SlideShare ist ein Scribd-Unternehmen logo
1 von 22
Downloaden Sie, um offline zu lesen
THE PRESENT AND FUTURE
STATE-OF-THE-ART REVIEW
Cardiac Pacemakers: Function,
Troubleshooting, and Management
Part 1 of a 2-Part Series
Siva K. Mulpuru, MD, Malini Madhavan, MBBS, Christopher J. McLeod, MBCHB, PHD, Yong-Mei Cha, MD,
Paul A. Friedman, MD
ABSTRACT
Advances in cardiac surgery toward the mid-20th century created a need for an artificial means of stimulating the
heart muscle. Initially developed as large external devices, technological advances resulted in miniaturization of
electronic circuitry and eventually the development of totally implantable devices. These advances continue to date,
with the recent introduction of leadless pacemakers. In this first part of a 2-part review, we describe indications,
implant-related complications, basic function/programming, common pacemaker-related issues, and remote
monitoring, which are relevant to the practicing cardiologist. We provide an overview of magnetic resonance imaging
and perioperative management among patients with cardiac pacemakers. (J Am Coll Cardiol 2017;69:189–210)
© 2017 by the American College of Cardiology Foundation.
A BRIEF HISTORY OF
CARDIAC PACING
Cardiac pacing, electrical stimulation to modify or
create cardiac mechanical activity, began in the 1930s
with Hyman’s “artificial pacemaker” (his term), in
which a hand crank created an electric current that
drove a DC generator whose electrical impulses were
directed to the patient’s right atrium through a needle
electrode placed intercostally. At that time, Hyman
faced professional skepticism, litigation, and accusa-
tions of creating “an infernal machine that interferes
with the will of God,” and he never found a manu-
facturer for his machine (1).
After World War II, public perception changed and
daring pioneers made great advances. Large,
external, alternating current–powered pacemakers
tethered to an extension cord gave way to battery-
powered, transistorized, “wearable” pacemakers
(Central Illustration). The birth of pacing was linked to
cardiac surgery, which was burgeoning. In 1957, at the
University of Minnesota, C. Walton Lillehei had per-
formed over 300 open-heart operations on young
adults and children with congenital defects. Dr. Lil-
lehei and coworkers developed a myocardial wire for
post-operative pacing. On October 31, 1957, a munic-
ipal power failure in Minneapolis lasted 3 h and led to
the tragic death of a baby (2). The following day, Dr.
Lillehei asked Earl Bakken, a hospital equipment en-
gineer, to build a battery-powered device to prevent
future tragedies. Bakken modified a circuit for an
electronic metronome he had seen in the April 1956
issue of Popular Electronics that used transistors,
which had been invented 10 years before. He modi-
fied the 2-transistor circuit so that the electrical pul-
ses would pace the heart, rather than power a
speaker. The device was immensely successful. He
named the company he founded Medtronic. Other
innovations would lead to the founding of other, now
familiar manufacturers.
From the Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota. Dr. Friedman has received research support
from St. Jude Medical; and is a consultant and advisory board member for Medtronic and Boston Scientific. All other authors have
reported that they have no relationships relevant to the contents of this paper to disclose.
Manuscript received June 29, 2016; revised manuscript received October 6, 2016, accepted October 18, 2016.
Listen to this manuscript’s
audio summary by
JACC Editor-in-Chief
Dr. Valentin Fuster.
J O U R N A L O F T H E A M E R I C A N C O L L E G E O F C A R D I O L O G Y V O L . 6 9 , N O . 2 , 2 0 1 7
ª 2 0 1 7 B Y T H E A M E R I C A N C O L L E G E O F C A R D I O L O G Y F O U N D A T I O N
P U B L I S H E D B Y E L S E V I E R
I S S N 0 7 3 5 - 1 0 9 7 / $ 3 6 . 0 0
h t t p : / / d x . d o i . o r g / 1 0 . 1 0 1 6 / j . j a c c . 2 0 1 6 . 1 0 . 0 6 1
In 1960, Rune Elmqvist and Ake Senning
of Stockholm placed the first fully implant-
able pacemaker in Arne Larsson. Larsson’s
wife had pleaded with Senning to use the
experimental technology to help her
desperately ill husband, who had complete
heart block and frequent Stokes-Adams
syncopal attacks. To avoid publicity, the
initial implantation was performed at night,
when the operating rooms were empty.
The original system lasted 8 h. Arne Larsson
ultimately went on to undergo over
20 pacemaker replacements, and he outlived both
his surgeon and device engineer. He was an advo-
cate for pacing until his death at 86 years of age in
2002 (3).
Although significant advances in pacing technology
were developed over the next 50 years, including
multichamber pacing, rate responsiveness, device size
reduction, internet-based remote monitoring, and
marked increases in battery longevity, the basic sys-
tem paradigm of an extravascular pulse generator
connected to 1 or more leads that traverse the venous
system to contact myocardial tissue would not change
for 50 years (Central Illustration). However, many
pacemaker-related complications (infection, throm-
bosis, lead failure, and pneumothorax) are related to
this basic construct, particularly the leads. This has led
to a paradigm shift: the development of a leadless
pacemaker, in which the entire device is placed within
cardiac chambers. Batteryless devices, which harvest
cardiac motion to power pacing circuits, are on the
horizon as a coming paradigm shift. In this first part of
the 2-part review of cardiac pacing, we explore the
state-of-the-art: the basics of pacing physiology,
pacing modes and indications, periprocedural
management, complications, basic troubleshooting,
perioperative management for nonpacemaker
procedures, and cardiac magnetic resonance imaging
(CMR) of patients with pacemakers. In part 2 of our
review (4), we will examine recent advances and future
directions, including resynchronization for heart
failure, His bundle pacing, remote monitoring, and
leadless and batteryless devices.
BASICS OF CARDIAC PACING
Normal cardiac activity begins in the sinus node,
where cells with intrinsic automaticity act as pace-
maker cells. Electrical wave fronts then spread across
the atria to the atrioventricular (AV) node, which they
pass through to enter the His-Purkinje system to
rapidly spread to and depolarize the ventricles
(Figure 1). When intrinsic cardiac automaticity or
CENTRAL ILLUSTRATION An Overview of the History of Cardiac Pacing
Paradigm Shifts in Cardiac Pacemakers
1950s
AC-powered
pacemakers
tethered to an
extension cord
(Furman)
1950s
Battery-powered
transistorized
“wearable”
pacemakers
(Lillehei/Bakken)
1958
First fully
implantable
pacemaker
(Elmqvist/
Senning)
2015
Implantable
pacemaker—
basic system
had not evolved
significantly
2016
Leadless
pacemaker—the
entire device is
placed within
cardiac chambers
Future
Batteryless
devices, which
harvest cardiac
motion to power
pacing circuits
Mulpuru, S.K. et al. J Am Coll Cardiol. 2017;69(2):189–210.
Historically, pacing developed using large, external, alternating current (AC)–powered devices, which subsequently evolved to “wearable” transistorized battery
powered pacemakers—both comprise the era of external devices. A paradigm shift occurred with the introduction of the entirely implantable pacemaker, composed of
an extravascular pulse generator connected to a transvenous lead in contact with the myocardium. This paradigm continues to this day. An emerging and rapidly
developing new paradigm is that of leadless pacemakers, which are available for clinical use. Batteryless pacemakers that harvest cardiac mechanical motion to
generate current, or that modify or add cells to introduce biological pacing activity, are under active investigation.
A B B R E V I A T I O N S
A N D A C R O N Y M S
CIED = cardiac implantable
electronic device
CS = coronary sinus
PMT = pacemaker-mediated
tachycardia
PVARP = post-ventricular
atrial refractory period
RV = right ventricle
TARP = total atrial refractory
period
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
190
conduction integrity fails, the electrical excitability of
cardiac tissue allows a small, external electrical
stimulus to drive myocytes to threshold, leading to
depolarization of neighboring myocytes through
energy-consuming biological processes and the
consequent propagation of an electrical wave front,
with near-simultaneous muscular contraction via
excitation-contraction coupling. Pacemakers provide
that external stimulus.
Pacemakers consist of a pulse generator or can,
which contains the battery and electronics, and leads,
which travel from the can to contact the myocardium,
to deliver a depolarizing pulse and to sense intrinsic
cardiac activity (Figure 2). Insulation materials
separate the conductor cables and the lead tip elec-
trodes. Depending on the relationship between the
cables, the leads can be designed as coaxial (a tube
within a tube) or coradial (side-by-side coils). The
lead fixation to the myocardium may be active (with
an electrically active helix at its tip for mechanical
stability) or passive (electrically inert tines anchor the
lead). Disruption of conductor elements and insu-
lation materials results in either high impedance
(fracture) or low impedance due to short-circuiting
(insulation breach), respectively. Pacing occurs
when a potential difference (voltage) is applied be-
tween the 2 electrodes. In bipolar pacing, the poten-
tial difference occurs between the lead tip (cathode)
FIGURE 1 The Cardiac Conduction System
(A) Conduction begins in the SA node (left). Impulse propagation through the atria gives rise to the P-wave (bottom left). The impulse is then
delayed in the AV node to allow blood to flow to the ventricles; wave front travel through the AV node is not seen on the surface elec-
trocardiogram. The wave fronts then pass through the His–Purkinje system to rapidly activate the ventricular myocardium, giving rise to the
large amplitude QRS complex. (B) An anatomic specimen showing the location of key conduction system elements. (Left) An external view of
the heart with the region of the sinoatrial node in the epicardium at the juncture of the SVC and right atrium indicated. The structure itself is
not visible to the naked eye. (Right) The right atrial and ventricular free walls have been removed to reveal the position of the AV node
anterior to the CS and atrial to the TV, situated in Koch’s triangle (bounded by the TV, CS, and tendon of Todaro; not shown). Reproduced with
permission from Hayes et al. (40). AV ¼ atrioventricular; CS ¼ coronary sinus; FO ¼ fossa ovalis; IVC ¼ inferior vena cava; RAA ¼ right atrial
appendage; SA ¼ sinoatrial; SVC ¼ superior vena cava; TV ¼ tricuspid valve.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
191
and a proximal ring (anode). With unipolar pacing,
current is delivered between the lead tip and the
pulse generator can. The minimum amount of energy
required to depolarize myocardium is called the
stimulation threshold. The delivered stimulus is
described by 2 characteristics: its amplitude
(measured in volts) and its duration (measured in
milliseconds). The energy required to pace the
myocardium depends on the programmed pulse
width and on the voltage delivered between the
electrodes. An exponential relationship (strength-
duration curve) exists between the stimulation
threshold and the pulse amplitude and duration
(Figure 3). This is clinically relevant, in that opti-
mizing the pulse width and amplitude can signifi-
cantly affect current drain and battery longevity.
Another clinical use for these parameters includes
reprogramming to prevent extracardiac (e.g., phrenic)
stimulation by lowering the pacing voltage to mini-
mize the risk of far-field capture and increasing the
pulse width to ensure cardiac stimulation. At im-
plantation, a typical acceptable threshold is under
1.5 V with a pulse width of 0.5 ms, but this may vary,
and higher values are accepted for coronary sinus
leads. Pacemaker leads are built with steroid-eluting
collars to prevent tissue-lead fibrosis and to mitigate
the threshold rise over time that is otherwise seen.
The relationship among voltage, current, and
resistance is defined by Ohm’s Law (V ¼ IR), where
V ¼ voltage, I ¼ current, and R ¼ resistance. Classi-
cally, Ohm’s law uses impedance, which includes the
effects of inductance and capacitance in opposing
flow; however, for clinical purposes, these are
generally negligible and resistance alone is used.
Pacing lead conductors are designed to have a low
internal resistance, to minimize wastage of energy as
resistive heat. Because permanent pacemakers
generate a constant voltage, the higher the pacing
resistance (the load, resistance of current passage
through tissue) the lower the current drain (I ¼ V/R),
and the lower the rate of battery depletion per each
pacing pulse. Thus, lead tip electrodes are optimized
to have a relatively high resistance (typically 400 to
1,200 U) to minimize current flow and preserve bat-
tery (1). Up to 50% of the current drain from the
battery is used for pacing, whereas the other one-half
is used for sensing and housekeeping functions
(algorithms and storage of electrograms) (5).
FIGURE 2 Transvenous Pacemaker System and its Associated Components
(A) A pacemaker system primarily consists of a hermitically encased can containing the battery and all of the circuitry placed in the pre-
pectoral region. The can is connected to the myocardial tissue by a pacemaker lead. The leads contain conductor coils to the distal electrodes
separated by insulation material. (B) The leads are of coaxial (coil within a coil) or coradial (side-by-side coils) design, depending on the
arrangement of the conductor coils. (C) The lead tips are attached to the myocardium by a penetrating helix (active fixation) or by tines that
embed in the myocardial trabeculations (passive fixation). Reproduced with permission from Hayes et al. (40).
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
192
Resistance is affected by many factors: lead-tissue
interface, body position, and tissue edema, to name
a few, but abrupt changes (>30%) may suggest lead
malfunction.
Initial pacemakers functioned as pacing-only de-
vices without the capability to sense intrinsic cardiac
activity. Asynchronous pacing provided a minimum
heart rate, but was complicated by atrial and ven-
tricular dissociation, and by competition between
pacing impulses and intrinsic cardiac activity. This
led to the development of sensing and demand-
pacing modes (discussed later). Sensing is the pro-
cess whereby a pacemaker determines the timing of
the cardiac depolarization of the chamber that a lead
is in. To effectively sense, the pacemaker must opti-
mally sense near-field depolarization signals, reject
near-field repolarization signals (T-waves), and reject
far-field signals (signals generated by tissues that the
lead electrode is not in contact with) as well as non-
physiological signals (electromagnetic interference
generated by cell phones, and so on). Atrial channels
are optimized to sense in the frequency range of 80 to
100 Hz, and ventricular channels in the 10- to 30-Hz
range (6,7). Typical amplitude ranges for signals
recorded from atrial and ventricular leads are 1.5 to 5
mV and 5 to 25 mV, respectively. Electrogram ampli-
tudes below these values may lead to undersensing,
the failure to detect cardiac depolarization, with
possible inappropriate delivery of pacing pulses
(Figure 4).
PACING MODE
In response to a sensed intracardiac signal, a pace-
maker may inhibit output, trigger output, or pace in a
different chamber after a timed delay. This function is
governed by the programmed pacing mode. The
pacing mode is described with a 4- or 5-letter code
(e.g., DDDR), in which the first position identifies the
chamber paced (A for atrium, V for ventricle, D for
dual/both), the second position indicates the chamber
sensed, the third position denotes the device
response to sensed events (I for inhibit, T for trigger,
or D for dual [both]), the fourth position indicates
whether rate response is on, and the fifth position
(when used), indicates whether multisite pacing is
employed in the atrium (A), ventricle (V), or both (D).
With regard to the device response (position 3 in
the code), inhibition indicates that a sensed event
inhibits pacing and initiates a new timing cycle. If the
timing cycle (the length of which is determined by the
programmed pacing rate) elapses before another
event is sensed, then pacing will occur. This is most
commonly used with single-chamber pacing, such as
VVI or VVIR (also called demand pacing); the presence
of an intrinsic depolarization above the pacing rate
inhibits pacing. If the intrinsic rate drops below the
programmed rate, pacing occurs. Rate response (dis-
cussed in the following text) detects physical activity,
such as exercise, and functionally increases the lower
rate (shortens the cycle length) for pacing. With
triggered pacing, a sensed event may trigger pacing in
the same chamber or, typically after a programmed
delay, in the other chamber. A triggered mode alone is
uncommonly used. With the dual mode (e.g., DDDR)
both triggering and inhibition are used. In the DDD
mode (Figure 5), inhibition occurs in the atrium if the
intrinsic atrial rate exceeds the programmed lower
rate. An atrioventricular clock is then started. In the
absence of an intrinsic ventricular event, a ventricu-
lar pacing spike is triggered; a sensed intrinsic ven-
tricular event inhibits pacing. In all pacing modes, a
lower rate limit indicates the rate below which pacing
occurs (this is the slowest heart rate that should be
present, although some features and algorithms may
permit programmable exceptions), and an upper rate
limit indicates the fastest rate at which the pacemaker
will pace, although intrinsic cardiac activity has no
such limit. Common pacing modes and their clinical
utility follow:
FIGURE 3 Relationships Among Chronic Ventricular Strength–Duration Curves From a
Canine, Expressed as Potential, Charge, and Energy
0
0
1
2
3
4
5
0.2 0.4 0.6 0.8 1.0
Strength Duration Curve
Charge
Energy
1.5
Pulse Width (ms)
Threshold(V,μJ,μC)
With longer pulse widths, less voltage is required to stimulate the heart. The energy
requirements, which are a function of the programmed pulse width and voltage, are
larger at very short and very long pulse widths. The energy expended is lower when the
pulse width is kept constant at 0.4 to 0.5 ms, and the voltage is adjusted to have an
adequate safety margin. Reprinted with permission from Stokes et al. (41).
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
193
DDD. Standard dual-chamber pacing is used when
the sinus mode is intact, but AV conduction impaired.
Sinus activity is sensed and will trigger ventric-
ular pacing following a programmed AV delay
(p-synchronous pacing).
DDDR. Rate response is added when sinus and AV
nodal function are both abnormal; the rate responsive
feature provides chronotropic response. Most modern
devices use sensors to determine the rate respon-
siveness to physiological demands.
VVI AND VVIR. Ventricular-only pacing is used in
patients with chronic atrial fibrillation, or infrequent
pauses or bradycardias. The potential for tracking
atrial arrhythmias is eliminated. Rate response
provides chronotropic support when needed.
Single-chamber pacemakers with leads in the
ventricle can deliver these modes.
AAIR. This mode is reserved for isolated sinus node
dysfunction with intact AV nodal conduction. It
avoids ventricular pacing and, when delivered by a
single-chamber pacemaker, eliminates the need for a
lead that crosses the tricuspid valve.
VOO/DOO. Asynchronous modes are programmed to
avoid recognition of electrical activity, most
commonly electrocautery, CMR signals, or other
electromagnetic interference (EMI). These modes
prevent sensing of extrinsic electrical activity, which
may be “misinterpreted” as native cardiac events,
inhibiting pacing, or lead to rapid ventricular pacing
up to the upper rate limit if sensing occurs on the
FIGURE 4 Sensing Abnormalities in Modern Pacemakers
(A) Myopotential oversensing (star) and inhibition of pacing in a patient with a pacemaker programmed to VVI mode. (B) Atrial undersensing
on a tracing from an ambulatory monitor. This is best seen at the fifth ventricular complex. A P-wave precedes the intrinsic ventricular event,
but this is not sensed (green arrow), and an atrial pacing artifact (black arrow) occurs immediately before the intrinsic ventricular complex.
The ventricular complex is sensed in the cross-talk sensing window. As a result, the ventricular pacing artifact (red arrow) is delivered early
after the intrinsic ventricular event (i.e., ventricular safety pacing).
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
194
atrial lead. In current clinical practice, these modes
are only used temporarily to prevent oversensing.
BASIC PROGRAMMABLE FEATURES
Several basic programmable features are important
for arrhythmia management.
MODE SWITCHING. In dual-chamber systems, an
atrial tachyarrhythmia sensed in DDD mode can lead
to ventricular pacing at rates up to the upper rate
limit because atrial events are tracked in the
ventricle. Mode-switching algorithms identify the
presence of an atrial tachyarrhythmia and switch to a
nontracking mode (VVI, DVI, or DDI, with or without
rate response). Although the specific algorithms vary,
the presence of more atrial events than ventricular
events at a rate above a programmable value triggers
a mode switch, with the pacing rate then driven by
the programmed lower rate limit or the sensor-
indicated rate. Following a mode-switch event,
atrial activity is scanned so that a tracking mode re-
sumes upon arrhythmia termination. Mode-switching
events generate internet-based alerts when available
(discussed in part 2 [4]), and are reported upon device
interrogation. Mode-switch events indicate the pres-
ence of atrial arrhythmias and potential need for
anticoagulation. Inappropriate mode switching can
occur with far-field R-wave oversensing (Figure 6).
AVOIDING VENTRICULAR PACING. Isolated right
ventricular (RV) pacing activates the interventricular
septum before the left ventricular (LV) lateral wall,
seen as a left bundle branch block pattern on the
electrocardiogram (ECG) due to propagation of the
electrical wave front away from the sternum. This
results in LV dyssynchrony and mismatched timing
between chamber walls, with deleterious effects on
LV function and adverse clinical outcomes, including
FIGURE 5 Dual-Chamber Pacemaker Timing Cycles
TARP
AV
AV AV AV
ID
VA VA
LRLR
AP VP AP VS AS VP
PVARP
TARP
AV PVARP
TARP
AV PVARP
The timing cycle in DDD consists of a lower rate (LR) limit, an atrioventricular (AV) interval, a ventricular refractory period, a post-ventricular
atrial refractory period (PVARP), and an upper rate limit. If intrinsic atrial and ventricular activity occur before the LR limit times out, both
channels are inhibited and no pacing occurs. In the absence of intrinsic atrial and ventricular activity, AV sequential pacing occurs (first cycle).
If no atrial activity is sensed before the ventriculoatrial (VA) interval is completed, an atrial pacing artifact is delivered, which initiates the AV
interval. If intrinsic ventricular activity occurs before the termination of the AV interval, the ventricular output from the pacemaker is
inhibited (i.e., atrial pacing with intrinsic conduction [second cycle]). If a P-wave is sensed before the VA interval is completed, output from
the atrial channel is inhibited. The AV interval is initiated, and if no ventricular activity is sensed before the AV interval terminates, a ventricular
pacing artifact is delivered (i.e., P-synchronous ventricular pacing [third cycle]). Reproduced with permission from Hayes et al. (40).
AP ¼ atrial pace; AS ¼ atrial-sensed event; ID ¼ intrinsic deflection; TARP ¼ total atrial refractory period (which includes the AV interval and
PVARP); VP ¼ ventricular pace; VS ¼ ventricular-sensed event.
TABLE 1 Recommendations for Permanent Pacing in Sinus Node Dysfunction
Class I: Permanent Pacemaker Implantation Is Indicated for
1. Documented symptomatic bradycardia, including frequent sinus pauses that produce
symptoms.
2. Symptomatic chronotropic incompetence.
3. Symptomatic sinus bradycardia that results from required drug therapy for medical
conditions.
Class IIa: Permanent Pacemaker Implantation Is Reasonable for
1. Sinus node dysfunction with heart rates <40 beats/min when a clear association
between significant symptoms consistent with bradycardia and the actual presence of
bradycardia has not been documented.
2. Syncope of unexplained origin when clinically significant abnormalities of sinus node
function are discovered or provoked in electrophysiological studies.
Class IIb: Permanent Pacemaker Implantation May Be Considered in
1. Minimally symptomatic patients with chronic heart rate <40 beats/min while awake.
Class III: Permanent Pacemaker Implantation Is Not Indicated in
1. Asymptomatic patients.
2. Patients for whom the symptoms suggestive of bradycardia have been clearly
documented to occur in the absence of bradycardia.
3. Patients with symptomatic bradycardia due to nonessential drug therapy.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
195
heart failure and mortality (8). Several studies have
reported RV pacing-induced cardiomyopathy rates of
up to 20% with frequent RV pacing among patients
with preserved ejection fraction. Male sex, wide
native QRS complexes, and frequent RV pacing
(>20%) are reported predictors of RV pacing-
associated cardiomyopathy (9). For this reason, al-
gorithms to avoid or minimize RV pacing have been
developed for dual-chamber pacemakers. The
fundamental concept is adjustment of timing in-
tervals to deliver atrial pacing whenever required, but
to extend the AV interval as long as possible to permit
intrinsic conduction and thus eliminate the need for
ventricular pacing. Implementations include use of
an atrial pacing mode (AAI) with an automatic switch
to ventricular pacing (DDD) if AV conduction fails,
and AV interval prolongation to supraphysiological
values with shortening if intrinsic conduction does
not manifest. These algorithms effectively decrease
the percentage of ventricular pacing in patients
without permanent complete AV block (10,11). The
benefit of RV pacing avoidance algorithms on devel-
opment of atrial fibrillation, heart failure, and mor-
tality is less clear from clinical trials. Moreover, very
long AV delays (>400 ms) result in atrial contraction
occurring during early diastole, resulting in cannon A
waves and adverse hemodynamics, referred to as
“pseudopacemaker syndrome.” RV pacing avoidance
FIGURE 6 Far-Field R-Wave Oversensing, a Cause of Inappropriate Atrial Fibrillation
Detection by a Biventricular Pacemaker
(Top) The pacing lead is commonly positioned in the right atrial appendage (RAA), which
hangs over the right ventricle. The spacing between the distal helix and ring electrode
