Main
Main
Main
BACKGROUND: Despite Food and Drug Administration approval of 2 new drugs for idiopathic
pulmonary fibrosis (IPF), curative therapies remain elusive and mortality remains high.
Preclinical and clinical data support the safety of human mesenchymal stem cells as a
potential novel therapy for this fatal condition. The Allogeneic Human Cells (hMSC) in
patients with Idiopathic Pulmonary Fibrosis via Intravenous Delivery (AETHER) trial was
the first study designed to evaluate the safety of a single infusion of bone marrow–derived
mesenchymal stem cells in patients with idiopathic pulmonary fibrosis.
METHODS: Nine patients with mild to moderate IPF were sequentially assigned to 1 of 3
cohorts and dosed with a single IV infusion of 20, 100, or 200 106 human bone marrow–
derived mesenchymal stem cells per infusion from young, unrelated, men. All baseline
patient data were reviewed by a multidisciplinary study team to ensure accurate diagnosis.
The primary end point was the incidence (at week 4 postinfusion) of treatment-emergent
serious adverse events, defined as the composite of death, nonfatal pulmonary embolism,
stroke, hospitalization for worsening dyspnea, and clinically significant laboratory test
abnormalities. Safety was assessed until week 60 and additionally 28 days thereafter.
Secondary efficacy end points were exploratory and measured disease progression.
RESULTS: No treatment-emergent serious adverse events were reported. Two nontreatment-
related deaths occurred because of progression of IPF (disease worsening and/or acute
exacerbation). By 60 weeks postinfusion, there was a 3.0% mean decline in % predicted FVC
and 5.4% mean decline in % predicted diffusing capacity of the lungs for carbon monoxide.
CONCLUSIONS: Data from this trial support the safety of a single infusion of human
mesenchymal stem cells in patients with mild-moderate IPF.
TRIAL REGISTRY: ClinicalTrials.gov; No.: NCT02013700; URL: www.clinicaltrials.gov.
CHEST 2017; 151(5):971-981
KEY WORDS: bone marrow; idiopathic pulmonary fibrosis; mesenchymal stem cells; safety trial
ABBREVIATIONS: 6-MWT = 6-min walk test; DLCO = diffusing MedDRA = Medical Dictionary for Regulatory Activities;
capacity of the lungs for carbon monoxide; FDA = Food and Drug MSCs = mesenchymal stem cells
Administration; hMSCs = human mesenchymal stem cells; AFFILIATIONS: From the Departments of Medicine (Drs Glassberg,
HRCT = high-resolution CT; IPF = idiopathic pulmonary fibrosis; Minkiewicz, Shafazand, Khan, Fishman, and Hare; Mss Simonet,
journal.publications.chestnet.org 971
Idiopathic pulmonary fibrosis (IPF) is a progressive airway inflammation, and restore alveolar fluid balance
and debilitating lung disease characterized by in acute lung injury.10-15
interstitial fibrosis with decreasing lung volumes and
In addition to safety data from preclinical studies, human
pulmonary insufficiency, eventually resulting in death.1
trials have also demonstrated the safety and tolerability of
Because of the insidious onset of symptoms, however,
IV allogeneic mesenchymal stem cells (hMSCs).16-23 A
most patients receive a diagnosis at late stages of the
single-center, open-label phase Ib study assessed the safety
disease after significant fibrosis has occurred. Diagnosis
and tolerability of multiple IV doses of adipose-derived
is established by the pathologic finding of usual
stromal cell-stromal vascular fraction (n ¼ 14) for the
interstitial pneumonia and/or by high-resolution CT
treatment of IPF. Although short-term infusion toxicities
(HRCT).2-4
and long-term ectopic tissue formation were reported,
The natural history of this disease is characterized by no adverse events related to the study treatment were
inexorable progressive decline interspersed with observed.21 In another single-center phase I study,
“exacerbations” or periods of accelerated disease, which patients with IPF received IV placenta-derived hMSCs
are often fatal.1 Although 2 new drugs were recently (n ¼ 8). In this study, most adverse events were mild and
approved by the Food and Drug Administration (FDA) self-limiting and no deaths were reported.19
for patients with IPF, neither is curative.5,6
Our study, the Allogeneic Human Cells in Patients With
In preclinical studies, mesenchymal stem cells (MSCs) Idiopathic Pulmonary Fibrosis via Intravenous Delivery
have shown promise as a potential novel treatment for (AETHER) trial, was the first human trial designed to
lung disease.7-9 Studies of MSCs have shown that they evaluate the safety of bone marrow–derived human
contribute to tissue regeneration, home to sites of lung allogeneic mesenchymal stem cells in patients with mild
injury, contribute to tissue remodeling, decrease chronic to moderate IPF.
