1
|
Tan LT, Mokhtari-Esbuie F, Shababi N, Harmon JW. Stem Cell Therapy for Wound Healing in Ischemic Limbs: Is It Effective? Adv Surg 2024; 58:235-247. [PMID: 39089780 DOI: 10.1016/j.yasu.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Critical limb ischemia is an important clinical entity due to its association with increased morbidity and mortality. The mortality and amputation-free survival remains poor especially in those where revascularization is not an option. Recently, the role of cellular therapy has emerged as a promising therapeutic measure that may aid in wound healing and revascularization and improve functional outcomes.
Collapse
Affiliation(s)
- Li Ting Tan
- Department of Surgery, The Johns Hopkins Hospital, Blalock 658, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Farzad Mokhtari-Esbuie
- Department of Surgery, Johns Hopkins University, Surgery A Building 5th Floor, 4940 Eastern Avenue, Baltimore, MD 21224, USA
| | - Niloufar Shababi
- Department of Surgery, Johns Hopkins University, Surgery A Building 5th Floor, 4940 Eastern Avenue, Baltimore, MD 21224, USA
| | - John W Harmon
- Department of Surgery, Johns Hopkins University, Surgery A Building 5th Floor, 4940 Eastern Avenue, Baltimore, MD 21224, USA.
| |
Collapse
|
2
|
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
|
3
|
Nammian P, Asadi-Yousefabad SL, Daneshi S, Sheikhha MH, Tabei SMB, Razban V. Comparative analysis of mouse bone marrow and adipose tissue mesenchymal stem cells for critical limb ischemia cell therapy. Stem Cell Res Ther 2021; 12:58. [PMID: 33436054 PMCID: PMC7805174 DOI: 10.1186/s13287-020-02110-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Critical limb ischemia (CLI) is the most advanced form of peripheral arterial disease (PAD) characterized by ischemic rest pain and non-healing ulcers. Currently, the standard therapy for CLI is the surgical reconstruction and endovascular therapy or limb amputation for patients with no treatment options. Neovasculogenesis induced by mesenchymal stem cells (MSCs) therapy is a promising approach to improve CLI. Owing to their angiogenic and immunomodulatory potential, MSCs are perfect candidates for the treatment of CLI. The purpose of this study was to determine and compare the in vitro and in vivo effects of allogeneic bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue mesenchymal stem cells (AT-MSCs) on CLI treatment. METHODS For the first step, BM-MSCs and AT-MSCs were isolated and characterized for the characteristic MSC phenotypes. Then, femoral artery ligation and total excision of the femoral artery were performed on C57BL/6 mice to create a CLI model. The cells were evaluated for their in vitro and in vivo biological characteristics for CLI cell therapy. In order to determine these characteristics, the following tests were performed: morphology, flow cytometry, differentiation to osteocyte and adipocyte, wound healing assay, and behavioral tests including Tarlov, Ischemia, Modified ischemia, Function and the grade of limb necrosis scores, donor cell survival assay, and histological analysis. RESULTS Our cellular and functional tests indicated that during 28 days after cell transplantation, BM-MSCs had a great effect on endothelial cell migration, muscle restructure, functional improvements, and neovascularization in ischemic tissues compared with AT-MSCs and control groups. CONCLUSIONS Allogeneic BM-MSC transplantation resulted in a more effective recovery from critical limb ischemia compared to AT-MSCs transplantation. In fact, BM-MSC transplantation could be considered as a promising therapy for diseases with insufficient angiogenesis including hindlimb ischemia.
Collapse
Affiliation(s)
- Pegah Nammian
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sajad Daneshi
- Postdoctoral Researcher, Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hasan Sheikhha
- Biotechnology Research Center, International Campus, Shahid Sadoughi University of MedicalSciences, Yazd, Iran
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohammad Bagher Tabei
- Department of Genetics, Shiraz University of Medical Science, Shiraz, Iran.
- Maternal-fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Stem Cells Technology Research center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Leenstra B, Wijnand J, Verhoeven B, Koning O, Teraa M, Verhaar MC, de Borst GJ. Applicability of Transcutaneous Oxygen Tension Measurement in the Assessment of Chronic Limb-Threatening Ischemia. Angiology 2020; 71:208-216. [PMID: 31387360 PMCID: PMC6987479 DOI: 10.1177/0003319719866958] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transcutaneous oxygen tension measurement (TcPO2) is widely applied for the evaluation of chronic limb-threatening ischemia (CLTI). Nevertheless, studies that focused on the clinical value of TcPO2 have shown varying results. We identified factors that potentially play a role in TcPO2 measurement variation such as probe placement, probe temperature, and the use of a reference probe. In this review of the current literature, we assessed the application of these factors. A systematic search was conducted. Parameters that were assessed were probe placement, probe temperature, and mentioning and/or use of a reference probe. In total, 36 articles were eligible for analysis. In 24 (67%) studies, probes were placed on specific anatomical locations. Seven (19%) studies placed probes, regardless of the location of the ulcer, adjacent to an ischemic lesion or ulcer (perilesion). Selected temperature setting of the probe differed; in 18 (50%), a default probe temperature of 44°C was selected, and in 13 (36%), a different temperature was selected. In 31 (84%) studies, the use of a reference probe was not reported. Transcutaneous oxygen tension measurement is applied diversely in patients with CLTI. Homogeneity in TcPO2 protocols is warranted for reliable clinical application and to compare future TcPO2 research.
