5% plain

mepivacaine will provide 2-3 h of anesthesia. The addition of 1:400,000 epinephrine

prolongs the anesthesia to about 3-4 h. The residual analgesia post anesthesia is variable

in duration, although rarely persists for more than 2 h after block resolution. The addition

Fig 6-32. Needle entrance point.

Two cm caudal and two cm

medial from the coracoid process.

(On a model with permission).

Fig 6-33. Needle insertion. The

needle is introduced from anterior

to posterior. (On a model with

permission).

Page 99: Manual Regional Carlo 2010

99 | P a g e

of 2 mg/mL of lyophilized tetracaine to 1.5% mepivacaine, for a final concentration of

0.2% tetracaine provides 4-6 h of surgical anesthesia.

Ropivacaine 0.5% can be used in the same volume for more than 12 h of

anesthesia. Also 20-30 mL of 0.2% ropivacaine can be used to provide postoperative

analgesia for surgery performed under general anesthesia.

Side effects and complications

Muscle pain and hematomas, which can be large in size, can happen.

Pneumothorax can occur due to injury of the pleura through an intercostal space.

Clinical pearls

This is a good place to put a catheter because it is easier to fix it.

Use adequate sedation, as this block is more uncomfortable for patients than other

more superficial blocks.

The junction between lateral and middle third of the clavicle can be used to locate

the deltopectoral groove and the coracoid process.

Placing the arm in slight abduction (30-40 degrees) brings the neurovascular

bundle away from the thoracic cage (it follows the arm) and decreases the chance

of pneumothorax.

Page 100: Manual Regional Carlo 2010

100 | P a g e

INFRACLAVICULAR BLOCK

ULTRASOUND TECHNIQUE

Indications

The same indications mentioned for nerve stimulation techniques, basically

anesthesia/analgesia of elbow, forearm wrist and hand.

Two main infraclavicular techniques

Ultrasound introduces a degree of flexibility to our techniques of regional

anesthesia that we did not have before. It certainly gives the operator the chance to

choose the best needle path based on the anatomy and the ultrasound image obtained,

without necessarily having to conform strictly to any particular technique already

described.

When using ultrasound in the infraclavicular area I distinguish two main

approaches, a proximal one just under the clavicle and a more distal one at the level of

the coracoid process. As I mentioned in the anatomy section, the brachial plexus crosses

under the clavicle as divisions before forming three cords. The divisions and the proximal

trajectory of the cords below the clavicle are located lateral to the axillary artery. When

the cords approach the coracoid process they rotate and surround the artery to take the

position from which they get their names.

Based on these two different dispositions of the plexus with respect to the axillary

artery I will describe two techniques.

Patient position

We perform both techniques with the patient in the semi seated position with the

shoulder on the side to be blocked down and the arm in abduction of about 45 degrees, as

shown in figure 6-34. Abducting the arm improves the ultrasound image of the

neurovascular bundle, perhaps by stretching it and bringing it closer to the anterior wall.

Type of needle

A 5cm, 22-G, insulated needle can be used in some patients, but it is usually

necessary to use a 10cm, 21-G, insulated needle due to the depth of the neurovascular

bundle at this location. Because the needle crosses through muscle, good sedation is

important as well as injection of local anesthetic in the intended needle path to keep the

patient comfortable.

Fig 6-34. Patient position. The

patient is semi seated, shoulder

down, arm abducted. (On a model

with permission).

Page 101: Manual Regional Carlo 2010

101 | P a g e

PROXIMAL INFRACLAVICULAR TECHNIQUE

Type of transducer

Depending on the thickness of the patient’s chest wall the operator can use a

linear high frequency (8-15 MHz) probe or a curved low frequency (3-7 MHz) one.

Scanning

For this more proximal approach we place the transducer parallel and

immediately below to the midpoint of the clavicle, as shown in figure 6-35.

The image obtained at this proximal level is a cross section of the neurovascular

bundle as it aligns under the clavicle in a formation that has the axillary vein as the most

medial structure, followed by the axillary artery in the center and the divisions of the

plexus most laterally, as shown in figure 6-36.

Needle insertion

The needle can be advanced out of plane from caudal to cephalad, but we usually

prefer an in plane technique from lateral to medial, as shown in figure 6-37.

Fig 6-35. Proximal scanning, left

side. The transducer is placed

parallel to the midpoint of the

clavicle and immediately below it.

(On a model with permission).

Fig 6-36. Proximal scanning, left side. At this

proximal level pectoralis major (Pec major) is the

main muscle seen superficial to the

neurovascular bundle. Pectoralis minor is located

distally to this US section. Among the

neurovascular bundle structures the axillary vein

(AV) is the most medial, followed by the axillary

artery (AA) and then the divisions of the plexus

most laterally. (Author’s archive).

Page 102: Manual Regional Carlo 2010

102 | P a g e

Figures 6-38 A, B and C, show a sequence of ultrasound images showing needle

insertion and injection.

CORACOID INFRACLAVICULAR TECHNIQUE

This technique is performed around the coracoid, but as opposed to the nerve

stimulation technique the level is not dictated by a fixed measurement with respect to the

coracoid, but instead by an optimal ultrasound image of the axillary artery and the

surrounding cords.

Type of transducer

A linear high frequency (8-15 MHz) or a curved low frequency (3-7 MHz) probe is used

depending on the thickness of the patient’s thoracic wall.

Scanning

For this more distal approach we place the transducer in an oblique fashion in the mid

pectoral region, as shown in figure 6-39. This probe rotation is needed to get a better

cross section of the neurovascular bundle, which is traveling diagonally in the

infraclavicular region.

Fig 6-37. Needle insertion, left side.

The needle is introduced in plane from

lateral to medial. (On a model with

permission).

Figure 6-38; A, B and C. Needle insertion/injection, left side. A (left): the divisions of the plexus

are shown surrounded by a fascial sheath lateral to the axillary artery (AA); B (center): the shadow of

the needle path (pointed by arrows) is barely seen as the needle comes in at a 45 degree angle. The two

smaller arrow heads point to the indentation of the fascia produced by the piercing needle; C: the

spread of local anesthetic is seen as a hypoechoic shadow pointed by a large arrow and the resulting

expanded sheath is shown with the smaller arrows. (Author’s archive)

Page 103: Manual Regional Carlo 2010

103 | P a g e

The ultrasound image obtained at this level is shown in figure 6-40. At this level

the cords of the plexus have already rotated behind the axillary artery and adopted their

arrangement around it from which they take their names, medial, posterior and lateral.

Needle insertion

As it is the case with the more proximal approach, the needle can be inserted out

of plane, from caudal to cephalad, but we usually prefer to advance it in plane, from

lateral to medial (superior to inferior), as shown in figure 6-41.

Clinical pearls

The proximal infraclavicular approach is a block of the divisions of the plexus

and as such it can resemble a supraclavicular block in onset and density of

anesthesia.

Fig 6-40. Coracoid level scanning, right

side. With the probe at the level of the

coracoid process the neurovascular bundle

appears under both pectoralis muscles. The

axillary vein (V) is more medial, close to

the chest wall, while the axillary artery (A)

is more lateral, surrounded by the three

cords, lateral (L), posterior (P) and medial

(M). (Author’s archive).

Fig 6-41. Needle insertion, right

side. The needle is introduced in

plane, from lateral (superior) to

medial (inferior). (On a model with

permission).

Fig 6-39. Coracoid level scanning, right

side. The transducer is placed in an oblique

fashion to get a perpendicular cut of the

neurovascular bundle at the level of the

coracoid process. (On a model with

permission).

Page 104: Manual Regional Carlo 2010

104 | P a g e

AXILLARY BLOCK

NERVE STIMULATOR TECHNIQUE

Indications

It is best suited for anesthesia/analgesia of the upper extremity distal to the elbow.

Point of contact of the needle with the brachial plexus

The needle approaches the plexus at the level of its terminal branches.

Main characteristics The axillary block is not properly a brachial plexus block, but rather a block of its

terminal branches. The larger surface area that the branches as a whole occupy and the

tendency for the local anesthetic to follow the paths of low resistance along individual

nerves affect the circumferential spread of the local anesthetic within the sheath (please

see discussion on axillary brachial plexus sheath in the anatomy section). A single

injection technique is an option, but multiple injections have shown to increase the

success rate at this level. If a single injection is to be attempted, the operator needs to

specifically target the nerve feeding the surgical area. If the surgical area involves more

than one terminal nerve, the single injection technique should be performed in the

proximity of the radial nerve because, as mentioned in the anatomy discussion, the local

anesthetic solution tends to spread inside the sheath more easily from back to front that

vice versa. In addition, my observations in the anatomy lab show that better

circumferential spread of local anesthetic may be obtained with a slight elevation of the

elbow, because this maneuver releases some of the stretching of the neurovascular

bundle.

Some authors advice to perform the block high in the axilla to improve its overall

success. This can be uncomfortable to the patient and challenging to the anesthesiologist.

The only perceived advantage would be to increase the chances of blocking the

musculocutaneous nerve before it leaves the sheath, but since its take off is variable the

operator could never be certain. I believe that a better strategy is to start the axillary block

by first blocking the musculocutaneous nerve in the proximal arm and then complete the

block according to what is needed.

Although some variability exists, usually the median nerve is superficial (anterior)

to the axillary artery, following its same direction; the ulnar nerve (and medial

brachial/antebrachial cutaneous nerves) are medial and somewhat posterior to the artery;

the musculocutaneous nerve is lateral to the artery (and eventually under the biceps

muscle); and the radial nerve is posterior to the artery.

I believe that in the 21st century, with the variety of tools at our disposal, there is

no good reason to perform a trans axillary technique.

Patient position and landmarks

The patient is supine, the arm is abducted to about 80-90 degrees and the elbow is

slightly elevated 20-30 degrees by using a small pillow or folded blanket.

The biceps muscle is identified by visualization and/or palpation. The

coracobrachialis muscle is found immediately under it (posterior). While biceps is highly

mobile the coracobrachialis is palpated as a thick poorly movable mass. The pulsation of

Page 105: Manual Regional Carlo 2010

105 | P a g e

the axillary artery is found immediately under the coracobrachialis. Sometimes it helps to

displace the latter slightly anterior to feel the pulsation of the artery. Figure 6-42 shows

the arm position in abduction with a small pillow under the elbow and the trajectory of

the axillary artery marked in blue.

Type of needle This is usually a superficial block, even in obese patients. A 5cm, 22-G, insulated

needle usually suffices.

Single injection axillary block

As I mentioned before, evidence shows that success rate in the axillary region

increases with 2 and 3 injection techniques as opposed to a single injection. If a single

injection technique is employed the “epicenter” of the injection should occur at the nerve

that is more relevant to the surgical site. If more than one nerve is involved in the

innervation of the surgical site, the single injection technique should be performed

preferably in front of the radial nerve. The volume of local anesthetic needed for a single

injection technique is 40 to 50 mL. If more than one injection is performed the volume

should be divided accordingly. If only one nerve is needed 5 mL of local anesthetic

solution is enough for anesthesia. A solution of 1.5% mepivacaine plus 1:400,000

epinephrine provides 3-4 hr of anesthesia. If longer anesthesia is desired 0.5%

ropivacaine with epinephrine provides 12 hr plus of anesthesia. For analgesia 0.2%

ropivacaine is adequate.

In order to perform a targeted injection of a specific nerve in the axilla it is

necessary to know how to block each individual nerve. The following is a description of

each technique.

MUSCULOCUTANEOUS NERVE BLOCK

The musculocutaneous nerve originates from the lateral cord (it can take off from

the median nerve already in the arm) and because of its uncertain take off level we like to

block it first.

Nerve stimulator setting

The nerve stimulator is set to deliver a current of 0.8-09 mA, pulse frequency of 1

Hz and pulse duration of 0.1 msec (100 microsec). A small skin wheal is raised with 1%

lidocaine or 1% mepivacaine using a small needle, ideally 27-G.

Fig 6-42. Patient position and

axillary artery marking. The arm is

abducted about 80° to 90°, the elbow is

elevated slightly with a small pillow

and the axillary artery is marked. (On a

model with permission).

Page 106: Manual Regional Carlo 2010

106 | P a g e

Needle insertion

The operator identifies and holds the patient’s biceps muscle with one hand and

directs the needle with the other in a direction perpendicular to the main axis of the arm,

advancing it between biceps and coracobrachialis, as shown in figure 6-43.