Excluded
(n = 2)
1 patient excluded due to clinical instability and acute exacerbation
1 patient excluded due to unforseen inability to attend week for safety visit
Interim
Cohort 1 Cohort 2 Safety Cohort 3
Treated
(n = 3) (n = 3) Analysis (n = 3)
(n = 9)
2 × 107 hMSCs 1 × 108 hMSCs 4 weeks after 2 × 108 hMSCs
Cohort 2 complete
Discontinued
Completed Follow Up (n = 2)
(n = 7) 1 patient died of acute exacerbation and respiratory failure
1 patient died of progression of IPF
Patients in the study received a standard dose of hMSCs rather than Screening of allogeneic donors followed standard transplant practices
weight-based doses made on the basis of results from previous and all allogeneic donors met allogeneic donor eligibility criteria as
studies in patients with cardiovascular disease.16 Detailed study outlined in 21 CFR Part 1271. Donor eligibility screening included
procedures are listed in Table 2. At the initial screening visit, testing for antibodies against HIV-1/2, human T-lymphocyte virus
informed consent was obtained and medical history was reviewed. I/II, hepatitis C virus, hepatitis B core (IgG and IgM), and
Baseline studies included physical examination, routine bloodwork, cytomegalovirus; nucleic acid testing for HIV-1, hepatitis C virus,
urinalysis, ECG, echocardiogram, HRCT, spirometry, DLCO, lung and West Nile virus; and testing for the surface antigen of the
volumes, 6-min walk test (6-MWT), and quality of life hepatitis B virus, Trypanosoma cruzi enzyme-linked immunosorbent
questionnaires. Treatment infusion was considered day 1. Adverse assay, and rapid plasma reagin.
events were reviewed at day 1, week 1, and at all visits thereafter.
The primary end point was assessed starting at week 4 until week For each donor, a total of 60 mL of bone marrow was aspirated from
60 and additionally 28 days thereafter. Secondary efficacy end the posterior iliac crest. The mononuclear cell fraction was isolated
points were measured at baseline and every 12 weeks until week 60. using a density gradient with lymphocyte separation media (specific
gravity, 1.077). Low-density cells were collected and washed with
Plasma-Lyte A containing 1% human serum albumin. Washed cells
Isolation of hMSCs
were sampled and viable cell numbers determined. The bone marrow
Because of the potential for pregnancy-induced antibodies to men’s mononuclear cells were seeded into 225 cm2 tissue culture flasks in
antigens, hMSCs were obtained only from men. Two men aged 24 alpha Minimal Essential Medium containing 20% fetal bovine serum.
and 25 years underwent bone marrow aspiration. Donors were After 14 days of culture, passage zero (P0) cells were harvested by
neither related nor human leukocyte antigen–matched to recipients. trypsin treatment and expanded into 60 individual flasks. These
flasks were incubated for a further 7 to 10 days before harvesting of
MSCs by trypsin treatment (P1 cells). All procedures used in the
TABLE 1 ] Dosing Schedule of AETHER Participants preparation of the investigational product followed protocols
Cohort Subject ID Dosing Date previously published by the sponsor (Joshua M. Hare).26
journal.publications.chestnet.org 973
974 Original Research
QOL questionnaires x x x x x x x
151#5 CHEST MAY 2017
CBC ¼ complete blood count; Chem7 ¼ sodium, potassium, chloride, uric acid, glucose, blood urea nitrogen, creatinine; DLCO ¼ diffusing capacity of the lungs for carbon monoxide; HRCT ¼ high-resolution CT;
LFTs ¼ liver function tests (alanine transaminase, alkaline phosphatase, aspartate transaminase, bilirubin, albumin, total protein, gamma glutamyl transpeptidase); PT/INR ¼ prothrombin time (PT) along with its
derived measures of prothrombin ratio and international normalized ratio (INR); QOL ¼ quality of life. See Table 1 legend for expansion of other abbreviations.
