Sunteți pe pagina 1din 16

Cell Stem Cell

Perspective

Mesenchymal Stromal Cells:


Clinical Challenges and Therapeutic Opportunities
Jacques Galipeau1,3,* and Luc Sensébé2,3,*
1Department of Medicine and Carbone Cancer Center, University of Wisconsin in Madison, Madison, WI, USA
2UMR5273 STROMALab CNRS/EFS/UPS – INSERM U1031, Toulouse, France
3These authors contributed equally

*Correspondence: jgalipeau@wisc.edu (J.G.), luc.sensebe@efs.sante.fr (L.S.)


https://doi.org/10.1016/j.stem.2018.05.004

Mesenchymal stromal cells (MSCs) have been the subject of clinical trials for more than a generation, and the
outcomes of advanced clinical trials have fallen short of expectations raised by encouraging pre-clinical an-
imal data in a wide array of disease models. In this Perspective, important biological and pharmacological
disparities in pre-clinical research and human translational studies are highlighted, and analyses of clinical
trial failures and recent successes provide a rational pathway to MSC regulatory approval and deployment
for disorders with unmet medical needs.

Mesenchymal stromal cells (MSCs) were first tested as a cellular press releases and society meeting abstracts (Martin et al.,
pharmaceutical agent in human subjects in 1995 by Hillard 2010). Subset analysis suggested that children with GvHD
Lazarus (Lazarus et al., 1995) and have since become the most were responsive to MSCs (Kurtzberg et al., 2010). On this basis,
clinically studied experimental cell therapy platform worldwide on May 17, 2012, Health Canada issued marketing approval for
for which there is no marketing approval in the United States Prochymal to treat children with acute GvHD (Reicin et al., 2012).
(Fung et al., 2017). The enthusiasm for these mostly early-phase Health Canada approved the drug via a Notice of Compliance
clinical trials reflects the considerable ease for the enthusiast to with Conditions (NOC/c), which allows certain drugs into the
manufacture culture-adapted MSCs from readily accessible tis- market without full efficacy data. Under these terms, Health
sue sourced from normal volunteers and the use of tissue culture Canada subjected Osiris Therapeutics to heightened post-mar-
techniques dating back more than a quarter century. In the ket surveillance. In addition to long-term monitoring, the NOC/c
United States in particular, industrial sponsors have led virtually requirements restricted treatment to children with refractory
all advanced phase III clinical trials of MSCs, and the field as a GvHD and restricted access of Prochymal to physicians who
whole has been severely criticized for its ill-informed irrational have experience treating GvHD patients. As of 2018, 6 years af-
exuberance (Bianco et al., 2013). This criticism often reflects ter Health Canada approval, Osiris—or latterly Mesoblast—have
angst arising from the egregiously predatory business activities not marketed Prochymal in Canada. Debate at the Canadian
of unregulated stem cell clinics in the United States and world- House of Commons Standing Committee on Health held on
wide, riding the unproven promise of regenerative therapies, May 30, 2016 spoke to the conundrum at hand, where testimony
including MSCs, as a cure-all (Turner and Knoepfler, 2016). described the case of Prochymal, which was ‘‘probably the first
truly innovative technology to be put forward for a conditional
The Travails of MSC Marketing Approval Worldwide: licensing approval’’ [in Canada], and it ‘‘went nowhere because
The MSCs for GvHD Paradigm it couldn’t get reimbursed’’ (House of Commons Canada,
MSCs have secured conditional approval in 2012 to treat chil- 2016). There is no publicly available data showing that Prochy-
dren with graft versus host disease (GvHD) in Canada and New mal has been distributed outside of clinical trials on the terms
Zealand, and, latterly, approval in Japan was obtained as well. of marketing following conditional approvals in Canada or New
The historic road map and outcomes of these approvals and Zealand for the indication of pediatric steroid-refractory GvHD.
how they informed a clinical trial strategy to secure United States In Japan, the Act on the Safety of Regenerative Medicine and
approval of MSCs for GvHD can provide some insights into how the Pharmaceuticals, Medical Devices and Other Therapeutic
to best adapt advanced clinical trials designed for regulatory Products Act were enacted in November 2014, creating a new
approval. framework for clinical research and products related to regener-
In May 2009, Osiris Therapeutics (United States) completed ative medicine (Sipp, 2015). This set the stage for approval of
the first major industry-sponsored phase III trial of allogeneic, MSCs as the first allogeneic regenerative medicine product in
marrow-derived MSCs for treatment of steroid-refractory Japan. Utilizing the technology licensed in from Osiris Therapeu-
GvHD (NCT00366145). The MSCs were sourced from normal tics in 2003, JCR Pharmaceuticals (JCR) developed TEMCELL
volunteers from whom up to 10,000 doses were manufactured in Japan for the treatment of acute GvHD. JCR announced on
per donor, and the ensuing cryobanked product (Prochymal) September 15, 2015 that the Japanese Ministry of Labour and
was thawed and transfused at the point of care into eligible Welfare has approved TEMCELL for acute GvHD. Reimburse-
patients with steroid-refractory GvHD (Table 2). The overall ment for TEMCELL was authorized by Japanese National Health
response rate with Prochymal was 82% versus 73% for placebo Insurance at U868,680 ($7,079) per bag of 72 million cells.
(p = 0.12). The clinical trial results have been presented as part of In Japan, the average adult patient will receive 16 or up to

824 Cell Stem Cell 22, June 1, 2018 ª 2018 Elsevier Inc.
Cell Stem Cell

Perspective

Table 1. Comparative Analysis of Pre-clinical Murine Data and Human Clinical Trial Use of MSCs
Murine Pre-clinical Studies Industry-Sponsored Phase III MSC Clinical Trialsa
MSC immune compatibility overwhelmingly syngeneic 11:8 allogeneic:autologous
MSC fitness typically ‘‘fresh’’ mostly ‘‘thawed’’b
Cell dose typically 50 million cells/kg i.v. 2 million cells/kg i.v., up to 120-600 million cells subdermally or endomyocardially
Clinical outcomes predominantly positive first European commission marketing approval on record for MSC product:
Alofisel (darvadstrocel), granted March 2018
a
See Table S1 for a detailed description of 19 studies.
b
Notable exception of Cx401 and Cx601 (also known as Alofisel or darvadstrocel, NCT01541579) and mesenchymal cardiopoietic cells
(NCT01768702), which are live cell products that were culture-rescued prior to administration to human subjects (see Table S1).

24 bags of 72 million cells. On this basis, a treatment course of discards such as umbilical cord tissue and placental cells
TEMCELL in an adult Japanese patient to be reimbursed up to (Table S1). Industry-sponsored manufactured allogeneic MSCs
U20,848,320 ($170,000). TEMCELL generated U0.7 billion sales allow manufacture of up to 1 million doses per donor for mass
for JCR in fiscal year (FY) 2016 (JCR Pharmaceuticals, 2017). deployment, whereas a mix of autologous or allogeneic MSCs
Therefore, with the exception of Japan and its approval of is used by academic centers, which typically manufacture no
MSCs for acute GvHD, MSCs have remained available solely more than 10 doses per donor (Galipeau, 2013). The relative
through clinical trial mechanisms for all indications in other major merits of allogeneic versus autologous and distinct tissue MSC
regulatory jurisdictions, including North America and Europe. sourcing are typically driven by proprietary concerns rather
Notwithstanding, rigorous, peer-reviewed scientific inquiry and than compelling biological superiority of an MSC platform in re-
well-designed, regulation-compliant clinical trials can provide gards to potency and outcomes. Notwithstanding, after more
mechanistic and translational insights that may well demonstrate than 20 years of clinical research in the translational use of
a useful role for MSCs in disorders with unmet medical needs, MSCs, the question remains whether MSCs can fulfill the thera-
including GvHD and others (Phinney et al., 2013; Galipeau, peutic promise foreshadowed by pre-clinical animal research.
2013; Fibbe et al., 2013). Indeed, in March 2018, the European
Commission approved the first MSC pharmaceutical agent Cognitive Dissonance between Pre-clinical Murine and
(Alofisel) to treat Crohn’s-related enterocutaneous fistular Human MSC Clinical Trial Outcomes
disease, and this progress foreshadows such developments. The knowledge of MSC mechanisms of action are derived
predominantly from pre-clinical work in murine systems and
MSC Beginnings in vitro analysis of human MSCs. A typical experimental scenario
Prior to their branding as mesenchymal stem cells (Caplan, involves the harvest of mouse bone marrow and culture expan-
1991), marrow-derived fibroblasts were exploited in the Dexter sion of plastic adherent MSC progenitors in two-dimensional
assay as feeder cells to allow long-term study of murine hemato- culture flask systems in room air humidified incubators akin
poietic stem cells in a reductionist in vitro system (Dexter et al., to what was described by Alexander Friedenstein in 1974.
1977). This system spoke to the niche-like properties of marrow The resultant polyclonal mix of MSCs is harvested during the
fibroblasts, at least in regards to sustaining primitive hematopoi- log phase of growth, and these MSCs are adoptively transferred
etic elements. The advent of recombinant growth factors rele- to immune-competent experimental mice that are nearly always
gated the Dexter assay to the trash bin of scientific history, but syngeneic to the MSC product. Using similar manufacturing
culture-adapted marrow fibroblasts themselves were found to methods, human MSCs, typically allogeneic, are culture-
serve as a powerful surrogate in vitro model system to study expanded to their replicative limit and cryobanked for later use.
human hematopoiesis, bone developmental biology, and related Human subjects enrolled in advanced clinical trials designed
mesenchymal structural elements (Friedenstein et al., 1974; for marketing approval typically receive one or multiple doses
Kfoury and Scadden, 2015). Based on their niche properties of allogeneic MSCs directly retrieved from the freezer or
and mesodermal structural capacity, their ability to profoundly following culture rescue. Indeed, there are numerous peer-re-
affect the functionality of bystander innate and adaptive immune viewed scientific reports providing unambiguous demonstration
cells was discovered (Bernardo and Fibbe, 2013). These insights of the positive effect of MSC adoptive transfer in pre-clinical
led to first-in-human clinical trials, where transfusion of MSCs mouse models of disease. However, the effect of MSCs on
was successful in accelerating hematopoietic recovery following murine outcomes has not readily translated to equivalency in
high-dose myeloablative chemotherapy (Koç et al., 2000) and human phase III studies (Table 1). The dissonance between
reverse steroid-resistant GvHD (Le Blanc et al., 2008). mouse and human clinical outcomes may be best explained
From these pioneering translational studies, the exploitation of by the apparent discrepancies of immune compatibility, dosing,
MSCs’ niche-like regenerative properties and their anti-inflam- and fitness of culture-adapted MSCs.
matory action have spurned the use of both autologous and allo-
geneic MSCs for acute tissue injury syndromes, chronic degen- MSC Immune Compatibility
erative disorders, and inflammatory disease. By far the most In the early days of MSC translational development, it was
prevalent source of MSCs in clinical trials is adult bone marrow, part of the lore that MSCs possessed unique but unproven
followed by adipose tissue with an emergence of puerperal immune privilege, allowing adoptive transfer in allogeneic

