Liver Flashcards

1
Q

What are some functions of the liver?

A
  • 2-2.5% of TBW – largest organ/gland in body
    • Adults: 600-1800g average wt (smaller in women)
      • 5% neonates (150-170g)

Multiple life sustaining functions

    1. Filtration and storage of blood
      * Serves as a blood reservoir.
      * Kupfer cells within the liver sinusoids avidly remove bacteria and other substances from the portal blood.
    1. Metabolism of carbohydrates, proteins, fats, hormones, foreign chemicals
      * Critical energy metabolism
    1. Formation of bile
      * Critical for emulsification of fats
      * Increase surface area for absorption
    1. Storage of vitamins and iron
      * Ex: Vit K → formation of coag factors
    1. Formation of coagulation factors
      * Failure of these fx contributes to intraop/postop hypoperfusion, tissue ischemia and activation of systemic inflammatory response – progression to MODS.
  • Alters, excretes and modifies numerous gut derived substances.
  • Most vascular organ in the body ~30% CO
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is the difference in liver anatomy when looking at it via the surface anatomy vs physiologic anatomy?

A

Surface anatomy - four distinct topographical lobes: (in picture)

  • right
  • left
    • falciform ligament in between R & L
  • caudate
  • quadrate
  • The surface anatomic description doesn’t correspond to the branches of the liver’s vascular supply and therefore is of limited clinical significance.
    • hexagonal unit on cross section
    • central vein at center of hexagonal cross section
      • drains hepatic vein–> IVC
    • portal canal contains: connective tissue, lymphatics, nerves and portal triad (portal vein, hepatic artery, bile duct)

Physiologic anatomy → 8 functionally independent segments (AKA Couinaud system)

  • Each segment has its own vascular flow and biliary drainage
    • The physiologic segmental division of the liver and is based on divisions of the portal vein, hepatic artery, and biliary ducts.
    • This anatomic arrangement facilitates limited segmental resection of the liver w/ relatively bloodless surgical dissection along the planes between the segments and thereby prevents major disruption of hepatobiliary function.
    • Couinaud anatomy simplifies efforts to preserve healthy tissue and extirpate diseased regions, and has resulted in improved clinical outcomes for patients undergoing hepatic surgery for neoplasms or trauma-induced injuries
  • In this model, the liver forms around vertical axis (portal canal) with
    • hepatic arteriole,
    • portal venule,
    • bile ductule,
    • lymph vessels,
    • nerves
  • Blood flows vertical to portal triads directed toward central veins
  • as these vessels terminate, it forms the zones of the liver (Zone ,1, 2, 3)
  • terminal vein is at periphery of acinus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe the various zones of the liver in the acinus lobule concept.

A
  • Acinus
    • functional microvascular unit of the liver
    • Forms around a vertical axis (the portal canal
    • Consists:
      • hepatic arteriole
      • a portal venule
      • a bile ductule
      • lymph vessels
      • nerves
  • Blood flow vertical to the portal triads and directed radially toward the central veins.
    • Center of Acinus = terminal vascular supply
    • As terminate → form ZONES
      • Zone 1: Most O2 rich blood (center)- periportal zone, gets first dibs on O2
      • Zone 2: further away
      • Zone 3: receives BF after 2 other regions
  • New data suggests the “liver architecture more closely resembles the classic lobule more than it does the acinus.”
  • Blood enters the center of the acinus and flows centrifugally to the hepatic venules. Bile flows in the opposite direction. The simple liver acinus lies between two or more terminal hepatic venules.
  • The difference between lobule and acinus is that the terminal vein is at the center of the lobule and the terminal vein is at the periphery of the acinus.
  • The conceptual advantage of the acinus concept is that the blood supply and biliary drainage of a portion of parenchyma reside in the same portal tract, whereas multiple portal vein branches and arteries supply each classic lobule
  • Hepatocytes 75-80% liver volume
  • Zone 1 (periportal zone) – HIGHLY METABOLICALLY ACTIVE
    • Cells are closest to the portal axis
    • receive blood that is rich in O2 and nutrients
      • Major site of:
        • oxidative metabolism
          • aerobic metabolism
          • highest Kreb cycle enzymes/highest number mitochondria (gluconeogenesis, b-oxidation fatty acids, amino acid catabolism, bile acid secretion)
        • conversion ammonia to urea*
    • **Most prone to hepatic reperfusion injury*
  • Zone 2 (midzonal region)
    • arbitrary intermediary transition zone
    • “anatomic reserve”- overlap with zone 1
  • Zone 3 (pericentral) cells at margin of acinus
    • receive blood that has exchanged gases and metabolites with cells in zones 1 & 2
      • → least resistant to metabolic and anoxic damage (most sensitive)
      • Lowest O2 saturation
      • Major site of:
        • CYP450
        • anaerobic metabolism– high quantity of ER– (glycolysis and lipogenesis)
        • general detox and biotransformation drugs, chemicals, toxins) These cells exquisitely susceptible to injury from systemic hypo-perfusion and hypoxemia – zone 3 cell necrosis is characteristic of acetaminophen/halothane toxicity
    • **most prone to ischemic damage**
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the innervation of the liver?

A
  • Stimulation of SNS fibers post-ganglionic T3-T11
    • increases hepatic vascular resistance (decreased blood volume) → release blood into central circulation (referred to earlier as liver storing blood)
      • ​autotransfuion of up to 80% hepatic blood (400-500 mL) if needed in SNS stimulation
    • increases glycogenolysis and gluconeogenesis (increased BG)
      • (catabolic/mobilization of energy mode)
  • Stimulation of PSNS (vagus nerve)
    • increases glucose uptake and glycogen synthesis
      • (anabolic storage mode)
  • Branches of the splanchnic nerves (postganglionic sympathetic fibers from T6-11), vagus nerve, and phrenic nerve enter the liver w/ the major blood vessels and bile ducts. These nerve fibers form an intercommunicating plexus, w/ synapses on the terminal arterioles and venules.
  • Sympathetic innervation of the hepatic and splanchnic vasculature plays a major role in regulating the volume of whole blood stored in, and expelled from, the hepatic reservoir. Studies in canine model = sympathoadrenal stimulation (e.g., hypercarbia, pain, hypoxia) can abruptly decrease hepatic blood flow and splanchnic vascular capacitance. Within seconds, splanchnic nerve stimulation can autotransfuse up to 80% of the hepatic blood (400–500 mL) into the central circulation. Other studies = vagal stimulation alters the tone of presinusoidal sphincters and influences blood flow distribution within the liver rather than total hepatic blood flow.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are some characteristics of the hepatic vascular system?