]
Statistical Analysis for safety analyses and projected enrollment rates. A 2-tailed Student
No formal statistical justification was performed to determine sample t test was used to evaluate differences in secondary end points from
size. Cohort size was determined on the basis of expected requirements baseline. A P value < .05 was considered statistically significant.
TLC ¼ total lung capacity. See Table 1 and 2 legends for expansion of other abbreviations.
journal.publications.chestnet.org 975
TABLE 4 ] Modified Intention-to-Treat Set
Cohort 1 Cohort 2 Cohort 3
Subject Status 2 107 hMSCs/Infusion 1 108 hMSCs/Infusion 2 108 hMSCs/Infusion Total, No. (%)
Started, No. (%) 3 (100) 3 (100) 3 (100) 9 (100)
Completed, No. (%) 3 (100) 3 (100) 1 (33.3) 7 (78)
Not completed, No. (%) 0 (0) 0 (0) 2 (66.7) 2 (22)
Data are No. of participants (%). Modified intention-to-treat set ¼ participants treated with hMSCs, regardless of study completion. See Table 1 legend for
expansion of abbreviations.
failure [patient 007]) occurred in cohort 3. Patient 007 bone marrow–derived hMSCs in patients with IPF. All
experienced an acute exacerbation and subsequent study objectives followed the recommendations of the
respiratory failure resulting in death at 10 weeks and FDA and the American Thoracic Society.1
3 days postinfusion. Patient 010 experienced progression
AETHER trial met its primary end point of safety,
of IPF (defined as disease worsening according to
showing that the administration of hMSCs is safe in
MedDRA), resulting in death at 29 weeks and 6 days
patients with IPF up to 2 108 cells/infusion. The
postinfusion. None of these serious adverse events was
intervention was well-tolerated in all patients and there
determined to be treatment-related.
were no treatment-emergent serious adverse events
Table 7 shows the progression of lung function reported. A majority of patients (78%) experienced
parameters over the course of the study. Data for treatment unrelated adverse events including, but not
participants 007 and 010 are not available beyond limited to, bronchitis, common cold, and sinusitis
week 4. Figure 2 shows progression of select respiratory (Table 7), which one might expect given the long
parameters up to 60 weeks postinfusion. Data combined duration of the study and the characteristics of the
for all cohorts (n ¼ 7) demonstrated a mean absolute population being studied.
decline in % predicted FVC of 3.0% and a 5.4% decline
There were 2 events of nonstudy-related death from
in % predicted DLCO. Overall, 6-MWT improved by
disease progression and acute exacerbation of IPF. As
36 weeks postinfusion (þ1% improvement from
expected in IPF, those enrolled in the AETHER trial
baseline), and declined to -4.4% from baseline by week
had variable rates of disease progression. Although
60 (Fig 2). These data are considered exploratory
the clinical course of this disease is unpredictable, the
because the study was not powered for efficacy analyses
natural history is typically one of steady decline of lung
and lacked a placebo-control arm.
