Vous êtes sur la page 1sur 10

Cardiovascular Surgery

Effect of Repetitive Intra-Arterial Infusion


of Bone Marrow Mononuclear Cells in Patients
With No-Option Limb Ischemia
The Randomized, Double-Blind, Placebo-Controlled Rejuvenating
Endothelial Progenitor Cells via Transcutaneous Intra-arterial
Supplementation (JUVENTAS) Trial
Martin Teraa, MD, PhD*; Ralf W. Sprengers, MD, PhD*; Roger E.G. Schutgens, MD, PhD;
Ineke C.M. Slaper-Cortenbach, PhD; Yolanda van der Graaf, MD, PhD; Ale Algra, MD, PhD;
Ingeborg van der Tweel, PhD; Pieter A. Doevendans, MD, PhD;
Willem P.Th.M. Mali, MD, PhD; Frans L. Moll, MD, PhD; Marianne C. Verhaar, MD, PhD

Background—Patients with severe limb ischemia may not be eligible for conventional therapeutic interventions. Pioneering
clinical trials suggest that bone marrow–derived cell therapy enhances neovascularization, improves tissue perfusion, and
prevents amputation. The objective of this trial was to determine whether repetitive intra-arterial infusion of bone marrow
mononuclear cells (BMMNCs) in patients with severe, nonrevascularizable limb ischemia can prevent major amputation.
Methods and Results—The Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation
(JUVENTAS) trial is a randomized, double-blind, placebo-controlled clinical trial in 160 patients with severe,
nonrevascularizable limb ischemia. Patients were randomly assigned to repetitive (3 times; 3-week interval) intra-
arterial infusion of BMMNC or placebo. No significant differences were observed for the primary outcome, ie, major
amputation at 6 months, with major amputation rates of 19% in the BMMNC versus 13% in the placebo group (relative
risk, 1.46; 95% confidence interval, 0.62–3.42). The safety outcome (all-cause mortality, occurrence of malignancy, or
hospitalization due to infection) was not significantly different between the groups (relative risk, 1.46; 95% confidence
Downloaded from http://ahajournals.org by on December 27, 2018

interval, 0.63–3.38), neither was all-cause mortality at 6 months with 5% versus 6% (relative risk, 0.78; 95% confidence
interval, 0.22–2.80). Secondary outcomes quality of life, rest pain, ankle-brachial index, and transcutaneous oxygen
pressure improved during follow-up, but there were no significant differences between the groups.
Conclusions—Repetitive intra-arterial infusion of autologous BMMNCs into the common femoral artery did not reduce
major amputation rates in patients with severe, nonrevascularizable limb ischemia in comparison with placebo. The
general improvement in secondary outcomes during follow-up in both the BMMNC and the placebo group, as well,
underlines the essential role for placebo-controlled design of future trials.
Clinical Trial Registration—URL: http://www.clinicaltrials.gov. Unique identifier: NCT00371371.  (Circulation. 2015;
131:851-860. DOI: 10.1161/CIRCULATIONAHA.114.012913.)
Key Words: atherosclerosis ◼ cells ◼ peripheral vascular diseases

S evere limb ischemia, due to advanced peripheral artery dis-


ease (PAD), is associated with a high risk of cardiovascu-
lar events and all-cause mortality1–4 and a poor prognosis with
40%.3,4 Severe limb ischemia is associated with poor quality of
life (QoL)7 and high treatment costs,8 especially when amputa-
tion is inevitable.8,9 With an estimated annual incidence of 500
respect to limb preservation.5,6 In nonrevascularizable, so-called to 1000 new cases per million individuals in Western society,3
no-option patients with severe limb ischemia, 6-month major
amputation rates have been reported to range from 10% to
Clinical Perspective on p 860

Received August 22, 2014; accepted January 5, 2015.


From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S.,
W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius
Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.),
and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands.
*Drs Teraa and Sprengers contributed equally.
The online-only Data Supplement is available with this article at http://circ.ahajournals.org/lookup/suppl/doi:10.1161/CIRCULATIONAHA.114.
012913/-/DC1.
Correspondence to Marianne C. Verhaar, MD, PhD, University Medical Center Utrecht, Heidelberglaan 100, HP F.03.227, 3584 CX Utrecht, The
Netherlands. E-mail m.c.verhaar@umcutrecht.nl
© 2015 American Heart Association, Inc.
Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.114.012913

851
852  Circulation  March 10, 2015

which is ever increasing in concert with the increase in cardio- zones of ischemic tissue, which is particularly important in
vascular risk factors,10–12 severe limb ischemia poses a substan- multilevel disease, and facilitates homing to ischemic tissue
tial burden on patients, healthcare providers, and resources; with preserved nutrient blood supply, leading to improved
hence, new treatment modalities are urgently needed. local cell survival,17,18 and to the extrapolation of clinical data,
Cell-based regenerative therapies aiming at enhanced as well, suggesting the benefit of intracoronary injection of
neovascularization and improved limb perfusion have been BMMNC in myocardial infarction.19,20 We chose to admin-
proposed as novel treatment strategies. In 2002, a small first- ister BMMNC obtained from 100 mL of BM, which can be
in-man clinical trial reported safety and promising effects obtained under local anesthesia in an outpatient setting with
of autologous bone marrow (BM)–derived cell therapy in a low risk of complications. We adopted a repetitive infusion
patients with critical limb ischemia.13 Since then, several stud- scheme assuming that repeated administration would enhance
ies have suggested benefit of BM-derived cell therapy for cell retainment based on observations that only limited num-
advanced PAD. However, studies were small, lacked appro- bers of cells are retained in injured tissue after injection.
priate controls, often did not consider clinically relevant out-
comes, and thus did not provide definite proof on clinical
Methods
effectiveness.14,15 Our recent meta-analysis on 12 randomized
Detailed methods can be found in the online-only Data Supplement.
clinical trials (RCTs) that studied BM-derived cell therapy in
a total of 510 patients with critical limb ischemia underlined
the promising potential of this therapy but also showed diver- Trial Design and Study Population
gent results between placebo-controlled and non–placebo- In this single-center, double-blind, placebo-controlled RCT, we inves-
tigated the clinical effects of repetitive infusion of BMMNC into the
controlled RCTs, stressing the need for a large, well-designed,
common femoral artery in 160 patients with severe, nonrevasculariz-
placebo-controlled RCT with clinically relevant outcomes.15 able PAD included from September 2006 through June 2012 (clini-
We designed the Rejuvenating Endothelial Progenitor caltrials.gov NCT00371371; Figure 1). Study design and detailed
Cells via Transcutaneous Intra-arterial Supplementation inclusion and exclusion criteria are described in the Methods in the
(JUVENTAS) trial, a double-blind, placebo-controlled RCT online-only Data Supplement and have been reported previously.16
The institutional review board of the University Medical Center
(NCT00371371) to investigate whether repetitive intra-arte-
Utrecht approved the study protocol, the study was conducted accord-
rial infusion of BM mononuclear cells (BMMNCs) reduces ing to the Declaration of Helsinki, and all patients provided written
amputation rates in a large cohort of patients with severe, non- informed consent before the study interventions.
revascularizable limb ischemia.16 Patients were randomly assigned by means of computerized block
JUVENTAS was developed and initiated in 2006, and its randomization with variable block sizes to receive either 3 repetitive
intra-arterial infusions of BMMNC or placebo (3-week intervals) into
Downloaded from http://ahajournals.org by on December 27, 2018

design was based on the available literature at that time. The


the common femoral artery of the affected limb. BM aspirates (100
rationale has been published elsewhere.16 In short, we chose mL) were obtained from the right iliac crest in all patients. BMMNCs
the intra-arterial administration route based on preclinical data were isolated by density gradient centrifugation (Lymphoprep, Axis-
suggesting that this route allows the migration to different Shield Inc, Oslo, Norway). For the placebo group, a placebo was

