Вы находитесь на странице: 1из 8

ADR-12708; No of Pages 8

Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


journal homepage: www.elsevier.com/locate/addr

Stem cell treatment of erectile dysfunction☆,☆☆


Amjad Alwaal a,b,⁎, Uwais B. Zaid a, Ching-Shwun Lin a, Tom F. Lue a
a
Department of Urology, University of California, San Francisco, CA, USA
b
King Abdul Aziz University, Jeddah, Saudi Arabia

a r t i c l e i n f o a b s t r a c t

Article history: Erectile Dysfunction (ED) is a common disease that typically affects older men. While oral type-5 phosphodieserase
Accepted 8 November 2014 inhibitors (PDE5Is) represent a successful first-line therapy, many patients do not respond to this treatment leading
Available online xxxx researchers to look for alternative treatment modalities. Stem cell (SC) therapy is a promising new frontier for the
treatment of those patients and many studies demonstrated its therapeutic effects. In this article, using a Medline
Keywords:
database search of all relevant articles, we present a summary of the scientific principles behind SCs and their
Erectile dysfunction
Stem cell therapy
use for treatment of ED. We discuss specifically the different types of SCs used in ED, the methods of delivery tested,
and the methods attempted to enhance SC therapy effect. In addition, we review the current preclinical literature on
SC therapy for ED and present a summary of its findings in addition to the single clinical trial published.
© 2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.1. ED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.2. Mechanism of erection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.3. Aging and ED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.4. Metabolic syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.5. Prostate cancer therapy-related ED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
1.6. Peyronie's disease (PD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. Types of stem cells used in ED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.1. ESC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2. Endothelial progenitor stem cells (EPSCs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3. Bone marrow-derived stem cell (BMSC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4. Skeletal muscle-derived stem cell (SKMSC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.5. Neural crest SCs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.6. Adipose tissue-derived stem cell (ADSC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.7. Testis SC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.8. Human urine SC (USC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Methods of SC delivery for ED treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. Methods to enhance the therapeutic effects of SCs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5. Assessment of outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
6. Clinical trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7. Immunocompatibilty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
8. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Regenerative Medicine Strategies in Urology”
☆☆ Author disclosure statement: No competing financial interests exist.
⁎ Corresponding author at: University of California, San Francisco, Box 0738, 1600 Divisadero St., MZ Bldg A, Room A609, San Francisco, CA 94143, USA. Tel.: +1 415 885 7748; fax: +1
415 885 7443.
E-mail address: amjadwal@yahoo.com (A. Alwaal).

http://dx.doi.org/10.1016/j.addr.2014.11.012
0169-409X/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
2 A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

1. Introduction Diabetics are 3 times more likely than non-diabetics to have ED [5],
with a prevalence of 50–75% [6]. ED affects diabetics 10–15 years earlier
1.1. ED than non-diabetics [5], and PDE5Is are less efficacious in this population
[20]. The mechanism by which DM causes ED is multifactorial and there
It is an important health problem that significantly impacts the are different findings and proposed mechanisms for its occurrence. DM
patient's quality of life and can have a detrimental effect on his well- was found to diminish the amount of NO-releasing penile nerves [21,
being and relationship with his partner [1]. ED is defined as the inability 22], cavernosal endothelial cells, and cavernosal smooth muscle content
to achieve or maintain a penile erection satisfactory for sexual inter- [22,23]. Ning et al. [24,25] recently demonstrated that hyperglycemia
course [2]. ED is estimated to affect 20% of men above 40 years of age, induced mitochondrial fragmentation and cellular apoptosis of endo-
with the incidence increasing with increasing age [3], in addition to thelial cells. Hyperglycemia is also associated with smooth muscle dys-
other factors such as better diagnosis and increased awareness of the function through oxidation of low-density lipoproteins and excessive
disease [4]. Men between the age of 61–70 years are twice as likely to production of oxygen free radicals. DM also impairs vascular endothelial
be affected than men aging 51–60 years [5], with 67% of men at age growth factor (VEGF) signaling and upregulates the RhoA/RhoKinase
70 years affected with the disease [6]. Another important reason for pathway [26–28]. It also reduces the expression and activity of neuronal
the rise in ED is the rapidly growing prevalence of diabetes mellitus nitric oxide synthase (nNOS) [29–31]. Given the penile tissue damage
(DM) [7], which is a major risk factor for ED [4]. Several treatment associated with DM and the negative impact of ED on the quality of
modalities exist for ED, including oral PDE5Is, intraurethral alprostadil life in diabetics [32], SC therapy holds a great promise to treat DM-
suppository, intracorporal (IC) injection of erectogenic medications, associated ED due to SCs' regenerative ability and their potential to re-
vacuum device, and penile implant. Among these modalities, PDE5Is re- store cavernosal endothelial and smooth muscle cells. Hyperlipidemia
main the most widely used and have great success rates, mainly due to is another metabolic factor responsible for ED [33]. It was found that
their ease of administration as oral medications and proven efficacy. for every 1 mmol/L increase in serum total cholesterol there was a
However, despite such successes, there are several limitations to their 32% increased risk of ED, and for every 1 mmol/L increase in high-
use. For example, they are contraindicated in some patients who take density lipoprotein (HDL) there was a marked reduction in the risk of
nitrates due to the risk of developing severe hypotension; some men ED [34]. Hyperlipidemia causes ED through neuronal and endothelial
cannot tolerate their side effects; they are only partially effective in cer- dysfunction, leading to a reduction in cavernosal NO levels [35–37].
tain patients; and they have a considerable financial cost [8]. In addition,
they provide only symptomatic relief of ED and do not offer a cure 1.5. Prostate cancer therapy-related ED
for the disease. Therefore, there is a growing interest in developing
therapies that offer a cure for the disease, including gene therapy and Prostate cancer is the most commonly diagnosed cancer in US men
SC therapy [9,10]. [38]. Earlier detection and treatment have allowed the widespread use
of localized treatment options in about 80% prostate cancer patients
1.2. Mechanism of erection [38], including radical prostatectomy (RP) and radiation therapy (RT)
[39]. However, these treatment options carry a significant risk of post-
Penile erection occurs through a nitric oxide (NO)-mediated mecha- treatment ED [40]; for example, RP is associated with 60.8–93.9% risk
nism. NO, the main neurotransmitter involved in erection, is released by of ED [41]. The most widely accepted mechanism for post-RP ED is inju-
endothelial cells and non-adrenergic non-cholinergic nerve terminals in ry to the cavernosal nerve fibers that run along the posterolateral aspect
the cavernosal tissue. It causes relaxation of the cavernosal smooth of the prostate. Nerve-sparing RP has been introduced in order to pre-
muscles through a cGMP-mediated reduction of intracellular calcium, serve the cavernosal nerves. However, despite the continued refine-
after which cGMP gets degraded by PDE5 [11]. Once the lacunar spaces ment in the surgical technique, nerve-sparing RP is still associated
are filled with blood, they compress the subtunical venules, resulting in with nearly 20% risk of ED at 24 months post-op [42]. This is likely
erection. Detumescence occurs when adrenergic receptors are activat- due to neurapraxia, in which stretching, heating, or trauma to the
ed, inducing contraction of the cavernosal smooth muscles with resul- cavernosal nerve fibers during surgery induce Wallerian degeneration
tant diminished blood in-flow and reduced size of the lacunar spaces, [43]. Alternatively, NO-releasing nerves stop producing NO during the
which in turn allows more venous out-flow through the subtunical ve- period of neurapraxia, resulting in cavernosal smooth muscle apoptosis
nules [12]. and fibrosis. In support of this theory is the reduction of smooth muscle
content and subsequent fibrosis in post-RP penile tissue [43,44]. RT has
1.3. Aging and ED been much less studied than RP, but generally from the limited data
published, it is likely that post-RT ED follows the same mechanism.
Different diseases and conditions cause ED through different mech- That is, cavernosal nerve injury followed by smooth muscle degenera-
anisms. Aging results in ED through increased penile vascular tone [13] tion [23,45]. PDE5Is are being prescribed routinely in many centers
and inactivation of endothelial nitric oxide synthase (eNOS), the latter post-RP as part of the “penile rehabilitation protocol”. The exact mech-
of which is accomplished by decreased phosphorylation of eNOS anism for their effect is not known, as PDE5 sits downstream of NO,
positive regulatory site and increased phosphorylation of its negative which cannot be produced by damaged nerves post-RP. Thus, the
regulatory site [14]. In addition, it reduces nNOS nerve fibers, thereby exact mechanism for PDE5Is' effects in post-RP ED patients remains to
diminishing the erectile response to cavernosal nerve stimulation and be proven, and so does their true effect in this population [44,46].
reducing penile reflexes [15]. Aging also decreases NO bioavailability
by increasing reactive oxygen species (ROS). When ROS are in excess, 1.6. Peyronie's disease (PD)
superoxide anion interacts with NO to cause endothelial dysfunction
[16,17]. Aging was also found to be associated with abundance of colla- PD affects 3% of men, resulting in pain and penile deformities such as
gen fibers, reduction of smooth muscle cells, and degenerative changes curvature, indentation, and shortening [47]. Although further research
in elastic fibers [18]. is needed, it is thought that PD causes ED directly. Among several
mechanisms that have been proposed to describe this effect [48],
1.4. Metabolic syndrome veno-occlusive dysfunction resulting from the PD plaque is the most
commonly accepted although further research is needed to prove this
The prevalence of DM in the US is 8.3% of the population, with 26.9% theory [49]. The severity of PD occasionally necessitates incising or ex-
of the population 65 years or older affected with DM in 2010 [19]. cising the plaque followed by patch grafting the tunica albuginea (TA)

