Basic Science for Clinicians Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative ...

Page created by Ann Avila
 
CONTINUE READING
Basic Science for Clinicians Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative ...
Basic Science for Clinicians

  Progress toward the Clinical Application of
  Mesenchymal Stromal Cells and Other
  Disease-Modulating Regenerative Therapies: Examples
  from the Field of Nephrology
  LaTonya J. Hickson,1 Sandra M. Herrmann,2 Bairbre A. McNicholas,3,4,5 and Matthew D. Griffin                     3,4

  Abstract
  Drawing from basic knowledge of stem-cell biology, embryonic development, wound healing, and aging, re-
  generative medicine seeks to develop therapeutic strategies that complement or replace conventional treatments
  by actively repairing diseased tissue or generating new organs and tissues. Among the various clinical-translational
  strategies within the field of regenerative medicine, several can be broadly described as promoting disease
  resolution indirectly through local or systemic interactions with a patient’s cells, without permanently integrating
  or directly forming new primary tissue. In this review, we focus on such therapies, which we term disease-
  modulating regenerative therapies (DMRT), and on the extent to which they have been translated into the clinical
  arena in four distinct areas of nephrology: renovascular disease (RVD), sepsis-associated AKI (SA-AKI), diabetic
  kidney disease (DKD), and kidney transplantation (KTx). As we describe, the DMRT that has most consistently
  progressed to human clinical trials for these indications is mesenchymal stem/stromal cells (MSCs), which potently
  modulate ischemic, inflammatory, profibrotic, and immune-mediated tissue injury through diverse paracrine
  mechanisms. In KTx, several early-phase clinical trials have also tested the potential for ex vivo–expanded
  regulatory immune cell therapies to promote donor-specific tolerance and prevent or resolve allograft injury.
  Other promising DMRT, including adult stem/progenitor cells, stem cell–derived extracellular vesicles, and
  implantable hydrogels/biomaterials remain at varying preclinical stages of translation for these renal conditions.
  To date (2021), no DMRT has gained market approval for use in patients with RVD, SA-AKI, DKD, or KTx, and
  clinical trials demonstrating definitive, cost-effective patient benefits are needed. Nonetheless, exciting progress in
  understanding the disease-specific mechanisms of action of MSCs and other DMRT, coupled with increasing
  knowledge of the pathophysiologic basis for renal-tissue injury and the experience gained from pioneering early-
  phase clinical trials provide optimism that influential, regenerative treatments for diverse kidney diseases will
  emerge in the years ahead.
                                     KIDNEY360 2: 542–557, 2021. doi: https://doi.org/10.34067/KID.0005692020

  Introduction                                                         manufacturing procedures to reverse disease more
  Since it was first introduced into biomedical par-                    effectively than can currently be achieved by con-
  lance by William Haseltine, the term regenerative                    ventional pharmacotherapy and interventional
  medicine has become broadly recognizable to health-                  procedures (1,2).
  care providers and the general public alike. After two                  For the nephrologist, the promise of regenerative
  decades of intense interest and diverse research initia-             medicine is compelling. Most acute and chronic kidney
  tives, the original concept of “an approach to therapy               diseases remain incurable, life limiting, and are typi-
  that...employs human genes, proteins and cells to re-                cally managed with drug combinations or procedures
  grow, restore or provide mechanical replacements for                 that are costly and carry a high burden of adverse
  tissues that have been injured by trauma, damaged                    effects. Added to this is the nephrologist’s natural
  by disease or worn by time” still conveys a succinct                 affinity for the application of cellular and physio-
  and valid definition of the field (1). Critically, one of              logic science to patient management. In this article,
  the central tenets of regenerative medicine has been                 we describe recent progress within a specific aspect
  the merging of basic insights into organ/tissue devel-               of regenerative medicine, which we term “disease-
  opment, stem-cell science, and disease pathophysi-                   modulating regenerative therapies” (DMRT), in the field
  ology with innovative translational concepts and                     of nephrology. In focusing on DMRT, we specifically
  1
    Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, Florida
  2
    Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
  3
    Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of
  Ireland Galway, Ireland
  4
    Nephrology Services, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland
  5
    Critical Care Services, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland

  Correspondence: Prof. Matthew D. Griffin, Biomedical Sciences, Regenerative Medicine Institute, National University of Ireland Galway,
  Corrib Village, Dangan, Galway, H91 TK33, Ireland. Email: matthew.griffin@nuigalway.ie

542    Copyright © 2021 by the American Society of Nephrology                                                                www.kidney360.org Vol 2 March, 2021
Basic Science for Clinicians Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative ...
KIDNEY360 2: 542–557, March, 2021                                   Disease-Modulating Regenerative Therapies in Nephrology, Hickson et al.     543

refer to therapeutic concepts based on systemic or localized            DMRT have made varying degrees of progress along a sim-
administration of cells, subcellular components, biomateri-             ilar translational path: (1) regulatory T cells (T-reg) and
als, or combinations of these that engage in a complex                  other immunologic cells, which suppress inflammation or
molecular crosstalk with resident cells and tissues of the              harmful immune responses (8,9); (2) additional types of
host to modify or reprogram damaging biologic activity. We              adult progenitor cells (including those derived from the
distinguish DMRT from other regenerative strategies that                kidney) with prorepair paracrine effects (10,11); (3) stem/
are based on harnessing pluripotent/multipotent stem                    progenitor cell–derived extracellular vesicles (EVs), which
cells and advances in tissue engineering to directly repair             may transfer proregenerative biomolecules to target cells
or replace damaged organs and tissue. This aspect of re-                (12); and (4) injectable hydrogels and other biomaterials,
generative medicine, which might be termed “organ- and                  which may have inherent regenerative properties or serve to
tissue-replacing regenerative therapies,” will not be                   enhance the benefits of cell-based therapies (13). As sum-
addressed in this article but has been expertly reviewed                marized in Figure 1 and described in detail in subsequent
by others (3–5).                                                        sections of the review, we focus on the progress toward
   First and foremost among DMRT are mesenchymal stem/                  clinical translation of MSCs and other DMRT that has oc-
stromal cells (MSCs), the subject of two decades of trans-              curred to date in four important areas of clinical practice in
lational research, which have been administered safely to               nephrology: renovascular disease (RVD), sepsis-associated
patients in numerous clinical trials (6,7). Critically, MSCs are        AKI (SA-AKI), diabetic kidney disease (DKD), and kidney
now considered to mediate their therapeutic benefits pre-                transplantation (KTx). For each of these exemplars, we
dominantly through inducible secretion of paracrine medi-               highlight the extent to which early-phase clinical trial expe-
ators and reprogramming of myeloid and lymphoid im-                     riences with DMRT are being driven by increased under-
mune cells, and can be expanded in culture to large numbers             standing of their potential mechanisms of action and are
from a range of autologous or allogeneic tissue sources (6).            linked to advances in knowledge of the pathophysiologic
In addition to MSC-based cell therapies, other forms of                 basis of the targeted condition.

