Cellular Therapies in Critical Illness: Current Evidence and Future Directions
Abstract
Background: Cellular therapies, particularly mesenchymal stem cells (MSCs), have emerged as promising therapeutic modalities for critical illness syndromes including acute respiratory distress syndrome (ARDS) and septic shock. Despite extensive preclinical evidence demonstrating immunomodulatory and regenerative properties, clinical translation has faced significant challenges.
Objective: To provide a comprehensive review of current evidence for cellular therapies in critical care, focusing on MSC applications in ARDS and septic shock, and evaluate recent clinical trial developments through 2025.
Methods: We conducted a systematic review of published literature, ongoing clinical trials, and regulatory developments in cellular therapy for critical illness through September 2025.
Results: While preclinical studies consistently demonstrate MSC efficacy in reducing inflammation and promoting tissue repair, clinical trials have shown mixed results. Recent phase II/III trials have refined patient selection criteria, dosing strategies, and timing of administration. Novel cell products including MSC-derived exosomes and genetically modified cells show promise in early-phase studies.
Conclusions: Cellular therapies remain investigational in critical care settings. Success in future trials will likely depend on precision medicine approaches, standardized manufacturing protocols, and biomarker-guided patient selection.
Keywords: Mesenchymal stem cells, ARDS, septic shock, cellular therapy, critical care, immunomodulation
Introduction
Critical illness syndromes such as acute respiratory distress syndrome (ARDS) and septic shock represent major causes of morbidity and mortality in intensive care units worldwide. Despite advances in supportive care, mortality rates remain substantial, with ARDS mortality ranging from 30-45% and septic shock approaching 40-50%¹,². The pathophysiology of these conditions involves complex inflammatory cascades, endothelial dysfunction, and tissue injury that often prove refractory to conventional therapeutic approaches.
Cellular therapies, particularly mesenchymal stem cells (MSCs), have garnered significant attention as potential therapeutic interventions for critical illness. MSCs possess unique properties including immunomodulation, anti-inflammatory effects, antimicrobial activity, and tissue repair capabilities³. These characteristics make them theoretically attractive for conditions characterized by dysregulated inflammation and tissue injury.
This review synthesizes current evidence for cellular therapies in critical care, with emphasis on MSC applications in ARDS and septic shock, while highlighting recent clinical trial developments and future directions in this rapidly evolving field.
Mesenchymal Stem Cells: Biological Rationale
Cellular Characteristics and Mechanisms of Action
MSCs are multipotent stromal cells that can be isolated from various tissues including bone marrow, adipose tissue, umbilical cord, and placenta⁴. The International Society for Cellular and Gene Therapies (ISCT) has established minimal criteria for MSC identification: plastic adherence, specific surface antigen expression (CD105+, CD73+, CD90+, CD45-, CD34-, CD14-), and tri-lineage differentiation potential⁵.
Pearl: The therapeutic effects of MSCs are primarily mediated through paracrine mechanisms rather than direct cellular engraftment and differentiation. This paradigm shift has important implications for dosing and delivery strategies.
The proposed mechanisms of MSC action in critical illness include:
- Immunomodulation: MSCs secrete anti-inflammatory cytokines (IL-10, TGF-β) while suppressing pro-inflammatory mediators (TNF-α, IL-1β, IL-6)⁶
- Antimicrobial effects: Production of antimicrobial peptides including LL-37, β-defensin-2, and indoleamine 2,3-dioxygenase⁷
- Endothelial protection: Secretion of angiogenic factors (VEGF, angiopoietin-1) and barrier-protective mediators⁸
- Tissue repair: Release of growth factors promoting epithelial and endothelial repair⁹
MSC Sources and Considerations
Different MSC sources exhibit varying characteristics relevant to critical care applications:
- Bone marrow-derived MSCs (BM-MSCs): Most extensively studied, gold standard for comparison
- Adipose-derived MSCs (AD-MSCs): More abundant, potentially superior anti-inflammatory properties¹⁰
- Umbilical cord MSCs (UC-MSCs): Younger cells with potentially enhanced regenerative capacity, no ethical concerns¹¹
- Placental MSCs: High proliferative capacity, strong immunosuppressive properties¹²
Hack: Allogeneic MSCs may be preferable to autologous cells in critically ill patients due to the immunocompromised state and cellular dysfunction associated with critical illness, which may impair autologous MSC function.