Cell Stem Cell 22, June 1, 2018 825


Cell Stem Cell

Perspective

immune-competent recipients without risk of transferred cell weight-adjusted dosing of a cell drug in rodents may not pre-
rejection. This one-size-fits-all narrative informed the develop- cisely predict human pharmacology or response (Shanks et al.,
ment of mass-produced MSCs from a few donors for wide- 2009). However, the order of magnitude difference in MSC
spread use in allogeneic unrelated recipients for an array of ail- dosing between species would foreshadow a negative bias in
ments. However, nearly all pre-clinical mouse data supporting outcomes for humans if the operating biological mechanisms
the use of MSCs examined the use of syngeneic, major histo- are comparable between species and are dose-dependent.
compatibility complex (MHC)-matched cells when examining ef- Considering these variables, we can surmise that methods and
ficacy endpoints. It has since been demonstrated that MSCs, like scale of manufacture, point-of-care deployment, and dosing
all somatic tissues, express MHC class I molecules constitutively trump MSC paracrine potency independent of their tissue sourc-
and have the ability to express MHC class II when exposed to in- ing. Despite the challenges in demonstrating unequivocal effi-
flammatory cues such as interferon-g. Although MSCs have the cacy, there is little controversy that intravenous administration
capacity to express potent inhibitory molecules of both innate of MSCs (mostly marrow-derived but also adipose) is clinically
and adaptive immune effectors, these may not suffice to fend safe, with a post-infusion febrile reaction being the sole adverse
off acquired alloimmunization. Indeed, the notion of MSCs being event likely associated with their use (Lalu et al., 2012). Although
immunoevasive and not immune-privileged has been proposed murine culture-adapted MSCs are prone to spontaneous immor-
(Ankrum et al., 2014). Admittedly, the use of pre-banked alloge- talization and latter transformation, a feature observed nearly
neic MSCs is the only feasible deployment strategy for use in 50 years ago (Franks et al., 1970), claims of similar genetic insta-
acute tissue injury syndromes like stroke, sepsis, or myocardial bility and tumorigenicity for human culture-expanded MSCs
infarction, where the delays in manufacturing autologous have been debunked (Sensebé et al., 2012).
MSCs would forfeit their utility in affecting outcomes. Indepen-
dent of biological concerns for immune compatibility, a business Fitness
case can be made that only mass-produced allogeneic MSCs Using mouse models of tissue injury or inflammatory pathology,
will ever sustain a margin-driven marketing model where cost it is apparent that MSCs affect outcomes through paracrine
of goods is the main headwind to commercial success. How- secretion of multiple cytokines, morphogens, small molecules,
ever, immunological compatibility between donor MSCs and and cargo-bearing exosomes, including contact factors, which
recipient may be critically vital in certain situations, such as affect the biology of adjacent and distant responder cells and tis-
enhancing engraftment in bone marrow transplantation (Nauta sue (Wang et al., 2014). The vast majority of these mouse studies
et al., 2006), as well as in a clinical scenario where long-term examine the use of culture-adapted murine MSCs harvested
repeated administration would be required to affect outcomes during the log phase of growth with optimal metabolic fitness,
for a chronic disorder. When sought, measurable humoral high replication capacity, and syngeneic status (e.g., autolo-
alloimmunization in human subjects receiving mismatched gous) to the recipient. The usual disclaimers and biases of using
MSCs can be detected (Reinders et al., 2015), although its role in-bred mouse strains as experimental systems predictive for the
in failure to meet primary efficacy endpoints in advanced clinical human condition are valid but not specific to MSC science.
trials using allogeneic MSCs intravenously remains unknown. It Reductionist analyses of the MSC secretome in animal disease
is reasonable to assume that, for acute tissue injury syndromes models clearly identify an array of candidate molecule pathways
such as myocardial infarction and stroke, where long-term out- that, as an aggregate, provide a cogent matrix of mechanisms
comes are dictated by short-term biological recovery (typically of action, which may provide a road map for human MSC func-
in the first week after injury), allogeneic MSCs may still provide tionalities where comparable (Chinnadurai et al., 2018).
a benefit prior to an adaptive immune rejection response. In contrast, the vast majority of human clinical trials use alloge-
Indeed, secondary analysis of post-stroke outcomes 1 year neic cryobanked MSCs that are thawed immediately prior to
following MSC therapy (NCT01436487) are suggestive of a transfusion. MSCs display molecular signatures of cell injury in
possible benefit (Hess et al., 2017). the first 24 hr following retrieval from cryostorage, and these
correlate with defects in suppression function in vitro, increased
Dosing susceptibility to lysis by immune cells, and complement as well
Following tail vein intravenous transfusion in mice, the bulk of as shortened persistence in vivo following intravenous transfu-
MSCs are immediately trapped in the lung microvasculature, sion (Moll et al., 2016). Furthermore, there are emerging data
and a small subset may redistribute to sites of injury or damage showing that human MSCs attaining replicative exhaustion
(Sensebé and Fleury-Cappellesso, 2013), although there is no (also known as senescence) are impaired in their ability to sup-
evidence that the latter property is required for their clinical press inflammation (Sensebé and Fleury-Cappellesso, 2013), a
effect. Indeed, administration of MSCs in extravascular com- feature likely to occur in industrial-scale MSC expansion meth-
partments (subcutaneous, intramuscular, or peritoneal) also odologies. Considering that MSCs are typically transfused in pa-
affects outcomes for distant organs (e.g., inflamed brain) without tients within a few hours after thawing, we can hypothesize that
meaningful diseased tissue-tropic MSC redistribution, speaking allogeneic, likely senescent human MSCs directly retrieved from
to their systemic effect. When MSCs are given intravenously to cryostorage, with its associated effect on viability, functionality,
rodents, it is often at a dose of 50 million MSCs/kg. For most and in vivo persistence, are less optimal than metabolically fit
clinical indications, human MSCs are transfused intravenously materials routinely used in analogous murine systems. Distinct
at doses typically in the one to two million cells per kilogram from metabolic fitness arising from culture methods is the issue
and never more than 12 million cells/kg. From a comparative of donor fitness, in particular when manufacturing thousands of
therapeutics perspective, an argument can be made that body allogeneic doses from a single human volunteer. As an