Figure 1. Patient recruitment and trial flow showing


the patient flow during the screening and trial
phase. No patients were lost to follow-up. ABI
indicates ankle-brachial index; and BMMNC, bone
marrow mononuclear cell.
Teraa et al   Bone Marrow Mononuclear Cells in No-Option Limb Ischemia   853

Table 1.  Demographic and Clinical Characteristics


Characteristic BMMNC (n=81) Placebo (n=79)
Age, y 69 (57–76) 65 (55–74)
Male sex, n (%) 57 (70) 51 (65)
History of cardiovascular disease, n (%)
 Myocardial infarction or chest pain 33 (41) 33 (42)
 TIA or stroke 10 (12) 13 (17)
 Dialysis dependent renal disease 2 (3) 3 (4)
 Angioplasty contralateral leg 18 (22) 19 (24)
 Bypass contralateral leg 11 (14) 16 (20)
 Major amputation contralateral leg 5 (6) 6 (8)
Body mass index, kg/m2 26.2±4.1 26.6±5.1
Smoking, n (%)
 Currently 18 (23) 24 (31)
 Past 47 (59) 46 (59)
Diabetes mellitus, n (%) 29 (36) 31 (39)
 Nonfasting glucose, mmol/L 5.9 (5.2–7.1) 5.8 (5.1–6.9)
Hypertension, n (%)* 70 (86) 72 (91)
 Systolic blood pressure, mm  Hg 130±19 132±21
 Diastolic blood pressure, mm  Hg 73±10 72±10
Hyperlipidemia, n (%)† 74 (91) 71 (90)
 Total cholesterol, mmol/L 4.3±1.1 4.2±1.1
 LDL-cholesterol, mmol/L 2.4±0.9 2.3±1.0
 HDL-cholesterol, mmol/L 1.17 (0.88–1.47) 1.11 (0.84–1.50)
 Triglycerides, mmol/L 1.4 (1.0–2.0) 1.5 (0.9–1.9)
Renal insufficiency, n (%)‡ 20 (25) 16 (20)
 Creatinine, μmol/L 92 (74–122) 86 (74–110)
Cardioprotective drug use, n (%)
Downloaded from http://ahajournals.org by on December 27, 2018

 Antiplatelet therapy 55 (68) 57 (72)


 Coumadines 32 (39) 29 (37)
 Statins 67 (83) 66 (84)
 ACE inhibitors 31 (38) 31 (39)
Disease characteristics treated leg
 Right/ left limb treated, n (%) 47/34 (58/42) 45/34 (57/43)
 Previous bypass ipsilateral, n (%) 37 (46) 43 (54)
 Previous angioplasty ipsilateral, n (%) 48 (59) 47 (60)
 Aorta to popliteal artery patent, n (%) 34 (42) 32 (41)
 <2 patent crural vessels 67 (83) 70 (89)
 Rutherford stage, n (%)
  3 4 (5) 4 (5)
  4 26 (32) 25 (32)
  5 46 (57) 46 (58)
  6 5 (6) 4 (5)
 Ulcer area, cm2 2.0 (1.0–4.5) 1.7 (1.0–4.3)
 Pain-free walking distance, m 50 (20–105) 50 (20–130)
 Unreliable ankle-brachial index, n (%)§ 28 (35) 28 (35)
 Baseline ankle-brachial index║ 0.50 (0.36–0.66) 0.50 (0.39–0.77)
 Baseline tco2, mm Hg 35±22 36±22
Use of cardioprotective drugs was defined at time of inclusion. ACE indicates angiotensin-converting enzyme; BMMNC,
bone marrow mononuclear cell; HDL, high-density lipoprotein; LDL, low-density lipoprotein; and TIA, transient ischemic attack.
*Hypertension was defined as having systolic blood pressure ≥140 mm Hg, or diastolic blood pressure ≥90 mm Hg, or
antihypertensive drug use.
†Hyperlipidemia was defined as having blood levels of total cholesterol ≥6.50 mmol/L, triglycerides ≥2.3 mmol/L, or HDL-
cholesterol ≤1.0 mmol/L, or lipid-lowering drug use.
‡Renal insufficiency was defined as having creatinine blood levels >120 μmol/L.
§Unreliable ankle-brachial index was defined as incompressible arteries or an ankle-brachial index >0.6 not in proportion to
the Fontaine classification.
║Only patients with a reliable ankle-brachial index were included in the analysis (n=53 in the BMMNC and n=51 in the
placebo group).
854  Circulation  March 10, 2015

prepared by using autologous peripheral blood erythrocytes to match 4.4),29 with adjustment of the point and interval estimates for cumula-
the color of the BMMNC product. Of both products, one-third was tive testing. Additionally, the risks for major amputation and combined
prepared for direct infusion, and two-thirds were cryopreserved and major amputation and death were analyzed by the Kaplan-Meier method
stored for subsequent infusions. For cryopreservation of the BMMNC with the use of the log-rank test. Primary analyses were performed at 6
product, 10% dimethyl sulfoxide was added. Because dimethyl sulf- months of follow-up. Major amputation, mortality, and combined major
oxide releases a specific odor at the time of infusion, the same amount amputation and death were also analyzed beyond 6 months. Data for
of dimethyl sulfoxide was added to the cryopreserved placebo prod- patients were censored at the date of death, last visit, or last known to
uct to guarantee blinding of the trial staff. Syringes without informa- be alive. Because of the lack of power, no subgroup analyses were per-
tion about the product were provided to the clinical staff at the time formed. With the exception of the sequential analysis, no adjustments
of infusion. At infusion, the product was slowly administered into the for multiple statistical testing were made.30
common femoral artery of the affected limb by hand injection. Furthermore, the results with respect to major amputation were
Numbers of white blood cells, CD34+ hematopoietic progenitor added to our recently published meta-analysis, with an updated lit-
cells, colony-forming unit granulocytes and monocytes, and burst- erature review, and reanalyzed in Review Manager (The Cochrane
forming unit-erythroid capacity of the BM product were assessed. Collaboration, version 5.3) with the use of a random-effects model
according to the methods published previously.15 Other statistical
analyses were performed by using SPSS for Windows version 20.0
Surveillance Protocol and Outcome Assessment (SPSS Inc., Chicago, IL).
Primary outcome was major amputation, defined as amputation
through or above the ankle joint, within 6 months after randomiza-
tion. The combined safety outcome was all-cause mortality, occur- Role of Funding Sources
rence of malignancy, or hospitalization due to infection. Secondary The sponsors of the study had no role in the study design, data collec-
outcomes were the combined occurrence of major amputation or tion, analysis, interpretation, or writing of the report. All authors had
death, minor amputations, changes in clinical status, ulcer size, rest full access to all data in the study and had final responsibility for the
pain, pain-free walking distance, ankle-brachial index (ABI), trans- decision to submit for publication.
cutaneous oxygen pressure measurements (tco2; Radiometer Medical
ApS, Copenhagen, Denmark), and health-related QoL (Short Form 36 Results
[SF-36] and EuroQoL 5D [EQ-5D]). SF-36 scores were converted to
a norm-based score and the EQ-5D was converted to the Preference- Enrollment and Baseline Patient Characteristics
Based EuroQoL Tariff as reported previously.7,21,22 To allow compari- One hundred sixty patients with severe, nonrevasculariz-
sons with recently published clinical trials, we included previously
published composite outcomes for success and failure of cell-based able PAD were randomly assigned to repetitive intra-arterial
therapy.23,24 Clinical evaluation by the same investigator, treadmill BMMNC (n=81) or placebo (n=79) infusions. A detailed
exercise testing, and tco2 measurement were performed at baseline overview of the trial flow is depicted in Figure 1. The 2 trial
and at 2 and 6 months follow-up. arms were well comparable with respect to baseline charac-
The assessment of renal and liver function–related laboratory mea- teristics and concomitant pharmacological therapy during
Downloaded from http://ahajournals.org by on December 27, 2018

