1
|
Setia O, Lee SR, Dardik A. Modalities to Deliver Cell Therapy for Treatment of Chronic Limb Threatening Ischemia. Adv Wound Care (New Rochelle) 2024; 13:253-279. [PMID: 37002893 PMCID: PMC11305013 DOI: 10.1089/wound.2022.0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/29/2023] [Indexed: 04/04/2023] Open
Abstract
Significance: Chronic limb threatening ischemia (CLTI) is a severe form of peripheral arterial disease (PAD) that is associated with high rates of morbidity and mortality, and especially limb loss. In patients with no options for revascularization, stem cell therapy is a promising treatment option. Recent Advances: Cell therapy directly delivered to the affected ischemic limb has been shown to be a safe, effective, and feasible therapeutic alternative for patients with severe PAD. Multiple methods for cell delivery, including local, regional, and combination approaches, have been examined in both pre-clinical studies and clinical trials. This review focuses on delivery modalities used in clinical trials that deliver cell therapy to patients with severe PAD. Critical Issues: Patients with CLTI are at high risk for complications of the disease, such as amputations, leading to a poor quality of life. Many of these patients do not have viable options for revascularization using traditional interventional or surgical methods. Clinical trials have shown therapeutic benefit for cell therapy in these patients, but methods of cell treatment are not standardized, including the method of cell delivery to the ischemic limb. Future Directions: The ideal delivery approach for stem cell therapy in PAD patients remains unclear. Further studies are needed to determine the best modality of cell delivery to maximize clinical benefits.
Collapse
Affiliation(s)
- Ocean Setia
- Vascular Biology and Therapeutics Program, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Shin-Rong Lee
- Vascular Biology and Therapeutics Program, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Inoue O, Goten C, Hashimuko D, Yamaguchi K, Takeda Y, Nomura A, Ootsuji H, Takashima S, Iino K, Takemura H, Halurkar M, Lim HW, Hwa V, Sanchez-Gurmaches J, Usui S, Takamura M. Single-cell transcriptomics identifies adipose tissue CD271 + progenitors for enhanced angiogenesis in limb ischemia. Cell Rep Med 2023; 4:101337. [PMID: 38118404 PMCID: PMC10772587 DOI: 10.1016/j.xcrm.2023.101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/10/2023] [Accepted: 11/21/2023] [Indexed: 12/22/2023]
Abstract
Therapeutic angiogenesis using mesenchymal stem/stromal cell grafts have shown modest and controversial effects in preventing amputation for patients with critical limb ischemia. Through single-cell transcriptomic analysis of human tissues, we identify CD271+ progenitors specifically from subcutaneous adipose tissue (AT) as having the most prominent pro-angiogenic gene profile distinct from other stem cell populations. AT-CD271+ progenitors demonstrate robust in vivo angiogenic capacity over conventional adipose stromal cell grafts, characterized by long-term engraftment, augmented tissue regeneration, and significant recovery of blood flow in a xenograft model of limb ischemia. Mechanistically, the angiogenic capacity of CD271+ progenitors is dependent on functional CD271 and mTOR signaling. Notably, the number and angiogenic capacity of CD271+ progenitors are strikingly reduced in insulin-resistant donors. Our study highlights the identification of AT-CD271+ progenitors with in vivo superior efficacy for limb ischemia. Furthermore, we showcase comprehensive single-cell transcriptomics strategies for identification of suitable grafts for cell therapy.
Collapse
Affiliation(s)
- Oto Inoue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chiaki Goten
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Daiki Hashimuko
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kosei Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Takeda
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ayano Nomura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Ootsuji
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shinichiro Takashima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kenji Iino
- Department of Thoracic, Cardiovascular and General Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hirofumi Takemura
- Department of Thoracic, Cardiovascular and General Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Manasi Halurkar
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Vivian Hwa
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Premium Research Institute for Human Medicine (WPI-PRIMe), Osaka University, Osaka, Japan; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Soichiro Usui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
3
|
Inoue O, Goten C, Hashimuko D, Yamaguchi K, Takeda Y, Nomura A, Ootsuji H, Takashima S, Iino K, Takemura H, Halurkar M, Lim HW, Hwa V, Sanchez-Gurmaches J, Usui S, Takamura M. Single cell transcriptomics identifies adipose tissue CD271+ progenitors for enhanced angiogenesis in limb ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527726. [PMID: 36865239 PMCID: PMC9980009 DOI: 10.1101/2023.02.09.527726] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Therapeutic angiogenesis using mesenchymal stem/stromal cell grafts have shown modest and controversial effects in preventing amputation for patients with critical limb ischemia. Through single-cell transcriptomic analysis of human tissues, we identified CD271 + progenitors specifically from subcutaneous adipose tissue (AT) as having the most prominent pro-angiogenic gene profile distinct from other stem cell populations. AT-CD271 + progenitors demonstrated robust in vivo angiogenic capacity, over conventional adipose stromal cell grafts, characterized by long-term engraftment, augmented tissue regeneration, and significant recovery of blood flow in a xenograft model of limb ischemia. Mechanistically, the angiogenic capacity of CD271 + progenitors is dependent on functional CD271 and mTOR signaling. Notably, the number and angiogenic capacity of CD271 + progenitors was strikingly reduced in insulin resistant donors. Our study highlights the identification of AT-CD271 + progenitors with in vivo superior efficacy for limb ischemia. Furthermore, we showcase comprehensive single-cell transcriptomics strategies for identification of suitable grafts for cell therapy. HIGHLIGHTS Adipose tissue stromal cells have a distinct angiogenic gene profile among human cell sources. CD271 + progenitors in adipose tissue have a prominent angiogenic gene profile. CD271 + progenitors show superior therapeutic capacities for limb ischemia. CD271 + progenitors are reduced and functionally impaired in insulin resistant donors. GRAPHICAL ABSTRACT
Collapse
|
4
|
PBMNCs Treatment in Critical Limb Ischemia and Candidate Biomarkers of Efficacy. Diagnostics (Basel) 2022; 12:diagnostics12051137. [PMID: 35626293 PMCID: PMC9139406 DOI: 10.3390/diagnostics12051137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/01/2022] [Accepted: 05/01/2022] [Indexed: 01/27/2023] Open
Abstract
When in critical limb ischemia (CLI) the healing process aborts or does not follow an orderly and timely sequence, a chronic vascular wound develops. The latter is major problem today, as their epidemiology is continuously increasing due to the aging population and a growth in the incidence of the underlying diseases. In the US, the mean annualized prevalence of necrotic wounds due to the fact of CLI is 1.33% (95% CI, 1.32–1.34%), and the cost of dressings alone has been estimated at USD 5 billion per year from healthcare budgets. A promising cell treatment in wound healing is the local injection of peripheral blood mononuclear cells (PBMNCs). The treatment is aimed to induce angiogenesis as well to switch inflammatory macrophages, called the M1 phenotype, into anti-inflammatory macrophages, called M2, a phenotype devoted to tissue repair. This mechanism is called polarization and is a critical step for the healing of all human tissues. Regarding the clinical efficacy of PBMNCs, the level of evidence is still low, and a considerable effort is necessary for completing the translational process toward the patient bed site. From this point of view, it is crucial to identify some candidate biomarkers to detect the switching process from M1 to M2 in response to the cell treatment.
Collapse
|
5
|
Chiang KJ, Chiu LC, Kang YN, Chen C. Autologous Stem Cell Therapy for Chronic Lower Extremity Wounds: A Meta-Analysis of Randomized Controlled Trials. Cells 2021; 10:3307. [PMID: 34943815 PMCID: PMC8699089 DOI: 10.3390/cells10123307] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Lower extremity chronic wounds (LECWs) commonly occur in patients with diabetes mellitus (DM) and peripheral arterial disease (PAD). Autologous stem cell therapy (ASCT) has emerged as a promising alternative treatment for those who suffered from LECWs. The purpose of this study was to assess the effects of ASCT on LECWs. Two authors searched three core databases, and independently identified evidence according to predefined criteria. They also individually assessed the quality of the included randomized controlled trials (RCTs), and extracted data on complete healing rate, amputation rate, and outcomes regarding peripheral circulation. The extracted data were pooled using a random-effects model due to clinical heterogeneity among the included RCTs. A subgroup analysis was further performed according to etiology, source of stem cells, follow-up time, and cell markers. A total of 28 RCTs (n = 1096) were eligible for this study. The pooled results showed that patients receiving ASCT had significantly higher complete healing rates (risk ratio (RR) = 1.67, 95% confidence interval (CI) 1.28-2.19) as compared with those without ASCT. In the CD34+ subgroup, ASCT significantly led to a higher complete healing rate (RR = 2.70, 95% CI 1.50-4.86), but there was no significant difference in the CD34- subgroup. ASCT through intramuscular injection can significantly improve wound healing in patients with LECWs caused by either DM or critical limb ischemia. Lastly, CD34+ is an important cell marker for potential wound healing. However, more extensive scale and well-designed studies are necessary to explore the details of ASCT and chronic wound healing.
