Transcript Slide 1

Thoracic organ
transplantation: an
overview for perfusionists
Andreas Hoschtitzky
Overview





OCTx
HCTx
HLTx
DLTx
SLTx
History OCTx


1905 Alexis Carrel transplanted a puppy's heart into the neck of a dog;
because of the lack of immunosuppression, the experiment was
unsuccessful.
Early investigators included Frank C. Mann of the Mayo Clinic, V.P. Demikov
of the Soviet Union, and Marcus Wong. These early efforts in
transplantation were thwarted by the infancy of cardiopulmonary bypass
and a lack of understanding of the immune system. As knowledge in these
areas advanced, so did the field of cardiac transplantation.

Shumway developed modern day transplantation protocols.

1967 Christian Barnard: first successful heart transplant in a human.


1983 The clinical use of cyclosporine as an immunosuppressant
revolutionized the field of transplantation. Recipient survival rates improved,
thus producing an explosive increase in the number of transplant centers
offering cardiac transplantation.
The remaining limiting factor: number of available organ donors.
OCTx transplantation



Indications: patients with end-stage
congestive heart failure with a prognosis of less
than a year to live without the transplant and
who are not candidates for conventional medical
therapy or have not been helped by
conventional medical therapy.
In the US approximately 4000 individuals are
waiting for hearts. In 1999, about 2000 heart
transplants were performed in the US. UK
around 300-400 per year.
Availability of organs is a major issue.
OCTx transplantation


Frequency: The annual frequency of the
procedure is about 1% of the general
population with heart failure
Etiology:
adults:
– Idiopathic cardiomyopathy 54%
– Ischemic cardiomyopathy 45%
– Congenital heart disease
and other diseases
1%
children: congenital heart disease and
cardiomyopathy most common: HLHS
commonest
Pathophysiology OCTx

Transplanted heart is unique:
– denervation of the organ makes it dependent on its intrinsic rate.
– as a result of the lack of neuronal input, some left ventricular hypertrophy
results.
– right ventricular function is directly dependent upon ischaemic time and
adequacy of preservation.
– right ventricle is easily damaged and may initially function as a passive conduit
until recovery occurs.

Allograft rejection 2 forms: cellular and humoral.
– Cellular rejection is the classic form of rejection: perivascular infiltration of
lymphocytes with subsequent myocyte damage and necrosis if left untreated.
– Humoral rejection is much more difficult to characterize and diagnose.
Generalized antibody response initiated by several unknown factors. The
antibody deposition into the myocardium results in global cardiac dysfunction.
Diagnosis is generally made on the basis of clinical suspicion and exclusion
because endomyocardial biopsy is of little value.

Coronary artery disease: late process, common to all cardiac allografts.
Diffuse myointimal hyperplasia of the small- and medium-sized vessels, occuring from
3 months to several years after implantation.
Etiology: still unclear, though cytomegalovirus (CMV) infection and chronic rejection
have been implicated. The mechanism of the process is thought to be dependent
upon growth-factor production in the allograft initiated by circulating lymphocytes.
Treatment: re-transplantation.
Indications OCTx








Deteriorating cardiac function and having a
prognosis of less than 1 year to live: NYHA class III
or IV symptoms
EF < 25%
Intractable angina or malignant cardiac arrhythmias
for which conventional therapy has been exhausted
PVR < 6-8 Wood units
Age < 65 years
Normal renal, hepatic, pulmonary and CNS function
Absence of malignancy, infection, recent pulmonary
infarct, severe peripheral vascular or cerebrovascular disease
Ability to comply with medical follow-up care
OCTx Donor criteria







Brain death
Consent next of kin
ABO compatible with recipient >
1year old
Within 20% size as recipient
No cardiac disease in medical history
Normal ventricular wall motion on
ECHO
Normal heart as assessed by donor
team

20% recipients die on waiting list
though….
Management of the Potential Cardiac Recipient
TAILORED MEDICAL THERAPY FOR END-STAGE CARDIAC FAILURE
Conventional outpatient management of congestive heart failure includes ACE inhibitors, AR blockers,
beta blockers, and diuretics (especially spironolactone).
PHARMACOLOGIC BRIDGE TO TRANSPLANTATION
Critically compromised patients require admission to the intensive care unit for intravenous inotropic
therapy. Milrinone, dobutamine, and dopamine are the agents of choice. Placement of an intra-aortic
balloon pump (IABP) also may be necessary in heart failure refractory to initial pharmacologic
measures. Patients with continued pulmonary congestion or global hypoperfusion despite maximal
pharmacologic and IABP therapies have been shown to improve with placement of mechanical devices
as bridges to transplantation.
MECHANICAL BRIDGE TO TRANSPLANTATION
The increased success of cardiac transplantation in conjunction with the static number of available
organs has created a need for mechanical assist devices as a bridge to transplantation. Ventricular
assist devices (VAD) or total artificial hearts (TAH) may be indicated in potential cardiac recipients who
remain unstable after 24 to 48 hours of maximal pharmacologic support. Since these devices are rarely
weaned, however, it is imperative that the patient's candidacy for transplantation be scrutinized prior to
placement of a VAD or TAH. Patient selection for a mechanical device is a complex, evolving field.
Recent data shows that approximately 70% of patients are successfully bridged to transplantation and
the actuarial survival is 80% at one year. Most large series suggest an improvement in survival because
the devices allow patients to be rehabilitated while on the device. Initial results from the Randomized
Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure (REMATCH) study
indicate that patients with devices have improved survival and quality of life at 1 year compared to
medical therapy and may prove to be an acceptable long term option in those patients who are not
candidates for cardiac transplantation.
LIFE-THREATENING VENTRICULAR ARRHYTHMIAS
Symptomatic VT or VF and a history of sudden cardiac death (SCD) are indications for placement of an
automatic implantable cardioverter-defibrillator (AICD), long-term amiodarone therapy, or occasionally
radiofrequency catheter ablation. SCD is the most common cause of death in patients awaiting heart
transplantation and is most common within the first 3 months after referral for transplantation. Several
studies have shown that implantation of a defibrillator improved survival in patients with either a history
of or inducible ventricular tachycardia or fibrillation.
Management of the Cardiac Donor





Complex physiological phenomenon of brain death and the need to coordinate procurement with other organ donor teams. Optimal
care requires that the donor be treated as any other intensive care unit patient with invasive hemodynamic monitoring, ventilatory
support, and meticulous attention to intravascular volume status and electrolytes.
Continuous monitoring of arterial pressure, central venous pressure, and urinary output is mandatory. As the number of marginal donors
increases with the acceptance of more lenient eligibility criteria, some transplant centers have established mobile intensive care teams
that are dispatched to ensure appropriate management of these highly labile patients.
Haemodynamic instability in the donor may result from vasomotor dysfunction, hypovolemia, hypothermia, and dysrhythmias.
Increased intracranial pressure may lead to massive sympathetic discharge with elevated levels of circulating endogenous
catecholamines. The resultant episodes of systemic hypertension and coronary vasospasm place the allograft at significant risk of
ischemic injury. Rapid afterload reduction may be achieved with sodium nitroprusside, whereas volatile anesthetics assist in reducing the
intensity of sympathetic bursts. To minimize cerebral edema prior to the declaration of brain death, potential donors have been
intravascularly volume depleted via strict fluid restriction and osmotic diuresis. Aggressive volume resuscitation is sometimes necessary
and may require use of a Swan-Ganz catheter.
Fluid overload, however, should be avoided to prevent postoperative allograft dysfunction caused by chamber distention and myocardial
edema. Blood transfusions are indicated to optimize oxygen delivery if the hemoglobin falls below 10 g/dL. Mean arterial pressure should
be maintained between 80 and 90 mm Hg. If fluid resuscitation is inadequate to restore blood pressure in the hypotensive donor, a
dopamine infusion is initiated for inotropic support. Vasopressors are occasionally indicated for hypotension caused by loss of
systemic vasomotor tone. Prolonged administration of high-dose catecholamine therapy (dopamine >10–15 µg/ kg/min) has been
associated with poor cardiac function in the posttransplant period because of depletion of myocardial norepinephrine stores.
Traditionally, these patients were rejected for use as cardiac donors, but high-dose inotropic support is no longer an absolute
contraindication for donation.
Maintenance of normal temperatures, electrolyte levels, osmolarity, acid-base balance, and oxygenation is critical for optimal donor
management. Common electrolyte disturbances include hypernatremia, hypokalemia, hypomagnesemia, and hypophosphatemia.97
Central diabetes insipidus develops in more than 50% of donors because of pituitary dysfunction, and massive diuresis complicates fluid
and electrolyte management.

A low-dose aqueous vasopressin (Pitressin) infusion is initiated at 0.8 to 1.0 U/h and titrated to keep urinary output at approximately
100 to 200 mL/h. Alternatively, vasopressin may be administered periodically subcutaneously or intramuscularly (10 U every 4 hours).

Standard ventilator management with endotracheal suctioning is essential in these vulnerable patients.

Broad-spectrum antibiotic therapy with a cephalosporin is initiated following collection of blood, urine, and tracheal aspirate for culture.

