Sie sind auf Seite 1von 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/221849692

Adult Stem Cells and the Clinical Arena: Are we Able to Widely Use this Therapy in
Patients with Chronic Limbs Arteriopathy and Ischemic Ulcers without Possibility of
Revasculariza...

Article  in  Cardiovascular & hematological agents in medicinal chemistry · February 2012


DOI: 10.2174/187152512800388920 · Source: PubMed

CITATIONS READS

7 151

6 authors, including:

Amelia Casamassimi Vincenzo Grimaldi


Università degli Studi della Campania "Luigi Vanvitelli Università degli Studi della Campania "Luigi Vanvitelli
150 PUBLICATIONS   2,178 CITATIONS    58 PUBLICATIONS   596 CITATIONS   

SEE PROFILE SEE PROFILE

Teresa Infante Mohammed Al-Omran


SDN Istituto di Ricerca Diagnostica e Nucleare University of Toronto
65 PUBLICATIONS   599 CITATIONS    153 PUBLICATIONS   2,118 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Kidney transplantation View project

An interview-based survey to assess the knowledge of peripheral arterial disease among medical students View project

All content following this page was uploaded by Vincenzo Grimaldi on 22 February 2016.

The user has requested enhancement of the downloaded file.


Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, 10, 99-108 99

Adult Stem Cells and the Clinical Arena: Are we Able to Widely Use this
Therapy in Patients with Chronic Limbs Arteriopathy and Ischemic Ulcers
without Possibility of Revascularization?

Amelia Casamassimi1,#, Vincenzo Grimaldi1,#,*, Teresa Infante2,#, Mohammed Al-Omran3,


Valeria Crudele1 and Claudio Napoli1,2

1
Department of General Pathology, Chair of Clinical Pathology, U.O.C. Immunohematology, and Excellence Research
Centre on Cardiovascular Disease, 1st School of Medicine, Second University of Naples, 80138 Naples, Italy (AC, VG,
VC, C.N.) 2Fondazione Studio Diagnostica Nucleare (SDN), IRCCS, 80134 Naples, Italy (T.I., C.N.); 3Peripheral
Vascular Disease Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia (M.AO.)

Abstract: The following paper is an overview on stem cells therapy in patients with peripheral vascular diseases. Recent
research shows the ability of stem cells to develop and strengthen the collateral network in ischemic legs. Here,
we discuss this clinical and therapeutic approach. To date, research has been mainly focused on patients with ischemic
ulcers without possibility of revascularization. Non-invasive stem cell therapy has been proposed as an alternative to the
amputation of such patients, but when the ulcers appear it is sometime too late. In our point of view, the selection of
patients is a very important issue and we believe that the best candidate for this treatment is the patient with intermittent
claudication before the development of ulcers. This choice could allow the optimization of results by the type of treated
patients and not only by the type of infused cells. Indeed, several variables still remain to be elucidated for stem cell
therapy, including the type of cells to be used, the infusion route, and more importantly, the stage of patients to be treated.
Keywords: Angiogenesis, autologous transplantation, bone marrow cells, circulating endothelial progenitor cells, clinical trials,
Fontaine’s stage, gangrene, intermittent claudication, limb ischemia, peripheral arterial disease, revascularization.

INTRODUCTION ulcers lesions and leg savage. Only few studies were
concentrated on pain free walking distance without lesions.
The peripheral arterial disease (PAD) is caused by an We think that this point is very important in the research of
inadequate supply of oxygen to the limbs. PAD has two this therapeutic approach, since, of course, not all patients
main stages: initially, the lack of oxygen occurs after can get the same outcome after cell therapy. Thus, proper
muscular exertion, causing the Intermittent Claudication patient selection is necessary to change this therapy
(IC); then, necrosis and gangrene are the result of chronic assessment and to provide new opportunities of positive
oxygenation absence [1]. Unfortunately, many patients cannot outcomes.
be helped with surgery or endovascular revascularization.
Moreover, this type of intervention is not conclusive, In this review, we focus on clinical trials that use stem
because after a few years occlusion and thrombosis can cells to treat patients suffering from limb ischemia.
occur. To date, we have not yet found a way to prevent
THE PREVALENCE OF PERIPHERAL VASCULAR
this complication; antiplatelet therapy has only prolonged DISEASE
the time of thrombosis occurrence, as well as, monitoring
with echocolordoppler and endovascular technique (PTA). The recent prevalence of the disease has been derived
Stem cell therapy has been proposed as an alternative to from numerous studies. In the Limburg study on 3654
amputation of limbs [2]. Some clinical studies have shown individuals, the prevalence of IC was 0.6% in aged between
that injection of bone marrow cells (BMC) in ischemic limbs 45 and 54 years, 2.5% between 55 and 65 years and 8.8%
is an effective therapy and can improve tissue vascularization between 67 and 74 years [4]. The Edinburgh Artery Study
[3]. (1592 patients) showed a prevalence of 4.5% between 55 and
74 years [5]. In the Scotland study, performed on 10,042
But our question is: which patients may actually benefit patients, the prevalence of IC between 40 and 59 years
from progenitor cells therapy? During the past years the turned out to be 1,1% [6]. In the Palermo study on 1558
main end-point of most studies has been the improvement of patients, a prevalence of 4.7% IC was observed in the range
between 40 and 49 years and 9.2% between 50 and 59 years
[7]. Finally, in the Rotterdam Study evaluating 7715 subjects,
*Address correspondence to this author at the Department of General the presence of PAD and IC was determined through the
Pathology, U.O.C. Immunohematology, Second University of Naples, Piazza measurement of ankle-arm index and PAD was considered
Miraglia, 2, 80138, Naples, Italy; Tel: +390815667567; Fax: +390819700943;
E-mail: vincenzo.grimaldi@policliniconapoli.it present with an index was below a 0.9 value. In these
#
Contributed equally to this study subjects, the prevalence of PAD was 19.1%, whereas IC was

1875-6182/12 $58.00+.00 © 2012 Bentham Science Publishers


100 Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 Casamassimi et al.