Collapse
Affiliation(s)
- Kuan-Ju Chiang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-J.C.); (L.-C.C.)
| | - Li-Cheng Chiu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (K.-J.C.); (L.-C.C.)
| | - Yi-No Kang
- Department of Health Care Management, College of Health Technology, National Taipei University of Nursing Health Sciences, Taipei 112, Taiwan
- Evidence-Based Medicine Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Research Center of Big Data and Meta-Analysis Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Cochrane Taiwan, Taipei Medical University, Taipei 110, Taiwan
- Institute of Health Policy & Management, College of Public Health, National Taiwan University, Taipei 100, Taiwan
| | - Chiehfeng Chen
- Evidence-Based Medicine Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Cochrane Taiwan, Taipei Medical University, Taipei 110, Taiwan
- Division of Plastic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
6
|
A Meta-Analysis of Randomized Controlled Trials on Therapeutic Efficacy and Safety of Autologous Cell Therapy for Atherosclerosis Obliterans. J Vasc Surg 2021; 75:1440-1449.e5. [PMID: 34788653 DOI: 10.1016/j.jvs.2021.10.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Atherosclerosis obliterans (ASO) is a chronic occlusive arterial disease and the most common type of peripheral arterial disease. Current treatment options like medication and vascularization have limited effects for "no-option" patients, and stem cell therapy is considered a viable option although its application and efficacy have not been standardized. The objective of this review was to assess the safety and efficacy of autologous stem cell therapy in patients with ASO. METHODS We performed a literature search of published RCTs for ASO patients receiving stem cell therapy without a revascularization option. PubMed, Embase, and the Cochrane Library were searched. This study was conducted by a pair of authors independently and audited by a third author. Data were synthesized with a random-effect model. RESULTS 630 patients in 12 RCTs were included. The results showed that cell therapy significantly improved total amputation (RR: 0.64, p = 0.004, 95% CI: [0.47, 0.87]), major amputation (RR: 0.69, p = 0.02, 95% CI: [0.50, 0.94]), ankle-brachial index (ABI) (MD = 0.08, p = 0.004, 95% CI: [0.02, 0.13]), transcutaneous oxygen tension (TcO2) (MD = 11.52, p = 0.004, 95% CI: [3.60, 19.43]) and rest pain score (MD = -0.64, p = 0.007, 95% CI: [-1.10, -0.17]) compared to placebo or standard care. However, current studies showed cell therapy was not superior to placebo or standard care in all-cause death (RR: 0.75, p = 0.34, 95% CI: [0.41, 1.36]) and ulcer size (MD = -8.85, p = 0.39, CI: [-29.05,11.36]). LIMITATION The number of trials included was limited. Moreover, most trials were designed for "no-option" patients and thus the results should be applied with caution to other PAD patients. CONCLUSION ASO patients can benefit from autologous cell therapy in limb salvage, limb blood perfusion, and rest pain alleviation.
Collapse
|
7
|
Dubský M, Fejfarová V, Bem R, Jirkovská A, Nemcová A, Sutoris K, Husáková J, Skibová J, Jude EB. Main Factors Predicting Nonresponders to Autologous Cell Therapy for Critical Limb Ischemia in Patients With Diabetic Foot. Angiology 2021; 72:861-866. [PMID: 33783233 DOI: 10.1177/00033197211005614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Autologous cell therapy (ACT) is a new treatment for patients with no-option critical limb ischemia (NO-CLI). We evaluated the factors involved in the nonresponse to ACT in patients with CLI and diabetic foot. Diabetic patients (n = 72) with NO-CLI treated using ACT in our foot clinic over a period of 8 years were divided into responders (n = 57) and nonresponders (n = 15). Nonresponder was defined as an insufficient increase in transcutaneous oxygen pressure by <5 mm Hg, 3 months after ACT. Patient demographics, diabetes duration and treatment, and comorbidities as well as a cellular response to ACT, limb-related factors, and the presence of inherited thrombotic disorders were compared between the 2 groups. The main independent predictors for an impaired response to ACT were heterozygote Leiden mutation (OR 10.5; 95% CI, 1.72-4) and homozygote methylenetetrahydrofolate reductase (MTHFR 677) mutation (OR 3.36; 95% CI, 1.0-14.3) in stepwise logistic regression. Univariate analysis showed that lower mean protein C levels (P = .041) were present in nonresponders compared with responders. In conclusion, the significant predictors of an impaired response to ACT in diabetic patients with NO-CLI were inherited thrombotic disorders.
Collapse
Affiliation(s)
- Michal Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.,First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Robert Bem
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Andrea Nemcová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Karol Sutoris
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jitka Husáková
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.,First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jelena Skibová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Edward B Jude
- Diabetes Centre, Tameside Hospital NHS Foundation Trust and University of Manchester, Lancashire, United Kingdom
| |
Collapse
|
8
|
Outcomes of Conservative Treatment in Patients with Chronic Limb Threatening Ischaemia: A Systematic Review and Meta-Analysis. Eur J Vasc Endovasc Surg 2021; 62:214-224. [PMID: 33674157 DOI: 10.1016/j.ejvs.2021.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 12/07/2020] [Accepted: 01/07/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Chronic limb threatening ischaemia (CLTI) is the most severe form of peripheral arterial disease. International guidelines recommend arterial revascularisation in patients with CLTI. However, these patients are often fragile elderly people with significant comorbidities, whose vascular anatomy is not always suitable for open or endovascular revascularisation. Recent studies have suggested acceptable outcomes of conservative treatment. A systematic review of the available literature was conducted to obtain best estimates of outcomes of conservative treatment in patients with CLTI. DATA SOURCES MEDLINE, Embase, and Cochrane Central. REVIEW METHODS A systematic review and meta-analysis was carried out following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines. MEDLINE, Embase, and Cochrane Central were searched from inception until September 2019. All studies reporting on outcomes of conservative treatment for CLTI were considered. Study selection, data extraction, and risk of bias assessment were done by two investigators independently. Risk of bias was evaluated with a modified version of the Cochrane tool for observational studies. Outcomes of interest were all cause mortality, major amputation, and amputation free survival (AFS) after at least 12 months of follow up. A random effects model was used for meta-analyses. RESULTS Twenty-seven publications were included, consisting of 12 observational studies and 15 placebo arms from randomised clinical trials, totalling 1 642 patients. Most studies included patients with non-reconstructable CLTI. Overall study quality was moderate. The pooled 12 month all cause mortality rate in 14 studies comprising 1 003 patients was 18% (95% confidence interval [CI] 13 - 25, I2 = 73%). The pooled major amputation rate from 14 studies comprising 755 patients was 27% (95% CI 20 - 36, I2 = 65%) after one year, and pooled AFS rate after 12 months in 11 studies with 970 patients was 60% (95% CI 52 - 67, I2 = 75%). CONCLUSION Conservative treatment for patients with CLTI may be considered and does not always result in loss of limb or patient demise. The results of this review can be used to inform patients with CLTI about conservative treatment as part of a shared decision making process.