                                                                                                       nslation
                                                                                           Clinical Tra
                                                                             Progress to

                                          Basic Mechanistic   Pre-clinical Development of      Case Series /       Phase 2/3        Market Approval
                                               Studies      Disease Models Clinical Products   Phase 1 Trials     Clinical Trials   for Clinical Use

                               RVD
  Mesenchymal                  SA-AKI
  Stromal Cells
                               DKD
                               KTx

                               RVD
   Regulatory                  SA-AKI
  Immune Cells
                               DKD
                               KTx

                               RVD
   Other Adult                 SA-AKI
 Progenitor Cells
                               DKD
                               KTx

                               RVD
   Extracellular               SA-AKI
    Vesicles
                               DKD
                               KTx

                               RVD
 Hydrogels and
                               SA-AKI
Other Biomaterials
                               DKD
                               KTx

Figure 1. | Variable progress has been made toward clinical translation of five categories of disease-modulating regenerative therapies
(DMRT) for four different areas of nephrology practice. The figure summarizes current translational status of mesenchymal stromal/stem cells,
regulatory immune cells, other adult progenitor cells, extracellular vesicles, and hydrogel/biomaterials as potential DMRT for renovascular
disease (RVD), sepsis-associated AKI (SA-AKI), diabetic kidney disease (DKD), and kidney transplantation (KTx). *Broken line indicates that
clinical trials of MSCs have been reported in sepsis, including some patients with SA-AKI, but not with kidney function as a primary outcome.
Figure created using Biorender.com.
Basic Science for Clinicians Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative ...
544   KIDNEY360

Renovascular Disease                                            derived EVs in renal diseases has been highlighted by
   Hypertension is a major risk factor for CKD, with hyper-     showing decreased renal inflammation and injury through
tensive kidney disease accounting for approximately 30% of      intrarenal delivery of EVs in pigs with RVD and concom-
all ESKD cases (14). Hypertensive kidney disease—charac-        itant metabolic syndrome (27). Other studies have demon-
terized by vascular damage, endothelial dysfunction, and        strated their role as carriers of anti-inflammatory genes and
loss of endogenous vasodilators—results in progressive          proteins and their capacity to be engineered to deliver
loss of the renal microvasculature (15). RVD is a common        specific substances or to have enhanced uptake by target
cause for secondary hypertension in individuals aged $65        cells (28–30). Therefore, MSC-derived EVs may serve as an
years, and RVD attributed to atherosclerotic plaque devel-      acellular therapeutic option to attenuate inflammation and
opment with reduction in renal-artery dimension repre-          fibrosis in RVD and other forms of renal disease. As illus-
sents a unique intersection between hypertension and CKD/       trated in Figure 2 for the clinical target of RVD and asso-
ESKD leading to progressive renal insufficiency (16). In         ciated ischemic nephropathy, the paracrine, regenerative
the setting of significant RVD, further reduction of renal       activities of MSCs, including both secreted soluble media-
blood flow (RBF) and hypoxia trigger inflammation, oxida-         tors and released EVs, are now recognized to be “tunable.”
tive stress, and profibrotic pathways, leading to scarring       Thus, as described later in greater detail, disease-associated
and further deterioration of renal function (ischemic ne-       dysfunction of ex vivo–expanded, autologous MSCs may
phropathy) (17). Importantly, recent clinical trials and ex-    be reversed through hypoxic preconditioning and other
perimental studies indicate that restoration of large-vessel    manipulations (31,32).
patency alone is not enough to regain kidney function in           There is now promising evidence that observations of
most patients with atherosclerotic RVD (18,19). The limited     RVD modulation by MSCs in animal models can be trans-
number of currently available strategies for effectively mod-   lated into clinical benefits. In a phase 1 trial in patients with
ulating RVD, and the realization that the natural history       RVD, we (L.J.H. and S.M.H.) recently demonstrated that
of this disease involves transition from a hemodynamic          intra-arterial infusion of autologous adipose tissue–
component to a proinflammatory and profibrotic disease,           derived MSCs into poststenotic kidneys (without con-
highlight the need for a paradigm shift in therapy (20).        comitant renal-artery angioplasty) resulted in increased
Importantly, RVD-associated ischemic nephropathy and            cortical RBF and GFR compared with the baseline val-
hypertensive kidney disease, in the absence of RVD, share       ues. These improvements in hemodynamic and func-
common pathophysiologic features that include activated         tional indices were associated with attenuation of tissue
renin-angiotensin-aldosterone system (RAAS), increased so-      hypoxia, inflammatory cytokines, and angiogenic bio-
dium retention, and—consequently—increased oxygen con-          markers, along with a fall in BP between baseline and
sumption (21). In evidence of this, studies in hypertensive     3 month follow-up. These changes were more prominent
rats using oxygen microelectrodes found pronounced med-         in the patients treated with a higher MSC dose (26,33).
ullary and cortical hypoxia in spontaneously hypertensive       Of further interest, we also observed RBF increases in the
rats compared with normotensive controls (22). Further-         (non-MSC-infused) contralateral kidneys (26,33). This
more, the presence of ischemic, rather than hypertrophic,       phenomenon likely occurred due to “crosstalk” signal-
glomeruli in hypertensive kidney disease suggests that          ing between kidneys and/or wider systemic effects of
hypoxia and ischemia are the predominant mechanisms             signaling and homing signals for MSCs. Moreover, ben-
(23). Therefore, in subjects with prolonged hypertension        eficial off-target effects, beyond the kidney, are also
and relevant genetic-, environmental-, and lifestyle-related    possible because infusion of MSCs or endothelial pro-
risk factors, limited blood flow and oxygenation to areas of     genitor cells into the renal artery has been associated
renal parenchyma, where the oxygen tension is ,10 mm Hg,        with attenuation of hypertensive cardiomyopathy in
makes the kidney vulnerable to ischemic injury which            experimental models of renovascular hypertension
resembles that due to RVD. In this context, implementation      (34). Despite the promising evidence base of preclinical
of DMRT, in particular the use of MSCs, can be viewed as        and clinical application of MSCs and MSC-derived EVs
a novel therapeutic option for RVD and hypertensive kid-        in RVD, there has been limited exploration to date of
ney disease.                                                    alternative DMRT (such as regulatory immune cells,
   Importantly, MSCs have immunomodulatory, anti-               other adult progenitor cells, and implantable hydrogels)
inflammatory, and proangiogenic properties that have been        in this area (Figure 1). Nonetheless, experimental evi-
demonstrated in experimental studies in animals and             dence in models of renal ischemia-reperfusion injury
humans with RVD (24–26). Arterial delivery of MSCs in           could provide a basis for the translation of such thera-
the swine model of RVD was associated with protection of        pies for chronic ischemic nephropathy in the future
the stenotic kidney and improved RBF and function, with         (13,35,36).
reduction of oxidative stress and inflammation, contributing
to tissue repair (24). The strong paracrine effect of MSCs,
and not their differentiation capacity, seems to be the prin-   Sepsis-Associated Acute Kidney Injury
cipal mechanism of their therapeutic action. In clinically        AKI has a prevalence ranging from 1% to 20% of patients
relevant, large-animal models of RVD, MSCs have been            who are hospitalized and 50% to 60% of patients in the
shown to release a variety of soluble mediators that act        intensive care unit. Mortality is proportional to the severity
locally within the kidney to ameliorate ischemic nephrop-       of AKI, and 30% of survivors die within the first year after
athy through proangiogenic, anti-inflammatory, and anti-         hospital discharge. Sepsis, which is the most frequent cause
oxidative mechanisms (24). Also consistent with a paracrine     of AKI in patients who are critically ill (37), is a life-
model, the therapeutic potential of stem/progenitor cell–       threatening syndrome resulting from a disordered immune
Basic Science for Clinicians Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative ...
KIDNEY360 2: 542–557, March, 2021                                   Disease-Modulating Regenerative Therapies in Nephrology, Hickson et al.   545

                                                                        Hypertension

                                                         Renovascular                   lschemic
                                                           Disease                     Nephropathy

                                                                        Stromal Cell
                                                                        Dysfunction

                                Disease Modulation

                                                                                             Limited MSC Paracrine
                                                                                              Regenerative Factors