826 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective

aggregate, culture-adapted MSCs from otherwise healthy volun- Chamley, 2011). Herein lies a plausible mechanism that concili-
teer donors invariably express canonical cell surface markers of ates the observation that intravenous administration of MSCs
identity for MSCs and maintain progenitor properties to varying and their subsequent lung entrapment may recapitulate a feto-
degrees. However, when interrogating the functional response maternal tolerance strategy evolved by placental vertebrates.
of MSCs to stimulatory cues, there can be substantial variance Indeed, it may well be that the phagocytic clearance of MSCs
in the magnitude of expression of key effector pathways, elicit an IL-10, IDO, and transforming growth factor b (TGF-b)-
such as indoleamine dioxygenase (IDO) upregulation following suppressive response akin to that observed when recycling
interferon-g stimulation (François et al., 2012), suggesting that apoptotic cellular debris by efferocytosis (Elliott et al., 2017).
biomarker-driven donor screening (or the lack thereof) may
affect the potency of a mass-manufactured product. Limits to the Potency of Apoptotic MSCs
The concept of efferocytosis as a means by which MSC can
Efferocytosis as a Theorem for a Cell-Autonomous MSC drive immune suppression in a cell function-autonomous
Effect In Vivo manner was first suggested by Moll et al. (2016) and provides
In 2009, Németh et al. (2009) at the NIH unequivocally demon- further credence to the observation that functionally compro-
strated that intravenously transfused mouse MSCs accumulate mised, apoptotic, or dead MSCs may drive a suppressive
in the lung and transmigrate outside to the vascular space, where response, albeit bereft of the added benefit of living MSC func-
more than half of lung-trapped MSCs are rapidly phagocytosed tional properties. This unifying theorem may also explain how
by lung-resident tissue macrophages. They went on to show that xenogenic, allogeneic, and syngeneic MSCs, especially when
MSCs led to a protective effect against lethal sepsis because of they display cell surface markers of cell injury such as phospha-
interleukin-10 (IL-10) produced by endogenous macrophages. tidylserine, may trigger a physiological mechanism of tissue
Since this seminal report, there has been a plurality of reports clearance that promotes immune suppression or tolerance.
validating a unique cross-talk between exogenous MSCs and Intriguingly, it has been demonstrated that the infusion of
recipient monocyte and/or macrophages as part of the anti- MSCs rendered apoptotic in vitro can deliver some immunosup-
inflammatory effect of MSCs (Carty et al., 2017). Intriguingly, pressive activity in GvHD mice. However, there are limitations
xenotransplantation of human MSCs into mice teaches us of using apoptotically rendered MSCs for the treatment of
that there are also likely cell function-autonomous effects of GvHD. In contrast to live MSCs, apoptotic MSCs are completely
MSCs. Namely, intravenous infusion of live or dead human ineffective when administered intravenously in mice. However,
MSCs evokes a similar molecular genetic response in murine apoptotic MSCs only improved GvHD when infused intraperito-
host lungs (Luk et al., 2016). Taking into consideration the neally and with a therapeutic efficacy substantially reduced in
inherent biases in studying the immune physiology of interspe- comparison with that of comparably sourced live MSCs (Galleu
cies cell transfer, these data speak to the possibility that MSCs et al., 2017). Considering that the potency of apoptotic MSCs
may elicit a non-specific immune-suppressive effect through is substantially less than that of live MSCs suggests that, in addi-
their phagocytosis by the host reticuloendothelial system tion to efferocytotic clearance, biological fitness of MSCs and
(Poon et al., 2014). Furthermore, the suppressive effect can their functionalities play an important role in their utility as a
take place independent of MSC viability. In contrast to blood- cell pharmaceutical agent (Figure 1). The phenomenon of MSC
borne lymphomyeloid cells, MSCs are the only anchorage- efferocytosis is not undesirable per se, but it certainly mightily
dependent nucleated cellular product ever examined as a complicates the interpretation of pre-clinical animal data where
transfusion pharmaceutical. xenotransfer of human MSCs is examined as a primary method
of analysis. It also adds a layer of complexity when attempting
Plazentallen as the Rosetta Stone of MSC Function to distinguish MSC effects arising from cell metabolic activity
Intriguingly, pregnancy is a normal physiological circumstance from non-specific immune modulation, arising from clearance
where a large bolus of circulating nucleated allogeneic non- of immune-mismatched or functionally marginal MSCs.
blood elements give us biological guidance regarding the
pharmacology of transfused MSCs. First described in 1893 as Phase III Industry-Sponsored MSC Clinical Trials:
plazentallen, it was observed that pregnant women having suc- Adaptive Clinical Trial Design and Outcomes
cumbed to the complications of eclampsia had fetal tropho- A query of the Clincaltrials.gov international database for self-re-
blast-derived stromal cellular elements shed in uterine veins ported industry-sponsored phase III clinical trials provides
that embolized to the lung microvasculature. It was later discov- important insights into the state of the field (Table S1). Among
ered that this phenomenon is part of normal pregnancy, and it these 19 studies, ten are completed, of which there are three ter-
was estimated that several grams of apoptotic fetal stromal minal studies that have outcomes that have been disclosed in a
cellular elements travel from the uterus to the lung on a daily ba- public forum that allows analysis: allogeneic marrow MSCs for
sis. These fetal stromal elements are cleared by maternal lung- GvHD, autologous marrow MSCs for heart disease, and alloge-
resident phagocytes within a few days and may lead to an upre- neic adipose MSCs for Crohn’s fistular disease.
gulation of IL-10 and IDO by these same phagocytes (Abumaree
et al., 2006). It is now believed that this phenomenon may be an Allogeneic Marrow MSCs for GvHD
important component of feto-maternal tolerance and that effero- The first major industry-sponsored phase III trial of MSCs
cytosis of fetal apoptotic stromal trophoblastic cells by maternal (Prochymal) was for treatment of steroid-refractory GvHD
phagocytic lung macrophages may provide a mechanism to (NCT00366145) and completed in 2009. The primary endpoint
provide tolerance to the semi-allogeneic fetus (Askelund and of GvHD, complete remission at day 28 after infusion, was not

Cell Stem Cell 22, June 1, 2018 827


Cell Stem Cell

Perspective
Figure 1. MSC Fitness, Function, and Fate
Theorem
Culture-adapted fit MSCs express inflammation-
suppressing paracrine factors that augment
T-regulatory cell (Treg) function and M2 macro-
phage polarization as well as suppress effector
lymphoid cell function. MSCs are also habilitated
to produce morphogens and exosomes that pro-
mote tissue repair. The sum effect of these additive
functionalities is to drive tissue regeneration.
MSCs progressing to apoptosis express ‘‘eat me’’
signals such as phosphatidylserine (PtS) and are
susceptible to the alternate efferocytosis pathway,
where their engulfment by phagocytic macro-
phages leads to expression of immune tolerance
factors. The reciprocal relationship between
fitness and apoptosis dictates whether MSC
metabolism or their efferocytosis, respectively, is
responsible for their in vivo biological effects.

significantly increased relative to placebo (Martin et al., 2010). Day 28 OR was 69% and was significantly improved (p = 0.0003)
In 2013, the Prochymal assets were divested from Osiris Thera- compared with the protocol-defined historical control rate of
peutics to Mesoblast (Australia), who is now the sponsor of 45%. This outcome likely foreshadows the first Food and Drug
more than four active phase III studies examining the use of Pro- Administration (FDA)-approved MSC product in the United
chymal for Crohn’s disease (NCT00482092), chronic heart failure States. In the absence of robust predictive biomarkers of
(NCT02032004), back pain (NCT02412735), and pediatric GvHD response, the judicious use of clinical observation and subgroup
(NCT02336230). The common denominator in all of these studies analysis of responders and non-responders in early-phase
is the focused use of banked allogeneic, marrow-derived MSCs, clinical trials may inform the rational selection of patients enrolled
thawed and infused. The adaptive clinical trial design from the in advanced clinical trials to bias toward outcomes meeting pri-
original use of MSCs for GvHD (NCT00366145) to the recently mary clinical endpoints of success.
completed study of MSCs in pediatric GvHD (NCT02336230)
provides important insights into the importance of empirical clin- Autologous Marrow MSCs for Heart Failure
ical observation informing selective patient enrollment to meet Marrow-derived MSCs have also been tested in a phase III trial by
primary clinical endpoints. In the original unsuccessful indus- Celyad (Belgium) for treatment of chronic advanced ischemic
try-sponsored placebo-controlled study of Prochymal for treat- heart failure (NCT01768702). A critical distinction with the identi-
ment of steroid-resistant GvHD (NCT00366145), both children cally marrow-sourced Prochymal product is that the Celyad
and adults with any grade B–D GvHD were treated when ste- protocol utilized autologous marrow-sourced MSCs that were
roid-resistant for at least 3 days up to 14 days (Martin et al., subsequently polarized toward a ‘‘cardiopoietic’’ phenotype
2010). Since then, some of the best empirical insights into patient and administered without interval freezing between culture and
selection for MSCs therapy of GvHD have been obtained from endoventricular delivery of 600 million MSCs in subjects. An
the large number of GvHD patients having received MSCs either earlier Celyad-sponsored study performed with the same MSC
through clinical trials in the United States or Europe or through product in subjects with heart failure secondary to ischemic
hospital exemption in Europe. In GvHD, it has been observed cardiomyopathy (NCT00810238) was suggestive of improved
that children respond better to allogeneic MSCs than adults cardiac outcomes (Bartunek et al., 2013). However, results
overall (Kurtzberg et al., 2010) and that treating patients early published in 2016 of an adequately powered clinical trial
on is better than delaying therapy after onset of acute GvHD (NCT01768702) were unable to demonstrate a significant change
(Ball et al., 2013). Gut and liver GvHD are more responsive between the MSC and placebo groups from baseline and
than skin GvHD. In the absence of robust predictive biomarkers, 39 weeks in a hierarchical composite primary endpoint (Bartunek
these observations provide guidance in clinical trial design et al., 2016). Distinct from prior studies in this space, these trials
biased toward subject selection likely to be responders. These utilized metabolically fit and autologous marrow MSC products,
data likely informed an adaptive clinical trial design for mitigating any negative functional bias arising from immune in-
NCT02336230, where identical MSC products and dosing compatibility between the MSC product and recipient as well
schemes in both studies were maintained but the definition of as optimizing functionality. Here again issues arise because of
response, age of inclusion, severity of disease, and exclusion practical limits in cell dosing. Subjects received 600 million to
of skin-only GvHD as well as a more aggressive start time for 1.2 billion autologous MSCs resuspended in 10 mL and delivered
MSC transfusion were implemented (Table 2). The latter Meso- by an average of 18 separate 0.5-mL endoventricular injections at
blast-sponsored study of marrow MSCs in pediatric GvHD least 1 cm apart. The technical challenge in delivering MSCs
completed recruitment in December 2017. In February 2018, it in this study is likely an important headwind to achieving biolog-
was announced by press release that the study had successfully ical effect. The delivery of multiple endoventricular injections
met the primary endpoint of improved day 28 overall response to a heart of 0.5 mL each while circumventing areas of thin
(OR) in steroid-refractory pediatric subjects with severe disease. myocardium (<8 mm thickness) to avoid perforation of the

828 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective

Table 2. Clinical Trial Adaptive Design for the Study of Prochymal or Remestemcel-L for GvHD
Study NCT00366145 NCT02336230
Product/sponsor Prochymal/Osiris Therapeutics Remestemcel-L/Mesoblast
Cell dosing 2 million cells/kg twice a week for 4 weeks 2 million cells/kg twice a week for 4 weeks
Primary outcome complete GvHD response of greater than overall GvHD response rate on day 28 to include both complete
or equal to 28 days duration response and partial response; complete response (CR):
resolution of GvHD in all involved organs; partial response (PR):
organ improvement by at least one stage without worsening
of any other organ
Target enrollment 240 60
Ages eligible 6 months to 70 years 2 months and 17 years
GvHD grade at any grade B–D (international bone marrow transplant grade C or D GvHD involving the skin, liver, and/or
enrollment registry [IBMTR] grading) of acute GvHD gastrointestinal (GI) tract; grade B GvHD involving the liver
and/or GI tract with or without concomitant skin disease;
exclusion: grade B GvHD with skin-only involvement
Steroid-refractory no improvement after 3 days and a duration of no progression within 3 days or no improvement within
definition greater than 2 weeks 7 consecutive days
Study sites 70 23
Completed May 2009 February 2018
Meaningful study design adaptations in NCT02336230 are shown in italics.