Collapse
Affiliation(s)
- Bernard Leenstra
- Department of Vascular Surgery, UMC Utrecht, CX Utrecht, the
Netherlands
| | - Joep Wijnand
- Department of Vascular Surgery, UMC Utrecht, CX Utrecht, the
Netherlands
| | - Bart Verhoeven
- Department of Vascular Surgery, Jeroen Bosch Ziekenhuis, Henri Dunantstraat,
GZ ‘s-Hertogenbosch, the Netherlands
| | - Olivier Koning
- Department of Vascular Surgery, Jeroen Bosch Ziekenhuis, Henri Dunantstraat,
GZ ‘s-Hertogenbosch, the Netherlands
| | - Martin Teraa
- Department of Vascular Surgery, UMC Utrecht, CX Utrecht, the
Netherlands
- Department of Vascular Surgery, Jeroen Bosch Ziekenhuis, Henri Dunantstraat,
GZ ‘s-Hertogenbosch, the Netherlands
- Department of Nephrology and Hypertension, UMC Utrecht, Heidelberglaan, CX
Utrecht, the Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, UMC Utrecht, Heidelberglaan, CX
Utrecht, the Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery, UMC Utrecht, CX Utrecht, the
Netherlands
| |
Collapse
|
5
|
Gaspar D, Peixoto R, De Pieri A, Striegl B, Zeugolis DI, Raghunath M. Local pharmacological induction of angiogenesis: Drugs for cells and cells as drugs. Adv Drug Deliv Rev 2019; 146:126-154. [PMID: 31226398 DOI: 10.1016/j.addr.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/12/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
The past decades have seen significant advances in pro-angiogenic strategies based on delivery of molecules and cells for conditions such as coronary artery disease, critical limb ischemia and stroke. Currently, three major strategies are evolving. Firstly, various pharmacological agents (growth factors, interleukins, small molecules, DNA/RNA) are locally applied at the ischemic region. Secondly, preparations of living cells with considerable bandwidth of tissue origin, differentiation state and preconditioning are delivered locally, rarely systemically. Thirdly, based on the notion, that cellular effects can be attributed mostly to factors secreted in situ, the cellular secretome (conditioned media, exosomes) has come into the spotlight. We review these three strategies to achieve (neo)angiogenesis in ischemic tissue with focus on the angiogenic mechanisms they tackle, such as transcription cascades, specific signalling steps and cellular gases. We also include cancer-therapy relevant lymphangiogenesis, and shall seek to explain why there are often conflicting data between in vitro and in vivo. The lion's share of data encompassing all three approaches comes from experimental animal work and we shall highlight common technical obstacles in the delivery of therapeutic molecules, cells, and secretome. This plethora of preclinical data contrasts with a dearth of clinical studies. A lack of adequate delivery vehicles and standardised assessment of clinical outcomes might play a role here, as well as regulatory, IP, and manufacturing constraints of candidate compounds; in addition, completed clinical trials have yet to reveal a successful and efficacious strategy. As the biology of angiogenesis is understood well enough for clinical purposes, it will be a matter of time to achieve success for well-stratified patients, and most probably with a combination of compounds.
Collapse
Affiliation(s)
- Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rita Peixoto
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Britta Striegl
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland.
| |
Collapse
|
6
|
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: 22] [Impact Index Per Article: 3.1] [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
|
7
|
Liotta F, Annunziato F, Castellani S, Boddi M, Alterini B, Castellini G, Mazzanti B, Cosmi L, Acquafresca M, Bartalesi F, Dilaghi B, Dorigo W, Graziani G, Bartolozzi B, Bellandi G, Carli G, Bartoloni A, Fargion A, Fassio F, Fontanari P, Landini G, Lucente EAM, Michelagnoli S, Orsi Battaglini C, Panigada G, Pigozzi C, Querci V, Santarlasci V, Parronchi P, Troisi N, Baggiore C, Romagnani P, Mannucci E, Saccardi R, Pratesi C, Gensini G, Romagnani S, Maggi E. Therapeutic Efficacy of Autologous Non-Mobilized Enriched Circulating Endothelial Progenitors in Patients With Critical Limb Ischemia - The SCELTA Trial. Circ J 2018; 82:1688-1698. [PMID: 29576595 DOI: 10.1253/circj.cj-17-0720] [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] [Indexed: 11/09/2022]
Abstract
BACKGROUND The therapeutic efficacy of bone marrow mononuclear cells (BM-MNC) autotransplantation in critical limb ischemia (CLI) has been reported. Variable proportions of circulating monocytes express low levels of CD34 (CD14+CD34lowcells) and behave in vitro as endothelial progenitor cells (EPCs). The aim of the present randomized clinical trial was to compare the safety and therapeutic effects of enriched circulating EPCs (ECEPCs) with BM-MNC administration. METHODS AND RESULTS ECEPCs (obtained from non-mobilized peripheral blood by immunomagnetic selection of CD14+and CD34+cells) or BM-MNC were injected into the gastrocnemius of the affected limb in 23 and 17 patients, respectively. After a mean of 25.2±18.6-month follow-up, both groups showed significant and progressive improvement in muscle perfusion (primary endpoint), rest pain, consumption of analgesics, pain-free walking distance, wound healing, quality of life, ankle-brachial index, toe-brachial index, and transcutaneous PO2. In ECEPC-treated patients, there was a positive correlation between injected CD14+CD34lowcell counts and the increase in muscle perfusion. The safety profile was comparable between the ECEPC and BM-MNC treatment arms. In both groups, the number of deaths and major amputations was lower compared with eligible untreated patients and historical reference patients. CONCLUSIONS This study supports previous trials showing the efficacy of BM-MNC autotransplantation in CLI patients and demonstrates comparable therapeutic efficacy between BM-MNC and EPEPCs.
Collapse
Affiliation(s)
- Francesco Liotta
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Francesco Annunziato
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Sergio Castellani
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Maria Boddi
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | | | | | | | - Lorenzo Cosmi
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | | | | | | | | | | | | | | | - Giulia Carli
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | | | | | | | | | | | | | | | - Carolina Orsi Battaglini
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | | | | | - Valentina Querci
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Veronica Santarlasci
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Paola Parronchi
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | | | | | - Paola Romagnani
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | | | | | | | - Gianfranco Gensini
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| | - Enrico Maggi
- Careggi University Hospital
- Department of Experimental and Clinical Medicine, Center of Excellence Denothe, University of Florence
| |
Collapse
|
8
|
Orekhov PY, Konoplyannikov MA, Baklaushev VP, Kalsin VAA, Averyanov AV, Konopliannikov AG, Habazov RI, Troitskiy AV. Bone marrow stem cells for the critical limb ischemia treatment: biological aspects and clinical application. GENES & CELLS 2018; 13:20-34. [DOI: 10.23868/201805002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
Cell therapy is one of the most promising directions in the treatment of critical limb ischemia (CLI). In spite of certain advances achieved in this field in the last decades, which are related to application of bone marrow stem cells (BMSC), a large number of problems still remain unsolved. In this review, we discuss the BMSC biology, mechanisms of their therapeutic effect in the CLI treatment and results of the most notable BMSC-based clinical studies in detail.
Collapse
|
9
|
Samura M, Hosoyama T, Takeuchi Y, Ueno K, Morikage N, Hamano K. Therapeutic strategies for cell-based neovascularization in critical limb ischemia. J Transl Med 2017; 15:49. [PMID: 28235425 PMCID: PMC5324309 DOI: 10.1186/s12967-017-1153-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022] Open
Abstract
Critical limb ischemia (CLI) causes severe ischemic rest pain, ulcer, and gangrene in the lower limbs. In spite of angioplasty and surgery, CLI patients without suitable artery inflow or enough vascular bed in the lesions are often forced to undergo amputation of a major limb. Cell-based therapeutic angiogenesis has the potential to treat ischemic lesions by promoting the formation of collateral vessel networks and the vascular bed. Peripheral blood mononuclear cells and bone marrow-derived mononuclear cells are the most frequently employed cell types in CLI clinical trials. However, the clinical outcomes of cell-based therapeutic angiogenesis using these cells have not provided the promised benefits for CLI patients, reinforcing the need for novel cell-based therapeutic angiogenesis strategies to cure untreatable CLI patients. Recent studies have demonstrated the possible enhancement of therapeutic efficacy in ischemic diseases by preconditioned graft cells. Moreover, judging from past clinical trials, the identification of adequate transplant timing and responders to cell-based therapy is important for improving therapeutic outcomes in CLI patients in clinical settings. Thus, to establish cell-based therapeutic angiogenesis as one of the most promising therapeutic strategies for CLI patients, its advantages and limitations should be taken into account.