1,6% [8]. In most of these studies there is a reasonable surgical intervention or prostanoids usage. However, most
correlation of results with an average prevalence of PAD patients, after failure of surgery, have to undergo to major
varying between 3% and 6% around the age of 60 years. amputation after few years or months. Moreover, as stated
above, not all patients can have surgery revascularization [9].
CLINICAL TREATMENT Indeed, the assessment of run-off is the prerequisite to
achieve a by-pass. Then, a lot of randomized studies show
Usually PAD patients are subdivided according to the
the no effect of surgical treatment on amputation tax [1]. In
Fontaine’s classification (Table 1). The risk factors associated
this context, therapy with progenitor cells can be introduced.
with PAD are old age, diabetes, smoking, hypertension, and
In most preclinical and clinical studies on progenitor cells
dyslipidemia (TASC II) [1]. Interestingly, the presence of
the end-point used has been the amputation of limbs or the
these risks is associated with a lower number and reduced
functionality of circulating vascular progenitor cells [9, 10]. regression of necrosis. But, how long is needed to obtain a
collateral network in the stem cell therapy? Is the gangrene
The patient with critical limb ischemia has a poor prognosis
faster than stem cell implantation?
and is at high risk for limb amputation [1].
ADULT STEM-LIKE CELL THERAPY
Table 1. Leriche Fontaine’s classification.
The main goal of cell therapy is to improve peripheral
vascularization by inducing therapeutic angiogenesis. Two
Fontaine’s Stages Symptomatology distinct post-embryogenesis mechanisms are essentially
responsible for new blood vessel formation in adult life:
Ist stage Asymptomatic
angiogenesis, where local capillaries proliferate and thereby
IInd stage Claudicatio intermittens increase local distribution of blood flow [10]; arteriogenesis,
rd
where pre-existing high resistance collaterals enlarge,
III stage Chronic pain thereby decreasing flow resistance and increasing perfusion
IVth stage Trophic lesion-gangrene in the ischemic region [11]. Angiogenesis occurs in response
to local hypoxia and it is mediated by hypoxia-inducible
factor 1-alpha (HIF-1) release of chemokines and
The first step of PAD treatment is deleting the risk cytokines, particularly VEGF and related signalling growth
factors. In the Ist and IInd stages medical procedures are used factors, FGF and nitric oxide; then, endothelial cells loosen
(antiplatelet, muscular work, carnitin, Pentoxifylline, etc). their intracellular connections, sprout, migrate and form new
The aim of therapy is to increase the free pain walking thin-walled vessels [12]. An increase in the number of
distance as well as to obtain a normal relationship life. In capillaries without a concomitant increase in the number and
patients without response to conservative and pharma- caliber of conducting vessels is insufficient to augment tissue
cological therapies, the revascularization intervention and perfusion.
control of pain can save the limbs. It is the second mechanism, arteriogenesis, involving
In patients at IIIrd and IVth stages, revascularization remodelling of pre-existing small arterioles into larger
intervention is directly used because of the promptness vessels, that is the essential component of a developing
of disease progression rapidly leading to amputation functional collateral network [13, 14]. Under normal
for massive gangrene in a few months. Two types of circumstances, flow through pre-existing collaterals (lying in
revascularization procedures can be used: surgical by-pass or parallel to the native artery) is low because of their small
endovascular techniques. The choice depends on the caliber and resulting high resistance. However, following
localization of the atherosclerotic obstruction and on the parent vessel occlusion, the consequent high pressure
lesion type. A significant proportion of patients are not leads to an increase of blood flow exerted on the wall of
eligible for these revascularization procedures, due to the arterioles connecting the vessels proximal and distal to the
distal localization of the obstruction or its length or the occlusion [15]. This pressure gradient induces a concomitant
absence of receiving vessels [9]. Indeed, the aim of these shear stress, which promotes proliferation, differentiation
interventional procedures is to maintain the blood on feet. and tube formation of endothelial cells, other than a higher
To this purpose, an artery “giver” and an artery “receiving” mitotic activity of both endothelial cells and smooth-muscle
(run off) are necessary; but, unfortunately, they are not cells required to form the neovasculature or to remodel
always present. In the last patients, to date, therapy is based existing collaterals [16, 17]. At molecular level, shear stress
on pain control (either pharmacological or with spinal cord induces a cascade triggered by nitric oxide production; then,
stimulation). endothelial cell activation leads to the infiltration and
In the patient at the IVth stage with important gangrene, adhesion of monocytes that secrete proteases, growth factors
sometimes, the amputation is the only option to save life. and chemokines. Finally, the whole process promotes the
enlargement of arterioles, which can form the collateral
In the TASC II manuscript [1], there are only few words vasculature turning blood flow beyond the occlusion [14-18].
about the implantation of borrow marrow cells, simply
indicated as a promising research. Particularly, it mentioned Bone Marrow Cells and Circulating Endothelial
the ability to stimulate vascular growth by bone marrow cells Progenitor Cells
or treatment with vascular endothelial growth factor (VEGF)
and basic fibroblast growth factor (bFGF). At this time, Several studies suggest an additional contribution derived
therapy for critical ischemia of lower limbs is based on from bone marrow progenitors, which are mobilized in the
Adult Stem Cells and the Clinical Arena Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 101

limb setting, migrate to ischemic tissues and incorporate into specificity. For instance, in EPCs functional assays, different
new vessels [18]. It is proposed that tissue ischemia induces culture conditions produce different EPC phenotypes: early
a local increase of chemokines, such as VEGF, thus EPCs (CD133+CD34+ cells) and late outgrowth EPCs
promoting migration of progenitor cells, expressing vascular (CD133CD34+ cells) or mixed ones. More importantly,
endothelial growth factor receptor 1- and 2- (VEGFR1 and these cell populations (early and late EPCs) seem to show a
VEGFR2), to the ischemic territory [19]. Thus, one rationale different contribution to neovascularization of ischemic
and proposed advantage of cell therapy versus conventional tissues and re-endothelization of injured vessels [17, 21, 25].
therapies is that cell therapy can directly increase the number
of stem/progenitor cells in the ischemic tissue. Paracrine Mechanisms in Adult Stem Cell Therapy
The increase of local stem/progenitor cells could be Despite some evidence that bone marrow-derived stem or
achieved either by systemic delivery (i.e. systemic injection progenitor cells incorporate into vascular structures, several
of progenitor cells or inducing them to mobilize from the studies suggest that only a small number of vessels contain
bone marrow microenvironment by chemokine therapy) or donor cells. A recent study shows that EPCs are not
by direct cell injection into or around the ischemic zone. frequently observed in the femoral muscle after 8 weeks and
Regardless of the pathway taken, the number of potential they preferentially home to the sciatic nerve. The authors
candidate cells to incorporate into the vessel wall is hypothesize that the role of EPCs on muscle angiogenesis is
substantially increased [17, 20]. Many clinical studies have mediated by indirect mechanism with cytokines on resident
used adult stem cells derived from autologous bone marrow. progenitor cells [26].
This choice is due to the easy availability, security and
lack of ethical controversies associated with the use of In addition, despite heterogeneous bone marrow cell
embryo derived stem cells. The bone marrow contains populations (i.e., mononuclear cells or unfractionated cells)
several subpopulations of cells, which can be isolated by contain very small numbers of stem cells (0.01% of total
direct extraction or by mobilization into peripheral blood cells), injection of these cells significantly enhances
by using cytokines (such as G-CSF) [17, 20]. The two collateral development. Thus, other mechanisms are likely to
main groups of BMCs are hematopoietic stem cells contribute to the overall improvement of vascular function
and mesenchymal stem cells, which can be further divided and benefits observed after bone marrow cell therapy [27].
into different subpopulations according to the expression An alternative or perhaps complementary hypothesis to
of specific cell surface receptors. A subpopulation of the concept of BMC and EPC incorporation is that the
heterogeneous cells includes endothelial progenitor cells injected cells act in a supportive role, optimizing the milieu
(EPC), which are also found, although in less amount, in for host vasculature to respond to tissue ischemia. Many
other districts like peripheral blood, fetal liver or umbilical bone marrow subpopulations (for example EPCs) are a
cord blood [21, 22]. source of growth factors previously demonstrated to be
In 1997, the existence of bone marrow-derived EPCs in central in the initiation and coordination of angiogenesis
the peripheral circulation was reported for the first time [21, [28]. Thus, a paracrine hypothesis has been recently
23]. These cells contribute to the re-endothelization of advanced establishing that the injected stem cells release into
injured vessels as well as to neovascularization of ischemic the surrounding tissue soluble factors, such as cytokines,
lesions, suggesting that EPCs play a major role in the chemokines and growth factors, that contribute to vascular
pathogenesis of atherosclerosis and cardiovascular diseases. repair and regeneration by paracrine mechanisms [29].
The number and function of EPCs are regulated both by Indeed, these released factors are able to induce angiogenesis
several angiogenic growth factors, cytokines and chemokines and neovascularization also through the recruitment and
as well as by cardiovascular risk factors or lifestyle activation of resident stem cells. The whole understanding of
modifications. An increase in the number of EPCs induced these novel mechanisms can have many future clinical
by ischemic stimuli contributes to reduction in ischemic implications in the field of cell therapy for vascular diseases.
damage through neovascularization. EPCs number and Since the fate of cells injected into peripheral muscle is
function directly influence the maintenance and development unknown, even the rationale of intramuscular injection is
of cardiovascular diseases [24]. based on the paracrine activity provided by injected cells in
Although a precise definition of EPCs is still lacking, the ischaemic area [29].
these cells are identified through the expression of a
CLINICAL STUDIES AND CRITICISMS
combination of hematopoietic stem cell markers, such as
CD34, CD45 and CD133, and endothelial-cell markers, The first pilot study on the treatment of patients with
including VEGFR2, von Willebrandfactor, endothelial nitric limb ischemia by autologous transplantation of BMCs was a
oxide synthase among others [17, 21, 22]. However, there randomized controlled trial published in 2002 [30] (Table 2).
is no a gold standard marker, resulting in confusion Since then, considering the whole published studies, more
regarding their definition. Moreover, EPCs have also been than 900 patients have been enrolled. Most of these studies
characterized by other methods like the uptake of have analysed patients in the IVth stage of Fontaine’s
diacetylated low-density lipoprotein and binding of classification, without any possibility of revascularization
fluorescently labeled Ulex europaeus agglutinin 1 lectin or [30-64]. Only two studies [42, 44] made a clear distinction of
by several in-vitro and in-vivo functional assays, such as the patients in subgroups by differentiating the clinical stages
colony formation, tube formation, and vascular integration and trophic lesions of patients with diabetes, haemodialysis,
[17, 21]. However, all these identification methods lack vasculitis etc. Noteworthy, the most favourable results were
102 Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 Casamassimi et al.