Brain death is associated with the depletion of a variety of hormones, including free triiodothyronine (T3), cortisol, and insulin. Donor
pretreatment with hormone replacement therapy has proven to be beneficial.
Donor Heart Procurement
The heart is inspected and palpated for evidence of
cardiac disease or injury. The SVC, IVC and azygous
vein are encircled with ties. The aorta is dissected
from the pulmonary artery and isolated with tape.
To facilitate access to the epigastrium by the liver
procurement team, the cardiac team often then
temporarily retires from the operating room table or
assists with retraction.
Once preparation for liver, pancreas, lung, and kidney
explantation is completed, the patient is
administered 30,000 IU of heparin intravenously.
OCTx donor operation
The azygous vein and SVC are ligated and divided distal to the
azygous vein leaving a long segment of superior vena cava.
The inferior vena cava is clamped at the level of the diaphragm
(if the abdominal IVC is vented) and divided proximal to the
clamp to permit efflux of the cardioplegia. Additional venting is
achieved with transection of the right superior pulmonary vein.
The cross-clamp is applied at the takeoff of the innominate
artery and the heart is arrested with a single flush (500 mL) of
cardioplegic solution infused through a 14-gauge needle inserted
proximal to the cross-clamp. Rapid cooling of the heart is
achieved with copious amounts of cold saline and cold saline
slush.
The apex of the heart is elevated cephalad and the pulmonary
veins are divided. This maneuver is appropriately modified to
retain adequate left atrial cuffs for both lungs and the heart if the
lungs also are being procured.
While applying caudal traction to the heart with the nondominant hand, the ascending aorta is transected proximal to the
innominate artery and the pulmonary arteries are divided distal
to bifurcation (modification is necessary if the lungs are being
procured).
More generous segments of the great vessels and superior vena
cava may be required for recipients with congenital heart
disease.
Donor heart for OCTx
Donor heart is removed from the
transport cooler and placed in a basin
of cold saline. Preparation of the
donor heart is accomplished.
Electrocautery and sharp dissection
are used to separate the aorta and
pulmonary artery.
The left atrium is incised by
connecting the pulmonary vein
orifices and excess atrial tissue is
trimmed forming a circular cuff
tailored to the size of the recipient left
atrial remnant.
Organ preservation OCTx
Safe ischaemic period is around 4 to 6 hours, beyond this “marginal donors”.
Postoperative myocardial dysfunction is secondary to suboptimal donor management,
hypothermia, ischemia-reperfusion injury, and depletion of energy stores.
A single flush of a cardioplegic or preservative solution followed by static hypothermic
storage.
No single preservation regimen has demonstrated consistent, clinically significant superior
myocardial protection when used within the current safe limits of ischemia.
Controversy surrounds optimal storage temperature, composition of cardioplegic and storage
solutions, techniques of solution delivery, additives, and reperfusion modification.
Hypothermia remains the cornerstone of organ preservation. The ideal storage temperature
is unknown, but most institutions aim for temperatures between 4°C and 10°C.
Crystalloid solutions of widely different compositions are available and the debate over
them speaks for the fact that no ideal solution currently exists. Depending on their ionic
composition, solutions are classified as intracellular or extracellular.
Intracellular solutions, characterized by moderate to high concentrations of potassium and
low concentrations of sodium, purportedly reduce hypothermia-induced cellular edema
by mimicking the intracellular milieu. Commonly used examples of these solutions
include University of Wisconsin, Euro-Collins, and in Europe, Bretschneider (HTK)
solutions.
Extracellular solutions, characterized by low to moderate potassium and high sodium
concentrations, avoid the theoretical potential for cellular damage and increased vascular
resistance associated with hyperkalemic solutions. Stanford, Hopkins, and St. Thomas
Hospital solutions are representative extracellular cardioplegic solutions.
Organ preservation OCTx
Additives for cardioplegic storage solutions:
The greatest potential for future routine use may lie with impermeants, substrates, and
antioxidants.
Impermeants (mannitol, lactobionate, raffinose, and histidine) counteract intracellular
osmotic pressure to reduce hypothermia-induced cellular edema in the allograft.
The preservation of myocardial high-energy phosphates during ischemia (to prevent
contracture bands) and their rapid regeneration at reperfusion (to fuel the newly
contracting heart) are the primary objectives for the use of substrate-enhanced
media. Adenosine, L-pyruvate, and L-glutamate have been studied most intensely.
Recognizing that oxygen-derived free radicals and neutrophils likely are critical
mediators of myocardial reperfusion injury, considerable investigative effort has
been undertaken to modify the untoward effects of ischaemia-reperfusion with
antioxidant additives including allopurinol, glutathione, superoxide dismutase,
catalase, mannitol, and histidine.
A variety of pharmacologic and mechanical strategies for leukocyte inhibition and
depletion are also being explored.
Benefits of continuous perfusion preservation techniques are currently overshadowed by
exacerbation of extracellular cardiac edema and logistical problems inherent to a
complex perfusion apparatus.
Experimental low-pressure (microperfusion) and intermittent flush techniques theoretically
provide sufficient oxygen and substrates for basal metabolic demands without causing
significant edema.
20% of peri-operative deaths are still caused by cardiac dysfunction….
OCTx vs HCTx


Orthotopic cardiac transplantation: replacement of
part (or occasionally all) of the recipient's heart
with a healthy donor allograft.
Heterotopic cardiac transplantation, the piggybacking of an allograft onto the patient's heart, is
rarely performed today. Indicated if orthotopic
transplantation is not possible because of elevated
pulmonary vascular resistance or when a donor
heart is too small to sustain the recipient. Results
are not equivalent to orthotopic transplant.
OCTx: ANAESTHETIC MANAGEMENT



Once donor team has given go-ahead, recipient
induction commences. High-dose narcotics (e.g.
fentanyl) usually are employed for induction and
maintenance anaesthesia.
In light of the poor ventricular function of the recipient,
all anesthetic agents should be titrated carefully with
inotropic and vasoactive agents readily accessible for the
rapid management of induction-induced hypotension.
Inhaled agents may be added if necessary, but their
potential myocardial depressant effects limit widespread
use in this patient population.
Prior to skin incision, some centers initiate aprotinin or
aminocaproic acid therapy to minimize perioperative
blood loss.
OCTx: OPERATIVE
RECIPIENT PREPARATION



Median sternotomy and vertical pericardiotomy, the patient is heparinized and
prepared for cardiopulmonary bypass. Bicaval venous cannulation and distal
ascending aortic cannulation just proximal to the origin of the innominate
artery is optimal. Umbilical tape snares are passed around the superior and
inferior vena cava. Bypass is initiated, the patient is cooled to 28°C, caval
snares are tightened, and the ascending aorta is cross-clamped. The great
vessels are transected above the semilunar commissures, whereas the atria
are incised along the atrioventricular grooves leaving cuffs for allograft
implantation. Removal of the atrial appendages reduces the risk of
postoperative thrombus formation.
Following cardiectomy, the proximal 1 to 2 cm of aorta and pulmonary artery
are separated from one another with electrocautery, taking care to avoid
injuring the right pulmonary artery. Continuous aspiration of pulmonary
venous return from bronchial collaterals is achieved by insertion of a vent into
the left atrial remnant, either directly or via the right superior pulmonary vein.
Timing of donor and recipient cardiectomies is critical to minimize allograft
ischaemic time and recipient bypass time. Frequent communication between
the procurement and transplant teams permits optimal coordination of the
procedures. Ideally, the recipient cardiectomy is completed just prior to the
arrival of the cardiac allograft.
OCTx implantation
A double-ended 3-0 Prolene is taken through
the recipient left atrial cuff at the level of the
left superior pulmonary vein and then
through the donor left atrial cuff near the
base of the atrial appendage.
The allograft is lowered into the recipient
mediastinum atop a cold sponge to insulate it
from direct thermal transfer from adjacent
thoracic structures. The suture is continued in
a running fashion caudally and then medially
to the inferior aspect of the interatrial
septum.
Upon completion of the posterior left atrial
suture line, continuous topical cold saline
irrigation of the pericardial well is initiated,
and the patient is oriented in a left side
down–head up position to allow drainage of
the saline away from the operative field and
maximal cold saline exposure of the left and
right ventricles.
OCTx implantation
OCTx implantation
The second arm of the suture is run along the roof of
the left atrium and down the interatrial septum. It is
important to continually assess size discrepancy
between donor and recipient atria so that appropriate
plication of excess tissue may be performed.
The left atrium is filled with saline and the two arms of
suture are tied together on the outside of the heart.
Some centers introduce a line into the left atrial
appendage for continuous endocardial cooling of the
allograft (50–75 mL/min) and evacuation of intracardiac
air Left atrial anastomosis is complete, a curvilinear
incision is made from the IVC toward the RA appendage
of the allograft. This reduces the risk of injury to the
sinoatrial node and accounts for the preservation of
sinus rhythm observed in most recipients.
The tricuspid apparatus and interatrial septum are
inspected. Recipients are predisposed to increased
right-sided heart pressures in the early postoperative
period owing to preexisting pulmonary hypertension
and volume overload. Both conditions are poorly
tolerated by the recovering right ventricle.
To avoid refractory arterial desaturation associated with
right-to-left shunting, patent foramen ovale is
oversewn.
OCTx implantation
RA anastomosis is performed in a
running fashion similar to the left
with the initial anchor suture placed
either at the most superior or
inferior aspect of the interatrial
septum so that the ends of the
suture meet in the middle of the
anterolateral wall.
The end-to-end pulmonary artery
anastomosis is next performed using
a 4-0 Prolene suture beginning with
the posterior wall from inside of the
vessel and then completing the
anterior wall from the outside.
It is crucial that the pulmonary
artery ends be trimmed to eliminate
any redundancy in the vessel that
might cause kinking.
OCTx implantation
Rewarming is initiated at this time. Finally, the aortic
anastomosis is performed using a technique similar to
the pulmonary artery except that some redundancy is
desirable in the aorta as it facilitates visualization of the
posterior suture line.
Rewarming is usually begun prior to the aortic
anastomosis, which is performed in a standard end-toend fashion.
Routine de-airing techniques are then employed.
Cold saline lavage is discontinued, lidocaine (100–200 mg
IV) is administered, and the aortic cross-clamp is
removed. Half of patients require electrical defibrillation.
A needle vent is inserted in the ascending aorta for final
de-airing with the patient in steep Trendelenburg.
Inotrope infusion is initiated and titrated to achieve a
heart rate between 90 and 110 bpm. Temporary
epicardial pacing wires are placed in the donor right
atrium and ventricle.
The patient is weaned from cardiopulmonary bypass and
the cannulae are removed.
Anatomy
Recent trend: bicaval anastomoses rather than right
atrial anastomoses in an attempt to decrease the
incidence of postoperative tricuspid insufficiency.
In the transplantation process, the sinoatrial nodes of
the donor and recipient remain intact, and both are
present within the recipient. For approximately 3
weeks after surgery, the electrocardiogram
demonstrates 2 P waves; however, the heart rate
and electrical activity of the new heart are purely
dependent on the intrinsic electrical system of the
heart and not on the neurological input from the
recipient.
ALTERNATIVE TECHNIQUES FOR
ORTHOTOPIC HEART TRANSPLANTATION

Two alternative techniques for orthotopic heart transplantation have
been gaining popularity over the past several years:
– total heart transplantation involves complete excision of the recipient
heart with bicaval end-to-end anastomoses
– bilateral pulmonary venous anastomoses.