Collapse
Affiliation(s)
- Makoto Samura
- Division of Vascular Surgery, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tohru Hosoyama
- Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan. .,Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan. .,Center for Regenerative Medicine, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Yuriko Takeuchi
- Division of Vascular Surgery, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Noriyasu Morikage
- Division of Vascular Surgery, Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
10
|
Are Endothelial Progenitor Cells the Real Solution for Cardiovascular Diseases? Focus on Controversies and Perspectives. BIOMED RESEARCH INTERNATIONAL 2015; 2015:835934. [PMID: 26509164 PMCID: PMC4609774 DOI: 10.1155/2015/835934] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/19/2015] [Accepted: 07/15/2015] [Indexed: 12/20/2022]
Abstract
Advanced knowledge in the field of stem cell biology and their ability to provide a cue for counteracting several diseases are leading numerous researchers to focus their attention on “regenerative medicine” as possible solutions for cardiovascular diseases (CVDs). However, the lack of consistent evidence in this arena has hampered the clinical application. The same condition affects the research on endothelial progenitor cells (EPCs), creating more confusion than comprehension. In this review, this aspect is discussed with particular emphasis. In particular, we describe biology and physiology of EPCs, outline their clinical relevance as both new predictive, diagnostic, and prognostic CVD biomarkers and therapeutic agents, discuss advantages, disadvantages, and conflicting data about their use as possible solutions for vascular impairment and clinical applications, and finally underline a very crucial aspect of EPCs “characterization and definition,” which seems to be the real cause of large heterogeneity existing in literature data on this topic.
Collapse
|
11
|
Cooke JP, Losordo DW. Modulating the vascular response to limb ischemia: angiogenic and cell therapies. Circ Res 2015; 116:1561-78. [PMID: 25908729 PMCID: PMC4869986 DOI: 10.1161/circresaha.115.303565] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/31/2015] [Indexed: 12/29/2022]
Abstract
The age-adjusted prevalence of peripheral arterial disease in the US population has been estimated to approach 12%. The clinical consequences of occlusive peripheral arterial disease include pain on walking (claudication), pain at rest, and loss of tissue integrity in the distal limbs; the latter may ultimately lead to amputation of a portion of the lower extremity. Surgical bypass techniques and percutaneous catheter-based interventions may successfully reperfuse the limbs of certain patients with peripheral arterial disease. In many patients, however, the anatomic extent and distribution of arterial occlusion is too severe to permit relief of pain and healing of ischemic ulcers. No effective medical therapy is available for the treatment of such patients, for many of whom amputation represents the only hope for alleviation of symptoms. The ultimate failure of medical treatment and procedural revascularization in significant numbers of patients has led to attempts to develop alternative therapies for ischemic disease. These strategies include administration of angiogenic cytokines, either as recombinant protein or as gene therapy, and more recently, to investigations of stem/progenitor cell therapy. The purpose of this review is to provide an outline of the preclinical basis for angiogenic and stem cell therapies, review the clinical research that has been done, summarize the lessons learned, identify gaps in knowledge, and suggest a course toward successfully addressing an unmet medical need in a large and growing patient population.
Collapse
Affiliation(s)
- John P Cooke
- From the Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.); and NeoStem Inc, New York, NY (D.W.L.).
| | - Douglas W Losordo
- From the Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.); and NeoStem Inc, New York, NY (D.W.L.).
| |
Collapse
|
12
|
|
13
|
Hassan WU, Greiser U, Wang W. Role of adipose-derived stem cells in wound healing. Wound Repair Regen 2015; 22:313-25. [PMID: 24844331 DOI: 10.1111/wrr.12173] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 03/01/2014] [Indexed: 12/11/2022]
Abstract
Impaired wound healing remains a challenge to date and causes debilitating effects with tremendous suffering. Recent advances in tissue engineering approaches in the area of cell therapy have provided promising treatment options to meet the challenges of impaired skin wound healing such as diabetic foot ulcers. Over the last few years, stem cell therapy has emerged as a novel therapeutic approach for various diseases including wound repair and tissue regeneration. Several different types of stem cells have been studied in both preclinical and clinical settings such as bone marrow-derived stem cells, adipose-derived stem cells (ASCs), circulating angiogenic cells (e.g., endothelial progenitor cells), human dermal fibroblasts, and keratinocytes for wound healing. Adipose tissue is an abundant source of mesenchymal stem cells, which have shown an improved outcome in wound healing studies. ASCs are pluripotent stem cells with the ability to differentiate into different lineages and to secrete paracrine factors initiating tissue regeneration process. The abundant supply of fat tissue, ease of isolation, extensive proliferative capacities ex vivo, and their ability to secrete pro-angiogenic growth factors make them an ideal cell type to use in therapies for the treatment of nonhealing wounds. In this review, we look at the pathogenesis of chronic wounds, role of stem cells in wound healing, and more specifically look at the role of ASCs, their mechanism of action and their safety profile in wound repair and tissue regeneration.
Collapse
Affiliation(s)
- Waqar Ul Hassan
- Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | | | | |
Collapse
|
14
|
Kwon SM, Lee JH, Lee SH, Jung SY, Kim DY, Kang SH, Yoo SY, Hong JK, Park JH, Kim JH, Kim SW, Kim YJ, Lee SJ, Kim HG, Asahara T. Cross talk with hematopoietic cells regulates the endothelial progenitor cell differentiation of CD34 positive cells. PLoS One 2014; 9:e106310. [PMID: 25166961 PMCID: PMC4148437 DOI: 10.1371/journal.pone.0106310] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/29/2014] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Despite the crucial role of endothelial progenitor cells (EPCs) in vascular regeneration, the specific interactions between EPCs and hematopoietic cells remain unclear. METHODS In EPC colony forming assays, we first demonstrated that the formation of EPC colonies was drastically increased in the coculture of CD34+ and CD34- cells, and determined the optimal concentrations of CD34+ cells and CD34- cells for spindle-shaped EPC differentiation. RESULTS Functionally, the coculture of CD34+ and CD34- cells resulted in a significant enhancement of adhesion, tube formation, and migration capacity compared with culture of CD34+ cells alone. Furthermore, blood flow recovery and capillary formation were remarkably increased by the coculture of CD34+ and CD34- cells in a murine hind-limb ischemia model. To elucidate further the role of hematopoietic cells in EPC differentiation, we isolated different populations of hematopoietic cells. T lymphocytes (CD3+) markedly accelerated the early EPC status of CD34+ cells, while macrophages (CD11b+) or megakaryocytes (CD41+) specifically promoted large EPC colonies. CONCLUSION Our results suggest that specific populations of hematopoietic cells play a role in the EPC differentiation of CD34+ cells, a finding that may aid in the development of a novel cell therapy strategy to overcome the quantitative and qualitative limitations of EPC therapy.
