Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Received: 11 January 2017

| Revised: 6 February 2017


| Accepted: 27 February 2017
DOI 10.1002/jca.21534

CONCISE REVIEWS

Extracorporeal photopheresis in pediatric patients:


Practical and technical considerations

Robert A. DeSimone1 | Joseph Schwartz2 | Jennifer Schneiderman3

1
Department of Pathology and Laboratory
Abstract
Medicine, New York-Presbyterian
Hospital-Weill Cornell Medicine, In adults, extracorporeal photopheresis (ECP) is widely utilized for a variety of indi-
New York, New York cations, most commonly cutaneous T-cell lymphoma, acute or chronic graft-versus-
2
Department of Pathology and Cell host disease (GVHD), solid organ transplant rejection, and other autoimmune and T-
Biology, New York-Presbyterian Hospital- cell-mediated disorders. In pediatric patients, the majority of case series and reports
Columbia University Medical Center, have focused on its use in the management of acute and chronic GVHD. Currently
New York, New York utilized ECP technologies were designed for adult patients and there are several chal-
3
Department of Pediatrics, Division of lenges in adapting these technologies for use in children. In our review, we focus on
Hematology/Oncology/Stem Cell
practical considerations and procedural modifications for ECP use in pediatric
Transplantation, Ann & Robert H. Lurie
patients, with special attention to patient safety.
Children’s Hospital of Chicago, Feinberg
School of Medicine, Northwestern
University, Chicago, Illinois KEYWORDS
extracorporeal photopheresis (ECP), graft-versus-host disease (GVHD), pediatrics, therapeutic apheresis
Correspondence
Robert A. DeSimone, M.D., Weill
Cornell Department of Pathology and
Laboratory Medicine, 525 East 68th St.
Starr-1036, New York, NY 10065.
Email: rod9096@nyp.org

1 | INTRODUCTION 2 | ECP DEVICE SELECTION


AND MODE
Apheresis procedures performed on pediatric patients are
often challenging because of these patients’ small sizes and ECP is a type of leukocytopheresis. During the procedure, a
blood volumes. When planning apheresis procedures in pedi- small percentage of the patient’s circulating white blood cells
atric patients, practitioners must adapt equipment designed (WBCs) are collected utilizing a centrifuge bowl, subse-
for adults to children, taking into account many details often quently incubated with a psoralen compound, and finally,
considered routine in adults, including placement of an exposed to ultraviolet A (UVA) light irradiation. The treated
appropriately sized central line, fluid status, tolerance to cells are then reinfused into the patient.3 Approved by the
extracorporeal volume, procedure length, anticoagulant FDA in 1988 for the treatment of cutaneous T-cell lym-
selection and dosing, and overall safe modification of the phoma (CTCL), ECP is categorized by the American Society
procedure.1 Data on the use of extracorporeal photopheresis for Apheresis (ASFA) as a primary, first-line adjunctive or
(ECP) in particular is limited in pediatric patients because supportive therapy for CTCL. ASFA also acknowledges
one of the more commonly used photopheresis systems is ECP as a second-line therapy, either as a standalone treat-
FDA approved only for patients >40 kg.2 Of note, within ment or in conjunction with other therapies, for cellular/
published case series to date, there are very few patients recurrent rejection and rejection prophylaxis in cardiac trans-
weighing <15 kg. Here, we review ECP’s use in pediatric plants, as well as acute and chronic graft-versus-host disease
patients, with special attention to technical considerations for (GVHD).4 Published retrospective experience utilizing ECP
safe and efficacious procedures. in pediatric patients has focused on the management of acute

J Clin Apher. 2017;1–10 wileyonlinelibrary.com/journal/jca V


C 2017 Wiley Periodicals, Inc. | 1
2 | DESIMONE ET AL.

