Open access peer-reviewed chapter

Allogeneic Mesenchymal Stem Cells as a Treatment for Aging Frailty

Written By

Bryon A. Tompkins, Ana Marie Landin, Victoria Florea, Makoto Natsumeda, Angela C. Rieger, Wayne Balkan, Ivonne Hernandez Schulman and Joshua M. Hare

Submitted: 22 February 2017 Reviewed: 13 April 2017 Published: 30 August 2017

DOI: 10.5772/intechopen.69194

From the Edited Volume

Frailty and Sarcopenia - Onset, Development and Clinical Challenges

Edited by Yannis Dionyssiotis

Chapter metrics overview

1,820 Chapter Downloads

View Full Metrics

Abstract

As life expectancy is projected to increase in the ensuing decades, individuals of older age continue to exceed the previous generation’s lifespan. Advancing age is associated with a reduction in physical and mental functional capacity, and chronic inflammation is a major factor contributing to this decline. A heightened inflammatory state can lead to exhaustion, weakness, weight loss, slow gate speed, and an overall decrease in activity level. These phenotypes define the onset of the disease process known as frailty. Frailty is a growing epidemic, which severely undermines a person’s ability to deal with outside stressors, and increases their rate of hospitalization, institutionalization, and mortality. Current interventions focus on preventative care by improving exercise capacity, strength, nutritional supplementation, diet, and mobility. However, a biological cure has heretofore remained elusive. Here, we introduce the novel therapeutic principle that mesenchymal stem cell (MSC) therapy may represent a safe, practical, and efficacious both the treatment and prevention of frailty in individuals of advancing age. To date, a phase I safety trial reveals an excellent safety profile and suggests that mesenchymal stem cells can ameliorate signs and symptoms of frailty. These early studies lay the groundwork for future large-scale clinical trials of this exciting and novel therapeutic concept that has the potential to expand health span in the aging population.

Keywords

  • mesenchymal stem cells
  • immunomodulation
  • frailty
  • tumor necrosis factor-alpha
  • regenerative medicine

1. Introduction

Projected life expectancy continues to grow worldwide owing to the advancement of new treatments and technologies for leading causes of death such as cardiovascular disease and cancer [1]. Meanwhile, frailty is gaining relevance as a significant clinical syndrome that is associated with increased risk of falls, depression, and disability, leading to higher mortality [2]. Frailty is defined by an age-related decline in reserve and function leading to a reduced ability to cope with acute or external stressors [3] and is characterized by easy tiring, decreased libido, mood disturbance, accelerated osteoporosis, diminished muscle strength, and susceptibility to disease. However, the pathophysiology underlying this syndrome is complex and not clearly understood [4].

Inflammation is a pathophysiologic change that is closely linked with frailty [5]. Aging is associated with immunosenescence or the dysregulation of the innate immune system, resulting in an increase of pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β, further leading to a chronic low-grade inflammatory state [6]. Chronic inflammatory response inhibits the repair, turnover, and adaptation of many tissues, including skeletal muscle [6]. Regeneration of skeletal muscle involves the cross-talk of muscle cells with immune cells, where the pro-inflammatory phenotype of immune cells promotes migration of satellite cells to the injured area, activates satellite cells, and matures newly formed muscle fibers [7]. Muscular degeneration caused by the altered pro-inflammatory state in frail patients is referred to as sarcopenia, another critical physiologic component of frailty.

Aging also produces physiologic changes in the brain, contributing to the development of frailty. Neurons with high metabolic demands, such as the hippocampal pyramidal neurons, are an important mediator in the pathophysiology of cognitive decline and are a key component of the stress response [3]. Concomitant with changes in the immune system, microglial cells, which are the resident immune cells of the central nervous system, are also structurally and functionally altered with aging and undergo senescence, likely causing damage and neuronal death [8]. Accumulating evidence supports an association between frailty, cognitive impairment, and dementia [9, 10].

The brain and endocrine system are intrinsically linked through the hypothalamo-pituitary axis, controlling metabolism via a series of homeostatic hormones. Four major circulating hormones are affected by aging: first, the decrease of insulin-like growth factor 1 (IGF-1) is associated with lower strength and decreased mobility [11]. Second, decreased sex hormone (estradiol and testosterone) increases the release of luteinizing hormone and follicle-stimulating hormone [12]. Third, decreased activity of the adrenocortical cells produces the major sex steroid precursor dehydroepiandrosterone sulfate (DHEA-S), which is associated with a gradual rise in cortisol. DHEA-S directly maintains muscle mass and indirectly prevents the inflammatory pathways that contribute to muscle decline [13, 14]. Finally, the reduced level of 25(OH) vitamin D is associated with the development of osteoporosis [15].

Due to the complexity of multiple inter-related physiological systems that contribute to frailty, there is no gold standard for diagnosing the syndrome. There are currently two models for evaluating frailty: the phenotype model and the cumulative deficit model that forms the basis of the Canadian Study of Health and Aging (CSHA) frailty index [16]. The phenotype model defines frailty as meeting three or more of five criteria: weight loss (>5% of body weight in the previous year), exhaustion (positive response to questions regarding effort required for activity), weakness (decreased grip strength), slow gait speed (>6–7 seconds for walking 15 feet), decreased physical activity, or low energy expenditure (kcal spent per week: males expending <383 kcal, females expending <270 kcal) (Table 1) [17]. This model is simple and easy to use; however, it fails to include factors such as cognitive impairment and highly prevalent conditions associated with functional decline and disability [3].

Frailty phenotypesMSC responsePostulated mechanism of action
Weight lossMaintains total caloric expenditure↓ Inflammation which suppresses the onset of sarcopenia
Exhaustion↑ Pulmonary function, ↓ chronic inflammation↑ Endothelial function, ↓ markers of inflammation
Weakness↑ Physical performance↑ Mitochondrial transfer, ↑endogenous stem cell function
Slow gate speed↑ 6-minute walk distance↑ Endothelial function, ↑ cardiac performance, ↑skeletal muscle performance
Decreased activity level↓ Chronic inflammation, ↑ quality of life↓ TNF-α, ↓ IL-1β, ↑ IL-10

Table 1.

The effect of MSCs on the phenotypes of frailty.

Notes: MSCs home to sites of injury and to enhance repair of damaged tissue (heart, joints, muscle, and blood vessels) and exert their regenerative effects via paracrine signaling, mitochondrial transfer, direct cellular contact, and exosome excretion.

The cumulative deficit model is based on the accumulation of illnesses, functional and cognitive declines, and social situations that are added together to calculate frailty [16]. This model utilizes 20 or more medically and functionally related questions and 92 baseline variables. The deficit model fashioned the origin of the CSHA frailty index [3]. The frailty index measures a number of age-associated health deficits (signs, symptoms, and laboratory values) [18], which is calculated by dividing each ‘deficit’ by the total number tested [3]. The higher the number of deficits, the higher the score, with ‘1’ being the maximum index, which indicates a poorer prognosis. Specifically, Rockwood and Mitnitski noted that an index >0.7 indicates a high risk of mortality [19]. In an effort to simplify this index, the same authors proposed the CSHA clinical frailty score, a 7-point rapid screening tool that was highly correlated with the frailty index; with 7 being the maximum score, indicating “severe frailty” [20]. The clinical frailty score is currently widely used in clinical practice [21].

Advertisement

2. Epidemiology

Universal characteristics that are associated with an increased prevalence of frailty include: chronological age, female sex, racial, and ethnic minority, those in supportive residential settings, and lower income [22]. The prevalence among community-dwelling people over 65 years of age ranges from 4 to 17% in studies with varying geographic features [23, 24]. There are similar trends in Japan, the country with the highest life expectancy in the world. The reason for longevity in Japan is multifactorial. The universal health insurance system, the high population density with close access to hospitals, fish-based diets, awareness of healthy aging in the general public, and lower prevalence of lung cancer despite a higher population of smokers are some contributors [25]. Interestingly, a meta-analyses of frail Japanese patients using the phenotype model demonstrated that the age-stratified-weighted prevalence of frailty was lower in younger age groups (1.9% in 65–69 years, 3.8% in 70–74 years), the same in the 75–79-year age groups (10.0%), but higher in older age groups (20.4% in 80–84 years, 35.1% in ≥85 years) when compared to Western countries [25]. One explanation is that non-Japanese frail older individuals die younger, while Japanese frail older individuals survive longer, leading to a higher prevalence of frailty in their 80s.