Type of response

As the needle approaches the musculocutaneous nerve a motor twitch of biceps

with flexion of the elbow is obtained. The current is reduced to 0.5 mA and, if a response

is still visible at this level, the injection is started.

MEDIAN NERVE BLOCK

The median nerve is most frequently located anterior (superficial) to the axillary

artery running in the same direction, making it a very superficial block.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 0.8-09 mA, pulse frequency of 1

Hz and pulse duration of 0.1 msec (100 microsec). A small skin wheal is raised with 1%

lidocaine or 1% mepivacaine using a small needle, ideally 27-G.

Needle insertion

Using the mark of the axillary artery on the skin as a reference, the needle is

introduced very tangential to the skin (shallow angle), in the same direction of the artery,

as shown in figure 6-44.

Fig 6-43. Blocking the musculo

cutaneous nerve. The needle is

introduced under the biceps

perpendicular to the main axis of the

arm. (On a model with permission).

Fig 6-44. Median nerve block. The

needle is introduced in reference to the

axillary artery with a very shallow angle

and in the same direction than the artery.

(On a model with permission).

Page 107: Manual Regional Carlo 2010

107 | P a g e

It is better to mark the course of the artery on the skin than to keep the fingers on

the pulse to avoid bringing the artery even closer to the skin and increasing the chances

for accidental artery puncture.

ULNAR NERVE BLOCK

The ulnar nerve is located immediately medial to the artery, slightly deeper than

the median nerve. It gives sensory innervation to the medial side of the hand. Because the

medial brachial and the medial antebrachial cutaneous nerves run along with the ulnar

nerve on the medial side of the axillary artery, the ulnar nerve technique is performed for

anesthesia of the medial arm and medial forearm.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 0.8-09 mA, pulse frequency of 1

Hz and pulse duration of 0.1 msec (100 microsec).

Needle insertion

Using the mark of the axillary artery on the skin as a reference, the needle is

directed slightly medial to the artery, as shown in figure 6-45.

RADIAL NERVE BLOCK

The radial nerve is most frequently located posterior (deeper) to the axillary

artery. It is the largest of the terminal branches of the plexus.

Nerve stimulator setting

The nerve stimulator is set to deliver a current of 0.8-09 mA, pulse frequency of 1

Hz and pulse duration of 0.1 msec (100 microsec).

Needle insertion

The operator uses two fingers of one hand as “hooks” to slightly displace the

artery out of the way in order to reach the radial nerve located posterior to it. The needle

is inserted posterior with a 30 degree cephalad orientation, as shown in figure 6-46.

Fig 6-45. Ulnar nerve block. The needle

is introduced slightly medial to the line

representing the axillary artery. Notice the

small difference in the angle of insertion

compared to the median nerve block. (On

a model with permission).

Page 108: Manual Regional Carlo 2010

108 | P a g e

Complications

Pneumothorax is virtually impossible to get from this location. Hematomas from

vascular puncture are more common and can be associated with nerve damage.

Pearls

This is a block mainly indicated for surgery on the distal forearm, wrist and hand.

It is not a good choice for elbow surgery.

Tourniquet pain is an issue and not necessarily due to intercostobrachial nerve,

but mainly due to insufficient proximal anesthesia of the deeper planes of the arm.

Two and three injection techniques have proven more successful, but if a single

injection is preferred this injection should be in front of the nerve most

responsible for the sensory innervation of the surgical site. If more than one nerve

is involved the injection should be performed in front of the radial nerve.

Fig 6-46. Radial nerve block. The axillary

artery is displaced towards the biceps to

gain entrance to its posterior aspect. The

needle is then introduced in reference to the

mark on the skin with a 30 degree cephalad

orientation. (On a model with permission).

Page 109: Manual Regional Carlo 2010

109 | P a g e

AXILLARY BLOCK

ULTRASOUND TECHNIQUE

Indications

The same indications mentioned for the nerve stimulation technique.

Patient position

The patient is semi seated with the arm in abduction and the elbow flexed, as

shown in figure 6-47.

Type of needle

This is a superficial block for which a 5cm, 22-G, insulated needle suffices.

Type of transducer

We use a high frequency (8-15 MHz) linear probe.

Scanning

The probe is placed across the neurovascular bundle in the proximal part of the

arm, as shown in figure 6-48.

At this level the neurovascular bundle of the axilla is usually very superficial and

the terminal nerves can be seen surrounding the axillary artery. The median nerve is

usually superficial (anterior) to the artery, the ulnar nerve is medial and somewhat

posterior, and the radial nerve is posterior, as shown in figure 6-49.

Fig 6-47. Patient position. The

patient is semi seated, with the arm

abducted and the elbow flexed. (On a

model with permission).

Fig 6-48. Scanning. The probe is

place perpendicular to the main axis

of the neurovascular bundle. (On a

model with permission).

Page 110: Manual Regional Carlo 2010

110 | P a g e

Distally in the axilla the radial nerve starts shifting more lateral, but it still

remains posterior to the artery. The musculocutaneous is lateral to the artery at all times

and it can be traced from its origin in the lateral cord proximally to its location between

biceps and coracobrachialis distally. If a single injection is planned it should be made in

the proximity of the radial nerve. Individual injections of terminal nerves can be done as

needed. An image of the neurovascular bundle of the axilla in cross section is shown in

figure 6-50.

Needle insertion

The needle is advanced in plane from lateral to medial, as shown in figure 6-51.

Fig 6-49. Terminal branches. The

axillary sheath has been removed to

show the relative location of the

nerves with respect to the axillary

artery. MACN: medial antebrachial

cutaneous nerve; axi: axillary

nerve. Cadaver dissection by Dr

Franco. Image is copyrighted.

Fig 6-50. Axillary scanning. With the

probe across the axilla the axillary

artery (AA) is seen surrounded by

three main nerves, median (M), Ulnar

(U) and Radial (R). Also seen is

Musculocutaneous nerve (MC),

axillary vein (AV) and some muscles.

(Author’s archive).

Fig 6-51. Needle insertion. The

needle is advanced in plane from

lateral (superior) to medial

(inferior) and aimed toward the

desired nerve. (On a model with

permission).

Page 111: Manual Regional Carlo 2010

111 | P a g e

We usually block first the musculocutaneous nerve located in between biceps and

coracobrachialis. To target this nerve the needle needs to be inserted at an angle of 30-45

degrees. Then the rest of the terminal branches are targeted as needed. These branches are

more superficial so they need a much smaller angle of insertion, which facilitates needle

visualization.

Local anesthetic and volume Because the nerves can be targeted individually it is possible to inject about 5 mL

of local anesthetic solution per nerve until all the needed nerves are completely

surrounded by it, usually requiring a total volume of 20-30 mL. For anesthesia we use

1.5% mepivacaine plus 1:400,000 epinephrine, which gives 3-4 hours of surgical

anesthesia. For more prolonged anesthesia 0.5% ropivacaine with epinephrine can be

used. For postoperative analgesia we recommend 0.2% ropivacaine.

Side effects and complications

The most common complication at the axillary level is hematoma at the site, but

ultrasound provides a good visualization of vessels and nerves at this location making

this occurrence more rare.

Page 112: Manual Regional Carlo 2010

112 | P a g e

References

1. Brown DL. Brachial plexus anesthesia: an analysis of options. Yale J Biol Med

1993; 66: 415-431

2. Winnie AP. Interscalene brachial plexus block. Anesth Analg 1970; 49: 455-466

3. Kulenkampff D, Persky MA. Brachial plexus anesthesia. Ann Surg 1928; 87: 883-

891

4. Winnie AP, Collins VJ. The subclavian perivascular technique of brachial plexus

anesthesia. Anesthesiology 1964; 25: 353-363

5. Franco CD, Vieira Z. 1,001 subclavian perivascular brachial plexus blocks:

success with a nerve stimulator. Reg Anesth Pain Med 2000; 25: 41-46

6. Franco CD. The subclavian perivascular block. Tech Reg Anesth Pain Med 1999;

3: 212-216

7. De Andres J, Sala-Blanch X. Peripheral nerve stimulation in the practice of

brachial plexus anesthesia: a review. Reg Anesth Pain Med 2001; 26: 478-483

8. Greenblatt Gm, Denson GS. Needle nerve stimulator-locator: nerve blocks with a

new instrument for locating nerves. Anesth Analg 1962; 41: 599-602

9. Hadzic A, Vloka J, Hadzic N, et al. Nerve stimulators used for peripheral nerve

blocks vary in their electrical characteristics. Anesthesiology 2003; 98: 969-974

10. Passannante AN. Spinal anesthesia and permanent neurologic deficit after

interscalene block. Anesth Analg 1996; 82: 873-874

11. Urmey WF, Grossi P, Sharrock NE, Stanton J, Gloeggler PJ. Digital pressure

during interscalene block is clinically ineffective in preventing anesthetic spread

to the cervical plexus. Anesth Analg 1996; 83: 366-370

12. Silverstein WB, Saiyed M, Brown AR. Interscalene block with a nerve stimulator:

A deltoid motor response is a satisfactory endpoint for successful block. Reg

Anesth pain Med 2000; 25: 356-359

13. Urmey WF, Talts KH, Sharrock NE. One hundred percent incidence of

hemidiaphragmatic paresis associated with interscalene brachial plexus anesthesia

as diagnosed by ultrasonography. Anesth Analg 1991; 72: 498-503

14. Urmey WF. Interscalene block: The truth about twitches (editorial). Reg Anesth

pain Med 2000; 25: 340-342

15. Brand L, Papper EM. A comparison of supraclavicular and axillary techniques for

brachial plexus blocks. Anesthesiology 1961; 22: 226-229

16. Brown DL. Atlas of regional anesthesia. Philadelphia, PA: W.B. Saunders, 1992

17. Mulroy MF. Regional anesthesia: An illustrated procedural guide. 3rd

edition.

Philadelphia, PA; Lippincott Williams & Wilkins 2002

18. Urmey WF, Stanton J. Inability to consistently elicit a motor response following

sensory paresthesia during interscalene block administration. Anesthesiology

2002; 96: 552-554

19. Neal JM, Moore JM, Kopacz DJ, Liu SS, Kramer DJ, Plorde JJ. Quantitative

analysis of respiratory, motor, and sensory function after supraclavicular block.

Anesth Analg 1998; 86: 1239-1244

20. Franco CD, Domashevich V, Voronov G, Rafizad A, Jelev T. The supraclavicular

block with a nerve stimulator: To decrease or not to decrease, that is the question.

Anesth Analg 2004; 98: 1167-1171

Page 113: Manual Regional Carlo 2010

113 | P a g e

21. Franco CD, Gloss FJ, Voronov G, Tyler SG, Stojiljkovic LS. Supraclavicular

block in the obese population: An analysis of 2020 blocks. Anesth Analg 2006;

102: 1252-1254

22. Perlas A, Chan V: Ultrasound-assisted nerve blocks. In: Textbook of Regional

Anesthesia, Hadzic A (ed). New York, McGraw Hill, 2007, pp 663-672

23. Franco CD, et al. Gross anatomy of the brachial plexus sheath in human cadavers.

Reg Anesth Pain Med 2008; 33: 64-69

24. Neal JM, Gerancher JC, Hebl JR, Ilfeld BM, McCartney CJL, Franco CD, Hogan

QH. Upper Extremity Regional Anesthesia: Essentials of Our Current

Understanding. Reg Anesth Pain Med 2009; 34: 134-170

Page 114: Manual Regional Carlo 2010

114 | P a g e

CHAPTER 7

LOWER EXTREMITY NERVE BLOCKS

Page 115: Manual Regional Carlo 2010

115 | P a g e

LOWER EXTREMITY BLOCKS

The innervation of the lower extremity comes from the lumbar and sacral

plexuses. The different nerve elements of the lower extremity run more distant from each

other than those of the upper extremity, without having a point of convergence like the

one at the level of the brachial plexus trunks. Therefore, no single peripheral block

technique is able to provide anesthesia of the whole lower extremity. This anatomical

fact, combined with the high success of neuraxial anesthesia, has traditionally affected

the popularity of lower extremity peripheral nerve blocks.

The introduction of low molecular weight heparin (LMWH) in the United States

in the early 1990s produced a renewed interest in lower extremity nerve blocks because

of the increased risk of epidural hematoma after neuraxial anesthesia in patients receiving

LMWH. The use of ultrasound in regional anesthesia has also been a major reason for the

increased popularity of all sort of peripheral nerve blocks.