                                                                  Hypoxic Pre-conditioning
                                                                    Inflammatory Stimuli

                           Enhanced MSC Paracrine
                             Regenerative Factors

Figure 2. | Hypoxic pre-conditioning is a potential strategy for enhancing MSC therapeutic potency in hypertensive kidney disease. As
illustrated, chronic hypertension, RVD, and ischemic nephropathy lead to stromal-cell dysfunction, which is associated with limited production
of paracrine regenerative factors (released extracellular vesicles and soluble mediators) by patient-derived, culture-expanded, mesenchymal
stem/stromal cells (MSCs). Culture under low oxygen tension (hypoxic preconditioning) may restore the production of extracellular vesicles and
soluble mediators, resulting in enhanced paracrine regenerative activity and increased potential for disease modulation after localized or
systemic delivery of autologous MSCs. Figure created using Biorender.com.

response to uncontrolled microbial infection (38). The                  and resident parenchymal cells (37). In SA-AKI, there is
pathophysiology of sepsis is dominated by dysregulated                  an abnormal repair process due to prolonged hypoxia,
inflammation and immune suppression, with endothelial                    cytokine expression, and defective adaptive immune cell
and epithelial injury, leukocyte aggregation, mitochondrial             function. Patients who are critically ill with persistent
dysfunction, apoptosis, and impaired regeneration (37). SA-             or recurrent AKI are at very high risk for secondary
AKI differs from ischemic and toxic AKI, being characterized            infection and increased mortality, and represent a key
by global renal hyperemia with altered RBF distribution                 target for novel and more-effective therapies (39). In this
and inflammation incited by both infiltrating immune cells                regard, MSCs have demonstrated benefits in multiple
Basic Science for Clinicians Progress toward the Clinical Application of Mesenchymal Stromal Cells and Other Disease-Modulating Regenerative ...
546   KIDNEY360

sepsis models, including LPS administration, bacterial                    crosstalk with immune cells that result in modulatory effects
pneumonia, and polymicrobial abdominal sepsis (40–42).                    on cytokine expression, vascular permeability, removal of
In animal models of sepsis, MSC administration is report-                 apoptotic cells, and clearance of bacteria by neutrophils and
edly associated with improved survival; reduced organ                     macrophages (Figure 3) (46). In addition to release of soluble
injury; increased clearance of bacteria, cells, and fluid; and             mediators and reprogramming of immune cells by viable
resolution of inflammation (43–45). Some animal-model                      MSCs, it has recently been shown that disease modula-
studies of SA-AKI have demonstrated improvement in                        tion may occur as a result of the induction of apoptosis
tubular-injury scores and kidney function (42,45), whereas                of intravenously infused MSCs by cytotoxic lymphocytes
others have not (43,44)—an inconsistency that may reflect                  followed by their phagocytosis by resident myeloid cells
differences among the models used.                                        (monocytes and macrophages). This process, referred to as
  From a mechanistic perspective, it is now clear that MSCs               efferocytosis, results in polarization of myeloid cells toward
administered intravenously in models of sepsis localize                   alternatively activated (M2-like) phenotypes with potent
predominantly in the lungs. From this location, they medi-                anti-inflammatory effects (47,48). Furthermore, either di-
ate their systemic benefits through mechanisms involving                   rectly or through their effects on myeloid cells, MSCs also

                                    Sepsis + AKI

                                                                                        Increased bacterial, cellular
                                                                                            and fluid clearance;
                                                                                         resolution of inflammation

                                                                                                                 MSC/Immune Cell
                                                                                                                  Cross-talk and
                                                                                                                   Efferocytosis

                                                       Neutralization of LPS;
                                          Increased secretion of IL-10, PGE2, VEGF, KGF,
                                             HGF; Enhanced macrophage phagocytosis;

                                                                                          Reduced pro-inflammatory cytokines;
                                                                                            Improved vascular permeability;
                                                                                          Enhanced clearance of apoptotic cells

             Renal functional improvement
                secondary to improved
               cardiorespiratory function
                                                                                                               AKI Resolution

Figure 3. | The systemic therapeutic effects of intravenousMSC therapy in SA-AKI are triggered by immune cell interactions in the lungs. As
illustrated, intravenous administration of MSCs in the setting of SA-AKI results in MSC trapping in the lungs, where complex interactions
(crosstalk and efferocytosis) with resident immune cells (mononuclear phagocytes [macrophages] and lymphocytes) result in beneficial lo-
calized effects within the alveolar spaces and systemic effects (LPS neutralization, secretion of anti-inflammatory factors, enhanced
phagocytosis) with potential to promote resolution of inflammation, disrupted vascular integrity, and increased cell death in the kidneys.
Improved cardiorespiratory function as a result of MSC local and systemic effects may provide further indirect effects to more effectively resolve
SA-AKI. Figure created using Biorender.com. HGF, hepatocyte growth factor; KGF, keratinocyte growth factor.
KIDNEY360 2: 542–557, March, 2021                             Disease-Modulating Regenerative Therapies in Nephrology, Hickson et al.   547