A
  • 25-30% of CO = 1350ml/min
  1. Portal Vein = 1050 ml/min
    1. BF: 70-75%
    2. Oxygenation: 50%
      • Blood drained from GI tract into here
        • formed by confluence of splenic and superior mesenteric veins and receives blood from GI tract, spleen, pancreas and gallbladder
        • portal vein has numerous tributaries of little importance until portal HTN present, and then these connections form large portosystemic shunts which allow venous blood flow to bypass liver and produce pathologies (ie esophageal varices)
  2. Hepatic Artery= 300 ml/min
    1. BF: 25%
      • Hepatic artery arises from the celiac trunk in 80% of the population, the rest the superior mesenteric artery
    2. Oxygenation: 50%
      1. Branch off celiac artery (Also supplies rest of GI tract)
      2. 100% oxygenation going through even though less BF
  • Portal vein pressure = 9mmHg
  • Hepatic vein leading to vena cava = 0mmHg
    • Normal Physiology:
      • HIGH flow
      • LOW resistance/pressure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What regulates hepatic blood flow?

A
  • Hepatic arterioles have a myogenic response to stretching keeps local blood flow constant, despite changes in BP
    • Increase in transmural pressure (BP) → vasoconstriction (preventing elevations of local BF)
    • Decrease in transmural pressure (Bp)→ vasodilation (preserving perfusion during systemic hypotension)
    • don’t see this same regulation in portal vein!!
    • Considerations:
      • Auto regulation of hepatic artery is present in metabolically active liver (postprandial hyperosmolarity)
        • usually absent in fasted state → most OR patients are NPO
      • VA dose-dependently decrease response
      • Pressure-flow auto regulation does not exist in the portal circulation
      • Drop in BP → directly influence hepatic BF
        • Liver sensitive to hypoTN episodes
      • Although the liver receives 25% of CO, regional blood flow within the organ is such that certain areas are highly prone to ischemia. Intrinsic (regional microvascular) and extrinsic (neural and hormonal). Also decreases in ph or O2 and inc in CO2 of the portal blood promote increases in hepatic artery flow
  • Hepatic arterial buffer response
    • change in portal venous flow causes a reciprocal change in heaptic arterial blood flow
    • as protal blood flow decreases, adenosine builds up, transfers to hepatic arterial wall causing a DILATION, which increases hepatic arterial blood flow
    • as portal venous flow increases, this washes out the adenosine in the periportal regional and causes arteriolar resistance to rise, lowering hepatic arterial blood flow back to normal
  • Extrinsic influences
    1. tone of pre-portal splanchnic organs regulate portal vein flow
    2. decrease in pH or increase PaCO2 (acidosis) causes hepatic arterial flow to increase
    3. postprandial hyperosmolarity
      • increases both hepatic arterila and portal venous flow
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the humoral influences on portal circulation in the hepatic arteries vs portal veins and what are the effects of common vasopressors and inotropes on hepatic blood flow?

A
  • Hepatic arterial bed
    • α1 -, α2 -, and β2 -adrenergic receptors
  • Portal vein
    • only α-receptors

Vasopressin

  • intensely constricts splanchnic arterial bed- distributes BF out and into central system
    • Ex: tx varices → dec BF going to liver and therefore decreasing the blood flow bypassing liver and going to varices
  • Lowers portal venous resistance
  • effective treatment for portal hypertension/esophageal varices*

Epinephrine

  • Epinephrine (all equal efficacy in receptors)
    • Arterial side: initial vasoconstriction (α-receptor), followed by vasodilation (β-receptor)
  • More balanced effect of epi on arterial beds vs vein because vein only has alpha receptor (missing Beta-2 dilation)
  • Portal Vein: only vasoconstriction (α-receptor)
  • Drop in portal vein flow

DOpamine

  • vasoactive effects weak compared w/ epi and norepi

Glucagon

  • dose-dependent relaxation of hepatic arterial smooth muscle
  • antagonizes vasoconstrictor responses of the hepatic artery to various physiologic stimuli-including increases in SNS tone (vasodilation)
  • ex: liver dx → liver not metabolizing glucagon → excessive levels of glucagon → wide spread dilation)–> causes hyperdynamic circulation seen in liver failure

ANG II

  • severely constricts hepatic arterial & portal venous beds, & markedly ↓ both mesenteric & portal venous flow
  • blood flow to liver may plummet​
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Describe how the liver is considered a blood reservoir?

A
  • Expandable organ
    • Hepatic arteries, veins and capillaries contain 450 ml blood = 10-15% TBV
      • a large, expandable venous organ capable of acting as a valuable blood reservoir in times of excess blood volume and capable of supplying extra blood in times of diminished blood volume.
    • ~20% of blood is in arteries, 10% is in capillaries, and 70% is in veins
  • Translocation of blood:
    • With increased right heart failure/ increased right atrial pressure liver
      • accommodate an extra liter of blood (pushes blood back to liver)
    • Intense SNS stimulation (pain, hypoxia, hypercarbia) can abruptly decrease hepatic blood flow and volume
      • 80% of flow (400-500ml) can be expelled in a matter of seconds (ex: for hemorrhage)
        • Autonomic innervation of the liver coupled w/ the neurohumoral input from the systemic circulation allows for rapid precise control of the reservoir volume.
    • Anesthetics & liver disease impair this response and severe liver dx pts have impaired vasoconstriction as well incapacitating the splanchnic reservoir – prevent redirection of blood flow to heart and brain
    • The normal liver moderates the hypotensive response to acute blood loss and hypovolemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Why is the liver considered an endocrine ordgan?