Collapse
|
9
|
Current Status of Angiogenic Cell Therapy and Related Strategies Applied in Critical Limb Ischemia. Int J Mol Sci 2021; 22:ijms22052335. [PMID: 33652743 PMCID: PMC7956816 DOI: 10.3390/ijms22052335] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Critical limb ischemia (CLI) constitutes the most severe form of peripheral arterial disease (PAD), it is characterized by progressive blockade of arterial vessels, commonly correlated to atherosclerosis. Currently, revascularization strategies (bypass grafting, angioplasty) remain the first option for CLI patients, although less than 45% of them are eligible for surgical intervention mainly due to associated comorbidities. Moreover, patients usually require amputation in the short-term. Angiogenic cell therapy has arisen as a promising alternative for these "no-option" patients, with many studies demonstrating the potential of stem cells to enhance revascularization by promoting vessel formation and blood flow recovery in ischemic tissues. Herein, we provide an overview of studies focused on the use of angiogenic cell therapies in CLI in the last years, from approaches testing different cell types in animal/pre-clinical models of CLI, to the clinical trials currently under evaluation. Furthermore, recent alternatives related to stem cell therapies such as the use of secretomes, exosomes, or even microRNA, will be also described.
Collapse
|
10
|
Blanton CM, Clougherty CO. The Role of Bone Marrow Aspirate in Osseous and Soft Tissue Pathology. Clin Podiatr Med Surg 2021; 38:1-16. [PMID: 33220739 DOI: 10.1016/j.cpm.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bone marrow aspirate (BMA) is an emerging therapy that is gaining popularity for orthoplastic reconstruction. The stem cells collected are multipotent and regenerative in nature. In addition to stem cells, other biological components collected augment the mitogen of local cells, proliferation, and angiogenesis, and inhibit proinflammatory cytokine and bacteria to optimize an environment to heal. The most common site for harvest is the iliac crest. Techniques for harvesting BMA are simple to perform, financially modest, and associated with low morbidity. Additional research is needed to evolve and standardize the technology; however, BMA is proven to be advantageous for tissue repair.
Collapse
Affiliation(s)
- Casie M Blanton
- The Reconstruction Institute of The Bellevue Hospital, 102 Commerce Park Drive, Suite D, Bellevue, OH 44811, USA.
| | - Coleman O Clougherty
- The Reconstruction Institute of The Bellevue Hospital, 102 Commerce Park Drive, Suite D, Bellevue, OH 44811, USA
| |
Collapse
|
11
|
Sharma A, Sinha M, Pandey NN, Chandrashekhara SH. Stem cell therapy in critical limb ischemia: Current scenario and future trends. Indian J Radiol Imaging 2019; 29:397-403. [PMID: 31949342 PMCID: PMC6958876 DOI: 10.4103/ijri.ijri_385_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/29/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Critical limb ischemia (CLI) represents the most severe manifestation of peripheral arterial disease (PAD). It imposes a huge economic burden and is associated with high short-term mortality and adverse cardiovascular outcomes. Prompt recognition and early revascularization, surgical or endovascular, with the aim of improving the inline bloodflow to the ischemic limb, are currently the standard of care. However, this strategy may not always be feasible or effective; hence, evaluation of newer pharmacological or angiogenic therapies for alleviating the symptoms of this alarming condition is of utmost importance. Cell-based therapies have shown promise in smaller studies; however, large-scale studies, demonstrating definite survival benefits, are entailed to ascertain their role in the management of CLI.
Collapse
Affiliation(s)
- Arun Sharma
- Department of Cardiovascular Radiology and Endovascular Interventions, All India Institute of Medical Sciences, New Delhi, India
| | - Mumun Sinha
- Department of Cardiovascular Radiology and Endovascular Interventions, All India Institute of Medical Sciences, New Delhi, India
| | - Niraj Nirmal Pandey
- Department of Cardiovascular Radiology and Endovascular Interventions, All India Institute of Medical Sciences, New Delhi, India
| | - S H Chandrashekhara
- Department of Radiodiagnosis, BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
Gao W, Chen D, Liu G, Ran X. Autologous stem cell therapy for peripheral arterial disease: a systematic review and meta-analysis of randomized controlled trials. Stem Cell Res Ther 2019; 10:140. [PMID: 31113463 PMCID: PMC6528204 DOI: 10.1186/s13287-019-1254-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023] Open
Abstract
Background Peripheral arterial disease (PAD) is a common cause of disability and mortality. The reconstruction of blood circulation presents to be the key to treatment, which can be achieved by surgery and interventional therapy. Since 40% patients have lost the chance for the therapy, a new method is needed to reduce the amputation and mortality rate for “no-option” patients. The objective of our systematic review and meta-analysis was to evaluate the efficacy and safety of autologous implantation of stem cells in patients with PAD critically, compared with active controls and placebo. Methods Randomized controlled trials (RCTs) of autologous implantation of stem cells compared with placebo and control for PAD were included. Electronic medical databases including MEDLINE, Embase, the Cochrane Central Register of Controlled Trials (CENTRAL), the Chinese Biomedical Literature Database, China National Knowledge Infrastructure (CNKI), and ClinicalTrials.gov were searched from initial period to September 2018. Independently, two reviewers screened citations, extracted data, and assessed the risk of bias according to the criteria of the Cochrane handbook. The quality of evidence was evaluated by GRADE evidence profile. The primary outcomes consisted of amputation rate, major amputation rate, ulcer healing rate, and side effects. The second outcomes included ankle-brachial index (ABI), transcutaneous oxygen tension (TcO2), pain-free walking distance (PFWD), and rest pain score. Statistical analysis was conducted via RevMan 5.3 and Stata 12.0. Results According to the twenty-seven RCTs, 1186 patients and 1280 extremities were included and the majority of studies showed a high risk of bias. Meta-analysis indicated that autologous stem cell therapy was more effective than conventional therapy on the healing rate of ulcers [OR = 4.31 (2.94, 6.30)]. There was also significant improvement in ABI [MD = 0.13 (0.10, 0.17)], TcO2 [MD = 0.13 (0.10, 0.17)], and PFWD [MD = 178.25 (128.18, 228.31)] while significant reduction was showed in amputation rate [OR = 0.50 (0.36, 0.69)] and rest pain scores [MD = − 1.61 (− 2.01, − 1.21)]. But the result presented no significant improvement in major limb salvage [0.66 (0.42, 1.03)]. Besides, stem cell therapy could reduce the amputation rate [OR = 0.50 (0.06, 0.45] and improve the ulcer healing rate [OR = 4.34 (2.96, 6.38] in DM subgroup. Eight trials reported the side effects of autologous stem cell therapy, and no serious side effects related to stem cells were reported. GRADE evidence profile showed all the quality evidence of outcomes were low. Conclusions Based on the review, autologous stem cell therapy may have a positive effect on “no-option” patients with PAD, but presented no significant improvement in major limb salvage. However, the evidence is insufficient to prove the results due to high risk of bias and low-quality evidence of outcomes. Further researches of larger, randomized, double-blind, placebo-controlled, and multicenter trials are still in demand. Electronic supplementary material The online version of this article (10.1186/s13287-019-1254-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei Gao
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.,Health Management Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Dawei Chen
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Guanjian Liu
- Chinese Cochrane Centre, Chinese EBM Centre, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xingwu Ran
- Diabetic Foot Care Center, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
13
|
Li X, Wan Q, Min J, Duan L, Liu J. Premobilization of CD133+ cells by granulocyte colony- stimulating factor attenuates ischemic acute kidney injury induced by cardiopulmonary bypass. Sci Rep 2019; 9:2470. [PMID: 30792422 PMCID: PMC6385363 DOI: 10.1038/s41598-019-38953-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/11/2019] [Indexed: 02/05/2023] Open
Abstract
Ischemic acute kidney injury (IAKI) is a common but severe complication after a cardiopulmonary bypass (CPB). Multiple studies have demonstrated that peripheral CD133+ or differentiated cells are able to home and repair the damaged tissues, but the number of available CD133+ cells is limited, and no efficient method published previously to mobilize them immediately. We analyzed the relationship between CD133+ cells and renal function in CPB patients, in addition, the efficacy of granulocyte colony-stimulating factor (G-CSF) pre-mobilized CD133+ cells in treating of mouse IAKI model have been investigated. In the clinical study, the prospective cohort study analyzed the correlation between BUN/Crea level and the peripheral CD133+ cell numbers. CPB was associated with postoperative renal dysfunction. The significant negative correlation was observed between patients' Crea and CD133+ cells (P < 0.05). The proposed mechanism studies were performed on the mouse IAKI model. The experimental mice were treated by G-CSF to mobilize CD133+ cells before implementing CPB. Data on cell count, inflammatory index, renal function/injury, and CD133+ cell mobilization were analyzed. The result demonstrated that pretreatment by G-CSF resulted in tremendous increase in the number of mouse peripheral blood and renal CD133+ cells, significantly reduces renal tissue inflammation and dramatically improves the renal function after CPB. In summary, we concluded that premobilization of CD133+ cells abated CPB induced IAKI, by promoting both repairing damaged epithelium and by its anti-inflammatory activity. Our findings stress the remarkable applications of CD133+ or differentiated cells-based therapies for potential preventing ischemic acute kidney injury.