determines the size of the antenna of the pacemaker lead. The large signal generated by
the right ventricle may be oversensed by the atrial lead (V deflection, inset), leading to
overcounting of atrial events and inappropriate detection. (Bottom) An example of a
device tracing in which the far-field ventricular signals were sensed on the atrial channel
(*), and an inappropriate mode switch (MS) (þ) occurs. Also present are paced atrial im-
pulses (AP), atrial events sensed during the blanking period (Ab), and atrial events sensed
in the PVARP period (AR). Reproduced with permission from Seet et al. (42). BV ¼
biventricular pacing; PVARP ¼ post-ventricular atrial refractory period; RA ¼ right atrium.
TABLE 2 Recommendations for Pacing in Acquired
Atrioventricular Conduction Abnormalities
Class I: Permanent Pacemaker Implantation Is Indicated for
1. Third- or advanced–second-degree AV block:
a) If associated with symptoms or ventricular arrhythmias.
b) In awake, asymptomatic patients:
i. In sinus rhythm, with documented asystole $3.0 s or any
escape rate <40 beats/min, or originating from below
the AV node.
ii. With AF and bradycardia with 1 or more pauses >5 s.
iii. With neuromuscular diseases such as myotonic muscular
dystrophy, Kearns-Sayre syndrome, Erb dystrophy (limb-
girdle muscular dystrophy), and peroneal muscular
atrophy, with or without symptoms.
iv. That is precipitated by exercise, and in the absence of
myocardial ischemia.
v. With average awake ventricular rates of 40 beats/min or
faster if cardiomegaly or LV dysfunction is present or if
the site of block is below the AV node.
2. Recurrent syncope, reproduced by CSM induction of ventricular
asystole >3 s.
Class IIa: Permanent Pacemaker Implantation Is Reasonable for
1. Persistent third-degree AV block with an escape rate >40
beats/min in asymptomatic adult patients without
cardiomegaly.
2. Asymptomatic second-degree AV block at intra- or infra-His
levels found at electrophysiological study (HV >100 ms).
3. First- or second-degree AV block with symptoms similar to
those of pacemaker syndrome or hemodynamic compromise.
4. Syncope not demonstrated to be due to AV block when other
likely causes have been excluded.
5. Symptomatic neurocardiogenic syncope associated with
bradycardia documented spontaneously or at the time of
tilt-table testing.
Class IIb: Permanent Pacemaker Implantation May Be Considered for
1. Neuromuscular diseases such as myotonic muscular dystrophy,
Erb dystrophy (limb-girdle muscular dystrophy), and peroneal
muscular atrophy with any degree of AV block (including first-
degree AV block), with or without symptoms, because of
unpredictable progression of AV conduction disease.
2. AV block in the setting of drug use and/or drug toxicity when
the block is expected to recur even after the drug is withdrawn.
Class III: Permanent Pacemaker Implantation Is Not Indicated in
1. Asymptomatic first-degree or fascicular block AV block; and
type I second-degree AV block at the supra-His (AV node) level.
2. AV block that is expected to resolve and is unlikely to recur
(e.g., drug toxicity, Lyme disease, or transient increases in
vagal tone or during hypoxia in sleep apnea syndrome in the
absence of symptoms).
3. Asymptomatic hypersensitive cardioinhibitory response to
carotid sinus stimulation.
4. Situational vasovagal syncope in which avoidance behavior is
effective and preferred.
AF ¼ atrial fibrillation; AV ¼ atrioventricular; CSM ¼ carotid sinus massage;
HV ¼ His-ventricle; LV ¼ left ventricular.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
196
algorithms can lead to pacing below the programmed
lower rate. Pause-dependent cases of ventricular
proarrhythmia were reported in patients with RV
pacing avoidance algorithms (12–14).
In contrast to standard dual-chamber pacemakers,
biventricular pacemakers aim to maximize ventricu-
lar pacing to deliver the highest possible dose of
cardiac resynchronization therapy (CRT, discussed in
part 2 [4]). Algorithms that shorten the AV interval
when intrinsic conduction or PVCs are sensed have
been developed for CRT systems.
RATE RESPONSE
Rate response (also called rate-adaptive pacing) refers
to an increase in the pacing rate in response to
physical, mental, or emotional exertion. Rate-
adaptive pacing improves exercise capacity in pa-
tients with chronotropic incompetence. The most
TABLE 3 Pacemaker-Related Complications
Complication Reported Frequency (%)
Pneumothorax 0.9–1.2 (22)
Cardiac perforation <1 (44)
Hemothorax <1
Significant pocket hematoma requiring 3.5 (19)
Surgery
Interruption of anticoagulation
Prolongation of hospital stay
Lead dislodgement
Right-sided leads 1.8 (22)
LV leads 5.7 (22)
Venous thrombosis and obstruction 1–3 (45)
CIED device infection 1.0–1.3 (46)
Mechanical lead complications <1
CIED ¼ cardiac implantable electronic device; LV ¼ left ventricular.
FIGURE 7 Immediate Complications Associated With Pacemaker Implantation
(A) Chest x-ray after device placement showing pneumothorax. The arrow highlights the border of the collapsed lung. (B) Hematoma
associated with blood drainage from the pocket. Chest x-ray (C) and computed tomography (CT) image (D) of arterial placement of a pacing
lead. Note that the lead is pointing posteriorly on a lateral chest x-ray while traversing through the ascending aorta on the CT scan. (E) Acute
pericardial effusion (arrow) with hemodynamic collapse in a patient after right ventricular lead implantation. Also see Online Video 1.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
197
common sensor used to drive rate response is the
accelerometer, which rapidly detects physical mo-
tion. Limitations include a need for upper extremity
motion to drive the heart rate (so that walking with
limited arm motion on a treadmill leads to a poor rate
response) and abrupt deceleration after a period of
moderate-intensity exercise when the metabolic de-
mands are still high, although programmable func-
tionality can mitigate these limitations. Physiological
sensors have used minute ventilation, cardiac
contractility, blood temperature, or volume changes
to drive rate response. Although more specific, they
introduce greater latency and are often used with
accelerometers to provide a blended response.
Minute ventilation sensors transmit a low-energy
current from the lead tip to the pulse generator to
measure the variations in pulmonary impedance that
occur with respiration. They may not be suitable for
patients with tachypnea related to pulmonary
disease, and may inappropriately increase the heart
rate when patients are connected to ECG monitors
that inject small currents into the body, which results
in sensor error.
INDICATIONS FOR PACING
Diseases of the sinus node, AV node, or His-Purkinje
system due to aging, fibrosis, inflammation, infarc-
tion, or other conditions disrupt cardiac electrical
signaling. In general, when symptomatic bradycardias
ensue, pacing is indicated. An important consider-
ation is whether the bradyarrhythmia is reversible, in
which case a temporary pacemaker is preferred. Ex-
amples include Lyme disease or inferior myocardial
ischemia, which can present with alarming brady-
cardia, but often with spontaneous recovery within
1 week. Treatable causes of bradycardia should be
considered, and include medications (beta-blockers,
calcium-channel blockers, most antiarrhythmic drugs,
ivabradine, and others), obstructive sleep apnea
(especially during apnea), infections (Lyme disease,
Chagas disease, Legionnaires’ disease, psittacosis, Q
fever, typhoid fever, typhus, among others), and
metabolic conditions (hypothyroidism, anorexia
nervosa, hypothermia, and hypoxia). In younger pa-
tients, hypervagotonia in association with athleticism
or vasovagal spells may lead to slow heart rates. When
pacing is warranted, the type of pacemaker (atrial,
ventricular, dual-chamber, or biventricular) is deter-
mined by the nature of the conduction system defect
(sinus node, AV node, or intraventricular conduction
delay, such as a left bundle branch block).
SINUS NODE DYSFUNCTION. Class I indications for
permanent pacemaker implantation are present when
it is clear that sinus bradycardia is responsible for
symptoms. The bradycardia may manifest as pauses
or chronotropic incompetence. The latter is defined
as a failure to achieve 70% of the age-predicted
maximum heart rate or 100 beats/min during
maximal exertion. Class II indications are present
when a sinus node abnormality is the likely cause of
the symptoms, but correlation is difficult. Sinus
bradycardia <40 beats/min in a patient with symp-
toms suggestive of bradycardia constitutes a class II
indication for pacing when an association between
bradycardia and symptoms cannot be definitively
demonstrated. Similarly, unexplained syncope in a
patient with evidence of sinus node dysfunction is a
class II indication. In many patients, medications
cause sinus bradycardia; if the medications (such as
calcium-channel blockers or beta-blockers) are
necessary, then permanent pacemaker implantation
is reasonable (Table 1).
FIGURE 8 Implant Predictors of Lead Stability
V. EGM (unfiltered) Peak to Peak: 13.0 mV
+20 mV
+10 mV
-10 mV
-20 mV
A
B
(A) Typical electrogram (EGM) changes at 100 mm/s sweep speed. The EGM widening
and ST-segment elevation seen here are good long-term predictors of lead fixation. (B)
EGMs in a patient who developed cardiac perforation. Negative current of injury
(ST-segment) changes to a positive pattern as the pacemaker lead is pulled back into the
right ventricle. Reprinted with permission from van Gelder et al. (21). VS ¼ ventricular-
sensed event.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
198
ACQUIRED ATRIOVENTRICULAR CONDUCTION DISEASE.
Class I indications are present in patients with severe
AV conduction disease who are at risk for catastrophic
events, such as syncope, falls, or sudden death. With
these conditions, conduction disease is commonly
infranodal, and is often characterized by a wide QRS
bradycardia. High-risk conditions include symptom-
atic Mobitz type I (Wenckebach) or type II second-
degree AV block, advanced AV block (block of 2 or
more consecutive P waves), or complete (third-degree)
AV block. Patients with asymptomatic complete heart
block or advanced AV block also warrant pacing,
despite being asymptomatic (Table 2).
NEUROCARDIOGENIC SYNCOPE. Neurally-mediated
syncope is a difficult syndrome for the clinician to
diagnose and treat. Patients with confirmed carotid
sinus hypersensitivity (demonstrated by a pause of
3 s or more, with carotid sinus pressure reproducing
symptoms) or other neurally-mediated car-
dioinhibitory pauses may benefit from permanent
pacemaker implant. Although pacing may be
FIGURE 9 X-Ray of a Patient With Twiddler Syndrome Who Developed Loss of Capture
and High Impedance on the Ventricular Lead
Note that several loops on the ventricular lead can put strain on the lead, leading to
fracture and high impedance.
FIGURE 10 Intermediate-Term Complications
(A) Hypertrophic scar after pacemaker implantation. (B) Keloid formation at the site of device surgery. (C and D) Echocardiographic images of
the right ventricular lead through the tricuspid valve (arrow) associated with severe tricuspid regurgitation.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
199
effective in patients with an isolated car-
dioinhibitory response, often a vasodepressor
response coexists, limiting the utility of pacing. Up
to 20% of patients with carotid hypersensitivity
continue to experience syncopal spells during a
5-year follow-up after pacemaker implantation, and
the syncope may related to the vasodepressor
component of the syndrome (15). In patients with
situational syncope (cough, micturition, and so on),
trigger avoidance is emphasized.
NEUROMUSCULAR DISEASES. Many progressive
neuromuscular diseases, such as the muscular dys-
trophies, are of special concern, given the unpre-
dictable disease course and their predilection for
cardiac muscle and fibrosis in and around the
FIGURE 11 Electrical Diagnosis of Lead Failure
3,000
2,500
2,000
1,600
1,200
800
400
Jul 4, 2013
V Unipolar
tip
Leadless
ECG
V Bipolar
Oct 4, 2013
Configurations
Unipolar
Bipolar
Episode: Noise Reversion (Continued)
First Measurement
360 Ω
840 Ω
Episode 1 of
Page 2 of
Ventricular Lead Impedance
Ventricular Lead Monitoring: 1-year trend 630 Ω (Unipolar)
Last 7 DaysAuto Polarity SwitchA
Lifetime Range
330-650 Ω
740->3,000 Ω
Jan 3, 2014 Apr 4, 2014 Jul 4, 2014 Jun 16, 2014
BiBi
Ω
Uni
0
A
B
(A) Abrupt increase in ventricular lead impedance, consistent with conductor fracture. (B) Electrograms obtained from the patient’s
pacemaker. Intermittent, short-duration, high-frequency signals with nonphysiological intervals (red arrow) in a patient with an abrupt
change in lead impedance universally points to lead fracture. The red star shows obligatory under sensing during DOO mode and functional
noncapture (blue arrow). After reversion to DDDR mode, appropriate sensing (blue triangle) is noted. Modern pacemakers can detect noise
and switch to a synchronous mode. Most often the fractures are not radiologically detectable because they are result of very small
conductor filar disruptions. ECG ¼ electrogram; SIR ¼ sensor indicated rate; other abbreviations as in Figure 5.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
200
His-Purkinje system. A class I indication for pacing is
present when second- or third-degree AV block is
seen, irrespective of symptoms.
Congestive heart failure. Patients with depressed
ventricular function, a wide QRS interval, and
symptomatic heart failure benefit from cardiac
resynchronization pacing, with reductions in heart
failure and mortality, discussed in detail in part 2 (4).
IMPLANT-RELATED COMPLICATIONS
Complication rates range from <1% to 6% with cur-
rent implant tools and techniques. These are
broadly divided into immediate/procedure-related,
intermediate-term, and long-term or late complica-
tions (Table 3). Their prompt recognition permits
timely management.
IMMEDIATE PROCEDURE-RELATED COMPLICATIONS.
Transvenous lead placement requires venous
puncture in the pre-pectoral region. Due to the
proximity of the apex of the lung to vascular targets,
pneumothorax (Figure 7A) and hemothorax occur in
up to 1% of cases. During implantation, the risk is
lowered when vascular access using anatomic land-
marks is replaced by vein visualization with cephalic
vein cut down (16), contrast venography (17), or ul-
trasound guidance (18). Arterial puncture and inad-
vertent placement of pacing leads in the arterial
system (Figures 7C and 7D) are avoided by advancing
the guidewire into the inferior vena cava before
advancing sheaths to deliver the leads. Inadvertent
placement of the RV lead in the LV through a patent
foramen ovale or the middle cardiac vein is avoided
during implant by advancing the pacing lead across
the pulmonary valve, and then withdrawing it to
allow it to drop into the RV cavity. Post-operatively, if
lead position is uncertain, the lateral chest x-ray,
oblique imaging (right anterior oblique, left anterior
FIGURE 12 Long-Term Complications Associated With Pacemakers
(A) Lead fracture on a chest x-ray. Lead fracture is associated with high impedance due to structural discontinuity (arrow) of the lead. (B) Lead
insulation break, which is typically associated with low impedance. Reprinted with permission from Kutarski et al. (43). (C) Deposition of
crystals in the lead, which may be associated with high thresholds and impedances (functional in nature). Reprinted with permission from
Marshall et al. (30). (D) Development of fibrosis at the lead myocardial interface, resulting in high thresholds, and impaired sensing, resulting
in exit block.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
201
oblique), echocardiography, and computed tomogra-
phy scan may be helpful. Right bundle branch
morphology during pacing suggests a left-sided lead.
Intracavitary LV leads may result in thromboembo-
lism, and are generally repositioned, or if discovered
late after implant, anticoagulation is instituted.
Pocket hematoma requiring intervention is infre-
quent (3.5%), and occurs more commonly in patients
who are taking combinations of anticoagulant and
antiplatelet drugs (Figure 7B). Uninterrupted anti-
coagulation with warfarin lowers the rate of hema-
toma formation compared with perioperative heparin
bridging (19). The optimal use of periprocedural novel
oral anticoagulants remains unresolved, and when
possible, they are discontinued 2 to 3 days before
pacemaker placement (20). Pericarditis and cardiac
tamponade are mechanical complications associated
with cardiac perforation (Figure 7E, Online Video 1).
Indications for repositioning a lead when micro-
perforation is suspected include refractory peri-
carditic pain, persistent effusion, or unacceptable
pacing or sensing function. Passive fixation leads
have a lower risk of perforation, but are used less
frequently than active fixation leads due to the
greater ease of site selection and ease of extraction
with the latter. Use of soft stylets and recognition of
negative current of injury (21) on the electrogram
recording enables implanters to minimize the risk of
cardiac perforation (Figure 8). Micro- and macro-
dislodgements are uncommon after device implant.
The risk of dislodgement is higher for coronary sinus
pacing leads (22) and passive fixation leads.
INTERMEDIATE-TERM COMPLICATIONS. Acute he-
matoma increases the risk of pocket and systemic
infection. Device movement and subsequent lead
dislodgements are infrequently seen in current prac-
tice. Manipulation of the pacemaker pocket can put
undue strain on the pacemaker lead, resulting in lead
malfunction (Figure 9) (23).
Excess scar at the incision site is associated with
unfavorable cosmetic result, pain, and discomfort.
Hypertrophic scar (Figure 10A) and keloid formation
(Figure 10B) are due to excess interstitial tissue
FIGURE 13 Signs of Pocket Infection
(A) Redness and skin changes in a patient 2 weeks after device surgery. (B) Purulence within the pocket. (C) Erosion in a patient who
subsequently developed systemic signs of infection after pacemaker surgery.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
202
formation during the healing process. The latter en-
tity has a raised surface and is frequently associated
with itching and photosensitivity. The risk of keloid
and hypertrophic scar formation is reduced by use of
monofilament suture, good surgical technique, and
avoidance of excess tension on the suture line. Ke-
loids near the suture line are treated with intrale-
sional steroid injection, silicone sheeting, and laser
phototherapy.
Pacemaker lead placement through the tricuspid
valve (TV) is infrequently associated with leaflet
perforation and impingement of leaflet motion,
resulting in valve dysfunction (Figures 10C and 10D)
(24). When this leads to chronic fibrotic changes in
the TV, tethering of the leaflet often ensues. Com-
plications associated with TV dysfunction are avoided
by using soft stylets and by crossing the valve at a
lower plane, thereby avoiding chordae on the septal
aspect of the tricuspid valve. Pacemaker leads often
accumulate adherent fibrous strands composed of
fibrotic material or thrombi. Recurrent emboli
of mobile echo-dense structures increase the risk of
pulmonary hypertension and, in patients with a pat-
ent foramen ovale, possibly stroke (25).
Other complications may include pocket pain or
arm swelling. Pocket pain is infrequently reported
after device implant. Pocket pain in the absence of
device infection occurs when the device is placed in
the subcuticular plane, leading to the stimulation of
pain corpuscles (26). Occasionally ventricular pacing
is associated with uncomfortable sensation or frank
pain (27,28). In the absence of clinically significant
coronary artery disease, treatment is often conser-
vative, using ventricular pacing avoidance algorithms
and occasionally lead repositioning to an area where
pacing-related discomfort is minimal. The presence of
multiple leads in the venous system or endothelial
injury during implantation can predispose patients to
development of venous thrombosis and stenosis,
which can manifest as unilateral edema or superior
vena cava syndrome.
LATE COMPLICATIONS. Because it is subject to re-
petitive mechanical stress with each cardiac cycle and
with shoulder girdle movement in the hostile envi-
ronment of the human body, the lead is the most
common pacemaker system component to fail.
Conductor fracture typically results in non-
physiological noise caused by the lead itself (high-
frequency, saturated electrograms generated by
intermittent contact between disrupted conductor
elements, called filars) and can be associated with
high lead impedance (Figures 11 and 12A). An insu-
lation break results in low impedance and
FIGURE 14 Summary of CIED Device Infection Management
CIED Device
Infections
TEE
Pocket examination
Blood Cultures
Pocket infection
(neg. blood cultures)
Lead erosion
(neg. blood cultures)
AHA Guidelines for
Endocarditis
4-6 weeks of
antibiotics
Non- S. Aureus -
2 weeks of antibiotics
S. Aureus
2-4 weeks of
antibiotics
10-14 days of
antibiotics
7-10 days of
antibiotics
Valve endocarditis Lead endocarditis
Positive blood
cultures with
negative TEE
The duration of antibiotic therapy depends on imaging (TEE), cultures, microbiology and clinical presentation. AHA ¼ American Heart Association;
CIED ¼ cardiac implantable electronic device; neg. ¼ negative; TEE ¼ transesophageal echocardiography.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
203
oversensing of signals generated by surrounding
structures (e.g., muscles) as conductors are exposed
(Figure 12B) (29). Acute venous entry angle, medial
venous access near the costoclavicular ligament,
sharp turns in the pocket, young age, subpectoral
placement of device, tight sutures, and silicone
insulation are risk factors associated with lead frac-
ture and insulation break. Occasionally, pacing
thresholds and impedance rise, typically gradually
over months, without any detectable fracture. This
results from the development of scar tissue at the
electrode myocardial interface (exit block) or the
deposition of calcium hydroxyapatite crystals (30) at
the lead-tissue interface, which can grow into the
lead (Figures 12C and 12D) (30). Steroid elution in
current-generation pacing leads has virtually elimi-
nated the risk of exit block. Gradual impedance may
be misinterpreted as a lead failure, but it is an
important distinction; gradual impedance rise re-
quires no action in an otherwise functional lead,
whereas lead failure may require surgical
intervention.
Pacemaker infections can involve the pocket,
associated endovascular leads, and the valves. The
risk is higher for subsequent generator changes than
for the initial implant procedure. Diabetes, heart
failure, renal failure, corticosteroid use, post-
operative hematoma, lack of antibiotic prophylaxis,
oral anticoagulation, previous cardiac implantable
electronic device (CIED) infection, generator change,
FIGURE 15 Pacemaker-Mediated Tachycardia and Upper Rate Behavior
VA VA
AV PVARP
PVARP
PVARP
PVC, retrograde P
PVARP
40 60 80 100
Upper tracking rate: 130 bpm
PVARP: 250ms AV delay:150ms
VRate
120 140 160
160
140
120
100
80
60
40
20
0
***
A
B
C
D
E
Atrial Rate
Continued in the next column
FIGURE 15 Continued
(A) Pacemaker-mediated tachycardia. The first beat illustrates
AV sequential pacing, followed by a PVC beat. The PVC starts a
new VA interval. Any AV decoupling event (most frequently
PVCs) can retrogradely conduct to the atrium. An atrial event
is sensed if it falls outside of the PVARP. The VA interval
expires and a new AV interval is triggered. Ventricular pacing
occurs at the end of the AV interval setting up a circuitous series
of events termed pacemaker-mediated tachycardia. (B to E)
Upper-rate behavior in a patient with a dual-chamber pace-
maker. The patient was programmed to an upper tracking rate
of 130 beats/min. 1:1 AV conduction occurs at baseline and up to
130 beats/min (B). With an increase in sinus rate above 130
beats/min, some of the P waves fall in the PVARP period
(arrows) and are not tracked (C). Progressive PR prolongation
is seen before blocked beats (pacemaker Wenckebach). When
the sinus rate reaches the TARP (AV interval þ PVARP), every
other P-wave falls in the PVARP and is not tracked, resulting
in an effective ventricular rate of 75 beats/min (D and E), and
an abrupt drop in heart rate (2:1 block rate in this patient,
around 150 beats/min). AV ¼ atrioventricular interval;
PVC ¼ premature ventricular beat; other abbreviations as in
Figure 5.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
204
and use of a temporary pacemaker are known risk
factors for CIED infection (31). Timing of CIED infec-
tion depends on the virulence of the organism. It
most commonly occurs within a few weeks, but may
present up to a year after device surgery. Early rein-
tervention is the strongest risk factor for CIED infec-
tion. Pocket re-entry should be avoided with needle
aspiration or surgery, unless the swelling is very
tense and painful. Redness, purulence, erosion,
discharge of gelatinous material, chronic pocket pain,
and nonhealing incision site with or without thinning
are signs of pocket infection (Figure 13). Systemic
blood stream infections and endocarditis of the lead
or the valves are often associated with fever, chills,
positive blood cultures, and the presence of intra-
cardiac vegetations. Appropriate referral to a
specialized center managing device infections, com-
plete device removal, and targeted antibiotic therapy
are associated with better outcomes (32). The dura-
tion of antibiotic therapy is summarized in Figure 14.
BASIC PACEMAKER TROUBLESHOOTING
BATTERY. Malfunction may occur with battery
depletion. Elective replacement indicator denotes that
90 days of reliable function remain, whereas end of
life indicates a battery depleted to the point of
unpredictable function. Recalls are uncommon in
pacemakers, in contrast to implantable cardioverter-
defibrillators, and most often involve software
updates to remedy the unintended consequences of
the problem.
LEAD DIAGNOSTICS. Pacemakers can be programmed
to pace and to sense in a bipolar mode (lead tip to lead
ring) or a unipolar mode (lead tip to pulse generator).
Bipolar sensing is preferred due to its smaller sensing
“antenna,” which minimizes oversensing of extrac-
ardiac signals. Bipolar pacing is favored to minimize
pectoral muscle stimulation. However, when bipolar
thresholds are elevated or sensing is impaired (e.g.,
due to a damaged conductor to the ring electrode),
unipolar pacing or sensing may be attractive. The
programmed sensitivity is the smallest signal that will
be detected as a cardiac event; thus, a setting of
0.5 mV is twice as sensitive as 1.0 mV. In general,
a sensing safety margin of 2 is preferred (sensitivity
set to 0.5 mV if atrial electrogram is 1.0 mV). Sensi-
tivity is adjusted to improve oversensing or under-
sensing. Some devices can automatically adjust
sensitivity on the basis of the measured amplitude of