and chronic GVHD, most often when refractory to first-line 3 | ECP’S PROPOSED MECHANISM
therapies.2,5–20 No randomized controlled trials investigating OF ACTION IN GVHD
ECP in pediatric patients have been published to-date.
ECP is commonly performed using the UVAR XTS or GVHD is a multisystem, potentially life-threatening com-
CELLEX photopheresis systems (Therakos Inc., Exton, PA), plication of allogeneic hematopoietic stem cell transplanta-
which are automated. Heparin or acid-citrate-dextrose can be tion. ECP has been shown to be an effective therapy for
used as anticoagulants for either system, although both sys- both acute and chronic GVHD, and its use has enabled clini-
tems are only officially approved for heparin. For the UVAR cians to successfully wean, and in some cases even discon-
XTS, which is a discontinuous flow, single-needle proce- tinue, immunosuppressive medications in both adults and
dure, 125 mL or 225 mL of whole blood is collected into a children.19,23–25
centrifuge bowl; the buffy coat is collected and the remaining ECP’s mechanism of action in GVHD has not been fully
blood components (plasma, red blood cells [RBCs], and pla- elucidated, but it has been proposed to result in a shift from
telets) are returned to the patient. Given the discontinuous an inflammatory (Th1) state to a tolerance (Th2) state, thus
nature of procedures performed with this device, 6 cycles inducing tolerance of donor T lymphocytes to host antigens.
(125 mL bowl) or 3 cycles (225 mL bowl) of whole blood A typical ECP treatment results in reinfusion of approxi-
must be processed to produce a 270 mL mononuclear cell mately 5 3 109 MNCs; during their photoactivation, mono-
(MNC) suspension. Following collection, the suspension is functional and bifunctional DNA adducts form, leading to
incubated with methoxsalen and pumped through a photoac- MNC apoptosis within 24–48 hours. The apoptotic MNCs
tivation chamber, at which time it is exposed to UVA light are engulfed by antigen presenting cells, and their antigens
and then reinfused to the patient.21 The general procedure for are presented in the context of major histocompatibility com-
the CELLEX is similar to the UVAR XTS, however the plex class I and II.26,27 The downstream effects described in
CELLEX is a continuous flow procedure that has both both GVHD and cardiac transplant include reduced pro-
single-needle and double-needle modes and has only one inflammatory cytokines (IL-2, TNF-a, IFN-g) and increased
size centrifuge bowl; for most patients, 1500 mL of whole anti-inflammatory cytokines (TGF-b) through arginine
blood is processed. As a result of these differences, the over- metabolism pathways, as well as increased populations of
all extracorporeal volume of the CELLEX is significantly regulatory T-cells.28,29 The emergence of regulatory T-cell
less than the UVAR XTS (216–266 mL for CELLEX populations during ECP treatments has been correlated with
vs.220–620 mL for UVAR XTS), and is therefore better clinical response.30 Other proposed mechanisms of action in
suited for pediatric patients with less total blood volumes.21 GVHD include expansion of memory CD81 T-cells31 and
In addition, for the CELLEX, the double-needle mode may differentiation of monocytes to immature dendritic cells,32
be safer for low-weight patients since this reduces extracor- which then may acquire the ability to secrete IL-10.32,33 In
poreal volume by 63% relative to the single-needle mode.21 addition, decreased proportions of immature CD19 1 CD21-
To reduce the risk of hypovolemia and hypotension, we rec- lymphocytes have been correlated with ECP response in
ommend selecting and adapting ECP technologies to mini- chronic GVHD, highlighting dysregulation of B lymphocytes
mize extracorporeal volume to <10–15% of total blood as a potential contributing factor.34
volume, if possible.
ECP can also be performed in an off-line fashion using 4 | FLUID STATUS AND
two devices, with a standard cell separator for MNC collec- TRANSFUSION SUPPORT
tion followed by UVA irradiation, as is the practice in sev-
eral centers in Europe. Options for MNC collection include The majority of pediatric case series utilized a RBC prime
the COBE Spectra Apheresis System (Terumo BCT, Lake- for their low-weight patients (Table 1), but weight cutoffs for
wood, CO), COM.TEC (Fresenius SE & Co., Bad Homburg, RBC priming varied greatly, ranging from <15 kg to
Germany), Amicus (Fenwal, Inc., Lake Zurich, IL), and Hae- <40 kg. The manufacturer of UVAR XTS and CELLEX
monetics MCS plus (Haemonetics Corp., Braintree, MA). systems recommends a RBC prime for patients weighing
UVA irradiation can be performed with the UV-MATIC <35 kg while utilizing the CELLEX to avoid symptomatic
(Vilber Lourmat, Eberhardzell, Germany), PIT System (Med hypovolemia.18 For patients >35 kg, the photopheresis cir-
Tech Solutions, Modena, Italy), or Theraflex-ECP (Maco- cuit can be primed with heparinized normal saline. Accord-
Pharma, Mouvaux, France). In the United States, this off-line ing to consensus recommendations published by the Italian
method has largely fallen out of favor following the develop- Society of Hemapheresis and Cell Manipulation and the Ital-
ment of automated systems.22 Of note, many ECP case series ian Group for Bone Marrow Transplantation, all patients
in pediatric patients utilized the COBE Spectra/UV-MATIC <20 kg should receive a RBC prime, irrespective of pre-
technology (Tables 1 and 2).5–10,13,14,16–18 procedure hematocrit.35 In one retrospective study, fluid
TAB LE 1 Review of patients (indications for extracorporeal photopheresis [ECP], weight, age) blood prime criteria, venous access, and ECP schedule in published pediatric case series

Mean or Mean or Blood


DESIMONE

Total no. Total no. median weight median age in prime


Reference patients procedures Indication in kg (range) yr (range) (Y/N) Venous access Schedule
ET AL.