Anatomy

Lateral femoral cutaneous nerve

It is an exclusively sensory nerve originating from the ventral rami of spinal

nerves L2-L3. It appears in the pelvis, lateral to the psoas muscle, caudal to the

ilioinguinal nerve. It runs anteriorly under the iliac fascia, parallel to the iliac crest. It

emerges from the pelvis, under the inguinal ligament, between the anterior superior and

anterior inferior iliac spines, as shown in figure 7-1 and 7-2. It provides sensory

innervation to the lateral thigh.

Femoral nerve

It is a motor and sensory nerve derived from the posterior divisions of the ventral

rami of spinal nerves L2-L3-L4. In the pelvis it is also located lateral to the psoas muscle,

in the cleavage between psoas and iliacus muscles. As it passes under the inguinal

ligament the nerve is superficial to the combined iliopsoas muscle. Under the inguinal

ligament the femoral nerve has the femoral artery medial to it, while the femoral vein is

located medial to the artery (VAN from medial to lateral), as shown in figure 7-2.

Fig 7-1. Lateral femoral cutaneous nerve

(LFCN), left side. The LFCN shown with

arrows perforates the fascia lata below the

inguinal ligament to become a superficial

nerve. Cadaver dissection by Dr Franco.

Image is copyrighted.

Page 116: Manual Regional Carlo 2010

116 | P a g e

Approximately 3-4 cm below the inguinal ligament, the femoral nerve divides

into anterior and posterior divisions. The anterior division has two sensory branches that

supply the antero medial thigh, and two muscular branches that supply the sartorius and

pectineus muscles. The posterior division has one sensory branch, the saphenous nerve,

and muscular branches to the quadriceps. The nerve is covered by the iliac fascia, which

separates it from the main vessels, and more superficially by the fascia lata, the deep

fascia of the thigh.

The muscular branch to the rectus femoris also supplies the hip joint while the

muscular branches to the three vasti muscles also supply the knee joint.

Obturator nerve

It is usually a mixed nerve (motor and sensory) derived from the anterior

divisions of the ventral rami of spinal nerves L2-L3-L4. It emerges on the medial side of

the psoas muscle just above the pelvic brim running down between this muscle and the

lumbar spine. As the nerve enters the pelvis it turns laterally to run along its lateral wall

until it reaches the obturator foramen, through which it reaches the thigh. In the thigh the

nerve divides into anterior and posterior branches, as shown in figure 7-3.

The anterior division runs caudally, first located between the pectineus muscle in

front and the obturator externus behind. A few cm distally the nerve runs between the

Fig 7-2. Femoral nerve, left side. The

femoral nerve (FN) passes under the

inguinal ligament lateral to the femoral

artery (A). Also shown are the femoral

vein (V) and the LFCN pointed with

arrows. Cadaver dissection by Dr

Franco. Image is copyrighted.

Fig 7-3. Obturator nerve. The

obturator nerve (OB) comes out of

the obturator foramen where its

two branches eventually straddle

the adductor brevis muscle (AB).

Also shown are femoral nerve

(FN), femoral artery (FA), femoral

vein (FV), pectineus (Pec),

obturator externus (OE) and

adductor longus (AL).Cadaver

dissection by Dr Franco. Image is

copyrighted.

Page 117: Manual Regional Carlo 2010

117 | P a g e

adductor longus anteriorly and the adductor brevis posteriorly. It gives innervation to the

gracilis, adductor brevis and adductor longus, and sometimes to the pectineus. It gives

also articular branches to the hip joint. On occasions it supplies the skin of the medial

side of the thigh.

The posterior division after a short trajectory it usually pierces the obturator

externus to then run caudally between the adductor brevis in front and the adductor

magnus behind. It supplies the obturator externus, the adductor magnus and the knee

joint. The anterior sensory branch can be frequently missing and in that case the medial

thigh is also supplied by the femoral nerve.

The highly variable distribution of the cutaneous branch of the obturator nerve has

contributed to the confusion about how much anesthesia can be obtained from a single

block performed at the femoral level (“3-in-1” block). Most of the studies have used

pinprick testing of the medial, anterior and lateral thigh to assess anesthesia of obturator,

femoral and lateral femoral cutaneous nerves territories. This testing does not take into

account the fact that many variations exist in the innervation patterns of the thigh

including the absence of a cutaneous branch of obturator nerve. Nevertheless many

authors believe that a block at the femoral level could also produce anesthesia of the

lateral femoral cutaneous nerve by lateral diffusion of the local anesthetic under the

fascia iliaca (“2-in-1 block”). Spread of local anesthetic to the obturator nerve either,

medially under the vessels or proximally toward the pelvis is more unlikely.

Sciatic nerve

It is the largest nerve in the body. It originates from the ventral rami of spinal

nerves L4-L5, S1-S3. Part of the anterior ramus of L4 joins the anterior ramus of L5 to

originate the lumbosacral trunk, which together with the first three sacral roots form the

sciatic nerve. The nerve has two components, the tibial nerve (on its medial side), which

is derived from the anterior divisions of the ventral rami of L4-L5, S1-S3 and the

common peroneal nerve (on its lateral side), which is derived from the posterior divisions

of the ventral rami of L4-L5, S1-S2. The nerve comes out of the pelvis through the

greater sciatic foramen, entering the gluteal region anterior (deep as seen from the gluteal

region) to the piriformis muscle. The nerve curves above the ischial tuberosity and then

turns vertically downwards to run between the ischium medially and the greater

trochanter laterally, as shown in figure 7-4.

Fig 7-4. The sciatic nerve (1) enters the

gluteal region covered superficially by the

piriformis muscle (2). It then travels parallel

to the midline (5), between the ischium (3)

and greater trochanter (4). Cadaver dissection

by Dr Franco. Image is copyrighted.

Page 118: Manual Regional Carlo 2010

118 | P a g e

For most of its trajectory in the buttocks, the sciatic nerve runs parallel to the

midline, at a distance of about 10 cm in adult patients. With the hips in adduction this

distance is maintained throughout adult life, not being influenced by gender or body

weight. This previously unknown fact has simplified enormously the approach to the

sciatic nerve in our practice (for more information see Franco. Anesthesiology 2003; 98:

723-728).

The tibial and common peroneal components can be easily identified as two

separate nerves during their entire trajectory in about 11% of the cases. However, even in

those cases the two components are surrounded by a common sheath of connective tissue,

as shown in figure 7-5. Therefore, it is important not to confuse this with a true separation

of the components, which invariably takes place always in the popliteal fossa.

The sciatic nerve enters the thigh deep to the biceps femoris muscle and not

lateral to it as usually mentioned in our literature, as shown in figure 7-6.

As opposed to what happens in the gluteal region, the position of the sciatic nerve

in the thigh with respect to the midline is influenced both by the degree of hip abduction

as well as by the amount of fat accumulating in the inner thigh.

Fig 7-5. The sciatic nerve components, tibial (T) and

common peroneal (CP) share a

common sheath. Cadaver

dissection by Dr Franco. Image

is copyrighted.

Fig 7-6. The sciatic nerve (SN) enters the thigh under

the cover of gluteus maximus

(sectioned) and biceps,

which is shown split between

two arrows. Cadaver

dissection by Dr Franco.

Image is copyrighted.

Page 119: Manual Regional Carlo 2010

119 | P a g e

The nerve runs in the posterior thigh under the cover of the hamstring muscles,

until it reaches the popliteal fossa. Upon entering the popliteal fossa, the two nerve

components, peroneal and tibial, finally diverge from each other, having never mixed

their fibers. The posterior tibial nerve continues to run in the direction of the main trunk,

at the center of the fossa. The common peroneal component turns laterally to run just

medial to the biceps tendon, as shown in figure 7-7.

Subgluteal fold

The fold that defines the buttocks inferiorly is a fold of the skin and does not

correspond with the lower border of the gluteus maximus muscle, as frequently thought.

In fact the inferior border of this muscle crosses the subgluteal fold diagonally as it runs

laterally to insert in the iliotibial tract, as shown in figure 7-8. Therefore, during a

subgluteal approach to the sciatic nerve, the needle crosses the same planes (fat and

gluteus maximus) than in more proximal approaches, although the fat layer can be

thinner.

Fig 7-8. The subgluteal fold. The

inferior border of the gluteus

maximus and subgluteal fold are

two different things. They cross

each other diagonally. Cadaver

dissection by Dr Franco. Image is

copyrighted.

Fig 7-7. The sciatic nerve division

after soft tissue removal. The

sciatic nerve (SN) divides into its

two components, tibial (TN) and

common peroneal (CP), in the

popliteal fossa. Also shown are the

popliteal vein (PV), popliteal artery

(PA) and muscles including

semimembranosus (SM). Cadaver

dissection by Dr Franco. Image is

copyrighted.

Page 120: Manual Regional Carlo 2010

120 | P a g e

Genitofemoral nerve

It derives from the ventral rami of spinal nerves L1-L2. Its genital branch

provides some of the innervation of the genital area, while its femoral component

provides sensory innervation of the medial upper thigh and the skin over the femoral

vessels.

Posterior cutaneous nerve of the thigh

It is also known as posterior femoral cutaneous nerve. It originates from the

ventral rami of spinal nerves S1-S3. It exits the pelvis through the greater sciatic foramen,

first medial and then superficial to the sciatic nerve. Somewhere caudal to the ischium,

the nerve pierces the deep fascia (fascia lata) and becomes a superficial structure. It is not

a branch of the sciatic nerve, although it has a close relationship with it in the gluteal

area, as shown in figures 7-9 and 7-10, before it becomes a superficial nerve as shown in

figure 7-11.

Fig 7-9. The posterior femoral cutaneous nerve shown with arrows is in close contact to the sciatic

nerve (SN) in the gluteal area. The sheath of the

nerve is partially intact. Cadaver dissection by Dr

Franco. Image is copyrighted.

Fig 7-10. The posterior femoral

cutaneous nerve shown with arrows and

sciatic nerve (SN) after removal of

connective tissue. Cadaver dissection by

Dr Franco. Image is copyrighted.

Fig 7-11. The posterior femoral

cutaneous nerve becomes a

superficial nerve around the

subgluteal fold, where it is separated

from the sciatic nerve which at this

point is running under the hamstring

muscles and fascia lata. Cadaver

dissection by Dr Franco. Image is

copyrighted.

Page 121: Manual Regional Carlo 2010

121 | P a g e

The posterior femoral cutaneous nerve innervates the lower part of the buttocks as

well as the posterior thigh, frequently reaching as far down as the proximal posterior

aspect of the leg. A block of the sciatic nerve performed in the gluteal area will

predictably produce anesthesia of this cutaneous nerve as well. A block performed at the

subgluteal level on the other hand, will not reliably block it.

Saphenous nerve

It is a sensory nerve that originates from the posterior division of the femoral

nerve (L3-L4) in the inguinal region. It is the largest cutaneous branch of the femoral

nerve. It runs down the femoral canal along with the femoral vessels, under the cover of

the sartorius muscle. It emerges on the medial side of the knee between the tendons of

sartorius and gracilis, as shown in figure 7-12

At a variable distance caudal to the knee, it pierces the deep fascia to become

superficial. Distal to the knee it gives off the subpatellar branch, which supplies the

medial side of the knee (chance for injury during knee arthroscopy). Once it becomes

superficial, it runs alongside the greater saphenous vein in the leg, passing in front of the

medial malleolus in the ankle, before terminating around the base of the first metatarsal

on the medial side of the foot.

Male and female pelvis issue

The female pelvis is adapted to accommodate child bearing and as a result the

pelvic cavity or inner pelvis is wider in females than in males. However, the total width

of the bony pelvis, that is the diameter between both iliac crests (bicrestal diameter), is

similar in both sexes, measuring an average of 280 mm in males and 275 mm in females

(see Cunningham’s Anatomy reference). The thicker bones in the male pelvis compensate

for a “roomier” female pelvis (see Hollinshead’s Anatomy reference). According to some

anthropologists (Hall et al reference) the human bony pelvis is “surprisingly” similar in

males and females at all ages. The perceived difference in pelvis size corresponds to

hormone-dependent, different patterns of fat deposition in both sexes. In other words the

difference in pelvic size among the sexes is mostly due to soft tissue and not due to

differences in the total width of the bony pelvis. It is the latter what determines the

position of the sciatic nerve in the buttocks as I will discuss further when I describe the

gluteal approaches to the sciatic nerve.

Fig 7-12. The saphenous nerve

(SN) emerges on the medial side of

the knee between sartorious and

gracilis to run along the greater

saphenous vein on the medial side

of the leg. ATT: anterior tibial

tuberosity. Cadaver dissection by

Dr Franco. Image is copyrighted.