The Wythenshawe bicaval technique is performed in a similar fashion
except that the recipient left atrium is prepared as a single cuff with
all four pulmonary vein orifices. Although these procedures are more
technically difficult than standard orthotopic transplantation, series
using these techniques have reported shorter hospital stays and
reduced postoperative dependence on diuretics, in addition to lower
incidences of atrial dysrhythmias, conduction disturbances, mitral
and tricuspid valve incompetence, and right ventricular failure.
Furthermore, a recently completed randomized study comparing biatrial versus bicaval transplant showed an improved twelve month
survival in the bicaval group. Long term outcomes and additional
randomized studies evaluating these alternative techniques are still
needed
RECIPIENTS WITH
CONGENITAL ANOMALIES
Unlike children and infants,
transplantation in adults with previous
palliative procedures for congenital
anomalies is uncommon.
Generous donor cardiectomy be
performed so that sufficient tissue is
available for optimal reconstruction.
There are a variety of anomaly-specific
implantation techniques.
Heterotopic Heart Transplantation
Pulmonary hypertension and right heart failure has remained one of the leading causes of death
in cardiac transplantation. This has led to an interest in heterotopic heart tranplantation.
Currently, heterotopic heart transplants are indicated in patients with irreversible pulmonary
hypertension or significant donor-recipient size mismatch.
DONOR ALLOGRAFT PREPARATION
Like the cardiectomy for patients with congenital disease, the maximal length of aorta, superior
vena cava, and pulmonary arteries is procured. The inferior vena cava and the right pulmonary
veins are oversewn, and a common left pulmonary vein orifice is created. A linear incision is made
along the long axis of the posterior right atrium extending 3 to 4 cm into the superior vena cava.
Domino Donor Procedure
The Domino donor procedure was used to avoid wasting relatively healthy hearts from selected heartlung transplant recipients. These organs were transplanted into a different recipient using
standard orthotopic or heterotopic techniques.
Heterotopic CTx
The sequence of anastomoses is as
following: donor left pulmonary vein orifice
to recipient left atrium, donor superior
vena cava-right atrial orifice to recipient
right atrium, end-to-side aortic-aortic
anastomosis, and finally an end-to-side
anastomosis joining the pulmonary arteries
of donor and recipient.
By employing this technique, the strengths
of both the native and transplanted heart
are utilized.
The conserved recipient's right ventricle
provides the necessary assistance to the
transplanted heart to overcome significant
pulmonary hypertension.
The incidence of tricuspid
regurgitation is reported to be as
high as 47-98% following heart
transplantation (Chan, 2001).
Some centers have now begun to
prophylactically perform tricuspid
annuloplasty on donor grafts
before performing the
transplantation (McGee, 2004).
POSTOPERATIVE MANAGEMENT
Because of denervation the SA node of the transplanted heart fires at its increased intrinsic resting rate of 90 to 110 bpm. The allograft relies on
distant noncardiac sites as its source for catecholamines; thus, its response to stress (e.g. hypovolemia, hypoxia, anemia) is somewhat
delayed until circulating catecholamines can exert their positive chronotropic effect on the heart. Careful examination of the
electrocardiogram occasionally may reveal a distinct P wave originating from the innervated atrial remnant of the recipient, and an
increase in its rate may be used as an early indicator of stress. The absence of a normal reflex tachycardia in response to venous pooling
accounts for the frequency of orthostatic hypotension in transplant patients.
Denervation alters the heart's response to therapeutic interventions that act directly through the cardiac autonomic nervous system.
Carotid sinus massage, Valsalva maneuver, and atropine have no effect on sinoatrial node firing or atrioventricular conduction. Because of
depletion of myocardial catecholamine stores associated with prolonged inotropic support of the donor, the allograft often requires high
doses of catecholamines.
ROUTINE HEMODYNAMIC MANAGEMENT
Donor myocardial performance is transiently depressed in the immediate postoperative period. Allograft injury associated with donor
hemodynamic instability and the hypothermic, ischemic insult of preservation contribute to the reduced ventricular compliance and
contractility characteristics of the newly transplanted heart. Abnormal atrial dynamics owing to the midatrial anastomosis exacerbate the
reduction in ventricular diastolic loading. An infusion of epinephrine or dobutamine is initiated routinely in the operating room to provide
temporary inotropic support. Restoration of normal myocardial function usually permits the cautious weaning of inotropic support within 2
to 4 days.
EARLY ALLOGRAFT FAILURE
Early cardiac failure accounts for up to 25% of perioperative deaths of transplant recipients. The cause may be multifactorial, but the most
important etiologies are pulmonary hypertension, ischemic injury during preservation, and acute rejection. Mechanical support with an
intra-aortic balloon pump or ventricular assist device is indicated in cases refractory to pharmacologic interventions. Re-transplantation in
this setting is associated with very high mortality.
Chronic left ventricular failure frequently is associated with elevated pulmonary vascular resistance, and the unprepared donor right
ventricle may be unable to overcome this increased afterload. Although recipients are screened to ensure that those with irreversible
pulmonary hypertension are not considered for transplantation, right heart failure remains a leading cause of early mortality. Initial
management involves employing pulmonary vasodilators such as inhaled nitric oxide, nitroglycerin, or sodium nitroprusside. Pulmonary
hypertension refractory to these vasodilators will often respond to prostaglandin E1 (PGE1). Inhalation nitric oxide is considered the
standard at several institutions. Intra-aortic or pulmonary artery balloon counterpulsation and right ventricular assist devices have been
utilized in patients unresponsive to medical therapy.
DYSRHYTHMIAS
Sinus or junctional bradycardia occurs in more than half of transplant recipients. The primary risk factor for sinus node dysfunction is
prolonged organ ischemia. Adequate heart rate is achieved with inotropic drug infusions and/or temporary epicardial pacing. Most
bradyarrhythmias resolve over 1 to 2 weeks, although recovery may be further delayed in patients who received preoperative amiodarone
therapy. Theophylline has been effective in patients with bradyarrhythmias and has decreased the need for permanent pacemakers in this
patient population. Ventricular arrhythmias, primarily premature ventricular beats (PVCs) and nonsustained ventricular tachycardia, have
been reported in up to 60% of recipients when monitored continuously. AF/flutter is treated with digoxin, but at a higher dose than used
in the setting of an innervated heart. Arrhythmias occasionally are markers for acute rejection.
SYSTEMIC HYPERTENSION
Mean arterial pressures greater than 80 mm Hg should be treated to prevent unnecessary afterload stress on the allograft. In the early
postoperative period, intravenous sodium nitroprusside or nitroglycerin is administered. Nitroglycerin is associated with less pulmonary
shunting because of a relative preservation of the pulmonary hypoxic vasoconstrictor reflex. If hypertension persists, an oral
antihypertensive can be added to permit weaning of the parenteral agents.
Respiratory Management
The respiratory management is the same as following routine cardiac surgery.
Renal Function
Preoperative renal insufficiency owing to chronic heart failure and the
nephrotoxic effects of cyclosporine places the recipient at increased risk of
renal insufficiency. Acute cyclosporine-induced renal insufficiency usually will
resolve with the reduction in cyclosporine dose. Patients at risk for renal
failure initially may receive cyclosporine as a continuous intravenous infusion
to eliminate the wide fluctuations in levels associated with oral dosing.
Furthermore, concurrent administration of mannitol with cyclosporine may
reduce its nephrotoxicity. Alternatively, some centers administer a cytolytic
agent in the immediate postoperative period and delay the initiation of
cyclosporine therapy.
Intermediate Care Unit and Convalescent Ward
The increasing risk of nosocomial infections with resistant organisms has led
to shorter hospital stays for cardiac transplant recipients. Most patients are
discharged 7 to 14 days following transplantation. Patient education is
performed by the cardiac nursing staff. Topics include medications (regimens
and potential side effects), diet, exercise (routines and restrictions), and
infection recognition.
Outpatient Follow-up
Close follow-up by an experienced transplant team is the cornerstone for
successful long-term survival after cardiac transplantation. This
comprehensive team facilitates the early detection of rejection, opportunistic
infections, patient noncompliance, and adverse sequelae of
immunosuppression. Clinic visits routinely are scheduled concurrently with
endomyocardial biopsies and include physical examination, a variety of
laboratory studies, CXR and ECG.
IMMUNOSUPPRESSIVE
THERAPY
An organism's ability to distinguish self from non-self
is critical for its survival in a hostile environment. In
transplantation, the recipient's host defense
mechanisms recognize the human leukocyte
antigens (HLA) on allograft cells as being non-self
and, if permitted, will respond to eradicate the
foreign cells.
The ultimate goal of immunosuppressive therapy is
the selective modulation of the recipient's immune
response to prevent rejection, whilst sparing
immune defenses against infections or neoplasia
and minimizing the toxicity associated with
immunosuppressive agents
Pharmacologic
Immunosuppressive Strategies
Early induction phase followed by a long-term maintenance phase. This basic
strategy essentially is universal, although the choice of immunosuppressive
agents, dosages, and combination protocols vary among transplantation
centers.
Tendency for allograft rejection is greatest in the early postoperative period: the
most intense immunosuppression is administered during this induction phase.
Most programs employ a triple immunosuppressive regimen while some
centers also provide additional induction prophylaxis with potent polyclonal
antibodies, and OKT3 or IL-2 blockers.
After several months, immunosuppression and rejection surveillance are gradually
reduced to chronic maintenance phase levels and frequencies.
Most centers use triple drug therapy: cyclosporine, steroids, and mycophenolate
mofentil or azathioprine. The use of a multidrug regimen permits adequate
immunosuppression with reduced doses of individual agents to minimize their
toxicity. The use of cyclosporine has allowed for steroid-free maintenance
immunosuppression, thus avoiding the multiple untoward sequelae associated
with chronic corticosteroid therapy immunosuppression.The timing of steroid
withdrawal varies as some clinicians discontinue prednisone within several
weeks of transplantation, whereas others delay the taper until 6 to 12 months
posttransplantation.
Recently, it has been suggested that the majority of patients can be completely
tapered off steroids without an increased incidence of rejection. Attempts at
corticosteroid withdrawal in patients with history of rejection, however, have
usually been unsuccessful.
Hyperacute Rejection
Results from pre-formed, donor-specific antibodies in the recipient. ABO
blood group and panel reactive antibody screening have made this
condition a rare complication.
The onset of hyperacute rejection occurs within minutes to several hours
after transplantation and the results are catastrophic.
Gross inspection reveals a mottled or dark red, flaccid allograft, and
histologic examination confirms the characteristic global interstitial
hemorrhage and edema without lymphocytic infiltrate.
Immunofluorescence techniques reveal deposits of immunoglobulins and
complement on the vascular endothelium.
No treatment is effective except retransplantation, and even this
aggressive strategy frequently is unsuccessful.
Complications