sensed events.
Two commonly encountered device-related ar-
rhythmias are pacemaker-mediated tachycardia and
FIGURE 16 Role of Current Pathway and Pacemaker Interference
(A) High risk of interference: pre-operative reprogramming or magnet use required.
(B) Low risk of electromagnetic interference.
FIGURE 17 Algorithm for Pre-Operative Pacemaker Reprograming
Pacemaker
Is surgical site above the hip?
yes
PM dependent?
No further device
programing needed
Reprogram to
asynchronous
pacing mode
Or
Place a magnet
no
no
Devices are programmed to asynchronous mode when surgical procedures involving
electrocautery are performed above the hip in pacemaker-dependent patients.
PM ¼ pacemaker.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
205
upper rate behavior resulting in high or low ventric-
ular rates.
1. Pacemaker-mediated tachycardia (PMT). Ventricu-
lar events (premature ectopic beats or paced beats),
when conducted retrograde to the atria, set up a
circuitous cycle of ventricular pacing at the upper
tracking rate (Figure 15A). The retrograde wave front
(ventriculoatrial conduction) is sensed on the atrial
channel, which then triggers ventricular pacing at
the end of the programmed AV interval. Each
ventricular-paced beat conducts retrogradely,
setting up a circuitous pacemaker-mediated tachy-
cardia. Atrioventricular decoupling due to retro-
grade ventriculoatrial conduction results in an
unfavorable hemodynamic profile, and is associated
TABLE 4 Potential Adverse Effects of CMR in Patients With Pacemaker
Adverse Effect on
Exposure to External
Electromagnetic Fields Mechanism Incidence and Risk Factors Prevention
Mechanical movement of
device or dislocation
of lead
Effect of static magnetic field on ferromagnetic
components of device and lead.
Risk is proportional to: 1) strength of magnetic
field (1.5-T vs. 3-T); 2) amount of
ferromagnetic material in lead and device; 3)
distance of CMR bore from device; and 4)
stability of device and lead
(time from implant).
 Movement of device or lead is very rare.
 Current leads have almost no
ferromagnetic material.
 Torsional force from 1.5-T scanner
typically not significant (47).
 Delay CMR in fresh implants to allow
lead stabilization.
 CMR-conditional devices and
leads minimize/eliminate
ferromagnetic material.
 Avoid 3-T CMR due to lack of
clinical experience.
Current induction and
heating of tissue at
lead interface
Radiofrequency and pulsed gradient magnetic
fields can induce current in the lead, and the
body can complete the circuit.
Radiofrequency pulses can heat tissue. Device
and leads can amplify this (the “antenna”
effect) (48).
Tissue edema and necrosis at the lead tip
can occur.
Lead parameters may change including
impedance change, reduced sensing, and
elevated threshold.
 The SAR describes the CMR
radiofrequency energy deposition
in tissues.
 Risk of tissue heating depends on lead
length and lead configuration, such as
loops (48).
 Epicardial, fractured, and abandoned
leads are more likely to cause local
heating (49).
 Risk is higher with thoracic CMR.
 Changes in lead impedance, sensing,
and threshold have been reported
immediately after CMR and in the long
term. Changes that warrant device
reprogramming or revision are rare
(38,50–52). Others have reported no
changes to lead parameters (53,54).
 Temporary changes in battery voltage
can also be seen and often recover
over time (50,54).
 Elevation in cardiac enzymes, typically
of small magnitude, has been
reported (39).
 Minimize SAR while still ensuring
image quality; input from
magnetic resonance physicist
should be considered.
 Avoid CMR in patients with
epicardial, fractured, and aban-
doned leads.
 Appropriate selection of CMR
landmark.
 Assessment of lead immediately
after CMR to detect changes that
may need intervention.
Electromagnetic effects
EMI leading to inhibition
of pacing or tacking
at a faster rate
RF pulses in magnetic fields are sensed as true
cardiac signals.
 Magnet mode and pauses in
dependent patients are rarely
reported (53,55).
 Newer devices are better
shielded from EMI.
 Program asynchronous pacing in
pacemaker-dependent patients.
 Program demand pacing in
nondependent patients.
 Continuous monitoring during
scan.
Induction of arrhythmias A strong pulsed gradient field results in
myocardial electrical stimulation causing
arrhythmias.
 Demonstrated mostly in
animal experiments.
 Current generated under clinical
circumstances is too low to capture
myocardium (56).
 Continuous monitoring of rhythm
by personnel trained in cardiac
resuscitation.
 Limit lead length and looped
leads (56).
Change in Reed
switch behavior
Static magnetic field can activate the
Reed switch.
Consequence: asynchronous pacing
detection/therapy.
 Reported incidence variable
from 0%–38% (38,54,57).
 Determined by distance from bore
of the magnet.
 Some CMR-conditional devices
use Hall sensor, which has more
predictable behavior.
Power on reset causes the
device to revert to
default factory settings;
in some devices, this
can be VVI mode
Combination of static and dynamic RF pulses
that induce currents in various pacemaker
components in an electromagnetic field.
 Incidence 0.7%–3.5% (38,39).
 More likely in devices released
before 2002 (39).
 Discontinue scan if noted during
monitoring.
 Avoid CMR in dependent patients
with devices from before 2002.
CMR ¼ cardiac magnetic resonance; EMI ¼ electromagnetic interference; RF ¼ radiofrequency; SAR ¼ specific absorption rate.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
206
with a sensation of palpitations and fullness in the
neck due to canon A waves. PMT is terminated by
magnet application (which results in asynchronous
pacing and prevents tracking of the retrograde
P-wave, thereby breaking the cycle) or by use of PMT
algorithms. Algorithms recognize ventricular pac-
ing at the upper tracking rate, and they either
extend the post-ventricular atrial refractory period
(PVARP) or pace the atrium simultaneously with
ventricular pacing for 1 cycle. PVARP extension re-
sults in functional atrial undersensing, whereas
atrial pacing makes the atria refractory to the
retrograde impulse.
2. Upper rate behavior. In dual-chamber pacing mode,
the upper rate is determined by the total atrial re-
fractory period (TARP). The TARP is a combination
of AV delay and the PVARP. If properly pro-
grammed, the device tracks in a 1:1 fashion until
reaching the programmed upper rate. As the atrial
rate increases, ventricular pacing cannot violate the
upper rate limit, resulting in progressively longer
AV intervals. This is referred to as pacemaker
Wenckebach. However, if the TARP is excessively
long, abrupt 2:1 block may develop with a sudden
slowing of the ventricular rate, which may cause
symptoms. This occurs because when the atrial rate
hits the TARP, every other event falls in the PVARP
and is not tracked. Effectively, the ventricular
pacing rate is one-half of the maximum tracking
rate. Undesirable upper rate behavior is avoided by
meticulous attention to intervals, by programming
to allow a Wenckebach interval between the
maximum tracking rate and the 2:1 block rate, and
by allowing rate-adaptive shortening of the AV and
PVARP intervals (Figures 15B to 15E).
3. Inappropriate mode switches. Occasionally, far field
events or frequent ectopic beats result in high
atrial counters and inappropriate detection and
classification of events as atrial fibrillation. It is
imperative to evaluate electrograms before thera-
peutic interventions for management of atrial
arrhythmias.
PERIOPERATIVE MANAGEMENT
OF PACEMAKERS
Electrosurgery is the application of a 100-kHz to
4-MHz electric current to cut or coagulate biological
tissue. With bipolar electrosurgery, both the active
and return electrodes are affixed to the cautery pen
placed at the surgical site, maintaining a local current
pathway; the more commonly used monopolar elec-
trosurgery delivers current from an active electrode
in the surgical field to a larger dispersive electrode
usually placed on the patient’s thigh or back. If elec-
trosurgical current enters a pacemaker lead electrode,
it may result in EMI. EMI sensed on the ventricular
channel is misinterpreted by the pacemaker as
intrinsic cardiac events that inhibit pacing, poten-
tially resulting in bradycardia or asystole. When
sensed on the atrial lead, EMI may result in tracking
with rapid ventricular pacing up to the programmed
upper rate limit, or inappropriate mode switch.
Electrosurgery-related EMI may also cause a power-
on reset (abrupt reversion to nominal mode and
pacing parameters), pulse generator damage, atrial
and ventricular arrhythmia, or tissue injury at the
lead-tissue interface, although these are uncommon
(33,34). The risk of electrosurgical EMI depends on
the surgical site and dispersive pad location, with the
highest risk for surgery of the heart and chest, fol-
lowed by the head and neck, shoulder/upper ex-
tremity, and abdomen-pelvis. In our experience,
pacemaker EMI is nonexistent with hip and lower
extremity surgery when the dispersive pad is applied
to the lower extremities (Figures 16A and 16B).
FIGURE 18 Algorithm for Cardiac Magnetic Resonance in Patients With Legacy,
Nonconditional Pacemakers
Other imaging
modality option
(radiology review)
MRI Scanning in CIED Patients
Yes
Don’t scan
Consult
Imaging may
be reasonable
Will depend
on consult
MRI
requested
FDA approved
MRI conditional
Pacemaker
dependent
Temporary PM
SICD; recall/advisory
CIED at ERI
• HRS RN eval pre/post
• Radiology/physicist
eval
• Consent by radiology
• Troponin if
abandoned lead
Scan: Mayo
MRI/CIED Protocol
Yes
No
No
No
No
Yes
Yes
Collaborative working relationships involving heart rhythm professions, radiology service
providers, and physicists are required for an efficient and safe imaging program in
patients with pacemakers. CIED ¼ cardiac implantable electronic device; ERI ¼ elective
replacement indicator; eval ¼ evaluation; FDA ¼ Food and Drug Administration;
HRS ¼ heart rhythm services; MRI ¼ magnetic resonance imaging; PM ¼ pacemaker;
RN ¼ registered nurse; SICD ¼ subcutaneous implantable cardioverter defibrillator.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
207
Professional society guidelines recommend the
following steps when surgery is needed in a patient
with a pacemaker: 1) use bipolar electrocautery when
possible; 2) use short-duration cautery bursts 5 s,
allowing for 5 s between bursts when using
monopolar electrocautery; and 3) apply pads to direct
the current pathway away from the pacemaker and
leads (35). When surgery is performed below the
umbilicus in pacemaker-dependent patients, or when
significant pacing is anticipated during the proced-
ure, the dispersive pad is placed on a lower extremity
and pacemaker reprogramming is unnecessary
(Figure 17). When the surgical site is above the um-
bilicus, pre-operative pacemaker interrogation is
performed, and the device is reprogrammed to an
asynchronous mode for the surgery (VOO, AOO, or
DOO); magnet application (which causes the pace-
maker to enter an asynchronous mode while the
magnet is applied) is an alternative. Pre-operative
device reprogramming to an asynchronous mode
(VOO, AOO, or DOO) is a must for pacemaker-
dependent patients (Figure 17) to prevent device in-
hibition. After surgery, the device is interrogated to
ensure appropriate function and reprogrammed to its
original settings.
CMR OF PATIENTS WITH PACEMAKERS
CMR has become ubiquitous, so that 75% of patients
with pacemakers will likely need a scan at some time
following implantation (36). In the 1980s, adverse
events, including death, occurred when patients with
pacemakers were (usually unknowingly) scanned, as
pacemaker leads could act as antennae in the CMR
environment, with induced currents during imaging
leading to risk of arrhythmia induction, capture
threshold changes, and device damage (Table 4). For
this reason, governmental organizations and profes-
sional society guidelines considered CMR scanning to
be contraindicated in patients with pacemakers.
Manufacturers subsequently developed CMR-
conditional pacemakers, which are specifically
designed to be safe in the CMR environment. How-
ever, the vast majority of implanted patients have
legacy, non–CMR-conditional pacemakers.
A growing body of evidence has indicated the
safety of CMR in patients with legacy pacemakers
implanted after the early 1990s, when imaging is
performed in a program established by integrated
teams that include radiologists, cardiologists,
specialized nurses, and physicists, with continuous
monitoring during the scan. Pacemakers made since
the 1990s include pass-through filters and other
technology to make them more resistant to EMI.
In several large series, including the Mayo Clinic se-
ries that now exceeds 1,000 scans, imaging of pa-
tients with legacy pacemakers has been safely
performed (Figure 18) (37,38). The greatest potential
risk is that of power-on reset in pacemaker-
dependent patients, which may change the pro-
grammed mode from asynchronous to synchronous,
permitting oversensing of CMR signals and inhibition
of pacing output. This was seen in a limited number
of older devices from a specific manufacturer (39).
Nearly all patients with legacy systems can be safely
imaged at centers with dedicated programs. Although
pacemaker pulse generator artifacts (particularly
when the can is low on the chest wall) may lead to
artifact obscuring the image, a large case series
observed that the clinical question was answered in
over 98% of nonthoracic scans (38), and newer CMR
processing algorithms facilitate thoracic imaging.
CMR-CONDITIONAL DEVICE AND LEADS. All major
device manufacturers have developed leads and
pacemakers, cardiac resynchronization devices, and
implantable cardioverter-defibrillators that are mag-
netic resonance–conditional. CMR-conditional refers
to devices that have no known hazards or risks un-
der specific magnetic resonance conditions. It is
notable that there are no “CMR-safe” devices, which
refers to devices that have no known hazards or
risks under any condition. CMR-conditional devices
have minimal ferromagnetic material, altered
filtering, and redesigned lead conductors to mini-
mize current induction and heating of tissue. There
are specific device and CMR conditions under which
such devices can be considered safe. Device re-
quirements include: CMR-conditional device paired
with CMR-conditional leads, and absence of aban-
doned, fractured, or epicardial leads. The device
must be implanted in the pectoral region. Magnetic
resonance scanning requirements are technical, and
are designed to limit energy deposition on CIEDs.
These include: static magnetic field of 1.5-T,
maximum specific absorption rate (SAR) value of
2 W/kg, maximum gradient slew rate of 200 T/m/s,
horizontal closed bore magnet, and supine or dorsal
positioning of patients (not on their sides). Although
several devices are approved for whole-body scan-
ning, some have more limited approval for scanning
parts other than the thorax. Continuous monitoring
during the scan (ECG, hemodynamics, and symp-
toms) by an advanced cardiac life support–trained
provider, and interrogation before and after scan-
ning are also required.
To summarize, CMR can be safely performed in pa-
tients with magnetic resonance–conditional devices
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
208
and at centers with dedicated programs, in nearly all
patients with legacy, non-CMR conditional devices.
CONCLUSIONS
Advances in pacemakers have led to their widespread
use to treat patients with bradycardias and congestive
heart failure. A basic understanding of their function,
troubleshooting, and management at the time of
surgery or imaging enhances patient care. In part 2 of
this review (4), we explore recent innovations and
future directions.
REPRINT REQUESTS AND CORRESPONDENCE: Dr.
Paul A. Friedman, Department of Cardiovascular
Medicine, Mayo Clinic, 200 First Street SW, Roches-
ter, Minnesota 55902. E-mail: pfriedman@mayo.edu.
R E F E R E N C E S
1. Nelson GD. A brief history of cardiac pacing.
Texas Heart Inst J 1993;20:12–8.
2. Aquilina O. A brief history of cardiac pacing.
Images Paediatr Cardiol 2006;8:17–81.
3. Altman LK, Arne HW. Larsson, 86; had first
internal pacemaker. New York Times January 18,
2002. Available at: http://www.nytimes.com/
2002/01/18/world/arne-h-w-larsson-86-had-first-
internal-pacemaker.html. Accessed November 3,
2016.
4. Madhavan M, Mulpuru SK, McLeod CJ, Cha Y-M,
Friedman PA. Advances and future directions in
cardiac pacemakers: part 2 of a 2-part series. J Am
Coll Cardiol 2017;69:211–35.
5. Lindemans FW, Denier Van der Gon JJ. Current
thresholds and liminal size in excitation of heart
muscle. Cardiovasc Res 1978;12:477–85.
6. Kleinert M, Elmqvist H, Strandberg H. Spectral
properties of atrial and ventricular endocardial
signals. Pacing Clin Electrophysiol 1979;2:11–9.
7. Parsonnet V, Myers GH, Kresh YM. Character-
istics of intracardiac electrograms II: atrial endo-
cardial electrograms. Pacing Clin Electrophysiol
1980;3:406–17.
8. DAVID Trial Investigators. Dual-chamber pacing
or ventricular backup pacing in patients with an
implantable defibrillator: the Dual Chamber and
VVI Implantable Defibrillator (DAVID) Trial. JAMA
2002;288:3115–23.
9. Khurshid S, Epstein AE, Verdino RJ, et al. Inci-
dence and predictors of right ventricular pacing-
induced cardiomyopathy. Heart Rhythm 2014;11:
1619–25.
10. Sweeney MO, Bank AJ, Nsah E, et al., for the
Search AV Extension and Managed Ventricular
Pacing for Promoting Atrioventricular Conduction
(SAVE PACe) Trial. Minimizing ventricular pacing
to reduce atrial fibrillation in sinus-node disease.
N Engl J Med 2007;357:1000–8.
11. Stockburger M, Boveda S, Moreno J, et al.
Long-term clinical effects of ventricular pacing
reduction with a changeover mode to minimize
ventricular pacing in a general pacemaker popu-
lation. Eur Heart J 2015;36:151–7.
12. Pascale P, Pruvot E, Graf D. Pacemaker syn-
drome during managed ventricular pacing mode:
what is the mechanism? J Cardiovasc Electro-
physiol 2009;20:574–6.
13. Cheng A, Calkins H, Berger RD. Managed ven-
tricular pacing below the lower rate limit: is it
simply caused by ventricular-based pacing? Heart
Rhythm 2009;6:1240–1.
14. Mansour F, Khairy P. Electrical storm due to
managed ventricular pacing. Heart Rhythm 2012;
9:842–3.
15. Brignole M, Menozzi C. The natural history of
carotid sinus syncope and the effect of cardiac
pacing. Europace 2011;13:462–4.
16. Parsonnet V, Roelke M. The cephalic vein
cutdown versus subclavian puncture for pace-
maker/ICD lead implantation. Pacing Clin Electro-
physiol 1999;22:695–7.
17. Burri H, Sunthorn H, Dorsaz PA, et al. Pro-
spective study of axillary vein puncture with or
without contrast venography for pacemaker and
defibrillator lead implantation. Pacing Clin Elec-
trophysiol 2005;28 Suppl 1:S280–3.
18. Seto AH, Jolly A, Salcedo J. Ultrasound-guided
venous access for pacemakers and defibrillators.
J Cardiovasc Electrophysiol 2013;24:370–4.
19. Birnie DH, Healey JS, Essebag V. Device sur-
gery without interruption of anticoagulation.
N Engl J Med 2013;369:1571–2.
20. Birnie DH, Healey JS, Essebag V. Management
of anticoagulation around pacemaker and defi-
brillator surgery. Circulation 2014;129:2062–5.
21. van Gelder BM, Bracke FA. Current of injury
(COI) pattern recorded from catheter delivered
active fixation pacing leads. Pacing Clin Electro-
physiol 2008;31:786–7, author reply 787.
22. van Rees JB, de Bie MK, Thijssen J, et al.
Implantation-related complications of implantable
cardioverter-defibrillators and cardiac resynchro-
nization therapy devices: a systematic review of
randomized clinical trials. J Am Coll Cardiol 2011;
58:995–1000.
23. Alvarez-Acosta L, Romero Garrido R, Farrais-
Villalba M, et al. Reel syndrome: a rare cause of
pacemaker malfunction. BMJ Case Rep 2014;2014.
24. Al-Bawardy R, Krishnaswamy A, Bhargava M,
et al. Tricuspid regurgitation in patients with
pacemakers and implantable cardiac defibrillators:
a comprehensive review. Clin Cardiol 2013;36:
249–54.
25. DeSimone CV, Friedman PA, Noheria A, et al.
Stroke or transient ischemic attack in patients with
transvenous pacemaker or defibrillator and echo-
cardiographically detected patent foramen ovale.
Circulation 2013;128:1433–41.
26. Furman S, Curtis AB, Conti JB. Recognition and
correction of subcuticular malposition of
pacemaker pulse generators. Pacing Clin Electro-
physiol 2001;24:1224–7.
27. Shvilkin A, Ellis ER, Gervino EV, et al. Painful
left bundle branch block syndrome: clinical and
electrocardiographic features and further direc-
tions for evaluation and treatment. Heart Rhythm
2016;13:226–32.
28. Squara F, Scarlatti D, Bres M, et al. Pacemaker-
induced painful left bundle-branch block. Acta
Cardiologica 2015;70:735.
29. Swerdlow CD, Ellenbogen KA. Implantable
cardioverter-defibrillator leads: design, diagno-
stics, and management. Circulation 2013;128:
2062–71, 1–9.
30. Marshall MVP, Baker P, Kiser A. Impedance
rise leading to unnecessary ICD lead extraction.
Paper presented at: The 5th Asia-Pacific Heart
Rhythm Society Scientific Session; October 3–6,
2012; Taipei, Taiwan.
31. Rohacek M, Baddour LM. Cardiovascular
implantable electronic device infections: associ-
ated risk factors and prevention. Swiss Med Wkly
2015;145:w14157.
32. Baddour LM, Epstein AE, Erickson CC, et al.,
for the Council on Cardiovascular Surgery and
Anesthesia, Council on Cardiovascular Nursing,
Council on Clinical Cardiology, Interdisciplinary
Council on Quality of Care and Outcomes
Research. Update on cardiovascular implantable
electronic device infections and their manage-
ment: a scientific statement from the American
Heart Association. Circulation 2010;121:458–77.
33. Pinski SL, Trohman RG. Interference in
implanted cardiac devices, part II. Pacing Clin
Electrophysiol 2002;25:1496–509.
34. Schoenfeld MH. Contemporary pacemaker and
defibrillator device therapy: challenges confront-
ing the general cardiologist. Circulation 2007;115:
638–53.
35. Crossley GH, Poole JE, Rozner MA, et al. The
Heart Rhythm Society (HRS)/American Society of
Anesthesiologists (ASA) expert consensus state-
ment on the perioperative management of
patients with implantable defibrillators, pace-
makers and arrhythmia monitors: facilities and
patient management; this document was devel-
oped as a joint project with the American Society
of Anesthesiologists (ASA), and in collaboration
with the American Heart Association (AHA), and
the Society of Thoracic Surgeons (STS). Heart
Rhythm 2011;8:1114–54.
J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al.
J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1
209
36. Kalin R, Stanton MS. Current clinical issues for
MRI scanning of pacemaker and defibrillator
patients. Pacing Clin Electrophysiol 2005;28:326–8.
37. Higgins JV, Watson RE Jr., Jaffe AS, et al.
Cardiac troponin T in patients with cardiac
implantable electronic devices undergoing mag-
netic resonance imaging. J Interv Card Electro-
physiol 2016;45:91–7.
38. Nazarian S, Hansford R, Roguin A, et al.
A prospective evaluation of a protocol for mag-
netic resonance imaging of patients with implan-
ted cardiac devices. Ann Intern Med 2011;155:
415–24.
39. Higgins JV, Sheldon SH, Watson RE Jr., et al.
“Power-on resets” in cardiac implantable elec-
tronic devices during magnetic resonance imaging.
Heart Rhythm 2015;12:540–4.
40. Hayes DL, Asirvatham SJ, Friedman PA.
Cardiac Pacing, Defibrillation and Resynchroniza-
tion: A Clinical Approach. Third Edition.
Chichester, UK: Wiley-Blackwell, 2013.
41. Stokes K, Bornzin G. The electrode–biointerface
stimulation. In: Barold SS, editor. Modern Cardiac
Pacing. Mount Kisco, NY: Futura Publishing Co,
1985:33–77.
42. Seet RC, FriedmanPA, Rabinstein AA.Prolonged
rhythm monitoring for the detection of occult
paroxysmal atrial fibrillation in ischemic stroke of
unknown cause. Circulation 2011;124:477–86.
43. Kutarski A, Małecka B, Kołodzinska A, et al.
Mutual abrasion of endocardial leads: analysis of
explanted leads. Pacing Clin Electrophysiol 2013;
36:1503–11.
44. Migliore A, Broccoli S, Bizzi E, et al. Indirect
comparison of the effects of anti-TNF biological
agents in patients with ankylosing spondylitis by
means of a mixed treatment comparison performed
on efficacy data from published randomised,
controlled trials. J Med Econ 2012;15:473–80.
45. Rozmus G, Daubert JP, Huang DT, et al.
Venous thrombosis and stenosis after implanta-
tion of pacemakers and defibrillators. J Interv Card
Electrophysiol 2005;13:9–19.
46. Polyzos KA, Konstantelias AA, Falagas ME.
Risk factors for cardiac implantable electronic
device infection: a systematic review and meta-
analysis. Europace 2015;17:767–77.
47. Luechinger R, Duru F, Scheidegger MB, et al.
Force and torque effects of a 1.5-Tesla MRI scan-
ner on cardiac pacemakers and ICDs. Pacing Clin
Electrophysiol 2001;24:199–205.
48. Yeung CJ, Karmarkar P, McVeigh ER. Mini-
mizing RF heating of conducting wires in MRI.
Magn Reson Med 2007;58:1028–34.
49. Langman DA, Goldberg IB, Finn JP, et al.
Pacemaker lead tip heating in abandoned and
pacemaker-attached leads at 1.5 Tesla MRI.
J Magn Reson Imaging 2011;33:426–31.
50. Cohen JD, Costa HS, Russo RJ. Determining
the risks of magnetic resonance imaging at 1.5
Tesla for patients with pacemakers and implant-
able cardioverter defibrillators. Am J Cardiol 2012;
110:1631–6.
51. Martin ET, Coman JA, Shellock FG, et al.
Magnetic resonance imaging and cardiac pace-
maker safety at 1.5-Tesla. J Am Coll Cardiol 2004;
43:1315–24.
52. Sommer T, Naehle CP, Yang A, et al. Strategy
for safe performance of extrathoracic magnetic
resonance imaging at 1.5 Tesla in the presence of
cardiac pacemakers in non-pacemaker-dependent
patients: a prospective study with 115 examina-
tions. Circulation 2006;114:1285–92.
53. Boilson BA, Wokhlu A, Acker NG, et al. Safety
of magnetic resonance imaging in patients with
permanent pacemakers: a collaborative clinical
approach. J Interv Card Electrophysiol 2012;33:
59–67.
54. Vahlhaus C, Sommer T, Lewalter T, et al.
Interference with cardiac pacemakers by magnetic
resonance imaging: are there irreversible changes
at 0.5 Tesla? Pacing Clin Electrophysiol 2001;24:
489–95.
55. Gimbel JR, Johnson D, Levine PA, et al. Safe
performance of magnetic resonance imaging on
five patients with permanent cardiac pacemakers.
Pacing Clin Electrophysiol 1996;19:913–9.
56. Tandri H, Zviman MM, Wedan SR, et al.
Determinants of gradient field-induced current in
a pacemaker lead system in a magnetic resonance
imaging environment. Heart Rhythm 2008;5:
462–8.
57. Sommer T, Vahlhaus C, Lauck G, et al. MR
imaging and cardiac pacemakers: in-vitro evalua-
tion and in-vivo studies in 51 patients at 0.5 T.
Radiology 2000;215:869–79.
KEY WORDS atrial fibrillation, bradycardia,
cardiac pacing, heart conduction system,
magnetic resonance imaging, tachycardia
APPENDIX For a supplemental video
and legend, please see the online version
of this article.
Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7
Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0
210