surements was performed at inclusion and before and after each intra-
arterial infusion. During conduct of the trial, all adverse events (AEs) the study (Table 1). The majority of patients had tissue loss
and serious adverse events (SAEs) were recorded and processed (Rutherford stage 5 or 6; 63% in both groups). Until primary
according to national guidelines. trial follow-up at 6 months, no patients were lost to follow-up.
Major amputation, mortality, and combined major amputation
Interim Analyses and Data Monitoring and death were also analyzed beyond the prespecified primary
An independent Data and Safety Monitoring Board evaluated the trial follow-up at 6 months; because this was not a prespecified
results of sequential interim analyses. Every 6 months, or after every outcome, follow-up beyond 6 months was not available for all
fourth major amputation, or occurrence of a safety outcome, unblinded patients (n=127; 79% of the total study population, median
analyses were performed on the cumulative database. Based on the
follow-up of 9 [6–20] months, not different between groups).
results of these analyses, the Data and Safety Monitoring Board
informed the trial’s executive committee on whether the data provided
sufficient evidence for either efficiency or harm of the intervention Intervention
with respect to the primary outcome and on safety and advised the Beside a slightly higher number of CD34+ hematopoietic pro-
executive committee whether to (dis)continue the trial (see Methods genitor cells in the BMMNC group, the characteristics of the
in the online-only Data Supplement). During the conduct of the trial,
BM aspirate did not differ between the 2 groups (Tables 2 and 3).
no recommendations were made to discontinue the trial.

Table 2.  Characteristics of the Bone Marrow Aspirate and


Sample-Size Calculation and Statistical Analyses Cell Product
Estimation of the sample size of the JUVENTAS trial was based on
the best available evidence at trial initiation in 2006, which reported Characteristics BM Aspirate BMMNC (n=81) Placebo (n=79)
a 6-month risk of major amputation in patients with nonrevasculariz-
Volume of BM, mL 100 (100–106) 100 (100–106)
able chronic critical limb ischemia of 42%25 and a 50% risk reduc-
tion of major amputation by BMMNC infusion.26,27 Based on these Total amount of BMMNC, ×10 6
657 (422–965) 574 (407–801)
assumptions, it was estimated that with a 2-sided α of 0.05 and a Total amount of CD34+ HPC, ×106 11.4 (7.4–15.8) 9.2 (4.8–13.9)
power of 80%, ≈110 to 160 patients should be included in the trial. For
a sequential design, no fixed sample size estimate can be provided.28 CFU-GM, per ×105 cells plated 264 (176–396) 245 (174–354)
Continuous variables are expressed as means±standard deviation or as BFU-e, per ×105 cells plated 156 (110–213) 148 (106–205)
medians and interquartile ranges for nonnormal data. Depending on data Erythrocytes, ×10 /mL*
9
0.08 (0.02–0.14) NA
characteristics, group differences for continuous variables were tested
with the independent t test or the Mann-Whitney U test. Dichotomous BFU-e indicates burst-forming unit-erythroid; BM, bone marrow; BMMNC,
variables were analyzed with the Fisher exact test. Analyses were per- bone marrow mononuclear cell; CFU-GM, colony forming unit granulocytes and
formed in accordance with the intention-to-treat principle. Sequential monocytes; HPC, hematopoietic progenitor cell; and NA, not applicable for the
analyses28 of the primary outcome, ie, major amputation, and of the placebo group.
safety outcome were performed by using the PEST program (version *Number of erythrocytes per milliliter after density centrifugation.
Teraa et al   Bone Marrow Mononuclear Cells in No-Option Limb Ischemia   855

Table 3.  Characteristics of the Cell Product per Infusion


Total Number of
First Infusion Second Infusion Third Infusion Infused Cells
Cell number per infusion
 BMMNC, ×106 199 (133–295) 144 (93–214)* 144 (87–217)* 500 (313–717)
 CD34 HPC, ×10
+ 6
3.6 (2.3–4.9) 2.5 (1.4–4.0)* 2.4 (1.5–3.6)* 8.4 (5.8–11.9)
Colony forming capacity per infusion*
 CFU-GM, per ×105 cells plated 264 (176–396) 151 (95–239)* 147 (92–217)*
 BFU-e, per ×10 cells plated
5
156 (110–213) 123 (92–170)* 129 (92–170)*
Data are only applicable to patients randomly assigned to the BMMNC group. BFU-e indicates burst-forming unit-
erythroid; BMMNC, bone marrow mononuclear cell; CFU-GM, colony forming unit granulocytes and monocytes; and HPC,
hematopoietic progenitor cell.
*P<0.001 compared with first infusion.

Additionally, the colony-forming capacity of patients’ BM was Hemodynamic Measurements and Ulcer Healing
compared with that of healthy controls (n=32; median age, 32 Baseline measurements were comparable for ABI, tco2, and ulcer
[21–36]), which showed no significant differences for both the area (Table 1). For the analyses of ABI, only patients with a reli-
colony-forming unit granulocytes and monocytes and burst- able index at baseline were included (n=53 BMMNC group; n=51
forming unit-erythroid (colony-forming unit granulocytes and placebo group). ABI increased in both groups during follow-up
monocytes 250 [174–355] versus 282 [197–394], P=0.23;
burst-forming unit-erythroid 149 [108–204] versus 174 [139–
Table 4.  Major Amputation, Death, and Composite Outcomes
242], P=0.07, for patients and healthy controls, respectively).
at 2 and 6 Months and Beyond
A total of 15 patients, 6 in the BMMNC and 9 in the pla-
cebo group, did not complete the scheduled 3 infusions Outcome BMMNC Control RR (95% CI) P Value
owing to the occurrence of a major amputation or mortality Major amputation, n (%)
before the completion of the scheduled infusions (Figure 1).  At 2 mo 6/81 (7) 6/79 (8) 0.98 (0.33–2.90) 1.0
In patients who completed 3 BMMNC infusions, total num-  At 6 mo 15/81 (19) 10/79 (13) 1.46 (0.62–3.42) 0.31
bers of 500×106 (313–717) BMMNC and 8.4×106 (5.8–11.9)  Overall* 21/81 (26) 19/79 (24) 1.08 (0.58–2.02) 0.81
Downloaded from http://ahajournals.org by on December 27, 2018

CD34+ hematopoietic progenitor cells were infused.