augment tissue-repair processes through promoting expan-          SA-AKI. Indeed, trials of MSCs and other DMRT in sepsis
sion of T-reg (49).                                               may be better suited to detecting their effects on the de-
   A number of specific soluble factors have been identified        velopment or severity of AKI. Because sepsis is a multiorgan
as mediating the paracrine effects of MSCs and their              disorder, favorable effects of systemically administered
immune-cell partners in models of sepsis. These include           MSCs on kidney function may derive from improved func-
IL-10, keratinocyte growth factor, PGE2, vascular endothe-        tion of other organ systems and from reprogramming of
lial growth factor (VEGF), antibacterial peptides LL-37 and       immune cells at distant sites. Indeed, preclinical studies
hepcidin, and other proresolution factors (50). The role of IL-   indicate that infusion of apoptotic versus viable MSCs
10 has been most convincingly demonstrated. In the mouse          within the lung led to greater suppression of inflammation,
cecal ligation and puncture model of polymicrobial sepsis,        oxidative stress, cellular markers of immune reactivity, and
Németh et al. (45) first reported that intravenously admin-        a less marked kidney injury in a cecal ligation and perfo-
istered MSCs stimulate IL-10 production by macrophages            ration model of sepsis (59). Given the high prevalence of
through PGE2/EP2-receptor interaction, resulting in the           AKI among patients with sepsis and its implications for
prevention of neutrophil extravasation into tissue. This          morbidity and mortality, it will be important for future
MSC-induced pulse of IL-10 production has since been              clinical trials of DMRT in sepsis to include patients with
replicated in several other studies (49). Transfer of specific     —or at risk for developing—AKI, for equal numbers of
microRNAs or mitochondria via nanotubes may also un-              patients with similar stages of AKI to be randomized,
derlie some of the effects of MSCs to enhance macrophage          and for specific renal end points to be included in the trial
phagocytic activity or endogenous stem-cell fitness in the         design.
septic environment (51). A further strategy to enhance the
immunomodulatory features of MSCs in sepsis is through
preconditioning (“licensing”) with proinflammatory cyto-           Diabetic Kidney Disease
kines, toll-like receptor ligands, carbon monoxide, and eico-        Due to the growth of the aging population, the estimated
sapentaenoic acid (52–54). In the case of carbon-monoxide         number of individuals with diabetes mellitus (DM) world-
licensing of MSCs, this was reported by Tsoyi et al. (52) to      wide increased from 108 million in 1980 to 422 million in
result in reduced organ damage (including kidney injury)          2014 (60). Moreover, the global epidemic of DM has con-
and superior survival in mouse models of sepsis, while            tributed approximately 50% of the increased health burden
also allowing for later MSC administration. Mechanisms            due to CKD (61). DKD is characterized by vascular damage,
by which preconditioning has been reported to enhance             resulting from cumulative effects of a wide range of pre-
MSC activity include activation of the lipoxygenase pathway       dominantly hyperglycemia-driven maladaptive processes,
and enhanced exosome delivery of microRNA to macro-               including chronic inflammation, increased oxidative stress,
phages. As already described in the context of RVD, MSC-          advanced accumulation of glycation end products, steatosis,
derived EVs also have the potential to be developed as            insulin resistance, renal hypoxia, apoptosis, cellular dedif-
a subcellular DMRT for sepsis and SA-AKI through the              ferentiation and senescence, and altered RAAS activation
transfer of a wide range of bioactive molecules (49,55).          (62–64). Intrinsic renal regenerative capacity is limited in
   Although the clinical application of MSCs in sepsis and        DM, exacerbating chronic glomerulosclerosis, tubulointer-
SA-AKI is at an early stage, they have shown promising            stitial fibrosis, and chronic inflammation (62,64,65). Al-
safety profiles in early-phase human trials (49). For example,     though recent clinical trials of sodium-glucose cotransporter-
in a phase 1, dose-escalation trial involving nine patients       2 inhibitors and other pharmacologic agents have shown
with sepsis, treatment with MSCs was found to be safe and         that the rate of renal functional loss can be slowed in DKD
well tolerated, albeit with no overt effect on sepsis param-      due to type 2 DM (66–68), successful targeting of multi-
eters. An analysis of cytokine levels in treated patients         ple injurious pathways—such as those mediating inflam-
demonstrated no increase in known proinflammatory medi-            mation, oxidative stress, renal hypoxia, and fibrosis—may
ators or biomarkers of organ dysfunction after MSC treat-         be necessary to truly halt DKD.
ment (56,57). In a phase 2, multicenter, randomized,                 With this goal in mind, DMRT represent novel therapeu-
placebo-controlled, clinical trial, Swaminathan et al. exam-      tic options for the delivery or induction of a wide range of
ined the effect of allogeneic MSC therapy delivered intra-        mediators to simultaneously target maladaptive processes
aortically in patients undergoing cardiac surgery who de-         that contribute to DKD progression. As with other renal
veloped postbypass AKI. More than half of these patients          diseases, the most extensively studied DMRT in DKD is the
had impaired renal function at baseline (58). Although this       MSC (64). In many preclinical experimental models of DM
trial was carried out in patients with a sterile form of AKI,     and diabetic nephropathy (64,68), the paracrine-mediated
the design and results have important implications for the        actions and cell-cell interactions of exogenously adminis-
future planning of clinical trials of DMRT in SA-AKI. Dis-        tered MSCs have shown potential to modulate a range of
appointingly, in this trial, MSC administration resulted in       pathophysiologic processes that contribute, both locally and
no difference in recovery of renal function, dialysis, or death   systemically, to the progressive renal injury and functional
compared with placebo (58). Although carried out in a rel-        loss that characterize DKD (Figure 4). External to the kid-
atively homogenous patient population, differences in renal       neys, MSCs delivered intravenously or by other routes have
reserve, complexity of cardiac surgical procedure, bypass         been experimentally shown in models of DM to modulate
time, and other postoperative complications may yet have          adipose-tissue inflammation, preserve islet function, and
hindered the ability to observe any modest clinical benefit of     enhance insulin sensitivity, leading indirectly to beneficial
MSCs. This negative study in sterile AKI does not neces-          renal effects through reducing glycemia and the proinflam-
sarily blunt interest in the clinical translation of MSCs for     matory systemic environment (64,69,70). Concomitantly,
548   KIDNEY360

                                                                          Preserved                                  Modulation of:
            Extra-renal Modulatory Effects                                                                           Tubular atrophy
                                                                        islet function
                                                                                                                     Interstitial inflammation
                                                                                                                     Oxidative stress
                            Reduced adipose                                                                          Fibrogenesis
                           tissue inflammation                                                                       Senescence

                                                                                         Modulation of:
                                                                                         Podocyte oxidative stress
                                                                                          and apoptosis
                                                                                         Mesangial expansion
                                                                                         Glomerular fibrosis

                                   Key Mediators
                              Hepatocyte Growth Factor
                            Indoleamine 2,3 Dioxygenase
Enhanced insulin                  Prostaglandin E2
   sensitivity           Vascular Endothelial Growth Factor
                                  Hemoxygenase 1
                                    Interleukin 10
                                Extracellular Vesicles                                                     Modulation of:
                      Re-programmed Macrophages and T-cells                                                Capillary rarefaction
                                                                                                           Hypoxia

                         Improved glycemic control
                       Reduced systemic inflammation
                           and oxidative stress                                                    Intra-renal Modulatory Effects

Figure 4. | Multiple potential therapeutic effects have been identified for systemically administered MSCs in DKD. Extensive preclinical and
limited clinical trial data indicate that MSCs (center) may exert both extrarenal and intrarenal modulatory effects, through a range of key
mediators, after intravenous administration in diabetes and DKD. Upper left: Extrarenal effects which diminish adipose-tissue inflammation,
enhance insulin sensitivity, and preserve islet function can stabilize or reverse the course of DKD by improving glycemic control and reducing
systemic inflammation and oxidative stress. Lower right: Intrarenal effects by which key MSC-generated and -induced mediators have been
shown experimentally to modulate multiple aspects of DKD pathophysiology within the glomerulus, the tubulointerstitial compartment, and the
microvasculature. Figure created using Biorender.com.

MSCs themselves, their released mediators, and regulatory               T-reg induced by interactions with, or uptake of, exoge-
immune cells induced as a result of MSC administration                  nously administered MSCs. One of the most important
may transfer to the kidneys to mediate beneficial effects                growth factors, HGF, reduces kidney fibrosis by blocking
within distinct renal compartments, including the glomer-               tubular epithelial cell dedifferentiation and inhibiting intra-
ulus, microvasculature, tubules, and interstitium. Reduc-               renal expression of monocyte chemoattractant protein-1 and
tions in glomerular size, podocyte apoptosis, glomerular                macrophage infiltration (70,71). Other key mediators asso-
matrix expansion/sclerosis, peritubular interstitial fibrosis,           ciated with the direct and induced paracrine effects of MSCs
renal tubular epithelial cell death and dedifferentiation,              in DKD include indoleamine 2,3-dioxygenase, a potent im-
tubulointerstitial fibrosis, and microvascular rarefaction               munomodulatory enzyme; PGE2, a likely mediator of T-reg
have been observed in association with reduced albumin-                 differentiation; and IL-10, an anti-inflammatory cytokine
uria and stabilization of GFR (64,69,70).                               released by macrophages after phagocytosis of apoptotic
   Preclinical, MSC-based, experimental studies have dem-               MSCs (64,72,73). The many observations from experimental
onstrated benefits derived from a variety of therapeutically             models of DM and DKD that key soluble and released
relevant mediators (Figure 4). These include soluble factors            factors mediate the disease-modulatory effects of MSCs
with antifibrotic (hepatocyte growth factor [HGF]), proan-               have also stimulated interest in the use of MSC-derived
giogenic (VEGF), antiapoptotic/homeostasis (HGF, VEGF,                  conditioned medium and EVs as alternative DMRT (74,75).
stromal cell–derived factor [SDF-1/CXCL-12]), and immu-                 Despite the focus on paracrine mechanisms in many
nomodulatory (indoleamine 2,3-dioxygenase, PGE2, and IL-                preclinical studies, however, it remains unclear whether
10) activity. Such soluble factors may be secreted inherently           soluble factors released by MSCs after systemic delivery
by MSCs, triggered in MSCs by signaling from proinflam-                  can explain all of the beneficial effects reported in experi-
matory cytokines and immune cells, or secondarily pro-                  mental DKD. Specifically, the transient survival of intrave-
duced by alternatively activated (M2) macrophages and                   nously administered MSCs, and reports in other disease
KIDNEY360 2: 542–557, March, 2021                            Disease-Modulating Regenerative Therapies in Nephrology, Hickson et al.   549