Weitere ähnliche Inhalte

Was ist angesagt?

Percutaneous Pulmonary Valve Interventions
Percutaneous Pulmonary Valve InterventionsPercutaneous Pulmonary Valve Interventions
Percutaneous Pulmonary Valve InterventionsPraveen Nagula
 
Ventricular Assist Device
Ventricular Assist DeviceVentricular Assist Device
Ventricular Assist DeviceNikki Ann Testa
 
Evaluation and management of Pacemaker malfunction
Evaluation and management of Pacemaker malfunctionEvaluation and management of Pacemaker malfunction
Evaluation and management of Pacemaker malfunctionPRAVEEN GUPTA
 
Basic EP study (part1)
Basic  EP  study (part1)Basic  EP  study (part1)
Basic EP study (part1)SolidaSakhan
 
Cardiac resynchronization therapy
Cardiac resynchronization therapyCardiac resynchronization therapy
Cardiac resynchronization therapyRamachandra Barik
 
Follow up and management of pacemaker programming and
Follow up and management of pacemaker programming andFollow up and management of pacemaker programming and
Follow up and management of pacemaker programming anddrskd6
 
Transesophageal echocardiography
Transesophageal echocardiographyTransesophageal echocardiography
Transesophageal echocardiographyAmit Gulati
 
Ventricular assist devices
Ventricular assist devicesVentricular assist devices
Ventricular assist devicesdegnarog
 
Advanced ventricular assist device ppt
Advanced ventricular assist device pptAdvanced ventricular assist device ppt
Advanced ventricular assist device pptkaushikbandopadhyay
 
Right heart catheterization
 Right heart catheterization Right heart catheterization
Right heart catheterizationToufiqur Rahman
 
Electrophysiology study basics
Electrophysiology study basicsElectrophysiology study basics
Electrophysiology study basicsSatyam Rajvanshi
 
Prosthetic heart valves types physiology
Prosthetic heart valves types physiologyProsthetic heart valves types physiology
Prosthetic heart valves types physiologyAswin Rm
 
Basic technical concepts in cardiac pacing
Basic technical concepts in cardiac pacingBasic technical concepts in cardiac pacing
Basic technical concepts in cardiac pacingSunil Reddy D
 

Was ist angesagt? (20)

Percutaneous Pulmonary Valve Interventions
Percutaneous Pulmonary Valve InterventionsPercutaneous Pulmonary Valve Interventions
Percutaneous Pulmonary Valve Interventions
 
fundamentals of pacemaker
fundamentals of pacemaker  fundamentals of pacemaker
fundamentals of pacemaker
 
Ventricular Assist Device
Ventricular Assist DeviceVentricular Assist Device
Ventricular Assist Device
 
Evaluation and management of Pacemaker malfunction
Evaluation and management of Pacemaker malfunctionEvaluation and management of Pacemaker malfunction
Evaluation and management of Pacemaker malfunction
 
Basic EP study (part1)
Basic  EP  study (part1)Basic  EP  study (part1)
Basic EP study (part1)
 
Cardiac resynchronization therapy
Cardiac resynchronization therapyCardiac resynchronization therapy
Cardiac resynchronization therapy
 
Basic ICD treatment_lecture
Basic ICD treatment_lectureBasic ICD treatment_lecture
Basic ICD treatment_lecture
 
Follow up and management of pacemaker programming and
Follow up and management of pacemaker programming andFollow up and management of pacemaker programming and
Follow up and management of pacemaker programming and
 
Transesophageal echocardiography
Transesophageal echocardiographyTransesophageal echocardiography
Transesophageal echocardiography
 
Ventricular assist devices
Ventricular assist devicesVentricular assist devices
Ventricular assist devices
 
Advanced ventricular assist device ppt
Advanced ventricular assist device pptAdvanced ventricular assist device ppt
Advanced ventricular assist device ppt
 
Pacemakers
PacemakersPacemakers
Pacemakers
 
Pacemaker
PacemakerPacemaker
Pacemaker
 
Right heart catheterization
 Right heart catheterization Right heart catheterization
Right heart catheterization
 
Electrophysiology study basics
Electrophysiology study basicsElectrophysiology study basics
Electrophysiology study basics
 
VAD
VADVAD
VAD
 
Atrial septostomy
Atrial septostomyAtrial septostomy
Atrial septostomy
 
Pacemaker
PacemakerPacemaker
Pacemaker
 
Prosthetic heart valves types physiology
Prosthetic heart valves types physiologyProsthetic heart valves types physiology
Prosthetic heart valves types physiology
 
Basic technical concepts in cardiac pacing
Basic technical concepts in cardiac pacingBasic technical concepts in cardiac pacing
Basic technical concepts in cardiac pacing
 

Andere mochten auch

Andere mochten auch (20)

Cardiac pacemakers part-II
Cardiac pacemakers  part-IICardiac pacemakers  part-II
Cardiac pacemakers part-II
 
pacemaker
pacemakerpacemaker
pacemaker
 
Cardiac Pacemaker
Cardiac PacemakerCardiac Pacemaker
Cardiac Pacemaker
 
Pacemakers
PacemakersPacemakers
Pacemakers
 
Pacemakers ppt1
Pacemakers  ppt1Pacemakers  ppt1
Pacemakers ppt1
 
Looping of heart
Looping of heartLooping of heart
Looping of heart
 
Cochlear implant
Cochlear implantCochlear implant
Cochlear implant
 
Defibrillator
Defibrillator Defibrillator
Defibrillator
 
Artificial Cardiac Pacemaker
Artificial Cardiac PacemakerArtificial Cardiac Pacemaker
Artificial Cardiac Pacemaker
 
Dialysis
DialysisDialysis
Dialysis
 
IImplantable Cardioverter Defibrillators (ICDs) - Dr Prithvi puwar
IImplantable Cardioverter Defibrillators (ICDs) - Dr Prithvi puwarIImplantable Cardioverter Defibrillators (ICDs) - Dr Prithvi puwar
IImplantable Cardioverter Defibrillators (ICDs) - Dr Prithvi puwar
 
Basics of medical statistics
Basics of medical statisticsBasics of medical statistics
Basics of medical statistics
 
Cochlear implants
Cochlear implantsCochlear implants
Cochlear implants
 
Cochlear Implants
Cochlear ImplantsCochlear Implants
Cochlear Implants
 
Heart Lung Mechine
Heart Lung MechineHeart Lung Mechine
Heart Lung Mechine
 
Defibrillation
DefibrillationDefibrillation
Defibrillation
 
AC & DC
AC & DCAC & DC
AC & DC
 
Cochlear Implant
Cochlear ImplantCochlear Implant
Cochlear Implant
 
Heart–lung machine
Heart–lung machineHeart–lung machine
Heart–lung machine
 
Pacemaker
PacemakerPacemaker
Pacemaker
 

Ähnlich wie Cardiac pacemakers part-I

Spinal cord Stimulation
Spinal cord StimulationSpinal cord Stimulation
Spinal cord Stimulationpulkit saxena
 
Components of Pacemaker and ICDs - understanding the hardware
Components of Pacemaker and ICDs - understanding the hardwareComponents of Pacemaker and ICDs - understanding the hardware
Components of Pacemaker and ICDs - understanding the hardwareRaghu Kishore Galla
 
3rd Year Project - Design and build of an Electrocardiogram
3rd Year Project - Design and build of an Electrocardiogram3rd Year Project - Design and build of an Electrocardiogram
3rd Year Project - Design and build of an ElectrocardiogramM. Yahia Al Kahf
 
Ieeepro techno solutions ieee 2013 embedded project a review on design of ...
Ieeepro techno solutions    ieee 2013 embedded project a review on design of ...Ieeepro techno solutions    ieee 2013 embedded project a review on design of ...
Ieeepro techno solutions ieee 2013 embedded project a review on design of ...srinivasanece7
 
The Development of Transcranial Magnetic Stimulation
The Development of Transcranial Magnetic StimulationThe Development of Transcranial Magnetic Stimulation
The Development of Transcranial Magnetic Stimulationbijnnjournal
 
DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...
DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...
DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...DrHeena tiwari
 
CM Pacemaker pp.pptx
CM Pacemaker pp.pptxCM Pacemaker pp.pptx
CM Pacemaker pp.pptxMostafaSbaa
 
What is New in Electrophysiology Technologies-Samir Rafla.pptx
What is New in Electrophysiology Technologies-Samir Rafla.pptxWhat is New in Electrophysiology Technologies-Samir Rafla.pptx
What is New in Electrophysiology Technologies-Samir Rafla.pptxAlexandria University, Egypt
 
Introduction to Electrocariography(ecg).doc
Introduction to Electrocariography(ecg).docIntroduction to Electrocariography(ecg).doc
Introduction to Electrocariography(ecg).docwosade3943
 
Senior Design Project
Senior Design ProjectSenior Design Project
Senior Design ProjectHaley Smith
 
Please upload your project drafts or comprehensive outlines (see an .docx
Please upload your project drafts or comprehensive outlines (see an .docxPlease upload your project drafts or comprehensive outlines (see an .docx
Please upload your project drafts or comprehensive outlines (see an .docxblazelaj2
 
A simple portable ecg monitor with iot
A simple portable ecg monitor with iotA simple portable ecg monitor with iot
A simple portable ecg monitor with iotArhamSheikh1
 
CM Function of the heart pp.pptx
CM Function of the heart pp.pptxCM Function of the heart pp.pptx
CM Function of the heart pp.pptxMostafaSbaa
 
B05507010
B05507010B05507010
B05507010inventy
 
Ecg signal processing for detection and classification of cardiac diseases
Ecg signal processing for detection and classification of cardiac diseasesEcg signal processing for detection and classification of cardiac diseases
Ecg signal processing for detection and classification of cardiac diseasesIAEME Publication
 

Ähnlich wie Cardiac pacemakers part-I (20)

Spinal cord Stimulation
Spinal cord StimulationSpinal cord Stimulation
Spinal cord Stimulation
 
Components of Pacemaker and ICDs - understanding the hardware
Components of Pacemaker and ICDs - understanding the hardwareComponents of Pacemaker and ICDs - understanding the hardware
Components of Pacemaker and ICDs - understanding the hardware
 
Pacemaker and anaesthesia
Pacemaker and anaesthesiaPacemaker and anaesthesia
Pacemaker and anaesthesia
 
3rd Year Project - Design and build of an Electrocardiogram
3rd Year Project - Design and build of an Electrocardiogram3rd Year Project - Design and build of an Electrocardiogram
3rd Year Project - Design and build of an Electrocardiogram
 
Ecg article
Ecg articleEcg article
Ecg article
 
Ieeepro techno solutions ieee 2013 embedded project a review on design of ...
Ieeepro techno solutions    ieee 2013 embedded project a review on design of ...Ieeepro techno solutions    ieee 2013 embedded project a review on design of ...
Ieeepro techno solutions ieee 2013 embedded project a review on design of ...
 