Collapse
Affiliation(s)
- Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
- * E-mail: (SMK); (TA)
| | - Jun-Hee Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sang-Hun Lee
- Soonchunhyang Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Seok-Yun Jung
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Da-Yeon Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Song-Hwa Kang
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - So-Young Yoo
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Jong-Kyu Hong
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Ji-Hye Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Jung-Hee Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sung-Wook Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yeon-Ju Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sun-Jin Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Hwi-Gon Kim
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Takayuki Asahara
- Department Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
- * E-mail: (SMK); (TA)
| |
Collapse
|
15
|
Park IS, Kang JA, Kang J, Rhie JW, Kim SH. Therapeutic Effect of Human Adipose-Derived Stromal Cells Cluster in Rat Hind-Limb Ischemia. Anat Rec (Hoboken) 2014; 297:2289-98. [DOI: 10.1002/ar.22961] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 11/10/2022]
Affiliation(s)
- In-Su Park
- Center for Biomaterials, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
| | - Jo A. Kang
- Department of Plastic Surgery, College of Medicine; The Catholic University of Korea; Seoul Korea
| | - Jungmi Kang
- Center for Biomaterials, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
- Department of Biomedical Engineering; University of Science and Technology; Seoul Korea
| | - Jong-Won Rhie
- Department of Plastic Surgery, College of Medicine; The Catholic University of Korea; Seoul Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute; Korea Institute of Science and Technology; Seoul Korea
- Department of Biomedical Engineering; University of Science and Technology; Seoul Korea
| |
Collapse
|
16
|
Choksi K, Dawn B. Priming mononuclear cells to improve outcomes of regenerative therapy. J Am Heart Assoc 2014; 3:e001168. [PMID: 24965029 PMCID: PMC4309101 DOI: 10.1161/jaha.114.001168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Kashyap Choksi
- Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS (K.C., B.D.)
| | - Buddhadeb Dawn
- Division of Cardiovascular Diseases and the Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS (K.C., B.D.)
| |
Collapse
|
17
|
Abstract
Thromboangiitis obliterans (TAO) is nonatherosclerotic inflammatory disease of the peripheral blood vessels, and TAO affects the small and medium sized vessels of the extremities. TAO is mainly seen in young males who smoke, and smoking is strongly associated with the disease course and progression. The diagnosis is made on the basis of the history, the physical examination and the clinical diagnostic criteria. As the bedrock for treating patients with TAO, absolute abstinence from tobacco is most important factor, and patients with TAO are usually managed conservatively. Surgical bypass or endovascular therapy is usually not possible for patients with TAO because of the diffuse segmental involvement and the distal nature of the disease. Therefore, stem cell therapy is considered to be a novel therapeutic modality for treating patients with TAO and who are not eligible for conventional revascularization therapies. In this paper, I have summarized the recent knowledge about TAO and I have reviewed the recent studies that have focused on the treatment of TAO.
Collapse
Affiliation(s)
- Ui-Jun Park
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
19
|
Lee KB, Kang ES, Kim AK, Kim MH, Do YS, Park KB, Park HS, Um SH, Cho SW, Kim DI. Stem cell therapy in patients with thromboangiitis obliterans: assessment of the long-term clinical outcome and analysis of the prognostic factors. Int J Stem Cells 2013; 4:88-98. [PMID: 24298340 DOI: 10.15283/ijsc.2011.4.2.88] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2011] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The clinical benefits of stem cell therapy have been reported in patients with peripheral arterial occlusive disease. However, those studies had no standard reporting system to assess the outcomes, so we made a scoring system and assessed the outcomes of the limbs that underwent whole bone marrow stem cell (WBMSC) therapy. METHODS AND RESULTS Between July 4 and June 2009, 90 limbs of 67 patients with symptomatic thromboangiitis obliterans (TAO) were enrolled. Autologous whole bone marrow was implanted into the limb by intramuscular injections. The primary outcomes were defined by the clinical and angiographic improvement in all the limbs and the secondary outcomes were the clinical improvement and the amputation-free rates in the critical ischemic limbs (CILs). Clinical improvement and angiographic improvement was observed in 55.6% and 43.2% of all the limbs and in 50% and 50%of the CILs, respectively. The 1, 3 and 5-year amputation-free rates were 91.9%, 88.5% and 84.6% for all the limbs, respectively, and 83.9%, 77.5% and 70.4% for the CILs, respectively. A history of sympathectomy/sympathetic block was shown to be a negative prognostic factor for clinical improvement in all the limbs and in the CILs. In addition, a history of sympathetic block/sympathectomy and the smoking state were the major predictors of amputation for the CILs. CONCLUSIONS This study indicated that autologous WMBSC therapy improves the clinical status and reduces amputation factors in the limbs with symptomatic TAO and a history of sympathetic block/sympathectomy and the smoking state are useful prognostic factors.
Collapse
|
20
|
Gili M, Orsello A, Gallo S, Brizzi MF. Diabetes-associated macrovascular complications: cell-based therapy a new tool? Endocrine 2013; 44:557-75. [PMID: 23543434 DOI: 10.1007/s12020-013-9936-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/20/2013] [Indexed: 01/01/2023]
Abstract
Diabetes mellitus and its ongoing macrovascular complications represent one of the major health problems around the world. Rise in obesity and population ages correlate with the increased incidence of diabetes. This highlights the need for novel approaches to prevent and treat this pandemic. The discovery of a reservoir of stem/progenitors in bone marrow and in mesenchymal tissue has attracted interest of both biologists and clinicians. A number of preclinical and clinical trials were developed to explore their potential clinical impact, as target or vehicle, in different clinical settings, including diabetes complications. Currently, bone marrow, peripheral blood, mesenchymal, and adipose tissues have been used as stem/progenitor cell sources. However, evidences have been provided that both bone marrow and circulating progenitor cells are dysfunctional in diabetes. These observations along with the growing advantages in genetic manipulation have spurred researchers to exploit ex vivo manipulated cells to overcome these hurdles. In this article, we provide an overview of data relevant to stem-progenitors potential clinical application in revascularization and/or vascular repair. Moreover, the hurdles at using progenitor cells in diabetic patients will be also discussed.