Halle5 8 254 cGVHD (n 5 8) 37 (18–49) 10 (5–15) N Tunnelized or double-lumen 2/wk for 2 wks, 1/wk or once
central catheter and/or every 2 wk for 3 mo
peripheral access

Salvaneschi6 23 NR aGVHD (n59), 35 (17–89) 10 (5–18) N Double-lumen central catheter aGVHD: 3/wk until
cGVHD (n514) (n 5 20); peripheral access improvement; cGVHD: 2
(n 5 3) consecutive days at 2 wk
interval for 3 mo

Messina7 77 NR aGVHD (n533), aGVHD: 30 9 (0.3–21) N Double-lumen central catheter 2 consecutive d/wk for 1 mo,
cGvHD (n544) (10–85), (n 5 50); peripheral access once every wks for 2 mo, then
cGVHD: 35 (n 5 27) monthly for 3 mo
(15–68)

Calore8 15 NR aGVHD (n515) NR 10 (1–18) Y NR 2 consecutive d/wk for 1 mo,


once every wks for 2 mo, then
monthly for 3 mo

Kanold9 23 750 aGVHD (n59), aGVHD: 48 14 (4–18) N Tunnelized or double-lumen 3/wk for 3 wks, then tapering
cGVHD (n514) (13–68), central catheter and/or
cGVHD: 43 peripheral access
(13–80)

Berger10 25 NR aGVHD (n515), 40 (17–72) aGVHD: 11 N NR 2 consecutive d/wk for 1 mo,


cGVHD (n510) (6–18), once every wks for 2 mo, then
cGVHD: monthly for 3 mo
12 (7–19)

Duzovali11 7 133 cGVHD (n57) 32 (16–89) 10 (8–17) Y, <40 kg Double-lumen central catheter 3-5 d/wk, then tapering

Landolfo12 8 157 GVHD 19 (7–35) NR Y, <20 kg Double-lumen central catheter 2 consecutive d/wk for 1 mo, 2
with or without peripheral consecutive d/wk every 2 wk
access for 2 mo, 2 consecutive d/wk
every mo for 6 mo

Linenberger13 7 287 GVHD median NR median NR Y, <25 kg NR NR


(11–38) (3–14)

Schneiderman14 11 334 cGVHD (n 5 9), 29 (19–39) NR N Single or double-lumen central Variable


GVHD catheter, peripheral access
prophylaxis (n 5 2)
|
3

(continues)
4
|

TAB LE 1 (continued)

Mean or Mean or Blood


Total no. Total no. median weight median age in prime
Reference patients procedures Indication in kg (range) yr (range) (Y/N) Venous access Schedule
15
Perotti 73 2360 aGVHD (n 5 50), aGVHD: 32 aGVHD: 10 Y, <15 kg Double-lumen central catheter 2-3/wk until improvement, then
cGVHD (n 5 23) (SD 16), (SD 5), (n526); peripheral access tapering
cGVHD: 39 cGVHD 12 (n547)
(SD 17) (SD 4)

Merlin16 12 NR aGVHD (n 5 12) NR 11 (2–18) NR Tunnelized or double-lumen 3/wk for 3 wks, then tapering
central catheter

Gonzalez Vicent17 27 225 aGVHD (n521), 30 (9–77) 10 (1–17) Y, <15 kg Double-lumen central catheter aGVHD: 2 consecutive d/wk
cGVHD (n56) until improvement then
tapering; cGVHD: 2
consecutive d at 2-wk intervals,
then tapering

Rangarajan18 9 385 aGVHD (n 5 1), 49 (19–86) 14 (4–24) Y, <35 kg Double-lumen central catheter 2/wk until improvement, then
cGVHD (n 5 8) tapering

Uygun2 12 194 aGVHD (n 5 6), 28 (7–68) 12 (2–17) Y, <35 kg Double-lumen central catheter 2/wk for 2 mo, 2/wk every 2
cGVHD (n 5 62) (69%), single-lumen central wks for 2 mo, 2/wk every mo
catheter with peripheral access for at leat 1 yr
(10%), peripheral access only
(21%)

Kapadia19 10 440 aGVHD (n 5 5), 32 (22–65) 10 (8–27) Y, <30 kg Double-lumen central catheter, 2 consecutive sessions/wk, then
cGVHD (n 5 5) peripheral access, vortex port tapering