Death, n (%)

Major Amputation, All-Cause Mortality,  At 2 mo 1/81 (1) 2/79 (3) 0.49 (0.05–5.27) 0.62
and Composite Outcomes  At 6 mo 4/81 (5) 5/79 (6) 0.78 (0.22–2.80) 0.74
The primary outcome was not different between the 2 groups  Overall* 9/81 (11) 11/79 (14) 0.79 (0.53–3.04) 0.60
with a major amputation rate of 19% and 13% at 6 months in Major amputation
the BMMNC and control group (adjusted relative risk BMMNC or death, n (%)†
versus placebo, 1.46; 95% confidence interval [CI], 0.62–3.42;  At 6 mo 19/81 (23) 13/79 (16) 1.43 (0.76–2.69) 0.27
Table 4 and Figure 2A). The combined safety outcome was
 Overall* 28/81 (35) 26/79 (33) 1.07 (0.63–1.84) 0.80
not different between the groups (15% and 10%, respectively;
Other (at 6 mo), n (%)
relative risk, 1.46; 95% CI, 0.63–3.38). All-cause mortality at
6 months was also not different between the groups with 5%  Minor amputation 9/81 (11) 10/79 (13) 0.88 (0.38–2.05) 0.95
versus 6% (relative risk, 0.78; 95% CI, 0.22–2.80), nor was the  Composite therapy 59/81 (73) 63/78 (81) 0.90 (0.76–1.07) 0.26
combined risk for major amputation and death, with a 6-month success Powell et al23‡
risk of 23% in the BMMNC and 16% in the placebo group (rel-  Composite therapy 35/81 (43) 34/77 (44) 0.98 (0.69–1.39) 1.0
ative risk, 1.43; 95% CI, 0.76–2.69; Table 4 and Figure 2B). In success Iafrati et al30a§
the BMMNC group, the number of infused cells did not relate BMMNC indicates bone marrow mononuclear cell; CI, confidence interval;
with any of the outcome measures (data not shown). When fol- and RR, relative risk.
low-up beyond 6 months was considered, no differences with *Overall denotes follow-up also including time beyond 6 months if available;
median follow-up was 9 months, with an interquartile range of 6 to 20 months
respect to major amputation, mortality, and combined major
(the overall results indicate hazard ratios calculated by the Kaplan–Meier
amputation and death were observed between groups (Table 4). method).
†Amputation-free survival defined as being alive without major amputation
Meta-Analysis may be directly derived as (100% – percentage of major amputation or death).
When adding the results of the JUVENTAS trial and newly ‡Composite end point as defined by Powell et al.23 Success is defined as
published RCTs31–34 to our recently published meta-analysis,15 being alive, without major amputation of the index leg, and no doubling in
a subtle, but significant difference was observed for major wound size or de novo gangrene.
§Composite end point as defined by Iafrati et al.30a Success is defined
amputation (Figure 3A) in favor of the cell therapy–treated as the subject A, is alive; B, did not undergo major amputation of the index
group. However, when only the blinded placebo-controlled limb; C, did not worsen in Rutherford classification or visual analogue scale
trials were included, no significant difference between the >30 mm; and D, improved in either Rutherford classification or visual
cell- and placebo-treated groups was observed (Figure 3B). analogue scale >30 mm.
856  Circulation  March 10, 2015

Figure 2. A, Kaplan–Meier plot of cumulative probability of major amputation according to trial arm. The dashed and continuous lines
represent the BMMNC and placebo group, respectively. No significant differences existed between the groups (P=0.34). B, Kaplan–Meier
plot of cumulative probability of major amputation and death according to trial arm. The dashed and continuous lines represent the
BMMNC and placebo group, respectively. No significant differences existed between the groups (P=0.31). BMMNC indicates bone
marrow mononuclear cell.

(0.11; 95% CI, 0.06–0.15 and 0.08; 95% CI, 0.02–0.13 for the CI, 0.12–0.31 and 0.19; 95% CI, 0.08–0.31), without differ-
BMMNC and placebo group; Table 5), without difference in ence between the 2 groups (Table II in the online-only Data
ΔABI between the groups (0.03; 95% CI, –0.04 to 0.10). tco2 also Supplement). The visual analog scale pain score decreased in both
improved in the BMMNC (10.4 mm Hg; 95% CI, 4.2–16.6) and groups at 6 months, with the most pronounced change in visual
Downloaded from http://ahajournals.org by on December 27, 2018

placebo group (6.7 mm Hg; 95% CI, 1.3–12.1), without differ- analog scale score at 6 months in the placebo group (–2.4; 95%
ence between the groups (3.7; 95% CI, –4.4 to 11.9). In patients CI, –3.2 to –1.5; Table II in the online-only Data Supplement).
with tissue loss, the ulcer area tended to decrease in both groups
without a difference between the groups (difference in Δulcer Safety
area at 6 months –0.1; 95% CI, –6.1 to 5.9). Complete ulcer heal- During follow-up, no effects on liver or kidney function were
ing at 6 months was observed in 37% of the BMMNC in com- observed in either group. The total number of SAEs and AEs
parison with 28% of the placebo group (Table I in the online-only observed during the study was 213, with 108 (S)AEs and
Data Supplement). Improvement of Rutherford stage, defined as 105 (S)AEs in the BMMNC and placebo groups (difference
at least a decrease of 1 Rutherford stage, was observed in 36% of of mean number of [S]AEs, 0.0; 95% CI, –0.31 to 0.31). The
the BMMNC and 33% in the placebo group. number of patients experiencing a SAE in the BMMNC and
placebo groups was 38 and 34 (difference of mean number of
Quality of Life and Rest Pain SAE, 0.0; 95% CI, –0.30 to 0.32). All SAEs were identified
Throughout the study, the proportion of QoL forms completed as not related to the study interventions, with the exception of
to such an extent that total scores could be obtained ranged one, an inguinal hematoma as a result of intra-arterial infusion.
from 89% and 97% at inclusion to 80% and 85% at 6 months
follow-up (of those alive without amputation) for the SF-36 Discussion
and EQ-5D groups, respectively, with no difference between The JUVENTAS trial is the largest double-blind, placebo-
the groups at any time point. Baseline QoL scores were low on controlled RCT to study the effects of BM-derived cell admin-
the physical components of the SF-36, ie, physical function- istration in patients with severe PAD to date. The study shows
ing, role physical, bodily pain, and general health, reflected that repetitive intra-arterial autologous BMMNC administra-
by the low Physical Composite Score of 29.5±7.1 in the tion was not effective in reducing the incidence of the primary
BMMNC and 30.7±7.3 in the placebo group. During follow- outcome, ie, major amputation at 6 months, and even leaves
up, QoL scores improved over a wide range of SF-36 com- room for a potential negative effect of the intervention. During
ponents, the physical components in particular, resulting in a the 6-month follow-up period, improvement of secondary
significant increase of the Physical Composite Score in both outcomes, including QoL, ABI, tco2, and ulcer healing, was
the BMMNC, and in the placebo group, as well. No differ- observed in both treatment groups without any significant dif-
ence was observed in ΔPhysical Composite Score at 6 months ferences between the groups.
between the 2 groups (–0.1; 95% CI, –3.3 to 3.1; Table 5). Strategies for BM-derived cell therapy in patients with severe
The EQ-5D scores 6 months after the first infusion improved limb ischemia differ between studies in many aspects, includ-
significantly in the BMMNC and placebo groups (0.21; 95% ing administration route, cell source and cell-subtype, cell dose,
Teraa et al   Bone Marrow Mononuclear Cells in No-Option Limb Ischemia   857
Downloaded from http://ahajournals.org by on December 27, 2018