The Development of Transcranial Magnetic Stimulation
The Development of Transcranial Magnetic StimulationThe Development of Transcranial Magnetic Stimulation
The Development of Transcranial Magnetic Stimulation
 
DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...
DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...
DENTAL ARMAMENTARIUMS AND ITS EFFECT ON CARDIAC ARMAMENTARIUMS AND ITS FUNCTI...
 
Electrocardiograph: A Portable Bedside Monitor
Electrocardiograph: A Portable Bedside MonitorElectrocardiograph: A Portable Bedside Monitor
Electrocardiograph: A Portable Bedside Monitor
 
CM Pacemaker pp.pptx
CM Pacemaker pp.pptxCM Pacemaker pp.pptx
CM Pacemaker pp.pptx
 
What is New in Electrophysiology Technologies-Samir Rafla.pptx
What is New in Electrophysiology Technologies-Samir Rafla.pptxWhat is New in Electrophysiology Technologies-Samir Rafla.pptx
What is New in Electrophysiology Technologies-Samir Rafla.pptx
 
PACEMAKER
PACEMAKERPACEMAKER
PACEMAKER
 
Introduction to Electrocariography(ecg).doc
Introduction to Electrocariography(ecg).docIntroduction to Electrocariography(ecg).doc
Introduction to Electrocariography(ecg).doc
 
Senior Design Project
Senior Design ProjectSenior Design Project
Senior Design Project
 
Please upload your project drafts or comprehensive outlines (see an .docx
Please upload your project drafts or comprehensive outlines (see an .docxPlease upload your project drafts or comprehensive outlines (see an .docx
Please upload your project drafts or comprehensive outlines (see an .docx
 
A simple portable ecg monitor with iot
A simple portable ecg monitor with iotA simple portable ecg monitor with iot
A simple portable ecg monitor with iot
 
CM Function of the heart pp.pptx
CM Function of the heart pp.pptxCM Function of the heart pp.pptx
CM Function of the heart pp.pptx
 
D1103012736
D1103012736D1103012736
D1103012736
 
B05507010
B05507010B05507010
B05507010
 
Ecg signal processing for detection and classification of cardiac diseases
Ecg signal processing for detection and classification of cardiac diseasesEcg signal processing for detection and classification of cardiac diseases
Ecg signal processing for detection and classification of cardiac diseases
 

Mehr von Ramachandra Barik

Intensive care of congenital heart disease.pptx
Intensive care of congenital heart disease.pptxIntensive care of congenital heart disease.pptx
Intensive care of congenital heart disease.pptxRamachandra Barik
 
Management of Hypetension.pptx
Management of Hypetension.pptxManagement of Hypetension.pptx
Management of Hypetension.pptxRamachandra Barik
 
CRISPR and cardiovascular diseases.pdf
CRISPR and cardiovascular diseases.pdfCRISPR and cardiovascular diseases.pdf
CRISPR and cardiovascular diseases.pdfRamachandra Barik
 
Pacemaker Pocket Infection After Splenectomy
Pacemaker Pocket Infection After SplenectomyPacemaker Pocket Infection After Splenectomy
Pacemaker Pocket Infection After SplenectomyRamachandra Barik
 
A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...
A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...
A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...Ramachandra Barik
 
Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...
Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...
Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...Ramachandra Barik
 
Anticoagulation therapy during pregnancy
Anticoagulation therapy during pregnancyAnticoagulation therapy during pregnancy
Anticoagulation therapy during pregnancyRamachandra Barik
 
Intracoronary optical coherence tomography
Intracoronary optical coherence tomographyIntracoronary optical coherence tomography
Intracoronary optical coherence tomographyRamachandra Barik
 
A roadmap for the human development
A roadmap for the human developmentA roadmap for the human development
A roadmap for the human developmentRamachandra Barik
 
Left ventricular false tendons
Left ventricular false tendonsLeft ventricular false tendons
Left ventricular false tendonsRamachandra Barik
 

Mehr von Ramachandra Barik (20)

Willens's syndrome.pptx
Willens's syndrome.pptxWillens's syndrome.pptx
Willens's syndrome.pptx
 
Intensive care of congenital heart disease.pptx
Intensive care of congenital heart disease.pptxIntensive care of congenital heart disease.pptx
Intensive care of congenital heart disease.pptx
 
Management of Hypetension.pptx
Management of Hypetension.pptxManagement of Hypetension.pptx
Management of Hypetension.pptx
 
CRISPR and cardiovascular diseases.pdf
CRISPR and cardiovascular diseases.pdfCRISPR and cardiovascular diseases.pdf
CRISPR and cardiovascular diseases.pdf
 
Pacemaker Pocket Infection After Splenectomy
Pacemaker Pocket Infection After SplenectomyPacemaker Pocket Infection After Splenectomy
Pacemaker Pocket Infection After Splenectomy
 
Piccolo Duct Occluder.pdf
Piccolo Duct Occluder.pdfPiccolo Duct Occluder.pdf
Piccolo Duct Occluder.pdf
 
MISPLACED ECG LEADS.pptx
MISPLACED ECG LEADS.pptxMISPLACED ECG LEADS.pptx
MISPLACED ECG LEADS.pptx
 
A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...
A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...
A Case of Device Closure of an Eccentric Atrial Septal Defect Using a Large D...
 
Arrythmia-IV.pptx
Arrythmia-IV.pptxArrythmia-IV.pptx
Arrythmia-IV.pptx
 
Arrythmia-III.pptx
Arrythmia-III.pptxArrythmia-III.pptx
Arrythmia-III.pptx
 
Arrythmia-II.pptx
Arrythmia-II.pptxArrythmia-II.pptx
Arrythmia-II.pptx
 
Arrythmia-I.pptx
Arrythmia-I.pptxArrythmia-I.pptx
Arrythmia-I.pptx
 
Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...
Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...
Trio of Rheumatic Mitral Stenosis, Right Posterior Septal Accessory Pathway a...
 
Anticoagulation therapy during pregnancy
Anticoagulation therapy during pregnancyAnticoagulation therapy during pregnancy
Anticoagulation therapy during pregnancy
 
Coronary guidewire
Coronary guidewireCoronary guidewire
Coronary guidewire
 
Intracoronary optical coherence tomography
Intracoronary optical coherence tomographyIntracoronary optical coherence tomography
Intracoronary optical coherence tomography
 
Brugada syndrome
Brugada syndromeBrugada syndrome
Brugada syndrome
 
A roadmap for the human development
A roadmap for the human developmentA roadmap for the human development
A roadmap for the human development
 
Intra aortic balloon pump
Intra aortic balloon pumpIntra aortic balloon pump
Intra aortic balloon pump
 
Left ventricular false tendons
Left ventricular false tendonsLeft ventricular false tendons
Left ventricular false tendons
 

Kürzlich hochgeladen

See the 2,456 pharmacies on the National E-Pharmacy Platform
See the 2,456 pharmacies on the National E-Pharmacy PlatformSee the 2,456 pharmacies on the National E-Pharmacy Platform
See the 2,456 pharmacies on the National E-Pharmacy PlatformKweku Zurek
 
Kolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Kolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowKolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Kolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowNehru place Escorts
 
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service MumbaiLow Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbaisonalikaur4
 
Glomerular Filtration rate and its determinants.pptx
Glomerular Filtration rate and its determinants.pptxGlomerular Filtration rate and its determinants.pptx
Glomerular Filtration rate and its determinants.pptxDr.Nusrat Tariq
 
97111 47426 Call Girls In Delhi MUNIRKAA
97111 47426 Call Girls In Delhi MUNIRKAA97111 47426 Call Girls In Delhi MUNIRKAA
97111 47426 Call Girls In Delhi MUNIRKAAjennyeacort
 
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...narwatsonia7
 
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaCall Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaPooja Gupta
 
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...narwatsonia7
 
High Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near Me
High Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near MeHigh Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near Me
High Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near Menarwatsonia7
 
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking ModelsMumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Modelssonalikaur4
 
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceCollege Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceNehru place Escorts
 
VIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service Mumbai
VIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service MumbaiVIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service Mumbai
VIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service Mumbaisonalikaur4
 
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️saminamagar
 
Call Girls Viman Nagar 7001305949 All Area Service COD available Any Time
Call Girls Viman Nagar 7001305949 All Area Service COD available Any TimeCall Girls Viman Nagar 7001305949 All Area Service COD available Any Time
Call Girls Viman Nagar 7001305949 All Area Service COD available Any Timevijaych2041
 
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbersBook Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbersnarwatsonia7
 
Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.ANJALI
 
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original PhotosBook Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photosnarwatsonia7
 
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls ServiceCall Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Servicesonalikaur4
 
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️saminamagar
 
Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...narwatsonia7
 

Kürzlich hochgeladen (20)

See the 2,456 pharmacies on the National E-Pharmacy Platform
See the 2,456 pharmacies on the National E-Pharmacy PlatformSee the 2,456 pharmacies on the National E-Pharmacy Platform
See the 2,456 pharmacies on the National E-Pharmacy Platform
 
Kolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Kolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowKolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Kolkata Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
 
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service MumbaiLow Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
 
Glomerular Filtration rate and its determinants.pptx
Glomerular Filtration rate and its determinants.pptxGlomerular Filtration rate and its determinants.pptx
Glomerular Filtration rate and its determinants.pptx
 
97111 47426 Call Girls In Delhi MUNIRKAA
97111 47426 Call Girls In Delhi MUNIRKAA97111 47426 Call Girls In Delhi MUNIRKAA
97111 47426 Call Girls In Delhi MUNIRKAA
 
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
 
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaCall Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
 
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
Call Girls Frazer Town Just Call 7001305949 Top Class Call Girl Service Avail...
 
High Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near Me
High Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near MeHigh Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near Me
High Profile Call Girls Mavalli - 7001305949 | 24x7 Service Available Near Me
 
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking ModelsMumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
 
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceCollege Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
 
VIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service Mumbai
VIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service MumbaiVIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service Mumbai
VIP Call Girls Mumbai Arpita 9910780858 Independent Escort Service Mumbai
 
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
 
Call Girls Viman Nagar 7001305949 All Area Service COD available Any Time
Call Girls Viman Nagar 7001305949 All Area Service COD available Any TimeCall Girls Viman Nagar 7001305949 All Area Service COD available Any Time
Call Girls Viman Nagar 7001305949 All Area Service COD available Any Time
 
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbersBook Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
Book Call Girls in Kasavanahalli - 7001305949 with real photos and phone numbers
 
Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.
 
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original PhotosBook Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
Book Call Girls in Yelahanka - For 7001305949 Cheap & Best with original Photos
 
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls ServiceCall Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Service
 
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in green park  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in green park DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
 
Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Kanakapura Road Just Call 7001305949 Top Class Call Girl Service A...
 