Advertisement

3. Intervention and preventive care

While there is no cure for frailty, exercise is considered the most effective intervention to improve quality of life and functionality in frail adults. Improvements in muscle strength and mobility [26, 27] are among the most successful changes reported. Several studies have demonstrated an improvement in muscle strength in older persons with resistance exercise [2830]. A longitudinal study of aging showed that physical activity is associated with a slower progression in functional limitations in older adults over a follow-up period of 6 months [31]. A randomized, placebo-controlled trial compared resistance exercise training, multinutrient supplementation, both interventions, and neither intervention (control group) in frail adults over a period of 10 weeks. The study suggested that high-intensity resistance exercise training improved muscle strength, gait velocity, and stair-climbing power. However, nutritional supplementation did not reduce muscle weakness or physical frailty [28]. Similarly, the LIFE-P (lifestyle interventions and independence for Elders pilot) exploratory study demonstrated that regular physical activity reduces frailty prevalence at a 12-month follow-up time point [32].

Another approach to delay frailty is the use of nutritional supplements, which increase protein and caloric daily intake. Administration of leucine-enriched essential amino acids can increase muscle synthesis through stimulation of the mechanistic target of rapamycin (mTOR) signaling pathway [33]. The PROT-AGE study [34] reviewed the dietary protein intake in older healthy people (>65 years). They found that the optimal protein intake in older persons with sarcopenia is 1.0–1.2 g/kg body weight per day, and higher protein intake >1.2 g/kg body weight per day for those who are exercising. The PROVIDE trial [35] assessed protein supplements enriched in leucine and vitamin D in sarcopenic older adults at high risk for disability. The study demonstrated that the group receiving high quantity protein supplements gained significantly more muscle mass and improved their chair stand ability relative to the control group.

The randomized Controlled Trial of Community-based Nutritional, Physical and Cognitive Training Intervention Programmes for At Risk Frail Elderly (FIT) compared the effects of 6-month interventions with either nutritional supplementation, cognitive training, physical activity, the combination treatments, or no intervention (control) in prefrail and frail adults [36]. Frailty score and status were measured at 3, 6 and 12 months. While these parameters were reduced in all groups, including the controls, the other four groups were significantly improved compared to the controls, at all three time points. However, none of the interventions improved the secondary endpoints, which included: hospitalizations, falls, and performance of activities of daily living [36]. Recent studies suggest that vitamin D plays a role in the pathogenesis and management of frailty [37, 38]. One such study demonstrated that daily doses of ≥800 IU of vitamin D has beneficial effects on balance and muscle strength [39]; while another reported an improvement in balance with a single large dose of vitamin D [40]. While an analysis of 53 trials showed that vitamin D supplementation alone does not prevent fractures in older adults [41], supplementation of vitamin D in combination with calcium can prevent disabling hip fractures among others [41].

While diet and exercise have been thoroughly evaluated, hormone therapy has also been tested. Testosterone undecanoate plus a high-calorie supplement (2108–2416 kJ/day) was compared with a control group (placebo plus a low-calorie supplement (142–191 kJ/day)) in a randomized controlled trial. The results showed that there were no significant differences in frailty scores at either 6- or 12-month follow-up between the groups [42]. While testosterone treatment improves muscle strength, it also increases incidence of adverse cardiovascular events [43]. Estrogen in combination with progestin therapy in postmenopausal women increases the risk of incident breast cancer after 5.6- or 7.1-years of follow-up [44, 45]. Likewise, the benefits of dehydroepiandrosterone sulfate (DHEA-S) supplementation in frail patients have not been demonstrated. Older subjects who received DHEA for 2 years exhibited no beneficial effects on body composition, physical performance, or quality of life [46]. Similarly, 1 year of treatment with insulin-like growth factor 1 (IGF-1) did not alter bone mineral density, fat mass, muscle strength, blood lipid parameters, and measures of postprandial glucose disposal in postmenopausal women [47].

Frailty is increasingly recognized as a clinical state of vulnerability with increased risk of adverse health outcomes. The pathogenesis that underlies this syndrome is multifactorial and elusive, and there is not yet a gold standard for diagnosis or treatment. Exercise and nutritional supplementations are currently the key interventions for frailty. Although helpful, none of them have been proven to treat the disease process. Therefore, an alternative approach to treating frail adults needs to be investigated. Here, we focus on a novel intervention, allogeneic mesenchymal stem cells (allo-MSCs) as a potential therapy for the treatment of frailty. While the mechanism is as yet unclear, we propose that the beneficial effects of MSCs for frailty are due, in large part, to a combination of their immunomodulatory, antifibrotic, and pro-regenerative effects.

Advertisement

4. Regenerative medicine

Frailty is a multifactorial condition triggered by genetic and environmental factors. As previously described, there is a direct association between frailty and the loss of proliferative homeostasis, neurodegeneration, DNA/mitochondrial mutations, free radical accumulation, a rise in pro-inflammatory markers, and increased immunosenescence. Stem cell depletion is a key mechanism postulated to contribute to frailty and its epigenetic dysregulation [4850]. Thus, the repletion of stem cells is an appealing approach to treat this multifactorial dysregulation and MSCs are a particularly attractive candidate. MSCs are a multipotent, self-renewing somatic progenitor cell type that exhibits immunoprivileged properties [5153] and are relatively easy to collect (bone marrow harvest), isolate, and expand [54, 55].

MSCs home to sites of injury, upregulate endogenous stem cells, and reduce inflammation and organ dysfunction [5661]. With respect to age-related diseases, MSCs have demonstrated improvements in ischemic and nonischemic cardiomyopathies [6264], stroke [65], systemic inflammation [66], and Parkinson’s, among others [67]. Although not completely understood, the beneficial effects of stem cells are likely due primarily to paracrine signaling [68, 69] including microvesicle/exosome release [70, 71] and secondarily to direct cellular contact including gap junction formation [72] and mitochondrial exchange via tunneling nanotubes [73, 60].

MSC effects at the molecular level are secondary to the secretion of growth factors, chemokines, and metalloproteinases, including vascular endothelial growth factor (VEGF), angiopoetin-1, fibroblast growth factor, placental growth factor, stem cell-derived factor (SDF), plasminogen activator [74], hepatocyte growth factor/scatter factor (HGF/SF) [75], secreted frizzled-related protein 2 (Sfrp2), hypoxic-induced Akt-regulated stem cell factor (HASF) and IGF-1 and -2 [76]. These molecules stimulate the Akt pathway [77], promote vasculogenesis [56], and protect native cells under hypoxic conditions [68]. In the heart, injection of MSCs produces antifibrotic, anti-inflammatory, and proangiogenic [5759] effects and upregulates the proliferation of endogenous cardiac stem cells [60, 61], while improving the preservation of function of the cells surrounding the sites of injection.

MSCs mediate metabolic changes and stimulate resident cell activation after injury. In vitro studies have shown that MSC-conditioned culture media stimulates resident cardiac stem cells to proliferate, differentiate, and migrate [53, 78] via IGF-1. HASF and Sfrp2 prevent cardiomyocyte apoptosis, promote cardiac stem cell differentiation, and reduce fibrosis after myocardial infarction (MI) [7981].

The absence of major histocompatibility complex (MHC) class II antigens underlies the lack of allo-MSCs to stimulate a major immune response [82] and has generated an interest in their systemic and local application without the need for immunosuppression [62]. In the clinical setting, multiple trials have evaluated and proven the safety and efficacy of MSC therapy [63, 64, 8385]. Several disease processes have been studied in humans: autoimmune diseases, organ transplantation, and as a therapeutic agent after solid organ injury.