Bleeding from the suture lines is a rare occurrence but may require reexploration.

Hyperacute rejection can occur immediately after blood flow is restored to the

Infection is the primary concern. Preventive measures should be instituted. Early on

Psychiatric disturbances from steroid therapy can occur in the immediate


allograft and up to 1 week after surgery despite therapeutic immunosuppression.
bacterial and fungal infections. Fungal infections can appear if the patient is diabetic or
overimmunosuppressed. Prophylaxis for Pneumocystis carinii is universally administered,
as is therapy for CMV infection. Maintain vigilance for other uncommon infectious
processes including Listeria, Legionella, Chlamydia, and Nocardia infections.
posttransplant period. These disturbances may be predicted from the pre-transplantation
psychiatric evaluation and thus averted.
Cardiac rejection is to be expected and should be detected by endomyocardial biopsy.
Depending upon the severity of the incident, the process can be treated with steroid
therapy alone, polyclonal antibody therapy, or monoclonal antibody therapy.
Allograft vascular disease is the main cause of late graft failure and death. The
coronary arteries develop a progressive concentric myointimal hyperplasia. This
hyperplasia can develop as early as 3 months after transplantation. The cause of the
process is unclear. However, CMV infection and recurrent rejection episodes are thought
to be associated with the cause. Current research indicates that the initial
ischemia/reperfusion injury of the allograft coupled with repeated rejection episodes
might contribute to the process. The only available therapy is re-transplantation. The
process can sometimes be treated by stenting of the diseased vessels.
CHRONIC COMPLICATIONS
Allograft Coronary Artery Disease
Long-term survival of cardiac transplant recipients is primarily limited by the development of allograft coronary artery disease (ACAD), the leading cause of death after the first
posttransplantation year.343–345 Angiographically detectable ACAD is reported in approximately 50% of patients by 5 years after transplantation. The etiology of this
allograft vasculopathy is multifactorial and involves both immunologic and nonimmunologic components. Recently, it has been shown that immune-related risk factors
appear to be more significant in the development of ACAD.346–348 Likewise, many nonimmune-associated related risks have been implicated in ACAD including increased
donor age, hyperlipidemia, and CMV infection.349–352 These immune and nonimmune risk factors lead to unique coronary pathology characterized by diffuse, concentric
intimal proliferation with infiltration by smooth muscle cells and macrophages leading to narrowing along the entire length of the vessel.353–354 Furthermore, collateral
vessels are notably absent. ACAD may begin within several weeks posttransplantation and insidiously progress at an accelerated rate to complete obliteration of the
coronary lumen with allograft failure secondary to ischemia.355
The clinical diagnosis of ACAD is difficult and complicated by allograft denervation resulting in silent myocardial ischemia. Ventricular arrhythmias, congestive heart failure, and
sudden death are commonly the initial presentation of significant ACAD. Noninvasive screening tests (e.g., thallium scintigraphy) are unreliable in transplant recipients.356
Annual coronary angiogram is the current gold standard for ACAD surveillance. However, due to the previously mentioned pathological changes, it underestimates the
extent of disease and is insensitive to early atherosclerotic lesions.357 This has led to growing interest in intravascular ultrasound (IVUS) devices.
IVUS is better equipped to provide important quantitative information regarding vessel wall morphology and the degree of intimal thickening.358–359 Some centers have begun
to use IVUS for the early detection of ACAD; however, concerns have been raised concerning its ability to assess more long-term lesions.360 Currently, the only definitive
treatment for advanced ACAD is retransplantation due to the diffuse and distal nature of ACAD. Based on this lack of effective treatment options, an emphasis has been
placed on prevention of ACAD. Currently, prophylactic management focuses on empiric risk factor modification (dietary and pharmacologic reduction of serum cholesterol,
cessation of smoking, hypertension control, etc.). Several studies have demonstrated a decrease in ACAD in patients treated with a calcium channel blocker or HMG-CoA
reductase inhibitors.348,361
Renal Dysfunction
Irreversible interstitial fibrosis caused by cyclosporine nephrotoxicity is chiefly responsible for the chronic renal dysfunction observed in cardiac transplant recipients.362–363 Its
pathogenesis is unclear but is believed to be secondary to afferent arteriolar vasoconstriction with secondary ischemia.364–365 Direct tubular toxicity also may play a
contributory role.366 Most renal injury occurs during the first 6 months following transplantation concurrent with the highest levels of cyclosporine. Little additional decline
in renal function occurs after 1 year.367 Frequent monitoring of cyclosporine levels and avoidance of intravascular volume depletion are important preventive
measures.368 Approximately 3% to10% of patients develop end-stage renal failure requiring dialysis or renal transplantation.369
Hypertension
Moderate to severe systemic hypertension afflicts 50% to 90% of cardiac transplant recipients and is a difficult problem to manage. Peripheral vasoconstriction in combination
with fluid retention seem to play the greatest role. Although the exact mechanisms are unclear, it likely involves a combination of cyclosporine-induced tubular
nephrotoxicity and vasoconstriction of renal and systemic arterioles mediated by sympathetic neural activation. No single class of antihypertensive agents has proven
uniformly effective, and treatment of this refractory hypertension remains empiric and difficult.
Malignancy
Chronic immunosuppression is associated with an increased incidence of malignancy. The estimated risk of carcinoma is almost 100-fold greater than in the general population
Lymphoproliferative disorders and carcinoma of the skin are most common. The risk of these malignancies is increased further following monoclonal and polyclonal
antibody therapy There is a predilection for unusual extranodal locations (e.g., lung, bowel, and brain). Treatment options in transplantation include: a reduction in
immunosuppression and high-dose acyclovir (to attenuate EBV replication) in addition to conventional therapies for carcinoma (chemotherapy, radiation therapy, and
surgical resection).
Other
Hyperlipidemia eventually develops in the majority of recipients and is managed with dietary restrictions, exercise, and lipid-lowering agents.
Osteoporosis
Avascular necrosis of weight-bearing joints
Obesity
Cholelithiasis
RESULTS OF OCTx
Operative mortality: 5% to 10%. Primary graft failure is the most
frequent cause of early death. Overall 1-year survival is approx. 80%
with a 4% mortality per year for subsequent years.
Infection and rejection account for the majority of deaths in the first 6
months; thereafter, accelerated coronary artery disease eventually
claims the lives of most recipients. Risk factors associated with
increased mortality include ventilator dependence, previous cardiac
transplantation, preoperative VAD or IABP, recipient age greater than
65 years, female gender (donor or recipient), and donor age greater
than 50 years.
Health-related quality of life (HRQOL) in patients following cardiac
transplantation demonstrates that most experience a HRQOL that
approaches that of the normal population. Although cardiac reserve
is reduced, exercise tolerance is improved dramatically compared to
preoperative level, and recipients usually can enjoy an active
lifestyle.
Nevertheless, because of concerns about future disability, recipients
often encounter significant problems with postoperative employment
and health insurance coverage particularly if over 50 years of age.
CARDIAC RE-TRANSPLANTATION