models of therapeutic effects mediated by apoptotic or           administration (78). Although also promising, injection of
heat-inactivated MSCs, suggest the existence of other            DRMT-derived EVs is not yet underway in human DKD
mechanisms (47,48,72,76). Although transmigration and            studies.
prolonged engraftment of a minority of administered cells
to the kidneys remains theoretically possible, the phenom-
ena of MSC apoptosis and efferocytosis by macrophages            Kidney Transplantation
(48) and MSC-induced expansion of T-reg (77) repre-                 Beyond the “holy grail” of donor-specific tolerance, steady
sent more compelling mechanisms by which their anti-             advances in understanding pathologic processes that un-
inflammatory, prorepair effects within the kidneys could          derlie the common causes of early and late renal allograft
be augmented and prolonged beyond the initial release of         failure have revealed other important new therapeutic tar-
soluble mediators.                                               gets that are not adequately addressed by conventional
   In addition to MSCs from various tissue sources, similar      immunosuppressive drugs and clinical practices (82). These
renal regenerative effects have been observed for a variety of   include inflammatory and metabolic pathways that mediate
other stem/progenitor-like cells and their secreted trophic      donor-organ injury before and early after transplantation,
factors or EVs (36,68). Primary cells derived from the kidney    immunologic processes that drive the formation of donor-
may also exert paracrine-mediated, disease-modulating            specific antibodies and antidonor memory T cells, effector
effects in a similar fashion to MSCs, and are being actively     mechanisms responsible for subsequent acute or chronic
pursued as potential DMRT. For example, selected renal           immune-mediated rejection, and maladaptive cellular pro-
cells (SRC), composed of isolated tubular and aquaporin          cesses such as fibrosis and senescence. Against this back-
2–positive collecting duct cells, have advanced to the           drop, the potential for DMRT, such as MSCs and regulatory
clinical-translation phase (78). These primary cells induce      immune cells, to address some or all of these major unmet
tubular cell proliferation while attenuating TNF-a–induced       needs for improved long-term KTx survival is being ro-
NF-kB and TGF-b1–mediated plasminogen activator                  bustly pursued. In the following paragraphs and illustrated
inhibitor-1 signaling pathways that contribute to inflamma-       in Figure 5, we provide overviews of recent progress in the
tion and fibrosis in experimental DKD (79). Given the tran-       translation of these DMRT to the field of KTx, and how they
sient period in which exogenously administered cells reside      may address key mechanisms of graft injury.
in the diseased microenvironment, use of biomaterials, such
as hydrogels, to enrich cell delivery and duration of action     Mesenchymal Stem/Stromal Cells
have been pursued (13,80). As discussed below, this has             Extensive preclinical evidence that MSCs modulate harm-
since been translated to a locally delivered therapeutic         ful antidonor immune responses and maladaptive inflam-
strategy for DKD in which a gelatin-based hydrogel con-          mation associated with allogeneic organ transplants and
taining expanded autologous SRC is implanted into the            may promote immune tolerance has accumulated over the
kidneys (78).                                                    past 18 years (83). In 2012, Tan et al. (84) reported the results
   Despite numerous studies in experimental DKD, clinical        of a phase 2 clinical trial in which 104 recipients of living-
translation of DMRT has been limited. In 2016, Packham           related-donor KTx received a novel induction regimen con-
et al. reported the results of a randomized, placebo-            sisting of two intravenous infusions of autologous bone
controlled, dose-escalation study that tested the safety         marrow–derived MSCs at the time of transplantation and
and feasibility of intravenous infusion of allogeneic mesen-     2 weeks later, followed by maintenance therapy with con-
chymal precursor cells (rexlemestrocel-L) in adults with         ventional- or low-dose cyclosporine. For MSC-induced
type 2 DM and CKD stages 3/4. The cell infusions were            recipients, early recovery of renal function and frequency
well tolerated, and trends in kidney function during a 12-       of acute rejection and opportunistic infection during the first
week follow-up period favored stabilization or improve-          post-transplant year were comparable or superior to those
ment in 20 patients treated with cell infusions compared         of a control group induced with anti–IL-2 receptor antibody
with ten patients treated with placebo (81). Other early-        followed by conventional-dose cyclosporine (84). Although
phase clinical trials are now investigating allogeneic bone      the trial provided an encouraging demonstration of the
marrow–derived MSCs (M.D.G.; Italy, Ireland, United              safety and potential efficacy of peritransplant MSC infu-
Kingdom; ClinicalTrials.gov, NCT02585622), autologous            sions, the lack of mechanistic studies and of a measurable
adipose-derived MSCs (L.J.H., S.M.H.; NCT03840343), and          indicator of the in vivo activities of the infused cells pre-
allogeneic umbilical cord–derived MSCs (Japan, NCT04125329;      cluded any immediate progress toward wider clinical prac-
China, NCT04216849) in DKD. As mentioned above, the              tice. For this reason, several other centers have focused on
therapeutic combination of primary kidney cells (SRC)            evaluating both autologous and allogeneic MSC therapies in
in hydrogels (named Neo-Kidney Augment) is also being            smaller numbers of KTx recipients, along with longitudinal
investigated as a DMRT for DKD in phase 1 and 2 clinical         immunologic and, in some cases, histologic monitoring of
trials (NCT02008851, NCT03270956, NCT02836574). Of               the grafts. Details of the designs, major outcomes, and
note, a report of the phase 1 trial involving laparoscopically   documented immunologic consequences of MSC adminis-
assisted intracortical implantation of SRC, in seven male        tration to KTx recipients in such early-phase trials carried
patients with type 2 DM and CKD stages 3/4, indicated an         out to date have been summarized and expertly reviewed
unacceptable number of postprocedural complications,             by Podestà et al. (83). Results from one further phase 1
prompting changes in implantation methodology. None-             trial have also been very recently reported (85). In addi-
theless, renal function and urine albumin-creatinine ra-         tion to determining safety profiles, these trials have begun
tio remained relatively stable for 12 months, whereas            to address whether autologous or allogeneic MSC infu-
eGFR tended to decline from months 12 to 24 after SRC            sions can: (1) promote T-reg and/or donor-specific T-cell
550    KIDNEY360

                             DMRT                      MSC              T-reg               M-reg          Tol-DC

                             Site of Action                        Systemic                     Local

                                              Naive Lymphocytes         Activated Lymphocytes            Myeloid Cells
                             Target
                              Cells              Dendritic Cells              Fibroblasts               Epithelial Cells

               Promote Donor-specific Immune                                                        Modulate Pre-existing Anti-donor
                        Tolerance                                                                        Immune Responses

                Promote Epithelial Repair and                                                            Suppress Pro-fibrotic
                       Regeneration                                                                         Inflammation

                                                                                   Reduced burden of
      Reduced ischemic injury/             Increased maximum                                                          Stabilization of declining
                                                                                  immunosuppressive
       Delayed Graft Function            glomerular filtration rate                                                      transplant function
                                                                                      medications