Collapse
Affiliation(s)
- Xiaoqiang Li
- Department of Anesthesiology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Qin Wan
- Department of Anesthesiology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Jie Min
- Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Linjia Duan
- Department of Cardiology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
14
|
Barreto-Durán E, Mejía-Cruz CC, Leal-García E, Pérez-Núñez R, Rodríguez-Pardo VM. Impact of donor characteristics on the quality of bone marrow as a source of mesenchymal stromal cells. AMERICAN JOURNAL OF STEM CELLS 2018; 7:114-120. [PMID: 30697455 PMCID: PMC6334204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/10/2018] [Indexed: 06/09/2023]
Abstract
In recent years, the therapeutic use of mesenchymal stromal cells (MSC) has generated a valuable number of scientific studies that delve into their biological characteristics and their potential in regenerative medicine; however, the impact of the clinical characteristics of tissue donors, from which these cells are isolated, on their potential in applied clinical research is not yet clear. The objective of this study was to evaluate the impact of the clinical characteristics of bone marrow donors on the quality of this tissue as a source of MSC for therapeutic use. Human MSC were isolated, characterized and cultured (according to ISCT criteria) from bone marrow samples from volunteer donors (n = 70) attending the Department of Orthopedics and Traumatology of the Hospital Universitario San Ignacio (Bogota, Colombia) for surgery of prosthetic hip replacement that agreed to participate voluntarily in the study. Donor data such as age, gender, weight, smoker and type of anesthesia used during the surgical procedure were recorded, and the impact of these characteristics on the volume of tissue collection, mononuclear cell count and confluence time of cells with fibroblastoid morphology was evaluated. Correlation coefficients between quantitative variables were calculated with Spearman's correlation test, and the association between qualitative and quantitative variables was evaluated with biserial correlation coefficient. A significant correlation was observed between the age of the donors and the time necessary to obtain confluent cells in vitro (r = 0.2489, P = 0.0377); similarly, the correlation between the volume of bone marrow collected and the number of mononuclear cells obtained was significant (r = 0.7101, P = 0.0001). Although a negative correlation tendency was observed between the mononuclear cell count and the confluence time, this was not significant (r = -0.2041, P = 0.0950). No significant associations were observed between gender, smoking status or type of anesthesia and the expansion characteristics of human mesenchymal stromal cells. Bone marrow donor age and the tissue collection volume impact the time of obtaining MSC in vitro and the mononuclear cell count with which the culture starts. These conditions must be considered when the bone marrow is selected as the tissue for obtaining MSC.
Collapse
Affiliation(s)
- Emilia Barreto-Durán
- Immunobiology and Cell Biology Group, Department of Microbiology, Science Faculty, Pontificia Universidad JaverianaBogotá D.C., Colombia, South America
| | - Claudia Camila Mejía-Cruz
- Immunobiology and Cell Biology Group, Department of Microbiology, Science Faculty, Pontificia Universidad JaverianaBogotá D.C., Colombia, South America
| | - Efrain Leal-García
- Department of Orthopedics and Traumatology, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San IgnacioBogotá D.C., Colombia, South America
| | - Rafael Pérez-Núñez
- Department of Orthopedics and Traumatology, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San IgnacioBogotá D.C., Colombia, South America
| | - Viviana Marcela Rodríguez-Pardo
- Immunobiology and Cell Biology Group, Department of Microbiology, Science Faculty, Pontificia Universidad JaverianaBogotá D.C., Colombia, South America
| |
Collapse
|
15
|
Yokohama A, Yokote K, Maruhashi T. Apheresis on aged patients/donors with complicated backgrounds like ischemic heart disease, arrhythmia, and others. Transfus Apher Sci 2018; 57:619-622. [PMID: 30262215 DOI: 10.1016/j.transci.2018.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Peripheral blood stem cells (PBSCs) are currently one of the most important stem cell sources for hematopoietic stem cell transplantation as well as cell therapy for ischemic heart disease or critical limb ischemia. Thus, it is sometimes necessary to collect autologous PBSCs from donors who have comorbidities. In terms yield, a sufficient number of PBSCs can be collected from donors with comorbidities for performing cell therapy if their age is < 60 years or up to a maximum of 70 years, although the number of PBSCs collected from older donors would probably be lower than that obtained from younger donors. On the other hand, granulocyte colony-stimulating factor (G-CSF) administration sometimes results in severe adverse events (AEs), such as ischemic heart disease and vascular thrombosis. Therefore, it is very important to perform strict medical check-ups according to the standards for donor operations in each country before apheresis. The apheresis procedure and G-CSF administration should be performed after administering the appropriate treatment. There is very less information available regarding AEs related to citrate administration during apheresis in aged donors with complicated medical histories. Medical staff should have knowledge of the electrocardiogram (ECG) QTc prolongation that occurs during apheresis owing to hypocalcemia caused by citrate administration, necessitating electrocardiographic monitoring of patients. Calcium should be administered during apheresis to prevent citrate related symptoms.
Collapse
Affiliation(s)
- Akihiko Yokohama
- Transfusion Service, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan.
| | - Keiko Yokote
- Transfusion Service, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan; Division of Nursing, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan
| | - Takayuki Maruhashi
- Transfusion Service, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan
| |
Collapse
|
16
|
Clinical Trial Design for Investigational Cardio-Regenerative Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018. [PMID: 30238373 DOI: 10.1007/978-3-319-97421-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Human trials of cardio-regenerative biologic therapies are being performed worldwide to address a growing, unmet need for durable treatments of cardiovascular disease. A well-constructed clinical trial design for these novel therapies requires careful attention to defining a clear hypothesis, a patient population, and anticipated outcomes. The scope of screening, method of randomization, blinding approach, data monitoring, and statistical analysis plan are the foundational elements that must be addressed in any clinical trial. Although the experience of human trials involving extracellular matrix constructs for cardiovascular disease treatment is limited, numerous lessons have been learned in the field of cell therapy that are translatable across all biologic treatment options. Future progress in this field may include testing combinations of cells, gene-transfer agents, and matrix and identifying treatment responders versus nonresponders.