Re-transplantation accounts for fewer than 3% of the cardiac
transplants. Primary indications: allograft coronary artery
disease and refractory acute rejection.
Actuarial survival remains markedly reduced following retransplantation if performed within 6 months of the initial
procedure or in the setting of acute rejection. Recent data
suggest that the survival rate for cardiac re-transplantation at
1 year is 55%.
Recent data from the International Society for Heart and Lung
Transplantation (ISHLT) also shows though that if retransplantation occurs 2 years after the initial transplant
procedure, the 1-year survival rate markedly improves but
remains approximately 4% to 6% below that of primary
cardiac transplantation.
FUTURE
Clinical outcome of heart transplantation has dramatically improved. Although cardiac replacement remains the best
therapeutic option for patients with end-stage heart failure, a number of challenges await future investigators to
further improve survival and reduce transplant-related morbidity.
A major factor limiting long-term survival of recipients is allograft rejection and the untoward effects of
immunosuppression. Development of reliable, noninvasive diagnostic studies will permit more frequent
evaluations for the early detection of rejection and for monitoring the effectiveness of therapy. Ultimately, this will
allow more precise control of immunosuppression, and in turn a reduction in cumulative allograft injury and
infectious complications.
Immunosuppressive strategists will continue their efforts to establish specific unresponsiveness to antigens of
transplanted organs in hopes of preserving much of the recipient's immune responses. Novel immunosuppressive
agents and techniques are under continuous investigation for this purpose. Alternatively, donor organs may be
made less susceptible to immunologic attack through genetic engineering techniques by altering the expression of
cell membrane-bound molecules. This approach is being currently utilized in the pursuit of clinically applicable
xenotransplant sources.
Xenografts eventually may be an additional source of donor organs, although extended xenograft survival remains an
elusive goal. Complicating this alternative are unresolved ethical issues concerning transgenic experimentation
and the potential for transmission of veterinary pathogens to an immunosuppressed recipient.
Future improvements in organ preservation permitting extension of the storage interval will have several benefits.
In addition to a modest increase in the donor pool, extension of storage times would permit better allocation of
organs with respect to donor-recipient immunologic matching. There is growing evidence that human lymphocyte
antigen (HLA) matching may be important for long-term graft function through attenuation of chronic rejection.
Reducing the ischemic injury may also result in an attenuation of transplant coronary artery disease.
Mechanical assist devices are being used more frequently in patients with end-stage heart failure and may prove to
be the best solution for the current organ shortage. Assist devices are being currently used both as a bridge to
transplantation and a destination therapy. The Randomized Evaluation of Mechanical Assistance for the Treatment
of Congestive Heart Failure (REMATCH) study demonstrated a survival benefit in heart failure patients in which
assist devices were utilized versus all other forms of treatment for heart failure.61 It appears that as the
technology of assist devices continues to improve, it is only a matter of time before they become a long-term
solution for patients with severe congestive heart failure.
History heart-lung and
lung transplantation



First lung transplantation: James Hardy
1963. But it took another 20 years before
routine.
First Heart-Lung transplantation Demikhov
in dogs 1962, Reitz 1981 human
Initial graft failure secondary to:
inadequate preservation
long ischaemic times
lack of good immuno-suppressive drugs
technical difficulties with bronchial anastomoses




En-bloc double lung replacement: introduced by Patterson in
1988. This technique was later replaced by sequential bilateral
lung transplantation, by Pasque in 1990.
More recent operative innovations include living lobar
transplantation, an alternative to cadaveric bilateral lung
transplantation.
Combined heart-lung and isolated lung transplantation have
emerged as lifesaving procedures for patients with end-stage
cardiopulmonary or pulmonary disease.
To date, 2861 combined heart-lung transplants, 7204 single
lung transplants, and 5420 bilateral lung transplants have
been performed worldwide. While the number of heart-lung
transplants performed annually has declined in recent years,
the number of single and bilateral lung transplantation
procedures remains stable.
Indications HLTx
Indications for single and
bilateral lung transplantation
Contra-indications to HLTx and LTx















Age > 50 (heart-lung), > 55 (bilateral lung), > 60 (single lung)
Significant systemic or multisystem disease (e.g., peripheral or
cerebrovascular disease, portal hypertension, poorly controlled
diabetes mellitus)
Significant irreversible hepatic or renal dysfunction (e.g., bilirubin >
3.0 mg/dL, creatinine clearance < 50 mg/mL/min)
Active malignancy
Corticosteroid therapy (> 10 mg/day)
Panresistant respiratory flora
Cachexia or obesity (< 70% or > 130% ideal body weight)
Current cigarette smoking
Psychiatric illness or history of medical noncompliance
Drug or alcohol abuse
Previous cardiothoracic surgery (considered on a case-by-case basis)
Severe osteoporosis
Prolonged mechanical ventilation
HIV or HBsAg positivity
Hepatitis C infection with biopsy-proven liver disease
Recipient selection HLTx
and LTx




Progressively disabling cardiopulmonary or pulmonary disease who
still possess the capacity for full rehabilitation after transplantation..
Life expectancy of less than 18 to 24 months despite the use of
appropriate medical or alternative surgical strategies. On average,
waiting times can be from 6 to 36 months. Unfortunately, mortality
while on the waiting list remains nearly 20% for both lung and heartlung transplant candidates.
Disabling symptoms prompting consideration for transplantation
typically include dyspnea, cyanosis, syncope, and haemoptysis. NYHA
classes III or IV.
Evaluation includes a complete history, physical examination,
laboratory tests, specialized studies, and a psychosocial evaluation.
Tests and studies recipient
evaluation HLTx and LTx








Laboratory tests and studies routine haematology including clotting, blood type and
antibody screen, Immunology panel (FANA, RF), U&E, LFTs Electrolytes, including Mg2
+ CK with isoenzymes Serum protein electrophoresis Urinalysis Viral
serologies
Compromised host panel (cytomegalovirus, adenovirus, varicella-zoster,
herpes simplex, Epstein-Barr virus)
Hepatitis A, B, and C antibodies, hepatitis B
surface antigen (HBsAg)
Cytomegalovirus (quantitative antibodies and IgM) Human
immunodeficiency virus Electrocardiogram Chest x-rayStudies obtained as
indicated Echocardiogram with bubble study MUGA for right and left ventricular
ejection fraction Cardiac catheterization with coronary angiogram Thoracic CT
scan Quantitative ventilation-perfusion scans Carotid
duplex Mammogram Sputum for Gram stain, AFB smear, KOH, and routine bacterial,
mycobacterial, and fungal cultures
Required for listing (phase II)HLA and DR typing
Transplant antibody
Quantitative immunoglobulins
Histoplasma, Coccidiodes, and Toxoplasma titers PPD
Pulmonary function tests with arterial blood gases
12-hour urine collection for creatinine clearance and total protein
Urine viral culture







It is extremely important that a candidate's medical condition be optimized prior to heart-lung and lung
transplantation. Standard medical measures should be aggressively employed by the patient's local
physician, and the patient should have routine follow-up at the transplant center.
Supplemental oxygen is recommended for any patient exhibiting arterial hypoxemia, defined as either
an arterial oxygen saturation less than 90% or an arterial Po2 less than 60 mm Hg at rest, during
exertion, or while asleep.
For patients with heart failure, standard therapeutic measures are applied, including dietary restrictions,
diuretics, and vasodilators. Dietary water and salt restriction as well as diuretic therapy facilitate
intravascular fluid management. However, particular care must be exercised when using loop diuretics
in patients with underlying pulmonary disease; this class of potent diuretics results in a metabolic
alkalosis that depresses the effectiveness of carbon dioxide as a stimulus for breathing. Vasodilators
result in afterload reduction, and have been proven to effectively improve functional capacity and
prolong survival in patients suffering from severe cardiac failure.Commonly used vasodilators include
nitrates, hydralazine, and angiotensin-converting enzyme inhibitors.
Despite the clinical heterogeneity among patients with primary pulmonary hypertension, conventional
medical therapy targets the sequelae of the pulmonary vascular derangements associated with this
disease process. Supplemental oxygen therapy is recommended to eliminate the stimulus for hypoxic
pulmonary vasoconstriction and secondary erythropoiesis, thus lessening the burden placed on the right
side of the heart and diminishing the likelihood of cardiac arrhythmias. Pulmonary vasodilator therapy is
important in the treatment of primary pulmonary hypertension, and includes the use of calcium channel
blockers and continuous prostacyclin infusions. Because most standard vasodilators have potent
systemic effects, careful dosing and follow-up is essential. Approximately 20% of patients with primary
pulmonary hypertension will respond to calcium channel blockers, and this favorable response can
usually be predicted by the response to short-acting vasodilators during cardiac catheterization, but
response to the acute vasodilator challenge does not always predict the response to long-term
prostacyclin infusion.
Interstitial lung disease in patients awaiting transplantation results from a wide variety of diffuse
inflammatory processes, such as sarcoidosis, asbestosis, and collagen-vascular diseases. Increases in
pulmonary vascular resistance leading to right-sided heart failure are thought to result from interstitial
inflammatory infiltrates that entrap and eventually destroy septal arterioles, reducing the distensibility
of the remaining pulmonary vessels.This process, coupled with closure of peripheral bronchioles, results
in arterial hypoxemia, which further aggravates pulmonary hypertension. Corticosteroids are the
mainstay of treatment in this class of diseases. The adverse effects of steroids on airway healing are
well established, and mandate significant dose reductions in anticipation of heart-lung and isolated lung
transplantation.
The multisystem manifestations of cystic fibrosis, particularly chronic bronchopulmonary infection,
malabsorption, malnutrition, and diabetes mellitus, pose difficult management problems and require
aggressive chest physiotherapy, antibiotics, enteral or parenteral nutritional supplementation, and tight
serum glucose control.
Certain underlying diagnoses are associated with increased rates of pulmonary and systemic thrombosis
and embolization. These include dilated cardiomyopathy, congestive heart failure, and primary
pulmonary hypertension, and most centers recommend routine prophylactic anticoagulation with
heparin, warfarin, or antiplatelet agents.
HLTx and LTx donor selection
criteria
< 40 (heart-lung), < 50 (lung)
Smoking history less than 20 pack-years
Arterial Po2 of 140 mm Hg on an Fio2 of 40% or 300
mm Hg on an Fio2 of 100%
Normal chest x-ray
Sputum free of bacteria, fungus, or significant
numbers of white blood cells on Gram and fungal
staining
Bronchoscopy showing absence of purulent secretions
or signs of aspiration
Absence of thoracic trauma
HIV negative
Donor Management