Advertisement

5. Immune biomarkers in aging and frailty

Aging and frailty are associated with a dramatic impairment of the ability of the immune system to provide protection from new pathogens. Older frail individual has serious complications that cause adverse health outcomes including acute illness, heightened inflammatory state, and immune dysregulation, which cause a severe impairment in both innate and adaptive immunity and greater susceptibility to infectious diseases, comorbidities, and increased mortality [86, 87]. The accumulation of reactive oxygen species (ROS) in the aging process leads, in part, to chronic activation of Toll-like receptors (TLRs), which in turn leads to an increase in the inflammatory process [8890]. In frailty, the immune phenotype is dysregulated due to incrementing chronic inflammation known as inflammaging and includes increased IL-6, C-reactive peptide (CRP), and TNF-α [87]. Inflammaging plays an important role in the suppression of the immune system and the remodeling of the immune phenotype (Figure 1). The remodeling of the immune phenotype in aging is known as immunosenescence and is marked by several immune biomarkers described below.

Figure 1.

The role of stressors, aging, and inflammation in frailty and the effects of mesenchymal stem cells. As individuals grow older, several stressors (poor nutrition, diseases, oxidative stress, and environmental factors) increase the inflammaging process leading to frailty. Mesenchymal stem cells (MSCs) secrete several factors that block or reverse the inflammaging process and ultimately reverses the effects of frailty.

The immune risk phenotype (IRP), which is the ratio of CD4+ to CD8+ T cells, decreases to <1 in aging and frailty, has been linked to increased risk of mortality [91, 92]. The decreased IRP associated with aging and frailty is due to an expansion of the CD8+ compartment in comparison to the CD4+ compartment. In spite of the expansion of the CD8+ T-cell compartment, the effector T cells in frail individuals have diminished function mostly due to antigen experienced CD8+ T cells re-expressing the naïve marker CD45RA, also known as TEMRA T cells [87]. This expansion of the TEMRA T cell population is exacerbated by factors such as chronic activation due to Cytomegalovirus (CMV) exposure known to be present in >60% of the US population [93]. Finally, the ability to produce protective antibodies upon new antigenic exposure is also severely impaired in aging and frailty due to a remodeling of the B cell compartment. In addition, there is an intrinsic defect in B cells in aging, which causes a decrease in the enzyme, activation-induced cytidine deaminase (AID) leading to diminished ability to switch antibody isotype, which has been correlated to increased TNF-α [94]. The inflammaging process depletes the B cell compartment of switched memory B cells, which are a predictive biomarker for protective vaccine response. In addition, the refractory/exhausted B cell compartment is expanded filling up the B cell niche with unresponsive cells [86, 94, 95].

Advertisement

6. The role of MSCs in inflammaging and immune modulation

Human bone marrow-derived MSCs downregulate the expression of pro-inflammatory cytokines TNF-α, IL-1β, IL-6, and monocyte chemoattractant protein-1 (Figure 2) [96, 32]. Other immunomodulatory properties include the inhibition of dendritic cells, natural killer cells [9799], and T/B cell proliferation via the downregulation of the molecules programmed death-1 (PD-1) transforming growth factor-β, HGF, nitric oxide, indoleamine 2,3-dioxygenase, and prostaglandin-E2 release [52, 68]. Interestingly, MSCs are able to transform pro-inflammatory macrophages (M1) into anti-inflammatory macrophages (M2) by upregulating IGF-1 and IL-10 [100], thereby promoting angiogenesis and cardiomyocyte recovery [101, 102]. Most importantly, MSCs suppress T cell activation, which is crucial due to the high incidence of CMV virus among the US population, causing chronic activation of T cells leading to an exhausted immune phenotype [103].

Figure 2.

MSCs serve as an anti-inflammatory treatment. (A) Depiction of systemic inflammation (white asterisks’). (B). The anti-inflammatory effects of MSCs. Reproduced from Golpanian et al., with permission from the publisher.

Ground-breaking immune-modulatory results in the Randomized Comparison of Allogeneic Versus Autologous Mesenchymal Stem Cells for Nonischemic Dilated Cardiomyopathy: POSEIDON-DCM study demonstrated that 6 months post-TESI, allogeneic human mesenchymal stem cells (hMSCs) were more efficient than auto-hMSCs in reducing serum levels of TNF-α, increased switched memory B cells, decreased exhausted B cells concomitant with a decrease in the percentage of B cells expressing TNF-α, decreased T cell activation and decreased TEMRA T cells [62]. This reversal on the effects of chronic inflammation on these immune biomarkers opens up the feasibility of using allo-hMSC as a treatment to reverse the process of inflammaging and immunosenescence.

Together, these findings are indicative of the safety of MSC therapy in a variety of disease processes. Furthermore, given that MSCs are known to elicit immunomodulatory, neoangiogenic, endogenous cellular proliferative, and antifibrotic effects post-MI in both animal models and clinical trials alike, we anticipate that MSC therapy will act in a similar manner and prove beneficial in older individuals with frailty.

Advertisement

7. CRATUS phase I and II

The therapeutic interventions available to frail older individuals focus on improving the functionality of a precipitously declining quality of life. With the understanding that deteriorating endogenous stem cell function is a key mechanism behind this disease process, the allogeneic human mesenchymal stem cells (allo-hMSC) in patients with aging FRAilTy via intravenous delivery (CRATUS) trial (Figure 3) was established to reverse the untoward effects of frailty. Conducted at the University of Miami Miller School of Medicine, the pilot phase was designed to establish an optimal dose, and tested the hypothesis that allo-hMSCs were safe, well tolerated, and reduced the signs and symptoms of the disease [104]. The study was conducted in a nonrandomized, nonblinded, escalating dosage via peripheral intravenous infusion in 15 frail subjects [104]. Donors were healthy males and females between the ages of 20 and 45 [104]. Three groups of five patients each received either 20 million (M)-, 100M-, or 200M-cells [104]. Patients were followed out to 1-year postinfusion. The pilot phase revealed the two most salient doses (100M- and 200M-cells) [55] and was followed by phase II, a randomized, double-blinded, placebo-controlled trial that further tested the safety and efficacy of allo-hMSCs in frail older individuals.

Figure 3.

Schematic of the CRATUS pilot and randomized phases.

7.1. Results of the phase I study

The 15 subjects in the pilot phase had an average age of 78.4 ± 4.7 years [55]. Eight patients were categorized as vulnerable to frailty with a CSHA score of 4 and the others were mild, with a score of 6 (n = 6) and moderate with a score of 7 (n = 1) [55]. Most importantly, the primary endpoint, treatment-emergent serious adverse events (TE-SAE) at 1-month postinfusion, was met with no adverse reactions in any group [55].

Efficacy outcomes were measured out to 6 months postinfusion as a secondary endpoint. The 6-minute walk distance (6-MWD) test is a measure of functional exercise capacity [105], which can reflect a frail subject’s ability to perform basic activities of daily living. The test has been specifically used to assess subjects with diseases of the musculoskeletal, pulmonary, and cardiovascular systems [106108]; and as such, the subjects experienced a significant improvement in all treatment groups at 3 and 6 months postinfusion with the greatest improvement in the 100M cell-dose group in the phase I [55]. Pulmonary function was measured via FEV1, and improved in the 200M cell-dose group [55]. In regard to physical and quality of life improvement, the SF-36 questionnaire, which has a physical and mental component, yielded an improvement in the 100M-group in the pilot phase. Immunomodulation was also reported with a significant reduction in the pro-inflammatory biomarker serum TNF-α in the 100M and 200M cell-dose groups at 6 months postinfusion [55].

7.2. Update on the phase II study

The 30 subjects in phase II had a mean age of 75.5 ± 7.3 and a mean frailty score of 4 based on the CSHA Clinical Frailty Scale, and were equally randomized to receive 100M-, 200M-cells, or placebo [109]. Safety was the primary endpoint of the study and was measured via the occurrence of TE-SAE at 1-month postinfusion. The trial has completed enrollment, however, as reported in phase 1, preliminary findings show that the treatment is safe and produces significant improvements in the treated subjects in both quality of life and functional status [109].

Advertisement

8. Conclusion

Frailty has increasingly been recognized as a constellation of waning physical and mental qualities secondary to outside stressors, which relate to aging, and confer a vulnerability to adverse health outcomes [110]. Biologically, inflammation and stem cell depletion are at the forefront of this disease process. Early intervention is warranted at the onset of recognized symptoms to reduce the burden of disease progression, hospitalizations, and associated healthcare costs [111, 112]. To date, several important multimodal interventions are available to manage frailty; however, a disease-specific treatment has yet to emerge [112, 113]. Given the positive results of numerous studies utilizing MSCs in a variety of disease processes common to frailty as defined by the physical phenotype model, we believe stem cell therapy will be a treatment of choice for this disease process.