Cardiac pacemakers part-I

  • 1. THE PRESENT AND FUTURE STATE-OF-THE-ART REVIEW Cardiac Pacemakers: Function, Troubleshooting, and Management Part 1 of a 2-Part Series Siva K. Mulpuru, MD, Malini Madhavan, MBBS, Christopher J. McLeod, MBCHB, PHD, Yong-Mei Cha, MD, Paul A. Friedman, MD ABSTRACT Advances in cardiac surgery toward the mid-20th century created a need for an artificial means of stimulating the heart muscle. Initially developed as large external devices, technological advances resulted in miniaturization of electronic circuitry and eventually the development of totally implantable devices. These advances continue to date, with the recent introduction of leadless pacemakers. In this first part of a 2-part review, we describe indications, implant-related complications, basic function/programming, common pacemaker-related issues, and remote monitoring, which are relevant to the practicing cardiologist. We provide an overview of magnetic resonance imaging and perioperative management among patients with cardiac pacemakers. (J Am Coll Cardiol 2017;69:189–210) © 2017 by the American College of Cardiology Foundation. A BRIEF HISTORY OF CARDIAC PACING Cardiac pacing, electrical stimulation to modify or create cardiac mechanical activity, began in the 1930s with Hyman’s “artificial pacemaker” (his term), in which a hand crank created an electric current that drove a DC generator whose electrical impulses were directed to the patient’s right atrium through a needle electrode placed intercostally. At that time, Hyman faced professional skepticism, litigation, and accusa- tions of creating “an infernal machine that interferes with the will of God,” and he never found a manu- facturer for his machine (1). After World War II, public perception changed and daring pioneers made great advances. Large, external, alternating current–powered pacemakers tethered to an extension cord gave way to battery- powered, transistorized, “wearable” pacemakers (Central Illustration). The birth of pacing was linked to cardiac surgery, which was burgeoning. In 1957, at the University of Minnesota, C. Walton Lillehei had per- formed over 300 open-heart operations on young adults and children with congenital defects. Dr. Lil- lehei and coworkers developed a myocardial wire for post-operative pacing. On October 31, 1957, a munic- ipal power failure in Minneapolis lasted 3 h and led to the tragic death of a baby (2). The following day, Dr. Lillehei asked Earl Bakken, a hospital equipment en- gineer, to build a battery-powered device to prevent future tragedies. Bakken modified a circuit for an electronic metronome he had seen in the April 1956 issue of Popular Electronics that used transistors, which had been invented 10 years before. He modi- fied the 2-transistor circuit so that the electrical pul- ses would pace the heart, rather than power a speaker. The device was immensely successful. He named the company he founded Medtronic. Other innovations would lead to the founding of other, now familiar manufacturers. From the Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota. Dr. Friedman has received research support from St. Jude Medical; and is a consultant and advisory board member for Medtronic and Boston Scientific. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose. Manuscript received June 29, 2016; revised manuscript received October 6, 2016, accepted October 18, 2016. Listen to this manuscript’s audio summary by JACC Editor-in-Chief Dr. Valentin Fuster. J O U R N A L O F T H E A M E R I C A N C O L L E G E O F C A R D I O L O G Y V O L . 6 9 , N O . 2 , 2 0 1 7 ª 2 0 1 7 B Y T H E A M E R I C A N C O L L E G E O F C A R D I O L O G Y F O U N D A T I O N P U B L I S H E D B Y E L S E V I E R I S S N 0 7 3 5 - 1 0 9 7 / $ 3 6 . 0 0 h t t p : / / d x . d o i . o r g / 1 0 . 1 0 1 6 / j . j a c c . 2 0 1 6 . 1 0 . 0 6 1
  • 2. In 1960, Rune Elmqvist and Ake Senning of Stockholm placed the first fully implant- able pacemaker in Arne Larsson. Larsson’s wife had pleaded with Senning to use the experimental technology to help her desperately ill husband, who had complete heart block and frequent Stokes-Adams syncopal attacks. To avoid publicity, the initial implantation was performed at night, when the operating rooms were empty. The original system lasted 8 h. Arne Larsson ultimately went on to undergo over 20 pacemaker replacements, and he outlived both his surgeon and device engineer. He was an advo- cate for pacing until his death at 86 years of age in 2002 (3). Although significant advances in pacing technology were developed over the next 50 years, including multichamber pacing, rate responsiveness, device size reduction, internet-based remote monitoring, and marked increases in battery longevity, the basic sys- tem paradigm of an extravascular pulse generator connected to 1 or more leads that traverse the venous system to contact myocardial tissue would not change for 50 years (Central Illustration). However, many pacemaker-related complications (infection, throm- bosis, lead failure, and pneumothorax) are related to this basic construct, particularly the leads. This has led to a paradigm shift: the development of a leadless pacemaker, in which the entire device is placed within cardiac chambers. Batteryless devices, which harvest cardiac motion to power pacing circuits, are on the horizon as a coming paradigm shift. In this first part of the 2-part review of cardiac pacing, we explore the state-of-the-art: the basics of pacing physiology, pacing modes and indications, periprocedural management, complications, basic troubleshooting, perioperative management for nonpacemaker procedures, and cardiac magnetic resonance imaging (CMR) of patients with pacemakers. In part 2 of our review (4), we will examine recent advances and future directions, including resynchronization for heart failure, His bundle pacing, remote monitoring, and leadless and batteryless devices. BASICS OF CARDIAC PACING Normal cardiac activity begins in the sinus node, where cells with intrinsic automaticity act as pace- maker cells. Electrical wave fronts then spread across the atria to the atrioventricular (AV) node, which they pass through to enter the His-Purkinje system to rapidly spread to and depolarize the ventricles (Figure 1). When intrinsic cardiac automaticity or CENTRAL ILLUSTRATION An Overview of the History of Cardiac Pacing Paradigm Shifts in Cardiac Pacemakers 1950s AC-powered pacemakers tethered to an extension cord (Furman) 1950s Battery-powered transistorized “wearable” pacemakers (Lillehei/Bakken) 1958 First fully implantable pacemaker (Elmqvist/ Senning) 2015 Implantable pacemaker— basic system had not evolved significantly 2016 Leadless pacemaker—the entire device is placed within cardiac chambers Future Batteryless devices, which harvest cardiac motion to power pacing circuits Mulpuru, S.K. et al. J Am Coll Cardiol. 2017;69(2):189–210. Historically, pacing developed using large, external, alternating current (AC)–powered devices, which subsequently evolved to “wearable” transistorized battery powered pacemakers—both comprise the era of external devices. A paradigm shift occurred with the introduction of the entirely implantable pacemaker, composed of an extravascular pulse generator connected to a transvenous lead in contact with the myocardium. This paradigm continues to this day. An emerging and rapidly developing new paradigm is that of leadless pacemakers, which are available for clinical use. Batteryless pacemakers that harvest cardiac mechanical motion to generate current, or that modify or add cells to introduce biological pacing activity, are under active investigation. A B B R E V I A T I O N S A N D A C R O N Y M S CIED = cardiac implantable electronic device CS = coronary sinus PMT = pacemaker-mediated tachycardia PVARP = post-ventricular atrial refractory period RV = right ventricle TARP = total atrial refractory period Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 190
  • 3. conduction integrity fails, the electrical excitability of cardiac tissue allows a small, external electrical stimulus to drive myocytes to threshold, leading to depolarization of neighboring myocytes through energy-consuming biological processes and the consequent propagation of an electrical wave front, with near-simultaneous muscular contraction via excitation-contraction coupling. Pacemakers provide that external stimulus. Pacemakers consist of a pulse generator or can, which contains the battery and electronics, and leads, which travel from the can to contact the myocardium, to deliver a depolarizing pulse and to sense intrinsic cardiac activity (Figure 2). Insulation materials separate the conductor cables and the lead tip elec- trodes. Depending on the relationship between the cables, the leads can be designed as coaxial (a tube within a tube) or coradial (side-by-side coils). The lead fixation to the myocardium may be active (with an electrically active helix at its tip for mechanical stability) or passive (electrically inert tines anchor the lead). Disruption of conductor elements and insu- lation materials results in either high impedance (fracture) or low impedance due to short-circuiting (insulation breach), respectively. Pacing occurs when a potential difference (voltage) is applied be- tween the 2 electrodes. In bipolar pacing, the poten- tial difference occurs between the lead tip (cathode) FIGURE 1 The Cardiac Conduction System (A) Conduction begins in the SA node (left). Impulse propagation through the atria gives rise to the P-wave (bottom left). The impulse is then delayed in the AV node to allow blood to flow to the ventricles; wave front travel through the AV node is not seen on the surface elec- trocardiogram. The wave fronts then pass through the His–Purkinje system to rapidly activate the ventricular myocardium, giving rise to the large amplitude QRS complex. (B) An anatomic specimen showing the location of key conduction system elements. (Left) An external view of the heart with the region of the sinoatrial node in the epicardium at the juncture of the SVC and right atrium indicated. The structure itself is not visible to the naked eye. (Right) The right atrial and ventricular free walls have been removed to reveal the position of the AV node anterior to the CS and atrial to the TV, situated in Koch’s triangle (bounded by the TV, CS, and tendon of Todaro; not shown). Reproduced with permission from Hayes et al. (40). AV ¼ atrioventricular; CS ¼ coronary sinus; FO ¼ fossa ovalis; IVC ¼ inferior vena cava; RAA ¼ right atrial appendage; SA ¼ sinoatrial; SVC ¼ superior vena cava; TV ¼ tricuspid valve. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 191
  • 4. and a proximal ring (anode). With unipolar pacing, current is delivered between the lead tip and the pulse generator can. The minimum amount of energy required to depolarize myocardium is called the stimulation threshold. The delivered stimulus is described by 2 characteristics: its amplitude (measured in volts) and its duration (measured in milliseconds). The energy required to pace the myocardium depends on the programmed pulse width and on the voltage delivered between the electrodes. An exponential relationship (strength- duration curve) exists between the stimulation threshold and the pulse amplitude and duration (Figure 3). This is clinically relevant, in that opti- mizing the pulse width and amplitude can signifi- cantly affect current drain and battery longevity. Another clinical use for these parameters includes reprogramming to prevent extracardiac (e.g., phrenic) stimulation by lowering the pacing voltage to mini- mize the risk of far-field capture and increasing the pulse width to ensure cardiac stimulation. At im- plantation, a typical acceptable threshold is under 1.5 V with a pulse width of 0.5 ms, but this may vary, and higher values are accepted for coronary sinus leads. Pacemaker leads are built with steroid-eluting collars to prevent tissue-lead fibrosis and to mitigate the threshold rise over time that is otherwise seen. The relationship among voltage, current, and resistance is defined by Ohm’s Law (V ¼ IR), where V ¼ voltage, I ¼ current, and R ¼ resistance. Classi- cally, Ohm’s law uses impedance, which includes the effects of inductance and capacitance in opposing flow; however, for clinical purposes, these are generally negligible and resistance alone is used. Pacing lead conductors are designed to have a low internal resistance, to minimize wastage of energy as resistive heat. Because permanent pacemakers generate a constant voltage, the higher the pacing resistance (the load, resistance of current passage through tissue) the lower the current drain (I ¼ V/R), and the lower the rate of battery depletion per each pacing pulse. Thus, lead tip electrodes are optimized to have a relatively high resistance (typically 400 to 1,200 U) to minimize current flow and preserve bat- tery (1). Up to 50% of the current drain from the battery is used for pacing, whereas the other one-half is used for sensing and housekeeping functions (algorithms and storage of electrograms) (5). FIGURE 2 Transvenous Pacemaker System and its Associated Components (A) A pacemaker system primarily consists of a hermitically encased can containing the battery and all of the circuitry placed in the pre- pectoral region. The can is connected to the myocardial tissue by a pacemaker lead. The leads contain conductor coils to the distal electrodes separated by insulation material. (B) The leads are of coaxial (coil within a coil) or coradial (side-by-side coils) design, depending on the arrangement of the conductor coils. (C) The lead tips are attached to the myocardium by a penetrating helix (active fixation) or by tines that embed in the myocardial trabeculations (passive fixation). Reproduced with permission from Hayes et al. (40). Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 192
  • 5. Resistance is affected by many factors: lead-tissue interface, body position, and tissue edema, to name a few, but abrupt changes (>30%) may suggest lead malfunction. Initial pacemakers functioned as pacing-only de- vices without the capability to sense intrinsic cardiac activity. Asynchronous pacing provided a minimum heart rate, but was complicated by atrial and ven- tricular dissociation, and by competition between pacing impulses and intrinsic cardiac activity. This led to the development of sensing and demand- pacing modes (discussed later). Sensing is the pro- cess whereby a pacemaker determines the timing of the cardiac depolarization of the chamber that a lead is in. To effectively sense, the pacemaker must opti- mally sense near-field depolarization signals, reject near-field repolarization signals (T-waves), and reject far-field signals (signals generated by tissues that the lead electrode is not in contact with) as well as non- physiological signals (electromagnetic interference generated by cell phones, and so on). Atrial channels are optimized to sense in the frequency range of 80 to 100 Hz, and ventricular channels in the 10- to 30-Hz range (6,7). Typical amplitude ranges for signals recorded from atrial and ventricular leads are 1.5 to 5 mV and 5 to 25 mV, respectively. Electrogram ampli- tudes below these values may lead to undersensing, the failure to detect cardiac depolarization, with possible inappropriate delivery of pacing pulses (Figure 4). PACING MODE In response to a sensed intracardiac signal, a pace- maker may inhibit output, trigger output, or pace in a different chamber after a timed delay. This function is governed by the programmed pacing mode. The pacing mode is described with a 4- or 5-letter code (e.g., DDDR), in which the first position identifies the chamber paced (A for atrium, V for ventricle, D for dual/both), the second position indicates the chamber sensed, the third position denotes the device response to sensed events (I for inhibit, T for trigger, or D for dual [both]), the fourth position indicates whether rate response is on, and the fifth position (when used), indicates whether multisite pacing is employed in the atrium (A), ventricle (V), or both (D). With regard to the device response (position 3 in the code), inhibition indicates that a sensed event inhibits pacing and initiates a new timing cycle. If the timing cycle (the length of which is determined by the programmed pacing rate) elapses before another event is sensed, then pacing will occur. This is most commonly used with single-chamber pacing, such as VVI or VVIR (also called demand pacing); the presence of an intrinsic depolarization above the pacing rate inhibits pacing. If the intrinsic rate drops below the programmed rate, pacing occurs. Rate response (dis- cussed in the following text) detects physical activity, such as exercise, and functionally increases the lower rate (shortens the cycle length) for pacing. With triggered pacing, a sensed event may trigger pacing in the same chamber or, typically after a programmed delay, in the other chamber. A triggered mode alone is uncommonly used. With the dual mode (e.g., DDDR) both triggering and inhibition are used. In the DDD mode (Figure 5), inhibition occurs in the atrium if the intrinsic atrial rate exceeds the programmed lower rate. An atrioventricular clock is then started. In the absence of an intrinsic ventricular event, a ventricu- lar pacing spike is triggered; a sensed intrinsic ven- tricular event inhibits pacing. In all pacing modes, a lower rate limit indicates the rate below which pacing occurs (this is the slowest heart rate that should be present, although some features and algorithms may permit programmable exceptions), and an upper rate limit indicates the fastest rate at which the pacemaker will pace, although intrinsic cardiac activity has no such limit. Common pacing modes and their clinical utility follow: FIGURE 3 Relationships Among Chronic Ventricular Strength–Duration Curves From a Canine, Expressed as Potential, Charge, and Energy 0 0 1 2 3 4 5 0.2 0.4 0.6 0.8 1.0 Strength Duration Curve Charge Energy 1.5 Pulse Width (ms) Threshold(V,μJ,μC) With longer pulse widths, less voltage is required to stimulate the heart. The energy requirements, which are a function of the programmed pulse width and voltage, are larger at very short and very long pulse widths. The energy expended is lower when the pulse width is kept constant at 0.4 to 0.5 ms, and the voltage is adjusted to have an adequate safety margin. Reprinted with permission from Stokes et al. (41). J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 193
  • 6. DDD. Standard dual-chamber pacing is used when the sinus mode is intact, but AV conduction impaired. Sinus activity is sensed and will trigger ventric- ular pacing following a programmed AV delay (p-synchronous pacing). DDDR. Rate response is added when sinus and AV nodal function are both abnormal; the rate responsive feature provides chronotropic response. Most modern devices use sensors to determine the rate respon- siveness to physiological demands. VVI AND VVIR. Ventricular-only pacing is used in patients with chronic atrial fibrillation, or infrequent pauses or bradycardias. The potential for tracking atrial arrhythmias is eliminated. Rate response provides chronotropic support when needed. Single-chamber pacemakers with leads in the ventricle can deliver these modes. AAIR. This mode is reserved for isolated sinus node dysfunction with intact AV nodal conduction. It avoids ventricular pacing and, when delivered by a single-chamber pacemaker, eliminates the need for a lead that crosses the tricuspid valve. VOO/DOO. Asynchronous modes are programmed to avoid recognition of electrical activity, most commonly electrocautery, CMR signals, or other electromagnetic interference (EMI). These modes prevent sensing of extrinsic electrical activity, which may be “misinterpreted” as native cardiac events, inhibiting pacing, or lead to rapid ventricular pacing up to the upper rate limit if sensing occurs on the FIGURE 4 Sensing Abnormalities in Modern Pacemakers (A) Myopotential oversensing (star) and inhibition of pacing in a patient with a pacemaker programmed to VVI mode. (B) Atrial undersensing on a tracing from an ambulatory monitor. This is best seen at the fifth ventricular complex. A P-wave precedes the intrinsic ventricular event, but this is not sensed (green arrow), and an atrial pacing artifact (black arrow) occurs immediately before the intrinsic ventricular complex. The ventricular complex is sensed in the cross-talk sensing window. As a result, the ventricular pacing artifact (red arrow) is delivered early after the intrinsic ventricular event (i.e., ventricular safety pacing). Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 194
  • 7. atrial lead. In current clinical practice, these modes are only used temporarily to prevent oversensing. BASIC PROGRAMMABLE FEATURES Several basic programmable features are important for arrhythmia management. MODE SWITCHING. In dual-chamber systems, an atrial tachyarrhythmia sensed in DDD mode can lead to ventricular pacing at rates up to the upper rate limit because atrial events are tracked in the ventricle. Mode-switching algorithms identify the presence of an atrial tachyarrhythmia and switch to a nontracking mode (VVI, DVI, or DDI, with or without rate response). Although the specific algorithms vary, the presence of more atrial events than ventricular events at a rate above a programmable value triggers a mode switch, with the pacing rate then driven by the programmed lower rate limit or the sensor- indicated rate. Following a mode-switch event, atrial activity is scanned so that a tracking mode re- sumes upon arrhythmia termination. Mode-switching events generate internet-based alerts when available (discussed in part 2 [4]), and are reported upon device interrogation. Mode-switch events indicate the pres- ence of atrial arrhythmias and potential need for anticoagulation. Inappropriate mode switching can occur with far-field R-wave oversensing (Figure 6). AVOIDING VENTRICULAR PACING. Isolated right ventricular (RV) pacing activates the interventricular septum before the left ventricular (LV) lateral wall, seen as a left bundle branch block pattern on the electrocardiogram (ECG) due to propagation of the electrical wave front away from the sternum. This results in LV dyssynchrony and mismatched timing between chamber walls, with deleterious effects on LV function and adverse clinical outcomes, including FIGURE 5 Dual-Chamber Pacemaker Timing Cycles TARP AV AV AV AV ID VA VA LRLR AP VP AP VS AS VP PVARP TARP AV PVARP TARP AV PVARP The timing cycle in DDD consists of a lower rate (LR) limit, an atrioventricular (AV) interval, a ventricular refractory period, a post-ventricular atrial refractory period (PVARP), and an upper rate limit. If intrinsic atrial and ventricular activity occur before the LR limit times out, both channels are inhibited and no pacing occurs. In the absence of intrinsic atrial and ventricular activity, AV sequential pacing occurs (first cycle). If no atrial activity is sensed before the ventriculoatrial (VA) interval is completed, an atrial pacing artifact is delivered, which initiates the AV interval. If intrinsic ventricular activity occurs before the termination of the AV interval, the ventricular output from the pacemaker is inhibited (i.e., atrial pacing with intrinsic conduction [second cycle]). If a P-wave is sensed before the VA interval is completed, output from the atrial channel is inhibited. The AV interval is initiated, and if no ventricular activity is sensed before the AV interval terminates, a ventricular pacing artifact is delivered (i.e., P-synchronous ventricular pacing [third cycle]). Reproduced with permission from Hayes et al. (40). AP ¼ atrial pace; AS ¼ atrial-sensed event; ID ¼ intrinsic deflection; TARP ¼ total atrial refractory period (which includes the AV interval and PVARP); VP ¼ ventricular pace; VS ¼ ventricular-sensed event. TABLE 1 Recommendations for Permanent Pacing in Sinus Node Dysfunction Class I: Permanent Pacemaker Implantation Is Indicated for 1. Documented symptomatic bradycardia, including frequent sinus pauses that produce symptoms. 2. Symptomatic chronotropic incompetence. 3. Symptomatic sinus bradycardia that results from required drug therapy for medical conditions. Class IIa: Permanent Pacemaker Implantation Is Reasonable for 1. Sinus node dysfunction with heart rates <40 beats/min when a clear association between significant symptoms consistent with bradycardia and the actual presence of bradycardia has not been documented. 2. Syncope of unexplained origin when clinically significant abnormalities of sinus node function are discovered or provoked in electrophysiological studies. Class IIb: Permanent Pacemaker Implantation May Be Considered in 1. Minimally symptomatic patients with chronic heart rate <40 beats/min while awake. Class III: Permanent Pacemaker Implantation Is Not Indicated in 1. Asymptomatic patients. 2. Patients for whom the symptoms suggestive of bradycardia have been clearly documented to occur in the absence of bradycardia. 3. Patients with symptomatic bradycardia due to nonessential drug therapy. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 195