Page 122: Manual Regional Carlo 2010

122 | P a g e

Clinical pearls

The nerves of the lower extremity are more distant from each other than in the

upper extremity so it is not possible to block the entire lower extremity from a

single injection point.

The position of the sciatic nerve in the buttocks with respect to the midline is

dictated by the bony pelvis and as such it is not affected by gender or obesity.

Its relationship to bone structures and to the midline remains unchanged

throughout adulthood.

The inferior border of the gluteus maximus muscle does not correspond with

the subgluteal fold (Snell’s Clinical Anatomy for Medical Students, 3rd

edition, page 554). In fact both cross each other diagonally. The subgluteal

fold is a fold of the skin anchored to the deep fascia.

The gluteus maximus is the only gluteal muscle to cover the sciatic nerve

superficially, caudal to the piriformis muscle in the gluteal region. Gluteus

medius and minimus are located cephalad and lateral to the sciatic nerve.

The inguinal crease does not correspond with the inguinal ligament. Both

structures are parallel to each other. The inguinal crease runs about 1 inch (2.5

cm) caudal and parallel to the inguinal ligament.

Page 123: Manual Regional Carlo 2010

123 | P a g e

LATERAL FEMORAL CUTANEOUS NERVE BLOCK

Indications

This block can be performed alone to provide anesthesia of the lateral thigh (e.g.,

donor area for a skin graft). It can also be performed along with femoral, obturator and

sciatic blocks to provide anesthesia of the thigh for surgical procedures above the knee

and for thigh tourniquet. It is also one of the nerves targeted in a “3-in-1” block, a block

of the femoral nerve performed with a higher volume of local anesthetic to try to block

also the lateral femoral and obturator nerves (not supported by the evidence).

Point of contact with the nerve

The nerve is approached as it emerges from under the inguinal ligament, medial

and inferior to the anterior superior iliac spine (ASIS).

Main characteristics

This can be a superficial block (above the fascia lata) if the block is performed at

2 or more cm distal to the inguinal ligament. More proximally the nerve is under the

fascia lata. This is important because this fascia is thick enough to slow the transfer of

local anesthetic to the target nerve.

ANATOMICAL TECHNIQUE

Patient position and landmarks

The patient lies supine. The ASIS is identified by palpation.

Technique The needle entrance point is identified about 1 cm medial and 1 cm caudal to the

ASIS. The needle is advanced perpendicular to the skin and directed deep to the fascia

lata where the local anesthetic is injected in a fanwise fashion. A nerve stimulator with

pulse duration of 0.3 to 1 msec (300 to 1000 microsec) can be used to elicit a sensory

paresthesia in the lateral thigh.

Local anesthetic and volume

A volume of 5 to 10 mL of 1% mepivacaine is frequently used. A long acting

agent, as ropivacaine, can be used if necessary.

Complications

Very rare. Some patients can complain of dysesthesia in the lateral thigh that

usually goes away without sequelae.

ULTRASOUND TECHNIQUE

The use of ultrasound facilitates this block. As the lateral femoral cutaneous nerve

passes under the inguinal ligament it is located under the fascia lata in between tensor

fascia lata laterally and sartorius medially. Placing the probe across the gap in between

these two muscles usually allows a good visualization. A few centimeters distal to the

inguinal ligament the nerve can be located superficial to the sartorius muscle.

Page 124: Manual Regional Carlo 2010

124 | P a g e

FEMORAL NERVE BLOCK

NERVE STIMULATOR TECHNIQUE

Indications

An isolated femoral nerve block can be performed to provide anesthesia for

surgery on the anterior thigh, patella and some knee procedures. It is more commonly

performed along with sciatic to provide anesthesia of the entire lower extremity.

Point of contact with the nerve

The nerve is usually approached just below the inguinal crease. However, if

possible, the nerve can be approached immediately above the crease (1 cm), where it is

more compacted, before its branches start to diverge.

Main characteristics

This is a simple block performed lateral to the pulse of the femoral artery, deep to

the fascia lata (deep fascia of the thigh) and deep to the fascia iliaca (the fascia that

covers the iliopsoas muscle). The femoral artery pulse usually provides an easy and

reliable landmark to the nerve.

Patient position and landmarks

The patient lies supine. If necessary, the back of the bed can be slightly elevated

for patient’s comfort. If done in combination with a sciatic nerve block, we prefer to do

the sciatic block first because this block has a longer onset time than the femoral. The

femoral pulse at the inguinal crease is found by palpation. The point of entrance is

marked on the skin, proximal or distal to the inguinal crease, about 2 cm lateral to the

pulse of the femoral artery, as shown in figure 7-13.

Type of needle

A 5 cm, 22G, insulated needle usually suffices.

Nerve stimulator settings

The nerve stimulator is set to deliver a 1.0 mA current, at a frequency of 1 Hz and

pulse duration of 0.1 msec (100 microsec).

Fig 7-13. Patient position and landmarks. The

patient is supine or semi seated. The location of the

femoral artery (A) is found by palpation. The femoral

nerve (N) is lateral to the artery, while the femoral vein

(V) is medial to it. Also shown is the anterior superior

iliac spine (ASIS). (On a model with permission).

Page 125: Manual Regional Carlo 2010

125 | P a g e

Needle insertion

The needle is inserted 1-2 cm lateral to the pulsation of the femoral artery with a

30-45-degree cephalad orientation, as shown in figure 7-14 A and B.

The needle is advanced parallel to the midline in the direction of the inguinal

ligament. A twitch of the quadriceps muscle with movement of the patella is a good

response. The current is lowered and with a muscle twitch still visible at 0.5 mA a slow

injection is started. A response from the sartorius is usually considered not a good

response, because it could be the result of stimulation of the nerve to the sartorius, a

branch of the anterior division of the femoral nerve. If the block is performed 1 cm above

the inguinal crease, where the nerve has not branched off yet, a twitch from the sartorius

is equally acceptable.

Local anesthetic and volume

The femoral nerve is a collection of branches flat in the frontal plane that offers a

large area of absorption. For anesthesia we usually use 10-20 mL of 1.5% mepivacaine

plus 1:400,000 epinephrine for 3-4 hours of surgical anesthesia. For longer anesthesia

0.5% ropivacaine with epinephrine can be used. For analgesia we usually use 10-15 mL

of 0.2% ropivacaine. We always use epinephrine 1:400,000 as an intravascular marker.

Side effects and complications

Blocks at the femoral level are usually well tolerated and complications are rare.

Fig 7-14 A. Needle insertion, frontal view. The needle is inserted lateral to the artery,

about 1 cm above the inguinal crease and in

a 30-45 degree cephalad orientation. (On a

model with permission).

Fig 7-14 B. Needle insertion,

lateral view. The needle is inserted

lateral to the artery, about 1 cm

above the inguinal crease and in a

30-45 degree cephalad orientation.

(On a model with permission).

Page 126: Manual Regional Carlo 2010

126 | P a g e

FEMORAL NERVE BLOCK

ULTRASOUND TECHNIQUE

Indications

The same indications than for nerve stimulator techniques.

Patient position

The patient is either supine or semi seated for more comfort.

Type of needle

Usually a 5cm, 22-G, insulated needle is used.

Type of transducer

The femoral nerve is fairly superficial in most of patients, so a high frequency (8-

15 MHz) linear probe is usually adequate.

Scanning

The probe is placed across the upper thigh over the femoral vessels, as shown in

figure 7-15.

If possible we like to place the probe immediately (1 cm) above the crease, where

the nerve is more compacted. The femoral vein is the most medial structure of the

neurovascular bundle and is easily collapsible by the probe. The artery is situated lateral

to the vein and the femoral nerve is located lateral to the artery. The characteristic image

obtained at this level is shown in figure 7-16.

Fig 7-15. Femoral Scanning. The

probe is placed across the

neurovascular bundle, right above

the crease to obtain a short axis

view of the femoral nerve and

vessels. (On a model with

permission).

Fig 7-16. Femoral scanning.

The femoral nerve (FN) is seen

as a flat structure over the

iliopsoas muscle and lateral to

the femoral artery (FA).

Author’s archive.

Page 127: Manual Regional Carlo 2010

127 | P a g e

Needle insertion

The needle can be advanced out of plane, usually from caudal to cephalad, as

shown in figure 7-17, or, as we usually prefer, in plane from lateral to medial, as shown

in figure 7-18.

Local anesthetic and volume

The multiple branches that constitute this nerve provide an ample area of

absorption for the local anesthetic. Usually we use no more than 20 mL of local

anesthetic solution and as little as 10 mL in some occasions. For shorter cases 1.5%

mepivacaine plus 1:400,000 epinephrine provides 3-4 hr of anesthesia. For longer cases

0.5% ropivacaine plus epinephrine is used. For analgesia 0.2% ropivacaine is our drug of

choice.

Side effects and complications

Very rare. Hematomas from puncture of the femoral artery are possible, but

avoidable with meticulous technique, use of small gauge needles and thorough

compression of the arterial puncture when it occurs. The use of ultrasound almost

eliminates this problem.

Fig 7-17. Needle insertion,

out of plane. The needle is

inserted from distal to proximal

(cephalad direction) towards

the femoral nerve. (On a model

with permission).

Fig 7-18. Needle insertion, in

of plane. The needle is inserted

in plane from lateral to medial

towards the femoral nerve. (On a

model with permission).

Page 128: Manual Regional Carlo 2010

128 | P a g e

OBTURATOR NERVE BLOCK

NERVE STIMULATOR TECHNIQUE

Indications

It is rarely performed alone. It is more often combined with femoral, lateral

femoral and/or sciatic blocks.

Point of contact with the nerve

The needle is inserted, if possible, immediately (1 cm) above the inguinal crease

to approach the nerve just distal to the obturator foramen before its two main branches

diverge.

Main characteristics

Although the obturator nerve exits the obturator foramen usually already divided

into anterior and posterior branches, they both run for a short distance (2-3 cm)

physically contiguous in the plane between the pectineus anteriorly and the obturator

externus posteriorly. After reaching the lateral border of the adductor brevis muscle both

branches separate, with the anterior branch passing anterior to this muscle and the

posterior branch posterior to it. It is a common practice to perform separate injections of

both branches. However we believe that if the injection is attempted 1 cm above the

crease both main branches of the obturator nerve can be blocked by a single injection

deep to the pectineus muscle.

Patient position and landmarks

The patient lies supine with the head of the bed slightly elevated. The thigh is

slightly abducted and externally rotated. Many methods have been devised to locate the

obturator nerve. Our own method is to use as the main landmark the pulsation of the

femoral artery. To locate the right obturator nerve the operator uses the right hand and for

the left obturator the left hand. The middle finger is used on both sides to palpate the

pulse of the femoral artery. This way the index finger on either side always points to the

femoral nerve, the ring fingers to the femoral vein and the little finger to the obturator

nerve, as shown in figure 7-19.

Fig 7-19. Finding the obturator

nerve. Our own method to locate the

obturator nerve uses the femoral artery

pulse, which is palpated using the

operator’s middle finger of the same

side to be blocked (right hand to

palpate right side, left hand to left

side). This way the little finger will

always roughly indicate the position of

the obturator nerve (pointed with an

arrow). (On a model with permission).

Page 129: Manual Regional Carlo 2010

129 | P a g e

Type of needle

Depending on the patient, a 5cm, 22-G or a 10cm, 21-G, insulated needle is used.

Nerve stimulator settings

The nerve stimulator is set to deliver a 1.0 mA current, at a frequency of 1 Hz and

pulse duration of 0.1 msec (100 microsec).

Needle insertion

The needle is inserted almost perpendicular to the frontal plane with a slight

cephalad angulation, as shown in figure 7-20.

As the needle traverses the muscular plane, a localized twitch from the pectineus

muscle is usually elicited by direct stimulation. As the needle reaches the deep face of the

muscle and the proximity of the obturator nerve a more global twitch of the thigh in

adduction is obtained. At this point the current is lowered progressively to around 0.5

mA, and if a twitch is still visible, a slow injection is started. If the needle makes contact

with the pubis ramus, it is walked off caudally.

Local anesthetic and volume

A volume of 10-15 mL of local anesthetic is usually used. Mepivacaine 1.5% can

be used with 1:400,000 epinephrine for 3-4 hr of anesthesia. For longer anesthesia 0.5%

ropivacaine with epinephrine can be used. For analgesia 0.2% ropivacaine is commonly

used.