Figure 3. A, Meta-analysis of the effect of cell therapy on major amputation in RCTs in CLI. The meta-analysis shows a subtle significant
(P=0.02) beneficial effect on major amputation of cell therapy in comparison with placebo (RR, 0.66; 95% CI, 0.47–0.93). Within Review
Manager, it is not possible to manually adapt the width of the CI to correct for (sequential) interim analyses. This has led to slightly
narrower 95% CI for the RR for major amputation of our study in the meta-analysis in comparison with the results as described in
the text. B, Meta-analysis of the effect of cell therapy on major amputation in placebo-controlled RCTs in CLI. Only blinded placebo-
controlled RCTs with a separate control group were analyzed (ie, trials that use standard of care or the contralateral limb as a control or
lack any form of blinding were excluded). No effect of cell therapy in comparison with the placebo arm is observed on major amputation
(RR, 0.95; 95% CI 0.64–1.39; P=0.78). Within Review Manager, it is not possible to manually adapt the width of the CI to correct for
(sequential) interim analyses. This has led to slightly narrower 95% CI for the RR for major amputation of our study in the meta-analysis in
comparison with the results as described in the text. CI indicates confidence interval; CLI, critical limb ischemia; M-H, Mantel-Haenszel
method; RCT, randomized clinical trial; and RR, relative risk.

and single or repetitive treatments. These variables may influ- BM product and clinical improvement. The results of the
ence study results. For the JUVENTAS trial, initiated in 2006, Intraarterial Progenitor Cell Transplantation of Bone Marrow
we chose a strategy of infusing BMMNCs obtained from a fixed Mononuclear Cells for Induction of Neovascularization
amount of 100 mL of BM into the common femoral artery on in Patients With Peripheral Arterial Occlusive Disease
3 separate occasions. It is unlikely that the lack of benefit in (PROVASA) trial suggested benefit of repeated intra-arterial
our study is due to differences in cell administration or dose. BMMNC administration.38 This could not be confirmed in our
Previously, potential beneficial effects have been reported by study. It has also been suggested that assessing potential clini-
studies that used intra-arterial administration and by studies cal benefits of cell therapy in patients with severe limb ischemia
using intramuscular administration, as well. Studies that directly requires longer follow-up.38 However, also with longer follow-
compared the intra-arterial versus intramuscular route did not up (median, 9 months), we observed no differences between
show superiority of either route, but lacked the proper design groups in any of the outcome measures.
to draw definite conclusions.35,36 Furthermore, beneficial results Cell-manufacturing procedures have been reported to
have been reported after the administration of highly variable influence BMMNC potency.39–41 We used Lymphoprep for
doses of BMMNC with volumes of aspirated BM ranging from BMMNC isolation and included heparin in our isolation proto-
50 to 1000 mL, from which varying amounts of BMMNC and col, which is similar to previous studies that showed potential
CD34+ cells were retrieved.15 Although some studies have sug- clinical effects.13,38,42 Heparin has been reported to negatively
gested a dose-response relation between numbers of adminis- affect cellular homing by impairment of the SDF-1α/CXCR4
tered CD34+ cells or BMMNCs, this has not been a consistent axis.40 We cannot exclude that the use of heparin in our isola-
finding.31,37,38 In the JUVENTAS trial, no relation was found tion procedure could have reduced the potency of our product.
between cell numbers or cell characteristics of the administered Contamination of erythrocytes in the BMMNC product may
858  Circulation  March 10, 2015

Table 5.  Hemodynamic Measurements, Ulcer Healing, and Quality of Life


Difference Between
Outcome BMMNC Control N the Groups
ABI*
 Δ ABI 2 mo 0.05 (0.00 to 0.10) 0.05 (0.00 to 0.10) 40/37    0.00 (–0.07 to 0.06)
 Δ ABI 6 mo 0.11 (0.06 to 0.15) 0.08 (0.02 to 0.13) 40/37    0.03 (–0.04 to 0.10)
tco2, mm Hg*
 Δ tco2 2 mo 6.8 (1.6 to 12.0) 2.4 (–2.0 to 6.8) 62/62    4.4 (–2.4 to 11.2)
 Δ tco2 6 mo 10.4 (4.2 to 16.6) 6.7 (1.3 to 12.1) 57/56    3.7 (–4.4 to 11.9)
Ulcer area, cm2 *
 Δ ulcer area 2 mo –0.8 (–1.1 to 2.8) –0.7 (–1.7 to 0.3) 41/39 –0.1 (–1.1 to 0.8)
 Δ ulcer area 6 mo –0.5 (–4.1 to 5.1) –0.4 (–3.8 to 4.6) 34/37 –0.1 (–6.1 to 5.9)
QoL, SF-36*
 Δ PCS 2 mo 4.0 (2.1 to 6.0) 4.6 (2.8 to 6.5) 53/48 –0.6 (–3.3 to 2.1)
 Δ MCS 2 mo –0.5 (–3.8 to 2.9) 4.8 (1.8 to 7.9) 53/48 –5.3 (–9.8 to 0.8)
 Δ PCS 6 mo 6.3 (4.1 to 8.4) 6.4 (3.9 to 8.8) 46/49 –0.1 (–3.3 to 3.1)
 Δ MCS 6 mo 3.9 (0.0 to 7.8) 1.7 (–2.6 to 5.9) 46/49   2.3 (–3.4 to 7.9)
Values are means and 95% CI. ABI indicates ankle-brachial index; BMMNC, bone marrow mononuclear
cell; CI, confidence interval; MCS, Mental Health Composite Score; PCS, Physical Composite Score; and QoL,
quality of life.
*Patients who underwent major amputation, were deceased, or had missing values at a specific follow-up
occasion were not included in the analysis at that specific time point. With respect to ABI patients with unreliable
ABI at inclusion (incompressible or not in proportion to the clinical status) were not included in the analyses.

also impair its effectiveness.39 Assmus et al showed substantial in both BMMNC- and placebo-treated patients was observed
impairment of the BMMNC product when >0.2×109 erythro- emphasizes the essential role of accurate blinding, which,
cytes/mL are present. Erythrocytes in our product were far together with the large population size, is a major strength of
below this threshold (0.08×109/mL±0.06×109/mL) and there- our study. Similar observations have previously been reported
Downloaded from http://ahajournals.org by on December 27, 2018