Nelson20 30 NR aGVHD (n 5 30) 26 (7–138) 10 (1–33) NR NR NR

Abbreviations: NR, not reported; cGVHD, chronic graft-versus-host disease; aGHVD, acute graft-versus-host disease; SD, standard deviation; kg, kilograms; yr, years; Y, yes; N, no; wk, weeks; d, days; mo, months.
DESIMONE
ET AL.
T AB L E 2 Review of extracorporeal photopheresis (ECP) procedure length, number and duration of procedures, ECP devices used, transfusion support, and complications in published pediatric
case series
DESIMONE

Median procedure Device used (COBE


ET AL.

length in hh:mm Median number of Median duration Spectra, UVAR


Reference (range) procedures (range) of ECP (range) XTS, CELLEX) RBC transfusions PLT transfusions Complications

Halle5 02:20 (01:00–04:00) 29 (10–66) 9 mo (2–20) COBE Spectra 6/8 patients (23 units total) 2/8 patients Catheter-related
(4 units total) infections (n 5 2, 25%)

Salvaneschi6 NR NR aGVHD: 5 mo COBE Spectra aGVHD 7/9 patients, NR Hypotension (number of


(0.5-13), cGVHD: cGVHD 1/14 patients patients NR)
17 mo (1-32)

Messina7 03:30 aGVHD: 8 cycles aGVHD: 74 d COBE Spectra Threshold to transfuse: Hb All aGVHD patients Mild hypotension
(03:00-04:00) (2–20), cGVHD: NR (8–467); cGVHD: (n 5 44) and UVAR <12 (if <15 kg), all required for first 8 (n 5 21, 27%),
NR XTS (n 5 33) patients with aGVHD; all procedures abdominal pain (n 5 8,
aGVHD patients required 10%)
RBC transfusions for first 8
procedures

Calore8 03:30 NR 171 d (35-311) COBE Spectra Threshold to transfuse: Hb NR Catheter related
(03:00-04:00) <12 (if <15 kg) infection (n 5 1, 7%)

Kanold9 aGVHD: 02:02 24 (10-68) NR COBE Spectra aGVHD 16% required, aGHVD 15% re- Overall (n52, 8%);
(01:01-04:10); cGVHD 7% required; quired, cGVHD stress/unwillingness,
cGVHD: 01:59 <25 kg 16% required, 1.5% paresthesia, pain at site
(01:12-03:40) >25 kg 8% required required; < 25 kg 7% of line placement,
required, >25kg 7% nausea/vomiting,
required general complaints,
fever and/or sepsis,
interrupted sessions

Berger10 03:30 aGVHD: 12 (4-21), COBE Spectra Threshold to transfuse: NR Abdominal pain (20%),
(03:00-04:00) cGVHD: 22 (10-98) (<40kg), UVAR Hb < 9 2 required de novo
XTS (>40 kg) catheter placement (8%)

Duzovali11 03:30 21 (3-31) 3 mo (4 d–6 mo) UVAR XTS Threshold to transfuse: Hb Threshold to Catheter-related
(03:00-04:00) <9; no additional transfuse: infections (n 5 3, 42%),
transfusions required with <20,000/mL; 2 catheters replaced
blood priming 3/7 patients (29%)
12
Landolfo 2:03 (1:55-2:10) NR NR COBE Spectra NR NR NR
(continues)
|
5
6
|

T AB L E 2 (continued)

Median procedure Device used (COBE


length in hh:mm Median number of Median duration Spectra, UVAR
Reference (range) procedures (range) of ECP (range) XTS, CELLEX) RBC transfusions PLT transfusions Complications

Linenberger13 NR median NR (19–72) median NR (5–16 COBE Spectra (col- 2 RBC units for every 212 total PLT Fever (5%), hypotension
mo) lection) and UVAR/ patient requiring blood transfusions (5%), hypocalcemia
XTS (photoactiva- prime (1 in COBE Spectra, (5%), nausea (4%),
tion) 1 in UVAR/XTS) (40 units catheter occlusion (4%),
total) catheter-related
infections (0.7%)

Schneiderman14 2:58 (1:30-5:03) 15 (7–113) NR UVAR/XTS Median of 5 RBCs/patient 2/11 patients Overall in 31% of
(range 0-14), 9/11 patients (aGVHD); procedure days:
required RBCs; threshold to Threshold > 50,000/ tachycardia, dizziness,
transfuse: Hct < 30 mL nausea, vomiting,
(if 25-40 kg), Hct < 35 (if hypotension, headache,
<25 kg)