ventricle would introduce substantial patient-to-patient vari- acquired by TiGenix in May 2011. The data obtained from
ability, considering that, for a disease like ischemic cardiomyop- NCT00475410 informed the adaptive design of a distinct,
athy with patchy scars often scattered all around the left ventricle, TiGenix-sponsored, phase III trial of culture-rescued ASCs for
this could have been a challenge. Thus, there is concern that the enterocutaneous fistular disease but with four meaningful
injections are concentrated in areas of good myocardium that changes: allogeneic ASCs were used rather than autologous
may increase the risk of disruption of the tissue and worsening ones; the cell dose was substantially increased to 120 million
outcomes. Indeed, post hoc analysis of ventricular remodeling cells; no fibrin glue matrix was used for intrafistular injection;
52 weeks after treatment of NCT01768702 study participants re- and only patients with Crohn’s disease were enrolled, whereas
vealed that the largest reverse remodeling was evident in patients these patients were excluded in the previous trial (Table 3).
receiving a moderate number of injections (i.e., <20). These data The resultant TiGenix-sponsored trial, NCT01541579, was
suggest an inverted U-shaped dose/response curve with worse completed in 2015 and represents the first unambiguously suc-
outcomes at higher dose delivery attempts. This counterintuitive cessful use of MSCs and/or ASCs in an advanced clinical trial.
observation informs that the number of injections may well be an Results were published in 2016, and it was shown that allogeneic
important factor in improving outcomes because more injections ASCs were significantly and substantially superior to placebo in
may cause potential myocardial damage through multiple mech- treating Crohn’s-associated perianal fistulas. Indeed, a signifi-
anisms, both mechanical and biological (Teerlink et al., 2017). cantly greater proportion of patients treated with ASCs versus
A path forward for this approach may well be to consider alternate placebo achieved remission of fistular disease 24 weeks after
means of augmenting the biology of infused MSCs as a method to treatment (50% versus 34%, p = 0.24) and suffered fewer
enhance potency, considering the practical limits of dosing via treatment-related adverse events (17%) compared with placebo
the endoventricular route. (29%) (Panés et al., 2016). These results were sustained for
at least 1 year after treatment as well (Panes et al., 2018).
‘‘Fit’’ Adipose MSCs for Enterocutaneous Fistular There are three meaningful distinctions that set this study
Disease apart from the precedent discussed negative MSC trials: MSC
Akin to the Celyad studies that utilized metabolically fit autolo- fitness, route of delivery, and dose intensity. First, batch-manu-
gous MSCs, Cellerix sponsored a study (NCT00475410) of autol- factured allogeneic, adipose-derived MSC cell doses were
ogous adipose MSCs (also known as adipose stromal cells retrieved from cryostorage and subsequently allowed to fully
[ASCs]) that were culture-revived for 48 hr prior to local injection recover in tissue culture for a few days before being shipped to
for treatment of complex perianal fistulas in patients without in- the point of care and administered to patients; second, 120
flammatory bowel disease. Up to 60 million ASCs were admixed million MSCs were formulated in a total volume of 24 mL and
with fibrin glue (or not), administered as multiple local injections delivered at the cutaneous site of disease (intrafistular) rather
in and around the fistula, and compared with fibrin glue alone as than transfused intravenously (i.v.), allowing for a local mass
the control. The study completed enrollment of 214 subjects allo- effect not otherwise achievable. Although the study made a pivot
cated to the three arms in 2009, and the ASC treatment groups from autologous ASCs used in NCT00475410 to allogeneic
were non-superior to fibrin glue alone when examining the pri- ASCs used in NCT01541579, likely to mitigate the cost of
mary endpoint of sustained closure and healing of fistulas goods as part of a marketing deployment strategy, the trial’s
6 months after treatment (Herreros et al., 2012). Cellerix was use of replication-fit MSCs parallels the approach used in murine

Cell Stem Cell 22, June 1, 2018 829


Cell Stem Cell

Perspective

Table 3. Clinical Trial Adaptive Design for the Study of ASCs for Enterocutaneous Perianal Fistular Disease
Study NCT00475410 NCT01541579
Product/sponsor Cx401/Cellerix Cx601/TiGenix
Source autologous ASCs allogeneic ASCs
Clinical indication adults with cryptoglandular complex fistula-in-ano adults with treatment-refractory complex perianal fistulas
excluding Crohn’s disease in patients with Crohn’s disease
Cell dosing 20 million cells + optional repeat of 40 million cells 120 million cells intralesionally in a single course of treatment
intralesionally (with and without fibrin glue) (24 mL in and around lesion), no fibrin glue
Primary outcome closure of fistula at 24 weeks and 1 year closure of fistula at 24 weeks and 2 years
Target enrollment 214 278
Study sites 19 49
Completed August 2009 July 2015
Meaningful study design adaptations in NCT01541579 are shown in italics.

pre-clinical models (Table 1) and also biases toward a local anti- driven informed guesses that are in part cognizant of pre-clinical
inflammatory effect through a cell dosing strategy that maxi- animal systems and in vitro cell biology (Galipeau et al., 2016).
mizes a locoregional paracrine MSC biology. Indeed, a substan- Their utility lies in defining a cell function signature that serves
tial number of European academic MSC clinical trials are testing as a yardstick against which distinct MSC manufacturing runs
the use of fit rather than thawed MSCs for systemic and local can be compared. A cogent argument can be made that the
use for osteoarticular and inflammatory disorders. Therein lies focus on MSC cell biology provides only part of the answer of
a pathway for clinical utility of MSCs where practical remedies predictive potency testing and that measuring patient parame-
maintain fitness and address the dose intensity required to ters that would be predictive of responsiveness to MSC therapy
achieve a biological endpoint. is likely as important. In industry-sponsored trials, where a large
number of patients are treated with an identical lot of allogeneic
The Importance of MSC Potency Assays and cells, outcomes, not surprisingly, vary between subjects. Vari-
Development of Biomarkers Predictive of Response ables such as severity of disease, co-morbidities, and other clin-
MSC identity and potency assays are a mandatory component ical parameters always affect the outcomes of any therapeutic
for advanced clinical trials and are required for securing an intervention. Considering that MSCs act in part indirectly through
FDA biologics license application (BLA) for marketing in the the in vivo licensing of host immune bystander cells, the respon-
United States (Mendicino et al., 2014). The core notion is that siveness of individuals to MSCs may be predicated by their idio-
these assays serve as means to validate the potency of distinct matic immune status at the time of infusion. It may well be that
MSC manufactured lots and are a guarantor of functionality pre- the examination of donor MSC and recipient leukocyte interac-
dictive of effectiveness in clinical practice. The extensive discov- tion in itself may serve as a biomarker of response. Similar as-
ery pre-clinical literature informs us that MSCs, similar to many says have been examined in solid organ transplantation to
cell platforms dependent on metabolic fitness for optimal utility, predict functional immune compatibility. Indeed, the advent of
affect outcomes in a multifunctional manner. Culture-adapted modern bone marrow transplantation was informed by the
MSCs likely mirror the homeostatic functionality of their endog- development of the mixed lymphocyte reaction by Fritz Bach
enous counterparts and likely deploy pharmaceutical properties in 1964 that paved the way for donor and recipient pairing
because of their interaction with host tissues and cells following permissive for good bone marrow transplant (BMT) outcomes
their bolus delivery in vivo. No singular physiological property is (Bach and Hirschhorn, 1964). A similar approach could be devel-
entirely predictive of mechanism of action, especially because oped where the MSC biological interplay on recipient lymphoid
most MSCs after intravenous infusion are likely subjected to or myeloid cells serves as a surrogate of host response to
phagocytosis and apoptosis and are cleared by tissue-resident MSC products. These latter data can only be inferred by post
perivascular macrophages within no more than a few days. hoc analysis of select biomarkers from ‘‘responders’’ and
Within that window of time, metabolically active MSCs, possibly ‘‘non-responders’’ enrolled in clinical trials that would be predic-
responsive to environmental cues, produce cytokines, chemo- tive of response to MSCs. The group, led by Francesco Dazzi at
kines, morphogens, and microparticles that alter the biology of King’s College London, recently provided compelling evidence
host bystander lymphomyeloid cells, and these likely mitigate that such an analysis may provide robust pre-treatment
tissue damage and loss (Figure 1). It is anybody’s guess which predictive values of response to MSC therapy (Galleu et al.,
combination of factors is most critical to MSC regenerative func- 2017). Indeed, in patients with GvHD, it was found that, when
tionality in humans, and the issue cannot be ethically resolved in recipient-derived CD8+ T cells and CD56+ natural killer (NK) cells
human subjects by the type of reductionist experiments readily were able to induce perforin- and granzyme-dependent
feasible in rodents. apoptosis of MSCs in vitro, it was predictive of at least a partial
The effort to map such in vitro potency assays, even in reduc- clinical response or better following transfusion of MSCs.
tionist systems, may not provide clear guidance on predictive These data suggest that, at least in GvHD, subjects poised to
potency assays. At best, potency assays mirror hypothesis- be responsive to MSC therapy display an enhanced innate