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 3

to maintain the length of the penis [50], and this can further increase the The SC niche is a microenvironment that supports SCs in their quies-
risk of ED [51]. Several strategies have been proposed for treatment of cent state and promote SC self-renewal or tissue regeneration upon tis-
PD including the use of long-term PDE5Is as anti-fibrotic strategy [52]. sue damage [66]. Niche cells can be classified as epithelial or stromal
Recent advances in SC research allowed the utilization of porcine depending on their location and proximity to SCs [66]. SC characteristics
small intestinal submucosa (SIS) with seeded SCs to reduce the risk of within the niche are influenced by many factors, such as cell–cell inter-
ED [53]. Another recent development is the use of SCs in transforming action between SCs and their neighboring cells, interaction with sur-
growth factor (TGF)-induced PD rat model to study the anti-fibrosis rounding adherent molecules and the extracellular matrix, presence of
effect of SCs in PD [54,55]. These are promising new strategies in the cytokines and other growth factors, and oxygen tension and other
treatment of PD and its associated ED. Table 1 summarizes some of physiochemical properties. The SC niche is usually present in the
the ED etiologies and their proposed mechanism. perivascular space, which allows direct access to the blood stream
In this article, we aimed at identifying the different types of SCs in when damaged tissues are in need of SCs for their regenerative poten-
the treatment of ED as well as their routes of delivery, modifications tial. SCs in their niche microenvironment were found to be in a state
(e.g. gene modifications), clinical applications, and possible future use. of relative hypoxia with oxygen tension as low as 2% [67,68], thereby
Using the terms “stem cell” and “erectile dysfunction”, we performed possibly shaping their unique characteristics and enhancing the produc-
a search of the Medline database for all the English language articles tion of growth factors leading some researchers to replicate this niche
that have been published from 2001 to present. Additional articles environment in vitro [69].
were identified through examination of citations.
2.1. ESC
2. Types of stem cells used in ED
ESCs are pluripotent cells that had been studied only once in ED re-
SCs have the potential to divide indefinitely with the daughter cells search. Bochinski et al. was the first study investigating the use of SCs in
having the ability to either become a SC or differentiate into a special- ED, and it investigated the effect of ESCs on neurogenic ED induced by
ized cell type, such as brain cell, red blood cell, or muscle cell [56,57]. cavernosal nerve injury and demonstrated improved erectile function
Their self-renewal capacity and ability to regenerate damaged tissues when ESCs were injected into the corpora cavernosa or major pelvic
depend on the various surrounding stimuli and factors [56,57]. They ganglia (MPG) [70]. Given the ethical concerns mentioned earlier, no
are classified into (totipotent, pluripotent, multipotent, or unipotent) further studies had been done using such cells.
depending on their hierarchical differentiation potential [58]. Totipo-
tent SCs, like the zygote and its morula daughter cells, have the greatest 2.2. Endothelial progenitor stem cells (EPSCs)
differentiation potential and can divide into any tissue type originating
from the ectoderm, mesoderm, and endoderm tissues [59]. Pluripotent They are bone marrow-derived cells that were discovered in 1997
cells give rise to the three germ cell layers but not to extra-embryonic [71]. When peripheral blood mononuclear cells (PBMCs) are enriched
tissues. The most widely acknowledged example of pluripotent SC is by selection for CD34 expression, they exhibit mature endothelial cell-
the embryonic SC (ESC) derived from the inner cell mass of the blasto- like characteristics and display several endothelial markers such as
cyst [60,61]. Multipotent SCs, such as the mesenchymal SC (MSC), are CD31, CD34, VEGFR2, Tie-2, and eNOS. In addition, they form vessel-
isolated from the developing germ layer or its developed adult organ like structures on matrigel [71]. This discovery has generated promise
[59]. Unipotent SCs are precursor cells within the developed tissue for using EPSCs in angiogenic therapy. However, there have been de-
and can give rise to only one cell type, such as epithelial cells. They bates over the characterization of these cells with some arguing that
have limited self-renewal capacity, therefore labeling them as SCs is de- EPSCs can be recognized through their antigenic receptors [72,73]
batable [62]. while others believe that they need to be recognized by using cell be-
There have been ethical concerns regarding the use of ESCs, due to havior and growth dynamics in vitro [74]. Generally, when PBMCs are
the need to destroy embryos for their isolation, and this has heightened grown in culture, the late cell outgrowth colonies at 1–3 weeks possess
the interest in adult SCs (ASCs) as alternative SC source [62]. In addition, antigenic properties restricted to endothelial cells and are highly prolific
induced pluripotent SCs (iPSC) can be generated from somatic cells by making this subset of cells the most viable source for angiogenic therapy
overexpression of ESC-specific transcription factors [63,64]. These cells [74,75]. Most research done thus far using EPSC has been in heart failure
share some characteristics with ESCs although the exact differences be- after myocardial infarction [76]. Low circulating levels of EPSCs have
tween them have not been fully elucidated [65]. been found to be an independent predictor for ED [77]. Gou et al. [78],

Table 1
Some etiologies and proposed mechanisms for erectile dysfunction.

Etiology Summary of proposed mechanisms

Aging – Increased penile vascular tone


– Inactivation of eNOS
– Reduction of nNOS nerve fibers
– Reduction of NO bioavailability by increasing reactive oxygen species (ROS)
– endothelial dysfunction
– Abundance of collagen fibers, reduction of smooth muscle cells, and degenerative changes in elastic fibers
DM – Reduction of NO-releasing penile nerves, cavernosal endothelial cells, and cavernosal smooth muscle content
– Mitochondrial fragmentation and cellular apoptosis of endothelial cells
– Smooth muscle dysfunction through oxidation of low-density lipoproteins and excessive production of
oxygen free radicals
– Impairment of VEGF signaling
– Upregulation of the RhoA/RhoKinase pathway
– Reduction of the expression and activity of nNOS
Hyperlipidemia – Neuronal and endothelial dysfunction
– Reduction in cavernosal NO levels
Prostate cancer treatment (RP and RT) – Injury to the CN fibers followed by smooth muscle degeneration
PD – Veno-occlusive dysfunction
– Intracorporal fibrosis