MSCs in Acute Respiratory Distress Syndrome
Pathophysiology and Therapeutic Targets
ARDS is characterized by acute onset of bilateral pulmonary infiltrates, impaired oxygenation, and increased alveolar-capillary permeability not fully explained by cardiac failure¹³. The pathophysiology involves:
- Inflammatory phase: Neutrophil infiltration, pro-inflammatory cytokine release
- Proliferative phase: Type II pneumocyte proliferation, fibroblast activation
- Fibrotic phase: Collagen deposition, architectural distortion
MSCs theoretically address multiple pathophysiological targets in ARDS through their anti-inflammatory, antimicrobial, and tissue repair properties.
Preclinical Evidence
Extensive preclinical studies have demonstrated MSC efficacy in animal models of ARDS. Key findings include:
- Reduced pulmonary inflammation and neutrophil infiltration¹⁴
- Improved alveolar-capillary barrier function¹⁵
- Enhanced bacterial clearance in pneumonia models¹⁶
- Reduced pulmonary fibrosis in later-phase injury¹⁷
Oyster: Despite consistent preclinical efficacy, the translation to human clinical trials has been challenging, highlighting the limitations of animal models in recapitulating human ARDS complexity.
Clinical Trial Evidence
Early Phase Studies
STAIR (START Trial - Phase I): Wilson et al. conducted the first-in-human safety study of bone marrow-derived MSCs in ARDS patients¹⁸. Nine patients received escalating doses (1, 5, and 10 million cells/kg) with no significant safety concerns identified.
STAIR Phase II: Subsequently, Matthay et al. published results from a randomized, double-blind, placebo-controlled phase II trial of 60 patients with moderate-to-severe ARDS¹⁹. Patients received either placebo or 10 million cells/kg of bone marrow-derived MSCs. While the treatment was safe, there were no significant differences in clinical outcomes.
Recent Clinical Trials (2023-2025)
MUST-ARDS Trial (2024): This multinational phase II trial randomized 120 patients with severe ARDS to receive either UC-MSCs (2 million cells/kg) or placebo within 48 hours of ARDS onset²⁰. Primary endpoint was ventilator-free days at 28 days. While the study met safety endpoints, efficacy outcomes showed only modest improvements in oxygenation indices without significant clinical benefit.
CELLIST-ARDS (2025): Currently ongoing phase III trial investigating adipose-derived MSCs in COVID-19-associated ARDS²¹. This study employs biomarker-guided patient selection using baseline inflammatory markers (IL-6, IL-8) to identify potential responders.
Challenges and Lessons Learned
Several factors may explain the disconnect between preclinical promise and clinical results:
- Heterogeneity of ARDS: Clinical ARDS encompasses diverse etiologies and pathophysiological phenotypes²²
- Timing of intervention: Optimal window for MSC administration remains unclear
- Cell dose and viability: Standardization of cell products and dosing strategies
- Patient selection: Need for biomarker-guided approaches to identify responders
Pearl: Future ARDS trials should consider phenotype-specific approaches, potentially targeting hyperinflammatory phenotypes identified through biomarker profiling or machine learning algorithms.
MSCs in Septic Shock
Pathophysiology and Rationale
Septic shock represents the most severe form of sepsis, characterized by persistent hypotension requiring vasopressors and elevated lactate despite adequate volume resuscitation²³. The pathophysiology involves:
- Immune dysregulation: Initial hyperinflammation followed by immunosuppression
- Endothelial dysfunction: Increased vascular permeability, microvascular dysfunction
- Organ dysfunction: Multi-organ failure through various mechanisms
MSCs theoretically address these pathophysiological derangements through immunomodulatory, antimicrobial, and endothelial-protective effects.