  • 8. heart failure and mortality (8). Several studies have reported RV pacing-induced cardiomyopathy rates of up to 20% with frequent RV pacing among patients with preserved ejection fraction. Male sex, wide native QRS complexes, and frequent RV pacing (>20%) are reported predictors of RV pacing- associated cardiomyopathy (9). For this reason, al- gorithms to avoid or minimize RV pacing have been developed for dual-chamber pacemakers. The fundamental concept is adjustment of timing in- tervals to deliver atrial pacing whenever required, but to extend the AV interval as long as possible to permit intrinsic conduction and thus eliminate the need for ventricular pacing. Implementations include use of an atrial pacing mode (AAI) with an automatic switch to ventricular pacing (DDD) if AV conduction fails, and AV interval prolongation to supraphysiological values with shortening if intrinsic conduction does not manifest. These algorithms effectively decrease the percentage of ventricular pacing in patients without permanent complete AV block (10,11). The benefit of RV pacing avoidance algorithms on devel- opment of atrial fibrillation, heart failure, and mor- tality is less clear from clinical trials. Moreover, very long AV delays (>400 ms) result in atrial contraction occurring during early diastole, resulting in cannon A waves and adverse hemodynamics, referred to as “pseudopacemaker syndrome.” RV pacing avoidance FIGURE 6 Far-Field R-Wave Oversensing, a Cause of Inappropriate Atrial Fibrillation Detection by a Biventricular Pacemaker (Top) The pacing lead is commonly positioned in the right atrial appendage (RAA), which hangs over the right ventricle. The spacing between the distal helix and ring electrode determines the size of the antenna of the pacemaker lead. The large signal generated by the right ventricle may be oversensed by the atrial lead (V deflection, inset), leading to overcounting of atrial events and inappropriate detection. (Bottom) An example of a device tracing in which the far-field ventricular signals were sensed on the atrial channel (*), and an inappropriate mode switch (MS) (þ) occurs. Also present are paced atrial im- pulses (AP), atrial events sensed during the blanking period (Ab), and atrial events sensed in the PVARP period (AR). Reproduced with permission from Seet et al. (42). BV ¼ biventricular pacing; PVARP ¼ post-ventricular atrial refractory period; RA ¼ right atrium. TABLE 2 Recommendations for Pacing in Acquired Atrioventricular Conduction Abnormalities Class I: Permanent Pacemaker Implantation Is Indicated for 1. Third- or advanced–second-degree AV block: a) If associated with symptoms or ventricular arrhythmias. b) In awake, asymptomatic patients: i. In sinus rhythm, with documented asystole $3.0 s or any escape rate <40 beats/min, or originating from below the AV node. ii. With AF and bradycardia with 1 or more pauses >5 s. iii. With neuromuscular diseases such as myotonic muscular dystrophy, Kearns-Sayre syndrome, Erb dystrophy (limb- girdle muscular dystrophy), and peroneal muscular atrophy, with or without symptoms. iv. That is precipitated by exercise, and in the absence of myocardial ischemia. v. With average awake ventricular rates of 40 beats/min or faster if cardiomegaly or LV dysfunction is present or if the site of block is below the AV node. 2. Recurrent syncope, reproduced by CSM induction of ventricular asystole >3 s. Class IIa: Permanent Pacemaker Implantation Is Reasonable for 1. Persistent third-degree AV block with an escape rate >40 beats/min in asymptomatic adult patients without cardiomegaly. 2. Asymptomatic second-degree AV block at intra- or infra-His levels found at electrophysiological study (HV >100 ms). 3. First- or second-degree AV block with symptoms similar to those of pacemaker syndrome or hemodynamic compromise. 4. Syncope not demonstrated to be due to AV block when other likely causes have been excluded. 5. Symptomatic neurocardiogenic syncope associated with bradycardia documented spontaneously or at the time of tilt-table testing. Class IIb: Permanent Pacemaker Implantation May Be Considered for 1. Neuromuscular diseases such as myotonic muscular dystrophy, Erb dystrophy (limb-girdle muscular dystrophy), and peroneal muscular atrophy with any degree of AV block (including first- degree AV block), with or without symptoms, because of unpredictable progression of AV conduction disease. 2. AV block in the setting of drug use and/or drug toxicity when the block is expected to recur even after the drug is withdrawn. Class III: Permanent Pacemaker Implantation Is Not Indicated in 1. Asymptomatic first-degree or fascicular block AV block; and type I second-degree AV block at the supra-His (AV node) level. 2. AV block that is expected to resolve and is unlikely to recur (e.g., drug toxicity, Lyme disease, or transient increases in vagal tone or during hypoxia in sleep apnea syndrome in the absence of symptoms). 3. Asymptomatic hypersensitive cardioinhibitory response to carotid sinus stimulation. 4. Situational vasovagal syncope in which avoidance behavior is effective and preferred. AF ¼ atrial fibrillation; AV ¼ atrioventricular; CSM ¼ carotid sinus massage; HV ¼ His-ventricle; LV ¼ left ventricular. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 196
  • 9. algorithms can lead to pacing below the programmed lower rate. Pause-dependent cases of ventricular proarrhythmia were reported in patients with RV pacing avoidance algorithms (12–14). In contrast to standard dual-chamber pacemakers, biventricular pacemakers aim to maximize ventricu- lar pacing to deliver the highest possible dose of cardiac resynchronization therapy (CRT, discussed in part 2 [4]). Algorithms that shorten the AV interval when intrinsic conduction or PVCs are sensed have been developed for CRT systems. RATE RESPONSE Rate response (also called rate-adaptive pacing) refers to an increase in the pacing rate in response to physical, mental, or emotional exertion. Rate- adaptive pacing improves exercise capacity in pa- tients with chronotropic incompetence. The most TABLE 3 Pacemaker-Related Complications Complication Reported Frequency (%) Pneumothorax 0.9–1.2 (22) Cardiac perforation <1 (44) Hemothorax <1 Significant pocket hematoma requiring 3.5 (19) Surgery Interruption of anticoagulation Prolongation of hospital stay Lead dislodgement Right-sided leads 1.8 (22) LV leads 5.7 (22) Venous thrombosis and obstruction 1–3 (45) CIED device infection 1.0–1.3 (46) Mechanical lead complications <1 CIED ¼ cardiac implantable electronic device; LV ¼ left ventricular. FIGURE 7 Immediate Complications Associated With Pacemaker Implantation (A) Chest x-ray after device placement showing pneumothorax. The arrow highlights the border of the collapsed lung. (B) Hematoma associated with blood drainage from the pocket. Chest x-ray (C) and computed tomography (CT) image (D) of arterial placement of a pacing lead. Note that the lead is pointing posteriorly on a lateral chest x-ray while traversing through the ascending aorta on the CT scan. (E) Acute pericardial effusion (arrow) with hemodynamic collapse in a patient after right ventricular lead implantation. Also see Online Video 1. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 197
  • 10. common sensor used to drive rate response is the accelerometer, which rapidly detects physical mo- tion. Limitations include a need for upper extremity motion to drive the heart rate (so that walking with limited arm motion on a treadmill leads to a poor rate response) and abrupt deceleration after a period of moderate-intensity exercise when the metabolic de- mands are still high, although programmable func- tionality can mitigate these limitations. Physiological sensors have used minute ventilation, cardiac contractility, blood temperature, or volume changes to drive rate response. Although more specific, they introduce greater latency and are often used with accelerometers to provide a blended response. Minute ventilation sensors transmit a low-energy current from the lead tip to the pulse generator to measure the variations in pulmonary impedance that occur with respiration. They may not be suitable for patients with tachypnea related to pulmonary disease, and may inappropriately increase the heart rate when patients are connected to ECG monitors that inject small currents into the body, which results in sensor error. INDICATIONS FOR PACING Diseases of the sinus node, AV node, or His-Purkinje system due to aging, fibrosis, inflammation, infarc- tion, or other conditions disrupt cardiac electrical signaling. In general, when symptomatic bradycardias ensue, pacing is indicated. An important consider- ation is whether the bradyarrhythmia is reversible, in which case a temporary pacemaker is preferred. Ex- amples include Lyme disease or inferior myocardial ischemia, which can present with alarming brady- cardia, but often with spontaneous recovery within 1 week. Treatable causes of bradycardia should be considered, and include medications (beta-blockers, calcium-channel blockers, most antiarrhythmic drugs, ivabradine, and others), obstructive sleep apnea (especially during apnea), infections (Lyme disease, Chagas disease, Legionnaires’ disease, psittacosis, Q fever, typhoid fever, typhus, among others), and metabolic conditions (hypothyroidism, anorexia nervosa, hypothermia, and hypoxia). In younger pa- tients, hypervagotonia in association with athleticism or vasovagal spells may lead to slow heart rates. When pacing is warranted, the type of pacemaker (atrial, ventricular, dual-chamber, or biventricular) is deter- mined by the nature of the conduction system defect (sinus node, AV node, or intraventricular conduction delay, such as a left bundle branch block). SINUS NODE DYSFUNCTION. Class I indications for permanent pacemaker implantation are present when it is clear that sinus bradycardia is responsible for symptoms. The bradycardia may manifest as pauses or chronotropic incompetence. The latter is defined as a failure to achieve 70% of the age-predicted maximum heart rate or 100 beats/min during maximal exertion. Class II indications are present when a sinus node abnormality is the likely cause of the symptoms, but correlation is difficult. Sinus bradycardia <40 beats/min in a patient with symp- toms suggestive of bradycardia constitutes a class II indication for pacing when an association between bradycardia and symptoms cannot be definitively demonstrated. Similarly, unexplained syncope in a patient with evidence of sinus node dysfunction is a class II indication. In many patients, medications cause sinus bradycardia; if the medications (such as calcium-channel blockers or beta-blockers) are necessary, then permanent pacemaker implantation is reasonable (Table 1). FIGURE 8 Implant Predictors of Lead Stability V. EGM (unfiltered) Peak to Peak: 13.0 mV +20 mV +10 mV -10 mV -20 mV A B (A) Typical electrogram (EGM) changes at 100 mm/s sweep speed. The EGM widening and ST-segment elevation seen here are good long-term predictors of lead fixation. (B) EGMs in a patient who developed cardiac perforation. Negative current of injury (ST-segment) changes to a positive pattern as the pacemaker lead is pulled back into the right ventricle. Reprinted with permission from van Gelder et al. (21). VS ¼ ventricular- sensed event. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 198
  • 11. ACQUIRED ATRIOVENTRICULAR CONDUCTION DISEASE. Class I indications are present in patients with severe AV conduction disease who are at risk for catastrophic events, such as syncope, falls, or sudden death. With these conditions, conduction disease is commonly infranodal, and is often characterized by a wide QRS bradycardia. High-risk conditions include symptom- atic Mobitz type I (Wenckebach) or type II second- degree AV block, advanced AV block (block of 2 or more consecutive P waves), or complete (third-degree) AV block. Patients with asymptomatic complete heart block or advanced AV block also warrant pacing, despite being asymptomatic (Table 2). NEUROCARDIOGENIC SYNCOPE. Neurally-mediated syncope is a difficult syndrome for the clinician to diagnose and treat. Patients with confirmed carotid sinus hypersensitivity (demonstrated by a pause of 3 s or more, with carotid sinus pressure reproducing symptoms) or other neurally-mediated car- dioinhibitory pauses may benefit from permanent pacemaker implant. Although pacing may be FIGURE 9 X-Ray of a Patient With Twiddler Syndrome Who Developed Loss of Capture and High Impedance on the Ventricular Lead Note that several loops on the ventricular lead can put strain on the lead, leading to fracture and high impedance. FIGURE 10 Intermediate-Term Complications (A) Hypertrophic scar after pacemaker implantation. (B) Keloid formation at the site of device surgery. (C and D) Echocardiographic images of the right ventricular lead through the tricuspid valve (arrow) associated with severe tricuspid regurgitation. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 199
  • 12. effective in patients with an isolated car- dioinhibitory response, often a vasodepressor response coexists, limiting the utility of pacing. Up to 20% of patients with carotid hypersensitivity continue to experience syncopal spells during a 5-year follow-up after pacemaker implantation, and the syncope may related to the vasodepressor component of the syndrome (15). In patients with situational syncope (cough, micturition, and so on), trigger avoidance is emphasized. NEUROMUSCULAR DISEASES. Many progressive neuromuscular diseases, such as the muscular dys- trophies, are of special concern, given the unpre- dictable disease course and their predilection for cardiac muscle and fibrosis in and around the FIGURE 11 Electrical Diagnosis of Lead Failure 3,000 2,500 2,000 1,600 1,200 800 400 Jul 4, 2013 V Unipolar tip Leadless ECG V Bipolar Oct 4, 2013 Configurations Unipolar Bipolar Episode: Noise Reversion (Continued) First Measurement 360 Ω 840 Ω Episode 1 of Page 2 of Ventricular Lead Impedance Ventricular Lead Monitoring: 1-year trend 630 Ω (Unipolar) Last 7 DaysAuto Polarity SwitchA Lifetime Range 330-650 Ω 740->3,000 Ω Jan 3, 2014 Apr 4, 2014 Jul 4, 2014 Jun 16, 2014 BiBi Ω Uni 0 A B (A) Abrupt increase in ventricular lead impedance, consistent with conductor fracture. (B) Electrograms obtained from the patient’s pacemaker. Intermittent, short-duration, high-frequency signals with nonphysiological intervals (red arrow) in a patient with an abrupt change in lead impedance universally points to lead fracture. The red star shows obligatory under sensing during DOO mode and functional noncapture (blue arrow). After reversion to DDDR mode, appropriate sensing (blue triangle) is noted. Modern pacemakers can detect noise and switch to a synchronous mode. Most often the fractures are not radiologically detectable because they are result of very small conductor filar disruptions. ECG ¼ electrogram; SIR ¼ sensor indicated rate; other abbreviations as in Figure 5. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 200
  • 13. His-Purkinje system. A class I indication for pacing is present when second- or third-degree AV block is seen, irrespective of symptoms. Congestive heart failure. Patients with depressed ventricular function, a wide QRS interval, and symptomatic heart failure benefit from cardiac resynchronization pacing, with reductions in heart failure and mortality, discussed in detail in part 2 (4). IMPLANT-RELATED COMPLICATIONS Complication rates range from <1% to 6% with cur- rent implant tools and techniques. These are broadly divided into immediate/procedure-related, intermediate-term, and long-term or late complica- tions (Table 3). Their prompt recognition permits timely management. IMMEDIATE PROCEDURE-RELATED COMPLICATIONS. Transvenous lead placement requires venous puncture in the pre-pectoral region. Due to the proximity of the apex of the lung to vascular targets, pneumothorax (Figure 7A) and hemothorax occur in up to 1% of cases. During implantation, the risk is lowered when vascular access using anatomic land- marks is replaced by vein visualization with cephalic vein cut down (16), contrast venography (17), or ul- trasound guidance (18). Arterial puncture and inad- vertent placement of pacing leads in the arterial system (Figures 7C and 7D) are avoided by advancing the guidewire into the inferior vena cava before advancing sheaths to deliver the leads. Inadvertent placement of the RV lead in the LV through a patent foramen ovale or the middle cardiac vein is avoided during implant by advancing the pacing lead across the pulmonary valve, and then withdrawing it to allow it to drop into the RV cavity. Post-operatively, if lead position is uncertain, the lateral chest x-ray, oblique imaging (right anterior oblique, left anterior FIGURE 12 Long-Term Complications Associated With Pacemakers (A) Lead fracture on a chest x-ray. Lead fracture is associated with high impedance due to structural discontinuity (arrow) of the lead. (B) Lead insulation break, which is typically associated with low impedance. Reprinted with permission from Kutarski et al. (43). (C) Deposition of crystals in the lead, which may be associated with high thresholds and impedances (functional in nature). Reprinted with permission from Marshall et al. (30). (D) Development of fibrosis at the lead myocardial interface, resulting in high thresholds, and impaired sensing, resulting in exit block. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 201
  • 14. oblique), echocardiography, and computed tomogra- phy scan may be helpful. Right bundle branch morphology during pacing suggests a left-sided lead. Intracavitary LV leads may result in thromboembo- lism, and are generally repositioned, or if discovered late after implant, anticoagulation is instituted. Pocket hematoma requiring intervention is infre- quent (3.5%), and occurs more commonly in patients who are taking combinations of anticoagulant and antiplatelet drugs (Figure 7B). Uninterrupted anti- coagulation with warfarin lowers the rate of hema- toma formation compared with perioperative heparin bridging (19). The optimal use of periprocedural novel oral anticoagulants remains unresolved, and when possible, they are discontinued 2 to 3 days before pacemaker placement (20). Pericarditis and cardiac tamponade are mechanical complications associated with cardiac perforation (Figure 7E, Online Video 1). Indications for repositioning a lead when micro- perforation is suspected include refractory peri- carditic pain, persistent effusion, or unacceptable pacing or sensing function. Passive fixation leads have a lower risk of perforation, but are used less frequently than active fixation leads due to the greater ease of site selection and ease of extraction with the latter. Use of soft stylets and recognition of negative current of injury (21) on the electrogram recording enables implanters to minimize the risk of cardiac perforation (Figure 8). Micro- and macro- dislodgements are uncommon after device implant. The risk of dislodgement is higher for coronary sinus pacing leads (22) and passive fixation leads. INTERMEDIATE-TERM COMPLICATIONS. Acute he- matoma increases the risk of pocket and systemic infection. Device movement and subsequent lead dislodgements are infrequently seen in current prac- tice. Manipulation of the pacemaker pocket can put undue strain on the pacemaker lead, resulting in lead malfunction (Figure 9) (23). Excess scar at the incision site is associated with unfavorable cosmetic result, pain, and discomfort. Hypertrophic scar (Figure 10A) and keloid formation (Figure 10B) are due to excess interstitial tissue FIGURE 13 Signs of Pocket Infection (A) Redness and skin changes in a patient 2 weeks after device surgery. (B) Purulence within the pocket. (C) Erosion in a patient who subsequently developed systemic signs of infection after pacemaker surgery. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 202
  • 15. formation during the healing process. The latter en- tity has a raised surface and is frequently associated with itching and photosensitivity. The risk of keloid and hypertrophic scar formation is reduced by use of monofilament suture, good surgical technique, and avoidance of excess tension on the suture line. Ke- loids near the suture line are treated with intrale- sional steroid injection, silicone sheeting, and laser phototherapy. Pacemaker lead placement through the tricuspid valve (TV) is infrequently associated with leaflet perforation and impingement of leaflet motion, resulting in valve dysfunction (Figures 10C and 10D) (24). When this leads to chronic fibrotic changes in the TV, tethering of the leaflet often ensues. Com- plications associated with TV dysfunction are avoided by using soft stylets and by crossing the valve at a lower plane, thereby avoiding chordae on the septal aspect of the tricuspid valve. Pacemaker leads often accumulate adherent fibrous strands composed of fibrotic material or thrombi. Recurrent emboli of mobile echo-dense structures increase the risk of pulmonary hypertension and, in patients with a pat- ent foramen ovale, possibly stroke (25). Other complications may include pocket pain or arm swelling. Pocket pain is infrequently reported after device implant. Pocket pain in the absence of device infection occurs when the device is placed in the subcuticular plane, leading to the stimulation of pain corpuscles (26). Occasionally ventricular pacing is associated with uncomfortable sensation or frank pain (27,28). In the absence of clinically significant coronary artery disease, treatment is often conser- vative, using ventricular pacing avoidance algorithms and occasionally lead repositioning to an area where pacing-related discomfort is minimal. The presence of multiple leads in the venous system or endothelial injury during implantation can predispose patients to development of venous thrombosis and stenosis, which can manifest as unilateral edema or superior vena cava syndrome. LATE COMPLICATIONS. Because it is subject to re- petitive mechanical stress with each cardiac cycle and with shoulder girdle movement in the hostile envi- ronment of the human body, the lead is the most common pacemaker system component to fail. Conductor fracture typically results in non- physiological noise caused by the lead itself (high- frequency, saturated electrograms generated by intermittent contact between disrupted conductor elements, called filars) and can be associated with high lead impedance (Figures 11 and 12A). An insu- lation break results in low impedance and FIGURE 14 Summary of CIED Device Infection Management CIED Device Infections TEE Pocket examination Blood Cultures Pocket infection (neg. blood cultures) Lead erosion (neg. blood cultures) AHA Guidelines for Endocarditis 4-6 weeks of antibiotics Non- S. Aureus - 2 weeks of antibiotics S. Aureus 2-4 weeks of antibiotics 10-14 days of antibiotics 7-10 days of antibiotics Valve endocarditis Lead endocarditis Positive blood cultures with negative TEE The duration of antibiotic therapy depends on imaging (TEE), cultures, microbiology and clinical presentation. AHA ¼ American Heart Association; CIED ¼ cardiac implantable electronic device; neg. ¼ negative; TEE ¼ transesophageal echocardiography. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 203
  • 16. oversensing of signals generated by surrounding structures (e.g., muscles) as conductors are exposed (Figure 12B) (29). Acute venous entry angle, medial venous access near the costoclavicular ligament, sharp turns in the pocket, young age, subpectoral placement of device, tight sutures, and silicone insulation are risk factors associated with lead frac- ture and insulation break. Occasionally, pacing thresholds and impedance rise, typically gradually over months, without any detectable fracture. This results from the development of scar tissue at the electrode myocardial interface (exit block) or the deposition of calcium hydroxyapatite crystals (30) at the lead-tissue interface, which can grow into the lead (Figures 12C and 12D) (30). Steroid elution in current-generation pacing leads has virtually elimi- nated the risk of exit block. Gradual impedance may be misinterpreted as a lead failure, but it is an important distinction; gradual impedance rise re- quires no action in an otherwise functional lead, whereas lead failure may require surgical intervention. Pacemaker infections can involve the pocket, associated endovascular leads, and the valves. The risk is higher for subsequent generator changes than for the initial implant procedure. Diabetes, heart failure, renal failure, corticosteroid use, post- operative hematoma, lack of antibiotic prophylaxis, oral anticoagulation, previous cardiac implantable electronic device (CIED) infection, generator change, FIGURE 15 Pacemaker-Mediated Tachycardia and Upper Rate Behavior VA VA AV PVARP PVARP PVARP PVC, retrograde P PVARP 40 60 80 100 Upper tracking rate: 130 bpm PVARP: 250ms AV delay:150ms VRate 120 140 160 160 140 120 100 80 60 40 20 0 *** A B C D E Atrial Rate Continued in the next column FIGURE 15 Continued (A) Pacemaker-mediated tachycardia. The first beat illustrates AV sequential pacing, followed by a PVC beat. The PVC starts a new VA interval. Any AV decoupling event (most frequently PVCs) can retrogradely conduct to the atrium. An atrial event is sensed if it falls outside of the PVARP. The VA interval expires and a new AV interval is triggered. Ventricular pacing occurs at the end of the AV interval setting up a circuitous series of events termed pacemaker-mediated tachycardia. (B to E) Upper-rate behavior in a patient with a dual-chamber pace- maker. The patient was programmed to an upper tracking rate of 130 beats/min. 1:1 AV conduction occurs at baseline and up to 130 beats/min (B). With an increase in sinus rate above 130 beats/min, some of the P waves fall in the PVARP period (arrows) and are not tracked (C). Progressive PR prolongation is seen before blocked beats (pacemaker Wenckebach). When the sinus rate reaches the TARP (AV interval þ PVARP), every other P-wave falls in the PVARP and is not tracked, resulting in an effective ventricular rate of 75 beats/min (D and E), and an abrupt drop in heart rate (2:1 block rate in this patient, around 150 beats/min). AV ¼ atrioventricular interval; PVC ¼ premature ventricular beat; other abbreviations as in Figure 5. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 204
  • 17. and use of a temporary pacemaker are known risk factors for CIED infection (31). Timing of CIED infec- tion depends on the virulence of the organism. It most commonly occurs within a few weeks, but may present up to a year after device surgery. Early rein- tervention is the strongest risk factor for CIED infec- tion. Pocket re-entry should be avoided with needle aspiration or surgery, unless the swelling is very tense and painful. Redness, purulence, erosion, discharge of gelatinous material, chronic pocket pain, and nonhealing incision site with or without thinning are signs of pocket infection (Figure 13). Systemic blood stream infections and endocarditis of the lead or the valves are often associated with fever, chills, positive blood cultures, and the presence of intra- cardiac vegetations. Appropriate referral to a specialized center managing device infections, com- plete device removal, and targeted antibiotic therapy are associated with better outcomes (32). The dura- tion of antibiotic therapy is summarized in Figure 14. BASIC PACEMAKER TROUBLESHOOTING BATTERY. Malfunction may occur with battery depletion. Elective replacement indicator denotes that 90 days of reliable function remain, whereas end of life indicates a battery depleted to the point of unpredictable function. Recalls are uncommon in pacemakers, in contrast to implantable cardioverter- defibrillators, and most often involve software updates to remedy the unintended consequences of the problem. LEAD DIAGNOSTICS. Pacemakers can be programmed to pace and to sense in a bipolar mode (lead tip to lead ring) or a unipolar mode (lead tip to pulse generator). Bipolar sensing is preferred due to its smaller sensing “antenna,” which minimizes oversensing of extrac- ardiac signals. Bipolar pacing is favored to minimize pectoral muscle stimulation. However, when bipolar thresholds are elevated or sensing is impaired (e.g., due to a damaged conductor to the ring electrode), unipolar pacing or sensing may be attractive. The programmed sensitivity is the smallest signal that will be detected as a cardiac event; thus, a setting of 0.5 mV is twice as sensitive as 1.0 mV. In general, a sensing safety margin of 2 is preferred (sensitivity set to 0.5 mV if atrial electrogram is 1.0 mV). Sensi- tivity is adjusted to improve oversensing or under- sensing. Some devices can automatically adjust sensitivity on the basis of the measured amplitude of sensed events. Two commonly encountered device-related ar- rhythmias are pacemaker-mediated tachycardia and FIGURE 16 Role of Current Pathway and Pacemaker Interference (A) High risk of interference: pre-operative reprogramming or magnet use required. (B) Low risk of electromagnetic interference. FIGURE 17 Algorithm for Pre-Operative Pacemaker Reprograming Pacemaker Is surgical site above the hip? yes PM dependent? No further device programing needed Reprogram to asynchronous pacing mode Or Place a magnet no no Devices are programmed to asynchronous mode when surgical procedures involving electrocautery are performed above the hip in pacemaker-dependent patients. PM ¼ pacemaker. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 205