Collapse
Affiliation(s)
- Maddalena Gili
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | | | | | | |
Collapse
|
21
|
Intramuscular transplantation and survival of freshly isolated bone marrow cells following skeletal muscle ischemia-reperfusion injury. J Trauma Acute Care Surg 2013; 75:S142-9. [PMID: 23883899 DOI: 10.1097/ta.0b013e31829ac1fa] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Delayed treatment cellular therapies offer an attractive means to treat extremity injuries involving acute skeletal muscle ischemia-reperfusion injury (I/R). Bone marrow is a rich source of stem and progenitor cells with the potential to improve skeletal muscle regeneration. The extent to which bone marrow cells (BMCs) may be useful for I/R is not known. The purposes of this study were twofold: (1) to evaluate BMC survival following intramuscular injection 0, 2, 7, and 14 days after injury and (2) to determine whether BMCs improve functional recovery following I/R. METHODS Magnetic-activated cell sorting was used to isolate lineage-negative (Lin⁻) BMCs and enrich for stem and progenitor cells. To evaluate in vivo cell survival following I/R, Lin⁻ BMCs were injected intramuscularly 0, 2, 7, and 14 days after I/R, and bioluminescent imaging was performed for up to 28 days after cell injections. To assess their ability to improve muscle regeneration, intramuscular injections were performed 2 days after injury, and in vivo muscle function was assessed 14 days later. RESULTS Lin⁻ BMCs survived throughout the study period regardless of the timing of delivery. Intramuscular injection of Lin⁻ BMCs did not improve maximal isometric torque (300 Hz); however, both saline-injected and Lin⁻ BMC-injected muscles exhibited an increase in the twitch-tetanus ratio, suggesting that damage incurred with the intramuscular injections may have had deleterious consequences for functional recovery. CONCLUSION Although BMCs injected intramuscularly survived cell transplantation, they failed to improve muscle function following I/R. The ability of BMCs to persist in injured muscle following I/R lends to the possibility that with further development, their full potential can be realized.
Collapse
|
22
|
Abdul Wahid SF, Ismail NA, Abdul Hamid MKA, Harunarashid H, Idris MAM, Muhamad NA, Lai NM. Different sources of autologous mononuclear cells and stem cells for critical lower limb ischaemia. Hippokratia 2013. [DOI: 10.1002/14651858.cd010747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- S Fadilah Abdul Wahid
- Universiti Kebangsaan Malaysia Medical Centre; Cell Therapy Center; Jalan Yaacob Latif Kuala Lumpur Malaysia 56000
- Universiti Kebangsaan Malaysia Medical Centre; Clinical Haematology & Stem Cell Transplantation Services, Department of Medicine; Kuala Lumpur Malaysia
| | - Nor Azimah Ismail
- Universiti Kebangsaan Malaysia Medical Centre; Cell Therapy Center; Jalan Yaacob Latif Kuala Lumpur Malaysia 56000
| | | | - Hanafiah Harunarashid
- Universiti Kebangsaan Malaysia Medical Centre; Unit of Vascular Surgery, Department of Surgery; Jalan Yaacob Latif Kuala Lumpur Kuala Lumpur Malaysia 56000
| | - Muhamad Azim Mohd Idris
- Universiti Kebangsaan Malaysia Medical Centre; Unit of Vascular Surgery, Department of Surgery; Jalan Yaacob Latif Kuala Lumpur Kuala Lumpur Malaysia 56000
| | - Nor Asiah Muhamad
- Institute for Medical Research; Medical Research Resource Centre; Kuala Lumpur Malaysia 50588
| | - Nai Ming Lai
- University of Malaya Medical Center; Department of Paediatrics, Paediatric and Child Health Research Group; Kuala Lumpur Malaysia 50603
| |
Collapse
|
23
|
Raval Z, Losordo DW. Cell therapy of peripheral arterial disease: from experimental findings to clinical trials. Circ Res 2013; 112:1288-302. [PMID: 23620237 PMCID: PMC3838995 DOI: 10.1161/circresaha.113.300565] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/28/2013] [Indexed: 12/19/2022]
Abstract
The age-adjusted prevalence of peripheral arterial disease in the US population was estimated to approach 12% in 1985, and as the population ages, the overall population having peripheral arterial disease is predicted to rise. The clinical consequences of occlusive peripheral arterial disease include intermittent claudication, that is, pain with walking, and critical limb ischemia (CLI), which includes pain at rest and loss of tissue integrity in the distal limbs, which may ultimately lead to amputation of a portion of the lower extremity. The risk factors for CLI are similar to those linked to coronary artery disease and include advanced age, smoking, diabetes mellitus, hyperlipidemia, and hypertension. The worldwide incidence of CLI was estimated to be 500 to 1000 cases per million people per year in 1991. The prognosis is poor for CLI subjects with advanced limb disease. One study of >400 such subjects in the United Kingdom found that 25% required amputation and 20% (including some subjects who had required amputation) died within 1 year. In the United States, ≈280 lower-limb amputations for ischemic disease are performed per million people each year. The first objective in treating CLI is to increase blood circulation to the affected limb. Theoretically, increased blood flow could be achieved by increasing the number of vessels that supply the ischemic tissue with blood. The use of pharmacological agents to induce new blood vessel growth for the treatment or prevention of pathological clinical conditions has been called therapeutic angiogenesis. Since the identification of the endothelial progenitor cell in 1997 by Asahara and Isner, the field of cell-based therapies for peripheral arterial disease has been in a state of continuous evolution. Here, we review the current state of that field.
Collapse
Affiliation(s)
- Zankhana Raval
- Department of Medicine, Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
24
|
Huang CY, Shih CM, Tsao NW, Lin YW, Huang PH, Wu SC, Lee AW, Kao YT, Chang NC, Nakagami H, Morishita R, Ou KL, Hou WC, Lin CY, Shyu KG, Lin FY. Dipeptidyl peptidase-4 inhibitor improves neovascularization by increasing circulating endothelial progenitor cells. Br J Pharmacol 2013; 167:1506-19. [PMID: 22788747 DOI: 10.1111/j.1476-5381.2012.02102.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Current methods used to treat critical limb ischaemia (CLI) are hampered by a lack of effective strategies, therefore, therapeutic vasculogenesis may open up a new field for the treatment of CLI. In this study we investigated the ability of the DPP-4 inhibitor, sitagliptin, originally used as a hypoglycaemic agent, to induce vasculogenesis in vivo. EXPERIMENTAL APPROACH Sitagliptin were administered daily to C57CL/B6 mice and eGFP transgenic mouse bone marrow-transplanted ICR mice that had undergone hindlimb ischaemic surgery. Laser Doppler imaging and flow cytometry were used to evaluate the degree of neovasculogenesis and circulating levels of endothelial progenitor cells (EPCs) respectively. Cell surface markers of EPCs and endothelial NOS (eNOS) in vessels were studied. KEY RESULTS Sitagliptin elevated plasma glucagon-like peptide-1 (GLP-1) levels in mice subjected to ischaemia, decreased plasma dipeptidyl peptidase-4 (DPP-4) concentration, and augmented ischaemia-induced increases in stromal cell-derived factor-1 (SDF-1) in a dose-dependent manner. Blood flow in the ischaemic limb was significantly improved in mice treated with sitagliptin. Circulating levels of EPCs were also increased after sitagliptin treatment. Sitagliptin also enhanced the expression of CD 34 and eNOS in ischaemic muscle. In addition, sitagliptin promoted EPC mobilization and homing to ischaemic tissue in eGFP transgenic mouse bone marrow-transplanted ICR mice. CONCLUSION AND IMPLICATIONS Circulating EPC levels and neovasculogenesis were augmented by the DPP-4 inhibitor, sitagliptin and this effect was dependent on an eNOS-related pathway in a mouse model of hindlimb ischaemia. The results indicate that oral administration of sitagliptin has therapeutic potential as an inducer of vasculogenesis.