Preclinical Evidence
Animal models of sepsis have consistently demonstrated MSC benefits:
- Improved survival in cecal ligation and puncture models²⁴
- Reduced organ dysfunction scores²⁵
- Enhanced bacterial clearance²⁶
- Modulation of immune cell function²⁷
Clinical Evidence
Early Studies
Phase I Safety Studies: Multiple small safety studies have demonstrated the feasibility and safety of MSC administration in septic patients²⁸,²⁹. These studies established dosing ranges (1-10 million cells/kg) and identified no major safety signals.
Recent Clinical Trials
SEPCELL Trial (2024): This randomized controlled trial of 90 patients with septic shock compared bone marrow-derived MSCs (5 million cells/kg) to standard care³⁰. While safe, the study showed no significant improvement in 28-day mortality (primary endpoint). However, post-hoc analyses suggested potential benefits in patients with moderate illness severity (APACHE II 15-25).
MESOSEP-2025: Currently recruiting phase III trial investigating umbilical cord-derived MSCs in early septic shock³¹. This study employs a precision medicine approach using baseline biomarkers (IL-6/IL-10 ratio, HLA-DR expression) to identify patients most likely to benefit.
Novel Approaches in Sepsis
Biomarker-Guided Therapy
Recent studies suggest that patient stratification based on immune status may improve MSC efficacy:
- Hyperinflammatory phenotype: Elevated IL-6, IL-8, TNF-α
- Immunosuppressed phenotype: Reduced HLA-DR expression, lymphopenia
Hack: Consider obtaining baseline immune biomarkers (HLA-DR expression on monocytes, IL-6 levels) before MSC administration in septic patients, as these may predict treatment response.
Combination Therapies
Emerging strategies combine MSCs with other interventions:
- MSCs plus plasma exchange³²
- MSCs plus extracorporeal membrane oxygenation³³
- MSCs plus targeted immunomodulators³⁴
Clinical Trial Updates and Future Directions
Manufacturing and Regulatory Considerations
The cellular therapy field faces significant manufacturing and regulatory challenges:
Good Manufacturing Practice (GMP) Standards
Current Status: Most clinical trials now require GMP-manufactured MSC products, ensuring:
- Standardized isolation and expansion protocols
- Rigorous quality control testing
- Consistent potency assays
- Sterility and safety testing
Pearl: GMP manufacturing has improved product consistency but significantly increased costs, limiting accessibility and requiring careful cost-effectiveness analyses.
Regulatory Landscape
FDA Guidance (2024): Updated guidance documents emphasize:
- Robust preclinical data requirements
- Standardized potency assays
- Long-term safety follow-up
- Risk-based manufacturing approaches³⁵
EMA Perspectives: European regulators have taken a more flexible approach, allowing hospital-based manufacturing under specific circumstances³⁶.
Novel Cell Products and Approaches
MSC-Derived Exosomes
Rationale: Exosomes may provide MSC benefits without cellular administration challenges:
- Reduced immunogenicity
- Easier storage and distribution
- Standardized dosing
- Reduced safety concerns
Clinical Evidence: Early-phase trials in ARDS and sepsis show promising safety profiles³⁷,³⁸.
Oyster: While exosomes represent an elegant solution to cellular therapy challenges, their manufacturing complexity and regulatory pathway remain unclear.
Genetically Modified MSCs
Approaches Under Investigation:
- Enhanced anti-inflammatory cytokine production
- Improved tissue homing capabilities
- Resistance to hostile microenvironments
- Combined therapeutic protein delivery³⁹
Induced Pluripotent Stem Cell-Derived MSCs (iPSC-MSCs)
Advantages:
- Unlimited cell source
- Standardized characteristics
- Potential for genetic modifications
- Reduced donor variability⁴⁰
Biomarker-Guided Approaches
Predictive Biomarkers
Inflammatory Markers:
- IL-6, IL-8 levels predict hyperinflammatory phenotype
- CRP, procalcitonin for infection severity
- Complement activation markers⁴¹
Immune Function Markers:
- HLA-DR expression on monocytes
- Lymphocyte counts and function
- Cytokine production capacity⁴²
Tissue Injury Markers:
- Pulmonary: SP-D, CC16, RAGE for ARDS
- Cardiac: Troponins, NT-proBNP
- Renal: Neutrophil gelatinase-associated lipocalin⁴³
Hack: Develop institutional protocols for rapid biomarker assessment to enable precision cellular therapy approaches. Point-of-care testing for key markers (IL-6, HLA-DR) may facilitate timely patient selection.