Table 2. Published studies of cell therapy in peripheral vascular diseases.

Published Studies Delivery Route Condition Subjects Cell Type Follow-UP Time End-Point and Clinical Indexes

2002 Tateishi-Yuyama BMCs or ABI and TcO2 improvement; rest


IM PAD, DM 45 4 and 24 weeks
et al., [30] PBMNCs pain; pain-free walking time

2002 Esato et al., [31] IA PAD, TAO 8 BMCs N/D Ulceration healing

2004 Saigawa et al., [32] IM PAD, DM 8 BMCs 4 weeks ABI and TcO2 improvement

2004 Higashi et al., BMCs or BM- ABI and TcO2 improvement; pain-
IM PAD 8 4 and 24 weeks
[33] MNCs free walking time

2004 Miyamoto ABI improvement; pain-free


IM PAD, CLI 12 BMCs - EPCs N/D
et al., [34] walking time

ABI, blood flow, laser Doppler blood


2004 Huang et al., [35] IM PAD 5 PBMNCs 3 months perfusion and angio-graphic scores
improvement

Normalization of limb temperature


2005 Kawamura
IM PAD, CLI 30 PBMNCs N/D was observed by thermograph, and
et al., [36]
symptoms also improved

ABI and TcO2 improvement; rest


2005 Lenk et al., [37] IA CLI 7 PBMNCs 20 weeks pain; pain-free walking time; flow-
dependent vasodilation

ABI, limb pain and ulcers, were


2005 Huang et al., [38] IM CLI, DM 28 M-PBMNCs 3 months
significantly improved

2005 Ishida et al., [39] IM TAO 6 M-PBMNCs 4 and 24 weeks ABI and ischemic ulcers improvement

ABI, rest pain scores and peak


2006 Durdu et al., [40] IM TAO 28 BM-MNCs 3 and 6 months
walking time improvement

2006 Miyamoto 4 weeks, 4 and 7 Rest pain and ischemic ulcers


IM TAO, CLI 8 BM-MNCs
et al., [41] months, 1 year improvement

2006 Kawamura
IM CLI 92 PBMNCs 1,5 month Capillary formation
et al., [42]

2007 Bartsch et al., ABI and pain-free walking distance


IM and IA PAD, CLI 13 BMCs 2 and 13 months
[43] improvement

BM-MNCs or
2007 Huang et al., [44] IM PAD 150 12 weeks ABI and rest pain improvement
M-PBMNCs

2007 Kajiiguchi et al., BM-MNCs or


IM CLI, TAO 7 1 month ABI improvement (for 3 patients)
[45] PBMNCs

2007 Hernandez ABI, SaO2, pain-free walking time


IM CLI, DM 12 BM-MNCs 24 months
et al., [46] and rest pain improvement

2007 Saito et al., [47] IM TAO 7 BM-MNCs N/D TcO2 improvement

2008 Matoba Leg pain scale, ulcer size and pain-


IM PAD, TAO 115 BM-MNCs 3 years
et al., [48] free walking distance improvement

3,6,12 and 18 ABI, ischemic ulcers and pain-free


2008 Napoli et al., [3] IA PAD 18 BM-MNCs
months walking distance improvement

ABI and TcO2 improvement and


2008 Gu et al., [49] IM and IA PAD, DM 16 BM-MNCs 4 weeks
limb salvage

2008 Chochola Collateral vessel development and


IA PAD, DM 28 BM-MNCs 1 year
et al., [50] indexes improvement

2008 Wester et al., [51] IM PAD/CLI 8 BMCs 4 and 8 months Pain relief and indexes improvement
Adult Stem Cells and the Clinical Arena Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 103

Table 2. contd….

Published Studies Delivery Route Condition Subjects Cell Type Follow-UP Time End-Point and Clinical Indexes

Pain-free walking distance, ABI


2008 Van Tongeren
IM and IA CLI, PAD 27 BMCs 6 and 12 months improvement and pain scores
et al., [52]
reduction

Modest improvement of resting pain,


2008 De Vriese
IM CLI 16 BM-MNCs 12 weeks TcO2 improvement, no ABI
et al., [53]
significant improvement

2008 Cobellis et al., ABI and pain-free walking distance


IA PAD 10 BM-MNCs 12 months
[54] improvement

No clinical or perfusion improvement.


2009 Amann et al., 3 months Limb salvage, ABI and TcO2 and
IM PAD 51 BM-MNCs
[55,56] 6 months pain-free walking distance
improvement

BM-MNCs or
2009 Capiod et al., [57] IM CLI 24 N/D Therapeutic angiogenesis
PBMNCs

Improvement in symptom severity.