fore unlikely to have influenced our product. for cell therapy trials for heart disease. Several recent well-
The JUVENTAS trial included mainly elderly (median age, designed and larger RCTs that studied BMMNC administration
67 years) white patients with a high systemic atherosclerotic in both acute myocardial infarction, and in chronic heart failure,
burden and prevalence of cardiovascular disease, a typical as well, failed to prove the beneficial effects of BMMNC admin-
Western population with advanced PAD. In contrast, several istration on cardiac function in comparison with placebo inter-
initial positive studies were conducted in Asian populations, vention.45,46 A recent meta-analysis showed that the observed
often including relatively young patients with thromboangi- improvement in left ventricular ejection fraction observed after
itis obliterans.14 Aging and comorbidities have been shown to BMMNC therapy in myocardial infarction disappeared when
induce functional impairment of BMMNCs which may limit only sham-controlled studies were analyzed.47
the therapeutic potential of autologous BM-derived cell therapy. In conclusion, this double-blind, placebo-controlled RCT
The lack of benefit in our patients could be partly due to BM of autologous BM-derived cell therapy in patients with severe,
cell dysfunction in patients with high atherosclerotic burden.43 nonrevascularizable PAD shows no effect of repetitive intra-
The major amputation rate in our study was lower than arterial infusion of autologous BMMNC on prespecified out-
expected based on the available literature at the time of the ini- comes. The lack of benefit of autologous BMMNCs in our
tiation of the trial.25 More recent data suggest that amputation study does not exclude the potential benefit of other cell
rates in no-option patients with advanced PAD have decreased sources or subpopulations, different administration locations
over the past decades, in line with the observation in our study.44 and routes, or in patients with milder disease. Further study is
The low number of major amputations decreased the statistical warranted to investigate whether cell therapy strategies with
power of the study with respect to major amputation, but the selected cell populations, enhanced BM cell function, or dif-
general lack of benefit on secondary outcomes in comparison ferent modes of administration can provide therapeutic ben-
with placebo, support a lack of benefit of autologous BMMNCs efit in patients with advanced PAD. The general improvement
as applied in our protocol. Moreover, our observations are in observed in clinical outcomes in both groups during follow-
line with our recently published meta-analysis on cell therapy in up stresses the need for future RCTs to implement a rigorous
critical limb ischemia.15 The inclusion of the JUVENTAS trial double-blinded, placebo-controlled design.
(and other new trial) results in the meta-analysis of all RCTs
shows a minor significant beneficial effect on major amputa-
tion, whereas analysis of only the blinded placebo-controlled
Acknowledgments
We thank Dr G.J. de Borst, Dr R.J. Toorop, Dr C.E.V.M. Hazenberg,
RCTs shows a clear lack of benefit. Dr J.A. van Herwaarden, Dr P. Berger, Dr E. Waasdorp, and Dr A.Kh.
The fact that, in our trial, where rigorous blinding of par- Jahrome for patient recruitment, screening, and general trial sup-
ticipants and investigators was applied, a general improvement port. Dr L. te Boome and Dr P.E. Westerweel are acknowledged for
Teraa et al   Bone Marrow Mononuclear Cells in No-Option Limb Ischemia   859

performing part of the bone marrow aspiration procedures. The Cell Peripheral Arterial Disease Committee of the Brazilian Society of Cardiology/
Therapy Facility of the University Medical Center Utrecht, in particular Funcor. Prevalence and risk factors associated with peripheral arterial disease
K. Westinga, is thanked for processing of the trial products. The nurs- in the Hearts of Brazil Project. Arq Bras Cardiol. 2008;91:370–382.
ing staff of the vascular surgery outpatient clinic and surgical wards of 12. Eraso LH, Fukaya E, Mohler ER 3rd, Xie D, Sha D, Berger JS. Peripheral
the University Medical Center Utrecht are thanked for their medical- arterial disease, prevalence and cumulative risk factor profile analysis. Eur
site support. Furthermore, all referring vascular surgeons, in particu- J Prev Cardiol. 2012;21:704–711. doi: 10.1177/2047487312452968.
lar, from the Sint Antonius Hospital Nieuwegein (Dr J.P.P.M de Vries, 13. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki
Dr H.D.W.M van de Pavoordt, and Dr R.H.W. van de Mortel) and the H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, Shimada K, Iwasaka
Meander Medical Center Amersfoort (Dr C.H.P. Arts, Dr M.C. Loubert, T, Imaizumi T; Therapeutic Angiogenesis using Cell Transplantation
Dr A.J.C. Mackaay, and Dr R. Voorhoeve) are kindly acknowledged for (TACT) Study Investigators. Therapeutic angiogenesis for patients with
their support. Authors’ contributions: Drs Sprengers, Verhaar, van der limb ischaemia by autologous transplantation of bone-marrow cells: a
Graaf, and Moll conceived and designed the study, with advise from pilot study and a randomised controlled trial. Lancet. 2002;360:427–435.
Drs Schutgens, Slaper-Cortenbach, Doevendans, and Mali. Dr Teraa doi: 10.1016/S0140-6736(02)09670-8.
and Sprengers enrolled the patients. Drs Teraa, Sprengers, Algra, van 14. Fadini GP, Agostini C, Avogaro A. Autologous stem cell therapy

der Tweel, van der Graaf, Schutgens, Slaper-Cortenbach, Moll, and for peripheral arterial disease meta-analysis and systematic review
of the literature. Atherosclerosis. 2010;209:10–17. doi: 10.1016/j.
Verhaar were involved in data acquisition, analysis, and interpretation.
atherosclerosis.2009.08.033.
Drs Teraa and Verhaar wrote the first draft of the report. All authors
15. Teraa M, Sprengers RW, van der Graaf Y, Peters CE, Moll FL, Verhaar
edited and approved the report for final submission.
MC. Autologous bone marrow-derived cell therapy in patients with critical
limb ischemia: a meta-analysis of randomized controlled clinical trials.
Sources of Funding Ann Surg. 2013;258:922–929. doi: 10.1097/SLA.0b013e3182854cf1.
The reported work was supported by the “Stichting Vrienden UMC 16. Sprengers RW, Moll FL, Teraa M, Verhaar MC; JUVENTAS Study

Utrecht” on behalf of the Dirkzwager-Assink foundation (The Group. Rationale and design of the JUVENTAS trial for repeated intra-
Netherlands, grant CS 06.007), the Dutch Heart Foundation (grant arterial infusion of autologous bone marrow-derived mononuclear cells in
2008B094), The Netherlands Organization for Scientific Research patients with critical limb ischemia. J Vasc Surg. 2010;51:1564–1568. doi:
10.1016/j.jvs.2010.02.020.
(ZonMw-TAS grant 116001026), and foundation “Stichting De Drie
17. Sprengers RW, Lips DJ, Moll FL, Verhaar MC. Progenitor cell therapy in
Lichten” (The Netherlands, grant 10/06). Dr Verhaar is supported by
patients with critical limb ischemia without surgical options. Ann Surg.
the Netherlands Organization for Scientific Research (NWO; The
2008;247:411–420. doi: 10.1097/SLA.0b013e318153fdcb.
Netherlands, Vidi grant 016.096.359).
18. Yoshida M, Horimoto H, Mieno S, Nomura Y, Okawa H, Nakahara K,
Sasaki S. Intra-arterial bone marrow cell transplantation induces angio-
Disclosures genesis in rat hindlimb ischemia. Eur Surg Res. 2003;35:86–91. doi:
None. 69401.
19. Abdel-Latif A, Bolli R, Tleyjeh IM, Montori VM, Perin EC, Hornung
CA, Zuba-Surma EK, Al-Mallah M, Dawn B. Adult bone marrow-derived
References cells for cardiac repair: a systematic review and meta-analysis. Arch Intern
Downloaded from http://ahajournals.org by on December 27, 2018

1. Adam DJ, Beard JD, Cleveland T, Bell J, Bradbury AW, Forbes JF, Fowkes Med. 2007;167:989–997. doi: 10.1001/archinte.167.10.989.
FG, Gillepsie I, Ruckley CV, Raab G, Storkey H; BASIL trial participants. 20. Lipinski MJ, Biondi-Zoccai GG, Abbate A, Khianey R, Sheiban I,

Bypass versus angioplasty in severe ischaemia of the leg (BASIL): mul- Bartunek J, Vanderheyden M, Kim HS, Kang HJ, Strauer BE, Vetrovec
ticentre, randomised controlled trial. Lancet. 2005;366:1925–1934. doi: GW. Impact of intracoronary cell therapy on left ventricular function
10.1016/S0140-6736(05)67704-5. in the setting of acute myocardial infarction: a collaborative systematic
2. Stoyioglou A, Jaff MR. Medical treatment of peripheral arterial disease: review and meta-analysis of controlled clinical trials. J Am Coll Cardiol.
a comprehensive review. J Vasc Interv Radiol. 2004;15:1197–1207. doi: 2007;50:1761–1767. doi: 10.1016/j.jacc.2007.07.041.
10.1097/01.RVI.0000137978.15352.C6. 21. Ware JE, Kosinski M. SF-36 Physical and Mental Health Summary Scales:
3. Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, Fowkes FG; A Manual for Users of Version 1. Lincoln, RI: QualityMetric Incorporated;
TASC II Working Group. Inter-society consensus for the management of 2001.
peripheral arterial disease (TASC II). J Vasc Surg. 2007;45(suppl S):S5– 22. Dolan P. Modeling valuations for EuroQol health states. Med Care.