Complications

Hematoma is the most frequent complication of this technique. Adductor muscles

spasm can occur.

Fig 7-20. Needle insertion. The

needle is inserted immediately

above the inguinal crease, almost

perpendicular to the frontal plane,

with a slight cephalad orientation.

(On a model with permission).

Page 130: Manual Regional Carlo 2010

130 | P a g e

OBTURATOR NERVE BLOCK

ULTRASOUND TECHNIQUE

Indications

The same indications mentioned for nerve stimulator techniques.

Patient position

The patient lies supine with the head of the bed slightly elevated. The thigh is

slightly abducted and externally rotated.

Type of needle

Depending on the patient, a 5cm, 22-G or a 10cm, 21-G, insulated needle is used.

Type of transducer

If at all possible a high frequency (8-15 MHz) linear probe is used.

Scanning

Before performing the scanning it is useful, if possible, to locate the adductor

longus, the most superficial of the three adductor muscles, as shown in figure 7-21.

This way the determination of the location of the obturator nerve is framed

between two easily identifiable structures, the femoral vessels on the lateral side and the

medial border of the adductor longus on the medial side. The probe is placed parallel and

slightly above the inguinal crease over the femoral vessels and then traced medially until

it rests over the pectineus muscle, as shown in figure 7-22.

Fig 7-21. Identifying the adductor

longus muscle. With the thigh in slight

abduction and slight external rotation

the adductor longus (AL) can usually

be easily palpated. (On a model with

permission).

Fig 7-22. Scanning the obturator

nerve. The probe is place across the

femoral vessels, as done for femoral

block, and then slowly displaced

medially until it rests over the pectineus

muscle, just cephalad to the inguinal

crease. (On a model with permission).

Page 131: Manual Regional Carlo 2010

131 | P a g e

With the probe over the pectineus muscle the obturator nerve can be seen as a

mostly hyperechogenic ovoid image under the pectineus muscle, as shown in figure 7-23.

If the scanning instead is performed a few centimeters more distally then the two

branches of the obturator can be seen, as shown in figure 7-24.

Needle insertion

My preferred method for this particular block is to use and out of plane technique

from distal to proximal, as shown in figure 7-25.

Fig 7-23. Obturator nerve,

proximal view. With the

probe just medial to the

femoral vein (FV) and just

above the crease, the obturator

nerve (arrow) appears as an

oval hyperechoic structure

under the pectineus muscle.

(FV). Author’s archive.

Fig 7-24. Obturator nerve,

distal view. With the probe

below the inguinal crease the

two main components (arrows)

of the obturator nerve can be

seen above and below the

adductor brevis (AB). Adductor

longus (AL), adductor magnus

(AM) also shown. Author’s

archive.

Fig 7-25. Needle

insertion. The needle

is introduced out of

plane, from distal to

proximal. On a model

with permission).

Page 132: Manual Regional Carlo 2010

132 | P a g e

Local anesthetic and volume

A volume of 10-15 mL of local anesthetic is usually used. Mepivacaine 1.5% can

be used with 1:400,000 epinephrine for 3-4 hr of anesthesia. For longer anesthesia 0.5%

ropivacaine with epinephrine can be used. For analgesia 0.2% ropivacaine is commonly

used.

Complications

Hematoma is the most frequent complication of this technique. Adductor muscles

spasm can occur.

Page 133: Manual Regional Carlo 2010

133 | P a g e

LUMBAR PLEXUS BLOCK (also called “psoas compartment block”)

NERVE STIMULATOR TECHNIQUE

Indications

Its goal is to produce anesthesia of the lateral femoral, femoral and obturator

nerves, so it can be used along with a proximal sciatic nerve block to provide anesthesia

of the entire lower extremity. It is also used to provide postoperative analgesia for hip and

knee surgery.

Point of contact with the nerve(s)

The plexus is accessed deeply in the lumbar area in the space limited by the

quadratus lumborum posteriorly (more superficial or closer to the skin of the back) and

the psoas muscle anteriorly (deeper).

Main characteristics

It is the posterior version of what a “3-in-1” block in the femoral area intends to

accomplish. It is a deep block, in which the needle goes through several layers, including

subcutaneous tissue, the mass of paraspinal muscles, and the quadratus lumborum muscle

before ending just posterior to the psoas muscle, in the retroperitoneal space.

Because of the depth at which the nerves are located and the long needles used,

the operator has little control over the exact location of the needle tip, increasing the

potential risk for complications. The most frequent complication is to produce an epidural

block, but also cases of total spinal anesthesia have been described. Because of the

relatively large volumes of local anesthetics used systemic toxicity can also develop.

Cases of penetration of the peritoneal cavity with injury of its contents as well as large

retroperitoneal hematomas and death have been reported with this block. It is essential

that the operator be familiar with the anatomy of this region before attempting this block,

which should be performed only by experienced people.

The lumbar plexus block perhaps should not be performed in obese patients.

Patient position and landmarks

The patient is placed in the lateral position with both hips and knees flexed like

for a neuraxial block. A line is drawn at the level of the iliac crest (L4-L5 interspace)

starting at the midline (spinous processes) and extending to the level of the posterior

superior iliac spine. The line is then divided into thirds, as shown in figure 7-26.

Fig 7-26. Landmarks. A line at the

level of the iliac crests is drawn from

the midline to the PSIS and divided

into thirds. The junction of the lateral

and middle thirds is the point of

needle insertion shown with an arrow.

On a model with permission).

Page 134: Manual Regional Carlo 2010

134 | P a g e

Type of needle At least a 10cm, 21-G, insulated needle is necessary for this block.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 1.5 mA, at a pulse frequency of

1 Hz and pulse duration of 0.1 msec (100 microsec).

Needle insertion

The needle is inserted parallel to the midline at the junction between the lateral

third and middle third of the line joining the midline with the level of the posterior

superior iliac spine, as shown in figure 7-27.

This insertion is more medial than the original technique. It is based on a study by

Capdevila et al (Anesth Analg 2002; 94: 1606-1613) demonstrating that the point of

needle insertion at the level of the PSIS falls lateral to the plexus mandating a medial

reposition of the needle that potentially could increase the chance for epidural or spinal

anesthesia.

As the needle is inserted through the mass of the paraspinal muscles a local

contraction is usually observed. The transverse process of L4 or the nerves of the lumbar

plexus should be contacted within 3 cm from the disappearance of the twitch from the

back muscles. If not, the needle is withdrawn superficially and redirected caudally or

cephalad. If the transverse process is contacted the needle should be walked off caudally

until a quad twitch is obtained, usually no deeper than 2 cm from the transverse process.

If no response is obtained within 2 cm the needle can be redirected cephalad from the

transverse process and again advanced for up to 2 cm.

When a muscle twitch from the quad is obtained the current in the nerve

stimulator is decreased to around 0.5-0.8 mA and with a visible response a gentle

aspiration is performed for blood or CSF before injecting a “test dose” amount of 3-5 mL

of epinephrine-containing local anesthetic. If no intravascular or subarachnoid injection is

detected the rest of the local anesthetic volume is slowly injected in small increments

with frequent gentle aspirations. The preferred response in this block is quad response.

An obturator response could mean that the needle is too medial and should be redirected.

A distal response (sciatic) could mean that the lumbosacral trunk is being

stimulated and could indicate a needle too medial. A medial position of the needle could

carry an increased risk of neuraxial injection.

Fig 7-27. Needle insertion. The

needle is inserted at the junction

between the lateral and middle

thirds of the line drawn from the

midline to the PSIS, and directed

parallel to the midline. On a

model with permission).

Page 135: Manual Regional Carlo 2010

135 | P a g e

Local anesthetic and volume

For anesthesia of 3-4 hours 1.5% mepivacaine with epinephrine 1:200,000 (a

larger concentration than we use at other sites) can be used. For longer anesthesia the

preferred drug is 0.5% ropivacaine plus 1:200,000 epinephrine. For analgesia 0.2%

ropivacaine is adequate.

Complications

I already mentioned that this block should be performed only by experienced

people. Epidural spread is the most common problem with an incidence of 1-16%, but

can be as high as 88% in some reports. Subarachnoid injection is a dangerous

complication not always avoided by a test dose. Death associated with total spinal has

been reported. Large retroperitoneal hematomas are also possible and therefore this block

should adhere to the same anticoagulation guidelines than neuraxial techniques. Kidney

and other injuries have also been reported.

LUMBAR PLEXUS BLOCK

ULTRASOUND TECHNIQUE

The ultrasound technique is based on the ability to map the sonoanatomy of the

lumbar spine and associated muscles. Because of the depth of these structures a curved,

low frequency probe is used. The technique can be “ultrasound assisted” rather than

ultrasound guided.

Page 136: Manual Regional Carlo 2010

136 | P a g e

SCIATIC NERVE BLOCK

Classic approach (Labat as modified by Winnie)

NERVE STIMULATOR TECHNIQUE

Indications

As an isolated block, it provides anesthesia of the back of the thigh (through

anesthesia of the posterior cutaneous nerve of the thigh, a branch of the sacral plexus) and

most of the lower extremity below the knee, with the exception of the medial side of the

leg (saphenous nerve). If used along with femoral, lateral femoral and obturator nerve

blocks (lumbar plexus block), it completes the anesthesia of the entire lower extremity.

Point of contact with the nerve The nerve is contacted in the gluteal area at the point where it is entering the

gluteal area caudal to the piriformis muscle. The needle on occasions could traverse

through the piriformis.

Main characteristics

Labat’s approach is a highly anatomical approach that requires the identification

of the posterior superior iliac spine (PSIS) and the greater trochanter (GT). A dissection

of the gluteal area shows that this is a reliable approach if the operator is able to

accurately determine the position of the PSIS and GT, disregarding ANY soft tissue (i.e.,

muscle, bursa, subcutaneous tissue and fat).

Position of the patient and landmarks

The patient is positioned in lateral decubitus, with the side to block up. The

dependent leg is extended. The non-dependent leg is flexed at the hip and at the knee,

while the buttock is rotated anteriorly (Sim’s position).

The PSIS is marked and so is the superior aspect of the GT. The midpoint of this

PSIS-GT transverse line is determined. From this midpoint a perpendicular line

measuring 3 cm, is directed caudally and medially. This is the point of needle insertion. It

is important that the marks placed on the skin truly represent the posterior projection of

the bony prominences on the skin. Marking the position of the GT on the lateral buttock

for example, would artificially lengthen the PSIS-GT line (because of soft tissue), making

its midpoint artificially more lateral and away from the sciatic nerve. The 3-cm

perpendicular line has also been a source of problems. Several authors have modified its

length to a range of 2 to 5 cm.

In 1974 Winnie and collaborators published in Anesthesiology Review a

modification to the Labat’s technique. This modification has been universally adopted

and it is now commonly known as the “classic” technique. In order to deal with the

controversy about the appropriate length of Labat’s original 3-cm perpendicular line, they

proposed to draw an additional transverse line extending from the sacral hiatus (SH) to

the tip of the greater trochanter to provide a distal point of intersection for the

perpendicular line of Labat. In this manner, the length of this line would be determined

by the distance between the two transverse lines, and would be “self adjustable” to every

particular patient. Quoting the authors, “with this technique the distance along the

perpendicular line will vary with the height of the patient”. This apparent solution is

Page 137: Manual Regional Carlo 2010

137 | P a g e

widely accepted but it might have some problems of its own. Because, as discussed in the

anatomy section, the transverse diameter of the pelvis is fairly constant in all adults, any

prolongation of the perpendicular line starting from a similar point would bring it closer

to the midline (its direction is caudal and medial). This will mean that a tall patient with a

long sacrum will have a sciatic nerve located closer to the midline (long perpendicular

line due to longer sacrum) than a short patient (short perpendicular line due to shorter

sacrum). This obviously could not be the case. The fact is that Labat’s perpendicular line

was not created to be adjustable.

The combined “classic” approach (Labat-Winnie), despite its shortcomings, is the

most commonly used posterior approach to the sciatic nerve in the gluteal area.

Technique

Usually the block can be completed with a 10cm, 21-G, insulated needle, but

sometimes a longer needle needs to be used. The needle is advanced, perpendicular to all

planes until a twitch from the sciatic nerve is found. If a twitch is still visible at 0.5 mA a

slow injection is started with frequent aspirations. If the nerve is not contacted, the

technique does not have a clear strategy for reposition of the needle. In fact the nerve

could be at any point around a 360-degree radius.