Figure 5. | Disease modulating regenerative therapies for kidney transplantation address diverse mechanisms and potential clinical benefits.
Upper panel: Diverse types of modulatory cellular therapies that have been the subject of early-phase clinical trials in KTx recipients along with
their potential sites of action and target cells. Middle panel: Four major mechanistic goals of DMRT applied to KTx with illustration of the most
relevant cellular therapies for each, along with their predicted sites of action and most significant cell targets for each (based on preclinical
studies and profiling/monitoring analyses of subjects from clinical trial). Lower panel: Significant clinical benefits that represent the most
immediate goals for the clinical translation of DMRT in KTx. Figure created using Biorender.com. M-reg, regulatory macrophages; Tol-DC,
tolerogenic dendritic cells; T-reg, regulatory T cell.

hypo-responsiveness (86–88), (2) reverse or stabilize subclinical           Taken together, the trial reports to date support the
tubulointerstitial inflammation and fibrosis/tubular atro-                  conclusion that intravenous or intrarenal infusion of MSCs
phy (89), and (3) allow for early or delayed reduction (or                in KTx recipients is safe and associated with comparable
even eventual withdrawal) of calcineurin inhibitor–based                  early-to-midterm patient/graft outcomes and potentially
immunosuppression (85,90,91). A further interesting ques-                 superior renal function compared with conventional thera-
tion, currently being addressed in preclinical studies (92–94)            peutic regimens. Where examined, they also provide evi-
and an early-phase clinical trial (NCT04388761), is whether               dence that pre- or early post-transplant MSC infusions may
ex vivo perfusion of kidneys procured for transplantation                 be associated with favorable immunologic effects, such as
with MSCs can modulate ischemic tissue injury and ame-                    increases in circulating T-reg or T-reg/effector T cell ratios.
liorate subsequent delayed graft function.                                Nonetheless, it should be acknowledged that overall patient
KIDNEY360 2: 542–557, March, 2021                              Disease-Modulating Regenerative Therapies in Nephrology, Hickson et al.   551

numbers remain small and the possibility of more subtle            macrophage, or one tolerogenic dendritic cell therapeutic
adverse effects—such as localized proinflammatory re-               product, in combination with a tapered conventional im-
sponse, reduced antiviral immunity, or (in the case of allo-       munosuppressive drug regimen, were collated and com-
geneic MSCs) stimulation of anti-HLA antibodies—should             pared with results for a group of 66 recipients that were
be carefully addressed by larger trials and longer follow-up       managed by a standard-of-care regimen across eight sites in
(83). Development of clinically applicable assays of potency       Europe and the United States. Although no conclusions
and in vivo effects in the context of KTx will also likely be      about the protolerogenic efficacy for any single regulatory
required for optimal translation of MSCs and other DMRT            cell therapy can be made from this report, the authors
into routine clinical practice. For example, for the clinical      convincingly show a good safety profile for such therapies
target of acute graft versus host disease after allogeneic         in KTx recipients. In addition, the combined cell-therapy
hematopoietic stem cell transplantation, Cheung et al. (95)        cohort had no increase in graft rejection or reduction in graft
recently demonstrated that an increase in serum PGE2 1–            survival, experienced a strikingly reduced rate of viral
4 days after MSC infusion distinguished patients with              infections, and appeared to revert to a more favorable
clinical response to cell therapy from nonresponders; this         peripheral blood immune cell profile compared with recip-
finding being consistent with in vitro cellular assays and          ients who were treated conventionally (100). Subsequently,
mechanistic animal model studies carried out by the same           Roemhild et al. (101) published a very detailed report of the
group (48).                                                        ONEnTreg13 Trial—a component of the ONE Study. In this
                                                                   phase 1/2a trial, autologous, ex vivo–expanded, natural
                                                                   T-reg (nTreg) were administered 7 days after transplanta-
Regulatory Immune Cells                                            tion to 11 recipients of living-donor KTx at Charité – Berlin
   Because the recognition that forkhead box P3 (FoxP3)–           University of Medicine (Berlin, Germany) and the results
positive T-reg are essential for maintaining peripheral im-        were compared with those of nine patients who were pre-
mune tolerance to autoantigens and nonthreatening foreign          viously transplanted and managed with the standard-of-
antigens, the concept that treatment with ex vivo–expanded         care regimen at the same center. These results demonstrated
regulatory immune cells could prevent allogeneic transplant        excellent safety and 3-year graft outcomes for the KTx
rejection and foster donor-specific tolerance in organ allo-        recipients treated with nTreg, along with successful tapering
graft recipients has been energetically pursued (96,97). In        of immunosuppression to tacrolimus monotherapy in eight
addition to T cells, it has also become clear that other major     of 11 subjects and evidence of oligoclonal (presumably
types of immune effector cells, including macrophages,             alloantigen-driven) expansion of nTreg at 60 weeks post-
dendritic cells, and B cells, incorporate subpopulations or        transplant (101). Finally, in another very recent, early-phase
alternative functional states that mediate counter-regula-         trial report, Morath et al. (104) describe favorable safety
tory/suppressive effects and may be amenable to clinical           and graft outcomes after pretransplant administration of
exploitation (97). In keeping with the concept of DMRT, the        donor-derived modified immune cell (mitomycin C–treated
potential therapeutic actions of regulatory immune cell            PBMC) infusions to ten recipients of living-donor KTx. Post-
therapies can be broadly viewed as modulating (as opposed          transplant immunologic studies also provided evidence for
to blocking) the interactions between donor-derived (allo-)        prolonged donor-specific T-cell hypo-responsiveness and in-
antigens or proinflammatory stimuli and recipient immune            creased numbers of circulating IL-10–producing regulatory
effectors to prevent or reverse acute and chronic organ            B cells (which have been associated with immune tolerance)
allograft injury. On the basis of evidence from almost two         (104).
decades of basic and preclinical research, early-phase clin-          Overall, recently reported and ongoing early-phase clin-
ical trials of ex vivo–expanded autologous T-reg have been         ical trials of several cell-based DMRT in the area of KTx
recently completed in recipients of KTx and liver transplant       provide important reassurances regarding the safety and
(8,96,98–101). Those carried out in KTx recipients have, to        feasibility of such therapies, both before and at early or later
date, consistently demonstrated safety in combination with         times after transplantation. The generally favorable short-
various conventional immunosuppressive regimens, pre-              to-midterm patient and graft outcomes, and promising im-
liminary evidence for persistence of infused T-reg in the          mune profiling and histologic studies, should be interpreted
blood for 1–3 months (99), and of an increase in total             with caution in the absence of larger, randomized controlled
circulating T-reg numbers for at least 12 months after ad-         trials. Nonetheless, cohesion among some of the observa-
ministration (98). Ongoing early-phase trials and planned          tions made in patients participating in these trials and the
phase 2 trials are likely to further clarify whether polyclonal,   growing scientific knowledge regarding immunologic tol-
autologous T-reg therapies robustly modulate post-                 erance and of the pathobiology of KTx complications should
transplant immune responses toward donor-specific toler-            provide a strong impetus for further progress. As illustrated
ance (96). Other regulatory immune cell types—specifically          in Figure 5, individual DMRT have the potential to target
regulatory macrophages and tolerogenic dendritic cells—            specific clinical and immunologic challenges associated
have also been developed to the point of early-phase clinical      with current limitations to the long-term health of KTx
trials in KTx recipients (102,103). Very recently, a report of     recipients.
the post-transplant outcomes and immunologic profiling
results for recipients of living-donor KTx enrolled into a suite
of early-phase regulatory cell therapy trials has been pub-        Autologous versus Allogeneic Cell Therapies: The
lished by the ONE Study consortium (100). In this unique           Influence of Patient-Specific Factors
study, the observed results for 38 recipients of living-donor        As we move toward clinical application of DMRT, optimi-
KTx receiving one of four different T-reg, one regulatory          zation of cell product remains vital to successful translation
552   KIDNEY360