Maintenance of haemodynamic stability and pulmonary function. Patients suffering from
acute brain injury are often haemodynamically unstable due to neurogenic shock,
excessive fluid losses, and bradycardia. Donor lungs are prone to neurogenic pulmonary
oedema, aspiration, nosocomial infection, and contusion. Continuous arterial and central
venous pressure monitoring, judicious fluid resuscitation, vasopressors, and inotropes are
usually required.
Meticulous fluid management prevents intra-operative blood pressure instability and
minimizes the need for inotropes and vasopressors that stress the myocardium.
Intravascular volume replacement should be given to maintain the central venous
pressure between 5 and 8 mm Hg, though fluids should not be administered at rates far
in excess of hourly urine output. In general, crystalloid fluid boluses are to be avoided.
Diabetes insipidus is common in organ donors and requires the use of intravenous
vasopressin (0.8 to 1.0 IU/hr) to reduce excessive urine losses.
Maintain adequate perfusion pressures: dopamine is the standard inotropic agent used,
although alpha agonists (e.g. phenylephrine) are often appropriate. Blood transfusions
should be used sparingly to maintain the haemoglobin concentration around 10 g/dL to
ensure adequate myocardial oxygen delivery. CMV-negative and leukocyte-filtered blood
should be used whenever possible. Hypothermia should be avoided because it
predisposes to ventricular arrhythmias and metabolic acidosis.
Mechanical ventilation: Fio2 values in excess of 40%, especially 100% oxygen
"challenges," should be avoided, since these oxygen levels may be toxic to the
denervated lung. Ventilator settings should include positive end-expiratory pressures
(PEEP) between 3 and 5 cm H2O to prevent atelectasis.
Donor Operation HLTx/LTx

Median sternotomy
Both pleural spaces are opened immediately with inspection of the lungs and
pleural spaces, particularly in cases of trauma. The lungs are briefly deflated, and
the pulmonary ligaments are divided inferiorly using electrocautery. The thymic
remnant is completely excised.
Pericardium is opened vertically and laterally on the diaphragm and cradled during
dissection of the great vessels and trachea. Umbilical tapes are placed around the
ascending aorta and venae cavae.
The pericardium overlying the trachea is incised vertically, and the trachea is encircled
with an umbilical tape between the aorta and superior vena cava at the highest
point possible and at least four rings above the carina. The entire anterior
pericardium is excised back to each hilum.
Donor operation HLTx
Cardioplegia and pulmonoplegia are
infused simultaneously into the
aorta and main pulmonary artery
after aortic cross-clamping.
Application of topical cold
Physiosol follows immediately.
The venae cavae and aorta are
divided, and the heart-lung bloc
is dissected free from the
esophagus and posterior hilar
attachments. After the trachea is
stapled and divided at the
highest point possible, the entire
heart-lung bloc is removed from
the chest.
Organ Preservation and
Transport
Maximum of 6 to 8 hours of ischaemia in lung and heart-lung allografts.
human studies: retrospective studies from the University of Pittsburgh and the University
of Virginia that showed comparable long-term survival and rates of acute rejection and
bronchiolitis obliterans among recipients of grafts with over 6 hours of ischaemia
compared with those with less than 4 or with 4 to 6 hours of ischaemia.
animal studies: successful transplantation of lung allografts with cold ischaemia times up
to 18 hours have been reported. However, it is believed that beyond a certain threshold,
organ ischaemia will likely lead to primary graft failure and/or impaired long-term
function.
The principle of preservation: minimise I/R injury, mediated by reactive oxygen species,
which disrupt the homeostatic mechanisms in myocyte and endothelial cells. As receptors
for leukocyte adhesion molecules are upregulated and leukocyte chemotactic factors are
released, an inflammatory response ensues, leading to cellular injury. Approaches to
minimise I-R injury: donor pre-treatment, specialized preservation solutions, and
recipient treatments.
Hypothermia reduces the tissue's metabolic demand by up to 99%.
Small number of centers, hypothermic preservation includes donor core cooling on CPB.
Universally, hypothermia is employed during explantation, storage, and implantation.
During explantation, organs are flushed with cold plegic solutions (between 0°C to 10
°C, depending on the institution and solution employed). They are stored at 0°C to 10°C,
and during implantation, they are covered with gauze soaked in saline slush or recipients
are cooled through CPB. The optimal temperature for flush and storage of organs
remains unknown, but common practice is to rely on ice bath temperature for
convenience.
Organ preservation and transport




Heart-lung and lung blocs are typically preserved with a cold pulmonary
artery flush in conjunction with standard crystalloid cardioplegic arrest.
A variety of crystalloid flush solutions are used worldwide, and they can
be divided into two categories based on their electrolyte compositions:
intracellular and extracellular. Euro-Collins is the most frequently used.
Prostaglandins are commonly used for donor pretreatment and as an
additive in pulmonary flush solutions. PGE1, a vasodilator, is given to
counteract reflex pulmonary vasoconstriction resulting from the cold flush and
to permit uniform distribution of the perfusate throughout the lung.
Experimental studies also suggest that PGE1 treatment may minimize
reperfusion injury through its anti-inflammatory properties.
Another commonly used donor pretreatment strategy is steroid
treatment. Experimental evidence suggests that donor lymphocytes may play
a role in ischaemic lung graft injury, so methylprednisolone is given
intravenously to the donor to inactivate them.
Lung graft function is improved when the explanted organ is inflated, when
100% oxygen is used for inflation, and when the lung is transported at 10°C.
Research in the field of lung preservation has recently focused on the role of
various flush and storage solution additives, such as antioxidants, which may
act as free radical scavengers. Other additives that have been shown to
decrease reperfusion injury in research models include nitric oxide donors and
phosphodiesterase inhibitors. Additional areas of research interest include the
development of leukocyte depletion strategies, examining the role of gene
therapy in modifying donor organ susceptibility to ischemia-reperfusion injury,
and the development of colloid-based perfusates.
HLTx RECIPIENT OPERATION



The recipient operation in heart-lung and lung transplantation
proceeds in two phases.
The first is excision of the native organ(s) and the second is
implantation of the allograft. Cardiopulmonary bypass is mandatory
in heart-lung transplantation and occasional in single and bilateral
lung transplantation. At all times, it should be available as stand-by.
Anaesthetic monitoring includes arterial pressure monitoring, pulse
oximetry, continuous electrocardiography, pulmonary artery catheter
monitoring, temperature monitoring, and urine output monitoring.
The use of double-lumen endotracheal tubes is particularly helpful,
allowing for single lung ventilation during certain portions of the
dissection. Large bore intravenous lines are placed for volume
infusion. TOE is often performed during the procedure.
HLTx recipient operation