Perotti15 NR aGVHD: 18 (12– 24 mo (2–145) COBE Spectra Threshold to transfuse: Threshold to Catheter-related
24), cGVHD: 34 Hb  8 transfuse: 20,000/ infections (n 5 10,
(16–43) mL 14%); chills (n 5 12,
0.5%), abdominal pain
(n 5 7, 0.2%), headache
(n 5 22, 0.9%), fever
(11, 0.4%)

Merlin16 NR 22 (10–56) 4 mo (0.7–9) COBE Spectra NR NR NR


17
Gonzalez Vicent 03:00 (average) 6 (2-25) 30 d (2-442) COBE Spectra NR NR Hypotension (n 5 3,
11%), catheter-related
infections (n 5 3, 11%)

Rangarajan18 1:46 (1:00–3:25) 47 (10–85) 10 mo (1–18) CELLEX Threshold to transfuse: Threshold to Catheter-associated
Hct < 27; 2 patients re- transfuse: <20,000/ thrombosis (n 5 1,
quired blood prime (1 pa- mL; 3 platelet doses 0.2%), delayed bleeding
tient received 53 RBCs, 1 given pre-ECP (n 5 1, 0.2%), catheter-
patient received 10 RBCs); related infections (n 5 4,
excluding blood prime, 10 1%); 15 (3.9%)
RBCs were given pre-ECP procedures cancelled
and 4 RBCs post-ECP
(continues)
DESIMONE
ET AL.
DESIMONE
ET AL.

T AB L E 2 (continued)

Median procedure Device used (COBE


length in hh:mm Median number of Median duration Spectra, UVAR
Reference (range) procedures (range) of ECP (range) XTS, CELLEX) RBC transfusions PLT transfusions Complications
2
Uygun NR 16 (4-36) NR CELLEX Threshold to transfuse: Threshold to Hypotension (n57, 4%),
initially Hct < 27, revised to transfuse: initially palpitation and
Hct <24 <50,000/mL, revised tachycardia (n56, 3%),
to <30,000/mL or increase in purpuric
<50,000/mL with lesions (n54, 2%),
bleeding gastrointestinal bleeding
(n52, 1%), pruritus
(n51, 0.5%),
catheter-related infection
(n51, 0.5%)

Kapadia19 UVAR/XTS: 3:09 UVAR/XTS: 14 8 mo (1-30) UVAR/XTS (n5225 Threshold to Threshold to Line occlusions (n523,
(average), CELLEX: (2-66), CELLEX: 17 procedures) or transfuse: Hct <30 transfuse: <30,000/ 5.2%), alarms (n529,
1:58 (average) (4-71) CELLEX (n5215 mL 7%), hypotension
procedures) (n518, 4%),
hypertension (n56, 2%),
blood products required
(n559, 13%), citrate
toxicity (n56, 1%)

Nelson20 NR NR NR CELLEX NR NR None

Abbreviations: NR, not reported; cGVHD, chronic graft-versus-host disease; aGHVD, acute graft-versus-host disease; hh, hours; mm, minutes; mo, months; d, days; kg, kilograms; RBC, red blood cells; Hb, hemoglobin;
Hct, hematocrit; PLT, platelets.
|
7
8 | DESIMONE ET AL.

boluses with 5% albumin (for patients <20 kg) or normal dose of 12,500 units in 500 mL normal saline, which is
saline (for patients >20 kg) were utilized during the proce- infused at an AC ratio ranging from 8:1 to 16:1, based on
dures utilizing the UVAR XTS without RBC priming, how- the platelet count. During the procedures, they adjust their
ever many patients in this study required peri-procedural AC ratio based on the activated clotting time (ACT), which
transfusion support.14 is maintained at >180 seconds. In their patients at a high
Published pediatric case series2,5–20 vary widely in their risk of bleeding, the authors use citrate anticoagulation,
recommendations for pre-procedural transfusions (Table 2). infused at an AC ratio ranging from 12:1 to 16:1. In patients
For the UVAR XTS and CELLEX systems, the patient’s <10 kg, the authors use 5,000 units of heparin, adjusting its
hematocrit (Hct) must be >27% in order to program the dose based on the ACT. When citrate is used, ionized cal-
machines for effective buffy coat collection. In line with cium is checked before and at multiple time points during
practices described in these published case series, we recom- the procedure. To correct for hypocalcemia, a 100 mg/mL
mend a Hct >27% and a platelet count >20,000/mL in non- calcium gluconate drip is administered at a dose of 0.5–
bleeding patients or >50,000/mL in bleeding patients prior to 1 mL/kg. Bleeding risk assessment in pediatric patients is
the start of the procedure. Many of the patients described in largely based on clinical judgement as no formally validated
these case series required pre- and post-procedural blood scoring systems exist at this time to predict bleeding.
product support, and patients with acute gastrointestinal
GVHD were more likely to require blood products, presum- 7 | SCHEDULE AND DURATION
ably due to their increased ongoing bleeding. The lowest OF ECP TREATMENTS FOR GVHD
weight patients were also more likely to require blood
products. The ideal schedule and duration of ECP for management of
GVHD has not been definitively established, and again, there
5 | ACCESS is great variation in practice. There is some degree of consen-
sus for treatment schedules, especially for acute GVHD, with
As with other apheresis procedures, the placement of an a single-day treatment 2–3 times a week (in some practices
adequately sized, functional line is essential for successful performed as consecutive days, and in others with 1–2 day
completion of the procedure. Central line placement and breaks in between ECP treatments) until clinical improve-
maintenance are especially challenging in pediatric patients. ment, followed by progressive tapering (Table 1). Patients
Successful ECP procedures have been performed in pediatric with chronic GVHD often require treatment for a year or
patients using single-or double-lumen central catheters, more. ECP can has been completely discontinued without a
peripheral access, and vortex ports (Table 1). Line sizes rebound in symptoms,36 but tapering decisions must be indi-
should be based on institutional recommendations regarding vidualized to patients’ symptoms. In addition, more intensive
patients’ age and weight, as well as ECP manufacturer rec- schedules consisting of 3–5 treatments per week for chronic
ommendations. In our pediatric patients undergoing ECP, GVHD have not been shown to provide additional benefit.11
lines with an internal diameter of at least 1.3 mm have been To date, clinical trials comparing different treatment sched-
well-tolerated and successfully used. ules have not been performed.