Collapse
|
17
|
Abdul Wahid SF, Ismail NA, Wan Jamaludin WF, Muhamad NA, Abdul Hamid MKA, Harunarashid H, Lai NM. Autologous cells derived from different sources and administered using different regimens for 'no-option' critical lower limb ischaemia patients. Cochrane Database Syst Rev 2018; 8:CD010747. [PMID: 30155883 PMCID: PMC6513643 DOI: 10.1002/14651858.cd010747.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Revascularisation is the gold standard therapy for patients with critical limb ischaemia (CLI). In over 30% of patients who are not suitable for or have failed previous revascularisation therapy (the 'no-option' CLI patients), limb amputation is eventually unavoidable. Preliminary studies have reported encouraging outcomes with autologous cell-based therapy for the treatment of CLI in these 'no-option' patients. However, studies comparing the angiogenic potency and clinical effects of autologous cells derived from different sources have yielded limited data. Data regarding cell doses and routes of administration are also limited. OBJECTIVES To compare the efficacy and safety of autologous cells derived from different sources, prepared using different protocols, administered at different doses, and delivered via different routes for the treatment of 'no-option' CLI patients. SEARCH METHODS The Cochrane Vascular Information Specialist (CIS) searched the Cochrane Vascular Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE Ovid, Embase Ovid, the Cumulative Index to Nursing and Allied Health Literature (CINAHL), the Allied and Complementary Medicine Database (AMED), and trials registries (16 May 2018). Review authors searched PubMed until February 2017. SELECTION CRITERIA We included randomised controlled trials (RCTs) involving 'no-option' CLI patients comparing a particular source or regimen of autologous cell-based therapy against another source or regimen of autologous cell-based therapy. DATA COLLECTION AND ANALYSIS Three review authors independently assessed the eligibility and methodological quality of the trials. We extracted outcome data from each trial and pooled them for meta-analysis. We calculated effect estimates using a risk ratio (RR) with 95% confidence interval (CI), or a mean difference (MD) with 95% CI. MAIN RESULTS We included seven RCTs with a total of 359 participants. These studies compared bone marrow-mononuclear cells (BM-MNCs) versus mobilised peripheral blood stem cells (mPBSCs), BM-MNCs versus bone marrow-mesenchymal stem cells (BM-MSCs), high cell dose versus low cell dose, and intramuscular (IM) versus intra-arterial (IA) routes of cell implantation. We identified no other comparisons in these studies. We considered most studies to be at low risk of bias in random sequence generation, incomplete outcome data, and selective outcome reporting; at high risk of bias in blinding of patients and personnel; and at unclear risk of bias in allocation concealment and blinding of outcome assessors. The quality of evidence was most often low to very low, with risk of bias, imprecision, and indirectness of outcomes the major downgrading factors.Three RCTs (100 participants) reported a total of nine deaths during the study follow-up period. These studies did not report deaths according to treatment group.Results show no clear difference in amputation rates between IM and IA routes (RR 0.80, 95% CI 0.54 to 1.18; three RCTs, 95 participants; low-quality evidence). Single-study data show no clear difference in amputation rates between BM-MNC- and mPBSC-treated groups (RR 1.54, 95% CI 0.45 to 5.24; 150 participants; low-quality evidence) and between high and low cell dose (RR 3.21, 95% CI 0.87 to 11.90; 16 participants; very low-quality evidence). The study comparing BM-MNCs versus BM-MSCs reported no amputations.Single-study data with low-quality evidence show similar numbers of participants with healing ulcers between BM-MNCs and mPBSCs (RR 0.89, 95% CI 0.44 to 1.83; 49 participants) and between IM and IA routes (RR 1.13, 95% CI 0.73 to 1.76; 41 participants). In contrast, more participants appeared to have healing ulcers in the BM-MSC group than in the BM-MNC group (RR 2.00, 95% CI 1.02 to 3.92; one RCT, 22 participants; moderate-quality evidence). Researchers comparing high versus low cell doses did not report ulcer healing.Single-study data show similar numbers of participants with reduction in rest pain between BM-MNCs and mPBSCs (RR 0.99, 95% CI 0.93 to 1.06; 104 participants; moderate-quality evidence) and between IM and IA routes (RR 1.22, 95% CI 0.91 to 1.64; 32 participants; low-quality evidence). One study reported no clear difference in rest pain scores between BM-MNC and BM-MSC (MD 0.00, 95% CI -0.61 to 0.61; 37 participants; moderate-quality evidence). Trials comparing high versus low cell doses did not report rest pain.Single-study data show no clear difference in the number of participants with increased ankle-brachial index (ABI; increase of > 0.1 from pretreatment), between BM-MNCs and mPBSCs (RR 1.00, 95% CI 0.71 to 1.40; 104 participants; moderate-quality evidence), and between IM and IA routes (RR 0.93, 95% CI 0.43 to 2.00; 35 participants; very low-quality evidence). In contrast, ABI scores appeared higher in BM-MSC versus BM-MNC groups (MD 0.05, 95% CI 0.01 to 0.09; one RCT, 37 participants; low-quality evidence). ABI was not reported in the high versus low cell dose comparison.Similar numbers of participants had improved transcutaneous oxygen tension (TcO₂) with IM versus IA routes (RR 1.22, 95% CI 0.86 to 1.72; two RCTs, 62 participants; very low-quality evidence). Single-study data with low-quality evidence show a higher TcO₂ reading in BM-MSC versus BM-MNC groups (MD 8.00, 95% CI 3.46 to 12.54; 37 participants) and in mPBSC- versus BM-MNC-treated groups (MD 1.70, 95% CI 0.41 to 2.99; 150 participants). TcO₂ was not reported in the high versus low cell dose comparison.Study authors reported no significant short-term adverse effects attributed to autologous cell implantation. AUTHORS' CONCLUSIONS Mostly low- and very low-quality evidence suggests no clear differences between different stem cell sources and different treatment regimens of autologous cell implantation for outcomes such as all-cause mortality, amputation rate, ulcer healing, and rest pain for 'no-option' CLI patients. Pooled analyses did not show a clear difference in clinical outcomes whether cells were administered via IM or IA routes. High-quality evidence is lacking; therefore the efficacy and long-term safety of autologous cells derived from different sources, prepared using different protocols, administered at different doses, and delivered via different routes for the treatment of 'no-option' CLI patients, remain to be confirmed.Future RCTs with larger numbers of participants are needed to determine the efficacy of cell-based therapy for CLI patients, along with the optimal cell source, phenotype, dose, and route of implantation. Longer follow-up is needed to confirm the durability of angiogenic potential and the long-term safety of cell-based therapy.
Collapse
Affiliation(s)
- S Fadilah Abdul Wahid
- Universiti Kebangsaan Malaysia Medical CentreCell Therapy CenterJalan Yaacob LatifKuala LumpurMalaysia56000
- Universiti Kebangsaan Malaysia Medical CentreClinical Haematology & Stem Cell Transplantation Services, Department of MedicineKuala LumpurMalaysia
| | - Nor Azimah Ismail
- Universiti Kebangsaan Malaysia Medical CentreCell Therapy CenterJalan Yaacob LatifKuala LumpurMalaysia56000
| | - Wan Fariza Wan Jamaludin
- Universiti Kebangsaan Malaysia Medical CentreCell Therapy CenterJalan Yaacob LatifKuala LumpurMalaysia56000
| | - Nor Asiah Muhamad
- Ministry of HealthInstitute for Public HealthKuala LumpurFederal TeritoryMalaysia50590
| | | | - Hanafiah Harunarashid
- Universiti Kebangsaan Malaysia Medical CentreUnit of Vascular Surgery, Department of SurgeryJalan Yaacob LatifKuala LumpurKuala LumpurMalaysia56000
| | - Nai Ming Lai
- Taylor's UniversitySchool of MedicineSubang JayaMalaysia
| | | |
Collapse
|
18
|
Raval AN, Cook TD, Duckers HJ, Johnston PV, Traverse JH, Abraham WT, Altman PA, Pepine CJ. The CardiAMP Heart Failure trial: A randomized controlled pivotal trial of high-dose autologous bone marrow mononuclear cells using the CardiAMP cell therapy system in patients with post-myocardial infarction heart failure: Trial rationale and study design. Am Heart J 2018; 201:141-148. [PMID: 29803986 DOI: 10.1016/j.ahj.2018.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/24/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Heart failure following myocardial infarction is a common, disabling, and deadly condition. Direct injection of autologous bone marrow mononuclear cells into the myocardium may result in improved functional recovery, relieve symptoms, and improve other cardiovascular outcomes. METHODS CardiAMP-HF is a randomized, double-blind, sham-controlled, pivotal trial designed to investigate the safety and efficacy of autologous bone marrow mononuclear cells treatment for patients with medically refractory and symptomatic ischemic cardiomyopathy. The primary end point is change in 6-minute walk distance adjusted for major adverse cardiovascular events at 12 months following treatment. Particularly novel aspects of this trial include a cell potency assay to screen subjects who have bone marrow cell characteristics that suggest a favorable response to treatment, a point-of-care treatment method, a high target dose of 200 million cells, and an efficient transcatheter intramyocardial delivery method that is associated with high cell retention. CONCLUSIONS This novel approach may lead to a new treatment for those with ischemic heart disease suffering from medically refractory heart failure.