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
4 A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

in the only study using EPSCs in ED, showed an improvement in rat transformation to other cell types. This has been suggested through
erectile function in streptozotocin-induced diabetic animals with ED studies showing improvement in erectile function when ADSC lysate
when they were transplanted with VEGF-transfected EPSCs. or ADSC conditioned growth medium was used [104,108]. However,
there are some problems associated with ADSCs that might make SVF
2.3. Bone marrow-derived stem cell (BMSC) more desirable to use, including the longer process of isolation relative
to SVF, the requirement for certain facilities and personnel, and possible
These are adult SCs that are considered the prototype for MSCs [10]. alteration of cellular characteristics [109].
They have pluripotent ability to differentiate into different tissues and
cells [79] and they secrete many cytokines that have trophic effects on 2.7. Testis SC
cytoprotection, cell survival, and immunomodulation [80]. They are
characterized by their ease of isolation and ex vivo expansion [81]. Choi et al. in 2013 [110], isolated CD34+/CD73+-double-positive
MSCs do not elicit an immune response due to their lack of surface co- testicular stromal cells (HTSCs). These cells demonstrated the ability
stimulatory molecules required for T cell activation, thus eliminating to proliferate for extended periods of time without forming tumors
the need for immunosuppression [82]. Several pre-clinical studies in vivo. They gave rise to all three germ cell lines, and more importantly,
have shown the beneficial effect of BMSCs on erectile function in differ- restored erections in a CN injury rat model without forming teratomas.
ent rat models such as DM [83–86], cavernous nerve crush injury
[87–90], and aging [81,91].
2.8. Human urine SC (USC)
2.4. Skeletal muscle-derived stem cell (SKMSC)
There has been one preclinical trial looking at USCs in the treatment
SKMSCs are another group of pluripotent MSCs. They were exten- of ED [111]. USCs are pluripotent SCs that have been shown to differen-
sively studied because of their prolonged proliferation, low immunoge- tiate into endothelial and smooth muscle cells. They have the advantage
nicity, and ability to convert to several cell lineages when implanted of being easy to obtain. The study showed a positive response on erectile
into different organs [92]. They should not be confused with satellite function in diabetic rats.
myoblasts, which are non-pluripotent and have been used to treat
heart disease in clinical trials as they can give rise to cardiomyocytes 3. Methods of SC delivery for ED treatment
[93]. SKMSCs can be easily obtained through muscle biopsy [94] and
have been shown to improve erectile function in a bilateral cavernous Intracavernosal (IC) SC delivery is by far the most common SC deliv-
injury ED rat model [95]. Nolazco et al. demonstrated their ability to ery route for ED treatment, given its already proven success and ease of
transform to SMs in penile tissue in addition to improving erectile func- administration [10]. In the CN injury rat models, the damage incited is
tion in aged rat models [96]. Woo et al. also confirmed such findings in a not in the cavernosa but rather in the axonal fibers, with the neurons lo-
cavernous nerve injury rat model and showed survival of these cells in cated in the MPG. By injecting into the corporal tissue, the SCs exert
the penile tissue [97]. their regenerative effect on the MPG by either secreting soluble factors
into the blood stream or by migrating to the MPG [10]. Our experiments
2.5. Neural crest SCs have demonstrated that SCs migrate to the MPG after IC injection [112,
113]. SC injection whether immediately at the time of CN injury or 4
Another form of pluriopotent SC is neural crest SC. These cells are weeks after injury had similar positive effects on erectile function
considered the progenitors of the peripheral nervous system, giving [103]. Injecting SCs directly into the MPG has also been tried and dem-
rise to neurons, Schwann cells, and adrenal chromaffin cells [98]. Song onstrated regenerative effects, however this route has not been exten-
et al. in 2007 demonstrated the ability of these cells to differentiate sively studied because of the technical difficulties involved and
into smooth muscle cells and endothelial cells in the rat penis [99]. To similarly positive results obtained with direct penile injection [70,89].
our knowledge, no studies have been conducted to test their therapeu- Application of SCs directly onto injured CNs followed by coverage
tic potential for ED. with poly(lactic-co-glycolic acid) (PLGA) or hydrogel has also been
studied. While recovery of erectile function was demonstrated, no di-
2.6. Adipose tissue-derived stem cell (ADSC) rect comparison to the IC method was done; therefore, we do not
know the relative efficacy of such approaches [114–117]. Three studies
ADSCs are pluripotent SCs that are considered the most widely used have been done by injecting SCs into the periprostatic tissue with or
type of SCs in ED research given that they are easy to obtain from abun- without simultaneous IC injection [90,110,118]. In one of these studies,
dant tissue sources [10]. There are two populations of SCs derived from all the groups showed similar recovery of erectile function, whether IC,
adipose tissue that have been used in ED and non-ED research. The ad- periprostatic, or both [118]. Recently, Ryu et al. demonstrated that intra-
ipose derived-stromal vascular fraction (AD-SVF) cells, which are de- peritoneal injection of SCs was less effective than IC injection in restor-
rived from the pellet fraction of collagenase-digested adipose tissue ing erectile function in CN injury mouse model [119].
homogenate [100], and ADSCs, which are derived from SVF and are ad- Our group has previously attempted to use adipose-derived acellular
herent to the culture dish and then further propagated [100]. One gram matrix seeded with ADSCs as nerve graft. While there was some recov-
of fat yields 250,000 SVF cells, and of those cells only 2% qualify as ADSC ery of erectile function, the results did not reach a statistical significance
[101]. SVF cells will likely be more commercially available than ADSC [120]. Further technical refinement is needed, but given the good re-
due to the availability of machines for automated isolation of SVF cells sponse with IC injection, this may not be necessary. We also previously
[102]. AD-SVF cells enhance angiogenesis by differentiating into endo- suggested that the effect seen with SC injection might be the result of SC
thelial cells or through paracrine effects by releasing growth factors homing to the injury site [121], and we have proven the beneficial effect
thereby enhancing erectile function [103,104]. ADSC on the other of intravenous ADSC injection on erectile function [121]. Tracking of the
hand demonstrated an improvement in erectile function in pre- injected cells showed that they migrate into the MPG in post-RT ED rats
clinical trials [30] through direct transformation to endothelial cells, [121]. This suggests that the intravenous route may be another valuable
smooth muscle cells, and neurons [105] as well as through release of method for SC delivery. Recently, Ying et al. demonstrated an improve-
stimulatory cytokines [106] such as VEGF and fibroblast growth factor ment in the erectile function of CN injury rat model when autologous
(FGF) [104,107]. It is likely that the erectile function improvement effect saphenous vein grafts were interposed at the nerve stumps and ADSCs
seen with ADSCs is due to cytokine release rather than the direct injected into the vein graft [122].

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 5

In regard to SC treatment for PD-associated ED, reconstruction of the sildenafil to MDSC application did not have any additive effect in
TA, with ADSC-seeded SIS resulted in better erectile function than preventing or reversing corporal veno-occlusive disease in the CN injury
with unseeded SIS [53]. In another study that simulates PD by using model [126]. FGF-transfected USCs had similar effect to USCs alone in
intratunical TGF injection, less TA fibrosis and better erectile function improving erectile function [111].
were observed when ADSC, as compared to saline, was injected into
the injured TA [54]. Similar results were obtained in a more recent
study [123]. 5. Assessment of outcome

The response to SC ED treatment in preclinical studies involves func-


4. Methods to enhance the therapeutic effects of SCs tional and histological assessment. The most widely used method is
measure the intracavernosal pressure (ICP) upon electrostimulation of
In order to boost the therapeutic potential of SC therapy, several pre- the cavernosal nerves. Typically, 1.5 mA, 20 Hz, and 0.2 ms pulse
clinical trials attempted to improve SC performance by genetically alter- width would result in elevation of ICP from 20 cm H2O to 100 cm H2O
ing their intrinsic characteristics or by combining them with scaffolds, in normal rats approaching systemic blood pressure, while in ED rats
growth factors, medications, etc. Two studies showed an improvement ICP is around 30 cm H2O. An elevation of ICP to above 70 cm H2O
in erectile function with increased endothelial and smooth muscle would be considered an indication of successful treatment. Alternative-
markers when VEGF-transfected ADSCs were injected in diabetic rats ly, ICP can be normalized to mean arterial pressure (MAP) in order to
[84,124]. The effect was greater than when either ADSC or VEGF was measure erectile function, which is around 0.8 (ICP/MAP) in normal
used alone [84,124]. In another study, BMSCs were transfected with rats.
KCNMA1, a gene involved in cellular excitement and neurotransmitter This method requires laparotomy, therefore it is usually done at the
release [85]. The results showed improved erectile function in diabetic time of sacrificing the animal for tissue harvesting, which is 4 weeks
rats [85]. As mentioned earlier, ADSC with PLGA membrane [114,116] post-SC transplantation. When tissues are harvested, they are examined
or hydrogel [115] improved angiogenesis and erectile function in CN in- using immunohistochemistry (IH) and immunofluorescence (IF) for lo-
jury model either alone or in combination with oral udenafil [84,116]. cation of SCs, correlation of their function with the function, and/or
Matrixen was also combined with BMSCs to increase their implantation presence of SC differentiation. In order to identify SCs, pre-labeling is
and was found to augment erectile function [125]. Adding low-dose typically done [10].

Table 2
Preclinical trials for stem cell treatment of erectile dysfunction.