2009-2011 Franz et al., 3 and 8 months ABI improvement; absence of rest
IM and IA PAD 18 BM-MNCs
[58,59] 3 months pain; absence of ulcers; and absence
of major limb amputations

2010 Zafarghandi et al., BM-MNCs ABI and pain-free walking distance


IM CLI 50 4 and 24 weeks
[60] and G-CSF improvement

2010 Prohazka et al., ABI improvement and absence of


IM PAD 42 BM-MSCs 4 months
[61] major limb amputations

2010 Lara-Hernandez 14, 18 months


IM CLI 28 mobilized EPCs ABI improvement, limb savage
et al., [62] and 1 year

Not significantly increase ankle-


2011 Walter et al., [63] IA CLI 40 BM-MNCs 3 months brachial index, ulcer healing and
improvement of resting pain

First toe pressure and toe-brachial


2011 Murphy et al., index increase, perfusion index by
IM CLI 29 BM-MNCs 1 year
[64] PET-CT increase and rest pain
improvement

ABI: ankle-brachial pressure index; BMCs: bone marrow cells; BM-MNCs, bone marrow-derived mononuclear cells; BM-MSCs: bone marrow-mesenchymal stem cells; CLI: critical
limb ischemia; DF: diabetic foot; DM, diabetes mellitus; EPCs: endothelial progenitor cells; G-CSF: granulocyte colony-stimulating factor; IA: intra-arterial injection; IM:
intramuscular injection; IV: intravenous injection; LASO: lower limb arteriosclerosis obliterans; LEI: lower extremity ischemia; M-PBMNCs: G-CSF-mobilized peripheral blood
mononuclear cells; PAD: peripheral artery disease; PET-CT: positron emission tomography-computed tomography; PB-MNCs: peripheral blood derived mononuclear cells; SaO2:
arterial oxygen saturation; SLI: severe leg ischemia; TAO, thromboangiitis obliterans; TcO2: transcutaneous oxygen pressure.

obtained in patients with diabetes or haemodialysis or patients should be the increase of the pain-free walking
vasculitis lesions with leg arteries in discrete function (good distance, the ulcers prevention and the rest pain loss, before
run-in). In these studies the best results were achieved in the skin lesions occur.
patients with stages II and III, whereas unsatisfactory
Most of the studies enrolled a limited number of patients
outcomes were obtained in patients with stage IV and and did not include a placebo arm, with no double-blinded
diabetic ulcers. This result can be rationally explained by the
randomized design and a very short follow-up period. Thus,
progression of the disease. Indeed, a long period is needed to
these studies do not provide definitive proof of the positive
detect the development of a collateral circulation stimulated
therapeutic effect of cell therapy.
by BMCs (at least 6 months to have an angiographic result).
However, patients with ulcers at stage IV (with the exception In most of the trials the preferred administration route
of patients with Burger’s disease or diabetic patients with was the injection into the ischemic muscle [30, 32-36, 38-42,
arterial permeability) generally do not have such a period 44-48, 49, 51-53, 55-62, 64] whereas only fewer studies with
before the disease progression into gangrene. In our opinion, a little number of treated patients utilized the intra-arterial
the main target of stem cell therapy is the patient with IC injection [3, 31, 37, 43, 49, 50, 52, 54, 58, 59, 63] (Table 2).
(stage II) or night pain (stage III). Thus, the outcome of Thus, to date we still do not know which is the better
104 Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 Casamassimi et al.

infusion way. In addition, a combination of both methods revascularization therapies. Several phase III studies
has been tested in some small studies showing no significant are currently undergoing to evaluate the efficacy of
differences in the clinical characteristics between patients intramuscular injection of autologous BMC in patients with
treated with intra-arterial or intra-arterial plus intramuscular CLI, such as the NCT00904501 and NCT01245335 trials
cell administrations [43, 49, 52, 58, 59]. However, there are (Table 3).
no direct comparisons of these two different routes of
Interestingly, in the study of Kawamura et al., [42],
administration of cell therapy in patients with limb ischemia,
where 92 patients were treated, the authors made a precise
although the intra-arterial administration offered similar
classification of patients in Fontaine’s stages with diabetes
levels of angiogenic activity as intramuscular injection in a
or haemodialysis treatment. We agree with the conclusions
rat model study [65].
of this study that non diabetic, non dialyzed patients
Most of the published studies are small studies and show with ischemic limbs are strongly indicated for stem cell
the results of short or medium term follow-up. To date, only transplantation regardless of Fontaine classification.
one study reported a long-term follow-up (36 months), Therapeutic angiogenesis is effective for critically ischemic
suggesting that intramuscular implantation of bone marrow limbs resulting from hemodialysis and diabetes until
mononuclear cells may lead to extension of amputation- Fontaine stage III, but it is of limited effectiveness for stage
free interval and improvement of ischemic pain, ulcer size IV cases. In the 92 patients only the stage I, II, III had a good
and walk without pain [56]. In addition, the safety and response, whereas it was of limited effectiveness for stage IV
efficacy of this therapy was not lower than conventional cases.

Table 3. NIH ongoing interventional trials with a number of patients  30.

Patients
NIH Registration Number Status Cell Type Condition Delivery Route Phases
Treated

NCT00904501 Unknown 110 BMCs CLI - PAD IM III

NCT00987363 Active, not recruiting 60 BMCs CLI - PAD IA I and II

NCT00539266 Recruiting 108 BMCs CLI - PAD IM II and III

NCT00411840 Unknown 100 BMCs CLI - PAD IM and IA I

NCT00468000 Active, not recruiting 150 BMCs CLI - PAD IM II

NCT01245335 Recruiting 210 BMCs CLI - PAD IM III

NCT00721006 Completed 35 BMCs CLI - PAD - SCI - PVD IM II

NCT01065337 Completed 30 BMCs CLI - DF IM and IA II

NCT01232673 Active, not recruiting 40 BMCs CLI - DF IM II

Local
NCT00292357 Unknown 40 BMCs DF I
application

NCT00595257 Active, not recruiting 60 MSCs CLI - PAD IM II

NCT01079403 Recruiting 36 MSCs CLI IA I and II

NCT01351610 Recruiting 30 MSCs CLI - PAD IV I and II

Umbilical
NCT01216865 Not yet recruiting 50 CLI IM I and II
cord MSCs

CD133+
NCT00753025 Completed 42 CLI - LEI IM II
from BM

NCT00922389 Unknown 36 M-PBMNCs CLI - DF IM I and II

GM-CSF
NCT01408901 Not yet recruiting 267 M-PBMNCs PAD ?
administration

GM-CSF
NCT01041417 Recruiting 180 M-PBMNCs PAD I
administration

BMCs: bone marrow cells; CLI: critical limb ischemia; DF: diabetic foot; G-CSF: granulocyte colony-stimulating factor; IA: intra-arterial injection; IM: intramuscular injection; IV:
intravenous injection; LEI: lower extremity ischemia; M-PBMNCs: G-CSF-mobilized peripheral blood mononuclear cells; MSCs: Mesenchymal Stem Cells; PAD: peripheral artery
disease; PB-MNCs: peripheral blood-derived mononuclear cells; SLI: severe leg ischemia.
Detailed information about the NIH registered clinical trials are reachable visiting the web site http://clinicaltrials.gov/.
Adult Stem Cells and the Clinical Arena Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 105

Good impact Middle impact Bad impact


Fig. (1). Selection of PAD patient and impact of cell therapy. Patients with peripheral artery disease have been subdivided into three major
subgroups based on the disease stage. Patients with Intermittent claudication are considered at high impact for cell therapy, whereas patients
with pain or ulcers are at middle and bad impact respectively.