function punctuated by acute exacerbations. Baseline
lung function parameters suggest that patients in cohort
Discussion 3 had significantly more advanced disease than patients
AETHER was the first clinical trial conducted over in cohorts 1 and 2. The 2 subjects who died were both
60 weeks to support the safety of a single IV infusion of in cohort 3 and had the lowest baseline FVC values and
TABLE 5 ] Respiratory and Hemodynamic Parameters at Baseline and After hMSC Infusion
2 h Before Infusion (Baseline) Start/During Infusion 2 h After Infusion
HR MAP SpO2 HR MAP SpO2 HR MAP
Subject ID (beats/min) (mm Hg) (%) (beats/min) (mm Hg) (%) (beats/min) (mm Hg) SpO2 (%)
001 69 120/73 95 76 121/70 96 79 115/74 96
002 67 116/71 97 75 108/63 95 74 115/60 97
003 65 158/68 99 63 150/49 99 68 134/55 98
004 54 132/61 98 56 120/68 100 62 129/72 99
005 54 153/83 97 58 162/77 98 56 154/76 94
006 70 152/72 99 65 148/82 100 67 130/80 99
007 61 127/63 94 58 137/58 94 58 140/55 95
010 61 158/76 97 60 165/74 98 66 155/74 96
011 56 139/78 98 57 126/71 98 61 97/49 95
HR ¼ heart rate; MAP ¼ mean arterial pressure; SpO2 ¼ peripheral capillary oxygen saturation.
6-MWT distances. One of the 2 patients also had the for IPF, we noted substantial inter- and intra-subject
lowest baseline DLCO overall. In light of a presumed variability in the direction and magnitude of change
1-year mortality risk of approximately 20% in patients of lung function parameters.19,21 The average absolute
with moderately advanced disease,27 the observed decline in % predicted FVC and DLCO by the end of this
death of 2 of 9 subjects was not unexpected. Prior trials study were below the previously mentioned thresholds
using intravenously delivered MSCs, primarily in the for IPF disease progression. However, this small phase I
cardiovascular literature, have not shown increased study was designed to evaluate safety and was not
mortality related to MSC treatment.16,24 Additionally, powered to detect significant changes in lung function.
although concerns that MSCs may contribute to lung Although encouraging, these data are preliminary and
fibrosis have been raised in the past,28-31 there are no should not be interpreted as proof of efficacy of hMSCs
preclinical or human studies that have demonstrated in IPF disease progression.
this relationship.
Results from the ASCEND (A Randomized, Double-
Secondary end points were exploratory and related to Blind, Placebo Controlled, Phase 3 Study of the Efficacy
disease progression. On the basis of Consensus and Safety of Pirfenidone in Patients With Idiopathic
Statement guidelines for patients with IPF, a decline in Pulmonary Fibrosis) trial of pirfenidone for the treatment
the absolute FVC $10% or $15% in the absolute DLCO of IPF suggest that the use of supplemental oxygen and
over 3 to 6 months represents progression of disease.1,32 FVC at baseline significantly correlates with rate of
As with other phase 1 trials of cell-based therapies disease progression.5 In the ASCEND trial, subjects who
journal.publications.chestnet.org 977
TABLE 7 ] Progression of Lung Function Parameters
Subject ID Baseline Week 12 Week 24 Week 36 Week 48 Week 60
TLC, L, Mean
001 3.60 3.21 3.90 3.12 3.16 3.12
002 4.08 4.59 4.04 4.63 4.76 4.80
003 4.78 5.08 4.07 4.39 4.39 3.34
004 5.79 4.39 4.97 4.50 5.81 5.62
005 3.85 3.66 3.53 4.45 4.17 4.39
006 3.54 3.47 3.31 3.62 4.29 4.52
007 3.73 N/A N/A N/A N/A N/A
010 4.14 N/A N/A N/A N/A N/A
011 3.91 4.09 4.25 4.18 4.67 4.85
FVC, L, Mean
001 2.48 2.14 2.56 2.20 2.26 1.95
002 3.38 3.64 2.98 3.39 3.34 3.34
003 2.91 2.85 2.92 2.65 2.69 2.83
004 3.76 3.50 3.67 3.62 3.75 3.61
005 2.18 2.20 2.17 2.05 2.07 2.03
006 2.58 2.62 2.4 2.54 2.42 2.48
007 2.25 N/A N/A N/A N/A N/A
010 2.51 N/A N/A N/A N/A N/A
011 2.70 2.76 2.50 2.47 2.75 2.94
N/A ¼ not applicable. See Table 2 legend for expansion of other abbreviations.
required supplemental oxygen experienced a higher rate progression. In line with these findings, 3 subjects in the
of disease progression, whereas those with a higher AETHER trial used supplemental oxygen at baseline and
baseline FVC experienced a lower rate of disease 2 died during the course of the study.