67. doi: 10.1016/j.jvs.2006.12.037. 1997;35:1095–1108.
4. Becker F, Robert-Ebadi H, Ricco JB, Setacci C, Cao P, de Donato G, 23. Powell RJ, Marston WA, Berceli SA, Guzman R, Henry TD, Longcore
Eckstein HH, De Rango P, Diehm N, Schmidli J, Teraa M, Moll FL, Dick AT, Stern TP, Watling S, Bartel RL. Cellular therapy with Ixmyelocel-T to
F, Davies AH, Lepäntalo M, Apelqvist J. Chapter I: Definitions, epide- treat critical limb ischemia: the randomized, double-blind, placebo-con-
miology, clinical presentation and prognosis. Eur J Vasc Endovasc Surg. trolled RESTORE-CLI trial. Mol Ther. 2012;20:1280–1286. doi: 10.1038/
2011;42(suppl 2):S4–12. doi: 10.1016/S1078-5884(11)60009-9. mt.2012.52.
5. Conte MS, Geraghty PJ, Bradbury AW, Hevelone ND, Lipsitz SR, Moneta 24. Benoit E, O’Donnell TF Jr, Kitsios GD, Iafrati MD. Improved amputation-
GL, Nehler MR, Powell RJ, Sidawy AN. Suggested objective performance free survival in unreconstructable critical limb ischemia and its implica-
goals and clinical trial design for evaluating catheter-based treatment of
tions for clinical trial design and quality measurement. J Vasc Surg.
critical limb ischemia. J Vasc Surg. 2009;50:1462–1473.
2012;55:781–789. doi: 10.1016/j.jvs.2011.10.089.
6. Van Hattum ES, Tangelder MJ, Lawson JA, Moll FL, Algra A. Long-term
25. Lepäntalo M, Mätzke S. Outcome of unreconstructed chronic critical leg
risk of vascular events after peripheral bypass surgery. A cohort study.
ischaemia. Eur J Vasc Endovasc Surg. 1996;11:153–157.
Thromb Haemost. 2012;108:543–553. doi: 10.1160/TH11-12-0844.
26. Kawamura A, Horie T, Tsuda I, Ikeda A, Egawa H, Imamura E, Iida
7. Sprengers RW, Teraa M, Moll FL, de Wit GA, van der Graaf Y, Verhaar
MC. Quality of life in patients with no-option critical limb ischemia under- J, Sakata H, Tamaki T, Kukita K, Meguro J, Yonekawa M, Kasai M.
lines the need for new effective treatment. J Vasc Surg. 2010;52:843–849. Prevention of limb amputation in patients with limbs ulcers by autolo-
8. Barshes NR, Chambers JD, Cohen J, Belkin M. Cost-effectiveness in gous peripheral blood mononuclear cell implantation. Ther Apher Dial.
the contemporary management of critical limb ischemia with tissue loss. 2005;9:59–63. doi: 10.1111/j.1774-9987.2005.00218.x.
J Vasc Surg. 2012;56:1015–1024. 27. Kawamura A, Horie T, Tsuda I, Abe Y, Yamada M, Egawa H, Iida J, Sakata
9. Peters EJ, Childs MR, Wunderlich RP, Harkless LB, Armstrong DG, H, Onodera K, Tamaki T, Furui H, Kukita K, Meguro J, Yonekawa M, Tanaka
Lavery LA. Functional status of persons with diabetes-related lower- S. Clinical study of therapeutic angiogenesis by autologous peripheral blood
extremity amputations. Diabetes Care. 2001;24:1799–1804. stem cell (PBSC) transplantation in 92 patients with critically ischemic
10. Hirsch AT, Hartman L, Town RJ, Virnig BA. National health care costs limbs. J Artif Organs. 2006;9:226–233. doi: 10.1007/s10047-006-0351-2.
of peripheral arterial disease in the Medicare population. Vasc Med. 28. Whitehead J. The Design and Analysis of Sequential Clinical Trials.
2008;13:209–215. doi: 10.1177/1358863X08089277. Chichester, UK: John Wiley & Sons Ltd; 1997.
11. Makdisse M, Pereira Ada C, Brasil Dde P, Borges JL, Machado-Coelho GL, 29. MPS Research Unit: PEST 4: Operating Manual. Reading, UK: University
Krieger JE, Nascimento Neto RM, Chagas AC; Hearts of Brazil Study and of Reading; 2000.
860  Circulation  March 10, 2015