Future implications of stem cell administration in frail or prefrail older individuals may be useful in settings where undo stress may cause rapid physical deterioration. There are a number of medical procedures (breast/colorectal cancer treatment, cardiac surgery, noncardiac elective surgery, etc.) that are taxing to a young healthy individual let alone older individuals [114116]. A preemptive or perioperative administration of MSCs may dampen the immune response, aid in the healing process, and keep at-risk older individuals from declining in functional status (Table 2) [56, 117120]. Given the results from the CRATUS, pilot dose finding [55] and randomized placebo controlled study [109] and the number of medical procedures older individuals undergo, large, randomized, double-blinded clinical trials are warranted to elucidate the efficacy of stem cell therapy in regard to the disease process in itself and its ability to suppress the progression of frailty after strenuous medical interventions.

Future indications applicable to frailtyResponse to MSCsProposed mechanisms of action
OsteoporosisImproved bone mineral content and reduced rate of fracturesReduction in ROS and increased AMPK
Heart diseaseImproved functionIncreased endothelial function, decreased inflammation, reduced cardiac fibrosis, and increased cardiomyogenesis
Delayed healing in InjuryIncreased wound healingDecrease inflammation (TNF-α and IL-1β), Increased IL-10
AutoimmunityDelayed onset/preventionSuppress TH17, induce T regs, and promote TH2 response

Table 2.

MSCs and future indications.

In summary, allogeneic MSCs are immunotolerant in frail older individuals providing clinically meaningful improvements in functional capacity, inflammatory biomarkers, and quality of life patient-reported outcomes. Frailty, the multimodal biologically mediated decline in physiologic reserve, may now have an optimistic therapeutic option.