A
  1. Insulin growth factor – 1- somatomedin
    • mediates actions of hormones from other endocrine glands (bone growth in children)
    • . Insulin-like growth factor promotes systemic growth, especially bone growth in children.
  2. Angiotensinogen - precursor of angiotensin II/ all angiotensin proteins, fluid and electrolyte balance
    • regulates SBP (systemic tone)
    • Regulates water and Na
  3. Thrombopoietin –
    • regulates plt production by stimulating bone marrow precursor cells to differentiate into plt-generating megakaryocytes.
  4. Hepcidin
    • responsible for iron homeostasis and regulates intestinal iron absorption, plasma iron concentrations, and tissue iron distribution by inducing degradation of the hepcidin receptor, ferroportin
  5. Conversion thyroxine (T4) to tri-iodothyronine (T3)
    • T4 (inactive) to T3 (active)
  6. Inactivation of corticosteroids, ADH, aldosterone, estrogen, androgens, insulin,
    • The interaction of altered hormone levels and diminished hepatic synthesis of hormone binding globulins w/ altered metabolism and receptor regulation leads to significant endocrine abnormalities in pts w/ liver disease.
    • Nearly half the insulin produced by the pancreas never reaches the systemic circulation bc it’s degraded during a single passage through the liver.
    • In addition to hormone synthesis, the liver participates in endocrine function by inactivating many hormones, including thyroxine, aldosterone, antidiuretic hormone, estrogens, androgens, and insulin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the coagulation factors formed in the liver?

A
  • All coagulation factors formed in liver
    • Except:
      • vWF
      • VIII
        • VIII is made in both the liver and endothelial cells – often patients have enough even in severe liver disease
      • III (tissue thromboplastin)
      • IV (calcium)
  • Vitamin K dependent: need bile salts to get vit K!
    • Prothrombin/Factor II
    • Factor VII
    • Factor IX
    • Factor X
    • Proteins C and S (anticoagulants)
  • Thrombopoietin → plts
    • Stimulate plt production!
  • Synthesizes Anticoagulant factors
    • antithrombin III, plasminogen activator inhibitor, Proteins C, S, Z, and fibrinolytic factors
      • liver dx: see coagulation issues
  • Clearance of activated coagulation factors including fibrinolysins, TPAs (clearance essential for control of fibrinolytic states)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is liver’s role in the formation of erythrocytes production and breakdown? (very unlikely to be a question?)

A
  • Liver responsible ~ 20% of heme production
    • Used to produce cytochrome P450 enzymes
    • Deficiency in porphobilinogen deaminase (an enzyme in the biosynthetic heme pathway and CYP450 enzymes) → acute intermittent porphyria
      • Consequences: NO BARBITUATES (or etomidate!)
    • The liver is the primary erythropoietic organ of the fetus between the 9th and 24th week of gestation. It continues to be a major site of hematopoiesis until an infant is about 2 months of age. In healthy adults, the liver is responsible for about 20% of heme production; bone marrow makes the rest
  • Metabolism of hgb produces bilirubin
    • Hepatocytes responsible for conjugating bilirubin and releasing it into the bile to be eliminated via alimentary tract

Secretion of bile:

  • Secretes 500 ml/day from common bile duct to duodenum
    • Contains conjugated bile salts, cholesterol, phospholipids, conjugated bilirubins, electrolytes
  • Bile acids
    • help alkalinize & emulsify large fat particles
      • → increase surface area for digestion/aiding absorption
    • excretion of several waste products from blood
      • ex: xenobiotics, bilirubin, calcium, & cholesterol
    • Excretes Compounds >300-500 Daltons = too large for the kidney
  • Considerations:
    • Opioids interfere w/ biliary flow→ increasing pressure in common bile duct or inducing spasm in sphincter of Oddi
      • Mimics angina in awake pt
    • Tx: Antagonize action w/ → (dilate bile duct)
      • VA’s
      • naloxone, nitroglycerin, atropine
      • glucagon *
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is the liver’s role in excreting drugs, hormones and other substances?

A
  • Detoxify or excrete into bile
    • Ex: sulfonamides, penicillin, ampicillin, calcium, & erythromycin
  • Termination of anesthetic effects via transformation by liver
    • Predictable termination of the pharmacologic effects of many anesthetic agents depends on the liver for metabolic biotransformation into inactive products that can be eliminated
  • Metabolize Endocrine gland hormones
    • chemically alter and excreted by liver
      • ex: thyroxine, (steroid hormones) estrogen, cortisol & aldosterone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Impact of advanced liver disease on drug pharmacokinetics? (from ppt notes)

A
  • Significant liver disease – portosystemic shunts allow orally administered drugs to bypass the first pass clearance + decreased liver blood flow= prolonged terminal half-life and increased systemic effects of high extraction drugs.
  • Hypoalbuminia increased free fraction – increased systemic effects, also increases elimination of those with low hepatic extraction ratios.
    • If ascites is present may have an increased Vd.
    • Doses should be decreased 50%
  • It is often difficult in liver disease to predict the pharmacokinetics and pharmacodynamics of drugs.
  • Often, cirrhotic patients with co-existing disease are better served receiving a hepatically cleared agent at reduced doses (careful titration) if it provides superior efficacy and side effects than less hepatically cleared agents.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Whatis liver’s role in the cyp450 system?

A
  • Liver has more than 20 different CYP enzymes
    • Lots of genetic differences in people
      • Up to four fold variation
  • Many of these isozymes contribute to:
    • oxidation of drugs
    • environmental toxins
    • steroid hormones
    • lipids
    • bile acids
  • Hepatocytes of zone 3 have the highest content of CYP proteins
    • Zone 3: most sensitive to ischemic damage
      • Also site of biotransformation → if toxic metabolites produced → impacts zone 3 cells
        • Ex: acetaminophen → produce toxic metabolites (zone 3 most vulnerable)
  • Advanced cirrhosis lowers both total CYP and hepatic perfusion and results in significantly reduced clearances of many substances.

CYP Inducers:

  • Phenobarbital, phenytoin-> induce several different cyp proteins
  • Smoke from cigarettes, cannabis–> induces CYP 1A2
  • Alcohol–> induces CYP2EI, CYP3A4
  • Induces of CYP not only affect own metabolism, but affect drug metabolism and biologic action of many other substances
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Refresh on cyp inducers (in notes)

A
  • Phenobarbital, phenytoin-> induce several different cyp proteins
  • Smoke from cigarettes, cannabis–> induces CYP 1A2
  • Alcohol–> induces CYP2EI, CYP3A4
  • Induces of CYP not only affect own metabolism, but affect drug metabolism and biologic action of many other substances
    • CYP induces activate nuclear orphan receptor transcriptional regulators and can participate in hepatic adaptation to chronic drug admin
    • Genetic and environmental influneces are most important variable affecting drug metabolism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is intrinsic clearance of the liver?