830 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective

cytotoxic reactivity to allogeneic MSCs. Whether such an Breakthrough Designation, and, more recently, the 21st Century
assay system would be predictive of MSC therapeutic utility for Cures Act, under which a drug is eligible for regenerative medi-
other inflammatory or tissue injury syndromes remains to be cine advanced therapy (RMAT) designation. An argument can
determined. be made that the creation of a novel regulatory path designed
to allow not-for-profit academic health centers to secure early
Headwinds to United States and European Union marketing approval (akin to the Japanese regulatory model) or
Marketing Approval and Sustainable Deployment of at least the ability to secure cost recovery of cell drug manufac-
MSC Therapies ture costs (akin to European hospital exemption) would serve
Clinical trials demonstrating an effect on clinical outcomes are the public good. Indeed, recent guidance from the FDA suggests
required to eventually secure FDA marketing approval and BLA such a path, where multiple manufacturers, which may be individ-
for reimbursement and sustainable large-scale clinical deploy- ual physicians or groups of physicians, enter into a cooperative
ment of MSCs. Although any accredited clinical center can development agreement (Marks and Gottlieb, 2018). These man-
readily adopt practice of medicine ‘‘minimal’’ cell therapy tech- ufacturers then produce the product at different sites according
nologies without FDA or European Medicines Agency (EMA) to the same protocol, which includes appropriate quality control
licensing requirements, the same is not true for adoption of procedures to help ensure consistency between different lots
more than minimally manipulated cell therapies, such as cul- produced at different sites. Patients are enrolled at each of the
ture-adapted MSCs. This regulation of cells as drugs favors a sites that are manufacturing the product in a multicenter clinical
development model driven by industry, where mass-produced trial protocol. When the data from the multicenter trial are
universal products amenable to batch manufacture with econo- analyzed to evaluate the safety and efficacy of the product, the
mies of scale are compatible with a sustainable business model. individual physicians or groups of physicians submit a BLA that
In contrast, there are many cell technologies that are likely to includes the manufacturing protocol used, the clinical data
have a substantial effect on patient outcomes whose develop- obtained at the individual site, and the results of the multicenter
ment will be overlooked by industry because of high cost of clinical trial, showing safety and efficacy. This ultimately results
goods, small market size, or highly specialized handling, such in the issuance of a site-specific biologics license for the product
as personalized MSC therapies, where a margin-driven business made by each physician or group of physicians. These types
model is less apparent. Further headwinds to deployment of of activities would complement the traditional industrial develop-
industrial MSC therapies include the $170,000 price point of ment and deployment of scalable MSC technologies.
MSC platforms for GvHD in Japan and how this informs the likely
pricing of analogous cell pharmaceuticals in the United States Conclusion
for pediatric GvHD. Cell drug affordability for marginally or un- There may well be very meaningful differences in MSC cell prep-
derinsured American patients will be a tremendous burden on aration, fitness, and functionality when comparing MSC tissue
the distributive justice notion embraced by many not-for-profit source, culture methods, and expansion levels. However,
academic healthcare institutions and hospital systems in the non-manufacturing variables, such as handling at the point of
United States and Europe. Alternate means of development care, including thawing, route of delivery, and dosing, may well
and deployment of promising but marginalized cell technologies override any MSC functionality differences. The recent success
are required to meet the public’s expectation of access to of using MSCs for treating fistular Crohn’s disease provides a
effective cell therapy. The remedy may well reside within bridgehead from which technical embellishments can be imple-
academic health centers affiliated with research-intensive mented to increase clinical utility. Pre-clinical animal data sup-
universities, which serve as the engines for discovery and port the notion that the use of augmented MSCs prior to infusion
development of cell technologies in a patient-beneficial manner, either by use of pharmaceutical or cytokine pre-activation
as espoused by the 21st Century Cures Act (United States 114th (Guess et al., 2017), genetic engineering, or reprogramming
Congress, 2016). enhances their pharmaceutical potency. The recognition of
In an effort to harness the technological benefits of this bio- MSC efferocytosis and its exploitation for immune modulation
pharmaceutical space, the Japanese FDA has put simplified reg- may provide novel translational strategies as well. Therein lies
ulatory requirements in place to expedite marketing approval of the path for MSC v2.0, where an understanding of potency and
cell technologies and therapies developed in Japan (Hara et al., failures informs engineered solutions in cell manufacturing,
2014). In essence, cell-based pharmaceuticals are granted banking, and point of care deployment and, importantly, rational
conditional marketing approval with evidence of safety and pre- selection of subjects based on clinical and biological parameters
sumption of efficacy, with a 7-year window to demonstrate scien- permissive to clinical effectiveness.
tifically sound efficacy. Early marketing approval allows for prod-
uct pricing and reimbursement that is permissive for SUPPLEMENTAL INFORMATION
development. In parallel, the European Medicines Agency has a
‘‘hospital exemption’’ clause that allows for not-for-profit public Supplemental Information includes one table and can be found with this article
institutions to make cellular therapies available whose online at https://doi.org/10.1016/j.stem.2018.05.004.

manufacturing costs, the lead headwind to development by aca-


ACKNOWLEDGMENTS
demic health centers, are borne by the public health payer sys-
tem. In the USA, there are 3 recent programs for accelerated
J.G. is the current, and L.S. is the former, Chair of the International Society of
approval at the end of a randomized double blind placebo Cell Therapy (ISCT) MSC Committee. This work was supported in part by NIH/
controlled (RDBPC) phase 2b trial, which include Fast Track, NIDDK R01DK109508 (to J.G.).

Cell Stem Cell 22, June 1, 2018 831


Cell Stem Cell

Perspective
AUTHOR CONTRIBUTIONS ronment of the hemopoietic tissues. Cloning in vitro and retransplantation
in vivo. Transplantation 17, 331–340.
J.G. wrote the paper and L.S. edited the paper.
Fung, M., Yuan, Y., Atkins, H., Shi, Q., and Bubela, T. (2017). Responsible
Translation of Stem Cell Research: An Assessment of Clinical Trial Registration
DECLARATION OF INTERESTS and Publications. Stem Cell Reports 8, 1190–1201.

The authors declare no competing interests. Galipeau, J. (2013). The mesenchymal stromal cells dilemma–does a negative
phase III trial of random donor mesenchymal stromal cells in steroid-resistant
graft-versus-host disease represent a death knell or a bump in the road?
REFERENCES Cytotherapy 15, 2–8.

Abumaree, M.H., Stone, P.R., and Chamley, L.W. (2006). The effects of Galipeau, J., Krampera, M., Barrett, J., Dazzi, F., Deans, R.J., DeBruijn, J.,
apoptotic, deported human placental trophoblast on macrophages: possible Dominici, M., Fibbe, W.E., Gee, A.P., Gimble, J.M., et al. (2016). International
consequences for pregnancy. J. Reprod. Immunol. 72, 33–45. Society for Cellular Therapy perspective on immune functional assays for
mesenchymal stromal cells as potency release criterion for advanced phase
Ankrum, J.A., Ong, J.F., and Karp, J.M. (2014). Mesenchymal stem cells: im- clinical trials. Cytotherapy 18, 151–159.
mune evasive, not immune privileged. Nat. Biotechnol. 32, 252–260.
Galleu, A., Riffo-Vasquez, Y., Trento, C., Lomas, C., Dolcetti, L., Cheung, T.S.,
Askelund, K.J., and Chamley, L.W. (2011). Trophoblast deportation part I: re- von Bonin, M., Barbieri, L., Halai, K., Ward, S., et al. (2017). Apoptosis in
view of the evidence demonstrating trophoblast shedding and deportation mesenchymal stromal cells induces in vivo recipient-mediated immunomodu-
during human pregnancy. Placenta 32, 716–723. lation. Sci. Transl. Med. 9, eaam7828.

Bach, F., and Hirschhorn, K. (1964). Lymphocyte Interaction: A Potential His- Guess, A.J., Daneault, B., Wang, R., Bradbury, H., La Perle, K.M.D., Fitch, J.,
tocompatibility Test in Vitro. Science 143, 813–814. Hedrick, S.L., Hamelberg, E., Astbury, C., White, P., et al. (2017). Safety Profile
of Good Manufacturing Practice Manufactured Interferon g-Primed Mesen-
Ball, L.M., Bernardo, M.E., Roelofs, H., van Tol, M.J., Contoli, B., Zwaginga, chymal Stem/Stromal Cells for Clinical Trials. Stem Cells Transl. Med. 6,
J.J., Avanzini, M.A., Conforti, A., Bertaina, A., Giorgiani, G., et al. (2013). Mul- 1868–1879.
tiple infusions of mesenchymal stromal cells induce sustained remission in
children with steroid-refractory, grade III-IV acute graft-versus-host disease. Hara, A., Sato, D., and Sahara, Y. (2014). New Governmental Regulatory
Br. J. Haematol. 163, 501–509. System for Stem Cell–Based Therapies in Japan. Ther. Innov. Regul. Sci. 48,
681–688.
Bartunek, J., Behfar, A., Dolatabadi, D., Vanderheyden, M., Ostojic, M., Dens,
J., El Nakadi, B., Banovic, M., Beleslin, B., Vrolix, M., et al. (2013). Cardio- Herreros, M.D., Garcia-Arranz, M., Guadalajara, H., De-La-Quintana, P., and
poietic stem cell therapy in heart failure: the C-CURE (Cardiopoietic stem Garcia-Olmo, D.; FATT Collaborative Group (2012). Autologous expanded ad-
Cell therapy in heart failURE) multicenter randomized trial with lineage-speci- ipose-derived stem cells for the treatment of complex cryptoglandular perianal
fied biologics. J. Am. Coll. Cardiol. 61, 2329–2338. fistulas: a phase III randomized clinical trial (FATT 1: fistula Advanced Therapy
Trial 1) and long-term evaluation. Dis. Colon Rectum 55, 762–772.
Bartunek, J., Davison, B., Sherman, W., Povsic, T., Henry, T.D., Gersh, B.,
Metra, M., Filippatos, G., Hajjar, R., Behfar, A., et al. (2016). Congestive Heart Hess, D.C., Wechsler, L.R., Clark, W.M., Savitz, S.I., Ford, G.A., Chiu, D.,
Failure Cardiopoietic Regenerative Therapy (CHART-1) trial design. Eur. J. Yavagal, D.R., Uchino, K., Liebeskind, D.S., Auchus, A.P., et al. (2017). Safety
Heart Fail. 18, 160–168. and efficacy of multipotent adult progenitor cells in acute ischaemic stroke
(MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial.
Bernardo, M.E., and Fibbe, W.E. (2013). Mesenchymal stromal cells: sensors Lancet Neurol. 16, 360–368.
and switchers of inflammation. Cell Stem Cell 13, 392–402.
House of Commons Canada (2016). Minutes of Proceedings of House of
Bianco, P., Cao, X., Frenette, P.S., Mao, J.J., Robey, P.G., Simmons, P.J., and Commons (Canada) Standing Committee on Health (2016). Published online
Wang, C.Y. (2013). The meaning, the sense and the significance: translating May 30, 2016. http://www.ourcommons.ca/DocumentViewer/en/42-1/HESA/
the science of mesenchymal stem cells into medicine. Nat. Med. 19, 35–42. meeting-12/evidence.
Caplan, A.I. (1991). Mesenchymal stem cells. J. Orthop. Res. 9, 641–650. JCR Pharmaceuticals (2017). Annual Report 2017. Published online
March 2017. http://www.jcrpharm.co.jp/wp2/wp-content/uploads/2017/10/
Carty, F., Mahon, B.P., and English, K. (2017). The influence of macrophages
JCR-AR2017_English_FINAL1.pdf.
on mesenchymal stromal cell therapy: passive or aggressive agents? Clin.
Exp. Immunol. 188, 1–11.
Kfoury, Y., and Scadden, D.T. (2015). Mesenchymal cell contributions to the
stem cell niche. Cell Stem Cell 16, 239–253.
Chinnadurai, R., Rajan, D., Qayed, M., Arafat, D., Garcia, M., Liu, Y., Kugatha-
san, S., Anderson, L.J., Gibson, G., and Galipeau, J. (2018). Potency Analysis
Koç, O.N., Gerson, S.L., Cooper, B.W., Dyhouse, S.M., Haynesworth, S.E.,
of Mesenchymal Stromal Cells Using a Combinatorial Assay Matrix Approach.
Caplan, A.I., and Lazarus, H.M. (2000). Rapid hematopoietic recovery after
Cell Rep. 22, 2504–2517.
coinfusion of autologous-blood stem cells and culture-expanded marrow
Dexter, T.M., Moore, M.A., and Sheridan, A.P. (1977). Maintenance of hemo- mesenchymal stem cells in advanced breast cancer patients receiving high-
poietic stem cells and production of differentiated progeny in allogeneic and dose chemotherapy. J. Clin. Oncol. 18, 307–316.
semiallogeneic bone marrow chimeras in vitro. J. Exp. Med. 145, 1612–1616.
Kurtzberg, J., Prasad, V., Grimley, M., Horn, B., Carpenter, P., Jacobsohn, D.,
Elliott, M.R., Koster, K.M., and Murphy, P.S. (2017). Efferocytosis Signaling and Prockop, S. (2010). Allogeneic human mesenchymal stem cell therapy
in the Regulation of Macrophage Inflammatory Responses. J. Immunol. 198, (Prochymal) as a rescue agent for severe treatment resistant GVHD in pedi-
1387–1394. atric patients. Biol. Blood Marrow Transplant. 16, S169.