Trial year First author Animal model Stem cell type Method of Reference
transplantation

2004 Bochinski CN injury rat Allogeneic ESC IC or Intra-MPG [70]


2006 Kim CN injury rat Allogeneic SKMSC IC [95]
2009 Fall CN injury rat Allogeneic BMSC IC [87]
2010 Albersen CN injury rat Autologous ADSC IC [108]
2010 Kendirci CN injury rat Allogeneic BMSC IC [88]
2011 Lin CN injury rat Autologous ADSC Nerve graft [120]
2011 Woo CN injury rat Allogeneic SKMSC IC [97]
2012 Qiu CN injury rat Autologous SVF IC [103]
2012 Kovanecz CN injury rat Mouse SKMSC IC [126]
2012 Kim CN injury rat Allogeneic BMSC CN scaffold [89]
2012 Fandel CN injury rat Autologous ADSC IC [112]
2012 Piao CN injury rat Human ADSC CN scaffold [114]
2013 Jeong CN injury rat Human ADSC CN scaffold [116]
2013 Kim CN injury rat Human ADSC CN scaffold [115]
2013 You CN injury rat Human BMSC IC + periprostatic [90]
2013 You CN injury rat Human ADSC IC + periprostatic [118]
2013 Choi CN injury rat Human testis SC Periprostatic [110]
2013 Ying CN injury rat Autologous ADSC IC [129]
2014 Ying CN injury rat Autologous ADSC Vein graft [122]
2014 Lee CN injury rat Human ADSC IC [117]
2014 Ryu CN injury mouse Allogenic clonal BMSC IC + IP [119]
2012 Qiu Radiation injury rat Allogeneic ADSC Intra-MPG [121]
2007 Bivalacqua Aging rat Allogeneic BMSC IC [91]
2008 Nolazco Aging rat Mouse SKMSC IC [96]
2010 Abdel Aziz Aging rat Allogeneic BMSC IC [81]
2010 Garcia DM rat Autologous ADSC IC [30]
2011 Gou DM rat Allogeneic EPC IC [78]
2011 Qiu DM rat Allogeneic BMSC IC [83]
2012 Qiu DM rat Allogeneic BMSC IC [84]
2012 Sun DM rat Allogeneic BMSC IC [86]
2012 Nishimatsu DM rat Allogeneic ADSC IC [130]
2013 He DM rat Allogeneic BMSC IC [85]
2013 Liu DM rat Human ADSC IC [124]
2014 Ouyang DM rat Human USC IC [111]
2012 Ryu DM mouse Syngeneic SVF IC [104]
2014 Das DM mouse Human SVF IC [131]
2010 Huang Hyperlipidemia rat Autologous ADSC IC [35]
2012 Ma TA injury rat Autologous ADSC SIS graft [53]
2013 Castiglione TA injury rat Human ADSC Intratunical [54]
2014 Gokce TA injury rat Autologous ADSC Intratunical [123]

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
6 A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

6. Clinical trials [5] A. Ponholzer, C. Temml, K. Mock, M. Marszalek, R. Obermayr, S. Madersbacher,


Prevalence and risk factors for erectile dysfunction in 2869 men using a validated
questionnaire, Eur. Urol. 47 (2005) 80–86.
There has been only one clinical trial assessing the efficacy of SCs for [6] R.W. Lewis, K.S. Fugl-Meyer, G. Corona, R.D. Hayes, E.O. Laumann, E.D. Moreira Jr.,
ED. Bahk et al. from Korea investigated the effect of IC injection of um- A.H. Rellini, T. Segraves, Original articles: definitions/epidemiology/risk factors for
sexual dysfunction, J. Sex. Med. 7 (2010) 1598–1607.
bilical cord SCs in 7 patients with DM [127]. This demonstrated im- [7] P.A. Zimmet, KGMM, Jonathan Shaw, Global and societal implications of the diabe-
proved erectile function and was initially promising. Clinical trials on a tes epidemic, Nature (2001) 782–787.
larger scale are needed in order to validate these results and clarify [8] A.A. Carvalheira, N.M. Pereira, J. Maroco, V. Forjaz, Dropout in the treatment of
erectile dysfunction with PDE5: a study on predictors and a qualitative analysis
their exact effects in humans. Currently, two phase I–II SC clinical trials of reasons for discontinuation, J. Sex. Med. 9 (2012) 2361–2369.
are recruiting patients with ED. In France, IC injection of BMSCs is [9] N. Yoshimura, R. Kato, M.B. Chancellor, J.B. Nelson, J.C. Glorioso, Gene therapy as fu-
being evaluated in post-RP patients (Identifier: NCT01089387), ture treatment of erectile dysfunction, Expert. Opin. Biol. Ther. 10 (2010)
1305–1314.
while in the United States the effects of IC injection of AD-SVF in organic
[10] C.-S. Lin, Z. Xin, J. Dai, Y.-C. Huang, T.F. Lue, Stem-cell therapy for erectile dysfunc-
ED (vasculogenic and neural) are being assessed (Identifier: tion, Expert. Opin. Biol. Ther. 13 (2013) 1585–1597.
NCT01601353). [11] T.F. Lue, Erectile dysfunction, N. Engl. J. Med. 342 (2000) 1802–1813.
[12] D. Prieto, Physiological regulation of penile arteries and veins, Int. J. Impot. Res. 20
(2007) 17–29.
7. Immunocompatibilty [13] T.J. Bivalacqua, M.F. Usta, H.C. Champion, P.J. Kadowitz, W.J.G. Hellstrom, Endothe-
lial dysfunction in erectile dysfunction: role of the endothelium in erectile physiol-
ogy and disease, J. Androl. 24 (2003) S17–S37.
The least immunogenic SC transplantation would require autolo- [14] B. Musicki, M.F. Kramer, R.E. Becker, A.L. Burnett, Age-related changes in phosphor-
gous SCs. However, even with some of the easier methods of SC extrac- ylation of endothelial nitric oxide synthase in the rat penis, J. Sex. Med. 2 (2005)
347–357.
tion such as in the case of ADSCs, a surgical procedure is still involved
[15] S. Carrier, P. Nagaraju, D.M. Morgan, K. Baba, L. Nunes, T.F. Lue, Age decreases nitric
and that by itself may adversely affect the outcome of SC transplanta- oxide synthase-containing nerve fibers in the rat penis, J. Urol. 157 (1997)
tion. Therefore, more recent SC preclinical studies have increasingly uti- 1088–1092.
lized allogeneic and xenogeneic SC transplantations as alternatives. [16] K.M. Azadzoi, R.N. Schulman, M. Aviram, M.B. Siroky, Oxidative stress in
arteriogenic erectile dysfunction: prophylactic role of antioxidants, J. Urol. 174
Allogeneic and xenogeneic SCs do not incite immune response because (2005) 386–393.
they lack T-cell co-stimulatory molecules, and thus, transplantation [17] T.J. Bivalacqua, M.F. Usta, H.C. Champion, D. Adams, D.B. McNamara, A.B. Abdel-
with such cells is an attractive alternative to the potentially costly and Mageed, P.J. Kadowitz, W.J.G. Hellstrom, Gene transfer of endothelial nitric oxide
synthase partially restores nitric oxide synthesis and erectile function in
inconvenient autotransplantation without the need for immunosup- streptozotocin diabetic rats, J. Urol. 169 (2003) 1911–1917.