  • 18. upper rate behavior resulting in high or low ventric- ular rates. 1. Pacemaker-mediated tachycardia (PMT). Ventricu- lar events (premature ectopic beats or paced beats), when conducted retrograde to the atria, set up a circuitous cycle of ventricular pacing at the upper tracking rate (Figure 15A). The retrograde wave front (ventriculoatrial conduction) is sensed on the atrial channel, which then triggers ventricular pacing at the end of the programmed AV interval. Each ventricular-paced beat conducts retrogradely, setting up a circuitous pacemaker-mediated tachy- cardia. Atrioventricular decoupling due to retro- grade ventriculoatrial conduction results in an unfavorable hemodynamic profile, and is associated TABLE 4 Potential Adverse Effects of CMR in Patients With Pacemaker Adverse Effect on Exposure to External Electromagnetic Fields Mechanism Incidence and Risk Factors Prevention Mechanical movement of device or dislocation of lead Effect of static magnetic field on ferromagnetic components of device and lead. Risk is proportional to: 1) strength of magnetic field (1.5-T vs. 3-T); 2) amount of ferromagnetic material in lead and device; 3) distance of CMR bore from device; and 4) stability of device and lead (time from implant). Movement of device or lead is very rare. Current leads have almost no ferromagnetic material. Torsional force from 1.5-T scanner typically not significant (47). Delay CMR in fresh implants to allow lead stabilization. CMR-conditional devices and leads minimize/eliminate ferromagnetic material. Avoid 3-T CMR due to lack of clinical experience. Current induction and heating of tissue at lead interface Radiofrequency and pulsed gradient magnetic fields can induce current in the lead, and the body can complete the circuit. Radiofrequency pulses can heat tissue. Device and leads can amplify this (the “antenna” effect) (48). Tissue edema and necrosis at the lead tip can occur. Lead parameters may change including impedance change, reduced sensing, and elevated threshold. The SAR describes the CMR radiofrequency energy deposition in tissues. Risk of tissue heating depends on lead length and lead configuration, such as loops (48). Epicardial, fractured, and abandoned leads are more likely to cause local heating (49). Risk is higher with thoracic CMR. Changes in lead impedance, sensing, and threshold have been reported immediately after CMR and in the long term. Changes that warrant device reprogramming or revision are rare (38,50–52). Others have reported no changes to lead parameters (53,54). Temporary changes in battery voltage can also be seen and often recover over time (50,54). Elevation in cardiac enzymes, typically of small magnitude, has been reported (39). Minimize SAR while still ensuring image quality; input from magnetic resonance physicist should be considered. Avoid CMR in patients with epicardial, fractured, and aban- doned leads. Appropriate selection of CMR landmark. Assessment of lead immediately after CMR to detect changes that may need intervention. Electromagnetic effects EMI leading to inhibition of pacing or tacking at a faster rate RF pulses in magnetic fields are sensed as true cardiac signals. Magnet mode and pauses in dependent patients are rarely reported (53,55). Newer devices are better shielded from EMI. Program asynchronous pacing in pacemaker-dependent patients. Program demand pacing in nondependent patients. Continuous monitoring during scan. Induction of arrhythmias A strong pulsed gradient field results in myocardial electrical stimulation causing arrhythmias. Demonstrated mostly in animal experiments. Current generated under clinical circumstances is too low to capture myocardium (56). Continuous monitoring of rhythm by personnel trained in cardiac resuscitation. Limit lead length and looped leads (56). Change in Reed switch behavior Static magnetic field can activate the Reed switch. Consequence: asynchronous pacing detection/therapy. Reported incidence variable from 0%–38% (38,54,57). Determined by distance from bore of the magnet. Some CMR-conditional devices use Hall sensor, which has more predictable behavior. Power on reset causes the device to revert to default factory settings; in some devices, this can be VVI mode Combination of static and dynamic RF pulses that induce currents in various pacemaker components in an electromagnetic field. Incidence 0.7%–3.5% (38,39). More likely in devices released before 2002 (39). Discontinue scan if noted during monitoring. Avoid CMR in dependent patients with devices from before 2002. CMR ¼ cardiac magnetic resonance; EMI ¼ electromagnetic interference; RF ¼ radiofrequency; SAR ¼ specific absorption rate. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 206
  • 19. with a sensation of palpitations and fullness in the neck due to canon A waves. PMT is terminated by magnet application (which results in asynchronous pacing and prevents tracking of the retrograde P-wave, thereby breaking the cycle) or by use of PMT algorithms. Algorithms recognize ventricular pac- ing at the upper tracking rate, and they either extend the post-ventricular atrial refractory period (PVARP) or pace the atrium simultaneously with ventricular pacing for 1 cycle. PVARP extension re- sults in functional atrial undersensing, whereas atrial pacing makes the atria refractory to the retrograde impulse. 2. Upper rate behavior. In dual-chamber pacing mode, the upper rate is determined by the total atrial re- fractory period (TARP). The TARP is a combination of AV delay and the PVARP. If properly pro- grammed, the device tracks in a 1:1 fashion until reaching the programmed upper rate. As the atrial rate increases, ventricular pacing cannot violate the upper rate limit, resulting in progressively longer AV intervals. This is referred to as pacemaker Wenckebach. However, if the TARP is excessively long, abrupt 2:1 block may develop with a sudden slowing of the ventricular rate, which may cause symptoms. This occurs because when the atrial rate hits the TARP, every other event falls in the PVARP and is not tracked. Effectively, the ventricular pacing rate is one-half of the maximum tracking rate. Undesirable upper rate behavior is avoided by meticulous attention to intervals, by programming to allow a Wenckebach interval between the maximum tracking rate and the 2:1 block rate, and by allowing rate-adaptive shortening of the AV and PVARP intervals (Figures 15B to 15E). 3. Inappropriate mode switches. Occasionally, far field events or frequent ectopic beats result in high atrial counters and inappropriate detection and classification of events as atrial fibrillation. It is imperative to evaluate electrograms before thera- peutic interventions for management of atrial arrhythmias. PERIOPERATIVE MANAGEMENT OF PACEMAKERS Electrosurgery is the application of a 100-kHz to 4-MHz electric current to cut or coagulate biological tissue. With bipolar electrosurgery, both the active and return electrodes are affixed to the cautery pen placed at the surgical site, maintaining a local current pathway; the more commonly used monopolar elec- trosurgery delivers current from an active electrode in the surgical field to a larger dispersive electrode usually placed on the patient’s thigh or back. If elec- trosurgical current enters a pacemaker lead electrode, it may result in EMI. EMI sensed on the ventricular channel is misinterpreted by the pacemaker as intrinsic cardiac events that inhibit pacing, poten- tially resulting in bradycardia or asystole. When sensed on the atrial lead, EMI may result in tracking with rapid ventricular pacing up to the programmed upper rate limit, or inappropriate mode switch. Electrosurgery-related EMI may also cause a power- on reset (abrupt reversion to nominal mode and pacing parameters), pulse generator damage, atrial and ventricular arrhythmia, or tissue injury at the lead-tissue interface, although these are uncommon (33,34). The risk of electrosurgical EMI depends on the surgical site and dispersive pad location, with the highest risk for surgery of the heart and chest, fol- lowed by the head and neck, shoulder/upper ex- tremity, and abdomen-pelvis. In our experience, pacemaker EMI is nonexistent with hip and lower extremity surgery when the dispersive pad is applied to the lower extremities (Figures 16A and 16B). FIGURE 18 Algorithm for Cardiac Magnetic Resonance in Patients With Legacy, Nonconditional Pacemakers Other imaging modality option (radiology review) MRI Scanning in CIED Patients Yes Don’t scan Consult Imaging may be reasonable Will depend on consult MRI requested FDA approved MRI conditional Pacemaker dependent Temporary PM SICD; recall/advisory CIED at ERI • HRS RN eval pre/post • Radiology/physicist eval • Consent by radiology • Troponin if abandoned lead Scan: Mayo MRI/CIED Protocol Yes No No No No Yes Yes Collaborative working relationships involving heart rhythm professions, radiology service providers, and physicists are required for an efficient and safe imaging program in patients with pacemakers. CIED ¼ cardiac implantable electronic device; ERI ¼ elective replacement indicator; eval ¼ evaluation; FDA ¼ Food and Drug Administration; HRS ¼ heart rhythm services; MRI ¼ magnetic resonance imaging; PM ¼ pacemaker; RN ¼ registered nurse; SICD ¼ subcutaneous implantable cardioverter defibrillator. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 207
  • 20. Professional society guidelines recommend the following steps when surgery is needed in a patient with a pacemaker: 1) use bipolar electrocautery when possible; 2) use short-duration cautery bursts 5 s, allowing for 5 s between bursts when using monopolar electrocautery; and 3) apply pads to direct the current pathway away from the pacemaker and leads (35). When surgery is performed below the umbilicus in pacemaker-dependent patients, or when significant pacing is anticipated during the proced- ure, the dispersive pad is placed on a lower extremity and pacemaker reprogramming is unnecessary (Figure 17). When the surgical site is above the um- bilicus, pre-operative pacemaker interrogation is performed, and the device is reprogrammed to an asynchronous mode for the surgery (VOO, AOO, or DOO); magnet application (which causes the pace- maker to enter an asynchronous mode while the magnet is applied) is an alternative. Pre-operative device reprogramming to an asynchronous mode (VOO, AOO, or DOO) is a must for pacemaker- dependent patients (Figure 17) to prevent device in- hibition. After surgery, the device is interrogated to ensure appropriate function and reprogrammed to its original settings. CMR OF PATIENTS WITH PACEMAKERS CMR has become ubiquitous, so that 75% of patients with pacemakers will likely need a scan at some time following implantation (36). In the 1980s, adverse events, including death, occurred when patients with pacemakers were (usually unknowingly) scanned, as pacemaker leads could act as antennae in the CMR environment, with induced currents during imaging leading to risk of arrhythmia induction, capture threshold changes, and device damage (Table 4). For this reason, governmental organizations and profes- sional society guidelines considered CMR scanning to be contraindicated in patients with pacemakers. Manufacturers subsequently developed CMR- conditional pacemakers, which are specifically designed to be safe in the CMR environment. How- ever, the vast majority of implanted patients have legacy, non–CMR-conditional pacemakers. A growing body of evidence has indicated the safety of CMR in patients with legacy pacemakers implanted after the early 1990s, when imaging is performed in a program established by integrated teams that include radiologists, cardiologists, specialized nurses, and physicists, with continuous monitoring during the scan. Pacemakers made since the 1990s include pass-through filters and other technology to make them more resistant to EMI. In several large series, including the Mayo Clinic se- ries that now exceeds 1,000 scans, imaging of pa- tients with legacy pacemakers has been safely performed (Figure 18) (37,38). The greatest potential risk is that of power-on reset in pacemaker- dependent patients, which may change the pro- grammed mode from asynchronous to synchronous, permitting oversensing of CMR signals and inhibition of pacing output. This was seen in a limited number of older devices from a specific manufacturer (39). Nearly all patients with legacy systems can be safely imaged at centers with dedicated programs. Although pacemaker pulse generator artifacts (particularly when the can is low on the chest wall) may lead to artifact obscuring the image, a large case series observed that the clinical question was answered in over 98% of nonthoracic scans (38), and newer CMR processing algorithms facilitate thoracic imaging. CMR-CONDITIONAL DEVICE AND LEADS. All major device manufacturers have developed leads and pacemakers, cardiac resynchronization devices, and implantable cardioverter-defibrillators that are mag- netic resonance–conditional. CMR-conditional refers to devices that have no known hazards or risks un- der specific magnetic resonance conditions. It is notable that there are no “CMR-safe” devices, which refers to devices that have no known hazards or risks under any condition. CMR-conditional devices have minimal ferromagnetic material, altered filtering, and redesigned lead conductors to mini- mize current induction and heating of tissue. There are specific device and CMR conditions under which such devices can be considered safe. Device re- quirements include: CMR-conditional device paired with CMR-conditional leads, and absence of aban- doned, fractured, or epicardial leads. The device must be implanted in the pectoral region. Magnetic resonance scanning requirements are technical, and are designed to limit energy deposition on CIEDs. These include: static magnetic field of 1.5-T, maximum specific absorption rate (SAR) value of 2 W/kg, maximum gradient slew rate of 200 T/m/s, horizontal closed bore magnet, and supine or dorsal positioning of patients (not on their sides). Although several devices are approved for whole-body scan- ning, some have more limited approval for scanning parts other than the thorax. Continuous monitoring during the scan (ECG, hemodynamics, and symp- toms) by an advanced cardiac life support–trained provider, and interrogation before and after scan- ning are also required. To summarize, CMR can be safely performed in pa- tients with magnetic resonance–conditional devices Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 208
  • 21. and at centers with dedicated programs, in nearly all patients with legacy, non-CMR conditional devices. CONCLUSIONS Advances in pacemakers have led to their widespread use to treat patients with bradycardias and congestive heart failure. A basic understanding of their function, troubleshooting, and management at the time of surgery or imaging enhances patient care. In part 2 of this review (4), we explore recent innovations and future directions. REPRINT REQUESTS AND CORRESPONDENCE: Dr. Paul A. Friedman, Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Roches- ter, Minnesota 55902. E-mail: pfriedman@mayo.edu. R E F E R E N C E S 1. Nelson GD. A brief history of cardiac pacing. Texas Heart Inst J 1993;20:12–8. 2. Aquilina O. A brief history of cardiac pacing. Images Paediatr Cardiol 2006;8:17–81. 3. Altman LK, Arne HW. Larsson, 86; had first internal pacemaker. New York Times January 18, 2002. Available at: http://www.nytimes.com/ 2002/01/18/world/arne-h-w-larsson-86-had-first- internal-pacemaker.html. Accessed November 3, 2016. 4. Madhavan M, Mulpuru SK, McLeod CJ, Cha Y-M, Friedman PA. Advances and future directions in cardiac pacemakers: part 2 of a 2-part series. J Am Coll Cardiol 2017;69:211–35. 5. Lindemans FW, Denier Van der Gon JJ. Current thresholds and liminal size in excitation of heart muscle. Cardiovasc Res 1978;12:477–85. 6. Kleinert M, Elmqvist H, Strandberg H. Spectral properties of atrial and ventricular endocardial signals. Pacing Clin Electrophysiol 1979;2:11–9. 7. Parsonnet V, Myers GH, Kresh YM. Character- istics of intracardiac electrograms II: atrial endo- cardial electrograms. Pacing Clin Electrophysiol 1980;3:406–17. 8. DAVID Trial Investigators. Dual-chamber pacing or ventricular backup pacing in patients with an implantable defibrillator: the Dual Chamber and VVI Implantable Defibrillator (DAVID) Trial. JAMA 2002;288:3115–23. 9. Khurshid S, Epstein AE, Verdino RJ, et al. Inci- dence and predictors of right ventricular pacing- induced cardiomyopathy. Heart Rhythm 2014;11: 1619–25. 10. Sweeney MO, Bank AJ, Nsah E, et al., for the Search AV Extension and Managed Ventricular Pacing for Promoting Atrioventricular Conduction (SAVE PACe) Trial. Minimizing ventricular pacing to reduce atrial fibrillation in sinus-node disease. N Engl J Med 2007;357:1000–8. 11. Stockburger M, Boveda S, Moreno J, et al. Long-term clinical effects of ventricular pacing reduction with a changeover mode to minimize ventricular pacing in a general pacemaker popu- lation. Eur Heart J 2015;36:151–7. 12. Pascale P, Pruvot E, Graf D. Pacemaker syn- drome during managed ventricular pacing mode: what is the mechanism? J Cardiovasc Electro- physiol 2009;20:574–6. 13. Cheng A, Calkins H, Berger RD. Managed ven- tricular pacing below the lower rate limit: is it simply caused by ventricular-based pacing? Heart Rhythm 2009;6:1240–1. 14. Mansour F, Khairy P. Electrical storm due to managed ventricular pacing. Heart Rhythm 2012; 9:842–3. 15. Brignole M, Menozzi C. The natural history of carotid sinus syncope and the effect of cardiac pacing. Europace 2011;13:462–4. 16. Parsonnet V, Roelke M. The cephalic vein cutdown versus subclavian puncture for pace- maker/ICD lead implantation. Pacing Clin Electro- physiol 1999;22:695–7. 17. Burri H, Sunthorn H, Dorsaz PA, et al. Pro- spective study of axillary vein puncture with or without contrast venography for pacemaker and defibrillator lead implantation. Pacing Clin Elec- trophysiol 2005;28 Suppl 1:S280–3. 18. Seto AH, Jolly A, Salcedo J. Ultrasound-guided venous access for pacemakers and defibrillators. J Cardiovasc Electrophysiol 2013;24:370–4. 19. Birnie DH, Healey JS, Essebag V. Device sur- gery without interruption of anticoagulation. N Engl J Med 2013;369:1571–2. 20. Birnie DH, Healey JS, Essebag V. Management of anticoagulation around pacemaker and defi- brillator surgery. Circulation 2014;129:2062–5. 21. van Gelder BM, Bracke FA. Current of injury (COI) pattern recorded from catheter delivered active fixation pacing leads. Pacing Clin Electro- physiol 2008;31:786–7, author reply 787. 22. van Rees JB, de Bie MK, Thijssen J, et al. Implantation-related complications of implantable cardioverter-defibrillators and cardiac resynchro- nization therapy devices: a systematic review of randomized clinical trials. J Am Coll Cardiol 2011; 58:995–1000. 23. Alvarez-Acosta L, Romero Garrido R, Farrais- Villalba M, et al. Reel syndrome: a rare cause of pacemaker malfunction. BMJ Case Rep 2014;2014. 24. Al-Bawardy R, Krishnaswamy A, Bhargava M, et al. Tricuspid regurgitation in patients with pacemakers and implantable cardiac defibrillators: a comprehensive review. Clin Cardiol 2013;36: 249–54. 25. DeSimone CV, Friedman PA, Noheria A, et al. Stroke or transient ischemic attack in patients with transvenous pacemaker or defibrillator and echo- cardiographically detected patent foramen ovale. Circulation 2013;128:1433–41. 26. Furman S, Curtis AB, Conti JB. Recognition and correction of subcuticular malposition of pacemaker pulse generators. Pacing Clin Electro- physiol 2001;24:1224–7. 27. Shvilkin A, Ellis ER, Gervino EV, et al. Painful left bundle branch block syndrome: clinical and electrocardiographic features and further direc- tions for evaluation and treatment. Heart Rhythm 2016;13:226–32. 28. Squara F, Scarlatti D, Bres M, et al. Pacemaker- induced painful left bundle-branch block. Acta Cardiologica 2015;70:735. 29. Swerdlow CD, Ellenbogen KA. Implantable cardioverter-defibrillator leads: design, diagno- stics, and management. Circulation 2013;128: 2062–71, 1–9. 30. Marshall MVP, Baker P, Kiser A. Impedance rise leading to unnecessary ICD lead extraction. Paper presented at: The 5th Asia-Pacific Heart Rhythm Society Scientific Session; October 3–6, 2012; Taipei, Taiwan. 31. Rohacek M, Baddour LM. Cardiovascular implantable electronic device infections: associ- ated risk factors and prevention. Swiss Med Wkly 2015;145:w14157. 32. Baddour LM, Epstein AE, Erickson CC, et al., for the Council on Cardiovascular Surgery and Anesthesia, Council on Cardiovascular Nursing, Council on Clinical Cardiology, Interdisciplinary Council on Quality of Care and Outcomes Research. Update on cardiovascular implantable electronic device infections and their manage- ment: a scientific statement from the American Heart Association. Circulation 2010;121:458–77. 33. Pinski SL, Trohman RG. Interference in implanted cardiac devices, part II. Pacing Clin Electrophysiol 2002;25:1496–509. 34. Schoenfeld MH. Contemporary pacemaker and defibrillator device therapy: challenges confront- ing the general cardiologist. Circulation 2007;115: 638–53. 35. Crossley GH, Poole JE, Rozner MA, et al. The Heart Rhythm Society (HRS)/American Society of Anesthesiologists (ASA) expert consensus state- ment on the perioperative management of patients with implantable defibrillators, pace- makers and arrhythmia monitors: facilities and patient management; this document was devel- oped as a joint project with the American Society of Anesthesiologists (ASA), and in collaboration with the American Heart Association (AHA), and the Society of Thoracic Surgeons (STS). Heart Rhythm 2011;8:1114–54. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Mulpuru et al. J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 Cardiac Pacing: Part 1 209
  • 22. 36. Kalin R, Stanton MS. Current clinical issues for MRI scanning of pacemaker and defibrillator patients. Pacing Clin Electrophysiol 2005;28:326–8. 37. Higgins JV, Watson RE Jr., Jaffe AS, et al. Cardiac troponin T in patients with cardiac implantable electronic devices undergoing mag- netic resonance imaging. J Interv Card Electro- physiol 2016;45:91–7. 38. Nazarian S, Hansford R, Roguin A, et al. A prospective evaluation of a protocol for mag- netic resonance imaging of patients with implan- ted cardiac devices. Ann Intern Med 2011;155: 415–24. 39. Higgins JV, Sheldon SH, Watson RE Jr., et al. “Power-on resets” in cardiac implantable elec- tronic devices during magnetic resonance imaging. Heart Rhythm 2015;12:540–4. 40. Hayes DL, Asirvatham SJ, Friedman PA. Cardiac Pacing, Defibrillation and Resynchroniza- tion: A Clinical Approach. Third Edition. Chichester, UK: Wiley-Blackwell, 2013. 41. Stokes K, Bornzin G. The electrode–biointerface stimulation. In: Barold SS, editor. Modern Cardiac Pacing. Mount Kisco, NY: Futura Publishing Co, 1985:33–77. 42. Seet RC, FriedmanPA, Rabinstein AA.Prolonged rhythm monitoring for the detection of occult paroxysmal atrial fibrillation in ischemic stroke of unknown cause. Circulation 2011;124:477–86. 43. Kutarski A, Małecka B, Kołodzinska A, et al. Mutual abrasion of endocardial leads: analysis of explanted leads. Pacing Clin Electrophysiol 2013; 36:1503–11. 44. Migliore A, Broccoli S, Bizzi E, et al. Indirect comparison of the effects of anti-TNF biological agents in patients with ankylosing spondylitis by means of a mixed treatment comparison performed on efficacy data from published randomised, controlled trials. J Med Econ 2012;15:473–80. 45. Rozmus G, Daubert JP, Huang DT, et al. Venous thrombosis and stenosis after implanta- tion of pacemakers and defibrillators. J Interv Card Electrophysiol 2005;13:9–19. 46. Polyzos KA, Konstantelias AA, Falagas ME. Risk factors for cardiac implantable electronic device infection: a systematic review and meta- analysis. Europace 2015;17:767–77. 47. Luechinger R, Duru F, Scheidegger MB, et al. Force and torque effects of a 1.5-Tesla MRI scan- ner on cardiac pacemakers and ICDs. Pacing Clin Electrophysiol 2001;24:199–205. 48. Yeung CJ, Karmarkar P, McVeigh ER. Mini- mizing RF heating of conducting wires in MRI. Magn Reson Med 2007;58:1028–34. 49. Langman DA, Goldberg IB, Finn JP, et al. Pacemaker lead tip heating in abandoned and pacemaker-attached leads at 1.5 Tesla MRI. J Magn Reson Imaging 2011;33:426–31. 50. Cohen JD, Costa HS, Russo RJ. Determining the risks of magnetic resonance imaging at 1.5 Tesla for patients with pacemakers and implant- able cardioverter defibrillators. Am J Cardiol 2012; 110:1631–6. 51. Martin ET, Coman JA, Shellock FG, et al. Magnetic resonance imaging and cardiac pace- maker safety at 1.5-Tesla. J Am Coll Cardiol 2004; 43:1315–24. 52. Sommer T, Naehle CP, Yang A, et al. Strategy for safe performance of extrathoracic magnetic resonance imaging at 1.5 Tesla in the presence of cardiac pacemakers in non-pacemaker-dependent patients: a prospective study with 115 examina- tions. Circulation 2006;114:1285–92. 53. Boilson BA, Wokhlu A, Acker NG, et al. Safety of magnetic resonance imaging in patients with permanent pacemakers: a collaborative clinical approach. J Interv Card Electrophysiol 2012;33: 59–67. 54. Vahlhaus C, Sommer T, Lewalter T, et al. Interference with cardiac pacemakers by magnetic resonance imaging: are there irreversible changes at 0.5 Tesla? Pacing Clin Electrophysiol 2001;24: 489–95. 55. Gimbel JR, Johnson D, Levine PA, et al. Safe performance of magnetic resonance imaging on five patients with permanent cardiac pacemakers. Pacing Clin Electrophysiol 1996;19:913–9. 56. Tandri H, Zviman MM, Wedan SR, et al. Determinants of gradient field-induced current in a pacemaker lead system in a magnetic resonance imaging environment. Heart Rhythm 2008;5: 462–8. 57. Sommer T, Vahlhaus C, Lauck G, et al. MR imaging and cardiac pacemakers: in-vitro evalua- tion and in-vivo studies in 51 patients at 0.5 T. Radiology 2000;215:869–79. KEY WORDS atrial fibrillation, bradycardia, cardiac pacing, heart conduction system, magnetic resonance imaging, tachycardia APPENDIX For a supplemental video and legend, please see the online version of this article. Mulpuru et al. J A C C V O L . 6 9 , N O . 2 , 2 0 1 7 Cardiac Pacing: Part 1 J A N U A R Y 1 7 , 2 0 1 7 : 1 8 9 – 2 1 0 210