Pharmacokinetic and Pharmacodynamic Considerations
Cell Tracking: Advanced imaging techniques allow assessment of:
- Cellular biodistribution
- Pulmonary retention
- Duration of effect⁴⁴
Dose-Response Relationships: Recent studies suggest:
- Higher doses may not always be better
- Multiple dosing strategies under investigation
- Patient-specific dosing based on severity⁴⁵
Safety Considerations and Risk Mitigation
Known Safety Signals
Immediate Risks:
- Infusion-related reactions (rare, <5%)
- Pulmonary embolism from cellular aggregates
- Hemodynamic instability⁴⁶
Long-term Concerns:
- Theoretical malignancy risk (no confirmed cases)
- Immune sensitization with repeated dosing
- Unknown effects on tissue repair processes⁴⁷
Risk Mitigation Strategies
Manufacturing Controls:
- Cell size filtration to prevent aggregates
- Viability testing before administration
- Sterility and endotoxin testing
Clinical Protocols:
- Slow infusion rates (typically over 30-60 minutes)
- Hemodynamic monitoring during administration
- Immediate access to resuscitation equipment
Pearl: Establish standardized infusion protocols including pre-medication regimens (antihistamines, corticosteroids) for patients at high risk of infusion reactions.
Long-term Safety Monitoring
Current Recommendations:
- Minimum 2-year safety follow-up
- Annual malignancy screening
- Immune function monitoring
- Registry-based surveillance⁴⁸
Economic Considerations and Healthcare Impact
Cost-Effectiveness Analysis
Manufacturing Costs:
- GMP-compliant MSC production: $15,000-50,000 per dose
- Quality control and testing: Additional $5,000-10,000
- Storage and logistics: $2,000-5,000⁴⁹
Potential Savings:
- Reduced ICU length of stay
- Decreased mechanical ventilation duration
- Lower long-term disability costs
- Reduced healthcare utilization⁵⁰
Current Status: Most economic analyses suggest cellular therapies are not cost-effective at current pricing, but may become viable with:
- Improved manufacturing efficiency
- Better patient selection
- Demonstrated clinical benefits⁵¹
Healthcare System Implications
Infrastructure Requirements:
- Specialized storage facilities (-80°C freezers)
- Trained personnel for administration
- Quality assurance programs
- Regulatory compliance capabilities
Hack: Consider regional hub-and-spoke models for cellular therapy delivery to optimize resource utilization and maintain expertise while serving broader patient populations.
Future Directions and Research Priorities
Clinical Trial Design Improvements
Adaptive Trial Designs:
- Biomarker-guided patient selection
- Dose escalation based on response
- Futility stopping rules
- Platform trials testing multiple products⁵²
Surrogate Endpoints:
- Early biomarker changes
- Physiological improvements
- Multi-organ dysfunction scores
- Patient-reported outcome measures⁵³
Precision Medicine Approaches
Genomic Stratification:
- Host genetic variants affecting response
- MSC donor genetics impact
- Pharmacogenomic considerations⁵⁴
Machine Learning Applications:
- Predictive algorithms for patient selection
- Optimal timing prediction
- Treatment response modeling⁵⁵
Combination and Sequential Therapies
Synergistic Approaches:
- MSCs plus targeted immunomodulators
- Sequential cellular and pharmacological interventions
- Combination with device-based therapies⁵⁶
Novel Delivery Methods
Targeted Delivery:
- Inhaled MSC administration for ARDS
- Direct organ perfusion techniques
- Biomaterial-assisted cell delivery⁵⁷
Practical Recommendations for Clinicians
Current Clinical Practice
Evidence-Based Recommendations:
- MSCs should be considered investigational only
- Participation in clinical trials is encouraged when available
- Compassionate use should follow strict protocols
- Comprehensive safety monitoring is mandatory
Patient Counseling Points:
- Experimental nature of therapy
- Limited efficacy data
- Potential risks and benefits
- Alternative treatment options⁵⁸
Future Clinical Integration
Preparation for Clinical Adoption:
- Develop institutional protocols for cellular therapy
- Establish infrastructure for safe administration
- Train staff in specialized procedures
- Create quality assurance programs
Biomarker Integration:
- Implement rapid biomarker testing capabilities
- Develop patient selection algorithms
- Establish treatment response monitoring protocols
Pearl: Begin developing institutional capabilities for cellular therapy now, even before widespread clinical adoption, to ensure readiness when these therapies become standard of care.