A
  • Reflects fraction of delivered drug load that is metabolized or extracted during a single pass through liver
    • High clearance – hepatic clearance approaches rates at which they transverse the liver
      • Hepatic metabolism/clearance DEPENDS on hepatic BF
        • Ex: decrease hepatic BF → highest impact on drugs listed below
      • Ex: (see Box 22-1 Miller 8
        • Lidocaine
        • Diphenhydramine
        • metoprolol
  • Low clearance - hepatic clearances are relatively independent of hepatic blood flow
    • Aka: capacity dependent elimination
      • Ex: anything changing free fraction of drug effects rate of clearing drug (low albumin [] )
    • Ex:
      • Diazepam
      • Acetaminophen
      • warfarin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are some lab tests that show hepatocellular damage?

A
  • ALT (Serum alanine aminotransfersase)- SELECTIVE
    • Only present in the liver (more selective for LIVER damage)
      • Found primarily in zone 1 hepatocytes (area of oxidative metabolism)
  • AST (Serum aspartate aminotransfersase)
    • Present in a wide variety of tissues (liver, heart, skeletal muscle, brain, heart)
      • Found primarily in zone 1 hepatocytes
        • No prognosis can be done with levels of AST/ALT
  • LDH (lactate dehydrogenase)
    • Nonspecific
  • GST
    • Isoenzyme B found exclusively in liver
      • Specifically ZONE 3
        • Worry about ischemic damage/toxic metabolite damage
    • *Highly sensitive
    • Half-life (60-90 min)- short
      • Track injury evolution
    • Found in all acinar hepatocyte zones
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are lab tests that assess hepatic synthetic function?

A
  • Albumin
    • 1/2 life = 20 days
      • Acute ischemic event → albumin levels wont reflect that
      • Better tracking long-term/chronic fx
    • Protein synthesis = function
      • Low levels → fx impairment
    • Poor specificity
      • Ex: fluid overload, pregnancy → dilutional effect
  • PT/INR
    • CF ½ lives: 4 hrs - 4 days
      • Factor VII- 4 hrs
      • Fibrinogen- 4 days
        • *helpful tracking severe deficits in hepatic fx
    • Issue: Excess CF produced normally → if seeing drop in PT/INR, pt pretty sick (ESLD)
  • Caffeine clearance
    • measure metabolites in saliva
  • Idocyanine green
    • extracted and metabolized exclusively by the liver
        • administer and measure elimination kinetics (plasma disappearance rate/measured transcutaneously)
    • Reflects both hepatic fx and hepatic BF
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What lab tests assess bile flow status?

A
  • Alkaline phosphatase (AP) isoenzymes
    • 2-4X increase → suggests cholestatic disease
    • Levels vary w/:
      • gender, age, blood type
      • increased in CHF, pregnancy, smokers, growth spurts, sepsis, hepatitis, etc.
        • general screen
  • 5’-nucleotidase (5’NT)
    • Helps differentiate between intrahepatic vs extrahepatic issue
  • Gamma glutamyl transferase (GGT)
    • Nonspecific
  • Serum bilirubin
    • HELPFUL → Reflects hepatic excretion capability
      • Conjugated levels
        • High levels: →
          • Mechanical outflow obstruction (→ jaundice)
      • Unconjugated levels:
        • High levels → indicates difficulty w/ liver conjugating (2)
          • High Hgb metabolism (Ex: hematoma or several blood tx → lots of bilirubin to conjugate)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the common causes of viral hepatitis?

A
  • Five types – similar clinical laboratory features
  • Hepatitis- inflammation of liver
  • Varied presentation among individual patients: asymptomatic VS influenza-like symptoms VS jaundice and acute hepatic failure
  1. Hepatitis A
    1. Transmission: Fecal/oral route- contamination of food
      1. Lasts ~21 days → fully recover
  2. Hepatitis B (50% US cases)
    1. Transmission: Blood/sexual intercourse
    2. Asymptomatic
  3. Hepatitis C (30% US cases)
    1. Transmission: Blood/sexual intercourse
    2. …. 50-85% progress to chronic disease with 5-25% risk of developing cirrhosis – leading cause of transplant. New medications are decreasing viral load and “curing” patients.
  4. Hepatitis D (20% US cases)
    1. Transmission: Blood/sexual intercourse
    2. RNA strand requiring co-infection w/ Hep B
  5. Hepatitis E (rare)
    1. Fecal/oral – similar to A
    2. Foundin asia, Africa, central america
  6. Miscellaneous Causes – CMV, Epstein Barr, Herpes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Describe the pathophysiology of halothane hepatitis including the classic presentation and risk factors?

A
  • All VA (except Sevo) metabolized by Cytochrome P450 2EI
    • oxidizes each anesthetic to yield highly reactive intermediatescovalently bind (acetylation) to hepatocytes (variety of hepatocellular macromolecules)
      • Immune system recognizes the intermediary and hepatocyte as FOREIGN protein → ATTACKS
    • Altered hepatic proteins trigger an immunologic response → causes massive hepatic necrosis
  • VA metabolism refresher (they would probably want to hear this?)
    • Halothane 46% metabolized
    • Enflurane 2.5-8.5% metabolized
    • Sevoflurane 2-5% metabolized
      • Not metabolized into intermediate (0% risk halothane hepatitis)
    • Isoflurane 0.2-2% metabolized
    • Desflurane 0.02% metabolized
      • Halothane gone → less and less risk of developing response
  • Classic presentation of VA associated hepatitis:
    • fever, anorexia, nausea, chills, myalgias, rash,
    • arthralgia (joint pain)
    • eosinophilia
    • jaundice (3-6 days later)
      • overt jaundice indicates severe disease and may indicate mortality rate of 40%
    • 2 types-
      • ​can be mild with small elevation AST/ALT
      • Second type is fulminant form known as halothane hepatitis. elevated AST/ALT/bili/AP/massive necrosis
  • Risk factors:
    • PRIOR EXPOSURE!
      • multiple brief procedures within brief duration of time
    • age >40 years old
    • obesity, female gender
    • Mexican ethnicity (chromosomal vulnerability)
    • enzyme induction (ex: pt on barbs/phenytonin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the effect of our anesthetic gases on hepatic blood flow?