References

  1. 1. Writing Group M, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB, American Heart Association Statistics C, Stroke Statistics S. Heart disease and stroke statistics-2016 update: A report from the american heart association. Circulation. 2016;133(4):e38–360. DOI: 10.1161/CIR.0000000000000350
  2. 2. Soysal P, Stubbs B, Lucato P, Luchini C, Solmi M, Peluso R, Sergi G, Isik AT, Manzato E, Maggi S, Maggio M, Prina AM, Cosco TD, Wu YT, Veronese N. Inflammation and frailty in the elderly: A systematic review and meta-analysis. Ageing Research Reviews. 2016;31:1–8. DOI: 10.1016/j.arr.2016.08.006
  3. 3. Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people. Lancet. 2013;381(9868):752–762. DOI: 10.1016/S0140-6736(12)62167-9
  4. 4. Gomes MJ, Martinez PF, Pagan LU, Damatto RL, Cezar MD, Lima AR, Okoshi K, Okoshi MP. Skeletal muscle aging: Influence of oxidative stress and physical exercise. Oncotarget. 2017;8(12):20428–20440. DOI: 10.18632/oncotarget.14670
  5. 5. Chen X, Mao G, Leng SX. Frailty syndrome: An overview. Clinical Interventions in Aging. 2014;9:433–441. DOI: 10.2147/CIA.S45300
  6. 6. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2014;69(Suppl 1):S4-9. DOI: 10.1093/gerona/glu057
  7. 7. Tidball JG, Villalta SA Regulatory interactions between muscle and the immune system during muscle regeneration. The American Journal of Physiology. 2010;298(5):R1173–1187. DOI: 10.1152/ajpregu.00735.2009
  8. 8. Luo XG, Ding JQ, Chen SD. Microglia in the aging brain: Relevance to neurodegeneration. Molecular Neurodegeneration. 2010;5:12. DOI: 10.1186/1750-1326-5-12
  9. 9. Boyle PA, Buchman AS, Wilson RS, Leurgans SE, Bennett DA. Physical frailty is associated with incident mild cognitive impairment in community-based older persons. Journal of the American Geriatrics Society. 2010;58(2):248–255. DOI: 10.1111/j.1532-5415.2009.02671.x
  10. 10. Song X, Mitnitski A, Rockwood K. Nontraditional risk factors combine to predict Alzheimer disease and dementia. Neurology. 2011;77(3):227–234. DOI: 10.1212/WNL.0b013e318225c6bc
  11. 11. Cappola AR, Xue QL, Ferrucci L, Guralnik JM, Volpato S, Fried LP. Insulin-like growth factor I and interleukin-6 contribute synergistically to disability and mortality in older women. The Journal of Clinical Endocrinology & Metabolism. 2003;88(5):2019–2025. DOI: 10.1210/jc.2002-021694
  12. 12. Travison TG, Nguyen AH, Naganathan V, Stanaway FF, Blyth FM, Cumming RG, Le Couteur DG, Sambrook PN, Handelsman DJ. Changes in reproductive hormone concentrations predict the prevalence and progression of the frailty syndrome in older men: The concord health and ageing in men project. The Journal of Clinical Endocrinology & Metabolism. 2011;96(8):2464–2474. DOI: 10.1210/jc.2011-0143
  13. 13. Leng SX, Cappola AR, Andersen RE, Blackman MR, Koenig K, Blair M, Walston JD. Serum levels of insulin-like growth factor-I (IGF-I) and dehydroepiandrosterone sulfate (DHEA-S), and their relationships with serum interleukin-6, in the geriatric syndrome of frailty. Aging Clinical and Experimental Research. 2004;16(2):153–157
  14. 14. Lamberts SW. The endocrinology of aging and the brain. Archives of Neurology. 2002;59(11):1709–1711
  15. 15. Puts MT, Visser M, Twisk JW, Deeg DJ, Lips P. Endocrine and inflammatory markers as predictors of frailty. Clin Endocrinology (Oxf). 2005;63(4):403–411. DOI: 10.1111/j.1365-2265.2005.02355.x
  16. 16. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of deficits. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2007;62(7):722–727
  17. 17. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, McBurnie MA, Cardiovascular Health Study Collaborative Research G. Frailty in older adults: Evidence for a phenotype. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2001;56(3):M146-156
  18. 18. Rockwood K, Blodgett JM, Theou O, Sun MH, Feridooni HA, Mitnitski A, Rose RA, Godin J, Gregson E, Howlett SE. A frailty index based on deficit accumulation quantifies mortality risk in humans and in mice. Scientific Reports. 2017;7:43068. DOI: 10.1038/srep43068
  19. 19. Rockwood K, Mitnitski A. Limits to deficit accumulation in elderly people. Mechanisms of Ageing and Development. 2006;127(5):494–496. DOI: 10.1016/j.mad.2006.01.002
  20. 20. Rockwood K, Song X, MacKnight C, Bergman H, Hogan DB, McDowell I, Mitnitski A. A global clinical measure of fitness and frailty in elderly people. Canadian Medical Association Journal. 2005;173(5):489–495. DOI: 10.1503/cmaj.050051
  21. 21. Ritt M, Ritt JI, Sieber CC, Gassmann KG. Comparing the predictive accuracy of frailty, comorbidity, and disability for mortality: A 1-year follow-up in patients hospitalized in geriatric wards. Clinical Interventions in Aging. 2017;12:293–304. DOI: 10.2147/CIA.S124342
  22. 22. Bandeen-Roche K, Seplaki CL, Huang J, Buta B, Kalyani RR, Varadhan R, Xue QL, Walston JD, Kasper JD. Frailty in older adults: A nationally representative profile in the United States. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2015;70(11):1427–1434. DOI: 10.1093/gerona/glv133
  23. 23. Collard RM, Boter H, Schoevers RA, Oude Voshaar RC. Prevalence of frailty in community-dwelling older persons: A systematic review. Journal of the American Geriatrics Society. 2012;60(8):1487–1492. DOI: 10.1111/j.1532-5415.2012.04054.x
  24. 24. Santos-Eggimann B, Cuenoud P, Spagnoli J, Junod J Prevalence of frailty in middle-aged and older community-dwelling Europeans living in 10 countries. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2009;64(6):675–681. DOI: 10.1093/gerona/glp012
  25. 25. Kojima G, Iliffe S, Taniguchi Y, Shimada H, Rakugi H, Walters K. Prevalence of frailty in Japan: A systematic review and meta-analysis. Journal of Epidemiology. 2016; pii:S0917-5040(16):30083–1. DOI: 10.1016/j.je.2016.09.008
  26. 26. Clegg AP, Barber SE, Young JB, Forster A, Iliffe SJ. Do home-based exercise interventions improve outcomes for frail older people? Findings from a systematic review. Reviews in Clinical Gerontology. 2012;22(1):68–78. DOI: 10.1017/S0959259811000165
  27. 27. Theou O, Stathokostas L, Roland KP, Jakobi JM, Patterson C, Vandervoort AA, Jones GR. The effectiveness of exercise interventions for the management of frailty: A systematic review. Journal of Aging Research. 2011;2011:569194. DOI: 10.4061/2011/569194
  28. 28. Fiatarone MA, O'Neill EF, Ryan ND, Clements KM, Solares GR, Nelson ME, Roberts SB, Kehayias JJ, Lipsitz LA, Evans WJ. Exercise training and nutritional supplementation for physical frailty in very elderly people. New England Journal of Medicine. 1994;330(25):1769–1775. DOI: 10.1056/NEJM199406233302501
  29. 29. Tieland M, Dirks ML, van der Zwaluw N, Verdijk LB, van de Rest O, de Groot LC, van Loon LJ. Protein supplementation increases muscle mass gain during prolonged resistance-type exercise training in frail elderly people: A randomized, double-blind, placebo-controlled trial. Journal of the American Medical Directors Association. 2012;13(8):713–719. DOI: 10.1016/j.jamda.2012.05.020
  30. 30. Churchward-Venne TA, Tieland M, Verdijk LB, Leenders M, Dirks ML, de Groot LC, van Loon LJ. There are no nonresponders to resistance-type exercise training in older men and women. Journal of the American Medical Directors Association. 2015;16(5):400–411. DOI: 10.1016/j.jamda.2015.01.071
  31. 31. Miller ME, Rejeski WJ, Reboussin BA, Ten Have TR, Ettinger WH. Physical activity, functional limitations, and disability in older adults. Journal of the American Geriatrics Society. 2000;48(10):1264–1272
  32. 32. Cesari M, Vellas B, Hsu FC, Newman AB, Doss H, King AC, Manini TM, Church T, Gill TM, Miller ME, Pahor M, Group LS. A physical activity intervention to treat the frailty syndrome in older persons-results from the LIFE-P study. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2015;70(2):216–222. DOI: 10.1093/gerona/glu099
  33. 33. Malafarina V, Uriz-Otano F, Iniesta R, Gil-Guerrero L. Effectiveness of nutritional supplementation on muscle mass in treatment of sarcopenia in old age: A systematic review. Journal of the American Medical Directors Association. 2013;14(1):10–17. DOI: 10.1016/j.jamda.2012.08.001
  34. 34. Bauer J, Biolo G, Cederholm T, Cesari M, Cruz-Jentoft AJ, Morley JE, Phillips S, Sieber C, Stehle P, Teta D, Visvanathan R, Volpi E, Boirie Y. Evidence-based recommendations for optimal dietary protein intake in older people: A position paper from the PROT-AGE Study Group. Journal of the American Medical Directors Association. 2013;14(8):542–559. DOI: 10.1016/j.jamda.2013.05.021