The same results have been confirmed by another study number of treated patients [62]. Thus, further research is
where a comparison of G-CSF-mobilized peripheral blood needed to decide which cells should be used for therapy and
mononuclear cells (M-PBMNCs) versus bone marrow- we have to wait for the results from the currently ongoing
mononuclear cells (BM-MNCs) for the treatment of patients clinical trials (Table 3).
with lower limb arteriosclerosis obliterans was made [44];
Another important consideration is about the patient at
also in this case a minor response in patients with diabetes
IVth stage. It is not correct to evaluate the patient at this stage
was observed. This result was associated with the
only for the presence of ulcers. We think it is necessary to
impairment of diabetic M-PBMNCs capacity to differentiate
divide this stage into subgroups for the presence or absence
into EPCs, to incorporate into vessel-like tubules in vitro,
to participate in vascular-like structure formation in a of best run-in for lesions of big artery in the legs. It is not
subcutaneous matrigel plug and to stimulate the recruitment possible to compare a Burger’s disease with pure chronic
of pericytes/smooth muscle cells. In addition, there was an arteriopathy or a diabetic foot with only a micro-angiopathy
impairment of vasculogenesis, which was related to the with diabetic patient that have also macro-angiopathy
reduced adhesion ability of EPCs from diabetic M-PBMNCs (obstruction of big artery). Indeed, these diseases have
[44]. different physiopathology and different evolution, especially
for the duration life of leg.
Another question to be addressed is the technique to
obtain stem cells. Of course, the borrow marrow cells CONCLUSIONS
aspirated from the iliac crest is the best technique and the
most tested. By contrast, this method requires a local or At the examination of the literature, there are no
general anaesthesia and a collection of a large amount of randomized, double-blinded trials, as well as only few
marrow by aspiration, which includes the risk of anaesthetic studies have reported the results obtained on more than 50
accident and excludes patients with contraindications to patients [42, 44, 48, 56]. Obviously, these data are not
anaesthesia [44]. About 250-500 ml bone marrow (5.8 ± enough to achieve significant conclusions about the
2.3108 mononuclear cells) can be usually aspirated from effectiveness of this therapy. The limit of most publications
the ileum, which is enough for only one implantation. is the selection of patients. Probably, not all the patients in
To aspirate or collect more bone marrow cells the patient the IVth Fontaine stage are suitable for the infusion of stem
risk increases to an unacceptable degree. In this study, cells. In the few studies where patients were divided into
comparative analysis indicated that M-PBMNCs should clinical sub-groups, the best results were achieved in patients
be more practical in comparison with BM-MNCs for with stages II and III, which can be explained by the
treatment [44]. However, to obtain M-PBMNCs, the patients progression of the disease. In our opinion, these patients
received recombinant human G-CSF, 300 μg twice a day should be the main targets of stem cell therapy. Obviously,
subcutaneously for five consecutive days. In the study of our considerations require further clinical studies to confirm
Huang [44] no collateral effects were found, but because of the effectiveness of therapy in patients with first disease
the possible dangerous consequences of G-CSF, a long time stages. It would be interesting to assess the impact on
observation would be necessary to validate the study. This pain-free walking distance not only in patients with
key point has been also discussed in a more recent study severe claudication in the absence of run-off (without
where mobilized EPCs have been administered in a small revascularization chance), but also in those with good run-
106 Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 Casamassimi et al.