A

VA

  • Hepatic BF and oxygenation decrease w/ VA
    • (Halothane > Enflurane > Desflurane > Isoflurane > Sevoflurane) → sevo reduces BF the LEAST
    • Portal BF reduced but hepatic arterial flow is maintained w/ des/iso/sevo
  • Decreased CO and stress response → catecholamine-induced vasoconstriction
    • Catecholamines preferentially vasoconstrict splanchnic circulation (normally feeds portal vein) → no BF to portal vein
      • Catecholamines improve BP but don’t help portal vein
  • Metabolic demands do decrease improving the supply-demand balance a bit

N2O

  • Increase SNS activity
    • Consequences:
      • mild vasoconstriction of splanchnic vasculature → decreased portal flow
      • increase flow through hepatic arterial system
  • Inhibition of methionine synthase activity
    • prolonged exposure → B12 deficiency
  • No concrete evidence it causes hepatic toxicity if O2 supply/demand normal
    • Dental providers 7 fold risk for liver dx
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Nitrous Oxide’s effect on HBF and O2 delivery?

A
  • Increase SNS activity
    • Consequences:
      • mild vasoconstriction of splanchnic vasculature → decreased portal flow
      • increase flow through hepatic arterial system
  • Inhibition of methionine synthase activity
    • prolonged exposure → B12 deficiency
  • No concrete evidence it causes hepatic toxicity if O2 supply/demand normal
    • Dental providers 7 fold risk for liver dx
24
Q

Spinal and epidural anesthesia effect on HBF?

A
  • Hepatic blood flow → decreases
    • appears to parallel reductions in systemic BP
  • Reduced PBF and unchanged HABF → cause decrease in THBF
  • Changes may not be reliably reversed & w/administration of vasopressors to maintain normal arterial BP (vasoconstriction of splenic vasculature)
    • Avoid high blocks > T5 (preserve O2 S/D to liver)
25
Q

What is nonalcoholic fatty liver disease?

A
  • Most common US chronic liver disease ~30% of the population
    • “hepatic manifestation of metabolic syndrome”
    • a/w:
      • DMII
      • obesity
    • Bariatric surgery often results in improvement
  • Dx: Liver fat accumulation >5% of weight
    • May have increased M& M with abdominal procedures
  • Spectrum of pathology from asymptomatic to cirrhosis (steatohepatitis 3-5%)
26
Q

What are the forms of alcoholic liver disease?

A
  • 3 forms of alcohol related liver disease:
  1. Steatosis- beginning
    1. Ingestion of moderate-large ETOH in short time
      1. s/s: malaise, N/V, anorexia, weakness, abd discomfort, mild LFT alterations
      2. Usually benign and resolves when drinking stopped
  2. Alcoholic hepatitis
    1. s/s: same as steatosis + fever & jaundice
    2. Altered LFTs, coags, hypoalbuminemia
      1. Tx: ETOH abstinence, high protein diet
  3. Cirrhosis
    1. chronic inflammation/necrosis causes fibrotic changes in liver, changing vasculature, making it more torturous and increasing resistance through liver
    2. Tx: liver transplant
  • Alcohol abusers → 2-3 fold increase peri-op morbidity
27
Q

What is the pathophysiology of cirrhosis?

A
  • Parenchymal liver disease
  • Most common etiologies:
    • alcoholism
    • steatohepatitis
    • chronic HCV
  • Chronic inflammation/necrosis result in fibrotic changes (via hepatic stellate cells?)
    • Changes vascular architecture → increase hepatic resistance
    • S/S: loss of liver fx
      • Anorexia
      • Weakness
      • N&V
      • abdominal pain
    • Other s/s: Hepatosplenomegaly, ascites, varices, jaundice, spider nevi, metabolic encephalopathy
  • Every organ and body system altered
  • End-stage Cirrhosis Hallmark = portal HTN
    • r/t
      • fibrosis and mechanical obstruction of vasculature +
      • hepatic endothelial dysfunction
        • less NO (typically vasodilator → reduces resistance)
        • more thromboxane (vasoconstrictor → increases resistance)
  • Portal HTN results in formation of portosystemic collaterals (varices)
    • develop by dilation and hypertrophy of existing vascular channels → potential for rupture/bleed because those vasculatures aren’t used to the higher flow of blood
  • Decompensated cirrhosis
    • Ascites
    • hepatic encephalopathy
    • esophageal varices
    • enhanced susceptibility to bacterial infections (Kupffer cells not filtering because blood is bypassing liver through protosystemic collaterals)
    • altered drug metabolism
28
Q

What are some CV abnormalities in cirrhosis?

A
  • Hyperdynamic circulation:
    • High CO
    • low SVR
    • low normal BP
    • normal/increased SV
    • normal filling pressures
    • high/normal HR
      • Systolic and diastolic dysfunction correlates with degree of liver disease
    • Prolonged QT 30-60% end stage patients
      • AVOID other meds prolonging QT: Erythromycin, Droperidol, Zofran
  • Increased TBV with decreased central “effective” volume
  • Hypervolemic splanchnic (extrahepatic) circulation
  • Extensive arteriovenous (AV) collateraliziation
    • BF directly going through artery → vein w/o intervening capillary bed
  • Decreased response to catecholamines
    • High [] circulating endogenous vasodilators
      • Ex: high glucagon, intestinal peptide, NO levels since not being metabolized
        • a lot of blood volume and big dilated vessels, difficult to maintain bp
29
Q

What are esophageal varices? treatment?

A
  • Portosystemic collaterals formed after preexisting vascular channels dilated by portal HTN
    • Hemizygous veins offer lower resistance path to liver → BLEED
      • CAUTION: Nearly 1/3 of initial bleeding episodes are fatal
  • Tx:
    • Non-selective B-blockers (propranolol and nadolol)
      • reduce risk of bleeding by 40-50%
    • Acute/suspected acute bleed:
      • IV somatostatin
      • IV synthetic somatostatin- octreotide
        • MOA: Decrease vasoactive mediator release from GI tract
          • ex: dec glucagon release → less dilation of splenetic vasculature → less BF through → less BF to varices
        • Stop 80% GI bleed by reducing portal pressure
    • Endoscopic band ligation/scelrotherapy
      • if not tolerate medical therapy
      • 90% effective in stopping active bleeding
30
Q

What are some pulmonary complications seen in cirrhosis?