Fibbe, W.E., Dazzi, F., and LeBlanc, K. (2013). MSCs: science and trials. Nat. Lalu, M.M., McIntyre, L., Pugliese, C., Fergusson, D., Winston, B.W., Marshall,
Med. 19, 812–813. J.C., Granton, J., and Stewart, D.J.; Canadian Critical Care Trials Group
(2012). Safety of cell therapy with mesenchymal stromal cells (SafeCell): a sys-
François, M., Romieu-Mourez, R., Li, M., and Galipeau, J. (2012). Human MSC tematic review and meta-analysis of clinical trials. PLoS ONE 7, e47559.
suppression correlates with cytokine induction of indoleamine 2,3-dioxyge-
nase and bystander M2 macrophage differentiation. Mol. Ther. 20, 187–195. Lazarus, H.M., Haynesworth, S.E., Gerson, S.L., Rosenthal, N.S., and Caplan,
A.I. (1995). Ex vivo expansion and subsequent infusion of human bone
Franks, L.M., Chesterman, F.C., and Rowlatt, C. (1970). The structure of tu- marrow-derived stromal progenitor cells (mesenchymal progenitor cells):
mours derived from mouse cells after ‘‘spontaneous’’ transformation in vitro. implications for therapeutic use. Bone Marrow Transplant. 16, 557–564.
Br. J. Cancer 24, 843–848.
Le Blanc, K., Frassoni, F., Ball, L., Locatelli, F., Roelofs, H., Lewis, I., Lanino, E.,
Friedenstein, A.J., Chailakhyan, R.K., Latsinik, N.V., Panasyuk, A.F., and Kei- Sundberg, B., Bernardo, M.E., Remberger, M., et al.; Developmental Commit-
liss-Borok, I.V. (1974). Stromal cells responsible for transferring the microenvi- tee of the European Group for Blood and Marrow Transplantation (2008).

832 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective
Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft- Phinney, D.G., Galipeau, J., Krampera, M., Martin, I., Shi, Y., and Sensebe, L.
versus-host disease: a phase II study. Lancet 371, 1579–1586. (2013). MSCs: science and trials. Nat. Med. 19, 812.

Luk, F., de Witte, S.F., Korevaar, S.S., Roemeling-van Rhijn, M., Franquesa, Poon, I.K., Lucas, C.D., Rossi, A.G., and Ravichandran, K.S. (2014). Apoptotic
M., Strini, T., van den Engel, S., Gargesha, M., Roy, D., Dor, F.J., et al. cell clearance: basic biology and therapeutic potential. Nat. Rev. Immunol. 14,
(2016). Inactivated Mesenchymal Stem Cells Maintain Immunomodulatory 166–180.
Capacity. Stem Cells Dev. 25, 1342–1354.
Reicin, C., McMahon, E., and Chung, C. (2012). Stem Cell Therapy Regulation
Marks, P., and Gottlieb, S. (2018). Balancing Safety and Innovation for Cell- in Canada: Implications of the Prochymal Approval. Westlaw Journal 28, 1–4.
Based Regenerative Medicine. N. Engl. J. Med. 378, 954–959. https://www.torys.com/Publications/Publications/AR2012-32.pdf.
Martin, P.J., Uberti, J.P., Soiffer, R.J., Klingemann, H., Waller, E.K., Daly, A.S.,
Herrmann, R.P., and Kebriaei, P. (2010). Prochymal Improves Response Rates Reinders, M.E., Dreyer, G.J., Bank, J.R., Roelofs, H., Heidt, S., Roelen, D.L.,
In Patients With Steroid-Refractory Acute Graft Versus Host Disease (SR- Zandvliet, M.L., Huurman, V.A., Fibbe, W.E., van Kooten, C., et al. (2015).
GVHD) Involving The Liver And Gut: Results Of A Randomized, Placebo- Safety of allogeneic bone marrow derived mesenchymal stromal cell therapy
Controlled, Multicenter Phase III Trial In GVHD. Biol. Blood Marrow Transplant. in renal transplant recipients: the neptune study. J. Transl. Med. 13, 344.
16, S169–S170.
Sensebé, L., and Fleury-Cappellesso, S. (2013). Biodistribution of mesen-
Mendicino, M., Bailey, A.M., Wonnacott, K., Puri, R.K., and Bauer, S.R. (2014). chymal stem/stromal cells in a preclinical setting. Stem Cells Int. 2013,
MSC-based product characterization for clinical trials: an FDA perspective. 678063.
Cell Stem Cell 14, 141–145.
Sensebé, L., Tarte, K., Galipeau, J., Krampera, M., Martin, I., Phinney, D.G.,
Moll, G., Geißler, S., Catar, R., Ignatowicz, L., Hoogduijn, M.J., Strunk, D., and Shi, Y.; MSC Committee of the International Society for Cellular Therapy
Bieback, K., and Ringdén, O. (2016). Cryopreserved or Fresh Mesenchymal (2012). Limited acquisition of chromosomal aberrations in human adult mesen-
Stromal Cells: Only a Matter of Taste or Key to Unleash the Full Clinical Poten- chymal stromal cells. Cell Stem Cell 10, 9–10, author reply 10–11.
tial of MSC Therapy? Adv. Exp. Med. Biol. 951, 77–98.
Shanks, N., Greek, R., and Greek, J. (2009). Are animal models predictive for
Nauta, A.J., Westerhuis, G., Kruisselbrink, A.B., Lurvink, E.G., Willemze, R., humans? Philos. Ethics Humanit. Med. 4, 2.
and Fibbe, W.E. (2006). Donor-derived mesenchymal stem cells are immuno-
genic in an allogeneic host and stimulate donor graft rejection in a nonmyeloa- Sipp, D. (2015). Conditional approval: Japan lowers the bar for regenerative
blative setting. Blood 108, 2114–2120. medicine products. Cell Stem Cell 16, 353–356.
Németh, K., Leelahavanichkul, A., Yuen, P.S., Mayer, B., Parmelee, A., Doi, K.,
Teerlink, J.R., Metra, M., Filippatos, G.S., Davison, B.A., Bartunek, J., Terzic,
Robey, P.G., Leelahavanichkul, K., Koller, B.H., Brown, J.M., et al. (2009).
A., Gersh, B.J., Povsic, T.J., Henry, T.D., Alexandre, B., et al.; CHART
Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent
Investigators (2017). Benefit of cardiopoietic mesenchymal stem cell therapy
reprogramming of host macrophages to increase their interleukin-10 produc-
on left ventricular remodelling: results from the Congestive Heart Failure
tion. Nat. Med. 15, 42–49.
Cardiopoietic Regenerative Therapy (CHART-1) study. Eur. J. Heart Fail. 19,
Panés, J., Garcı́a-Olmo, D., Van Assche, G., Colombel, J.F., Reinisch, W., 1520–1529.
Baumgart, D.C., Dignass, A., Nachury, M., Ferrante, M., Kazemi-Shirazi, L.,
et al.; ADMIRE CD Study Group Collaborators (2016). Expanded allogeneic Turner, L., and Knoepfler, P. (2016). Selling Stem Cells in the USA: Assessing
adipose-derived mesenchymal stem cells (Cx601) for complex perianal fis- the Direct-to-Consumer Industry. Cell Stem Cell 19, 154–157.
tulas in Crohn’s disease: a phase 3 randomised, double-blind controlled trial.
Lancet 388, 1281–1290. United States 114th Congress (2016). H.R.34 – 21st Century Cures Act.
Published online December 13, 2016. https://www.congress.gov/bill/114th-
Panes, J., Garcia-Olmo, D., Van Assche, G., Colombel, J.F., Reinisch, W., congress/house-bill/34/text.
Baumgart, D.C., Dignass, A., Nachury, M., Ferrante, M., Kazemi-Shirazi, L.,
et al.; ADMIRE CD Study Group Collaborators (2018). Long-term Efficacy Wang, Y., Chen, X., Cao, W., and Shi, Y. (2014). Plasticity of mesenchymal
and Safety of Stem Cell Therapy (Cx601) for Complex Perianal Fistulas in stem cells in immunomodulation: pathological and therapeutic implications.
Patients With Crohn’s Disease. Gastroenterology 154, 1334–1342.e4. Nat. Immunol. 15, 1009–1016.