6 | ANTICOAGULANT SELECTION 8 | ECP COMPLICATIONS


AND DOSING IN PEDIATRIC PATIENTS

There are no standardized or evidence-based protocols for In general, ECP is well-tolerated with few acute side effects.
anticoagulant selection and dosing in pediatric patients The most commonly reported adverse effects include hypo-
undergoing ECP, and practices vary widely among ECP tensive episodes, mild fever during reinfusion, and lethargy
practitioners. The manufacturer user manuals for the UVAR following the procedure for 1–2 days.37 In the reviewed
XTS and CELLEX only describe the use of heparin as an pediatric literature,2,5–20 the most commonly reported
anticoagulant solution, without mention of other anticoagu- adverse events include hypotension (range 0–27%), catheter-
lants. At our institution using CELLEX for pediatric patients, related infections (range 0–42%), and abdominal pain (range
heparin is always selected as the anticoagulant at a dose of 0–20%) (Table 2). In addition, smaller patients (<20 kg)
250 units/kg diluted in 500 mL of normal saline, and is seemed to be at a greater risk of developing adverse events,
infused at a whole blood:anticoagulant (AC) ratio set at 10:1. most commonly hypotension, likely related to their extracor-
Rutella et al. also published their anticoagulant protocol poreal volume constituting a greater fraction of total blood
for CELLEX use in pediatric patients.22 For patients at a low volume. We also recently reported an episode of acute
risk of bleeding, they select heparin as the anticoagulant at a mechanical hemolysis in a 10.6 kg patient, which is a rare
DESIMONE ET AL.
| 9