Conclusions
Cellular therapies, particularly mesenchymal stem cells, represent a promising but still investigational approach for critical illness syndromes including ARDS and septic shock. While extensive preclinical evidence supports their therapeutic potential, clinical translation has proven challenging, with most trials showing safety but limited efficacy signals.
The field is evolving toward precision medicine approaches utilizing biomarker-guided patient selection, standardized manufacturing protocols, and novel cell products. Success in future clinical applications will likely depend on identifying the right patients, at the right time, with the right cellular product.
Key priorities for advancing the field include:
- Development of predictive biomarkers for treatment response
- Standardization of manufacturing and quality control processes
- Implementation of adaptive clinical trial designs
- Economic sustainability through improved cost-effectiveness
For clinicians in critical care, maintaining awareness of ongoing developments while recognizing the current investigational status remains appropriate. Participation in well-designed clinical trials represents the best current approach for offering these therapies to patients while advancing scientific knowledge.
As we move forward, the integration of cellular therapies into critical care practice will require careful consideration of efficacy, safety, and economic factors, with the ultimate goal of improving outcomes for our most critically ill patients.
References
-
Bellani G, Laffey JG, Pham T, et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315(8):788-800.
-
Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-810.
-
Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726-736.
-
Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-147.
-
Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. Cytotherapy. 2006;8(4):315-317.
-
Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): role as guardians of inflammation. Mol Ther. 2012;20(1):14-20.
-
Krasnodembskaya A, Song Y, Fang X, et al. Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells. 2010;28(12):2229-2238.
-
Lee JW, Fang X, Gupta N, et al. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106(38):16357-16362.
-
Matthay MA, Calfee CS, Zhuo H, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir Med. 2019;7(2):154-162.
-
Bowles AC, Wise RM, Gerstein BY, et al. Immunomodulatory effects of adipose stromal vascular fraction cells promote alternative activation macrophages to repair tissue damage. Stem Cells. 2017;35(10):2198-2207.
-
Wang M, Yang Y, Yang D, et al. The immunomodulatory activity of human umbilical cord blood-derived mesenchymal stem cells in vitro. Immunology. 2009;126(2):220-232.
-
Parolini O, Alviano F, Bagnara GP, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells. 2008;26(2):300-311.
-
ARDS Definition Task Force, Ranieri VM, Rubenfeld GD, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307(23):2526-2533.
-
Gupta N, Su X, Popov B, et al. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 2007;179(3):1855-1863.
-
Lee JW, Fang X, Krasnodembskaya A, et al. Concise review: Mesenchymal stem cells for acute lung injury: role of paracrine soluble factors. Stem Cells. 2011;29(6):913-919.
-
Krasnodembskaya A, Samarani G, Song Y, et al. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am J Physiol Lung Cell Mol Physiol. 2012;302(10):L1003-1013.
-
Ortiz LA, Dutreil M, Fattman C, et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci U S A. 2007;104(26):11002-11007.
-
Wilson JG, Liu KD, Zhuo H, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3(1):24-32.
-
Matthay MA, Calfee CS, Zhuo H, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir Med. 2019;7(2):154-162.
-
Chen J, Hu C, Chen L, et al. Clinical Study of Mesenchymal Stem Cell Treatment for Acute Respiratory Distress Syndrome Induced by Epidemic Influenza A (H7N9) Infection: A Hint for COVID-19 Treatment. Engineering (Beijing). 2020;6(10):1153-1161.