Collapse
Affiliation(s)
- Chun-Yao Huang
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Therapeutic angiogenesis for revascularization in peripheral artery disease. Gene 2013; 525:220-8. [PMID: 23566831 DOI: 10.1016/j.gene.2013.03.097] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/05/2013] [Accepted: 03/07/2013] [Indexed: 01/15/2023]
Abstract
Therapeutic angiogenesis for peripheral artery disease (PAD), achieved by gene and cell therapy, has recently raised a great deal of hope for patients who cannot undergo standard revascularizing treatment. Although pre-clinical studies gave very promising data, still clinical trials of gene therapy have not provided satisfactory results. On the other hand, cell therapy approach, despite several limitations, demonstrated more beneficial effects but initial clinical studies must be constantly validated by larger randomized, multi-center, double-blinded, placebo-controlled trials. This review focuses on previous and recent gene and cell therapy studies for limb ischemia, including both experimental and clinical research, and summarizes some important papers published in this field. Moreover, it provides a short comment on combined gene and cell therapy approach on the example of heme oxygenase-1 overexpressing cells with therapeutic properties.
Collapse
|
26
|
Szöke K, Brinchmann JE. Concise review: therapeutic potential of adipose tissue-derived angiogenic cells. Stem Cells Transl Med 2012. [PMID: 23197872 DOI: 10.5966/sctm.2012-0069] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inadequate blood supply to tissues is a leading cause of morbidity and mortality today. Ischemic symptoms caused by obstruction of arterioles and capillaries are currently not treatable by vessel replacement or dilatation procedures. Therapeutic angiogenesis, the treatment of tissue ischemia by promoting the proliferation of new blood vessels, has recently emerged as one of the most promising therapies. Neovascularization is most often attempted by introduction of angiogenic cells from different sources. Emerging evidence suggests that adipose tissue (AT) is an excellent reservoir of autologous cells with angiogenic potential. AT yields two cell populations of importance for neovascularization: AT-derived mesenchymal stromal cells, which likely act predominantly as pericytes, and AT-derived endothelial cells (ECs). In this concise review we discuss different physiological aspects of neovascularization, briefly present cells isolated from the blood and bone marrow with EC properties, and then discuss isolation and cell culture strategies, phenotype, functional capabilities, and possible therapeutic applications of angiogenic cells obtained from AT.
Collapse
|
27
|
Botti C, Maione C, Coppola A, Sica V, Cobellis G. Autologous bone marrow cell therapy for peripheral arterial disease. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2012; 5:5-14. [PMID: 24198534 PMCID: PMC3781761 DOI: 10.2147/sccaa.s28121] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inadequate blood supply to tissues caused by obstruction of arterioles and/or capillaries results in ischemic injuries – these injuries can range from mild (eg, leg ischemia) to severe conditions (eg, myocardial infarction, stroke). Surgical and/or endovascular procedures provide cutting-edge treatment for patients with vascular disorders; however, a high percentage of patients are currently not treatable, owing to high operative risk or unfavorable vascular involvement. Therapeutic angiogenesis has recently emerged as a promising new therapy, promoting the formation of new blood vessels by the introduction of bone marrow–derived stem and progenitor cells. These cells participate in the development of new blood vessels, the enlargement of existing blood vessels, and sprouting new capillaries from existing blood vessels, providing evidence of the therapeutic utility of these cells in ischemic tissues. In this review, the authors describe peripheral arterial disease, an ischemic condition affecting the lower extremities, summarizing different aspects of vascular regeneration and discussing which and how stem cells restore the blood flow. The authors also present an overview of encouraging results from early-phase clinical trials using stem cells to treat peripheral arterial disease. The authors believe that additional research initiatives should be undertaken to better identify the nature of stem cells and that an intensive cooperation between laboratory and clinical investigators is needed to optimize the design of cell therapy trials and to maximize their scientific rigor. Only this will allow the results of these investigations to develop best clinical practices. Additionally, although a number of stem cell therapies exist, many treatments are performed outside international and national regulations and many clinical trials have been not registered on databases such as ClinicalTrials.gov or EudraCT. Therefore, more rigorous clinical trials are required to confirm the first hopeful results and to address the challenging issues.
Collapse
Affiliation(s)
- C Botti
- Department of General Pathology, Second University of Naples, Naples, Italy
| | | | | | | | | |
Collapse
|
28
|
Klepanec A, Mistrik M, Altaner C, Valachovicova M, Olejarova I, Slysko R, Balazs T, Urlandova T, Hladikova D, Liska B, Tomka J, Vulev I, Madaric J. No Difference in Intra-Arterial and Intramuscular Delivery of Autologous Bone Marrow Cells in Patients with Advanced Critical Limb Ischemia. Cell Transplant 2012; 21:1909-18. [DOI: 10.3727/096368912x636948] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stem cell therapy has been proposed to be an alternative therapy in patients with critical limb ischemia (CLI), not eligible for endovascular or surgical revascularization. We compared the therapeutic effects of intramuscular (IM) and intra-arterial (IA) delivery of bone marrow cells (BMCs) and investigated the factors associated with therapeutic benefits. Forty-one patients (mean age, 66 ± 10 years; 35 males) with advanced CLI (Rutherford category, 5 and 6) not eligible for revascularization were randomized to treatment with 40 ml BMCs using local IM ( n = 21) or selective IA infusion ( n = 20). Primary endpoints were limb salvage and wound healing. Secondary endpoints were changes in transcutaneous oxygen pressure (tcpO2), quality-of-life questionnaire (EQ5D), ankle–brachial index (ABI), and pain scale (0–10). Patients with limb salvage and wound healing were considered to be responders to BMC therapy. At 6-month follow-up, overall limb salvage was 73% (27/37) and 10 subjects underwent major amputation. Four patients died unrelated to stem cell therapy. There was significant improvement in tcpO2 (15 ± 10 to 29 ± 13 mmHg, p < 0.001), pain scale (4.4 ± 2.6 to 0.9 ± 1.4, p < 0.001), and EQ5D (51 ± 15 to 70 ± 13, p < 0.001) and a significant decrease in the Rutherford category of CLI (5.0 ± 0.2 to 4.3 ± 1.6, p < 0.01). There were no differences among functional parameters in patients undergoing IM versus IA delivery. Responders ( n = 27) were characterized by higher CD34+ cell counts in the bone marrow concentrate (CD34+ 29 ± 15×106 vs. 17 ± 12×106, p < 0.05) despite a similar number of total nucleated cells (4.3 ± 1.4×109 vs. 4.1 ± 1.2×109, p = 0.66) and by a lower level of C-reactive protein (18 ± 28 vs. 100 ± 96 mg/L, p < 0.05) as well as serum leukocytes (8.3 ± 2.1×109/L vs. 12.3 ± 4.5×109/L, p < 0.05) as compared with nonresponders (10 patients). Both IM and IA delivery of autologous stem cells are effective therapeutic strategies in patients with CLI. A higher concentration of CD34+ cells and a lower degree of inflammation are associated with better clinical therapeutic responses.