of preclinical findings. Allogeneic cell–based products offer         Conclusions and Future Directions
a readily available “off-the-shelf” treatment option. Yet,              As we have reviewed here, extensive preclinical research
patient-derived (autologous) cells may be preferable for             has greatly increased our understanding of the in vivo
individualized therapy or for repeated dosing, given the             distribution, longevity, and mechanisms of action of MSCs
lower risk for allosensitization. In this regard, the influ-          and other potential regenerative therapies in the setting of
ence of patient- or disease-specific factors on the growth and        kidney diseases. These insights have extended the rationale
functionality of culture-expanded stromal and other pri-             for regenerative therapies beyond the initial concept of
mary cell therapies represents a key research area—partic-           engraftment and differentiation into functional tissues to
ularly in settings of high relevance to kidney disease, such as      include modulatory effects of, typically, short-lived cells,
older age, DM, vascular disease, and reduced renal function          vesicles, or biomaterials that ameliorate disease processes
(105). In DM and kidney disease, oxidative stress, autoph-           through complex, paracrine interactions with host cells and
agy, and cellular senescence induce dysfunction of autolo-           tissues which promote inherent mechanisms of repair and
gous cells (106). In metabolic syndrome and DM, stem-cell            regeneration. Well-conducted small- and large-animal
mobilization and therapeutic effect are diminished (65,74).          model studies have been essential to determining the opti-
In older individuals with atherosclerotic RVD, we identified          mal parameters for clinical application of MSCs and other
altered MSC functional capacity (migration, angiogenesis)            DMRT to kidney diseases—including the route of delivery
and increased cellular-senescence burden compared with               and localization after administration; the key responder
controls (32). Despite this, our clinical trial results, described   compartments and cell responses within the kidney; and
above in RVD, confirm that intrarenal administration of               the source, timing of release, and activity of the most im-
autologous MSC was associated with improved RBF and                  portant soluble mediators. In keeping with the literature
preserved GFR (26,33). Similarly, in MSCs harvested from             reviewed in this article, we believe that these critical param-
adults with DKD and healthy controls, we have observed               eters must be defined for each specific disease and therapy.
transcriptome alterations and reduced in vitro MSC migra-            Conversely, lack of concordance between animal-model re-
tion but preserved or increased immunomodulatory and                 search and human clinical application of regenerative ther-
renal reparative activities in vitro (L.J.H., under review). In      apies continues to be a major challenge to the field. For the
ESKD and KTx recipients, autologous MSCs have been                   four distinct renal-disease areas we have focused on, trans-
shown to undergo comparable culture-expansion charac-                lation of DMRT into clinical nephrology practice remains at
teristics to those from individuals with normal kidney               an early stage. Indeed, of the translational strategies we
function, and to maintain the capacity to inhibit antidonor          review here, only MSC therapy for RVD and T-reg therapy
HLA immune response when compared with control MSCs                  in KTx could be said to have shown preliminary (early-
(83,107).                                                            phase) clinical trial evidence of superior efficacy compared
   Despite these encouraging results, counteracting biologic         with conventional pharmacotherapy and interventional
processes that potentially limit the regenerative functions of       procedures. Nonetheless, research in this area has gener-
manufactured cell products may prove to be important for             ated a wealth of novel scientific insight and a growing
maximizing the benefits of autologous cell therapies. Re-             number of informative clinical trial experiences with MSC-
cent developments in the understanding of cellular senes-            and regulatory immune cell–based investigational medici-
cence may offer exciting opportunities for the application           nal products. Reassuringly, the safety profiles for such
of DMRT to kidney disease. Premature senescence reduces              cell therapies in patients with kidney diseases and KTx
MSC replicative capacity and limits cell expansion in                enrolled into clinical trials has, thus far, proven to be very
manufacturing protocols. The abundance of senescent cells            good. For some of these clinical applications, preliminary
also fuels proinflammatory pathways in the pathogenesis of            signals of in vivo disease modulation have also emerged
disease processes, such as DM and DKD (108,109). Further-            (26,33,84,86,88,100,101,104), whereas others have lacked ev-
more, the microenvironmental stressors of kidney diseases            idence of efficacy (58).
(uremia, hyperglycemia, kidney aging, RAAS alteration,                  In considering how future clinical effect could be maxi-
oxidative stress, inflammation) contribute to senescent-cell          mized for cell-based DMRT that have undergone early-
accumulation, potentially diminishing endogenous and ex-             phase clinical trials in patients with kidney disease, a num-
ogenous MSC regenerative capacity (105,110). Emerging                ber of critical missing elements should be highlighted: (1)
therapeutic strategies offer the possibility of modulating           development of disease-specific assays to quantify potency
the microenvironments from which primary stromal cells               of, and patient response to, DMRT to account for complex-
are extracted through senescent-cell clearance in vivo. In           ities, such as interindividual heterogeneity and changes in
a pilot clinical trial, we recently observed that a 3-day oral       cell functionality, that occur during ex vivo expansion; (2)
senolytic regimen of dasatinib and quercetin can diminish            definition of optimal dosing, distribution, and frequency of
senescent-cell abundance in adipose and epithelial tissue            administration of DMRT for specific clinical targets; (3)
and improve MSC proliferation in subjects with DKD (111).            consensus on the influence of cryopreservation, which
We and others are also pursuing other preconditioning                may negatively affect the consistency of DMRT therapeutic
methods to optimize autologous MSC functionality. Inter-             effects (6,7,113); (4) increased understanding of relative
ventions such as exposure to hypoxia or melatonin during             clinical efficacy of autologous and allogeneic cell therapies
culture expansion may enhance the prorepair properties of            for specific patient groups; (5) innovations in manufacturing
MSCs (32,112). Taken together, these insights suggest that           procedures that will eventually allow for cost-effective de-
further integration of in vivo or ex vivo conditioning regi-         livery of DMRT to large numbers of patients. As is clear
mens could improve the success of autologous (and perhaps            from Figure 1, other DMRT for which preclinical evidence
also allogeneic) cell-based DMRT in kidney disease.                  bases and technological developments are building await
KIDNEY360 2: 542–557, March, 2021                                    Disease-Modulating Regenerative Therapies in Nephrology, Hickson et al.   553