% Predicted DLCO
80
70
70
60 60
50 50
40 40
30 30
20 20
10 10
0 0
Baseline 12 24 36 48 60 Baseline 12 24 36 48 60
Week Week
C
550
6-MWT distance (m)
500
450
400
350
300
Baseline 12 24 36 48 60
Week
Figure 2 – Secondary efficacy outcomes during the 60-week study period. A, Changes in mean % predicted FVC for overall study period. B, Changes in
% predicted diffusing capacity of the lungs for carbon monoxide (DLCO). C, Changes in 6-min walk test (6-MWT) distance. N ¼ 7 for all data.
Results from the ASCEND trial also suggest that single IV administration of hMSCs up to 2 108 cells/
baseline body weight is correlated with a decreased rate infusion for the treatment of IPF.19,21 Limitations of
of disease progression in treated subjects weighing up to the current study include its small sample size, lack of
approximately 85 kg (187.5 lb). For patients with body randomization, and lack of a placebo arm. Current
weights greater than 85 kg, disease progression was safety findings support the role of more extensive
found to be similar in both the treatment and placebo studies of the safety and efficacy of hMSCs in the
groups. The average body weight of subjects in the treatment of IPF. Before larger randomized placebo-
AETHER study was 86.36 (8.34) kg; only 4 of 9 controlled studies are done to evaluate efficacy, further
subjects weighed less than 85 kg at baseline. Both safety studies should be conducted with larger numbers
patients who died during the study weighed more than of patients as well as patients with more advanced IPF.
85 kg. AETHER did not analyze disease progression in One challenge will be to establish the optimal number
relation to body weight; however, future trials could aim of infusions and the appropriate dosing interval.
to enroll subjects with similar baseline body mass index Another challenge will be to identify early stage
to ensure a uniform weight-based dose of hMSCs. An patients most likely to benefit from the intervention.
alternative approach could be to dose patients by body Ultimately, well-designed and meticulously conducted
weight as in previous hMSC trials.16-20,23 phase II/III clinical trials of hMSCs for the treatment of
IPF will be required to evaluate their efficacy as a
In agreement with other studies of IPF patients treated potential therapeutic modality for this devastating
with cell therapies, the data support the safety of a disease.
journal.publications.chestnet.org 979
Acknowledgments pirfenidone in patients with idiopathic 18. Hare JM, Fishman JE, Gerstenblith G,
pulmonary fibrosis. N Engl J Med. et al. Comparison of allogeneic
Author contributions: Study conception: 2014;370(22):2083-2092. vs autologous bone marrow-derived
M. K. G., R. L. T., and J. M. H. Study design mesenchymal stem cells delivered by
6. Richeldi L, Cottin V, du Bois RM, et al.
and coordination: D. D., A. K., M. V. P., S. S., transendocardial injection in patients
Nintedanib in patients with idiopathic
V. F. L., L. H. L., G. D. R., J. F., E. S. S., A. M., with ischemic cardiomyopathy: the
pulmonary fibrosis: Combined
and Y. N. M. Data analysis and reporting: evidence from the TOMORROW and POSEIDON randomized trial. JAMA.
M. K. G., R. L. T., and J. M. Manuscript INPULSIS((R)) trials. Res Med. 2016;113: 2012;308(22):2369-2379.
preparation: M. K. G., J. M., R. L. T., and 74-79. 19. Chambers DC, Enever D, Ilic N, et al.