Collapse
|
19
|
Hausburg F, Müller P, Voronina N, Steinhoff G, David R. Protocol for MicroRNA Transfer into Adult Bone Marrow-derived Hematopoietic Stem Cells to Enable Cell Engineering Combined with Magnetic Targeting. J Vis Exp 2018. [PMID: 29985305 DOI: 10.3791/57474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
While CD133+ hematopoietic stem cells (SCs) have been proven to provide high potential in the field of regenerative medicine, their low retention rates after injection into injured tissues as well as the observed massive cell death rates lead to very restricted therapeutic effects. To overcome these limitations, we sought to establish a non-viral based protocol for suitable cell engineering prior to their administration. The modification of human CD133+ expressing SCs using microRNA (miR) loaded magnetic polyplexes was addressed with respect to uptake efficiency and safety as well as the targeting potential of the cells. Relying on our protocol, we can achieve high miR uptake rates of 80-90% while the CD133+ stem cell properties remain unaffected. Moreover, these modified cells offer the option of magnetic targeting. We describe here a safe and highly efficient procedure for the modification of CD133+ SCs. We expect this approach to provide a standard technology for optimization of therapeutic stem cell effects and for monitoring of the administered cell product via magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Frauke Hausburg
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University
| | - Paula Müller
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University
| | - Natalia Voronina
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University;
| |
Collapse
|
20
|
Pan T, Wei Z, Fang Y, Dong Z, Fu W. Therapeutic efficacy of CD34 + cell-involved mononuclear cell therapy for no-option critical limb ischemia: A meta-analysis of randomized controlled clinical trials. Vasc Med 2018; 23:219-231. [PMID: 29457540 DOI: 10.1177/1358863x17752556] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Early-phase clinical trials in patients with critical limb ischemia (CLI) have shown positive results of mononuclear cell therapy. The current meta-analysis investigated whether cluster of differentiation (CD) 34+ mononuclear cell therapy (CD34+MCT) is effective for no-option CLI. Ten randomized controlled clinical studies of CD34+MCT for no-option CLI with 479 patients were identified and analyzed for pooled results. Compared to control groups, the CD34+MCT was associated with lower total amputation (odds ratio (OR): 0.45, p=0.01; 95% confidence interval (CI): 0.24-0.85) and a higher complete ulcer healing rate (OR: 2.80, p=0.008; 95% CI: 1.31-6.02), but showed no advantage in major amputation (OR: 0.58, p=0.11; 95% CI: 0.29-1.14) and all-cause mortality (OR: 0.82, p=0.62; 95% CI: 0.36-1.83) . Studies with a high CD34+ cell dosage showed significant results in major amputation (OR: 0.38, p=0.002; 95% CI: 0.21-0.70), total amputation (OR: 0.31, p=0.0002; 95% CI: 0.17-0.57) and complete ulcer healing (OR: 7.58, p=0.0005; 95% CI: 2.40-23.88), which were not observed in the low-dose studies. However, inclusion of placebo-controlled studies showed no improvement of the CD34+MCT in total amputation (OR: 0.67, p=0.42; 95% CI: 0.25-1.79), major amputation (OR: 1.31, p=0.43; 95% CI: 0.67-2.54) or complete ulcer healing (OR: 1.52, p=0.27; 95% CI: 0.72-3.21), which were extremely significant in non-placebo-controlled studies ( p<0.001). In conclusion, the significant results of CD34+MCT might not support its therapeutic benefit due to high placebo-effect risk and considerable heterogeneity caused by distinct cell doses. More sizable double-blinded, randomized, placebo-controlled trials with higher CD34+ cell dosage are needed in the future.
Collapse
Affiliation(s)
- Tianyue Pan
- 1 Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Wei
- 2 Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Fang
- 1 Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihui Dong
- 1 Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiguo Fu
- 1 Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Autologous Stem Cell Therapy in Critical Limb Ischemia: A Meta-Analysis of Randomized Controlled Trials. Stem Cells Int 2018; 2018:7528464. [PMID: 29977308 PMCID: PMC5994285 DOI: 10.1155/2018/7528464] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/16/2018] [Indexed: 01/13/2023] Open
Abstract
Objective Critical limb ischemia (CLI) is the most dangerous stage of peripheral artery disease (PAD). Many basic researches and clinical treatment had been focused on stem cell transplantation for CLI. This systematic review was performed to review evidence for safety and efficacy of autologous stem cell therapy in CLI. Methods A systematic literature search was performed in the SinoMed, PubMed, Embase, ClinicalTrials.gov, and Cochrane Controlled Trials Register databases from building database to January 2018. Results Meta-analysis showed that cell therapy significantly increased the probability of ulcer healing (RR = 1.73, 95% CI = 1.45–2.06), angiogenesis (RR = 5.91, 95% CI = 2.49–14.02), and reduced the amputation rates (RR = 0.59, 95% CI = 0.46–0.76). Ankle-brachial index (ABI) (MD = 0.13, 95% CI = 0.11–0.15), TcO2 (MD = 12.22, 95% CI = 5.03–19.41), and pain-free walking distance (MD = 144.84, 95% CI = 53.03–236.66) were significantly better in the cell therapy group than in the control group (P < 0.01). Conclusions The results of this meta-analysis indicate that autologous stem cell therapy is safe and effective in CLI. However, higher quality and larger RCTs are required for further investigation to support clinical application of stem cell transplantation.
Collapse
|
22
|
Frangogiannis NG. Cell therapy for peripheral artery disease. Curr Opin Pharmacol 2018; 39:27-34. [PMID: 29452987 DOI: 10.1016/j.coph.2018.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/28/2022]
Abstract
Patients with severe peripheral artery disease (PAD) who are not candidates for revascularization have poor prognosis. Cell therapy using peripheral blood-derived or bone marrow-derived mononuclear cells, mesenchymal stem cells, or marker-specific subsets of bone marrow cells with angiogenic properties may hold promise for no-option PAD patients. Injected cells may exert beneficial actions by enhancing local angiogenesis (either through maturation of endothelial progenitors, or through secretion of angiogenic mediators), or by transducing cytoprotective signals that preserve tissue structure. Despite extensive research, robust clinical evidence supporting the use of cell therapy in patients with critical limb ischemia is lacking. Larger, well-designed placebo-controlled clinical trials did not support the positive results of smaller less rigorous studies. There is a need for high-quality clinical studies to test the effectiveness of cell therapy in PAD patients. Moreover, fundamental cell biological studies are needed to identify the optimal cell types, and to develop strategies that may enhance homing, survival and effectiveness of the injected cells.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
23
|
Fujita Y, Kawamoto A. Stem cell-based peripheral vascular regeneration. Adv Drug Deliv Rev 2017; 120:25-40. [PMID: 28912015 DOI: 10.1016/j.addr.2017.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Chronic critical limb ischemia (CLI) represents an end-stage manifestation of peripheral arterial disease (PAD). CLI patients are at very high risk of amputation and cardiovascular complications, leading to severe morbidity and mortality. Because many patients with CLI are ineligible for conventional revascularization procedures, it is urgently needed to explore alternative strategies to improve blood supply in the ischemic tissue. Although researchers initially focused on gene/protein therapy using proangiogenic growth factors/cytokines, recent discovery of somatic stem/progenitor cells including bone marrow (BM)-derived endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) has drastically developed the field of therapeutic angiogenesis for CLI. Overall, early phase clinical trials demonstrated that stem/progenitor cell therapies may be safe, feasible and potentially effective. However, only few late-phase clinical trials have been conducted. This review provides an overview of the preclinical and clinical reports to demonstrate the usefulness and the current limitations of the cell-based therapies.
Collapse
Affiliation(s)
- Yasuyuki Fujita
- Division of Vascular Regeneration, Unit of Regenerative Medicine, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation, Japan; Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Japan
| | - Atsuhiko Kawamoto
- Division of Vascular Regeneration, Unit of Regenerative Medicine, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation, Japan; Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Japan.
| |
Collapse
|
24
|
Dubský M, Jirkovská A, Bem R, Nemcová A, Fejfarová V, Jude EB. Cell therapy of critical limb ischemia in diabetic patients - State of art. Diabetes Res Clin Pract 2017; 126:263-271. [PMID: 28288436 DOI: 10.1016/j.diabres.2017.02.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/19/2016] [Accepted: 02/22/2017] [Indexed: 01/15/2023]
Abstract
In this review we report on the state of cell therapy of critical limb ischemia (CLI) with respect to differences between diabetic and non-diabetic patients mainly from the clinical point of view. CLI is the most severe form of peripheral arterial disease and its diagnosis and treatment in diabetic patients is very difficult. The therapeutic effect of standard methods of CLI treatment is only partial - more than one third of diabetic patients are not eligible for standard revascularization; therefore, new therapeutic techniques such as cell therapy have been studied in clinical trials. Presence of CLI in patients with diabetic foot disease is associated with worse clinical outcomes such as lack of healing of foot ulcers, major amputations and premature mortality. A revascularization procedure cannot be successful as the only method in contrast to patients without diabetes, but it must always be part of a complex therapy focused not only on ischemia, but also on treatment of infection, off-loading, metabolic control of diabetes and nutrition, local therapy, etc. Therefore, the main criteria for cell therapy may vary in diabetic patients and non-diabetic persons and results of this treatment method should always be assessed in the context of ensuring comprehensive therapy. This review carries out an analysis of the source of precursor cells, route of administration and brings a brief report of published data with respect to diabetic and non-diabetic patients and our experience with autologous cell therapy of diabetic patients with CLI. Analysis of the studies in terms of diabetes is difficult, because in most of them sub-analysis for diabetic patients is not performed separately. The other problem is that it is not clear if diabetic patients received adequate complex treatment for their foot ulcers which can strongly affect the rate of major amputation as an outcome of CLI treatment.