A

Pulmonary implications= hypoxemia

  • Hepatopulmonary syndrome – progressive hypoxemia
    • Intrapulmonary shunting → vascular dilations
      • Type I: precapillary – close to alveoli
        • Oxygenation improves with O2 in this type
      • Type II: arteriovenous- dilation resulting in large AV communications that may/may not be near gas exchange units
        • Alveolar to arterial gradient > 15 mmHg
      • Often causes development of orthodexia- SOB int sanding position because dilation is worse at the base of the lung
  • VQ mismatch
    • impaired hypoxic pulmonary vasoconstriction
    • pleural effusions
    • ascites
    • diaphragm dysfunction
  • Right shift of oxyhemoglobin dissociation curve
    • increased erythrocyte 2,3-diphosphoglycerate levels
  • Altered pulmonary diffusion capacity (decreased)- impairing ability to oxygenate
    • secondary to increased ECF
    • interstitial pneumonitis
    • porto-pulmonary HTN (2% of cirrhotic patients)
  • Hepatic hydrothorax (pleural effusions in absence of CV dx)
    • ascites + defects in the diaphragm
  • Pulmonary arterial hypertension (5% of patients with portal HTN – poor prognosis/unclear mechanism)
    • Aka portopulmonary HTN?
      • PAP >25, normal wedge pressure, elevated PVR >120 dyne/second)<
31
Q

What are the current theories why cirrhosis patients retain sodium which contributes to ascites development?

A
  • overflow theory“— Primary renal tubular retention of Na as a result of portal HTN
    • hepatorenal reflex→ increased plasma volume results, and then ascites develops as fluid is translocated out of splanchnic circulation
  • underfill theory“—simply reflects the normal homeostatic response to intravascular volume depletion (sodium and water retention)
    • Decrease in effective volume because of ascites/portal HTN– renal tubular reabsorption of sodium, increased ECF volume (and hypoalbuminemia causing low oncotic pressure) = increased ascites and edema
  • “peripheral arterial vasodilation hypothesis” - portal HTN causes the production of mediators such as nitric oxide cause vasodilation & enlargement of the intravascular compartment decreasing the effective volume
    • baroreceptors activated = SNS, renin-angiotensin- aldosterone and vasopressin release = Na and water retention
32
Q

Pathophys for cirrhosis-induced portal HTN?

A

. Cirrhosis and portal HTN induce circulatory changes that decrease the effective blood volume.

→ activates volume receptors and stimulates neurohumoral and intrarenal reflexes to decrease renal blood flow and to increase renal retention of sodium

  • vicious cycle, all leads to retaining more and more fluid, ascites getting worse, and risk of variceal bleeding increasing

Schematic of pathways for cirrhosis-induced portal hypertension:

  • the forward and backward theories.
  • Cirrhosis and portal hypertension induce circulatory changes that decrease the effective blood volume.
    • This activates volume receptors and stimulates neurohumoral and intrarenal reflexes to decrease renal blood flow and
    • increase renal retention of sodium.

PVBF, portal venous blood flow; HABF, hepatic arterial blood flow; THBF, total hepatic blood flow; A-V, arteriovenous; PG’s, prostaglandins; ADH, antidiuretic hormone; ANF, atrial natriuretic factor; PAF, platelet activating factor.

33
Q

What is hepatorenal syndrome?

A
  • Prerenal failure characterized by:
    • intense vasoconstriction of renal circulation
    • Low GFR
    • Preserved renal tubular function
    • normal renal histology
  • Type 1: Poor Prognosis (tx list quickly)
    • progressive oliguria
    • rapid rise in serum creatinine
  • Type 2
    • moderate more stable impairment in renal function
    • usually seen in patients with refractory ascites
34
Q

Treatment for hepatorenal syndrome?

A
  • Tx: aimed at reversing pathophysiological cause → splanchnic arterial vasodilation
  1. Vasoconstrictor therapy → increase renal perfusion/BP
    1. Vasopressin (AVP)
    2. NE
  2. Octreotide + midodrine (alpha-1 agonist)
    1. Octreotide: Synthetic somatostatin → decrease release of vasodilators (glucagon, VIP) → splanchnic vessels constrict/renal perf improve
    2. Midodrine (like neo)
      1. → splanchnic vessels constrict/renal perf improve
  3. Albumin + norepinephrine infusion
  4. Transplant usually curative
35
Q

What are some hematologic and coagulation implication of cirrhosis?

A
  • Anemia: may be d/t:
    • plasma volume expansion (dilution)
    • gastrointestinal bleeding
    • malnutrition/vitamin deficiencies
    • hemolysis
    • hypersplenism
    • bone marrow depression
  • Synthesis of vitamin K-dependent factors
    • DECREASE: II, VII, IX, X and anticoag proteins C and S.
      • Coagulopathies result → Factor VII level decrease by 60% - 70% before PT becomes prolonged
        • Ex: High PT/INR -→ SEVERE liver fx deficit (because we always make more coag factors than we need)
  • Thrombocytopenia (30-60% of patients) and thrombopathy
    • splenic sequestration syndrome (90% plts may be sequestered)
    • bone marrow suppression (esp with ETOH, interferon or other meds)
    • immune-mediated platelet destruction (platelet-associated IgG)
  • Dysfibrinogenemia (activation of fibrinolysis)
    • abnormal fibrinogen functioning and increases in fibrin degradation products and D-dimer occur.
    • The patient may have prolonged thrombin time with a normal to slightly prolonged PT,PTT, and “nml” fibrinogen levels and still have coagulation problems
  • Difficulty clotting WITH coagulopathic issues!
36
Q

What is the cause of hepatic encephalopathy in cirrhosis?

A
  • Reflects decompensated cirrhosis
    • d/t excessive ammonia (not converting ammonia to urea)
    • Pathogenesis- decreased hepatocellular function, reduced hepatic blood flow, and diversion of portal flow through extrahepatic collateral vessels
  • Complex, reversible w/highly variable clinical manifestations
    • ranging from minimal personality changes to confusion lethargy, somnolence & coma (Graded I-IV (coma))
      • Neuropsychologic (LOC/affect)
      • Neuromotor symptoms (hyperreflexiveness/nystagmus/ posturing)
      • Ammonia accumulation
      • inappropriate levels of inhibitory and/or excitatory neurotransmitters
        • glutamate/glutamine can contribute to excitotoxicity and cerebral edema
        • GABA and octopamine increase inhibitory outflow
  • 50-70% of cirrhotic patients have at least minimal encephalopathy
  • Increased ammonia production
    • excessive dietary protein
    • constipation
    • GI bleed
    • Infection
    • Azotemia
      • Tx: restrict protein
  • Factors generating Ammonia:
    • Fluid, electrolyte, and acid-base imbalance generate ammonia
    • Surgical stresses, diarrhea, vomiting, diuretics, paracentesis, general anesthetic related reductions in hepatic perfusion
      • AVOID these things
  • Altered liver and brain function r/t encephalopathy
    • Worsening factors → hypoxia, hypotension, anemia, hypoglycemia, sedation/hypnotics
  • Reduced hepatic metabolism – creation of portal-systemic shunt
37
Q

S/S of cholestatic disease?