pression [128]. [18] W. E., Erectile dysfunction in the ageing man, Curr. Opin. Urol. 10 (2000) 625–628.
[19] National Diabetes Statistics, National Diabetes Information Clearinghouse, National
Institute of Diabetes and Digestive, Kidney Diseases (NIDDK), National Institutes of
8. Future directions Health, Available from: http://diabetes.niddk.nih.gov/dm/pubs/statistics/index.
aspx.
[20] M.S. Rendell, J. Rajfer, P.A. Wicker, M.D. Smith, G. for the Sildenafil Diabetes Study,
SC therapy represents a very promising treatment option for ED Sildenafil for treatment of erectile dysfunction in men with diabetes: a randomized
cases not responsive or partially responsive to PDE5Is. IC injection is controlled trial, JAMA 281 (1999) 421–426.
the most commonly used method for SC delivery, in both preclinical [21] M.R. Dashwood, A. Crump, X. Shi-Wen, A. Loesch, Identification of neuronal nitric
oxide synthase (nNOS) in Human penis: a potential role of reduced neuronally-
and clinical trials. Several more cumbersome methods have been derived nitric oxide in erectile dysfunction, Curr. Pharm. Biotechnol. 12 (2011)
attempted but did not prove to be advantageous. The intravenous 1316–1321.
route has shown promise and should be further tested in additional pre- [22] F. Zhou, H. Xin, T. Liu, G.-Y. Li, Z.-Z. Gao, J. Liu, W.-R. Li, W.-S. Cui, G.-Y. Bai, N.C. Park,
Z.C. Xin, Effects of Icariside II on improving erectile function in rats with
clinical studies before adoption for clinical trials. The issue of allogeneic streptozotocin-induced diabetes, J. Androl. 33 (2012) 832–844.
versus autologous source also needs to be further clarified. Among the [23] X. Qiu, G. Lin, Z. Xin, L. Ferretti, H. Zhang, T.F. Lue, C.-S. Lin, Effects of low-energy
different SCs used, ADSC represents the most widely and easiest SC to shockwave therapy on the erectile function and tissue of a diabetic rat model, J.
Sex. Med. 10 (2013) 738–746.
work with. But, whether ADSC or AD-SVF is a better choice needs to [24] H. Ning, G. Lin, T.F. Lue, C.S. Lin, A coculture system of cavernous endothelial and
be answered through additional comparative studies. Timing of SC in- smooth muscle cells, Int. J. Impot. Res. 25 (2013) 63–68.
jection represents another issue despite the demonstration that both [25] H. Ning, X. Qiu, L. Baine, G. Lin, T.F. Lue, C.-S. Lin, Effects of high glucose on human
cavernous endothelial cells, Urology 80 (2012) (1162.e1167-1162.e1111).
immediate and delayed injections had similar long-term positive treat-
[26] C.R. Moore, R. Wang, Pathophysiology and treatment of diabetic erectile dysfunc-
ment outcome [103]. The dosage of SCs requires further testing as the tion, Asian J. Androl. 8 (2006) 675–684.
currently chosen doses of half million to 2 million cells in the rat were [27] S. Gur, P.J. Kadowitz, W.J.G. Hellstrom, A Protein tyrosine kinase inhibitor, imatinib
arbitrary. Pharmacological modifications to the SCs have not been mesylate (Gleevec), improves erectile and vascular function secondary to a reduc-
tion of hyperglycemia in diabetic rats, J. Sex. Med. 7 (2010) 3341–3350.
attempted in the treatment of ED, but might be a future target in [28] N. Villalba, C. Contreras, M. Hernández, A. García-Sacristán, D. Prieto, Impaired
order to influence self-renewal capabilities and thereby prolonging Ca2+ handling in penile arteries from prediabetic Zucker rats: involvement of
their therapeutic effect. In addition to the two current clinical trials, Rho kinase, Am. J. Physiol. Heart Circ. Physiol. 300 (2011) H2044–H2053.
[29] L. de Young, D. Yu, R.M. Bateman, G.B. Brock, Oxidative stress and antioxidant ther-
more should be conducted so as to adequately cope with increasing de- apy: their impact in diabetes-associated erectile dysfunction, J. Androl. 25 (2004)
mand from patients. All these tasks are difficult but extremely impor- 830–836.
tant in order to develop better treatment options for this common and [30] M.M. Garcia, T.M. Fandel, G. Lin, A.W. Shindel, L. Banie, C.-S. Lin, T.F. Lue, Treatment
of erectile dysfunction in the obese Type 2 diabetic ZDF rat with adipose tissue-
important disease. Table 2 provides a summary of all preclinical trials derived stem cells, J. Sex. Med. 7 (2010) 89–98.
using SC for ED treatment. [31] Reduction of penile nitric oxide synthase in diabetic BB/WORdp (type I) and BBZ/
WORdp (type II) rats with erectile dysfunction, Endocrinology 136 (1995)
5709–5717.
References [32] D.F. Penson, D.M. Latini, D.P. Lubeck, K.L. Wallace, J.M. Henning, T.F. Lue, Do impo-
tent men with diabetes have more severe erectile dysfunction and worse quality of
[1] J.J. Sánchez-Cruz, A. Cabrera-León, A. Martı́n-Morales, A. Fernández, R. Burgos, J. life than the general population of impotent patients?: Results from the Explorato-
Rejas, Male erectile dysfunction and health-related quality of life, Eur. Urol. 44 ry Comprehensive Evaluation of Erectile Dysfunction (ExCEED) database, Diabetes
(2003) 245–253. Care 26 (2003) 1093–1099.
[2] N.C.C. Impotence. NIH consensus development panel on impotence, JAMA 270 [33] R. Virag, P. Bouilly, D. Frydman, Is impotence an arterial disorder?: A study of arte-
(1993) 83–90. rial risk factors in 440 impotent men, Lancet 325 (1985) 181–184.
[3] E.O. Laumann, S. West, D. Glasser, C. Carson, R. Rosen, J.-h. Kang, Prevalence and [34] M. Wei, C.A. Macera, D.R. Davis, C.A. Hornung, H.A. Nankin, S.N. Blair, Total choles-
correlates of erectile dysfunction by race and ethnicity among men aged 40 or terol and high density lipoprotein cholesterol as important predictors of erectile
older in the United States: from the male attitudes regarding sexual health survey, dysfunction, Am. J. Epidemiol. 140 (1994) 930–937.
J. Sex. Med. 4 (2007) 57–65. [35] Y.-C. Huang, H. Ning, A.W. Shindel, T.M. Fandel, G. Lin, A.M. Harraz, T.F. Lue, C.-S.
[4] R. Shamloul, H. Ghanem, Erectile dysfunction, The Lancet 381 (2013) 153–165. Lin, The effect of intracavernous injection of adipose tissue-derived stem cells on