off and mild claudication, which currently do not undergo [2] Aranguren, X.L.; Varfaille C.M.; Luttun, A. Emerging hurdles in
revascularization intervention. Thus, we should consider the stem cell therapy for peripheral vascular disease. J. Mol. Med.,
2009, 87(1), 3-16.
entering of stem cell therapy in medical treatment before [3] Napoli, C.; Farzati, B.; Sica, V.; Iannuzzi, E.; Coppola, G.;
exposing the patient to surgical revascularization. By this Silvestroni, A.; Balestrieri, M.L.; Florio, A.; Matarazzo, A.
approach, the increase of collateral circulation can improve Beneficial effects of autologous bone marrow cell infusion and
the run-off and, conceivably, the potency of the bypass in the antioxidant / L-arginine in patient with chronic critical limb
ischemia. Eur. J. Cardiovasc. Prev. Rehabil., 2008, 15(6), 709-718.
successive clinical practice. Thus, the imperative would be: [4] Stoffers, H.-E.-J.-H.; Kaiser, V.; Knottnerus, J.-A. Prevalence in
do not wait ulcer development, but prevent it! This concept general practice. In: Fowkes FGR ed. Epidemiology of peripheral
is well explained in Fig. (1) where selected patients have vascular disease. Springer-Verlag: London, 1991.
been displayed as high, middle and bad impact of cell [5] Fowkes, F.G.; Housley, E.; Cawood, E.H.; Macintyre, C.C.;
Ruckley, C.V.; Prescott, R.J. Edinburgh Artery Study: prevalence
therapy. of asymptomatic and symptomatic peripheral arterial disease in the
general population. Int. J. Epidemiol., 1991, 20(2), 384-392.
CONFLICT OF INTEREST [6] Smith, W.-C.-S.; Woodward, M.; Tunstall-Pedoe, H. Intermittent
claudication in Scotland. In: Fowkes FGR ed. Epidemiology of
The authors have no conflict of interest peripheral vascular disease. Springer-Verlag: London, 1991.
[7] Novo, S.; Avellone, G.; Di Garbo, V.; Abrignani, M.G.; Liquori,
ACKNOWLEDGEMENTS M.; Panno, A.V.; Strano, A. Prevalence of risk factors in patients
with peripheral arterial disease: a clinical and epidemiological
This work was supported in part by Grants from PRIN- evaluation. Int. Angiol., 1992, 11(3), 218-229.
MIUR 2008, Foundation Jerome Lejeaune, and Regione [8] Meijer, W.T.; Hoes, A.W.; Rutgers, D.; Bots, M.L.; Hofman, A.;
Grobbee, D.E. Peripheral arterial disease in the elderly: the
Campania 2009. Rotterdam study. Arterioscler. Thromb. Vasc. Biol., 1998, 18(2),
185-192.
ABBREVIATIONS [9] Collinson, D.J.; Donnelly, R. Therapeutic angiogenesis in
peripheral arterial disease: can biotechnology produce an effective
bFGF = Basic fibroblast growth factor collateral circulation? Eur. J. Vasc. Endovasc. Surg., 2004, 28(1),
9-23.
BMCs = Bone Marrow Cells [10] Carmeliet, P. Angiogenesis in health and disease. Nat Med., 2003,
BM-MNCs = Bone marrow-derived mononuclear cells 9(6), 653-660.
[11] Schaper, W.; Buschmann, I. Collateral circulation and diabetes.
CLI = Critical Limb Ischemia Circulation, 1999, 99(17), 2224-2226.
[12] Rey, S.; Semenza, G.L. Hypoxia-inducible factor-1-dependent
DF = Diabetic foot mechanisms of vascularization and vascular remodelling.
Cardiovasc. Res., 2010, 86(2), 236-242.
DM = Diabetes mellitus [13] Unthank, J.L.; Sheridan, K.M.; Dalsing, M.C. Collateral growth in
EPCs = Endothelial progenitor cells the peripheral circulation: a review. Vasc. Endovascular Surg.,
2004, 38(4), 291-313.
G-CSF = Granulocyte colony-stimulating factor [14] Schierling, W.; Troidl, K.; Troidl, C.; Schmitz-Rixen, T.; Schaper,
W.; Eitenmuller, IK. The role of angiogenic growth factors in
IA = Intra-arterial injection arteriogenesis. J. Vasc. Res., 2009, 46(4), 365-374.
[15] Pan, S. Molecular mechanisms responsible for the atheroprotective
IC = Intermittent Claudication effects of laminar shear stress. Antioxid. Redox. Signal., 2009,
11(7), 1669-1682.
IM = Intramuscular injection [16] Virag, J.I.; Murry, C.E. Myofibroblast and endothelial cell
LASO = Lower limb arteriosclerosis obliterans proliferation during murine myocardial infarct repair. Am. J.
Pathol., 2003, 163(6), 2433-2440.
LEI = Lower extremity ischemia [17] Tongers, J.; Roncalli, J.G.; Losordo, D.W. Role of endothelial
progenitor cells during ischemia-induced vasculogenesis and
MSCs = Mesenchymal Stem Cells collateral formation. Microvasc. Res., 2010, 79(3), 200-206.
[18] Shintani, S.; Murohara, T.; Ikeda, H.; Ueno, T.; Honma, T.; Katoh,
PAD = Peripheral Artery Disease A.; Sasaki, K.; Shimada, T.; Oike, Y.; Imaizumi, T. Mobilization of
PB-MNCs = Peripheral blood derived mononuclear endothelial progenitor cells in patients with acute myocardial
infarction. Circulation, 2001, 103(23), 2776-2779.
cells [19] Asahara, T.; Takahashi, T.; Masuda, H.; Kalka, C.; Chen, D.;
SLI = Severe leg ischemia Iwaguro, H.; Inai, Y.; Silver, M.; Isner, J.M. VEGF contributes to
postnatal neovascularisation by mobilizing bone marrow-derived
TAO = Thromboangiitis obliterans endothelial progenitor cells. EMBO J., 1999, 18(14), 3964-3972.
[20] Kinnaird, T.; Stabile, E.; Burnett, M.S.; Epstein, S.E. Bone marrow-
VEGF = Vascular endothelial growth factor derived cells for enhancing collateral development: mechanisms,
animal data, and initial clinical experiences. Circ. Res., 2004,
VEGFR1 = Vascular endothelial growth factor 95(4), 354-363.
and -2 = receptor-1 and -2 [21] Napoli, C.; Hayashi, T.; Cacciatore, F.; Casamassimi, A.; Casini, C.;
Al-Omran, M.; Ignarro, L.J. Endothelial progenitor cells as therapeutic
REFERENCES agents in the microcirculation: an update. Atherosclerosis, 2011,
215(1), 9-22.
[1] Norgren, L.; Hiatt, W.R.; Dormandy, J.A.; Nehler, M.R.; Harris, [22] Richardson, M.R.; Yoder, M.C. Endothelial progenitor cells: quo
K.A.; Fowkes, F.G.; Bell, K.; Caporusso, J.; Durand-Zaleski, I.; vadis? J. Mol. Cell. Cardiol., 2011, 50(2), 266-272.
Komori, K.; Lammer, J.; Liapis, C.; Novo, S.; Razavi, M.; Robbs, [23] Asahara, T.; Murohara, T.; Sullivan, A.; Silver, M.; van der Zee,
J.; Schaper, N.; Shigematsu, H.; Sapoval, M.; White, C.; White, J.; R.; Li, T.; Witzenbichler, B.; Schatteman, G.; Isner, J.M. Isolation
Clement, D.; Creager, M.; Jaff, M.; Mohler, E., 3rd .; Rutherford, of putative progenitor endothelial cells for angiogenesis. Science,
R.B.; Sheehan, P.; Sillesen, H.; Rosenfield, K. Inter-society 1997, 275(5302), 964-967.
consensus for the management of peripheral arterial disease [TASC [24] Zampetaki, A.; Kirton, J.P.; Xu, Q. Vascular repair by endothelial
II]. Eur. J. Vasc. Endovasc. Surg., 2007, 33(Suppl. 1), S1-S75. progenitor cells. Cardiovasc. Res., 2008, 78(3), 413-421.
Adult Stem Cells and the Clinical Arena Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 107