The recipient is positioned supine on the operating table. The chest is entered through a median
sternotomy, a sternal retractor is placed, and both pleural spaces are opened anteriorly from the level
of the diaphragm to the level of the great vessels. Any pleural adhesions are divided using
electrocautery. In patients in whom dense pleural adhesions are anticipated, such as those with
previous thoracotomies or cystic fibrosis, a bilateral "clamshell" thoracotomy is performed. Combined
with the use of perioperative antifibrinolytic therapy (e.g., aprotinin) and an argon beam coagulator, this
approach improves exposure and facilitates both lysis of adhesions and hemostasis. The anterior
pericardium is excised, while lateral segments are preserved to support the heart and protect the
phrenic nerves. A 3-cm border of the pericardium should be left both anteriorly and posteriorly to each
phrenic nerve extending from the level of the diaphragm to the level of the great vessels. After fully
heparinizing the recipient, the ascending aorta is cannulated near the base of the innominate artery,
and the venae cavae are individually cannulated laterally and snared. Cardiopulmonary bypass with
systemic cooling to 28°C to 30°C is instituted, and the heart is excised at the midatrial level. The aorta
is divided just above the aortic valve, and the pulmonary artery is divided at its bifurcation. The left
atrial remnant is then divided vertically at a point halfway between the right and left pulmonary veins.
The posterior edge of the left atrial and pulmonary venous remnant is developed in a manner that
allows the left inferior and superior pulmonary veins to be displaced over into the left chest. Following
division of the pulmonary ligament, the left lung is moved into the field, allowing full dissection of the
posterior aspect of the left hilum, being careful to avoid the vagus nerve posteriorly. Once this is
completed, the left main pulmonary artery is divided, and the left main bronchus is stapled with a TA30 stapler and divided. The same technique of hilar dissection and division is repeated on the right
side, and both lungs are removed from the chest.
The native main pulmonary artery remnant is removed, leaving a portion of the pulmonary artery intact
adjacent to the underside of the aorta near the ligamentum arteriosum to preserve the left recurrent
laryngeal nerve. Attention is then turned to preparing the distal trachea for anastomosis. The stapled
ends of the right and left bronchi are grasped and dissection is carried up to the level of the distal
trachea. Bronchial vessels are individually identified and carefully ligated. Patients with congenital heart
disease and pulmonary atresia or severe cyanosis secondary to Eisenmenger's syndrome may have
large mediastinal bronchial collaterals that must be meticulously ligated. Perfect hemostasis is
necessary in this area of the dissection, because it is obscured once graft implantation is completed.
Once absolute hemostasis is achieved, the trachea is divided at the carina with a no. 15 blade. The
chest is now prepared to receive the heart-lung graft.
The donor heart-lung bloc is removed from its transport container and prepared by irrigating,
aspirating, and culturing the tracheobronchial tree and by trimming the trachea to leave one
cartilaginous ring above the carina. The heart-lung graft is then lowered into the chest, passing the
right lung beneath the right phrenic nerve pedicle. The left lung is then gently manipulated under the
left phrenic nerve pedicle. The tracheal anastomosis is performed using continuous 3-0 polypropylene
suture. The posterior membranous portion of the anastomosis is performed first, followed by
completion of the anastomosis anteriorly. The lungs are then ventilated with room air at half-normal
tidal volumes to inflate the lungs and reduce atelectasis. Topical cooling with a continuous infusion of
cold Physiosol into both thoraces is begun. To augment endomyocardial cooling and to exclude air from
the graft, a third cold "bubble-free" line is placed directly into the left atrial appendage.
Next, the bicaval venous anastomosis is performed. The recipient inferior vena cava is anastomosed to
the donor inferior vena cava-right atrial junction with a continuous 4-0 polypropylene suture. At this
point the patient is rewarmed toward 37°C, and the superior vena caval and aortic anastomoses are
performed end-to-end with continuous 4-0 polypropylene sutures. After the ascending aorta and
pulmonary artery are cleared of air, the aortic cross-clamp and caval tapes are removed. The left atrial
catheter is removed, and the atrium is allowed to drain. The amputated left atrial stump is oversewn,
and the pulmonoplegia infusion site on the pulmonary artery is closed. The heart is defibrillated, and
the patient is gradually weaned from cardiopulmonary bypass in the standard fashion.
Methyprednisolone (500 mg) is administered to the recipient following heparin reversal with protamine
sulfate.
PEEP at 3 to 5 cm H2O and an Fio2 of 40% are maintained. As in cardiac transplantation, isoproterenol
(0.005 to 0.01 µg/kg/min) is usually initiated on graft reperfusion to increase the heart rate to about
100 to 110 bpm and to lower pulmonary vascular resistance. Temporary right atrial and ventricular
pacing wires are placed. Right and left pleural chest tubes (right angle) are placed along each
diaphragm, as well as one mediastinal tube. The chest is closed in the standard fashion. Finally, the
double-lumen endotracheal tube is exchanged for a single-lumen tube and the tracheal anastomosis is
checked endoscopically before transporting the patient to the intensive care unit.
Lick et al at the University of Texas and the University of Arizona have recently described an interesting
alternative to the standard technique in which the pulmonary hila are placed anterior to the phrenic
nerves and direct caval anastomoses are used whenever feasible. This modification obviates extensive
dissection of the phrenic nerves and posterior mediastinum, decreasing the likelihood of phrenic and
vagus nerve injury. Furthermore, the posterior mediastinum can be inspected more easily for bleeding
after implantation by rotating the heart-lung bloc anteriorly and medially while still on bypass.
Bilateral Lung Transplantation
recipient operation


Sequential single lung transplants through a bilateral anterior
thoracosternotomy (clamshell) incision is made at the level of
the fourth intercostal space. The lung with the least amount
of function (as determined by a preoperative ventilationperfusion scan) is removed first and replaced with an allograft
as described for single lung transplantation above. Once
ventilation and perfusion are established in the first allograft,
the second native lung is explanted and the second allograft is
implanted. Bilateral chest tubes are placed and the chest is
closed. Bronchoscopy is performed to evaluate the bronchial
anastomoses.
Many centers use CPB routinely during bilateral lung
transplantation. It allows for improved exposure, shorter graft
ischemic times, controlled reperfusion, and the use of
leukocyte-depleting filters. Because the risk of bleeding may
be increased with CPB, strategies have been developed to
minimize the chance of haemorrhage. These include the
routine use of aprotinin and heparin-coated CPB circuits, as
well as the availability of an argon beam coagulator.
Single LTx recipient operation





If possible, the lung with the least function determined by preoperative ventilation-perfusion scan is
selected for replacement. The patient is placed in a standard thoracotomy position, with access to the
groin should CPB be needed. A posterior lateral thoracotomy is made at the level of the fourth or fifth
intercostal space. Adhesions are lysed and the hilar dissection is performed. The pulmonary artery, the
superior and inferior pulmonary veins, and the mainstem bronchus are isolated. A trial occlusion of the
pulmonary artery is used to determine whether the procedure will be tolerated without CPB. If it is
tolerated, the pulmonary artery is ligated and divided distal to the upper lobe branch. The pulmonary
veins are also ligated and divided. The mainstem bronchus is stapled and divided, and the native lung is
explanted.
The donor lung is removed from its transport container and prepared for implantation. The donor
bronchus is opened and secretions are aspirated and cultured. The bronchus is trimmed, leaving two
cartilaginous rings proximal to the orifice of the upper lobe. Any remaining pericardial and lymphatic
tissue is removed, and the left atrial cuff is trimmed as needed. The donor lung is then placed in the
recipient's chest and covered with saline slush and iced laparotomy pads.
The sequence of anastomoses is a matter of preference, though most perform the deepest anastomosis
(the bronchial anastomosis) first and then proceed to the more superficial ones. The bronchial
anastomosis is fashioned with 4-0 polypropylene suture. We favour a continuous suture technique;
alternatively, the membranous portion can be sewn with interrupted suture. Alternatively, the entire
anastomosis can be sewn with a running suture. Variations on the end-to-end bronchial anastomosis
include the use of a telescoping technique, in which the donor bronchus is intussuscepted into the
recipient bronchus, and the placement of an omental pedicle flap around the anastomosis. These
techniques were developed to prevent bronchial anastomotic dehiscence but are now rarely performed.
Once the bronchial anastomosis is complete, attention is then turned to making the pulmonary venous
anastomosis. A side-biting clamp is applied to the left atrium to include the pulmonary veins. The
recipient pulmonary vein stumps are opened and the intervening atrial tissue is cut. This creates a cuff
that is anastomosed to the donor atrial remnant using continuous 4-0 polypropylene suture; this suture
is not tied down until reperfusion. Donor and recipient pulmonary arteries are anastomosed with 5-0
polypropylene suture. Upon graft inflation, kinking can occur if the arteries are left too long, so they
must be carefully trimmed to an appropriate length before fashioning the anastomosis. The pulmonary
artery anastomosis is de-aired. The lung is inflated, and the pulmonary artery clamp is temporarily
released to allow flushing of air through the atrial suture line, and the left atrial clamp is removed to
allow retrograde de-airing of the atrial anastomosis. The pulmonary venous anastomosis is then
secured.
After haemostasis is ensured, apical and basal chest tubes are inserted. The ribs are reapproximated
and the chest is closed in a standard fashion. The double-lumen endotracheal tube is exchanged for a
single-lumen tube and bronchoscopy is performed to evaluate the bronchial anastomosis.
Arguments pro- and con
CPB in LTx
Pro:
 improved exposure of the hilar strutures, which is particularly
helpful in patients with dense adhesions and bronchial
collaterals
 allows for early pneumonectomies without hemodynamic or
respiratory instability
 ischemic time of the second lung is substantially reduced
when compared to off-CPB bilateral lung transplants
 Its use also prevents overperfusion of the first lung graft with
the entire cardiac output
 In patients with suppurative lung disease, the use of CPB
facilitates careful washout of the distal trachea and proximal
bronchi to prevent contamination of the first implanted lung.
Con:
 increased blood loss, transfusion needs, and reperfusion
injury.
POSTOPERATIVE MANAGEMENT
Heart-Lung and Lung Graft Physiology

Denervation of the lungs results in diminished cough reflex as well as impaired
mucociliary clearance mechanisms. This predisposes recipients to pulmonary infections
and necessitates aggressive postoperative pulmonary toilet.


Moreover, ischaemia and reperfusion injury in the transplanted lung, along with disrupted
pulmonary lymphatics, can result in increased vascular permeability and varying degrees
of interstitial oedema. For the heart-lung recipients, denervation of the cardiac allograft
leads to additional physiologic characteristics. The denervated heart has lost its
sympathetic and parasympathetic autonomic regulation; therefore recipients of heartlung grafts do not have normal autonomic regulation of heart rate, contractility, or
coronary artery caliber. The resting heart rate is generally higher due to the absence of
vagal tone. Respiratory sinus arrhythmia and carotid reflex bradycardia are absent.
Interestingly, the denervated heart develops an increased sensitivity to catecholamines;
this is due to an increase in beta-adrenergic receptor density and a loss of
norepinephrine uptake in postganglionic sympathetic neurons.
This augmented sensitivity plays an important role in maintaining an adequate cardiac
response to exercise and stress. During exercise, the recipient experiences a steady but
delayed increase in heart rate, primarily due to a rise in circulating catecholamines. This
initial rise in heart rate is subsequently accompanied by an immediate increase in filling
pressures resulting from augmented venous return. These changes lead to an
augmentation of stroke volume and cardiac output sufficient to sustain an increase in
activity. The ability of the coronary circulation to dilate and increase blood flow in
response to increased myocardial oxygen demand is normal in cardiac transplant
recipients and would likewise be expected to be so in recipients of heart-lung grafts.
Conversely, graft coronary vasodilator reserve is abnormal in the presence of rejection,
hypertrophy, or regional wall abnormalities.
Clinical Management in the Early
Postoperative Period