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 7

hyperlipidemia-associated erectile dysfunction in a rat model, J. Sex. Med. 7 (2010) [68] D.L. Jones, A.J. Wagers, No place like home: anatomy and function of the stem cell
1391–1400. niche, Nat. Rev. Mol. Cell Biol. 9 (2008) 1121.
[36] M. Bocchio, G. Desideri, P. Scarpelli, S. Necozione, G. Properzi, C. Spartera, F. [69] X. Song, M.D. Wong, E. Kawase, R. Xi, B.C. Ding, J.J. McCarthy, T. Xie, Bmp signals
Francavilla, C. Ferri, S. Francavilla, Endothelial cell activation in men with erectile from niche cells directly repress transcription of a differentiation-promoting
dysfunction without cardiovascular risk factors and overt vascular damage, J. gene, bag of marbles, in germline stem cells in the Drosophila ovary, Development
Urol. 171 (2004) 1601–1604. 131 (2004) 1353–1364.
[37] G. Yetik-Anacak, J.D. Catravas, Nitric oxide and the endothelium: history and im- [70] D. Bochinski, G.T. Lin, L. Nunes, R. Carrion, N. Rahman, C.S. Lin, T.F. Lue, The effect of
pact on cardiovascular disease, Vasc. Pharmacol. 45 (2006) 268–276. neural embryonic stem cell therapy in a rat model of cavernosal nerve injury, BJU
[38] A. Jemal, R. Siegel, J. Xu, E. Ward, Cancer statistics, 2010, CA Cancer J. Clin. 60 Int. 94 (2004) 904–909.
(2010) 277–300. [71] T. Asahara, T. Murohara, A. Sullivan, M. Silver, R. van der Zee, T. Li, B. Witzenbichler,
[39] J. Brandeis, C.L. Pashos, J.M. Henning, M.S. Litwin, A nationwide charge comparison G. Schatteman, J.M. Isner, Isolation of putative progenitor endothelial cells for an-
of the principal treatments for early stage prostate carcinoma, Cancer 89 (2000) giogenesis, Science 275 (1997) 964–966.
1792–1799. [72] N. Werner, S. Kosiol, T. Schiegl, P. Ahlers, K. Walenta, A. Link, M. Böhm, G. Nickenig,
[40] R.C. Dean, T.F. Lue, Neuroregenerative strategies after radical prostatectomy, Rev. Circulating endothelial progenitor cells and cardiovascular outcomes, N. Engl. J.
Urol. 7 (2005) 26. Med. 353 (2005) 999–1007.
[41] M.J. Resnick, T. Koyama, K.-H. Fan, P.C. Albertsen, M. Goodman, A.S. Hamilton, R.M. [73] M. Peichev, A.J. Naiyer, D. Pereira, Z. Zhu, W.J. Lane, M. Williams, M.C. Oz, D.J.
Hoffman, A.L. Potosky, J.L. Stanford, A.M. Stroup, R.L. Van Horn, D.F. Penson, Long- Hicklin, L. Witte, M.A.S. Moore, S. Rafii, Expression of VEGFR-2 and AC133 by circu-
term functional outcomes after treatment for localized prostate cancer, N. Engl. J. lating human CD34+ cells identifies a population of functional endothelial precur-
Med. 368 (2013) 436–445. sors, Blood 95 (2000) 952–958.
[42] V. Ficarra, G. Novara, W. Artibani, A. Cestari, A. Galfano, M. Graefen, G. Guazzoni, B. [74] G.C. Schatteman, M. Dunnwald, C. Jiao, Biology of bone marrow-derived endothe-
Guillonneau, M. Menon, F. Montorsi, V. Patel, J. Rassweiler, H. Van Poppel, Retropubic, lial cell precursors, Am. J. Physiol. Heart Circ. Physiol. 292 (2007) H1–H18.
laparoscopic, and robot-assisted radical prostatectomy: a systematic review and cu- [75] M.C. Yoder, L.E. Mead, D. Prater, T.R. Krier, K.N. Mroueh, F. Li, R. Krasich, C.J. Temm,
mulative analysis of comparative studies, Eur. Urol. 55 (2009) 1037–1063. J.T. Prchal, D.A. Ingram, Redefining endothelial progenitor cells via clonal analysis
[43] M. Albersen, M. Kendirci, F. Van der Aa, W.J.G. Hellstrom, T.F. Lue, J.L. Spees, and hematopoietic stem/progenitor cell principals, Blood 109 (2007) 1801–1809.
Multipotent stromal cell therapy for cavernous nerve injury-induced erectile dys- [76] T.D. Strong, M.A. Gebska, H.C. Champion, A.L. Burnett, T.J. Bivalacqua, Stem and en-
function, J. Sex. Med. 9 (2012) 385–403. dothelial progenitor cells in erection biology, Int. J. Impot. Res. 20 (2008) 243–254.
[44] M. Fode, D.A. Ohl, D. Ralph, J. Sønksen, Penile rehabilitation after radical prostatec- [77] C. Foresta, N. Caretta, A. Lana, A. Cabrelle, G. Palù, A. Ferlin, Circulating endothelial
tomy: what the evidence really says, BJU Int. 112 (2013) 998–1008. progenitor cells in subjects with erectile dysfunction, Int. J. Impot. Res. 17 (2005)
[45] S. Carrier, H. Hricak, S.S. Lee, K. Baba, D.M. Morgan, L. Nunes, G.Y. Ross, T.L. Phillips, 288–290.
T.F. Lue, Radiation-induced decrease in nitric oxide synthase–containing nerves in [78] H.W. Gou, M.Z. Xiao, et al., Transplantation of endothelial progenitor cells
the rat penis, Radiology 195 (1995) 95–99. transfected with VEGF165 to restore erectile function in diabetic rats, Asian J.
[46] M. Kendirci, J. Bejma, W.J.G. Hellstrom, Update on erectile dysfunction in prostate Androl. 13 (2011) 332–338.
cancer patients, Curr. Opin. Urol. 16 (2006) 186–195 (110.1097/1001.mou. [79] J.B. Jiang Y, R.L. Reinhardt, R.E. Schwartz, C.D. Keene, O.-G.X., Pluripotency of mes-
0000193407.0000105285.d0000193408). enchymal stem cells derived from adult marrow, Nature 418 (2011) 41–49.
[47] S.S. Gholami, N.F. Gonzalez-Cadavid, C.-S. Lin, J. Rajfer, T.F. Lue, Peyronie's disease: [80] F. Tögel, C. Westenfelder, The role of multipotent marrow stromal cells (MSCs) in
a review, J. Urol. 169 (2003) 1234–1241. tissue regeneration, Organogenesis 7 (2011) 96–100.
[48] G. Garaffa, L.W. Trost, E.C. Serefoglu, D. Ralph, W.J.G. Hellstrom, Understanding the [81] M.T. Abdel Aziz, S. El-Haggar, T. Mostafa, H. Atta, H. Fouad, S. Mahfouz, L. Rashed, D.
course of Peyronie's disease, Int. J. Clin. Pract. 67 (2013) 781–788. Sabry, A. Senbel, G.A. Ali, Effect of mesenchymal stem cell penile transplantation on
[49] J.A. Lopez, J.P. Jarow, Penile vascular evaluation of men with Peyronie's disease, J. erectile signaling of aged rats, Andrologia 42 (2010) 187–192.
Urol. 149 (1993) 53–55. [82] G. Chamberlain, J. Fox, B. Ashton, J. Middleton, Concise review: mesenchymal stem
[50] D. Martinez, C.E. Ercole, T.S. Hakky, A. Kramer, R. Carrion, Peyronie's disease: still a cells: their phenotype, differentiation capacity, immunological features, and poten-
surgical disease, Adv. Urol. (2012) 5. tial for homing, Stem Cells 25 (2007) 2739–2749.
[51] A. Kadioglu, O. Sanli, T. Akman, A. Ersay, S. Guven, F. Mammadov, Graft materials in [83] X. Qiu, H. Lin, Y. Wang, W. Yu, Y. Chen, R. Wang, Y. Dai, Intracavernous transplan-