[25] Yoder, M.C.; Mead, L.E.; Prater, D.; Krier, T.R.; Mroueh, K.N.; Autologous peripheral blood mononuclear cell implantation for
Li, F.; Krasich, R.; Temm, C.J.; Prchal, J.T.; Ingram, D.A. patients with peripheral arterial disease improves limb ischemia.
Redefining endothelial progenitor cells via clonal analysis and Circ. J., 2005, 69(10), 1260-1265.
hematopoietic stem/progenitor cell principals. Blood, 2007, 109(5), [40] Durdu, S.; Akar, A.R.; Arat, M.; Sancak, T.; Eren, N.T.; Ozyurda,
1801-1809. U. Autologous bone-marrow mononuclear cell implantation for
[26] Jeong, J.O.; Kim, M.O.; Kim, H.; Lee, M.Y.; Kim, S.W.; Ii, M.; patients with Rutherford grade II-III thromboangiitis obliterans. J.
Lee, J.U.; Lee, J.; Choi, Y.J.; Cho, H.J.; Lee, N.; Silver, M.; Vasc. Surg., 2006, 44(4), 732-739.
Wecker, A.; Kim, D.W.; Yoon, Y.S. Dual angiogenic and [41] Miyamoto, K.; Nishigami, K.; Nagaya, N.; Akutsu, K.; Chiku, M.;
neurotrophic effects of bone marrow-derived endothelial progenitor Kamei, M.; Soma, T.; Miyata, S.; Higashi, M.; Tanaka, R.;
cells on diabetic neuropathy. Circulation, 2009, 119(5), 699-708. Nakatani, T.; Nonogi, H.; Takeshita, S. Unblinded pilot study of
[27] Cho, H.J.; Lee, N.; Lee, J.Y.; Choi, Y.J.; Ii, M.; Wecker, A.; Jeong, autologous transplantation of bone marrow mononuclear cells in
J.O.; Curry, C.; Qin, G.; Yoon, Y.S. Role of host tissues for sustained patients with thromboangiitis obliterans. Circulation, 2006,
humoral effects after endothelial progenitor cell transplantation into 114(24), 2679-2684.
the ischemic heart. J. Exp. Med., 2007, 204(13), 3257-3269. [42] Kawamura, A.; Horie, T.; Tsuda, I.; Abe, Y.; Yamada, M., Egawa,
[28] Rehman, J.; Li, J.; Orschell, C.M.; March, K.L. Peripheral H.; Iida, J.; Sakata, H.; Onodera, K.; Tamaki, T.; Furui, H.; Kukita,
blood “endothelial progenitor cells” are derived from monocyte/ K.; Meguro, J.; Yonekawa, M.; Tanaka, S. Clinical study of
macrophages and secrete angiogenic growth factors. Circulation, therapeutic angiogenesis by autologous peripheral blood stem cell
2003, 107(8), 1164-1169. (PBSC) transplantation in 92 patients with critically ischemic
[29] Gnecchi, M.; Zhang, Z.; Ni, A.; Dzau, V.J. Paracrine mechanisms limbs. J. Artif. Organs, 2006, 9(4), 226-233.
in adult stem cell signaling and therapy. Circ. Res., 2008, 103(11), [43] Bartsch, T.; Brehm, M.; Zeus, T.; Kögler, G.; Wernet, P.; Strauer,
1204-1219. B.E. Transplantation of autologous mononuclear bone marrow
[30] Tateishi-Yuyama, E.; Matsubara, H.; Murohara, T.; Ikeda, U.; stem cells in patients with peripheral arterial disease (the TAM-
Shintani, S.; Masaki, H.; Amano, K.; Kishimoto, Y.; Yoshimoto, PAD study). Clin. Res. Cardiol., 2007, 96(12), 891-899.
K.; Akashi, H.; Shimada, K.; Iwasaka, T.; Imaizumi, T. Therapeutic [44] Huang, P.P.; Yang, X.F.; Li, S.Z.; Wen, J.C.; Zhang, Y.; Han, Z.C.
angiogenesis for patients with limb ischaemia by autologous Randomised comparison of G-CSF-mobilized peripheral blood
transplantation of bone-marrow cells: a pilot study and a mononuclear cells versus bone marrow-mononuclear cells for the
randomised controlled trial. Therapeutic Angiogenesis using Cell treatment of patients with lower limb arteriosclerosis obliterans.
Transplantation (TACT) Study Investigators. Lancet, 2002, Thromb. Haemost., 2007, 98(6), 1335-1342.
360(9331), 427-435. [45] Kajiguchi, M.; Kondo, T.; Izawa, H.; Kobayashi, M.; Yamamoto,
[31] Esato, K.; Hamano, K.; Li, T.S.; Furutani, A.; Seyama, A.; Takenaka, K.; Shintani, S.; Numaguchi, Y.; Naoe, T.; Takamatsu, J.; Komori,
H.; Zempo, N. Neovascularization induced by autologous K.; Murohara, T. Safety and efficacy of autologous progenitor cell
bone marrow cell implantation in peripheral arterial disease. Cell transplantation for therapeutic angiogenesis in patients with critical
Transplant., 2002, 11(8), 747-752. limb ischemia. Circ. J., 2007, 71(2), 196-201.
[32] Saigawa, T.; Kato, K.; Ozawa, T.; Toba, K.; Makiyama, Y.; [46] Hernández, P.; Cortina, L.; Artaza, H.; Pol, N.; Lam, R.M.;
Minagawa, S.; Hashimoto, S.; Furukawa, T.; Nakamura, Y.; Dorticós, E.; Macías, C.; Hernández, C.; del Valle, L.; Blanco, A.;
Hanawa, H.; Kodama, M.; Yoshimura, N.; Fujiwara, H.; Namura, Martínez, A.; Díaz, F. Autologous bone-marrow mononuclear cell
O.; Sogawa, M.; Hayashi, J.; Aizawa, Y. Clinical application of implantation in patients with severe lower limb ischaemia: a
bone marrow implantation in patients with arteriosclerosis comparison of using blood cell separator and Ficoll density
obliterans, and the association between efficacy and the number of gradient centrifugation. Atherosclerosis, 2007, 194(2), e52-e56.
implanted bone marrow cells. Circ. J., 2004, 68(12), 1189-1193. [47] Saito, S.; Nishikawa, K.; Obata, H.; Goto, F. Autologous bone
[33] Higashi, Y.; Kimura, M.; Hara, K.; Noma, K.; Jitsuiki, D.; marrow transplantation and hyperbaric oxygen therapy for patients
Nakagawa, K.; Oshima, T.; Chayama, K.; Sueda, T.; Goto, C.; with thromboangiitis obliterans. Angiology, 2007, 58(4), 429-434.
Matsubara, H.; Murohara, T.; Yoshizumi, M. Autologous bone- 48] Matoba, S.; Tatsumi, T.; Murohara, T., Imaizumi, T.; Katsuda, Y.;
marrow mononuclear cell implantation improves endothelium- Ito, M.; Saito, Y.; Uemura, S.; Suzuki, H.; Fukumoto, S.;
dependent vasodilation in patients with limb ischemia. Circulation, Yamamoto, Y.; Onodera, R.; Teramukai, S.; Fukushima, M.;
2004, 109(10), 1215-1218. Matsubara, H. Long-term clinical outcome after intramuscular
[34] Miyamoto, M.; Yasutake, M.; Takano, H.; Takagi, H.; Takagi, G.; implantation of bone marrow mononuclear cells (Therapeutic
Mizuno, H.; Kumita, S.; Takano, T. Therapeutic angiogenesis by Angiogenesis by Cell Transplantation [TACT] trial) in patients
autologous bone marrow cell implantation for refractory chronic with chronic limb ischemia. TACT Follow-up Study Investigators.
peripheral arterial disease using assessment of neovascularization Am. Heart J., 2008, 156(5), 1010-1018.
by 99mTc-tetrofosmin (TF) perfusion scintigraphy. Cell. [49] Gu, Y.Q.; Zhang, J.; Guo, L.R.; Qi, L.X.; Zhang, S.W.; Xu, J.; Li,
Transplant., 2004, 13(4), 429-437. J.X.; Luo, T.; Ji, B.X.; Li, X.F.; Yu, H.X.; Cui, S.J.; Wang, Z.G.
[35] Huang, P.P.; Li, S.Z.; Han, M.Z.; Xiao, Z.J.; Yang, R.C.; Qiu, Transplantation of autologous bone marrow mononuclear cells for
L.G.; Han, Z.C. Autologous transplantation of peripheral blood patients with lower limb ischemia. Chin. Med. J. (Engl.), 2008,
stem cells as an effective therapeutic approach for severe 121(11), 963-967.
arteriosclerosis obliterans of lower extremities. Thromb. Haemost., [50] Chochola, M.; Pytlík, R.; Kobylka, P.; Skalická, L.; Kideryová, L.;
2004, 91(3), 606-609. Beran, S.; Varejka, P.; Jirát, S.; Køivánek, J.; Aschermann, M.;
[36] Kawamura, A.; Horie, T.; Tsuda, I.; Ikeda, A.; Egawa, H.; Linhart, A. Autologous intra-arterial infusion of bone marrow
Imamura, E.; Iida, J.; Sakata, H.; Tamaki, T.; Kukita, K.; Meguro, mononuclear cells in patients with critical leg ischemia. Int.
J.; Yonekawa, M.; Kasai, M. Prevention of limb amputation Angiol., 2008, 27(4), 281-290.
in patients with limbs ulcers by autologous peripheral blood [51] Wester, T.; Jørgensen, J.J.; Stranden, E.; Sandbaek, G.; Tjønnfjord,
mononuclear cell implantation. Ther. Apher. Dial., 2005, 9(1), G.; Bay, D.; Kollerøs, D.; Kroese, A.J.; Brinchmann, J.E.
59-63. Treatment with autologous bone marrow mononuclear cells in
[37] Lenk, K.; Adams, V.; Lurz, P.; Erbs, S.; Linke, A.; Gielen, S.; patients with critical lower limb ischaemia. A pilot study. Scand. J.
Schmidt, A.; Scheinert, D.; Biamino, G.; Emmrich, F.; Schuler, G.; Surg., 2008, 97(1), 56-62.
Hambrecht, R. Therapeutical potential of blood-derived progenitor [52] Van Tongeren, R.B.; Hamming, J.F.; Fibbe, W.E.; Van Weel, V.;
cells in patients with peripheral arterial occlusive disease and Frerichs, S.J.; Stiggelbout, A.M.; Van Bockel, J.H.; Lindeman,
critical limb ischaemia. Eur. Heart J., 2005, 26(18), 1903-1909. J.H. Intramuscular or combined intramuscular/intra-arterial
[38] Huang, P.; Li, S.; Han, M.; Xiao, Z.; Yang, R.; Han, Z.C. administration of bone marrow mononuclear cells: a clinical trial in
Autologous transplantation of granulocyte colony stimulating patients with advanced limb ischemia. J. Cardiovasc. Surg., 2008,
factor-mobilized peripheral blood mononuclear cells improves 49(1), 51-58.
critical limb ischemia in diabetes. Diabetes Care, 2005, 28(9), [53] De Vriese, A.S.; Billiet, J.; Van Droogenbroeck, J.; Ghekiere, J.;
2155-2160. De Letter, J.A. Autologous transplantation of bone marrow
[39] Ishida, A.; Ohya, Y.; Sakuda, H.; Ohshiro, K.; Higashiuesato, Y.; mononuclear cells for limb ischemia in a caucasian population with
Nakaema, M., Matsubara, S.; Yakabi, S.; Kakihana, A.; Ueda, M.; atherosclerosis obliterans. J. Intern. Med., 2008, 263(4), 395-403.
Miyagi, C.; Yamane, N.; Koja, K.; Komori, K.; Takishita, S.
108 Cardiovascular & Hematological Agents in Medicinal Chemistry, 2012, Vol. 10, No. 2 Casamassimi et al.