Collapse
Affiliation(s)
- Michal Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | - Robert Bem
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Andrea Nemcová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Edward B Jude
- Diabetes Centre, Tameside Hospital NHS Foundation Trust and University of Manchester, Lancashire, UK
| |
Collapse
|
25
|
Jin E, Chae DS, Son M, Kim SW. Angiogenic characteristics of human stromal vascular fraction in ischemic hindlimb. Int J Cardiol 2017; 234:38-47. [PMID: 28258850 DOI: 10.1016/j.ijcard.2017.02.080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 02/03/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
Abstract
INTRODUCTION In this study, we sought to characterize the angio-vasculogenic property of human adipose tissue-derived stromal vascular fraction (SVF) and to determine the therapeutic potential of SVF in the context of experimental ischemia. Although human SVF is used for cell therapy, its angiogenic and vasculogenic characteristics have not been fully elucidated. METHODS AND RESULTS We conducted flow cytometry, microarray, quantitative (q)-PCR, Matrigel tube formation assays and in vivo therapeutic assays using an ischemic hind limb mouse model. Gene/micro RNA microarray, quantitative (q)-PCR results revealed that the representative pro-angiogenic factors were highly upregulated in SVF compared with human adipose-derived mesenchymal stem cells (ASCs). In addition, SVF exhibited high expression of endothelium-specific genes and showed robust in vitro micro-vascular formation. SVF was transplanted into ischemic mouse hind limbs and compared with ASC transplantation. SVF transplantation prevented limb loss and augmented blood perfusion, indicating that SVF promotes neovascularization in hind limb ischemia. Transplanted SVF showed high vasculogenic potential in vivo compared with transplanted ASCs. CONCLUSIONS Our data indicate that SVF has remarkable therapeutic effects on hind limb ischemia via robust angiogenic and vasculogenic activity.
Collapse
Affiliation(s)
- Enze Jin
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, Republic of Korea
| | - Mina Son
- Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Republic of Korea
| | - Sung-Whan Kim
- Department of Medicine, College of Medicine, Catholic Kwandong University, Gangneung, Republic of Korea.
| |
Collapse
|
26
|
Safety and Effectiveness of Bone Marrow Cell Concentrate in the Treatment of Chronic Critical Limb Ischemia Utilizing a Rapid Point-of-Care System. Stem Cells Int 2017; 2017:4137626. [PMID: 28194186 PMCID: PMC5282442 DOI: 10.1155/2017/4137626] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/14/2016] [Indexed: 01/09/2023] Open
Abstract
Critical limb ischemia (CLI) is the end stage of lower extremity peripheral vascular disease (PVD) in which severe obstruction of blood flow results in ischemic rest pain, ulcers and/or gangrene, and a significant risk of limb loss. This open-label, single-arm feasibility study evaluated the safety and therapeutic effectiveness of autologous bone marrow cell (aBMC) concentrate in revascularization of CLI patients utilizing a rapid point-of-care device. Seventeen (17) no-option CLI patients with ischemic rest pain were enrolled in the study. Single dose of aBMC, prepared utilizing an intraoperative point-of-care device, the Res-Q™ 60 BMC system, was injected intramuscularly into the afflicted limb and patients were followed up at regular intervals for 12 months. A statistically significant improvement in Ankle Brachial Index (ABI), Transcutaneous Oxygen Pressure (TcPO2), mean rest pain and intermittent claudication pain scores, wound/ ulcer healing, and 6-minute walking distance was observed following aBMC treatment. Major amputation-free survival (mAFS) rate and amputation-free rates (AFR) at 12 months were 70.6% and 82.3%, respectively. In conclusion, aBMC injections were well tolerated with improved tissue perfusion, confirming the safety, feasibility, and preliminary effectiveness of aBMC treatment in CLI patients.
Collapse
|
27
|
Magnet-Bead Based MicroRNA Delivery System to Modify CD133 + Stem Cells. Stem Cells Int 2016; 2016:7152761. [PMID: 27795713 PMCID: PMC5067480 DOI: 10.1155/2016/7152761] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022] Open
Abstract
Aim. CD133+ stem cells bear huge potential for regenerative medicine. However, low retention in the injured tissue and massive cell death reduce beneficial effects. In order to address these issues, we intended to develop a nonviral system for appropriate cell engineering. Materials and Methods. Modification of human CD133+ stem cells with magnetic polyplexes carrying microRNA was studied in terms of efficiency, safety, and targeting potential. Results. High microRNA uptake rates (~80-90%) were achieved without affecting CD133+ stem cell properties. Modified cells can be magnetically guided. Conclusion. We developed a safe and efficient protocol for CD133+ stem cell modification. Our work may become a basis to improve stem cell therapeutical effects as well as their monitoring with magnetic resonance imaging.
Collapse
|
28
|
Abstract
Diabetes is one of the main economic burdens in health care, which threatens to worsen dramatically if prevalence forecasts are correct. What makes diabetes harmful is the multi-organ distribution of its microvascular and macrovascular complications. Regenerative medicine with cellular therapy could be the dam against life-threatening or life-altering complications. Bone marrow-derived stem cells are putative candidates to achieve this goal. Unfortunately, the bone marrow itself is affected by diabetes, as it can develop a microangiopathy and neuropathy similar to other body tissues. Neuropathy leads to impaired stem cell mobilization from marrow, the so-called mobilopathy. Here, we review the role of bone marrow-derived stem cells in diabetes: how they are affected by compromised bone marrow integrity, how they contribute to other diabetic complications, and how they can be used as a treatment for these. Eventually, we suggest new tactics to optimize stem cell therapy.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| |
Collapse
|
29
|
Abstract
Clinical investigations using stem cell products in regenerative medicine are addressing a wide spectrum of conditions using a variety of stem cell types. To date, there have been few reports of safety issues arising from autologous or allogeneic transplants. Many cells administered show transient presence for a few days with trophic influences on immune or inflammatory responses. Limbal stem cells have been registered as a product for eye burns in Europe and mesenchymal stem cells have been approved for pediatric graft versus host disease in Canada and New Zealand. Many other applications are progressing in trials, some with early benefits to patients.
Collapse
Affiliation(s)
- Alan Trounson
- Hudson Institute for Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia.
| | - Courtney McDonald
- Hudson Institute for Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|
30
|
Ai M, Yan CF, Xia FC, Zhou SL, He J, Li CP. Safety and efficacy of cell-based therapy on critical limb ischemia: A meta-analysis. Cytotherapy 2016; 18:712-24. [PMID: 27067609 DOI: 10.1016/j.jcyt.2016.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/13/2016] [Accepted: 02/22/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND AIMS Critical limb ischemia (CLI) is a major health problem worldwide, affecting approximately 500-1000 people per million per annum. Cell-based therapy has given new hope for the treatment of limb ischemia. This study assessed the safety and efficacy of cellular therapy CLI treatment. METHODS We searched the PubMed, Embase and Cochrane databases through October 20, 2015, and selected the controlled trials with cell-based therapy for CLI treatment compared with cell-free treatment. We assessed the results by meta-analysis using a variety of outcome measures, as well as the association of mononuclear cell dosage with treatment effect by dose-response meta-analysis. RESULTS Twenty-five trials were included. For the primary evaluation index, cell-based therapy significantly reduced the rate of major amputation (odds ratio [OR] 0.44, 95% confidence interval [CI] 0.32-0.60, P = 0.000) and significantly increased the rate of amputation-free survival (OR 2.80, 95% CI 1.70-4.61, P = 0.000). Trial sequence analysis indicated that optimal sample size (n = 3374) is needed to detect a plausible treatment effect in all-cause mortality. Cell-based therapy significantly improves ankle brachial index, increases the rate of ulcer healing, increases the transcutaneous pressure of oxygen, reduces limb pain and improves movement ability. Subgroup analysis indicated heterogeneity is caused by type of control, design bias and transplant route. In the dose-response analysis, there was no significant correlation between cell dosage and the therapeutic effect. CONCLUSIONS Cell-based therapy has a significant therapeutic effect on CLI, but randomized double-blind placebo-controlled trials are needed to improve the credibility of this conclusion. Assessment of all-cause mortality also requires a larger sample size to arrive at a strong conclusion. In dose-response analysis, increasing the dosage of cell injections does not significantly improve the therapeutic effects of cell-based therapy.