A
  • Pruritus (early)
  • jaundice (later)
  • lighter colored stool
  • dark urine (bile pigments are diverted from the gastrointestinal tract to the kidney for excretion)
38
Q

CV dysfunction in cholestatic disease?

A
  • CV dysfunction very common:
    • circulating bile salts impair myocardial contractility
    • blunts response to:
      • norepi–> endogenous and exogenous
      • angiotensin II
      • isoproterenol
    • decreased SVR (vasodilation)
    • increased CO
    • increased portal venous pressure
    • decreased portal venous flow
      • Sensitive to extreme blood loss
39
Q

What would you look for on assessment of patient with liver dx?

A
  • Easy bruising?
  • Anorexia or weight changes
  • N&V or pain with fatty meals
  • Pruritus or fatigue
  • Abdominal distention/ascites
  • GI Bleeding
  • Scleral Icterus- yellowing of eyes
  • Hepatomegaly or splenomegaly
  • Palmer erythema
  • Gynecomastia (dec hormone met)
  • Asterixis- hand tremor
  • Spider angiomata, petechiae, and ecchymosis
40
Q

Preop H&P considerations for pt with liver disease?

A

If history of jaundice or abnormal lab (LFTs) results determine relationship to:

  • Prior surgical or anesthetic technique
    • Ex: prior halothane use in sx ?– may want to use sevo
  • Blood transfusions
    • Excessive blood tx/hematoma healing → large bilirubin → jaundice can occur
  • ETOH or recreational drug use
  • Sexual history (viral)
  • Current medications and herbal meds
    • Ex: Statins, tylenol, antifungals
    • Herbals not regulated
  • Family hx of jaundice/liver disease
    • Alpha-1 antitrypsin deficiency
      • Alpha 1 antitrypsin – enzyme helps protect lungs
      • Deficiency – pts develop dysfx/misshaped globule unable to exit liver → liver damage
    • Wilson’s disease
      • Buildup copper in liver → neurologic consequences
  • Travel history
    • Hep E- Asia/Africa
  • Occupational history/exposure to environmental toxins

If H&P does not reveal S&S of liver disease → LFT’s/laboratory work not indicated

  • Consider pts age
  • coexisting disease
  • type, location, and duration of surgery
  • If known or suspected liver disease use scoring systems: (2)
    • Child-Pugh
    • MELD
41
Q

What can be done to optimize patients with liver disease preop?

Additional considerations that may alter anesthetic plan?

A
  • Optimization” correction of:
    • ETOH dependency, coagulopathy, pH, electrolyte abnormalities (esp. K+) malnutrition, anemia
    • esophageal varices- careful NG/TEE
    • hepatic encephalopathy- address diet/dec N/V
    • PT or INR
      • (few days)parental vitamin K
      • recombinant factor VII, etc.
      • FFP in emergency
    • Plt infusion
      • Transfusion: < 100,000 cells/microliter
  • Assume full stomach (ascites, decreased gastric and intestinal motility):
    • Tx: H-2 receptor blocker, metoclopramide and sodium citrate
  • Sedative pre-medication – titrate to effect, altered pharmacodynamics and pharmacokinetics
    • VOD- altered
42
Q

IV anesthetic consideraitons in liver dysfunction? (Benzo, preceded, propofol?)

A
  • Benzodiazepines
    • increased cerebral uptake
    • decreased clearance
    • prolonged E1/2 life
  • Dexmedetomidine-
    • decreased clearance
    • prolonged ½ life (reduce dose)
      • CAUTION- already hypoTN
  • Propofol
    • single dose similar response as normal patients
    • Longer recovery times after infusions
      • *drug of choice w/ encephalopathy **
  • E1/2 unchanged in most studies
    • TPL
    • Etomidate
    • Ketamine
    • methohexital
43
Q

Considerations for opioids in liver dx?

morphine

meperidine

fentanyl

sufentanil

alfentanil

remifentanil

A
  • Morphine - AVOID
    • prolonged E1/2 life
    • increased bioavailability of oral form
    • decreased plasma protein binding- more free drug
    • exaggerated sedative and respiratory-depressant effects
  • Meperidine- AVOID
    • 50% reduction in clearance
    • doubling half-life
      • Poss experience neuro-toxicity from accumulation of normeperidine**
  • Fentanyl- Drug of choice
    • plasma clearance decreased
    • continuous infusions/repeated dosing in cirrhotic pts → produce exaggerated/pronounced effects
      • titrate w/ caution
  • Sufentanil
    • pharmacokinetics not significantly altered
    • some differences seen in E1/2 – so infusions/multiple doses → possible prolongation of effect.
  • Alfentanil
    • E1/2 life almost doubled- avoid
    • higher free fractions- more free drug available
      • potentially lead to prolonged DOA and enhanced effects
  • Remifentanil elimination unaltered.
44
Q

Consideraitons iwth NMB in liver disease?

A
  • Increased Vd → require HIGHER initial dose
  • Cirrhosis/advanced liver disease reduces elimination of:
    • vecuronium, rocuronium, pancuronium → increased DOA
      • especially after repeated doses/infusions
  • Atracurium and cisatracurium:
    • not dependent on hepatic elimination
    • can be used without modification
  • Decreased plasma cholinesterase levels
    • DOA of Succinylcholine and mivacurium prolonged
  • Increased Vd may require a higher initial dose
  • Sugammadex – excreted unchanged in the urine
    • effective in ESLD
    • no data on effectiveness
45
Q

Administration of catecholamines to patients with liver disease?

A
  • Decreased response bc circulating vasodilators
    • ex: increase bile acids and glucagon
  • Impaired ability to translocate blood from pulmonary and splanchnic blood reservoirs to systemic circulation
    • Impaired ability to compensate (don’t tolerate BL well)
  • Considerations:
    • Increased doses of vasopressors
    • Nonadrenergic vasoconstrictor (vasopressin) to support BP
      • Preferentially vasoconstrict splenetic vasculature → redirect BF to central circ
    • Pts with biliary obstruction are particularly intolerant of blood loss
46
Q

Considerations when choosing anesthesia technique in patient with liver disease?