[54] Cobellis, G.; Silvestroni, A.; Lillo, S.; Sica, G.; Botti, C.; Maione, [60] Zafarghandi, M.R.; Ravari, H.; Aghdami, N.; Namiri, M.;
C.; Schiavone, V.; Rocco, S.; Brando, G.; Sica, V. Long-term Moazzami, K.; Taghiabadi, E.; Fazel, A.; Pournasr, B.; Farrokhi, A.;
effects of repeated autologous transplantation of bone marrow cells Sharifian, R.A.; Salimi, J.; Moini, M.; Baharvand, H. Safety and
in patients affected by peripheral arterial disease. Bone Marrow efficacy of granulocyte-colony-stimulating factor administration
Transplant., 2008, 42(10), 667-672. following autologous intramuscular implantation of bone marrow
[55] Amann, B.; Lüdemann, C.; Rückert, R.; Lawall, H.; Liesenfeld, B.; mononuclear cells: a randomized controlled trial in patients
Schneider, M.; Schmidt-Lucke, J. Design and rationale of a with advanced lower limb ischemia. Cytotherapy, 2010, 12(6),
randomized, double-blind, placebo-controlled phase III study for 783-791.
autologous bone marrow cell transplantation in critical limb [61] Procházka, V.; Gumulec, J.; Jal vka, F.; Salounová, D.; Jonszta,
ischemia: the Bone Marrow Outcomes Trial in Critical Limb T.; Czern, D.; Kraja, J.; Urbanec, R.; Klement, P.; Martinek, J.;
Ischemia (BONMOT-CLI). Vasa, 2008, 37(4), 319-325. Klement, G.L. Cell therapy, a new standard in management of
[56] Amann, B.; Luedemann, C.; Ratei, R.; Schmidt-Lucke, J.A. chronic critical limb ischemia and foot ulcer. Cell Transplant.,
Autologous bone marrow cell transplantation increases leg 2010, 19(11), 1413-1424.
perfusion and reduces amputations in patients with advanced [62] Lara-Hernandez, R.; Lozano-Vilardell, P.; Blanes, P.; Torreguitart-
critical limb ischemia due to peripheral artery disease. Cell. Mirada, N.; Galmés, A.; Besalduch, J. Safety and efficacy of
Transplant., 2009, 18(3), 371-380. therapeutic angiogenesis as a novel treatment in patients with
[57] Capiod, J.C.; Tournois, C.; Vitry, F.; Sevestre, M.A.; Daliphard, S.; critical limb ischemia. Ann. Vasc. Surg., 2010, 24(2), 287-294.
Reix, T.; Nguyen, P.; Lefrère, J.J.; Pignon, B. Characterization and [63] Walter, D.H.; Krankenberg, H.; Balzer, J.O.; Kalka, C.;
comparison of bone marrow and peripheral blood mononuclear Baumgartner, I.; Schlüter, M.; Tonn, T.; Seeger, F.; Dimmeler, S.;
cells used for cellular therapy in critical leg ischaemia: towards a Lindhoff-Last, E.; Zeiher, A.M. Intraarterial administration of bone
new cellular product. Vox Sang., 2009, 96(3), 256-265. marrow mononuclear cells in patients with critical limb ischemia: a
[58] Franz, R.W.; Parks, A.; Shah, K.J.; Hankins, T.; Hartman, J.F.; randomized-start, placebo-controlled pilot trial (PROVASA).
Wright, M.L. Use of autologous bone marrow mononuclear PROVASA Investigators. Cardiovasc. Interv., 2011, 4(1), 26-37.
cell implantation therapy as a limb salvage procedure in patients [64] Murphy, M.P.; Lawson, J.H.; Rapp, B.M.; Dalsing, M.C.; Klein, J.;
with severe peripheral arterial disease. J. Vasc. Surg., 2009, 50(6) Wilson, M.G.; Hutchins, G.D.; March, K.L. Autologous bone
1378-1390. marrow mononuclear cell therapy is safe and promotes amputation-
[59] Franz, R.W.; Shah, K.J.; Johnson, J.D.; Pin, R.H.; Parks, A.M.; free survival in patients with critical limb ischemia. J. Vasc. Surg.,
Hankins, T.; Hartman, J.F.; Wright, M.L. Short- to mid-term results 2011, 53(6) 1565-1574.
using autologous bone-marrow mononuclear cell implantation [65] Yoshida, M.; Horimoto, H.; Mieno, S.; Nomura, Y.; Okawa, H.;
therapy as a limb salvage procedure in patients with severe Nakahara, K.; Sasaki, S. Intra-arterial bone marrow cell
peripheral arterial disease. Vasc. Endovascular Surg., 2011, 45(5), transplantation induces angiogenesis in rat hindlimb ischemia. Eur.
398-406. Surg. Res., 2003, 35(2) 86-91.

Received: December 06, 2011 Revised: January 19, 2012 Accepted: January 23, 2012

View publication stats

Das könnte Ihnen auch gefallen