30. Schulz KF, Grimes DA. Multiplicity in randomised trials I: end- 40. Seeger FH, Rasper T, Fischer A, Muhly-Reinholz M, Hergenreider E,
points and treatments. Lancet. 2005;365:1591–1595. doi: 10.1016/ Leistner DM, Sommer K, Manavski Y, Henschler R, Chavakis E, Assmus
S0140-6736(05)66461-66466. B, Zeiher AM, Dimmeler S. Heparin disrupts the CXCR4/SDF-1 axis
30a. Iafrati MD, Hallett JW, Geils G, Pearl G, Lumsden A, Peden E, Bandyk D, and impairs the functional capacity of bone marrow-derived mononucle-
Vijayaraghava KS, Radhakrishnan R, Ascher E, Hingorani A, Roddy S. ar cells used for cardiovascular repair. Circ Res. 2012;111:854–862. doi:
Early results and lessons learned from a multicenter, randomized, double- 10.1161/CIRCRESAHA.112.265678.
blind trial of bone marrow aspirate concentrate in critical limb ischemia. 41. Seeger FH, Tonn T, Krzossok N, Zeiher AM, Dimmeler S. Cell isolation
J Vasc Surg. 2011;54:1650–1658. doi: 10.1016/j.jvs.2011.06.118. procedures matter: a comparison of different isolation protocols of bone
31. Losordo DW, Kibbe MR, Mendelsohn F, Marston W, Driver VR, marrow mononuclear cells used for cell therapy in patients with acute
Sharafuddin M, Teodorescu V, Wiechmann BN, Thompson C, Kraiss myocardial infarction. Eur Heart J. 2007;28:766–772. doi: 10.1093/
L, Carman T, Dohad S, Huang P, Junge CE, Story K, Weistroffer T, eurheartj/ehl509.
Thorne TM, Millay M, Runyon JP, Schainfeld R; Autologous CD34+ 42. Britten MB, Abolmaali ND, Assmus B, Lehmann R, Honold J, Schmitt
Cell Therapy for Critical Limb Ischemia Investigators. A randomized, J, Vogl TJ, Martin H, Schächinger V, Dimmeler S, Zeiher AM. Infarct
controlled pilot study of autologous CD34+ cell therapy for critical remodeling after intracoronary progenitor cell treatment in patients with
limb ischemia. Circ Cardiovasc Interv. 2012;5:821–830. doi: 10.1161/ acute myocardial infarction (TOPCARE-AMI): mechanistic insights
CIRCINTERVENTIONS.112.968321. from serial contrast-enhanced magnetic resonance imaging. Circulation.
32. Szabó GV, Kövesd Z, Cserepes J, Daróczy J, Belkin M, Acsády G. Peripheral 2003;108:2212–2218. doi: 10.1161/01.CIR.0000095788.78169.AF.
blood-derived autologous stem cell therapy for the treatment of patients with 43. Teraa M, Sprengers RW, Westerweel PE, Gremmels H, Goumans MJ,
late-stage peripheral artery disease-results of the short- and long-term fol- Teerlink T, Moll FL, Verhaar MC; JUVENTAS study group. Bone mar-
low-up. Cytotherapy. 2013;15:1245–1252. doi: 10.1016/j.jcyt.2013.05.017. row alterations and lower endothelial progenitor cell numbers in critical
33. Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, limb ischemia patients. PLoS One. 2013;8:e55592. doi: 10.1371/journal.
Krishnamurthy S, Anthony N, Pherwani A, Majumdar AS. A double pone.0055592.
blind randomized placebo controlled phase I/II study assessing the 44. Benoit E, O’Donnell TF Jr, Iafrati MD, Asher E, Bandyk DF, Hallett
safety and efficacy of allogeneic bone marrow derived mesenchymal JW, Lumsden AB, Pearl GJ, Roddy SP, Vijayaraghavan K, Patel AN. The
stem cell in critical limb ischemia. J Transl Med. 2013;11:143. doi: role of amputation as an outcome measure in cellular therapy for criti-
10.1186/1479-5876-11-143. cal limb ischemia: implications for clinical trial design. J Transl Med.
34. Li M, Zhou H, Jin X, Wang M, Zhang S, Xu L. Autologous bone mar- 2011;9:165. doi: 10.1186/1479-5876-9-165.
row mononuclear cells transplant in patients with critical leg ischemia: 45. Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Silva GV,
preliminary clinical results. Exp Clin Transplant. 2013;11:435–439. doi: Lai D, Thomas JD, Kronenberg MW, Martin AD, Anderson RD, Traverse
10.6002/ect.2012.0129. JH, Penn MS, Anwaruddin S, Hatzopoulos AK, Gee AP, Taylor DA, Cogle
35. Gu YQ, Zhang J, Guo LR, Qi LX, Zhang SW, Xu J, Li JX, Luo T, Ji BX, CR, Smith D, Westbrook L, Chen J, Handberg E, Olson RE, Geither C,
Li XF, Yu HX, Cui SJ, Wang ZG. Transplantation of autologous bone Bowman S, Francescon J, Baraniuk S, Piller LB, Simpson LM, Loghin
marrow mononuclear cells for patients with lower limb ischemia. Chin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic
Med J (Engl). 2008;121:963–967. RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moyé LA, Simari
36. Klepanec A, Mistrik M, Altaner C, Valachovicova M, Olejarova I, Slysko RD; Cardiovascular Cell Therapy Research Network (CCTRN). Effect of
R, Balazs T, Urlandova T, Hladikova D, Liska B, Tomka J, Vulev I, Madaric transendocardial delivery of autologous bone marrow mononuclear cells
J. No difference in intra-arterial and intramuscular delivery of autolo- on functional capacity, left ventricular function, and perfusion in chronic
Downloaded from http://ahajournals.org by on December 27, 2018

gous bone marrow cells in patients with advanced critical limb ischemia. heart failure: the FOCUS-CCTRN trial. JAMA. 2012;307:1717–1726. doi:
Cell Transplant. 2012;21:1909–1918. doi: 10.3727/096368912X636948. 10.1001/jama.2012.418.
37. Onodera R, Teramukai S, Tanaka S, Kojima S, Horie T, Matoba S, Murohara 46. Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis
T, Matsubara H, Fukushima M; BMMNC Follow-Up Study Investigators; SG, Forder JR, Anderson RD, Hatzopoulos AK, Penn MS, Perin EC,
M-PBMNC Follow-Up Study Investigators. Bone marrow mononuclear Chambers J, Baran KW, Raveendran G, Lambert C, Lerman A, Simon
cells versus G-CSF-mobilized peripheral blood mononuclear cells for treat- DI, Vaughan DE, Lai D, Gee AP, Taylor DA, Cogle CR, Thomas
ment of lower limb ASO: pooled analysis for long-term prognosis. Bone JD, Olson RE, Bowman S, Francescon J, Geither C, Handberg E,
Marrow Transplant. 2011;46:278–284. doi: 10.1038/bmt.2010.110. Kappenman C, Westbrook L, Piller LB, Simpson LM, Baraniuk S,
38. Walter DH, Krankenberg H, Balzer JO, Kalka C, Baumgartner I, Loghin C, Aguilar D, Richman S, Zierold C, Spoon DB, Bettencourt J,
Schlüter M, Tonn T, Seeger F, Dimmeler S, Lindhoff-Last E, Zeiher AM; Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M,
PROVASA Investigators. Intraarterial administration of bone marrow Moyé LA, Simari RD; Cardiovascular Cell Therapy Research Network
mononuclear cells in patients with critical limb ischemia: a randomized- (CCTRN). Effect of the use and timing of bone marrow mononuclear
start, placebo-controlled pilot trial (PROVASA). Circ Cardiovasc Interv. cell delivery on left ventricular function after acute myocardial infarc-
2011;4:26–37. doi: 10.1161/CIRCINTERVENTIONS.110.958348. tion: the TIME randomized trial. JAMA. 2012;308:2380–2389. doi:
39. Assmus B, Tonn T, Seeger FH, Yoon CH, Leistner D, Klotsche J, 10.1001/jama.2012.28726.
Schächinger V, Seifried E, Zeiher AM, Dimmeler S. Red blood cell 47. Jeong H, Yim HW, Cho Y, Park HJ, Jeong S, Kim HB, Hong W, Kim H.
contamination of the final cell product impairs the efficacy of au- The effect of rigorous study design in the research of autologous bone
tologous bone marrow mononuclear cell therapy. J Am Coll Cardiol. marrow-derived mononuclear cell transfer in patients with acute myocar-
2010;55:1385–1394. doi: 10.1016/j.jacc.2009.10.059. dial infarction. Stem Cell Res Ther. 2013;4:82. doi: 10.1186/scrt233.

Clinical Perspective
The results of our double-blind, placebo-controlled, randomized Rejuvenating Endothelial Progenitor Cells via
Transcutaneous Intra-arterial Supplementation (JUVENTAS) trial show no benefit of repetitive intra-arterial infusion of
autologous bone marrow–derived mononuclear cells over placebo on prespecified clinical outcomes (eg, major amputation,
quality of life, wound healing) in patients with severe, nonrevascularizable peripheral artery disease. The lack of benefit of
autologous bone marrow–derived mononuclear cells in our study does not exclude the potential benefit of alternative cell
sources or subpopulations, different administration locations and routes, or that in patients with milder disease. Further
study is warranted to investigate whether cell therapy strategies with selected cell populations, enhanced bone marrow cell
function, or different modes of administration can provide therapeutic benefit in patients with advanced peripheral artery dis-
ease. The general improvement in clinical outcomes observed in both the placebo- and bone marrow–derived mononuclear
cell–treated groups during follow-up stresses the need for future randomized clinical trials to implement a rigorous double-
blinded, placebo-controlled design.

Vous aimerez peut-être aussi