Local anesthetic and volume

For anesthesia 1.5% mepivacaine plus 1:400,000 epinephrine in a volume of 30-

35 mL can provide 3-4 hrs of anesthesia. Ropivacaine 0.5-0.75% with epinephrine can be

used if longer duration is needed.

Complications

The literature mentions that the absorption from this site is minimal. However, it

is important to remember that the branches of the inferior gluteal vessels at this level are

large and multiple, therefore hematomas could develop. The patient lying supine

immediately post block could theoretically help to decrease the chance for a hematoma to

develop.

It is important to inject slowly, alternated with frequent and gentle aspirations.

Dysesthesias in the territories of the sciatic or posterior femoral cutaneous nerves are

reported more frequently after this block than any other. These problems usually resolve

spontaneously within 1-2 weeks.

Page 138: Manual Regional Carlo 2010

138 | P a g e

SCIATIC NERVE BLOCK

Franco’s approach

NERVE STIMULATOR TECHNIQUE

Indications

The same indications than for a classic technique.

Point of contact with the nerve

This is a mid-gluteal technique that approaches the sciatic nerve distal to the

piriformis in the proximity of the ischium at about the same level than the classic

technique does. However, because caudal to the piriformis the sciatic nerve runs almost

parallel to the midline, this technique can be performed at any point between mid-gluteal

to subgluteal levels. It can also be used for continuous catheter techniques.

Main characteristics

This is a simple technique that relies on one simple anatomical landmark, the

intergluteal sulcus (midline), making the palpation of any buried landmarks totally

unnecessary. It is based on simple, although not universally known facts:

1. The trajectory of the sciatic nerve in the gluteal region is for the most part parallel

to the midline.

2. The width of the adult pelvis is similar in all adults and according to some

anthropologists “surprisingly” similar in males and females at any given age.

Variations in hip width are mainly the result of hormone-dependent, different

patterns of fat deposition in both sexes and are not due to significant differences

in the width of the bony pelvis. Although male and female pelvises are indeed

different, most of those differences are limited to the diameters of the minor or

inner pelvis without affecting the total diameter of the pelvis. Thicker bones in

males compensate for the wider inner pelvis of females to make the average

bicrestal diameter (total width) 280 mm in males and 275 mm in females.

3. As determined by our own study (Anesthesiology 2003; 98: 723-728), the sciatic

nerve is located about 10 cm from the midline (intergluteal sulcus) in all adults.

What remains highly variable is the amount of adipose tissue that can accumulate

in the buttocks affecting the depth of the nerve and its distance to the lateral side

of the patient. The distance midline-nerve is, on the other hand, unaffected by fat

accumulation as it is dictated by the distance between the ischium and the midline

(fixed after puberty).

Position of the patient and landmarks

This block can be performed in the lateral decubitus or in the prone position. We

prefer to do it almost 100% of the times in the lateral position, because it is more

comfortable for the patient and faster to prepare for.

The patient is placed in the lateral position with both hips and knees slightly flexed.

In a true lateral decubitus, a tangential line to the buttocks, should form a 90-degree angle

with the table. Having the patient placed at straight angles with the table, makes his/her

midline parallel to the table.

Page 139: Manual Regional Carlo 2010

139 | P a g e

The midgluteal sulcus is identified and the point of needle insertion is marked at 10

cm from it around the midgluteal region, as shown in figure 7-28.

This is a linear measurement that, on purpose, disregards any particular curvature

or contour in the patient’s buttocks. The insertion point, always located at 10 cm from the

midline, can be moved distally at will, as far caudal as the subgluteal fold. This could be

necessary for example, if the buttock is large and the needle is not long enough.

Type of needle

A 10cm, 21-G, insulated needle is usually sufficient, although in some cases a

15cm needle is necessary.

Nerve stimulator settings

For this technique we set the nerve stimulator current at 1.5 mA (1.8 mA in

diabetic patients), with a frequency of 1 Hz and pulse duration of 0.1 ms (100 microsec).

Needle insertion

The needle is advanced parallel to the midline, as shown in figure 7-29.

When the needle reaches the gluteus maximus muscle a local muscular twitch of

the buttock is observed. This twitch is very reassuring, telling the operator that the

needle-stimulator unit is functional and most importantly, providing information on

sciatic nerve depth. If 8 cm or more, of a 10 cm needle, have been used to reach the

gluteus maximus, it is unlikely that the needle will be long enough to reach the sciatic

nerve.

Fig 7-28. Patient position and

point of needle insertion. The

patient lies on lateral decubitus. The

point of needle entrance is easily

found at 10 cm from the midline at

about midgluteal level. (On a

patient with permission).

Fig 7-29. Needle insertion. The needle is inserted parallel

to the midline.(On a patient

with permission).

Page 140: Manual Regional Carlo 2010

140 | P a g e

The needle is advanced through the gluteus muscle, producing a visible local

twitch that does not disappear until the needle exits the deep surface of this muscle. The

ensuing “silence” is evidence that the needle is passing through the connective tissue that

separates the gluteus maximus from the nerve. It should be soon followed by a twitch

resulting from stimulation of the sciatic nerve. The nerve is rarely more than 2 cm deeper

to the gluteus maximus.

I believe that any of the possible responses from the sciatic nerve (i.e. eversion,

dorsiflexion, inversion and plantar flexion) are adequate, provided that the injection is

made with a visible response at 0.5 mA or less. There are few reports in the literature that

argue in favor of inversion and against eversion. This is not our experience.

If no response from the sciatic nerve is obtained deeper to the gluteus maximus,

then a reposition of the needle is necessary. Here is very important to take into account

the “vector” effect, the impact of the angle of reinsertion in the final position of the

needle. According to my own calculations, at a theoretical depth of 9 cm, a 10-degree

correction angle, moves the needle tip 1.6 cm, while a 20-degree correction moves it 3.4

cm. Because the nerve is around 1.5 cm wide, it would be very easy to “overshoot” the

correction.

Some useful tips when trying to “pinpoint” the sciatic nerve

When an adequate twitch is found, the nerve stimulator current is lowered until a

twitch is still visible at 0.5 mA or less. This is done while maintaining visual contact with

the twitch. If before reaching 0.5 mA the twitch becomes too weak, the current is not

lowered any further and instead the operator slowly moves the needle closer to the nerve.

It is not infrequent to see the response fade as the needle is inserted deeper. This

can be the result of a needle approaching the nerve tangentially, overshooting the nerve

on one of its sides. In these cases we usually like to perform a small correction in order

to get a “bull’s eye” alignment with the nerve. Deciding whether to correct lateral or

medial depends on what type of response is being elicited. Eversion and dorsiflexion are

responses from the common peroneal nerve (lateral side), while inversion and plantar

flexion are responses from the tibial nerve (medial side). A small correction is then made

accordingly. A more controlled correction can be accomplished by only partially

removing the needle a couple of cm. The unburied portion of the needle is then bent and

directed in the desired direction. The buried portion of the needle keeps the needle from

overcorrecting. Bringing the needle out completely, and then reinserting it, carries a

chance of overshooting the correction.

Complications

Same as classic approach.

Pearls

The 10 cm measurement is a linear measurement that disregards, on purpose, the

contour of the patient’s buttock. This linear measurement tries to reflect only the

distance between the midline and the outer lip of the ischium, without soft tissue

interference.

Placing the patient in true lateral position, makes the patient’s midline parallel to

the table. If this position is not possible, the operator needs to ascertain the degree

Page 141: Manual Regional Carlo 2010

141 | P a g e

of inclination of the midline with respect to the table, so the needle still can be

advanced parallel to the patient’s midline.

When the nerve is not found at first attempt, it could only be located either lateral

or medial to the needle. Because of gravity, it is more frequent to underestimate

the midline-nerve distance (sagging midline). Therefore, the first correction

should be lateral.

When reposition is necessary, keep in mind the “vector” effect. At a theoretical

distance of 9 cm a 10-degree correction will move the needle app 1.6 cm. A 20-

degree correction will move it 3.4 cm. This big “jump” could easily overshoot the

correction. A small 10-degree correction usually is all it takes to localize the

nerve.

Page 142: Manual Regional Carlo 2010

142 | P a g e

MIDGLUTEAL SCIATIC BLOCK

ULTRASOUND TECHNIQUE

Indications

The same than for nerve stimulation techniques.

Patient position

For a midgluteal approach the patient can be placed prone, lateral or in Sim’s

position.

Type of needle

A 10 or 15cm, 21-G, insulated needle is used.

Type of transducer

Because of the depth at which the sciatic nerve is located in the gluteal area, most

of the times a curved, low frequency (5-7 MHz) probe is needed.

Scanning

The nerve is identified in cross section (short axis) by placing the transducer

across the midgluteal area at which point the sciatic nerve can be identified between the

greater trochanter and ischial tuberosity, as shown in figure 7-30.

Needle insertion

The easiest approach is to introduce the needle out of plane from distal to

proximal as observed in figure 7-31.

Fig 7-30. Sciatic nerve scanning.

With the probe in the midgluteal

region the sciatic nerve (SN) is

observed between the greater

trochanter (GT) and ischial

tuberosity (IT). Author’s archive.

Fig 7-31. Needle insertion. The

needle is inserted out of plane

from distal to proximal. On a

model with permission).

Page 143: Manual Regional Carlo 2010

143 | P a g e

Figure 7-32 shows an ultrasound image of an out of plane technique during

injection.

Local anesthetic and volume

For anesthesia 1.5% mepivacaine plus 1:400,000 epinephrine provides 3-4 hr of

anesthesia. For longer duration 0.5% ropivacaine can be used. For analgesia 0.2%

ropivacaine is appropriate.

Complications

The same as nerve stimulation techniques.

Fig 7-32. Sciatic nerve injection. Out of

plane technique. The sciatic nerve (SN) is

shown in between greater trochanter (GT)

and ischial tuberosity (IT). The tip of the

needle is shown above the nerve pointed

with an arrow and the injected local

anesthetic appears as a dark hypoechoic

shadow on top of the nerve. Author’s

archive.

Page 144: Manual Regional Carlo 2010

144 | P a g e

SCIATIC NERVE BLOCK, SUBGLUTEAL

di Benedetto’s approach

Indications

This is a block more suitable for surgery below the knee, because it does not

reliably block the posterior femoral cutaneous nerve (back of the thigh). It can also be

used for continuous catheter techniques.

Point of contact with the nerve The nerve is approached in the vicinity of the subgluteal fold.

Main characteristics

There are several techniques performed at or around the subgluteal fold. Some

authors mention Raj’s “supine approach” to sciatic nerve (Anesthesia & Analgesia 1975)

as being the first. In fact, this is a sciatic block performed between the ischium and

greater trochanter (mid-gluteal, not subgluteal level), just a few cm caudal to Labat’s

classic approach. In this technique the extremity is elevated and flexed at the hip and

knee, stretching the buttock tissues. This supposedly brings the sciatic nerve closer to the

skin. It is interesting to note that, even though this technique is universally known as

“Raj’s supine approach”, a completely similar technique was published a year earlier

(1974) by Winnie and colleagues in Anesthesiology Review. Raj’s technique was

correctly devised “for below-the-knee operations”. This fact is frequently forgotten and

we will revisit it later.

A popular infra or subgluteal technique is the technique introduced by di

Benedetto and colleagues in 2001.

Patient position and landmarks

This block is performed in the Sim’s position, as the classic technique. The greater

trochanter and the ischium are identified and a line is drawn in between the two. The

midpoint of this line is determined. A second line is drawn from this midpoint,

perpendicularly and caudally for 4 cm. This is the needle insertion point. According to

the authors, the operator should be able to palpate at this point a “skin depression”, which

would represent “the groove between the biceps femoris and semitendinosus muscles”.

This groove supposedly represents the trajectory of the sciatic nerve. This is just one

more instance in which anesthesiologists display their love affair with grooves. In fact

cadaver dissections show:

1. Ischium and greater trochanter are located at about the same transverse plane in

the buttocks, as shown in figure 7-1. Di Benedetto’s perpendicular line going

caudal and lateral, needs to have the trochanter located significantly higher than

the ischium.

2. The subgluteal fold is about 8 cm caudal to the midpoint between ischium and

greater trochanter and not 4 cm. On the other hand, being the subgluteal fold so

evident, would it suffice to extend the line until it intercepted the subgluteal fold?

3. At the subgluteal fold the three components of the hamstring muscles are

practically fused together in one single tendon, without any evident groove in

Page 145: Manual Regional Carlo 2010

145 | P a g e

between. More distally in the thigh a groove can be found between biceps and

semitendinosus, but it is too subtle to be easily palpable through several layers of

tissue (skin, subcutaneous tissue and thick fascia lata).