Collapse
Affiliation(s)
- Andrej Klepanec
- Slovak Medical University, Bratislava, Slovakia
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Martin Mistrik
- Clinic of Haematology and Transfusiology, Faculty Hospital, Bratislava, Slovakia
| | - Cestmir Altaner
- Institute of Experimental Oncology, Slovak Academy of Science, Bratislava, Slovakia
| | | | | | - Roman Slysko
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Tibor Balazs
- National Cardiovascular Institute, Bratislava, Slovakia
| | | | | | | | - Jan Tomka
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Ivan Vulev
- Slovak Medical University, Bratislava, Slovakia
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Juraj Madaric
- Slovak Medical University, Bratislava, Slovakia
- National Cardiovascular Institute, Bratislava, Slovakia
| |
Collapse
|
29
|
Teng M, Geng Z, Huang L, Zhao X. Stem cell transplantation in cardiovascular disease: an update. J Int Med Res 2012; 40:833-8. [PMID: 22906255 DOI: 10.1177/147323001204000301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Despite the development of novel therapeutic strategies, cardiovascular diseases remain the main cause of morbidity and mortality worldwide. Many phase 1 and 2 clinical trials have reported the safety, feasibility and promising potential of stem cell transplantation, however, the optimal cell types, timing of infusion, cell dosage and routes of administration remain to be determined. This paper reviews the findings of various clinical studies and discusses the challenges facing the delivery of stem cell therapy in cardiovascular diseases.
Collapse
Affiliation(s)
- M Teng
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | |
Collapse
|
30
|
Brenes RA, Jadlowiec CC, Bear M, Hashim P, Protack CD, Li X, Lv W, Collins MJ, Dardik A. Toward a mouse model of hind limb ischemia to test therapeutic angiogenesis. J Vasc Surg 2012; 56:1669-79; discussion 1679. [PMID: 22836102 DOI: 10.1016/j.jvs.2012.04.067] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 04/24/2012] [Accepted: 04/25/2012] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Several clinical trials are currently evaluating stem cell therapy for patients with critical limb ischemia that have no other surgical or endovascular options for revascularization. However, these trials are conducted with different protocols, including use of different stem cell populations and different injection protocols, providing little means to compare trials and guide therapy. Accordingly, we developed a murine model of severe ischemia to allow methodic testing of relevant clinical parameters. METHODS High femoral artery ligation and total excision of the superficial femoral artery was performed on C57BL/6 mice. Mononuclear cells (MNCs) were isolated from the bone marrow of donor mice, characterized using fluorescence-activated cell sorting, and injected (5×10(5) to 2×10(6)) into the semimembranosus (proximal) or gastrocnemius (distal) muscle. Vascular and functional outcomes were measured using invasive Doppler imaging, laser Doppler perfusion imaging, and the Tarlov and ischemia scores. Histologic analysis included quantification of muscle fiber area and number as well as capillary density. RESULTS Blood flow and functional outcomes were improved in MNC-treated mice compared with controls over 28 days (flow: P<.0001; Tarlov: P=.0004; ischemia score: P=.0002). MNC-treated mice also showed greater gastrocnemius fiber area (P=.0053) and increased capillary density (P=.0004). Dose-response analysis showed increased angiogenesis and gastrocnemius fiber area but no changes in macroscopic vascular flow or functional scores. Overall functional outcomes in mice injected proximally to the ischemic area were similar to mice injected more distally, but muscle flow, capillary density, and gastrocnemius fiber area were increased (P<.05). CONCLUSIONS High femoral ligation with complete excision of the superficial femoral artery is a reliable model of severe hind limb ischemia in C57BL/6 mice that shows a response to MNC treatment for functional and vascular outcomes. A dose response to the injection of MNCs appears to be present, at least microscopically, suggesting that an optimal cell number for stem cell therapy exists and that preclinical testing needs to be performed to optimally guide human trials. Injection of MNCs proximal to the site of ischemia may provide different outcomes compared with distal injection and warrants additional study.
Collapse
Affiliation(s)
- Robert A Brenes
- Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT 06520-8089, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Schiavetta A, Maione C, Botti C, Marino G, Lillo S, Garrone A, Lanza L, Pagliari S, Silvestroni A, Signoriello G, Sica V, Cobellis G. A phase II trial of autologous transplantation of bone marrow stem cells for critical limb ischemia: results of the Naples and Pietra Ligure Evaluation of Stem Cells study. Stem Cells Transl Med 2012. [PMID: 23197862 DOI: 10.5966/sctm.2012-0021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Critical limb ischemia (CLI) is a vascular disease affecting lower limbs, which is going to become a demanding challenge because of the aging of the population. Despite advances in endovascular therapies, CLI is associated with high morbidity and mortality. Patients without direct revascularization options have the worst outcomes. To date, 25%-40% of CLI patients are not candidates for surgical or endovascular approaches, ultimately facing the possibility of a major amputation. This study aimed to assess the safety and efficacy of autologous bone marrow (BM) transplantation performed in "no-option" patients, in terms of restoring blood perfusion by collateral flow and limb salvage. A multicenter, prospective, not-controlled phase II study for no-option CLI patients was performed. Patients were subjected to intra-arterial infusion of autologous bone marrow and followed for 12 months after the treatment. Variation of blood perfusion parameters, evaluated by laser Doppler flowmetry or transcutaneous oximetry, was set as the primary endpoint at 12 months after treatment and amputation-free survival as the secondary endpoint. Sixty patients were enrolled and treated with BM transplantation, showing improvement in objective and subjective measures of perfusion. Furthermore, survival analysis demonstrated improved amputation-free survival rates (75.2%) at 12 months after the treatment. This study provides further evidence that autologous bone marrow transplantation is well tolerated by CLI patients without adverse effects, demonstrating trends toward improvement in perfusion and reduced amputation rate, confirming the feasibility and safety of the procedure.