Peyronie's disease surgery: a comprehensive review, J. Sex. Med. 4 (2007) 581–595. tation of bone marrow-derived mesenchymal stem cells restores erectile function
[52] N.F. Gonzalez-Cadavid, J. Rajfer, Treatment of Peyronie's disease with PDE5 inhib- of streptozocin-induced diabetic rats, J. Sex. Med. 8 (2011) 427–436.
itors: an antifibrotic strategy, Nat. Rev. Urol. 7 (2010) 215–221. [84] X. Qiu, C. Sun, W. Yu, H. Lin, Z. Sun, Y. Chen, R. Wang, Y. Dai, Combined strategy of
[53] L. Ma, Y. Yang, S.C. Sikka, P.J. Kadowitz, L.J. Ignarro, A.B. Abdel-Mageed, W.J.G. mesenchymal stem cell injection with vascular endothelial growth factor gene
Hellstrom, Adipose tissue-derived stem cell-seeded small intestinal submucosa therapy for the treatment of diabetes-associated erectile dysfunction, J. Androl.
for tunica albuginea grafting and reconstruction, Proc. Natl. Acad. Sci. 109 (2012) 33 (2012) 37–44.
2090–2095. [85] Y. He, W. He, G. Qin, J. Luo, M. Xiao, Transplantation KCNMA1 modified bone
[54] F. Castiglione, P. Hedlund, F. Van der Aa, T.J. Bivalacqua, P. Rigatti, H. Van Poppel, F. marrow-mesenchymal stem cell therapy for diabetes mellitus-induced erectile
Montorsi, D. De Ridder, M. Albersen, Intratunical injection of human adipose tis- dysfunction, Andrologia (2013) 479–486.
sue-derived stem cells prevents fibrosis and is associated with improved erectile [86] C. Sun, H. Lin, W. Yu, X. Li, Y. Chen, X. Qiu, R. Wang, Y. Dai, Neurotrophic effect of
function in a rat model of Peyronie's disease, Eur. Urol. 63 (2013) 551–560. bone marrow mesenchymal stem cells for erectile dysfunction in diabetic rats,
[55] N.F. Gonzalez-Cadavid, J. Rajfer, Laboratory forum: experimental models of Int. J. Androl. 35 (2012) 601–607.
Peyronie's disease. Implications for new therapies, J. Sex. Med. 6 (2009) 303–313. [87] P.A. Fall, M. Izikki, L. Tu, S. Swieb, F. Giuliano, J. Bernabe, R. Souktani, C. Abbou, S.
[56] S.J. Morrison, N.M. Shah, D.J. Anderson, Regulatory mechanisms in stem cell biolo- Adnot, S. Eddahibi, R. Yiou, Apoptosis and effects of intracavernous bone marrow
gy, Cell 88 (1997) 287–298. cell injection in a rat model of postprostatectomy erectile dysfunction, Eur. Urol.
[57] C.-S. Lin, Z.-C. Xin, C.-H. Deng, H. Ning, G. Lin, T.F. Lue, Recent advances in 56 (2009) 716–726.
andrology-related stem cell research, Asian J. Androl. 10 (2008) 171–175. [88] M. Kendirci, L. Trost, B. Bakondi, M.J. Whitney, W.J.G. Hellstrom, J.L. Spees, Trans-
[58] G. Keller, Embryonic stem cell differentiation: emergence of a new era in biology plantation of nonhematopoietic adult bone marrow stem/progenitor cells isolated
and medicine, Genes Dev. 19 (2005) 1129–1155. by p75 nerve growth factor receptor into the penis rescues erectile function in a rat
[59] C. Becker, G. Jakse, Stem cells for regeneration of urological structures, Eur. Urol. 51 model of cavernous nerve injury, J. Urol. 184 (2010) 1560–1566.
(2007) 1217–1228. [89] S.J. Kim, S.W. Choi, K.J. Hur, S.H. Park, Y.C. Sung, Y-S. Ha, H.J. Cho, S-H. Hong, J.Y. Lee,
[60] M.J. Evans, M.H. Kaufman, Establishment in culture of pluripotential cells from T-K. Hwang, S.W. Kim, Synergistic effect of mesenchymal stem cells infected with
mouse embryos, Nature 292 (1981) 154–156. recombinant adenovirus expressing human BDNF on erectile function in a rat
[61] G.M. Strong TD, H.C. Champion, A.L. Burnett, T.J. Bivalacqua, Stem and endothelial model of cavernous nerve injury, Korean J. Urol. 53 (2012) 726–732.
progenitor cells in erection biology, Int. J. Impot. Res. 20 (2008) 243–254. [90] D. You, M.J. Jang, J. Lee, I.G. Jeong, H.S. Kim, K.H. Moon, N. Suh, C.-S. Kim,
[62] J.L. Yamzon, P. Kokorowski, C.J. Koh, Stem cells and tissue engineering applications Periprostatic implantation of human bone marrow-derived mesenchymal stem
of the genitourinary tract, Pediatr. Res. 63 (2008) 472–477. cells potentiates recovery of erectile function by intracavernosal injection in a rat
[63] L. Yao, X. Yu, N. Hui, S. Liu, Application of iPS in assisted reproductive technology: model of cavernous nerve injury, Urology 81 (2013) 104–110.
sperm from somatic cells? Stem Cell Rev. Rep. 7 (2011) 714–721. [91] T.J. Bivalacqua, W. Deng, M. Kendirci, M.F. Usta, C. Robinson, B.K. Taylor, S.N.
[64] K. Takahashi, K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka, K. Tomoda, S. Yamanaka, Murthy, H.C. Champion, W.J.G. Hellstrom, P.J. Kadowitz, Mesenchymal stem cells
Induction of pluripotent stem cells from adult human fibroblasts by defined fac- alone or ex vivo gene modified with endothelial nitric oxide synthase reverse
tors, Cell 131 (2007) 861–872. age-associated erectile dysfunction, Am. J. Physiol. Heart Circ. Physiol. 292
[65] M.H. Chin, M.J. Mason, W. Xie, S. Volinia, M. Singer, C. Peterson, G. Ambartsumyan, (2007) H1278–H1290.
O. Aimiuwu, L. Richter, J. Zhang, I. Khvorostov, V. Ott, M. Grunstein, N. Lavon, N. [92] K. Urish, Y. Kanda, J. Huard, Initial failure in myoblast transplantation therapy has
Benvenisty, C.M. Croce, A.T. Clark, T. Baxter, A.D. Pyle, M.A. Teitell, M. Pelegrini, K. led the way toward the isolation of muscle stem cells: potential for tissue regener-
Plath, W.E. Lowry, Induced pluripotent stem cells and embryonic stem cells are dis- ation, in: P.S. Gerald (Ed.), Current Topics in Developmental Biology, Academic
tinguished by gene expression signatures, Cell Stem Cell 5 (2009) 111–123. Press, 2005, pp. 263–280.
[66] J.C. Kiefer, Primer and interviews: the dynamic stem cell niche, Dev. Dyn. 240 [93] M.G. Engelmann, W.M. Franz, Stem cell therapy after myocardial infarction: ready
(2011) 737–743. for clinical application? Curr. Opin. Mol. Ther. 8 (2006) 396–414.
[67] J. Becerra, L. Santos-Ruiz, J. Andrades, M. Marí-Beffa, The stem cell niche should be [94] J.H. Hwang, S.H. Yuk, J.H. Lee, W.S. Lyoo, S-H. Ghil, S.S. Lee, I.G. Khang, S.Y. Paik, J.Y.
a key issue for cell therapy in regenerative medicine, Stem Cell Rev. Rep. 7 (2011) Lee, Isolation of muscle derived stem cells from rat and its smooth muscle differen-
248–255. tiation, Mol. Cell 17 (2004) 57–61.