4. A groove is visible in most people between the biceps and the iliotibial tract. This

groove has nothing to do with the trajectory of the sciatic nerve.

5. The sciatic nerve runs under the biceps femoris and not in a groove between

biceps and semitendinosus.

Technique

The authors suggest to insert the needle perpendicular to the skin until a twitch

from the sciatic nerve is obtained.

Local anesthetic and volume

The same than for classic approach

Complications

Common to other approaches to the sciatic nerve.

Page 146: Manual Regional Carlo 2010

146 | P a g e

SCIATIC NERVE BLOCK, SUBGLUTEAL

Franco’s approach

The subgluteal approach can be easily performed at 10 cm from the midline at the

subgluteal fold, with the patient lying in lateral decubitus, as shown in fig 7-33.

The 10-cm measurement is made from the midline at the level of the subgluteal

fold, in a way similar to the one described for the mid-gluteal approach. The needle is

advanced parallel to the midline, through the gluteus maximus muscle and into the sciatic

nerve. The current is lowered to around 0.5 mA and a slow injection is started. If the

nerve is missed at first pass it could only be located medial or lateral to the needle. The

needle is reinserted, with a small 10-degree correction in its orientation, first lateral

(toward the trochanter) and then medial (to the midline) if necessary.

Ultrasound technique

Although the same tissue layers cover the sciatic nerve at the midgluteal and

subgluteal levels, the fat layer is usually thinner. This makes the ultrasound visualization

of the sciatic nerve at this level usually easier than in the midgluteal area. Depending on

depth, the nerve can be visualized with a linear high frequency probe, but frequently a

lower frequency probe is needed. Curved low frequency probes are needed for bigger

patients. The patient is placed prone, lateral position or in Sim’s position. The nerve is

visualized in cross section (short axis) and the needle is advanced either out of plane

(usually) or in line with the probe.

A few facts on subgluteal approach

1. This approach consistently misses the posterior femoral cutaneous nerve, so

anesthesia of the back of the thigh is only obtained in about 30% of the cases (our

own data, Reg Anesth Pain Med 2006; 31: 215-20). The reason is that the

posterior femoral nerve is usually already a superficial nerve (above the fascia) at

the level of the subgluteal fold.

2. The inferior border of gluteus maximus and subgluteal fold are not the same

thing. Therefore, during a subgluteal approach the needle needs to pass through

the same layers of tissue than at more proximal approaches.

Fig 7-33. Needle insertion point.

It is easily found at 10 cm from

the midline as done for the

midgluteal approach. (On a

patient with permission).

Page 147: Manual Regional Carlo 2010

147 | P a g e

3. The sciatic nerve is relatively more superficial at the subgluteal fold because the

amount of fat decreases from mid-gluteal to subgluteal level, although the type of

layers (fat and muscle) remains the same.

4. The popliteal fossa is the only level in the trajectory of the sciatic nerve in which

the nerve is not covered superficially by muscle. Approaching the sciatic nerve,

without passing through muscle is the only true advantage of a popliteal approach.

5. In terms of anesthesia distribution, the subgluteal approach is more comparable to

the popliteal block than to other more proximal approaches.

Page 148: Manual Regional Carlo 2010

148 | P a g e

SCIATIC NERVE BLOCK, POPLITEAL

Franco’s approach

NERVE STIMULATOR TECHNIQUE

Indications

It is especially suitable for foot surgery. Along with femoral nerve block

(saphenous) it provides complete anesthesia below the knee.

Point of contact with the nerve

The needle approaches the sciatic nerve high in the popliteal fossa, before its main

components diverge from each other.

Main characteristics

This is the only place in the trajectory of the sciatic nerve where the nerve is not

covered superficially by muscle, perhaps its only true advantage over other more

proximal approaches to the sciatic nerve. Characteristically, a sciatic block done at this

level with a blind technique has a slower onset and lower success rate than more

proximal approaches. The fact that the two components of the nerve diverge from each

other could account for some of the partial blocks. However, slower onset and lower

success rates are sometimes observed in cases where there is reasonable evidence to

believe that the main trunk has been contacted. One of the possible reasons is that the

nerve sheath at this level fuses with the fat that fills the popliteal fossa soaking the local

anesthetic away from the nerve. Ultrasound techniques have a faster onset.

Patient position and landmarks

This block is most usually performed in the prone position. The patient’s patella is

palpated with two hands, to verify the neutral position of the knee on the bed (the natural

resting position of the knee is with a small degree of lateral rotation). The patient is then

asked to flex the knee slightly to make the biceps (lateral) and semitendinosus (medial)

tendons visible at the popliteal crease. A mark is placed on both tendons at the crease, as

shown in figure 7-34.

Fig 7-34. Landmarks. The biceps

and semitendinosus tendons (ST)

are marked at the crease. (On a

model with permission).

Page 149: Manual Regional Carlo 2010

149 | P a g e

The distance between these two points in adults is usually 6-7 cm in females and

7-8 cm in males. The midpoint between the two tendons is located and marked, as shown

in figure 7-35.

The needle insertion point is then found 7-9 cm above the crease, as shown in

figure 7-36.

Type of needle

A 5cm, 22-G, insulated needle is usually adequate.

Nerve stimulator settings

The nerve stimulator is set to deliver a current of 1.0 mA (higher in diabetics)

with a pulse frequency of 1 Hz and pulse duration of 0.1 msec (100 microsec).

Needle insertion

The needle is introduced with a 30-45 degree cephalad orientation, as shown in

figure 7-37.

Fig 7-35. Inter tendinous

distance. The midpoint between

biceps and semitendinosus

tendons is determined at the

crease. (On a model with

permission).

Fig 7-36. Point of needle insertion. The

point of insertion shown with an arrow, is

found 7-9 cm above the midpoint of the

tendons at the crease. (On a model with

permission).

Fig 7-37. Needle insertion.

The needle is inserted with

a cephalad orientation. (On

a model with permission).

Page 150: Manual Regional Carlo 2010

150 | P a g e

The needle is directed approximately 45-degrees cephalad, so the contact with the

nerve happens at 1-2 cm higher from the crease than the actual entrance point, increasing

the chances that the sciatic nerve is contacted prior to its division. The distance from the

crease at which the needle is inserted varies according to the patient’s height. A good

ballpark estimation is to insert the needle at a distance from the crease that is 1 cm longer

than the intertendinous distance.

Once a response from the sciatic nerve is elicited, and still present at 0.5 mA or

less, a slow injection is started with frequent aspirations.

Local anesthetic and volume

I believe that a block of the sciatic nerve in the popliteal fossa using nerve

stimulation requires a higher volume than more proximal approaches. As a general rule I

give about 10 mL more of local anesthetic solution than what I would give to the same

patient at more proximal locations. This comes to about 35-45 mL of 1.5% mepivacaine

with 1:400,000 epinephrine for 3-4 hr of anesthesia. If longer anesthesia is desired I

would use 10 mL of 1.5% mepivacaine with epinephrine followed by 30 mL of 0.5%

ropivacaine.

Complications

Small hematoma can develop. Residual dysesthesia lasting up to two weeks can

be seen.

Page 151: Manual Regional Carlo 2010

151 | P a g e

POPLITEAL BLOCK, LATERAL APPROACH

WITH NERVE STIMULATOR

Indications

It is especially suitable for any surgery below the knee including ankle and foot,

in patients who cannot be placed in any other position than supine

Point of contact with the nerve

Similar to the posterior technique. The needle approaches the sciatic nerve from

the lateral side, before this nerve’s main components diverge from each other. The needle

is advanced between the biceps (posteriorly) and vastus lateralis (anteriorly) into the

popliteal fossa.

Main characteristics

Blocking the sciatic nerve with this approach is a little bit more challenging than

the posterior approach. Biceps and vastus lateralis fibers are in close physical contact so

the needle usually stimulates some muscle fibers before reaching the sciatic nerve.

Patient position and landmarks

The patient lies supine in the semi sitting position. A pillow is placed under the

leg, so the hip and knee are slightly flexed. The patient can be asked to shift his/her

weight to the opposite side, so a small degree of lateral rotation is obtained. The popliteal

crease is identified and marked toward the lateral side of the knee. The cleavage between

the biceps and vastus lateralis is identified. A mark is placed in this groove 10 cm

proximal to the popliteal crease. This is the point of needle insertion.

Technique

The midpoint of the patella is found and a line is drawn from it proximally into

the thigh. This line represents roughly the projection of the sciatic nerve and therefore it

can be used to estimate the depth of the sciatic nerve, as measured from the lateral side.

With the thigh in slight lateral rotation the needle is advanced with a 30-degree posterior

orientation. A local twitch of biceps and/or vastus lateralis muscles can be found before

entering the popliteal fossa. If the needle overshoots the projection of the nerve without

eliciting a twitch, it is withdrawn to the skin and a small 10-degree posterior correction is

applied before reinsertion. With a visible twitch at 0.5 mA or less, a slow injection is

started with frequent aspirations.

Local anesthetic and volume

The same than for posterior approach.

Complications

The same than for posterior approach.

Page 152: Manual Regional Carlo 2010

152 | P a g e

POPLITEAL BLOCK

ULTRASOUND TECHNIQUE

Indications

The same indications than nerve stimulation techniques.

Patient position

There are basically two main positions in which this block can be performed,

supine and prone. The views obtained are similar, but in general the supine technique can

be more challenging, especially in larger patients. The supine technique usually involves

an in-plane lateral approach, while the prone technique provides the opportunity for out

of plane approaches also. Whether the technique is done supine or prone, having the

patient flex the knee improves the visualization of the sciatic nerve and its components.

Type of needle

If an out of plane technique is performed usually a 5cm, 22-G, insulated needle

suffices. If an in plane lateral approach is attempted usually a longer 10cm, 21-G,

insulated needle is needed.

Type of transducer

In most cases a linear, high frequency (8-15 MHz) is used. In larger patients it is

sometimes necessary to use a curved, low frequency (3-7 MHz) probe.

Scanning

The nerve is scanned in short axis. The scanning can be started at any level in the

popliteal fossa, but it is helpful to start at the crease where the popliteal vessels, vein and

artery, have an intimate relationship with the tibial component of the sciatic nerve. Figure

7-38 shows a sequence of images as the probe is moved from distal to proximal.

Fig 7-38, A and B.

Popliteal scanning. Image

A (left) obtained at the

crease shows vein (PV),

artery (PA) and tibial nerve

(arrow). Figure B (right)

shows both components of

the sciatic nerve (arrows). Author’s archive.

Fig 7-38, C and D.

Popliteal scanning. Image

C (left) shows both

components approaching

each other (arrows).

Figure D (right) shows the

sciatic nerve (SN) as a

single structure. Author’s

archive.

Page 153: Manual Regional Carlo 2010

153 | P a g e

Needle insertion

The needle can be inserted out of plane, usually from distal to proximal or in

plane from lateral to medial, as shown in figure 7-39.

A needle inserted in plane from the lateral side is easily seen in the screen as

shown in figure 7-40.

Local anesthetic and volume

For surgery 30-40 mL of 1.5% mepivacaine can provide 3-4 hr of anesthesia. For

longer cases 30 mL of 0.5% ropivacaine with epinephrine can be used. For analgesia

0.2% ropivacaine is adequate.

Fig 7-39. Needle insertion,

supine technique. The needle is

inserted in plane, from the lateral

side in the groove between vastus

lateralis and biceps. (On a model

with permission).

Fig 7-40. Popliteal block, in plane

technique. The needle (shown with 2

arrows) is seen approaching the

sciatic nerve (SN) from the lateral

side. The injected local anesthetic is

seen as a hypoechoic (dark) lagoon

surrounding the nerve. Author’s

archive.