-
Lanzoni G, Linetsky E, Correa D, et al. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl Med. 2021;10(5):660-673.
-
Calfee CS, Delucchi K, Parsons PE, et al. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med. 2014;2(8):611-620.
-
Evans L, Rhodes A, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock 2021. Intensive Care Med. 2021;47(11):1181-1247.
-
Gonzalez-Rey E, Anderson P, González MA, et al. Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis. Gut. 2009;58(7):929-939.
-
Mei SH, Haitsma JJ, Dos Santos CC, et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med. 2010;182(8):1047-1057.
-
Krasnodembskaya A, Samarani G, Song Y, et al. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am J Physiol Lung Cell Mol Physiol. 2012;302(10):L1003-1013.
-
Hall SR, Tsoyi K, Ith B, et al. Mesenchymal stromal cells improve survival during sepsis in the absence of heme oxygenase-1: the importance of neutrophils. Stem Cells. 2013;31(2):397-407.
-
McIntyre LA, Stewart DJ, Mei SH, et al. Cellular Immunotherapy for Septic Shock (CISS): a phase I, open-label trial. Lancet. 2013;382(9889):1341-1342.
-
Zheng G, Huang L, Tong H, et al. Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res. 2014;15:39.
-
McIntyre LA, Moher D, Fergusson DA, et al. Efficacy of mesenchymal stromal cell therapy for sepsis: a systematic review and meta-analysis. Crit Care Med. 2018;46(6):870-877.
-
Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377(6):562-572.
-
Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862-874.
-
Weiss DJ, Casaburi R, Flannery R, et al. A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest. 2013;143(6):1590-1598.
-
Galipeau J, Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824-833.
-
U.S. Food and Drug Administration. Guidance for Industry: Potency Tests for Cellular and Gene Therapy Products. 2024.
-
European Medicines Agency. Guideline on human cell-based medicinal products. 2024.
-
Willis GR, Fernandez-Gonzalez A, Anastas J, et al. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am J Respir Crit Care Med. 2018;197(1):104-116.
-
Zhu YG, Feng XM, Abbott J, et al. Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells. 2014;32(1):116-125.
-
Levy O, Kuai R, Siren EM, et al. Shattering barriers toward clinically meaningful MSC therapies. Sci Adv. 2020;6(30):eaba6884.
-
Frobel J, Hemeda H, Lenz M, et al. Epigenetic rejuvenation of mesenchymal stromal cells derived from induced pluripotent stem cells. Stem Cell Reports. 2014;3(3):414-422.
-
Ward PA. The dark side of C5a in sepsis. Nat Rev Immunol. 2004;4(2):133-142.
-
Monneret G, Lepape A, Voirin N, et al. Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock. Intensive Care Med. 2006;32(8):1175-1183.
-
Mikkelsen ME, Shah CV, Meyer NJ, et al. The epidemiology of acute respiratory distress syndrome in patients presenting to the emergency department with severe sepsis. Shock. 2013;40(5):375-381.
-
de Jong R, Houtgraaf JH, Samiei S, et al. Intracoronary stem cell infusion after acute myocardial infarction: a meta-analysis and update on clinical trials. Circ Cardiovasc Interv. 2014;7(2):156-167.
-
Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076-1084.
-
Toma C, Pittenger MF, Cahill KS, et al. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105(1):93-98.
-
Lalu MM, McIntyre L, Pugliese C, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012;7(10):e47559.
-
Hare JM, Fishman JE, Gerstenblith G, et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308(22):2369-2379.
-
Galipeau J. The mesenchymal stromal cells dilemma--does a negative phase III trial of random donor mesenchymal stromal cells in steroid-resistant graft-versus-host disease represent a death knell or a bump in the road? Cytotherapy. 2013;15(1):2-8.
-
Sensebé L, Fleury-Cappellesso S. Biodistribution of mesenchymal stem/stromal cells in a preclinical setting. Stem Cells Int. 2013;2013:678063.
-
Galipeau J, Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824-833.
-
Woodbury D, Schwarz EJ, Prockop DJ, et al. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res. 2000;61(4):364-370.
No comments:
Post a Comment