The primary objective in heart-lung and isolated lung graft recipients in the immediate postoperative
period is to maintain adequate perfusion and gas exchange in the recipient while minimizing
intravenous fluid administration, cardiac work, and barotrauma.
Cardiac rhythm and arterial and central venous pressures are monitored. Strict isolation precautions,
previously enforced to reduce the incidence of infection in these immunosuppressed patients, are no
longer required; simple handwashing is now considered sufficient.
Approximately 10% to 20% of heart-lung graft recipients experience some degree of transient sinus
node dysfunction in the immediate perioperative period, often manifested as sinus bradycardia, which
usually resolves within a week. The use of bicaval venous anastomoses has been reported to lower the
incidence of sinus node dysfunction and improve tricuspid valve function. Because cardiac output is
primarily rate dependent after heart-lung transplantation, the heart rate should be maintained between
90 and 110 bpm during the first few postoperative days using temporary pacing or isoprenalinel
(0.005–0.01 µg/kg/min) as needed. Although rarely seen, persistent sinus node dysfunction and
bradycardia may require a permanent transvenous pacemaker. The systolic blood pressure should be
maintained between 90 and 110 mm Hg using afterload reduction in the form of nitroglycerin or
nitroprusside if necessary. Renal-dose dopamine (3–5 µg/kg/min) is used frequently to augment renal
blood flow and urine output. The adequacy of cardiac output is indicated by warm extremities and a
urine output great than 0.5 mL/kg/hr without diuretics. Cardiac function generally returns to normal
within 3 to 4 days, at which time parenteral inotropes and vasodilators can be weaned.
Depressed global myocardial performance in the acute postoperative setting. The myocardium is
potentially subject to prolonged ischemia, inadequate preservation, or catecholamine depletion prior to
implantation. Hypovolemia, cardiac tamponade, sepsis, and bradycardia may also be contributory and
should be treated expeditiously if they are present. A Swan-Ganz pulmonary artery catheter should be
used in cases of persistently abnormal haemodynamics.
Ventilatory management is a key element in the postoperative management of both heart-lung and isolated lung graft
recipients. Barotrauma and high airway pressures that might compromise bronchial mucosal flow should be avoided. Lower
tidal volumes and flow rates may be necessary to limit peak airway pressures to less than 40 cm H2O. Upon arrival to the
ICU, an anteroposterior chest x-ray is obtained, and the ventilator is typically set to an Fio2 of 50%, tidal volume of 10 to
15 mL/kg, an assist-control rate of 10 to 14 breaths per minute, and PEEP of 3 to 5 cm H2O. These settings are adjusted
every 30 minutes to achieve an arterial Po2 greater than 75 mm Hg on an Fio2 of 40%, an arterial carbon dioxide pressure
(Paco2 between 30 and 40 mm Hg, and a pH between 7.35 and 7.45. Pulmonary toilet with endotracheal suctioning is an
effective means of reducing mucous plugging and atelectasis. Ventilatory weaning is initiated after the patient is deemed
stable, awake, and alert. Usually, weaning is accomplished through successive decrements in intermittent mandatory
ventilation rate followed by a trial of continuous positive airway pressure. Once ventilatory mechanics and arterial blood
gases are deemed acceptable, the patient is extubated. This usually occurs within the first 24 hours after transplantation.
Subsequent pulmonary care consists of vigorous diuresis, supplemental oxygen for several days, continued aggressive
pulmonary toilet and incentive spirometry, and serial chest x-rays.
A diffuse interstitial infiltrate is often found on early postoperative chest x-rays. Previously referred to as a reimplantation
response, this finding is better defined as graft edema due to inadequate preservation, reperfusion injury, or early rejection.
It appears that the degree of pulmonary edema is inversely related to the quality of preservation. Judicious administration
of fluid and loop diuretics is required to maintain fluid balance and minimize this pulmonary edema.
Early lung graft dysfunction manifested by persistent marginal gas exchange without evidence of infection or rejection
occurs in less than 15% of transplants.This primary graft failure is often the result of ischemia-reperfusion injury and is
manifested histologically by diffuse alveolar damage. Of course, technical causes of graft failure, such as pulmonary venous
anastomotic stenosis or thrombosis, must always be considered. In cases of persistent severe pulmonary graft dysfunction
refractory to mechanical ventilatory maneuvers, extracorporeal membrane oxygenation (ECMO) and inhaled nitric oxide
have been used successfully to stabilize gas exchange in several patients. In others, urgent retransplantation has been
performed.
Expedient removal of vascular lines has been shown to reduce the incidence of line sepsis. Pleural and mediastinal chest
tubes are removed when drainage has fallen to less than 25 mL/h. For heart-lung graft recipients, pacing wires are removed
between 7 and 10 days after transplantation, provided that pacing is not required. After several days, barring significant
complications, the patient is transferred from the ICU to a standard cardiothoracic surgical ward for the remainder of the
hospital stay.
Immunosuppressive Management:
Early and Late Postoperative Regimens
For heart-lung and lung graft recipients, immunosuppression begins intraoperatively and is
continued for the patient's lifetime. The conventional triple-drug combination consists of
cyclosporine, azathioprine, and prednisone. Initially, high doses of these drugs are given,
and they are later tapered for chronic administration.
Cyclosporine is initiated in the early postoperative period, initially intravenously (0.05–0.1
mg/kg/h) and subsequently orally when oral intake is well established (5–10 mg/kg/d in
two divided doses). Dosing is titrated to maintain a trough serum concentration between
150 and 250 ng/mL in the first few weeks after transplantation and from 100 to 150
ng/mL thereafter.
Azathioprine is administered intravenously at 4 mg/kg preoperatively and subsequently
maintained at approximately 2 to 3 mg/kg/d. Azathioprine dosages are adjusted to
maintain the white blood cell count greater than 4000 cells/mm3.
Methylprednisolone is administered intraoperatively at graft reperfusion (500 mg
intravenously) and then continued for the first 24 hours at 125 mg intravenously every 8
hours. Steroids are then suspended for 2 weeks, based on experimental and clinical
evidence that they impede bronchial anastomotic healing. After 2 weeks, prednisone is
started at a daily oral dose of 0.6 mg/kg and gradually tapered over the next 3 to 4
weeks to 0.1 to 0.2 mg/kg/d.
The conventional triple-drug combination of cyclosporine,
azathioprine, and prednisone is modified at some centers.
Tacrolimus (FK506) and mycophenolate mofetil are two drugs
that have been used widely in kidney and liver
transplantation; experience with use of these drugs in heartlung and lung transplant recipients is limited.
Many centers have added prophylactic induction therapy to the
standard triple-drug regimen. This includes the use of OKT3,
antithymocyte globulin (RATG and ATGAM), and daclizumab.
OKT3 is a murine monoclonal antibody preparation that
recognizes the CD3 antigen of human T cells.
Side effects: Cyclosporine is commonly associated with
nephrotoxicity, hypertension, hepatotoxicity, hirsutism, and an
increased incidence of lymphoma. The primary toxicity of
azathioprine is generalized bone marrow depression, which
manifests as leukopenia, anemia, and thrombocytopenia.
Steroids are associated with a myriad of side effects, including
the development of cushingoid features, hypertension,
diabetes, osteoporosis, and peptic ulcer disease. Initial doses
of OKT3 and antithymocyte globulin can be associated with a
"cytokine release syndrome"; significant hypotension,
bronchospasm, and fever can result. Therefore, patients
receiving these induction agents are premedicated with
acetominophen, antihistamines, and corticosteroids, and are
monitored closely. Interestingly, daclizumab is not associated
with the cytokine release syndrome.
Infection Prophylaxis
Antiviral and antifungal prophylaxis are important components of
postoperative management in heart-lung and lung transplant
recipients.
Cytomegalovirus prophylaxis (CMV) with ganciclovir is employed by
many centers in any CMV-positive recipient and in any CMV-negative
recipient receiving an allograft from a CMV-positive donor. Ganciclovir
is typically given for a several week course, and can be associated
with leukopenia. Some patients may require G-CSF if their white
blood cell count falls below 4000.
Fungal prophylaxis against mucosal Candida infection includes use of
daily nystatin swish and swallow.
Pneumocystis carinii prophylaxis consists of trimethoprim-
sulfamethoxazole or aerosolized pentamidine. In the immediate
postoperative period, Aspergillus colonization is inhibited by the use
of aerosolized amphotericin B. For Toxoplasma-negative recipients of
grafts from Toxoplasma-positive patients, pyrimethamine prophylaxis
is maintained for the first 6 months after transplantation.
Graft Surveillance: Patient
Follow-Up Schedule


Routine clinical follow-up for heart-lung and lung allograft recipients
is required to monitor graft function and modify immunosuppressive
regimens. Regular surveillance protocols have been developed to
monitor graft function, and these typically consist of serial pulmonary
function tests, arterial blood gases, and bronchoscopic evaluation
at 2 weeks, 4 to 6 weeks, 12 weeks, and 6 months after
transplantation, and yearly thereafter. Transbronchial biopsies are
obtained from each transplanted lung, and lavage specimens are
submitted for staining (i.e., Gram, fungal, acid-fast bacillus, and
silver), culture, and cytology. Surveillance endomyocardial
biopsies are performed at 3 months and then annually in heart-lung
graft recipients.
In addition to routine surveillance, follow-up is often needed to
address changes in clinical status. Complications related to
transplantation are many, and these must be addressed carefully and
expediently to prevent long-term graft failure.
POSTOPERATIVE
COMPLICATIONS








Haemorrhage
Early Graft Dysfunction and Primary
Graft Failure
Hyperacute Rejection
Acute Rejection
Chronic Rejection
Airway Complications
Infection
Neoplasm
Causes of death HLTx and
LTx
Long-term survival HLTx and LTx