definitive clinical translation (13,36,114). Increasingly, cross-           3. Oxburgh L, Carroll TJ, Cleaver O, Gossett DR, Hoshizaki DK,
discipline research and innovation has brought the potential                  Hubbell JA, Humphreys BD, Jain S, Jensen J, Kaplan DL,
                                                                              Kesselman C, Ketchum CJ, Little MH, McMahon AP, Shankland
for combinatorial advanced therapies to the forefront of                      SJ, Spence JR, Valerius MT, Wertheim JA, Wessely O, Zheng
translational initiatives in regenerative medicine. Incorpo-                  Y, Drummond IA: (Re)Building a kidney. J Am Soc Nephrol
ration of gene editing and biomaterials science holds future                  28: 1370–1378, 2017 https://doi.org/10.1681/
promise for stabilizing and enhancing the key mechanisms                      ASN.2016101077
of action of cellular therapies identified from preclinical                 4. Little MH, Hale LJ, Howden SE, Kumar SV: Generating kidney
                                                                              from stem cells. Annu Rev Physiol 81: 335–357, 2019 https://
studies or patient profiling in early-phase clinical trials.                   doi.org/10.1146/annurev-physiol-020518-114331
Similarly, clever use of combined pharmacotherapy and                      5. Nishinakamura R: Human kidney organoids: Progress and
DMRT is likely to be critical for optimizing and broaden-                     remaining challenges. Nat Rev Nephrol 15: 613–624, 2019
ing the clinical applications of regenerative medicine. For                   https://doi.org/10.1038/s41581-019-0176-x
                                                                           6. Galipeau J, Sensébé L: Mesenchymal stromal cells: Clinical
example, patient conditioning through the coadministra-                       challenges and therapeutic opportunities. Cell Stem Cell 22:
tion of antisenescence (senolytic) agents to enhance MSC                      824–833, 2018 https://doi.org/10.1016/j.stem.2018.05.004
survival and anti-inflammatory/immune regulatory respon-                    7. Squillaro T, Peluso G, Galderisi U: Clinical trials with mesen-
ses (31,111).                                                                 chymal stem cells: An update. Cell Transplant 25: 829–848,
   Viewed through the lens of four distinct areas of clinical                 2016 https://doi.org/10.3727/096368915X689622
                                                                           8. Zwang NA, Leventhal JR: Cell therapy in kidney transplantation:
nephrology practice, we conclude that the potential for our                   Focus on regulatory T cells. J Am Soc Nephrol 28: 1960–1972,
patients to substantially benefit from DMRT within the next                    2017 https://doi.org/10.1681/ASN.2016111206
decade is high and will be driven by a spirit of “joined-up                9. Wood KJ, Bushell A, Hester J: Regulatory immune cells in
thinking” among basic scientists, biomedical engineers,                       transplantation. Nat Rev Immunol 12: 417–430, 2012 https://
                                                                              doi.org/10.1038/nri3227
technology innovators, clinical trialists, clinicians, and fund-          10. Arcolino FO, Zia S, Held K, Papadimitriou E, Theunis K, Bus-
ing and regulatory bodies.                                                    solati B, Raaijmakers A, Allegaert K, Voet T, Deprest J, Vriens J,
                                                                              Toelen J, van den Heuvel L, Levtchenko E: Urine of preterm
Disclosures                                                                   neonates as a novel source of kidney progenitor cells. J Am Soc
   M. Griffin reports receiving honoraria from American Society of             Nephrol 27: 2762–2770, 2016 https://doi.org/10.1681/
Nephrology, Hebei Medical University (China), and National                    ASN.2015060664
                                                                          11. Leuning DG, Reinders ME, Li J, Peired AJ, Lievers E, de Boer HC,
Institutes of Health; being an associate editor for JASN, being on the        Fibbe WE, Romagnani P, van Kooten C, Little MH, Engelse MA,
editorial boards for Frontiers in Antigen Presenting Cell Biology,            Rabelink TJ: Clinical-grade isolated human kidney perivascular
Frontiers in Renal Pharmacology, Kidney International, and Trans-             stromal cells as an organotypic cell source for kidney re-
plantation; being a section editor for Mayo Clinic Proceedings; and           generative medicine. Stem Cells Transl Med 6: 405–418, 2017
                                                                              https://doi.org/10.5966/sctm.2016-0053
receiving research funding from Randox Laboratories for research
                                                                          12. Rani S, Ryan AE, Griffin MD, Ritter T: Mesenchymal stem cell-
not related to this article. S. Herrmann reports having patents and           derived extracellular vesicles: Toward cell-free therapeutic
inventions with Pfizer, but these are not related to this research. All        applications. Mol Ther 23: 812–823, 2015 https://doi.org/
remaining authors have nothing to disclose.                                   10.1038/mt.2015.44
                                                                          13. McFetridge ML, Del Borgo MP, Aguilar MI, Ricardo SD: The use
                                                                              of hydrogels for cell-based treatment of chronic kidney disease.
Funding
                                                                              Clin Sci (Lond) 132: 1977–1994, 2018 https://doi.org/10.1042/
  L. Hickson is supported by Regenerative Medicine Minnesota                  CS20180434
grant RMM 091718, National Institute of Diabetes and Digestive            14. US Renal Data System, US Renal Data System 2016 annual data
and Kidney Diseases (NIDDK) grants DK109134 and DK123492,                     report 2016. Available at: https://www.ajkd.org/article/S0272-
and NIDDK Diabetic Complications Consortium grants DK076169                   6386(16)30703-X/fulltext. Accessed February 12, 2020
                                                                          15. Long DA, Norman JT, Fine LG: Restoring the renal microvas-
and DK115255 (RRID:SCR_001415, www.diacomp.org). S. Herr-                     culature to treat chronic kidney disease. Nat Rev Nephrol 8:
mann is supported by NIDDK grant DK118120, and by a Mary                      244–250, 2012 https://doi.org/10.1038/nrneph.2011.
Kathryn and Michael B. Panitch Career Development Award. M.                   219
Griffin is supported by European Commission grants 634086                  16. Herrmann SM, Textor SC: Renovascular hypertension. Endo-
                                                                              crinol Metab Clin North Am 48: 765–778, 2019 https://doi.org/
(Horizon 2020 Collaborative Health Project NEPHSTROM) and                     10.1016/j.ecl.2019.08.007
602470 (FP7 Collaborative Health Project VISICORT), Science               17. Eirin A, Textor SC, Lerman LO: Emerging paradigms in chronic
Foundation Ireland grants 09/SRC-B1794 (REMEDI Strategic Re-                  kidney ischemia. Hypertension 72: 1023–1030, 2018 https://
search Cluster) and 13/RC/2073 (CÚRAM Research Centre), and                   doi.org/10.1161/HYPERTENSIONAHA.118.11082
                                                                          18. Cooper CJ, Murphy TP, Cutlip DE, Jamerson K, Henrich W, Reid
the European Regional Development Fund.                                       DM, Cohen DJ, Matsumoto AH, Steffes M, Jaff MR, Prince MR,
                                                                              Lewis EF, Tuttle KR, Shapiro JI, Rundback JH, Massaro JM,
                                                                              D’Agostino RB Sr, Dworkin LD; CORAL Investigators: Stenting
Author Contributions                                                          and medical therapy for atherosclerotic renal-artery stenosis.
   M. Griffin was responsible for funding acquisition; and M. Griffin,          N Engl J Med 370: 13–22, 2014 https://doi.org/10.1056/
S. Herrmann, L. Hickson, and B. McNicholas conceptualized the                 NEJMoa1310753
                                                                          19. Saad A, Herrmann SM, Crane J, Glockner JF, McKusick MA,
article, wrote the original draft, and reviewed and edited the
                                                                              Misra S, Eirin A, Ebrahimi B, Lerman LO, Textor SC: Stent re-
manuscript.                                                                   vascularization restores cortical blood flow and reverses tissue
                                                                              hypoxia in atherosclerotic renal artery stenosis but fails to re-
                                                                              verse inflammatory pathways or glomerular filtration rate. Circ
References                                                                    Cardiovasc Interv 6: 428–435, 2013 https://doi.org/10.1161/
  1. Haseltine WA: The emergence of regenerative medicine: A new              CIRCINTERVENTIONS.113.000219
     field and a new society. J Regen Med 2: 17–23, 2004                  20. Eirin A, Textor SC, Lerman LO: Novel therapeutic strategies for
  2. Mao AS, Mooney DJ: Regenerative medicine: Current therapies              renovascular disease. Curr Opin Nephrol Hypertens 28:
     and future directions. Proc Natl Acad Sci U S A 112:                     383–389, 2019 https://doi.org/10.1097/MNH.
     14452–14459, 2015 https://doi.org/10.1073/pnas.1508520112                0000000000000513
You can also read