Cell Stem Cell 22, June 1, 2018 833


Cell Stem Cell, Volume 22

Supplemental Information

Mesenchymal Stromal Cells:


Clinical Challenges and Therapeutic Opportunities
Jacques Galipeau and Luc Sensébé
Table S1. Industry Sponsored Phase III studies with MSCs, Related to Table 1 

ClinicalTrials.gov Sponsor Status Official Title Target MSC Dose/route Primary Outcome Measure Study outcome and
Identifier: enrollment source Reference
1 NCT00366145 Osiris Completed A Phase III, Randomized, Double Blind, 240 Marrow, 2 million cells/kg twice a week for 4 Complete Response of greater than or The overall response
Therapeutics May 2009 Placebo-Controlled Study to Evaluate the allogeneic weeks/intravenous equal to 28 days duration rate with Prochymal
Efficacy and Safety of Prochymal (Ex Vivo [ Time Frame: Day 100 ] was 82% vs 73% for
Cultured Adult Human Mesenchymal Stem placebo p=0.12.
Cells) Infusion for the Treatment of Steroid Study data presented
Refractory Acute GVHD as an abstract in
February 2010
supplement issue of
Biology of Blood and
Marrow
Transplantation.

There is no referenced
publication of
NCT00366145 study
outcome
2 NCT00543374 Mesoblast, Ltd. Discontinued A Phase III, Multicenter, Placebo- 98 Marrow, Intravenous infusion four times over two weeks; This was an extension
2009 controlled, Randomized, Double-blind allogeneic possibly repeated once. Two treatment groups Duration of clinical benefit (Crohn's study offered only to
Durability and Retreatment Study to receiving either a total of 1200 million (high dose) disease activity index) [ Time Frame: 6 those subjects who
Evaluate the Safety and Efficacy of or 600 million (low dose) cells months ] successfully achieved
PROCHYMAL® (ex Vivo Cultured Adult clinical benefit
Human Mesenchymal Stem Cells) Re-induction of clinical benefit (Crohn's (reduction of CDAI of
Intravenous Infusion for the Maintenance disease activity index) [ Time Frame: 6 at least 100 points) in
and Re-induction of Clinical Benefit and months ] NCT00482092. The
Remission in Subjects Experiencing study was
Treatment-refractory Moderate-to-severe discontinued to
Crohn's Disease (NCT00482092) remove the potential
for bias for study
NCT01233960 started
in 2010.

There is no referenced
publication of
NCT00543374 study
outcome
3 NCT00562497 Mesoblast, Ltd. Completed A Phase III, Randomized, Double-Blind, 184 Marrow, Intravenous infusion four times during the first two There is no referenced
April 2009 Placebo-Controlled Study to Evaluate the allogeneic weeks (twice weekly), then two infusions Achieved an induction of a complete publication of
Efficacy and Safety of Prochymal™ administered during the next two weeks (once response, Followed by 28 days of NCT00562497 study
Infusion in Combination With weekly). Subjects assigned to the active maintenance of a clinically meaningful outcome
Corticosteroids for the Treatment of Newly treatment group will receive Prochymal™ at 2 x response that does not did not require
Diagnosed Acute GVHD 106 hMSC/kg per infusion. an increase in corticosteroid dose , did
not require second line therapy and
Subjects must be randomized and treated with survived 90 days. [ Time Frame: 90
corticosteroid (1-2 mg/kg/d methylprednisolone, Days ]
or equivalent) and Prochymal™/placebo within 72
hours of onset of acute GVHD.
4 NCT00475410 Cellerix Completed A Phase III Multicenter, Single Blind, 214 Adipose, Cx401 is a cell suspension of human expanded Closure of fistulas defined as absence NCT00475410 study
NCT01020825 August 2009 Randomized, Comparative and add-on autologous adipose-derived stem cells of autologous origin. of suppuration and re-epithelization of did not meet primary
Clinical Trial, in Three Parallel Groups, to The cells will be given at a dose of 20 million cells the external opening and absence of endpoint of efficacy
Evaluate Efficacy and Safety of a New alone or with fibrin glue for intralesional injection collections>2 cm directly related to the and published in July
Therapy With Adipose-derived Autologous (5 mL). If fistula remained open at 12 weeks, a fistula tract treated, as measured by 2012 (Herreros et al.,
Stem Cells for the Treatment of Complex second dose of 40 million ASCs was MRI (healing) [Time Frame: weeks 1, 4, 2012)
Perianal Fistulas in Patients Without administered. 12 and 24. Week 26 in patients with a
Inflammatory Bowel Disease second dose ]
5 NCT01041001 Medipost Co Completed Randomized, Open-Label, Multi-Center 104 Umbilical mesenchymal stem cells are mixed with semi- ICRS Cartilage Repair Assessment will There is no referenced
Ltd. January 2011 and Phase 3 Clinical Trial to Compare the Cord solid polymer, and administered into the cartilage follow to determine the appropriate publication of
Efficacy and Safety of Cartistem® and tissue lesion by orthopedic surgery with a single grade. The treatment will be considered NCT01041001 study
Microfracture in Patients With Knee dose of 500㎕/㎠ of cartilage defect efficacious if the ICRS grade drops by outcome
Articular Cartilage Injury or Defect at least 1 grade or more from baseline
to week 48. [ Time Frame: Week 0 and
48 ]

6 NCT00810238 Celyad Completed C-Cure - Safety, Feasibility and Efficacy of 47 Marrow, 0,6 to 1,2 x 109 cells resuspended in 10 mLs Change in left ventricular ejection NCT00810238
(formerly January 2012. Guided Bone Marrow-derived autologous delivered to endomyocardium in 9 to 26 injections fraction [ Time Frame: 6 months ] demonstrated a 7%
named Cardio3 Mesenchymal Cardiopoietic Cells for the of 0,5 mL at least 0,5 cm apart increase in the LVEF
BioSciences) Treatment of Heart Failure Secondary to with cell therapy from
Ischemic Cardiomyopathy 27.5% (95%
confidence interval
[CI]: 25.5% to 29.5%)
to 34.5% (95% CI:
32.5% to 36.6%) (n =
21, p; 0.0001). Study
outcome was
published in 2013
(Bartunek et al., 2013)
7 NCT01626677 Medipost Co Completed Long Term Follow-Up Study of 103 Umbilical mesenchymal stem cells are mixed with semi- There is no referenced
Ltd. May 2015 CARTISTEM® Versus Microfracture for Cord solid polymer, and administered into the cartilage Degree of improvement in knee publication of
the Treatment of Knee. 60 month tissue lesion by orthopedic surgery with a single assessments [IKDC (International Knee NCT01626677 study
outcome of patients enrolled in Documentation Committee); Pain score outcome
dose of 500㎕/㎠ of cartilage defect on VAS (Visual Analogue Scale);
NCT01041001
WOMAC (Western Ontario and
McMaster Universities Arthritis Index)
compared to the active control
(microfracture) [ Time Frame: 36
months, 48 months, and 60 months ]

8 NCT01541579 Cellerix Completed A Phase III, Randomized, Double Blind, 278 Adipose, Cx601 is a cell suspension in aseptic buffered Combined Remission of perianal NCT00810238 study
(TiGenix July 2015 Parallel Group, Placebo Controlled, allogeneic solution containing human expanded adipose- fistulising Crohn's disease defined as met primary outcomes
S.A.U.) Multicentre Study to Assess Efficacy and derived stem cells (eASCs) of allogeneic origin in the clinical assessment of closure of all and published in
Safety of Expanded Allogeneic Adipose- disposable vials with no preservative agents. The treated external openings (EO) that September 2016
derived Stem Cells (eASCs) for the cells will be given at a dose of 120 million cells (5 were draining at baseline despite gentle (Panes et al., 2016)
Treatment of Perianal Fistulising Crohn's million cells / mL) for intralesional injection. (24 finger compression at week 24, and with one year follow-
Disease Over a Period of 24 Weeks and mL) absence of collections > 2 cm of the up published in
an Extended Follow-up Period up to 104 treated perianal fistulas confirmed by December 2017
Weeks. centrally blinded MRI assessment by (Panes et al., 2018)
week 24. [ Time Frame: 24 weeks ]
European Commission
approval of Alofisel®
(darvadstrocel),
previously Cx601,
announced on March
24, 2018
9 NCT01768702 Celyad Completed Efficacy and Safety of Bone Marrow- 315 Marrow, 600 × 106 bone marrow-derived and lineage- Change between groups from baseline NCT01768702 study
(formerly August 2017 derived Mesenchymal Cardiopoietic Cells autologous directed autologous cardiopoietic stem cells and 39 weeks in a hierarchical did not meet primary
named Cardio3 (C3BS-CQR-1) for the Treatment of administered via a retention-enhanced composite outcome comprising, from outcome published in
BioSciences) Chronic Advanced Ischemic Heart Failure. intramyocardial injection catheter most to least severe outcome, days to February 2016
death from any cause, number of (Bartunek et al., 2016)
worsening of heart failure events,
change in score for the Minnesota Post Hoc analysis
Living with Heart Failure Questionnaire demonstrated that
(MLHFQ) (10-point deterioration, no LVEDV and LVESV of
meaningful change,10-point treated patients
improvement), change in six-minute decreased by 17.0 mL
walk distance (40-m deterioration, no and 12.8 mL greater
meaningful change, 40-m improvement) than controls (P=0.006
and change in left ventricular end and P=0.017,
systolic volume (15-mL deterioration, respectively at 1 year.
no meaningful change, 15-mL (Teerlink et al., 2017)
improvement), and left ventricular
ejection fraction (4% absolute
deterioration, no meaningful change,
4% absolute improvement).
[ Time Frame: 39 weeks post-index ]