Collapse
Affiliation(s)
- Min Ai
- Pangang General Hospital, Panzhihua, Sichuan Provience, China
| | - Chang-Fu Yan
- Pangang General Hospital, Panzhihua, Sichuan Provience, China.
| | - Fu-Chun Xia
- Pangang General Hospital, Panzhihua, Sichuan Provience, China
| | - Shuang-Lu Zhou
- Pangang General Hospital, Panzhihua, Sichuan Provience, China
| | - Jian He
- Pangang General Hospital, Panzhihua, Sichuan Provience, China
| | - Cui-Ping Li
- Pangang General Hospital, Panzhihua, Sichuan Provience, China
| |
Collapse
|
31
|
Rurali E, Bassetti B, Perrucci GL, Zanobini M, Malafronte C, Achilli F, Gambini E. BM ageing: Implication for cell therapy with EPCs. Mech Ageing Dev 2016; 159:4-13. [PMID: 27045606 DOI: 10.1016/j.mad.2016.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/22/2016] [Accepted: 04/01/2016] [Indexed: 12/17/2022]
Abstract
The bone marrow (BM) is a well-recognized source of stem/progenitor cells for cell therapy in cardiovascular diseases (CVDs). Preclinical and clinical studies suggest that endothelial progenitor cells (EPCs) contribute to reparative process of vascular endothelium and participate in angiogenesis. As for all organs and cells across the lifespan, BM and EPCs are negatively impacted by ageing due to microenvironment modifications and EPC progressive dysfunctions. The encouraging results in terms of neovascularization observed in young animals after EPC administration were mitigated in aged patients treated for ischemic CVDs. The limited efficacy of EPC-based therapy in clinical setting might be ascribed at least partly to ageing. In this review, we comprehensively discussed the age-related changes of BM and EPCs and their implication for cardiovascular cell-therapies. Finally, we examined alternative approaches under investigation to enhance EPC potency.
Collapse
Affiliation(s)
- Erica Rurali
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Marco Zanobini
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | - Felice Achilli
- Cardiology Department, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| |
Collapse
|
32
|
Transcatheter Arterial Infusion of Autologous CD133(+) Cells for Diabetic Peripheral Artery Disease. Stem Cells Int 2016; 2016:6925357. [PMID: 26981134 PMCID: PMC4769775 DOI: 10.1155/2016/6925357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/10/2015] [Accepted: 01/04/2016] [Indexed: 01/06/2023] Open
Abstract
Microvascular lesion in diabetic peripheral arterial disease (PAD) still cannot be resolved by current surgical and interventional technique. Endothelial cells have the therapeutic potential to cure microvascular lesion. To evaluate the efficacy and immune-regulatory impact of intra-arterial infusion of autologous CD133(+) cells, we recruited 53 patients with diabetic PAD (27 of CD133(+) group and 26 of control group). CD133(+) cells enriched from patients' PB-MNCs were reinfused intra-arterially. The ulcer healing followed up till 18 months was 100% (3/3) in CD133(+) group and 60% (3/5) in control group. The amputation rate was 0 (0/27) in CD133(+) group and 11.54% (3/26) in control group. Compared with the control group, TcPO2 and ABI showed obvious improvement at 18 months and significant increasing VEGF and decreasing IL-6 level in the CD133(+) group within 4 weeks. A reducing trend of proangiogenesis and anti-inflammatory regulation function at 4 weeks after the cells infusion was also found. These results indicated that autologous CD133(+) cell treatment can effectively improve the perfusion of morbid limb and exert proangiogenesis and anti-inflammatory immune-regulatory impacts by paracrine on tissue microenvironment. The CD133(+) progenitor cell therapy may be repeated at a fixed interval according to cell life span and immune-regulatory function.
Collapse
|
33
|
Peeters Weem S, Teraa M, de Borst G, Verhaar M, Moll F. Bone Marrow derived Cell Therapy in Critical Limb Ischemia: A Meta-analysis of Randomized Placebo Controlled Trials. Eur J Vasc Endovasc Surg 2015; 50:775-83. [DOI: 10.1016/j.ejvs.2015.08.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/22/2015] [Indexed: 01/13/2023]
|
34
|
Liew A, Bhattacharya V, Shaw J, Stansby G. Cell Therapy for Critical Limb Ischemia: A Meta-Analysis of Randomized Controlled Trials. Angiology 2015. [PMID: 26195561 DOI: 10.1177/0003319715595172] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Early-phase trials showed the feasibility and potential efficacy of cell therapy in patients with critical limb ischemia (CLI). For systematic review, randomized controlled trials (RCTs) of cell therapy versus no cell therapy in CLI were searched from PubMed and the Cochrane library databases. Outcome measures included major amputation, complete ulcer healing, ankle-brachial index (ABI), and all-cause mortality. Data were pooled using 16 RCTs, involving 774 patients. Compared with no cell therapy, cell therapy significantly reduced major amputation (odds ratio [OR]: 0.54; 95% CI: 0.34-0.87:P= .01) and improved ulcer healing (OR: 2.90; 95% confidence interval [CI]: 1.44-5.82;P< .01) and ABI (OR: 5.91; 95% CI: 1.85-18.86:P< .01). Peripheral blood-derived mononuclear cells (PB-MNCs; OR: 0.29; 95% CI: 0.12-0.72;P< .01) and bone marrow concentrate (OR: 0.44; 95% CI: 0.21-0.93;P= .03) significantly lowered the risk of major amputation. The PB-MNCs also significantly increased ulcer healing (OR: 5.77; 95% CI: 1.77-18.87;P< .01). All-cause mortality was similar in both groups (OR: 0.78; 95% CI: 0.44-1.40;P= .41). However, all estimates were nonsignificant following reanalysis using placebo-controlled RCTs only. Cell therapy remains a potential therapeutic option in CLI, but further larger placebo-controlled RCTs are needed.
Collapse
Affiliation(s)
- Aaron Liew
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, United Kingdom
| | | | - James Shaw
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, United Kingdom
| | - Gerard Stansby
- Freeman Hospital, High Heaton, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
35
|
Almeida SO, Skelton RJ, Adigopula S, Ardehali R. Arrhythmia in stem cell transplantation. Card Electrophysiol Clin 2015; 7:357-70. [PMID: 26002399 DOI: 10.1016/j.ccep.2015.03.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Stem cell regenerative therapies hold promise for treating diseases across the spectrum of medicine. While significant progress has been made in the preclinical stages, the clinical application of cardiac cell therapy is limited by technical challenges. Certain methods of cell delivery, such as intramyocardial injection, carry a higher rate of arrhythmias. Other potential contributors to the arrhythmogenicity of cell transplantation include reentrant pathways caused by heterogeneity in conduction velocities between graft and host as well as graft automaticity. In this article, the arrhythmogenic potential of cell delivery to the heart is discussed.
Collapse
Affiliation(s)
- Shone O Almeida
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA
| | - Rhys J Skelton
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA; Murdoch Children's Research Institute, The Royal Children's Hospital, Cardiac Development, 50 Flemington Road, Parkville, Victoria 3052, Australia
| | - Sasikanth Adigopula
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA; Eli and Edyth Broad Stem Cell Research Center, University of California, 675 Charles E Young Drive South, MRL Room 3780, Los Angeles, CA 90095, USA.
| |
Collapse
|