but potentially fatal adverse event.38 The benefits and risks ease after haematopoietic stem cell transplantation. Br J
of ECP, and of having a central line if necessary, must be Haematol. 2003;122(1):118–127.
carefully weighed and discussed in an interdisciplinary fash- [8] Calore E, Calo A, Tridello G, et al. Extracorporeal photochemo-
ion before starting ECP procedures in pediatric patients. therapy may improve outcome in children with acute GVHD.
Bone Marrow Transplant. 2008;42(6):421–425.
Although this has not been reported, we hypothesize that
centers performing ECP more routinely in pediatric and low- [9] Kanold J, Merlin E, Halle P, et al. Photopheresis in pediatric
graft-versus-host disease after allogeneic marrow transplantation:
weight patients with established protocols will have lower
clinical practice guidelines based on field experience and review
adverse event rates. of the literature. Transfusion. 2007;47(12):2276–2289.
[10] Berger M, Pessolano R, Albiani R, et al. Extracorporeal photo-
9 | CONCLUSIONS pheresis for steroid resistant graft versus host disease in pediatric
patients: A pilot single institution report. J Pediatr Hematol
Although case series in pediatric patients have focused on Oncol. 2007;29(10):678–687.
ECP’s use in GVHD, ECP is a widely-used modality, with [11] Duzovali O, Chan KW. Intensive extracorporeal photochemo-
many centers exploring its use in diseases not mentioned or therapy in pediatric patients with chronic graft-versus-host dis-
ease (cGVHD). Pediatr Blood Cancer. 2007;48(2):218–221.
currently listed as category III indications in the ASFA guide-
lines, including inflammatory bowel diseases, scleroderma, [12] Landolfo A, Isacchi G. Extracorporeal photochemotherapy in
low weight patients with acute and chronic graft-versus-host dis-
and pemphigus vulgaris, to name a few.39 Even in adult
ease. J Clin Apher. 2004;20(1):17–18. abst 26).
patients, health care practitioners are using ECP without fully
[13] Linenberger M, Murtaugh A, Jarosek B, et al. A novel 2-
understanding its mechanism of action, how to adequately
process, 3-step method for extracorporeal photochemotherapy in
dose and schedule it, or an appropriate treatment and tapering small body weight children with steroid resistant GVHD: proc-
schedule. Given these uncertainties with adult patients, even essing efficiencies, treated cell doses, and clinical experience.
less is known with regards to pediatric patients. Researchers J Clin Apher. 2005;20(1):18–19. abst 28).
must continue to investigate ECP’s mechanism of action to [14] Schneiderman J, Jacobson DA, Colins J, et al. The use of fluid
close the gap in knowledge of these areas, with the ultimate boluses to safely perform extracorporeal photopheresis (ECP) in
goal of helping both adult and pediatric practitioners develop low-weight children: A novel procedure. J Clin Apher. 2010;25
standardized treatment protocols. Further investigation of (2):63–69.
ECP’s mechanism of action in various diseases may also lead [15] Perotti C, Del Fante C, Tinelli C, et al. Extracorporeal photoche-
motherapy in graft-versus-host disease: a longitudinal study on
to modification of the procedure to further enhance its efficacy
factors influencing the response and survival in pediatric
and safety in low-weight patients.
patients. Transfusion. 2010;50(6):1359–1369.
[16] Merlin E, Paillard C, Rochette E, et al. Extracorporeal photoche-
R EF ERE NC ES motherapy as second- or first-line therapy of acute GVHD?.
[1] Kim HC. Therapeutic pediatric apheresis. J Clin Apher. 2000;15 Bone Marrow Transplant. 2010;45(5):963–965.
(1–2):129–157. [17] Gonzalez Vicent M, Ramirez M, et al. Analysis of clinical out-
come and survival in pediatric patients undergoing extracorpor-
[2] Uygun V, Daloglu H, Karasu G, et al. Safety and outcomes of
eal photopheresis for the treatment of steroid-refractory GVHD.
extracorporeal photopheresis with the Therakos Cellex system
J Pediatr Hematol Oncol. 2010;32(8):589–593.
for graft-versus-host disease in pediatric patients. J Pediatr Hem-
atol Oncol. 2015;37(3):209–214. [18] Rangarajan HG, Punzalan RC, Camitta BM, et al. The use of
novel TherakosTM Cellex® for extracorporeal photopheresis in
[3] Zic JA, Miller JL, Stricklin GP, et al. The North American expe-
treatment of graft-versus-host disease in paediatric patients. Br J
rience with photopheresis. Ther Apher. 1999;3(1):50–56.
Haematol. 2013;163(3):357–364.
[4] Schwartz J, Padmanabhan A, Aqui N, et al. Guidelines on the
[19] Kapadia E, Wong E, Perez-Albuerne E, Jacobsohn D. Extracor-
use of therapeutic apheresis in clinical practice-evidence-based
poreal photopheresis performed on the CELLEX® compared
approach from the writing committee of the American Society
with the UVAR-XTS® instrument is more efficient and better
for Apheresis: The seventh special issue. J Clin Apher. 2016;31
tolerated in children with steroid-refractory graft-versus-host dis-
(3):149–338.
ease. Pediatr Blood Cancer. 2015;62(8):1485–1488.
[5] Halle P, Paillard C, D’incan M, et al. Successful extracorporeal
[20] Nelson AS, Pinkard SL, Carey P, et al. Extracorporeal photophe-
photochemotherapy for chronic graft-versus-host disease in pedi- resis (ECP) for treatment of refractory graft-versus-host disease
atric patients. J Hematother Stem Cell Res. 2002;11(3):501–512. in children after allogeneic hematopoietic stem cell transplanta-
[6] Salvaneschi L, Perotti C, Zecca M, et al. Extracorporeal photo- tion. Biol Blood Marrow Transplant. 2016;22(3):S255–S256
chemotherapy for treatment of acute and chronic GVHD in (abst 366).
childhood. Transfusion. 2001;41(10):1299–1305. [21] Wong ECC, Jacobsohn D. ECP in children and adolescents. In:
[7] Messina C, Locatelli F, Lanino E, et al. Extracorporeal photo- Greinix H, Knobler R, eds. Extracorporeal Photopheresis. Ber-
chemotherapy for paediatric patients with graft-versus-host dis- lin/Boston: Walter de Gruyter GmbH & Co. KG; 2012: 8–47.
10 | DESIMONE ET AL.