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012
8 A. Alwaal et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

[95] Y. Kim, F. de Miguel, I. Usiene, D. Kwon, N. Yoshimura, J. Huard, M.B. Chancellor, In- [114] S. Piao, I.G. Kim, J.Y. Lee, S.H. Hong, S.W. Kim, T-K. Hwang, S.H. Oh, J.H. Lee, J.C. Ra, J.
jection of skeletal muscle-derived cells into the penis improves erectile function, Y. Lee, Therapeutic effect of adipose-derived stem cells and BDNF-immobilized
Int. J. Impot. Res. 18 (2006) 329–334. PLGA membrane in a rat model of cavernous nerve injury, J. Sex. Med. 9 (2012)
[96] G. Nolazco, I. Kovanecz, D. Vernet, R.A. Gelfand, J. Tsao, M.G. Ferrini, T. Magee, J. 1968–1979.
Rajfer, N.F. Gonzalez-Cadavid, Effect of muscle-derived stem cells on the restora- [115] I.G. Kim, S. Piao, J.Y. Lee, S.H. Hong, T-K. Hwang, S.W. Kim, C.S. Kim, J.C. Ra, I. Noh, J.
tion of corpora cavernosa smooth muscle and erectile function in the aged rat, Y. Lee, Effect of an adipose-derived stem cell and nerve growth factor-incorporated
BJU Int. 101 (2008) 1156–1164. hydrogel on recovery of erectile function in a rat model of cavernous nerve injury,
[97] J.C. Woo, W.J. Bae, S.J. Kim, S.D. Kim, D.W. Sohn, S.H. Hong, J.Y. Lee, T.-K. Hwang, Tissue Eng. A 19 (2013) 14–23.
Y.C. Sung, S.W. Kim, Transplantation of muscle-derived stem cells into the corpus [116] H.H. Jeong, S. Piao, J.N. Ha, I.G. Kim, S.H. Oh, J.H. Lee, H.J. Cho, S.H. Hong, S.W. Kim,
cavernosum restores erectile function in a rat model of cavernous nerve injury, Ko- J.Y. Lee, Combined therapeutic effect of udenafil and adipose-derived stem cell
rean J. Urol. 52 (2011) 359–363. (ADSC)/brain-derived neurotrophic factor (BDNF)-membrane system in a rat
[98] P.A. Trainor, R. Krumlauf, M. Bronner-Fraser, Neural crest cells, in: B.H., R. Lanza, D. model of cavernous nerve injury, Urology 81 (2013) (1108.e1107-1108.e1114).
M (Eds.), Handbook of Stem Cells, Elsevier Academic Press, Burlington, MA, 2004, [117] L.S. Hwan, K.I. Gul, J.A. Ryang, S.K. Raj, L.J. Ho, P.K. Dong, C.B. Ha, K.S. Woong, K.K.
pp. 205–218. Hean, L.J. Youl, Combined effects of brain-derived neurotrophic factor immobilized
[99] Y.S. Song, H.J. Lee, I.H. Park, I.S. Lim, J.H. Ku, S.U. Kim, Human neural crest stem cells poly-lactic-co-glycolic acid membrane with human adipose-derived stem cells and
transplanted in rat penile corpus cavernosum to repair erectile dysfunction, BJU basic fibroblast growth factor hydrogel on recovery of erectile dysfunction, Tissue
Int. 102 (2008) 220–224. Eng. A 20 (2014) 2446–2454.
[100] C.S. Lin, Z.C. Xin, C.H. Deng, H. Ning, G. Lin, T.F. Lue, Defining adipose tissue-derived [118] D. You, M.J. Jang, J. Lee, N. Suh, I.G. Jeong, D.W. Sohn, S.W. Kim, T.Y. Ahn, C.-S. Kim,
stem cells in tissue and in culture, Histol. Histopathol. 25 (2010) 807–815. Comparative analysis of periprostatic implantation and intracavernosal injection of
[101] H.K. Strem BM, M. Zhu, et al., Multipotential differentiation of adipose tissue- human adipose tissue-derived stem cells for erectile function recovery in a rat
derived stem cells, Keio J. Med. 54 (2005) 132–141. model of cavernous nerve injury, Prostate 73 (2013) 278–286.
[102] C.S. Lin, T.F. Lue, Stem cell therapy for stress urinary incontinence: a critical review, [119] J.-K. Ryu, D.-H. Kim, K.M. Song, T. Yi, J.-K. Suh, S.U. Song, Intracavernous delivery of
Stem Cells Dev. 21 (2012) 834–843. clonal mesenchymal stem cells restores erectile function in a mouse model of cav-
[103] X. Qiu, T.M. Fandel, L. Ferretti, M. Albersen, H. Zhang, G. Lin, C.-S. Lin, T. Schroeder, ernous nerve injury, J. Sex. Med. 11 (2014) 411–423.
T.F. Lue, Both immediate and delayed intracavernous injection of autologous [120] G. Lin, M. Albersen, A.M. Harraz, T.M. Fandel, M. Garcia, M.H. McGrath, B.R. Konety,
adipose-derived stromal vascular fraction enhances recovery of erectile function T.F. Lue, C.-S. Lin, Cavernous nerve repair with allogenic adipose matrix and autol-
in a rat model of cavernous nerve injury, Eur. Urol. 62 (2012) 720–727. ogous adipose-derived stem cells, Urology 77 (2011) (1509.e1501-1509.e1508).
[104] J.-K. Ryu, M. Tumurbaatar, H.-R. Jin, W.J. Kim, M.-H. Kwon, S. Piao, M.J. Choi, G.N. [121] X. Qiu, J. Villalta, L. Ferretti, T.M. Fandel, M. Albersen, G. Lin, Y. Dai, T.F. Lue, C.S. Lin,
Yin, K.-M. Song, Y.-J. Kang, Y.J. Koh, G.Y. Koh, J.-K. Suh, Intracavernous delivery of Effects of intravenous injection of adiposederived stem cells in a rat model of radi-
freshly isolated stromal vascular fraction rescues erectile function by enhancing ation therapy-induced erectile dysfunction, J. Sex. Med. 9 (2012) 1834–1841.
endothelial regeneration in the streptozotocin-induced diabetic mouse, J. Sex. [122] C. Ying, W. Hu, B. Cheng, M. Yang, X. Zheng, X. Wang, Erectile function restoration
Med. 9 (2012) 3051–3065. after repair of resected cavernous nerves by adipose-derived stem cells combined
[105] A. Miranville, C. Heeschen, C. Sengenès, C.A. Curat, R. Busse, A. Bouloumié, Im- with autologous vein graft in rats, Cell. Mol. Neurobiol. 34 (2014) 393–402.
provement of postnatal neovascularization by human adipose tissue-derived [123] A. Gokce, Z.Y. Abd Elmageed, G.F. Lasker, M. Bouljihad, H. Kim, L.W. Trost, P.J.
stem cells, Circulation 110 (2004) 349–355. Kadowitz, A.B. Abdel-Mageed, S.C. Sikka, W.J. Hellstrom, Adipose tissue-derived
[106] J. Rehman, D. Traktuev, J. Li, S. Merfeld-Clauss, C.J. Temm-Grove, J.E. Bovenkerk, C.L. stem cell therapy for prevention and treatment of erectile dysfunction in a rat
Pell, B.H. Johnstone, R.V. Considine, K.L. March, Secretion of angiogenic and model of Peyronie's disease, Andrology 2 (2014) 244–251.
antiapoptotic factors by human adipose stromal cells, Circulation 109 (2004) [124] G. Liu, X. Sun, J. Bian, R. Wu, X. Guan, B. Ouyang, Y. Huang, H. Xiao, D. Luo, A. Atala,
1292–1298. Y. Zhang, C. Deng, Correction of diabetic erectile dysfunction with adipose derived
[107] H. Ning, G. Liu, G. Lin, R. Yang, T.F. Lue, C.-S. Lin, Fibroblast growth factor 2 pro- stem cells modified with the vascular endothelial growth factor gene in a rodent
motes endothelial differentiation of adipose tissue-derived stem cells, J. Sex. diabetic model, PLoS ONE 8 (2013) e72790.
Med. 6 (2009) 967–979. [125] S.J. Kim, S.H. Park, Y.C. Sung, S.W. Kim, Effect of mesenchymal stem cells associated
[108] M. Albersen, T.M. Fandel, G. Lin, G. Wang, L. Banie, C.-S. Lin, T.F. Lue, Injections of to matrixen on the erectile function in the rat model with bilateral cavernous
adipose tissue-derived stem cells and stem cell lysate improve recovery of erectile nerve crushing injury, Int. Braz. J. Urol. 38 (2012) 833–841.
function in a rat model of cavernous nerve injury, J. Sex. Med. 7 (2010) 3331–3340. [126] I. Kovanecz, S. Rivera, G. Nolazco, D. Vernet, D. Segura, S. Gharib, J. Rajfer, N.F.
[109] M.J. Varma, R.G.M. Breuls, T.E. Schouten, W.J.F.M. Jurgens, H.J. Bontkes, G.J. Gonzalez-Cadavid, Separate or combined treatments with daily sildenafil,
Schuurhuis, S.M. Van Ham, F.J. Van Milligen, Phenotypical and functional charac- molsidomine, or muscle-derived stem cells prevent erectile dysfunction in a rat
terization of freshly isolated adipose tissue-derived stem cells, Stem Cells Dev. model of cavernosal nerve damage, J. Sex. Med. 9 (2012) 2814–2826.
16 (2007) 91–104. [127] J.Y. Bahk, J.H. Jung, H. Han, S.K. Min, Y.S. Lee, Treatment of diabetic impotence with
[110] W.Y. Choi, H.G. Jeon, Y. Chung, J.J. Lim, D.H. Shin, J.M. Kim, B.S. Ki, S-H. Song, S.-J. umbilical cord blood stem cell intracavernosal transplant: preliminary report of 7
Choi, K.-H. Park, S.H. Shim, J. Moon, S.J. Jung, H.M. Kang, S. Park, H.M. Chung, J.J. cases, Exp. Clin. Transplant. 8 (2010) 150–160.
Ko, K.Y. Cha, T.K. Yoon, H. Kim, D.R. Lee, Isolation and characterization of novel [128] C.S. Lin, G. Lin, T.F. Lue, Allogeneic and xenogeneic transplantation of adipose-
highly proliferative human CD34/CD73-double-positive testis-derived stem cells derived stem cells in immunocompetent recipients without immunosuppressants,
for cell therapy, Stem Cells Dev. 22 (2013) 2158–2173. Stem Cell Dev. 21 (2012) 2770–2778.
[111] B. Ouyang, X. Sun, D. Han, S. Chen, B. Yao, Y. Gao, J. Bian, Y. Huang, Y. Zhang, Z. Wan, [129] C. Ying, M. Yang, X. Zheng, W. Hu, X. Wang, Effects of intracavernous injection of
B. Yang, H. Xiao, Z. Songyang, G. Liu, Y. Zhang, C. Deng, Human urine-derived stem adipose-derived stem cells on cavernous nerve regeneration in a rat model, Cell.
cells alone or genetically-modified with FGF2 improve type 2 diabetic erectile dys- Mol. Neurobiol. 33 (2013) 233–240.
function in a rat model, PLoS ONE 3 (2014) e92825. [130] H. Nishimatsu, E. Suzuki, S. Kumano, A. Nomiya, M. Liu, H. Kume, Y. Homma,
[112] T.M. Fandel, M. Albersen, G. Lin, X. Qiu, H. Ning, L. Banie, T.F. Lue, C.-S. Lin, Recruit- Adrenomedullin mediates adipose tissue-derived stem cell-induced restoration
ment of intracavernously injected adipose-derived stem cells to the major pelvic of erectile function in diabetic rats, J. Sex. Med. 9 (2012) 482–493.
ganglion improves erectile function in a rat model of cavernous nerve injury, [131] N.D. Das, K.-M. Song, G.N. Yin, D. Batbold, M.-H. Kwon, K.-D. Kwon, W.J. Kim, Y.S.
Eur. Urol. 61 (2012) 201–210. Kim, J.-K. Ryu, J.-K. Suh, Xenogenic transplantation of human breast adipose-
[113] Q.X., G. Lin, T. Fandel, et al., Tracking intracavernously injected adipose-derived derived stromal vascular fraction enhances recovery of erectile function in diabetic
stem cells to bone marrow, Int. J. Impot. Res. 23 (2011) 268–275. mice, Biol. Reprod. 90 (2014) 1–10 (66).

Please cite this article as: A. Alwaal, et al., Stem cell treatment of erectile dysfunction, Adv. Drug Deliv. Rev. (2014), http://dx.doi.org/10.1016/
j.addr.2014.11.012

Вам также может понравиться