10 NCT02336230 Mesoblast, Ltd. Completed A Prospective Study of Remestemcel-L, 60 Marrow, Intravenous remestemcel-L at a dose of 2 x 106 To evaluate the efficacy of In February 2018 it
February Ex-vivo Cultured Adult Human allogeneic MSC/kg (actual body weight at screening) twice remestemcel-L in pediatric subjects was announced that
2018 Mesenchymal Stromal Cells, for the per week for each of 4 consecutive weeks. with Grades B-D aGVHD who have the study had
Treatment of Pediatric Patients Who Have Infusions will be administered at least 3 days failed to respond to steroid treatment successfully met the
Failed to Respond to Steroid Treatment for apart and no more than 5 days apart for any post allogeneic HSCT (Overall primary endpoint of
Acute GVHD infusion. Response Rate) [ Time Frame: from improved Day 28
day of first infusion until 100 days post Overall Response in
first infusion ] steroid-refractory
pediatric subjects with
The primary endpoint for the population severe disease Day 28
under investigation (Subjects may have OR was 69% as was
Grades C and D aGVHD involving the significantly improved
skin, liver and/or gastrointestinal (GI) (p=0.0003) compared
tract or Grade B aGVHD involving the to protocol-defined
liver and/or GI tract, with or without historical control rate
concomitant skin disease.) is the of 45%.
Overall Response Rate at Day 28 post
initiation of therapy (Day 0) with No results published
remestemcel-L., defined as to include
both complete response and partial
response:
11 NCT00482092 Mesoblast Ltd Active since A Phase III, Multicenter, Placebo- 330 Marrow, Intravenous infusion of suspension of adult Disease remission (CDAI at or below In March 2009, Osiris
June 2007, controlled, Randomized, Double-blind allogeneic human mesenchymal stem cells, total of 1200 150) [ Time Frame: 28 days ] Therapeutics, Inc.
but not Study to Evaluate the Safety and Efficacy million (high dose) or 600 million (low dose) cells announced that it had
recruiting of PROCHYMAL® (ex Vivo Cultured Adult infused in four visits over two weeks elected to end
participants Human Mesenchymal Stem Cells) enrollment at 210
since March Intravenous Infusion for the Induction of patients in its Phase III
2009 Remission in Subjects Experiencing trial evaluating
Treatment-refractory Moderate-to-severe Prochymal for Crohn's
Crohn's Disease disease. The
Company believed
there was a design
flaw in the trial
resulting in
significantly higher
than expected placebo
response rates. The
decision was made
after the trial's final
scheduled interim
analysis showed that
one of the two
Prochymal dose arms
had crossed a futility
boundary. The dose
arm was unlikely to
achieve the primary
endpoint of remission
because of the high
placebo response rate.

There is no referenced
publication of
NCT00482092 study
outcome
12 NCT01233960 Mesoblast, Ltd. Active since A Multicenter, Open-label Study to 120 Marrow, Subjects will receive infusions of PROCHYMAL Disease remission [ Time Frame: 180 The Crohn's program
November Evaluate the Safety of PROCHYMAL® allogeneic on Day 42, Day 84, and Day 126 after initial Days after first infusion in Protocol 603 ] consisted of two linked
2010, but not (Remestemcel-L) Intravenous Infusion in infusion of PROCHYMAL in NCT00482092. Each CDAI at or below 150 and increase in trials - one aimed at
recruiting Subjects Who Have Received Previous infusion will contain 200 million cells. As subjects IBDQ inducing remission
participants. Remestemcel-L Induction Treatment for will be required to be in NCT00482092 during the (NCT00482092) and
Treatment-refractory Moderate-to-severe entire duration of their participation in the other at
Crohn's Disease (protocol 03) NCT01233960, all concomitant medication and maintaining response
safety information will be monitored by (NCT00543374) in
NCT01233960 and the combination of data from patients who had
the two protocols failed other available
treatments for the
disease. Enrollment
was suspended in
2009 over concerns
the trial design would
make it difficult to
detect a treatment
effect. The trial
remained blinded,
however, to permit an
interim analysis of all
207 patients enrolled
in the study. The
results showed that
despite the initial
concerns, the effect
size, or difference
between the
Prochymal and
placebo response
rates, of one dose arm
of Prochymal is
consistent with the
original statistical
assumptions of the
protocol and is
significantly
outperforming
placebo. The decision
to resume enrollment
was made following
discussions with the
Food and Drug
Administration (FDA)
about the results of the
interim analysis.
Enrollment was to
continue with the best-
performing Prochymal
dose arm and the
placebo arm,
according to the pre-
specified adaptive trial
design. The trial was
been repowered to
compensate for the
statistical penalty
incurred by the interim
analysis in the ITT
population. The follow-
on maintenance trial
(NCT00543374) was
discontinued to
remove the potential
for bias.

There is no referenced
publication of
NCT01233960 study
outcome
13 NCT01676441 Pharmicell Co., Active since A Phase II/III Clinical Trial to Evaluate the 32 Marrow, After posterior cervical laminectomy, 1.6X107 and Motor Score of the American Spinal No results posted
Ltd. August 2012, Safety and Efficacy of Bone Marrow- autologous 3.2 X107 autologous MSCs is injected into the Injury Association (ASIA) scale
but not derived Mesenchymal Stem Cell intramedullary and intrathecal space respectively. [ Time Frame: 12months ]
recruiting Transplantation in Patients With Chronic
Spinal Cord Injury. Phase I study published in May 2012 PMID:
22127044
14 NCT01716481 Pharmicell Co., Recruiting Intravenous Administration of Autoserum- 60 Marrow, Intravenous infusion of 1 × 106 cells/kg (maximum Categorical shift in modified Rankin No results posted
Ltd. since October cultured Autologous Mesenchymal Stem autologous 1.2 × 108) through the antecubital vein with 5 × scale (mRS) [ Time Frame: 90 days
2012 Cells in Ischemic Stroke: A Single Center, 106 cells/mL of normal saline over 10 minutes. after the cell treatment ]
Randomized, Open Label, Prospective,
Phase 3 Study PROBE Study published in October 2013 PMID:
24083670
15 NCT01652209 Pharmicell Co., Recruiting A Multi-center, Open-label, Comparison 135 Marrow, Within 30 days (+ / -7 days) after aspirating bone- LVEF by MRI [ Time Frame: 13 months No results posted
Ltd. since and a Parallel Group Study (3 Groups) autologous marrow, approximately 1×10^6/kg after the cell treatment ]
September Phase 3 Clinical Trial for a Comparative mesenchymal stem cells are administered into the
2013 Evaluation With the Existing Treatments, infarct coronary artery using balloon tipped
in Order to Verify the Long-term Efficacy catheter vs 2 doses 30 days apart
and Safety of the First Cell Treatment
Using Hearticellgram-AMI(Autologous
Human Bone Marrow Derived
Mesenchymal Stem Cells) in AMI Patients,
and to Observe the Efficacy of the Second
Cell Treatment.

16 NCT02032004 Mesoblast, Inc Recruiting Phase III Double-blind, Randomized, 600 Marrow, 150 million cells once/ transendocardial Time to non-fatal recurrent first In April 2017, the trial
since January Sham-procedure-controlled, Parallel- allogeneic decompensated heart failure (HF) achieved a successful
2014 Group Efficacy and Safety Study of events in the presence of terminal HF- pre-specified interim
Allogeneic Mesenchymal Precursor Cells related major adverse cardiac events futility analysis of the
(Rexlemestrocel-L) in Chronic Heart (HF-MACE) [ Time Frame: 12 Month efficacy endpoint in
Failure Due to LV Systolic Dysfunction minimum ] the first 270 patients of
(Ischemic or Nonischemic) (DREAM HF-1) the 600-patient trial
and an independent
data monitoring
committee formally
recommended the
continuation of the
trial.

No results posted
17 NCT02412735 Mesoblast, Ltd. Recruiting A Prospective, Multicenter, Randomized, 360 Marrow, 2.0 mL formulation of approximately 6 million Treatment Success (composite No results posted
since March Double-blind, Placebo-controlled Study to allogeneic rexlemestrocel-L cells - Intervention will be responder analysis of low back pain
2015 Evaluate the Efficacy and Safety of a injected into the painful intervertebral disc (with Visual Analogue Scale (VAS) score,
Single Injection of Rexlemestrocel-L Alone and without HA) Oswestry Disability Index (ODI) score
or Combined With Hyaluronic Acid (HA) in and no post-treatment interventions)
Subjects With Chronic Low Back Pain [ Time Frame: 24 Months ]
18 NCT03258164 Stemform Active as of Fat Transplantation Enriched With 32 Adipose, ASC injected into the breast tissue or face, both Change in volume over time after No results posted
March 2017, Expanded Adipose-derived Autologous autologous for cosmetic use. Dosing information not breast augmentation [ Time Frame: 12
but not Mesenchymal Stromal Cells in Cosmetic available on clinicaltrials.gov months ]
recruiting Breast Augmentation and Cosmetic Facial
participants Filling
19 NCT03280056 Brainstorm-Cell Recruiting A Phase 3, Randomized Double-Blind, 200 Marrow, Three Intrathecal administrations of NurOwn® To evaluate the efficacy and safety of No results posted
Therapeutics since Placebo-Controlled Multicenter Study to autologous (MSC-NTF cells) at bi-monthly intervals. Dosing NurOwn® (autologous MSC-NTF cells)
September Evaluate Efficacy and Safety of Repeated information not available on clinicaltrials.gov as compared to placebo as measured
2017 Administration of NurOwn® (Autologous by the amyotrophic lateral sclerosis
Mesenchymal Stem Cells Secreting functional rating scale (ALSFRS-R)
Neurotrophic Factors) in Participants With [ Time Frame: 28 weeks following the
ALS first treatment ]

S-ar putea să vă placă și