Page 154: Manual Regional Carlo 2010

154 | P a g e

References

1. Snell RS: Clinical anatomy for medical students, 3rd

edition. Boston, MA: Little,

Brown and Company; 1986

2. Labat G: Regional anesthesia: Its technique and clinical application. Philadelphia,

PA: W.B. Saunders, 1922

3. Shipman P, Walker A, Bichell D: Human skeleton. Cambridge, MA: Harvard

University Press; 1985

4. Hall J, Froster-Iskenius U, Allanton J: Handbook of normal physical

measurements. Oxford: Oxford University Press; 1989

5. Cunningham’s Textbook of Anatomy, 5th edition. Edited by Robinson A. New

York, William Wood and Company, 1928, pp 258

6. Hollinshead’s Textbook of Anatomy, 5th edition. Edited by Rosse C, Gaddum-

Rosse P. Philadelphia, Lippincott-Raven, 1997, pp 641–80

7. Winnie A, Ramamurthy S, Durrani Z, et al. Plexus blocks for lower extremity

surgery. Anesthesiology Review 1974; 1: 11-16

8. Franco, CD. Posterior approach to the sciatic nerve in adults: Is Euclidean

geometry still necessary? Anesthesiology 2003; 98: 723-728

9. Franco CD, Choksi N, Rahman A, Voronov G, Almachnouk M. A Subgluteal

Approach to the Sciatic Nerve in Adults at 10 cm from the Midline. Reg Anesth

Pain Med 2006; 31: 215-20

10. Di Benedetto P, Bertini L, Casati A, et al. A new approach to the sciatic nerve

block: A prospective, randomized comparison with the classic posterior approach.

Anesth Analg 2001; 93: 1040-1044

11. Rogers J, Ramamurthy S: Lower extremity blocks, Regional anesthesia and

analgesia. Edited by Brown DL. Philadelphia, PA: W.B. Saunders Company,

1996

12. Mulroy M: Regional Anesthesia, An illustrated procedural guide, 3rd

edition.

Philadelphia, PA: Lippincott Williams & Wilkins; 2002

13. Enneking FK, Chan V, Greger J, et al. Lower-extremity peripheral nerve

blockade: Essentials of our current understanding. Reg Anesth Pain Med 2005;

30: 4-35

14. Vloka JD, Hadzic A, Drobnik L, Ernest A, Reiss W, Thys DM. Anatomical

landmarks for femoral nerve block: A comparison of four needle insertion sites.

Anesth Analg 1999; 89: 1467-1470

15. Capdevila X, Macaire P, Dadure C, Choquet O, Biboulet P, Ryckwaert Y,

D’Athis F. Continuous psoas compartment block for postoperative analgesia after

total hip arthroplasty: New landmarks, technical guidelines, and clinical

evaluation. Anesth Analg 2002; 94: 1606-1613

16. Orebaugh SL. The femoral nerve and its relationship to the lateral circumflex

femoral artery. Anesth Analg 2006; 102: 1859-1862

Page 155: Manual Regional Carlo 2010

155 | P a g e

CHAPTER 8

CONTINUOUS NERVE BLOCKS

Page 156: Manual Regional Carlo 2010

156 | P a g e

Introduction

Single-shot peripheral nerve blocks provide quality anesthesia for a variety of

different procedures. In most cases postoperative pain is moderate and manageable with

either IV PCA (patient controlled analgesia) or oral analgesics. However, there are

surgical procedures known to be followed by intense pain in the postoperative period.

Pain is a very important and usually not well addressed problem. It does not only affect

patients physically and emotionally, but also affects their recovery time and

rehabilitation.

In those cases in which postoperative pain is expected to be more than moderate

and lasts longer than the duration of a single shot block, the anesthesiologist needs other

means to produce and prolong the analgesia. Ideally, analgesia could be provided by

slow-released analgesic products injected along with local anesthetics during single shot

techniques. Local anesthetics and other substances like morphine have been added to

liposome systems to deliver controlled and steady doses of analgesia. However, to date

only duromorph, a liposomal system delivering morphine, is the only one available. It has

been approved by the FDA for epidural analgesia. In this context continuous peripheral

nerve blocks with perineural catheters become an excellent option for postoperative

analgesia providing the versatility in duration and effect that single shot techniques lack.

F. Paul Ansbro published in 1946 what is widely considered the first account of a

continuous peripheral nerve block technique. He described a technique in the

supraclavicular area in which he used a needle passed through a cork for stabilization.

Once the needle was inserted to an adequate level, as judged by paresthesia, the cork was

advanced to the level of the skin and taped. A tubing connected to a syringe provided the

opportunity for what Ansbro called “fractional injections”. More recently in the 1970s,

Selander introduced continuous techniques in the axillary region using an IV cannula left

in place.

Benefits of continuous perineural catheters

Many authors have demonstrated the benefits of continuous techniques, mainly

prolonged analgesia without the undesirable side effects associated with opioid use (i.e.,

nausea, vomiting, constipation, dependency), better patient satisfaction and better

ability to participate in rehabilitation. Liu and Salinas published in 2003 an excellent

review on continuous perineural blocks. After an extensive review of the available

literature they concluded that there was enough evidence to support the claim of superior

analgesia of continuous perineural blocks as compared to IV PCA “for open shoulder

procedures and total knee replacement”. It is likely that may other surgical procedures

could also benefit from the ability to extend the analgesia provided by perineural

catheters.

Continuous techniques

Continuous blocks are usually performed in a similar way than single-shot

techniques with the addition of a catheter that provides the means to continuously deliver

the analgesic solution. Single-shot blocks (“primary block”) are generally associated with

Page 157: Manual Regional Carlo 2010

157 | P a g e

a high success rate. Catheters techniques (“secondary block”) do not generally achieve

the same degree of success. Catheters need to be closely placed in the proximity of target

nerve(s) in order to decrease the “secondary block failure”, a failure to achieve the same

degree of success than single shot techniques. In general catheters should not be

advanced more than 3-4 cm because the risks for catheter-related complications (e.g.,

knotting, vascular puncture, nerve injury, etc) potentially increase.

Stimulating versus non-stimulating catheters

There are proponents of both techniques. The non-stimulating catheters are

commonly inserted through an insulated, Tuohy type needle. The catheter can be a single

orifice catheter in which the hole is usually at the tip, or most commonly a multiorifice

catheter with a dead end (no hole at the tip) and three side holes, the distal one at about

0.5 cm from the tip. The proximal hole is separated from the distal one by a distance of

about 1 cm. After the needle is positioned the catheter is advanced to the desired location.

The technique is generally easy, but the success of the secondary block (through the

catheter) depends on a good perineural placement of the catheter.

The stimulating catheter uses for insertion a similar Tuohy type needle, but the

catheter itself has a wire connected to its tip, allowing for stimulation through it in a

similar fashion than through a needle. The ability to stimulate a nerve as the catheter is

advanced provides a measure of catheter tip-nerve proximity. If the elicited twitch

disappears the catheter is carefully withdrawn into the housing of the needle to avoid

cutting or otherwise damaging the catheter. The position of needle is then slightly

modified by rotation or by moving it in and out a few millimeters and a new attempt is

made. The needle and catheter together as a unit can be slightly rotated in its main axis

before reinserting the catheter. This technique can be more time consuming and more

difficult, but supposedly decreases secondary failure. The introduction of ultrasound into

regional anesthesia practice with its ability to visualize the needle and catheter as well as

the spread of the local anesthetic solution, has called into question the need for

stimulating catheters.

Catheter related problems

The most common problems with catheters include inability to achieve adequate

analgesia and other technical problems like accidental dislodgement and peri-catheter

leaks. Catheters tend to have a “mind of their own”. They can advance away from nerves

and into undesirable places. Capdevila et al in 2005 in a multicenter study that included

1,416 patients identified 17.9 % of “technical problems due to catheters and devices”.

Many techniques are used to increase the resistance to accidental dislodgement.

Perhaps the most successful is the subcutaneous tunnelization of the catheter. It does not

only increase the resistance to removal but also provides the opportunity to direct the

catheter away from the surgical site.

Severe nerve damage and infection are rare complications of continuous

techniques.

Page 158: Manual Regional Carlo 2010

158 | P a g e

References

1. Ansbro FP. A method of continuous brachial plexus block. Am J Surg 1946; 71: 716-

722

2. Selander D. Catheter technique in axillary plexus block. Acta Anaesthesiol Scand

1977; 21: 324-329

3. Liu SS, Salinas FV. Continuous plexus and peripheral nerve blocks for postoperative

analgesia. Anesth Analg 2003; 96: 263-272

4. Boezaart AP: Continuos Peripheral Nerve Blocks, In: Boezaart AP (ed): Anesthesia

and Orthopaedic Surgery. New York, McGraw-Hill, 2006, pp 257-264

5. Capdevila X, Pirat P, Bringuier S, et al. Continuous peripheral nerve blocks in

hospital wards after orthopedic surgery: A multicenter prospective analysis of the

quality of postoperative analgesia in 1,416 patients. Anesthesiology 2005; 103: 1035-

1045

Page 159: Manual Regional Carlo 2010

159 | P a g e

CHAPTER 9

OTHER NERVE BLOCKS

Page 160: Manual Regional Carlo 2010

160 | P a g e

TRANS ABDOMINAL PLANE (TAP) BLOCK

The innervation of the anterolateral abdominal wall is provided by the lower six

thoracic (intercostal) nerves and the first lumbar nerve. The 7th

intercostal nerve swings

up and terminates around the xiphoid of the sternum in the highest point of the abdominal

wall. The 10th

intercostal runs almost horizontally toward the umbilicus, while the 12th

intercostal (subcostal) innervates the area above the inguinal ligament and suprapubic

area. The first lumbar nerve originates the iliohypogastric and ilioinguinal nerves, both

branches of the lumbar plexus, which run above the iliac crest.

Main characteristics

This block was described before the use of ultrasound. It was performed in the

posterior abdominal wall at the level of the triangle of Petit. This triangle is formed

anteriorly by the posterior border of the external oblique muscle, posteriorly by the

anterior border of latissimus dorsi and inferiorly by the iliac crest. The area of the triangle

is covered superficially by the aponeurosis of insertion of the external oblique and deeper

by the external oblique and transversus abdominis muscles. The performance of this

block used to require the feeling of “pop” sensations as the needle crossed the different

fascia and muscle planes. Since the introduction of ultrasound I recommend doing this

block under direct vision.

Indications

To produce anesthesia or analgesia of the abdominal wall.

Point of contact with the nerves

The needle approaches the thoraco abdominal nerves as they travel between the

transversus abdominis (deep) and the internal oblique (superficial) muscles at the level of

the mid axillary line.

Patient position

The patient can be supine or in lateral position with the arm on the side to be

blocked elevated and turned to the opposite side.

Type of needle

A 5cm, 22-G, insulated needle is usually used. An 18-G epidural needle can also

been used with the advantage that the bigger needle is more readily seen and its curved

end could help minimize the accidental puncture of deeper planes.

Type of transducer

A linear high frequency (8-15 MHz) probe is usually sufficient. In larger patients

with thicker abdominal wall (more fat) a curved, low frequency (3-7 MHz) probe is

necessary.

Scanning

The probe is placed diagonally over the lateral abdominal wall at the level of the

mid axillary line.

Page 161: Manual Regional Carlo 2010

161 | P a g e

Needle insertion

The needle can be inserted in plane or out of plane. We prefer to insert the needle

in plane from anterior to posterior, as shown in figure 9-1.

The planes observed at this level are shown in figure 9-2.

Local anesthetic and volume

A volume of 15 to 20 mL on one or bilateral is used according to the incision site.

This block produces somatic pain relief (abdominal wall incision) but it does relieve

visceral pain. For analgesia 0.2% ropivacaine plus epinephrine is frequently used.

Fig 9-1. Needle insertion, in

plane. The needle is inserted in

plane, from anterior to posterior.

(On a model with permission).

Fig 9-2. TAP block. With the probe placed

diagonally over the lateral abdominal wall,

the external oblique (EXT), internal oblique

INT) and transversus abdominis (TRA) are

easily distinguished. The arrows show the

fascial plane between the internal oblique

and transversus muscles where the injection

is performed. Author’s archive.

Page 162: Manual Regional Carlo 2010

162 | P a g e

References

1. Snell RS: Clinical anatomy for medical students, 5th

edition. Boston, MA: Little,

Brown and Company; 1986, pp 133-182

2. Rafi A. Abdominal field block: A new approach via the lumbar triangle.

Anaesthesia 2001; 56: 1024-26.

3. Hebbard P, Fujiwara Y, Shibata Y, Royse C. Ultrasound-guided transversus

abdominis plane (TAP) block. Anaesthesia and Intensive Care 2007; 35: 616-7.

4. Carney J, McDonnell JG, Ochana A, et al. The transversus abdominis plane block

provides effective postoperative analgesia in patients undergoing total abdominal

hysterectomy. Anesth Analg 2008; 107:2056-60

5. McDonnell JG, Curley G, Carney J, et al. The analgesic efficacy of transversus

abdominis plane block after cesarean delivery: A randomized controlled trial.

Anesth Analg 2008; 106:186–91