  35. 35. Bauer JM, Verlaan S, Bautmans I, Brandt K, Donini LM, Maggio M, McMurdo ME, Mets T, Seal C, Wijers SL, Ceda GP, De Vito G, Donders G, Drey M, Greig C, Holmback U, Narici M, McPhee J, Poggiogalle E, Power D, Scafoglieri A, Schultz R, Sieber CC, Cederholm T. Effects of a vitamin D and leucine-enriched whey protein nutritional supplement on measures of sarcopenia in older adults, the PROVIDE study: A randomized, double-blind, placebo-controlled trial. Journal of the American Medical Directors Association. 2015;16(9):740–747. DOI: 10.1016/j.jamda.2015.05.021
  36. 36. Ng TP, Feng L, Nyunt MS, Feng L, Niti M, Tan BY, Chan G, Khoo SA, Chan SM, Yap P, Yap KB. Nutritional, physical, cognitive, and combination interventions and frailty reversal among older adults: A randomized controlled trial. The American Journal of Medicine. 2015;128(11):1225–1236 e1221. DOI: 10.1016/j.amjmed.2015.06.017
  37. 37. Wong YY, Flicker L. Hypovitaminosis D and frailty: Epiphenomenon or causal? Maturitas. 2015;82(4):328–335. DOI: 10.1016/j.maturitas.2015.07.027
  38. 38. Artaza-Artabe I, Saez-Lopez P, Sanchez-Hernandez N, Fernandez-Gutierrez N, Malafarina V. The relationship between nutrition and frailty: Effects of protein intake, nutritional supplementation, vitamin D and exercise on muscle metabolism in the elderly. A systematic review. Maturitas. 2016;93:89–99. DOI: 10.1016/j.maturitas.2016.04.009
  39. 39. Muir SW, Montero-Odasso M. Effect of vitamin D supplementation on muscle strength, gait and balance in older adults: A systematic review and meta-analysis. Journal of the American Geriatrics Society. 2011;59(12):2291–2300. DOI: 10.1111/j.1532-5415.2011.03733.x
  40. 40. Dhesi JK, Jackson SH, Bearne LM, Moniz C, Hurley MV, Swift CG, Allain TJ. Vitamin D supplementation improves neuromuscular function in older people who fall. Age and Ageing. 2004;33(6):589–595. DOI: 10.1093/ageing/afh209
  41. 41. Avenell A, Mak JC, O'Connell D. Vitamin D and vitamin D analogues for preventing fractures in post-menopausal women and older men. The Cochrane Database of Systematic Reviews. 2014;(4):CD000227. DOI: 10.1002/14651858.CD000227.pub4
  42. 42. Theou O, Chapman I, Wijeyaratne L, Piantadosi C, Lange K, Naganathan V, Hunter P, Cameron ID, Rockwood K, Visvanathan R. Can an intervention with testosterone and nutritional supplement improve the frailty level of under-nourished older people? The Journal of Frailty & Aging. 2016;5(4):247–252. DOI: 10.14283/jfa.2016.108
  43. 43. Basaria S, Coviello AD, Travison TG, Storer TW, Farwell WR, Jette AM, Eder R, Tennstedt S, Ulloor J, Zhang A, Choong K, Lakshman KM, Mazer NA, Miciek R, Krasnoff J, Elmi A, Knapp PE, Brooks B, Appleman E, Aggarwal S, Bhasin G, Hede-Brierley L, Bhatia A, Collins L, LeBrasseur N, Fiore LD, Bhasin S. Adverse events associated with testosterone administration. The New England Journal of Medicine. 2010;363(2):109–122. DOI: 10.1056/NEJMoa1000485
  44. 44. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J, Writing Group for the Women's Health Initiative I. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: Principal results from the women's health initiative randomized controlled trial. Journal of the American Medical Association. 2002;288(3):321–333
  45. 45. Prentice RL, Manson JE, Langer RD, Anderson GL, Pettinger M, Jackson RD, Johnson KC, Kuller LH, Lane DS, Wactawski-Wende J, Brzyski R, Allison M, Ockene J, Sarto G, Rossouw JE. Benefits and risks of postmenopausal hormone therapy when it is initiated soon after menopause. American Journal of Epidemiology. 2009;170(1):12–23. DOI: 10.1093/aje/kwp115
  46. 46. Nair KS, Rizza RA, O'Brien P, Dhatariya K, Short KR, Nehra A, Vittone JL, Klee GG, Basu A, Basu R, Cobelli C, Toffolo G, Dalla Man C, Tindall DJ, Melton 3rd LJ, Smith GE, Khosla S, Jensen MD. DHEA in elderly women and DHEA or testosterone in elderly men. The New England Journal of Medicine. 2006;355(16):1647–1659. DOI: 10.1056/NEJMoa054629
  47. 47. Friedlander AL, Butterfield GE, Moynihan S, Grillo J, Pollack M, Holloway L, Friedman L, Yesavage J, Matthias D, Lee S, Marcus R, Hoffman AR. One year of insulin-like growth factor I treatment does not affect bone density, body composition, or psychological measures in postmenopausal women. The Journal of Clinical Endocrinology and Metabolism. 2001;86(4):1496–1503. DOI: 10.1210/jcem.86.4.7377
  48. 48. Peffers MJ, Collins J, Loughlin J, Proctor C, Clegg PD.. A proteomic analysis of chondrogenic, osteogenic and tenogenic constructs from ageing mesenchymal stem cells. Stem Cell Research & Therapy. 2016;7(1):133. DOI: 10.1186/s13287-016-0384-2
  49. 49. Sethe S, Scutt A, Stolzing A2006. Aging of mesenchymal stem cells. Ageing Research Reviews. 2006;5(1):91–116. DOI: 10.1016/j.arr.2005.10.001
  50. 50. Stolzing A, Jones E, McGonagle D, Scutt A. Age-related changes in human bone marrow-derived mesenchymal stem cells: Consequences for cell therapies. Mechanisms of Ageing and Development. 2008;129(3):163–173. DOI: 10.1016/j.mad.2007.12.002
  51. 51. Chapel A, Bertho JM, Bensidhoum M, Fouillard L, Young RG, Frick J, Demarquay C, Cuvelier F, Mathieu E, Trompier F, Dudoignon N, Germain C, Mazurier C, Aigueperse J, Borneman J, Gorin NC, Gourmelon P, Thierry D. Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome. The Journal of Gene Medicine 2003;5(12):1028–1038. DOI: 10.1002/jgm.452
  52. 52. Augello A, Tasso R, Negrini SM, Amateis A, Indiveri F, Cancedda R, Pennesi G. Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. European Journal of Immunology. 2005;35(5):1482–1490. DOI: 10.1002/eji.200425405
  53. 53. Mourkioti F, Rosenthal N. IGF-1, inflammation and stem cells: Interactions during muscle regeneration. Trends in Immunology. 2005;26(10):535–542. DOI: 10.1016/j.it.2005.08.002
  54. 54. Sanina C, Hare JM. Mesenchymal stem cells as a biological drug for heart disease: Where are we with cardiac cell-Based Therapy? Circulation Research. 2005;117(3):229–233. DOI: 10.1161/CIRCRESAHA.117.306306
  55. 55. Golpanian S, DiFede DL, Khan A, et al. Allogeneic Human Mesenchymal Stem Cell Infusions for Aging Frailty. J Gerontol A Biol Sci Med Sci. DOI: 10.1093/gerona/glx056.
  56. 56. Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the damaged heart: Mesenchymal stem cells, cell-based therapy, and engineered heart tissue. Physiological Reviews. 2016;96(3):1127–1168. DOI: 10.1152/physrev.00019.2015
  57. 57. Cao Y, Gomes S, Rangel EB, Paulino EC, Fonseca TL, Li J, Telxelra MB, Gouvela CH, Bianco AC, Kapiloff MS, Balkan W, Hare JM. S-nitrosoglutathione reductase–dependent PPARγ denitrosylation participates in adipogenesis and osteogenesis. The Journal of Clinical Investigation. 2015;125(4):1679–91. DOI: 10.1172/JCI73780
  58. 58. Karantalis V, Hare JM. Use of mesenchymal stem cells for therapy of cardiac disease. Circulation Research. 2015;116(8):1413–1430. DOI: 10.1161/CIRCRESAHA.116.303614
  59. 59. Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, Fishman J, Pattany P, McNiece I, Conte J, Schulman S, Wu K, Shah A, Breton E, Davis-Sproul J, Schwarz R, Feigenbaum G, Mushtaq M, Suncion VY, Lardo AC, Borrello I, Mendizabal A, Karas TZ, Byrnes J, Lowery M, Heldman AW, Hare JM. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The prospective randomized study of mesenchymal stem cell therapy in patients undergoing Cardiac Surgery (PROMETHEUS) trial. Circulation Research. 2014;114(8):1302–1310. DOI: 10.1161/CIRCRESAHA.114.303180
  60. 60. Hatzistergos KE, Quevedo H, Oskouei BN, Hu Q, Feigenbaum GS, Margitich IS, Mazhari R, Boyle AJ, Zambrano JP, Rodriguez JE, Dulce R, Pattany PM, Valdes D, Revilla C, Heldman AW, McNiece I, Hare JM . Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circulation Research. 2010;107(7):913–922. DOI: 10.1161/CIRCRESAHA.110.222703
  61. 61. Premer C, Blum A, Bellio MA, Schulman IH, Hurwitz BE, Parker M, Dermarkarian CR, DiFede DL, Balkan W, Khan A, Hare JM. Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine. 2015;2(5):467–475. DOI: 10.1016/j.ebiom.2015.03.020