A
  • Local/MAC
    • adequate sedation essential to minimize SNS stimulation and resultant decreases in hepatic blood flow and O2 delivery – titrate carefully
  • Regional
    • nice option only if no coagulopathies
  • GA:
    • RSI or awake intubation
    • Sevoflurane and Isoflurane agents of choice (Des has SNS stim)
    • N2O O.K.
  • Fluids: No proof colloids better than crystalloids for resuscitation
47
Q

Considerations during maintenance phase of anesthesia in a patient with liver disease?

A

Avoid hepatic hypoxia! Consider effects of techniques…..

  • FiO2, right to left shunting, VQ mismatch
  • Anemia
  • Hypotension, decreased CO, hypovolemia
  • Stress response → Release of endogenous vasoconstrictors (renin-angiotensin, catecholamines, ADH)
    • Reduce stress response
  • Vasodilation (hypotension) and low CO increases O2 uptake in pre-portal areas and decreases the O2 content of portal blood = ESLD can no longer autoregulate hepatic arterial flow = hepatic hypoxia!
    • Maintain normal BP, Oxygenation

Intraoperative liver injury can develop from O2 deprivation, the stress response, drug toxicity, blood transfusion, and infection.

48
Q

Post-operative indications of liver dysfunction?

A
  • Jaundice appears in many patients following major surgery
    • overproduction or underexcretion of bilirubin
      • hematoma/blood tx
    • direct hepatocellular injury (low BP/hypoxia)
    • extrahepatic obstruction
  • Fulminant hepatic failure
    • Encephalopathy occurring w/in 2-8 weeks of symptoms
  • Other causes of jaundice:
    • Hemolytic anemia
    • Resorption of lg surgical hematomas
    • transfusions of RBCs
      • Unconjugated hyperbilirubinemia. Often post-op liver dysfunction is realted to asymtomatic and preexisting hepatic injury that escaped pre-op detected.
49
Q

What are TIPS procedures?

A

Transjugular Intrahepatic Portal-Systemic Shunt Procedure

  • percutaneously created intrahepatic connection of portal and systemic circulations
    • Uses: ESLD to decrease portal pressure and attenuate complications related to portal hypertension
      • Hook up PV to HV
        • (Uses ex: variceal bleeding or refractory ascites)
      • Improve QOL (not long term)–> minimizes need for paracentesis frequently
    • Diversion of PBF into the hepatic vein is achieved by placement of an expandable intraparenchymal tract

Pic:

Miller Figure 73-7 (TIPS) procedure.

A stent (or stents) passed through the IJ vein over a wire into the hepatic vein (A); dilated esophageal varices (EV) are apparent. The wire and stent or stents are then advanced into the portal vein (B), after which blood can pass through the portal vein into the hepatic vein and bypass and decompress dilated esophageal veins (C).

50
Q

What are some outcomes after a TIPS procedure?

A
  • Good:
    • Decrease risk of bleed
    • Decrease risk ascites
  • Bad:
    • No macrophage activity in blood going through shunt
      • Higher sepsis risk
    • No opportunity for ammonia → urea conversion
      • Cognitive fx change
51
Q

Anesthesia technique for TIPS procedure?

A
  • under local, MAC or GA (RSI) in selected patients
    • (length of procedure, ability to tolerate supine position, cirrhotic patients with underlying pulmonary dysfunction as a result of ascites and hypoxemia, aspiration risk → GA w/ RSI)
52
Q

Resuscitation/fluid/blood thresholds for TIPS procedure?

A
  • Resuscitation with fluid and blood products in patients w/variceal bleeding
    • Transfusion thresholds:
      • Hgb < 7-9 mg/dL
      • INR > 2.0 → FFP
      • plt < 50,000/uL
        • Patients frequently have a severe coagulopathy requires preprocedural correction
53
Q

Complications of TIPS procedures?

A
  • PTX or neck vessel injury can occur during vessel puncture
    • (complications reduced by US guidance during jugular vein puncture)
  • Cardiac dysrhythmias
    • mechanically induced during intracardiac catheter passage
  • Acute/life-threatening hemorrhage
    • caused by hepatic artery puncture, a hepatic capsular tear, or extrahepatic portal venous puncture
  • Increased risk of pulmonary edema and CHF in patients with borderline cardiac reserve
54
Q

Hepatic resection preop considerations?

A
  • Surgical indications: malignancy or benign hepatic masses
    • Smaller masses resected laparoscopically
  • M&M relates to extent of resection
    • (which occurs along one or more of the 8 functional segments of the liver)
  • Risk assessment similar as other major abdominal procedures
  • Pre-op:
    • CBC
    • Electrolytes
    • liver transaminases
    • albumin
    • coags
    • T&C
      • Anemia and coagulopathy should be corrected preop
  • Arterial line (usually)
  • +/- CVP
  • Choice/dosing of anesthetic drugs –
    • consider baseline hepatic parenchymal dysfunction and potential postop dysfunction resulting from resection of a major portion of the liver parenchyma
      • Avoid drugs depending on Hepatic BF
55
Q

Introperative considerations for hepatic resection?

A
  • Consider RSI
  • Risk of significant intra-op blood loss –
    • appropriate monitoring
    • sufficient vascular access to permit rapid transfusion
    • Fluid mgt
      • Controversial – liberal fluid and blood administration buffer against sudden blood loss VS maintenance of a low CVP during resection to minimize blood loss
        • (research inconclusive- see Miller 9th pg 439)
    • Other techniques:
      • vasopressor use
      • intermittent portal triad clamping
      • ischemic pre-conditioning- clamp for 10 min, unclamp, then increase time
      • acute normovolemic hemodilution
      • cell salvage
    • TXA:
      • Pre-incision: 500 mg IV
        • 250 mg q6 hrs X 3 days followed
  • Position: Modest degree of Trendelenburg position desired
    • reduction of intrahepatic venous pressure
      • reduce BL
    • maintain/increase cardiac preload and CO
    • reduce air embolism risk from disrupted hepatic veins
      • heart higher than sx site
  • Anticipate surgical clamping and hemodynamic responses
56
Q

Postoperative management of patients following hepatic resection?

A
  • Similar to those of other major abdominal procedures
  • Considerations:
    • Intravenous fluids:
      • include phosphates → facilitate liver regeneration and avoid severe hypophosphatemia
    • Decreased clearance of hepatically metabolized drugs
      • selecting and titrating methods of postoperative analgesia
    • ERAS protocols may improve outcomes – quality of evidence not adequate yet to make any clear recommendations