[22] Rutella S, Valentini CG, Ceccarelli S, et al. Extracorporeal pho- [33] Rutella S, Danese S, Leone G. Tolerogenic dendritic cells: cyto-
topheresis for paediatric patients experiencing graft-versus-host kine modulations comes of age. Blood. 2006;108(5):1435–1440.
disease (GVHD). Transfus Apher Sci. 2014;50(3):340–348.
[34] Kuzmina Z, Greinix HT, Knobler R, et al. Proportions of imma-
[23] Apisarnthanarax N, Donato M, Korbling M, et al. Extracorporeal ture CD191CD21- B lymphocytes predict the response to
photopheresis therapy in the management of steroid-dependent extracorporeal photopheresis in patients with chronic graft-
cutaneous graft-versus-host disease after allogenic stem cell versus-host disease. Blood. 2009;114(3):744–746.
transplantation: feasibility and results. Bone Marrow Transplant.
2003;31(6):459–465. [35] Pierelli L, Perseghin P, Marchetti M, et al. Extracorporeal photo-
pheresis for the treatment of acute and chronic graft-versus-host
[24] Child FJ, Ratnavel R, Watkins P, et al. Extracorporeal photophe-
resis (ECP) in the treatment of chronic graft-versus-host disease disease in adults and children: best practice recommendations
(GVHD). Bone Marrow Transplant. 1999;23(9):881–887. from an Italian Society of Hemapheresis and Cell Manipulation
(SIdEM) and Italian Group for Bone Marrow Transplantation
[25] Couriel DR, Hosing C, Saliba R, et al. Extracorporeal photoche-
(GITMO) consensus process. Transfusion. 2013;53(10):2340–
motherapy for the treatment of steroid-resistant chronic GVHD.
2352.
Blood. 2006;107(8):3074–3080.
[26] Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endoge- [36] Dippel E, Goerdt S, Orfanos CE. Long-term extracorporeal pho-
nous activators of dendritic cells. Nat Med. 1999;5(11):1249–1255. toimmunotherapy for treatment of chronic cutaneous graft-
versus-host disease: observation in four patients. Dermatology.
[27] Voll RE, Herrmann M, Roth EA, et al. Immunosuppressive
effects of apoptotic cells. Nature. 1997;390(6658):350–351. 1999;198(4):370–374.

[28] Peritt D. Potential mechanisms of photopheresis in hematopoietic [37] Marshall SR. Technology insight: ECP for the treatment of
stem cell transplantation. Biol Blood Bone Marrow Transplant. GvHD – can we offer selective immune control without general-
2006;12(1Suppl2):7–12. ized immunosuppression?. Nat Clin Pract Oncol. 2006;3(6):
[29] George JF, Gooden CW, Guo L, Kirklin JK. Role for CD4(1) 302–314.
CD25(1) T cells in inhibition of graft rejection by extracorporeal [38] DeSimone RA, Wontakal SN, Lyashchenko AK, Schwartz J.
photopheresis. J Heart Lung Transplant. 2008;27(6):616–622. Acute mechanical hemolysis as a complication of extracorporeal
[30] Di Biaso I, Di Maio L, Bugarin C, et al. Regulatory T cells and photopheresis in a low-weight child. J Clin Apher., in press.
extracorporeal photochemotherapy: correlation with clinical
[39] Dunbar NM, Raval JS, Johnson A, et al. Extracorporeal photo-
response and decreased frequency of proinflammatory T cells.
pheresis practice patterns: an international survey by the ASFA
Transplantation. 2009;87(9):1422–1425.
ECP subcommittee. J Clin Apher., in press.
[31] Yamashita K, Horwitz ME, Kwatemaa A, et al. Unique abnor-
malities of CD4(1) and CD8(1) central memory cells associ-
ated with chronic graft-versus-host disease improve after
How to cite this article: DeSimone RA, Schwartz J,
extracorporeal photopheresis. Biol Blood Marrow Transplant.
2006;12(1 Suppl 2):22–30.
Schneiderman J. Extracorporeal photopheresis in pedi-
atric patients: Practical and technical considerations.
[32] Spisek R, Gasova Z, Barunkova J. Maturation state of dendritic
cells during the extracorporeal photopheresis and its relevant for J Clin Apher. 2017;00:1–10. https://doi.org/10.1002/
the treatment of chronic graft-versus-host disease. Transfusion. jca.21534
2006;46(1):55–65.

You might also like