  62. 62. Hare JM, DiFede DL, Castellanos AM, Florea V, Landin AM, El-Khorazaty J, Khan A, Mushtaq M, Lowery MH, Byrnes JJ, Hendel RC, Cohen MG, Alfonso CE, Valasaki K, Pujol MV, Golpanian S, Ghersin E, Fishman JE, Pattany P, Gomes SA, Delgado C, Miki R, Abuzeid F, Vidro-Casiano M, Premer C, Medina A, Porras V, Hatzistergos KE, Anderson E, Mendizabal A, Mitrani R, Heldman AW. Randomized comparison of allogeneic vs. autologous mesenchymal stem cells for non-lschemic dilated cardiomyopathy: POSEIDON-DCM trial. Journal of the American College of Cardiology. 2016; pii: S0735-1097(16)36906–6. DOI:10.1016/j.jacc.2016.11.009
  63. 63. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB, Jr., Reisman MA, Schaer GL, Sherman W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. Journal of the American College of Cardiology. 2009;54(24):2277–2286. DOI: 10.1016/j.jacc.2009.06.055
  64. 64. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J, Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C, Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW, George R, Lardo A. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The POSEIDON randomized trial. Journal of the American Medical Association. 2012;308(22):2369–2379. DOI: 10.1001/jama.2012.25321
  65. 65. Gutierrez-Fernandez M, Rodriguez-Frutos B, Otero-Ortega L, Ramos-Cejudo J, Fuentes B, Diez-Tejedor E. Adipose tissue-derived stem cells in stroke treatment: From bench to bedside. Discovery Medicine. 2013;16(86):37–43
  66. 66. Weiss DJ, Casaburi R, Flannery R, LeRoux-Williams M, Tashkin DP. A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest. 2013;143(6):1590–1598. DOI: 10.1378/chest.12-2094
  67. 67. Trounson A, McDonald C. Stem cell therapies in clinical trials: Progress and challenges. Cell Stem Cell. 2015;17(1):11–22. DOI: http://dx. DOI.org/10.1016/j.stem.2015.06.007
  68. 68. Hodgkinson CP, Bareja A, Gomez JA, Dzau VJ. Emerging concepts in paracrine mechanisms in regenerative cardiovascular medicine and biology. Circulation Research. 2016;118(1):95–107. DOI: 10.1161/CIRCRESAHA.115.305373
  69. 69. Ranganath SH, Levy O, Inamdar MS, Karp JM. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell. 2012;10(3):244–258. DOI: 10.1016/j.stem.2012.02.005
  70. 70. Figeac F, Lesault PF, Le Coz O, Damy T, Souktani R, Trebeau C, Schmitt A, Ribot J, Mounier R, Guguin A, Manier C, Surenaud M, Hittinger L, Dubois-Rande JL, Rodriguez AM.. Nanotubular crosstalk with distressed cardiomyocytes stimulates the paracrine repair function of mesenchymal stem cells. Stem Cells. 2014;32(1):216–230. DOI: 10.1002/stem.1560
  71. 71. Akyurekli C, Le Y, Richardson RB, Fergusson D, Tay J, Allan DS. A systematic review of preclinical studies on the therapeutic potential of mesenchymal stromal cell-derived microvesicles. Stem Cell Reviews. 2014;11(1):150–60. DOI: 0.1007/s12015-014-9545-9
  72. 72. Hahn JY, Cho HJ, Kang HJ, Kim TS, Kim MH, Chung JH, Bae JW, Oh BH, Park YB, Kim HS. Pre-treatment of mesenchymal stem cells with a combination of growth factors enhances gap junction formation, cytoprotective effect on cardiomyocytes, and therapeutic efficacy for myocardial infarction. Journal of the American College of Cardiology. 2008;51(9):933–943. DOI: 10.1016/j.jacc.2007.11.040
  73. 73. Mazhari R, Hare JM. Mechanisms of action of mesenchymal stem cells in cardiac repair: Potential influences on the cardiac stem cell niche. Nature Clinical Practice Cardiovascular Medicine. 2007;4(Suppl 1):S21-26. DOI: 10.1038/ncpcardio0770
  74. 74. Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, Epstein SE. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circulation Research. 2004;94(5):678–685. DOI: 10.1161/01.RES.0000118601.37875.AC 01.RES.0000118601.37875.AC [pii]
  75. 75. Weimar IS, Miranda N, Muller EJ, Hekman A, Kerst JM, de Gast GC, Gerritsen WR. Hepatocyte growth factor/scatter factor (HGF/SF) is produced by human bone marrow stromal cells and promotes proliferation, adhesion and survival of human hematopoietic progenitor cells (CD34+). Experimental Hematology. 1998;26(9):885–894
  76. 76. Cheng SL, Zhang SF, Mohan S, Lecanda F, Fausto A, Hunt AH, Canalis E, Avioli LV. Regulation of insulin-like growth factors I and II and their binding proteins in human bone marrow stromal cells by dexamethasone. The Journal of Cellular Biochemistry. 1998;71(3):449–458
  77. 77. Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, Noiseux N, Zhang L, Pratt RE, Ingwall JS, Dzau VJ. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nature Medicine. 2005;11(4):367–368
  78. 78. Engels MC, Rajarajan K, Feistritzer R, Sharma A, Nielsen UB, Schalij MJ, de Vries AA, Pijnappels DA, Wu SM. Insulin-like growth factor promotes cardiac lineage induction in vitro by selective expansion of early mesoderm. Stem Cells. 2014;32(6):1493–1502. DOI: 10.1002/stem.1660
  79. 79. Schmeckpeper J, Verma A, Yin L, Beigi F, Zhang L, Payne A, Zhang Z, Pratt RE, Dzau VJ, Mirotsou M. Inhibition of Wnt6 by Sfrp2 regulates adult cardiac progenitor cell differentiation by differential modulation of Wnt pathways. Journal of Molecular and Cellular Cardiology. 2015;85:215–225. DOI: 10.1016/j.yjmcc.2015.06.003
  80. 80. Kobayashi K, Luo M, Zhang Y, Wilkes DC, Ge G, Grieskamp T, Yamada C, Liu TC, Huang G, Basson CT, Kispert A, Greenspan DS, Sato TN. Secreted frizzled-related protein 2 is a procollagen C proteinase enhancer with a role in fibrosis associated with myocardial infarction. Nature Cell Biology. 2009;11(1):46–55. DOI: 10.1038/ncb1811
  81. 81. Mastri M, Shah Z, Hsieh K, Wang X, Wooldridge B, Martin S, Suzuki G, Lee T. Secreted frizzled-related protein 2 as a target in antifibrotic therapeutic intervention. American Journal of Physiology Cell physiology. 2014;306(6):C531-539. DOI: 10.1152/ajpcell.00238.2013
  82. 82. Karantalis V, Schulman IH, Balkan W, Hare JM. Allogeneic cell therapy: A new paradigm in therapeutics. Circulation Research. 2015;116(1):12–15. DOI: 10.1161/CIRCRESAHA.114.305495
  83. 83. Hare JM, DiFede DL, Rieger AC, Florea V, Landin AM, El-Khorazaty J, Khan A, Mushtaq M, Lowery MH, Byrnes JJ, Hendel RC, Cohen MG, Alfonso CE, Valasaki K, Pujol MV, Golpanian S, Ghersin E, Fishman JE, Pattany P, Gomes SA, Delgado C, Miki R, Abuzeid F, Vidro-Casiano M, Premer C, Medina A, Porras V, Hatzistergos KE, Anderson E, Mendizabal A, Mitrani R, Heldman AW. Randomized comparison of allogeneic versus autologous mesenchymal stem cells for nonischemic dilated cardiomyopathy: POSEIDON-DCM trial. Journal of the American College of Cardiology. 2017;69(5):526–537. DOI: 10.1016/j.jacc.2016.11.009
  84. 84. Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, Zhang JJ, Chunhua RZ, Liao LM, Lin S, Sun JP. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. The American Journal of Cardiology. 2004;94(1):92–95. DOI: 10.1016/j.amjcard.2004.03.034
  85. 85. Chullikana A, Majumdar AS, Gottipamula S, Krishnamurthy S, Kumar AS, Prakash VS, Gupta PK. Randomized, double-blind, phase I/II study of intravenous allogeneic mesenchymal stromal cells in acute myocardial infarction. Cytotherapy. 2015;17(3):250–261. DOI: 10.1016/j.jcyt.2014.10.009
  86. 86. Frasca D, Blomberg BB . Inflammaging decreases adaptive and innate immune responses in mice and humans. Biogerontology. 2016;17(1):7–19. DOI: 10.1007/s10522-015-9578-8
  87. 87. Mitnitski A, Collerton J, Martin-Ruiz C, Jagger C, von Zglinicki T, Rockwood K, Kirkwood TB. Age-related frailty and its association with biological markers of ageing. BMC Medicine. 2015;13:161. DOI: 10.1186/s12916-015-0400-x
  88. 88. Cannizzo ES, Clement CC, Sahu R, Follo C, Santambrogio L. Oxidative stress, inflamm-aging and immunosenescence. Journal of Proteomics. 2011;74(11):2313–2323. DOI: 10.1016/j.jprot.2011.06.005
  89. 89. Albert C. Shaw SJ, Hannah Greenwood, Alexander Panda, and Janet M. Lord. Aging of the Innate Immune System. Current Opinion in Immunology. 2010;22(4):507–513. DOI: 10.1016/j.coi.2010.05.003
  90. 90. Dewan SK, Zheng SB, Xia SJ, Bill K . Senescent remodeling of the immune system and its contribution to the predisposition of the elderly to infections. Chinese Medical Journal(Engl)). 2012;125(18):3325–3331
  91. 91. Wikby A, Ferguson F, Forsey R, Thompson J, Strindhall J, Lofgren S, Nilsson BO, Ernerudh J, Pawelec G, Johansson B. An immune risk phenotype, cognitive impairment, and survival in very late life: Impact of allostatic load in Swedish octogenarian and nonagenarian humans. Journals of Gerontology Series A Biological Sciences and Medical Sciences. 2005;60(5):556–565
  92. 92. Pawelec G. Immunity and ageing in man. Experimental Gerontology. 2006;41(12):1239–1242. DOI: 10.1016/j.exger.2006.09.005