G. A. R. Critical revision of manuscript for A phase 1b study of placenta-derived
important intellectual content: all authors 7. Moodley Y, Atienza D, Manuelpillai U,
et al. Human umbilical cord mesenchymal mesenchymal stromal cells in patients
read and approved final manuscript. M. K. G. with idiopathic pulmonary fibrosis.
stem cells reduce fibrosis of bleomycin-
had full access to all of the data and takes Respirology. 2014;19(7):1013-1018.
induced lung injury. Am J Pathol. 2009;
responsibility for the integrity of the data and 175(1):303-313. 20. Le Blanc K, Frassoni F, Ball L, et al.
the accuracy of the data analysis. Mesenchymal stem cells for treatment of
8. Rojas M, Xu J, Woods CR, et al. Bone
Financial/nonfinancial disclosures: The marrow-derived mesenchymal stem cells steroid-resistant, severe, acute graft-
authors have reported to CHEST the in repair of the injured lung. Am J Resp versus-host disease: a phase II study.
following: M. K. G. is participating in a role Cell Mol Bio. 2005;33(2):145-152. Lancet. 2008;371(9624):1579-1586.
of an investigator at industry-sponsored 21. Tzouvelekis A, Paspaliaris V, Koliakos G,
9. Tashiro J, Elliot SJ, Gerth DJ, et al.
clinical trials for Genentech, Boehringer et al. A prospective, non-randomized, no
Therapeutic benefits of young, but not old,
Ingelheim, Bayer, and Biogen and serves placebo-controlled, phase Ib clinical trial
adipose-derived mesenchymal stem cells
as a consultant expert for Genentech and to study the safety of the adipose derived
in a chronic mouse model of bleomycin-
stromal cells-stromal vascular fraction in
Boehringer Ingelheim. L. H. L. served as induced pulmonary fibrosis. Transl Res.
an investigator for Boehringer Ingelheim, idiopathic pulmonary fibrosis. J Transl
2015;166(6):554-567.
Genentech, Global Blood Therapeutics, Med. 2013;11:171.
10. Ortiz LA, Gambelli F, McBride C, et al.
Celgene, Roche, and Veracyte and has 22. Weiss DJ, Casaburi R, Flannery R,
Mesenchymal stem cell engraftment in
served on advisory boards for Genentech LeRoux-Williams M, Tashkin DP.
lung is enhanced in response to bleomycin
and Boehringer Ingelheim. Y. N. M. has A placebo-controlled, randomized trial of
exposure and ameliorates its fibrotic
served on the speakers bureau for Boehringer mesenchymal stem cells in COPD. Chest.
effects. Proc Natl Acad Sci U S A. 2003;
Ingelheim. None declared (J. M., R. L. T., 2013;143(6):1590-1598.
100(14):8407-8411.
E. S. S., G. A. R., D. D.; S. S., A. K., M. V. P., 23. Wilson JG, Liu KD, Zhuo H, et al.
11. Ishizawa K, Kubo H, Yamada M, et al.
V. F. L., G. D. R., J. F., A. M., J. M. H.). Mesenchymal stem (stromal) cells for
Bone marrow-derived cells contribute to
treatment of ARDS: a phase 1 clinical trial.
Role of sponsor: The sponsor had no role in lung regeneration after elastase-induced
Lancet. 2015;3(1):24-32.
the design of the study, the collection and pulmonary emphysema. FEBS Lett. 2004;
analysis of the data, or the preparation of the 556(1-3):249-252. 24. Heldman AW, DiFede DL, Fishman JE,
manuscript. et al. Transendocardial mesenchymal stem
12. Spees JL, Pociask DA, Sullivan DE, et al.
cells and mononuclear bone marrow
Engraftment of bone marrow progenitor
Other contributions: We thank the Lester cells for ischemic cardiomyopathy: the
cells in a rat model of asbestos-induced
and Sue Smith Foundation for funding TAC-HFT randomized trial. JAMA.
pulmonary fibrosis. Am J Resp Crit Care
the AETHER trial and ongoing support; 2014;311(1):62-73.
Med. 2007;176(4):385-394.