  93. 93. Zhang LJ, Hanff P, Rutherford C, Churchill WH, Crumpacker CS. Detection of human cytomegalovirus DNA, RNA, and antibody in normal donor blood. Journal of Infectious Diseases. 1995;171(4):1002–1006
  94. 94. Frasca D, Diaz A, Romero M, Landin AM, Phillips M, Lechner SC, Ryan JG, Blomberg BB. Intrinsic defects in B cell response to seasonal influenza vaccination in elderly humans. Vaccine. 2010;28(51):8077–8084
  95. 95. Frasca D, Diaz A, Romero M, Phillips M, Mendez NV, Landin AM, Blomberg BB. Unique biomarkers for B-cell function predict the serum response to pandemic H1N1 influenza vaccine. International Immunology. 2012;24(3):175–182. DOI: 10.1093/intimm/dxr123
  96. 96. Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, Go K, Phinney DG. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proceedings of the National Academy of Sciences of the United States of America . 2007;104(26):11002–11007. DOI: 10.1073/pnas.0704421104
  97. 97. Djouad F, Charbonnier LM, Bouffi C, Louis-Plence P, Bony C, Apparailly F, Cantos C, Jorgensen C, Noel D. Mesenchymal stem cells inhibit the differentiation of dendritic cells through an interleukin-6-dependent mechanism. Stem Cells. 2007;25(8):2025–2032. DOI: 10.1634/stemcells.2006-0548
  98. 98. Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L. MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: Central role of MSC-derived prostaglandin E2. Blood. 2009;113(26):6576–6583. DOI: 10.1182/blood-2009-02-203943
  99. 99. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: Role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008;111(3):1327–1333. DOI: 10.1182/blood-2007-02-074997
  100. 100. Burchfield JS, Iwasaki M, Koyanagi M, Urbich C, Rosenthal N, Zeiher AM, Dimmeler S. Interleukin-10 from transplanted bone marrow mononuclear cells contributes to cardiac protection after myocardial infarction. Circulation Research. 2008;103(2):203–211. DOI: 10.1161/circresaha.108.178475
  101. 101. Yano T, Miura T, Whittaker P, Miki T, Sakamoto J, Nakamura Y, Ichikawa Y, Ikeda Y, Kobayashi H, Ohori K, Shimamoto K. Macrophage colony-stimulating factor treatment after myocardial infarction attenuates left ventricular dysfunction by accelerating infarct repair. Journal of the American College of Cardiology. 2006;47(3):626–634. DOI: 10.1016/j.jacc.2005.09.037
  102. 102. Troidl C, Mollmann H, Nef H, Masseli F, Voss S, Szardien S, Willmer M, Rolf A, Rixe J, Troidl K, Kostin S, Hamm C, Elsasser A. Classically and alternatively activated macrophages contribute to tissue remodelling after myocardial infarction. Journal of Cellular and Molecular Medicine. 2009;13(9b):3485–3496. DOI: 10.1111/j.1582-4934.2009.00707.x
  103. 103. van der Heiden M, van Zelm MC, Bartol SJ, de Rond LG, Berbers GA, Boots AM, Buisman A-M. Differential effects of Cytomegalovirus carriage on the immune phenotype of middle-aged males and females. Scientific Reports. 2016;6:26892
  104. 104. Golpanian S, DiFede DL, Pujol MV, Lowery MH, Levis-Dusseau S, Goldstein BJ, Schulman IH, Longsomboon B, Wolf A, Khan A, Heldman AW, Goldschmidt-Clermont PJ, Hare JM. Rationale and design of the allogeneiC human mesenchymal stem cells (hMSC) in patients with aging fRAilTy via intravenoUS delivery (CRATUS) study: A phase I/II, randomized, blinded and placebo controlled trial to evaluate the safety and potential efficacy of allogeneic human mesenchymal stem cell infusion in patients with aging frailty. Oncotarget. 2016;7(11):11899–11912. DOI: 10.18632/oncotarget.7727
  105. 105. Guyatt GH, Sullivan MJ, Thompson PJ, Fallen EL, Pugsley SO, Taylor DW, Berman LB.. The 6-minute walk: A new measure of exercise capacity in patients with chronic heart failure. Canadian Medical Association Journal. 1985;132(8):919–923
  106. 106. Castillo-Moreno JA, Garcia-Escribano IA, Martinez-Pascual-de-Riquelme M, Jaulent-Huertas L, Dau-Villarreal DF, Rubio-Paton R, Giner-Caro JA, Bonaque-Gonzalez JC, Consuegra-Sanchez L. Prognostic usefulness of the 6-minute walk test in patients with severe aortic stenosis. American Journal of Cardiology. 2016;118(8):1239–1243. DOI: 10.1016/j.amjcard.2016.07.042
  107. 107. Albarrati AM, Gale NS, Enright S, Munnery MM, Cockcroft JR, Shale DJ. A simple and rapid test of physical performance inchronic obstructive pulmonary disease. International Journal of Chronic Obstructive Pulmonary Disease. 2016;11:1785–1791. DOI: 10.2147/COPD.S106151
  108. 108. Bello L, Campadello P, Barp A, Fanin M, Semplicini C, Soraru G, Caumo L, Calore C, Angelini C, Pegoraro E. Functional changes in Becker muscular dystrophy: Implications for clinical trials in dystrophinopathies. Scientific Reports. 2016;6:32439. DOI: 10.1038/srep32439
  109. 109. Hare JM, Difede DL, Khan A, Landin AM, Tompkins B, Goldschmidt PJ, Mendizabal A. Allogeneic mesenchymal stem cells to treat aging frailty: A randomized, placebo-controlled intervention study. The Gerontologist. 2016;56(Suppl 3):50–51
  110. 110. Walston J, Hadley EC, Ferrucci L, Guralnik JM, Newman AB, Studenski SA, Ershler WB, Harris T, Fried LP. Research agenda for frailty in older adults: Toward a better understanding of physiology and etiology: Summary from the American Geriatrics Society/National Institute on Aging Research Conference on Frailty in Older Adults. Journal of the American Geriatrics Society. 2006;54(6):991–1001. DOI: 10.1111/j.1532-5415.2006.00745.x
  111. 111. Cerreta F, Eichler HG, Rasi G. Drug policy for an aging population--the European Medicines Agency's geriatric medicines strategy. The New England Journal of Medicine. 2012;367(21):1972–1974. DOI: 10.1056/NEJMp1209034
  112. 112. Morley JE, Vellas B, van Kan GA, Anker SD, Bauer JM, Bernabei R, Cesari M, Chumlea WC, Doehner W, Evans J, Fried LP, Guralnik JM, Katz PR, Malmstrom TK, McCarter RJ, Gutierrez Robledo LM, Rockwood K, von Haehling S, Vandewoude MF, Walston J. Frailty consensus: A call to action. Journal of the American Medical Directors Association 2013;14(6):392–397. DOI: 10.1016/j.jamda.2013.03.022
  113. 113. Xue QL. The frailty syndrome: Definition and natural history. Clinics in Geriatric Medicine 2011;27(1):1–15. DOI: 10.1016/j.cger.2010.08.009
  114. 114. van Abbema D, van Vuuren A, van den Berkmortel F, van den Akker M, Deckx L, Buntinx F, van Kampen R, Lambooij E, de Boer M, de Vos-Geelen J, Tjan-Heijnen VC. Functional status decline in older patients with breast and colorectal cancer after cancer treatment: A prospective cohort study. Journal of Geriatric Oncology. 2017; pii: S1879-4068(17)30023–1. DOI:10.1016/j.jgo.2017.01.003
  115. 115. Kim DH, Kim CA, Placide S, Lipsitz LA, Marcantonio ER. Preoperative frailty assessment and outcomes at 6 months or later in older adults undergoing cardiac surgical procedures: A systematic review. Annals of Internal Medicine. 2016;165(9):650–660. DOI: 10.7326/M16-0652
  116. 116. CA Polanczyk, E Marcantonio, L Goldman, LEP Rohde, J Orav, CM Mangione, TH Lee. Impact of Age on Perioperative Complications and Length of Stay in Patients Undergoing Noncardiac Surgery. Annals of Internal Medicine 2001;134:637–643
  117. 117. Sharaf-Eldin WE, Abu-Shahba N, Mahmoud M, El-Badri N. The modulatory effects of mesenchymal stem cells on osteoclastogenesis. Stem Cells International. 2016;2016:1908365. DOI: 10.1155/2016/1908365
  118. 118. Takano T, Li YJ, Kukita A, Yamaza T, Ayukawa Y, Moriyama K, Uehara N, Nomiyama H, Koyano K, Kukita T. Mesenchymal stem cells markedly suppress inflammatory bone destruction in rats with adjuvant-induced arthritis. Laboratory Investigation. 2014;94(3):286–296. DOI: 10.1038/labinvest.2013.152
  119. 119. Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: A new trend for cell therapy. Acta Pharmacologica Sinica. 2013;34(6):747–754. DOI: 10.1038/aps.2013.50
  120. 120. Rastegar F, Shenaq D, Huang J, Zhang W, Zhang BQ, He BC, Chen L, Zuo GW, Luo Q, Shi Q, Wagner ER, Huang E, Gao Y, Gao JL, Kim SH, Zhou JZ, Bi Y, Su Y, Zhu G, Luo J, Luo X, Qin J, Reid RR, Luu HH, Haydon RC, Deng ZL, He TC. Mesenchymal stem cells: Molecular characteristics and clinical applications. World Journal of Stem Cells. 2010;2(4):67–80. DOI: 10.4252/wjsc.v2.i4.67

Written By

Bryon A. Tompkins, Ana Marie Landin, Victoria Florea, Makoto Natsumeda, Angela C. Rieger, Wayne Balkan, Ivonne Hernandez Schulman and Joshua M. Hare

Submitted: 22 February 2017 Reviewed: 13 April 2017 Published: 30 August 2017