Kevin Anstrom, PhD, for reviewing the 25. Golpanian S, DiFede DL, Pujol MV, et al.
data; Jose Da Silva, PhD, for providing input 13. Spees JL, Whitney MJ, Sullivan DE, et al.
Rationale and design of the allogeneiC
for the conduction of the trial; and study Bone marrow progenitor cells contribute
human mesenchymal stem cells (hMSC)
to repair and remodeling of the lung and
coordinators Julio Sierra, MD, and Cindy in patients with aging fRAilTy via
heart in a rat model of progressive
Delgado, MA, who coordinated subject visits. intravenoUS delivery (CRATUS) study: a
pulmonary hypertension. FASEB J. 2008;
phase I/II, randomized, blinded and
22(4):1226-1236.
placebo controlled trial to evaluate the
References 14. Bonfield TL, Koloze M, Lennon DP, safety and potential efficacy of allogeneic
1. Raghu G, Collard HR, Egan JJ, et al. An Zuchowski B, Yang SE, Caplan AI. human mesenchymal stem cell infusion in
official ATS/ERS/JRS/ALAT statement: Human mesenchymal stem cells suppress patients with aging frailty. Oncotarget.
idiopathic pulmonary fibrosis: evidence- chronic airway inflammation in the 2016;7(11):11899-11912.
based guidelines for diagnosis and murine ovalbumin asthma model. Am J 26. Trachtenberg B, Velazquez DL,
management. Am J Resp Crit Care Med. Physiol. 2010;299(6):L760-L770. Williams AR, et al. Rationale and design
183(6):788-824. 15. Lee JW, Fang X, Gupta N, Serikov V, of the Transendocardial Injection of
2. Travis WD, Hunninghake G, King TE Jr., Matthay MA. Allogeneic human Autologous Human Cells (bone marrow
et al. Idiopathic nonspecific interstitial mesenchymal stem cells for treatment or mesenchymal) in Chronic Ischemic
pneumonia: report of an American of E. coli endotoxin-induced acute lung Left Ventricular Dysfunction and Heart
Thoracic Society project. Am J Resp Crit injury in the ex vivo perfused human lung. Failure Secondary to Myocardial
Care Med. 2008;177(12):1338-1347. Proc Natl Acad Sci U S A. 2009;106(38): Infarction (TAC-HFT) trial: a randomized,
16357-16362. double-blind, placebo-controlled study of
3. Nishimura K, Kitaichi M, Izumi T, safety and efficacy. Am Heart J. 2011;
Nagai S, Kanaoka M, Itoh H. Usual 16. Hare JM, Traverse JH, Henry TD, et al.
A randomized, double-blind, placebo- 161(3):487-493.
interstitial pneumonia: histologic
correlation with high-resolution CT. controlled, dose-escalation study of 27. Ley B, Bradford WZ, Weycker D,
Radiology. 1992;182(2):337-342. intravenous adult human mesenchymal Vittinghoff E, du Bois RM, Collard HR.
stem cells (prochymal) after acute Unified baseline and longitudinal
4. Johkoh T, Muller NL, Cartier Y, et al. myocardial infarction. J Am Coll Cardiol. mortality prediction in idiopathic
Idiopathic interstitial pneumonias: 2009;54(24):2277-2286. pulmonary fibrosis. Eur Resp J. 2015;45(5):
diagnostic accuracy of thin-section CT 1374-1381.
17. Liang J, Zhang H, Hua B, et al. Allogenic
in 129 patients. Radiology. 1999;211(2):
mesenchymal stem cells transplantation in 28. Lama VN, Phan SH. The extrapulmonary
555-560.
refractory systemic lupus erythematosus: a origin of fibroblasts: stem/progenitor cells
5. King TE Jr., Bradford WZ, Castro- pilot clinical study. Ann Rheum Dis. 2010; and beyond. Proc Am Thorac Soc. 2006;
Bernardini S, et al. A phase 3 trial of 69(8):1423-1429. 3(4):373-376.
journal.publications.chestnet.org 981