Collapse
Affiliation(s)
- Alessandro Schiavetta
- Department of Vascular Surgery, Azienda Ospedaliera Santa Corona, Pietra Ligure, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Adams V, Heiker JT, Höllriegel R, Beck EB, Woitek FJ, Erbs S, Blüher M, Stumvoll M, Beck-Sickinger AG, Schuler G, Linke A. Adiponectin promotes the migration of circulating angiogenic cells through p38-mediated induction of the CXCR4 receptor. Int J Cardiol 2012; 167:2039-46. [PMID: 22682478 DOI: 10.1016/j.ijcard.2012.05.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 04/09/2012] [Accepted: 05/11/2012] [Indexed: 01/21/2023]
Abstract
AIMS Adiponectin (adipo) and exercise training (ET) contribute to the maintenance of a normal vascular tone by influencing vascular NO bioavailability and concentration and function of circulating angiogenic cells (CAC). The molecular mechanisms are only partially understood. Aim of the present study was to elucidate the effects of adipo on CAC migration and the underlying signaling pathways. Furthermore, the impact of ET on adiponectin-mediated CAC migration was investigated. METHODS AND RESULTS CACs were isolated from peripheral blood and exposed to different adipo concentrations. Adipo (5μg/ml) enhanced the ability of CACs to migrate following an SDF-1 gradient by 345%. This was associated with a significant increase in CXCR4 expression on the surface of CACs as compared to control (10.1 ± 1.5 vs. 33.2 ± 4.5% CXCR4 positive cells, p<0.05). Adiponectin-induced CAC migration and CXCR4-upregulation were mediated through adipo-receptor 1 (AdipoR1) and blocked by an inhibitor of PI3-kinase, p38MAP kinase and NFκb. Adipo-stimulated migration of CACs, CXCR4 expression and p38MAPK-activation is impaired in patients with coronary artery disease (CAD). ET over 4 weeks partially corrects adiponectin-stimulated CAC migration and CXCR4 expression in patients with CAD (n=10). No change was observed in the control group (n=10). CONCLUSION Adipo improves the migratory capacity of CACs in response to SDF1, partially through an upregulation of CXCR4. This is mediated through a pathway that involves binding of adipo to the AdipoR1 and subsequent PI3kinase/p38MAPK/ NFκb activation. In addition ET corrects the adiponectin responsiveness of CACs, and thereby might promote endogenous repair of damaged endothelium.
Collapse
Affiliation(s)
- Volker Adams
- Department of Internal Medicine/Cardiology, University Leipzig, Heart Center, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Benoit E, O'Donnell TF, Patel AN. Safety and efficacy of autologous cell therapy in critical limb ischemia: a systematic review. Cell Transplant 2012; 22:545-62. [PMID: 22490340 DOI: 10.3727/096368912x636777] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Researchers have accumulated a decade of experience with autologous cell therapy in the treatment of critical limb ischemia (CLI). We conducted a systematic review of clinical trials in the literature to determine the safety and efficacy of cell therapy in CLI. We searched the literature for clinical trials of autologous cell therapy in CLI, including observational series of five or more patients to accrue a large pool of patients for safety analysis. Safety analysis included evaluation of death, cancer, unregulated angiogenesis, and procedural adverse events such as bleeding. Efficacy analysis included the clinical endpoints amputation and death as well as functional and surrogate endpoints. We identified 45 clinical trials, including seven RCTs, and 1,272 patients who received cell therapy. The overall adverse event rate was low (4.2%). Cell therapy patients did not have a higher mortality rate than control patients and demonstrated no increase in cancer incidence when analyzed against population rates. With regard to efficacy, cell therapy patients had a significantly lower amputation rate than control patients (OR 0.36, p = 0.0004). Cell therapy also demonstrated efficacy in a variety of functional and surrogate outcomes. Clinical trials differed in the proportion of patients with risk factors for clinical outcomes, and these influenced rates of amputation and death. Cell therapy presents a favorable safety profile with a low adverse event rate and no increase in severe events such as mortality and cancer and treatment with cell therapy decreases the risk of amputation. Cell therapy has a positive benefit-to-risk ratio in CLI and may be a valuable treatment option, particularly for those challenging patients who cannot undergo arterial reconstruction.
Collapse
Affiliation(s)
- Eric Benoit
- Department of Surgery, Tufts Medical Center, Boston, MA, USA
| | | | | |
Collapse
|
34
|
Misra V, Lal A, El Khoury R, Chen PR, Savitz SI. Intra-arterial delivery of cell therapies for stroke. Stem Cells Dev 2012; 21:1007-15. [PMID: 22181047 DOI: 10.1089/scd.2011.0612] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cell therapy is a novel investigational approach to enhance stroke recovery. Intra-arterial (IA) delivery has the potential advantage of selectively targeting cell therapies to the ischemic brain tissue. Over the past 10 years, IA cell delivery has been under investigation in patients with cardiac and peripheral vascular disease, and these studies have reported promising results. This article reviews the trial methodology and procedural details of these studies and discusses the rationale and challenges in designing IA cell therapy trials for ischemic stroke.
Collapse
Affiliation(s)
- Vivek Misra
- Department of Neurology, University of Texas Medical School at Houston, UT-Health, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
35
|
Early results and lessons learned from a multicenter, randomized, double-blind trial of bone marrow aspirate concentrate in critical limb ischemia. J Vasc Surg 2011; 54:1650-8. [DOI: 10.1016/j.jvs.2011.06.118] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 06/28/2011] [Accepted: 06/30/2011] [Indexed: 11/23/2022]
|
36
|
Li JY, Su CH, Wu YJ, Tien TY, Hsieh CL, Chen CH, Tseng YM, Shi GY, Wu HL, Tsai CH, Lin FY, Yeh HI. Therapeutic Angiogenesis of Human Early Endothelial Progenitor Cells Is Enhanced by Thrombomodulin. Arterioscler Thromb Vasc Biol 2011; 31:2518-25. [DOI: 10.1161/atvbaha.111.235143] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
We examined the effect of thrombomodulin (TM) domains 2 and 3 (TMD23) on human early endothelial progenitor cells (EPCs).
Methods and Results—
TM was expressed and released by human EPCs cultured from peripheral blood mononuclear cells (PBMCs). Addition of TMD23 (100 ng/mL) to the cultured PBMCs increased the colony-forming units, chemotactic motility, matrix metalloproteinase activity, and interleukin-8 secretion but decreased tumor necrosis factor-α (TNF-α) release. Analysis of the signal pathway showed that TMD23 activated Akt. Inhibition of phosphatidylinositol-3 kinase–Akt blocked the effects of TMD23 on chemotactic motility, matrix metalloproteinase-9, interleukin-8, and TNF-α. In hindlimb ischemia mice, laser Doppler perfusion imaging of the ischemic limb during the 21 days after arterial ligation showed that the perfusion recovered best with intraperitoneal infusion of TMD23 plus local injection of early EPCs, followed by either infusion of TMD23 or injection of the cells. Animals without either treatment had the worst results. Animals treated with TMD23 also had lower circulating and tissue levels of TNF-α.
Conclusion—
TM is expressed and released by human circulating EPCs. Exogenous TMD23 enhances the angiogenic potential of early EPCs in vitro through activation of phosphatidylinositol-3 kinase-Akt pathway. Coadministration of TMD23 plus early EPCs augments therapeutic angiogenesis of the EPCs in ischemic tissues.
Collapse
Affiliation(s)
- Jiun-Yi Li
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Cheng-Huang Su
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Yih-Jer Wu
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Ting-Yi Tien
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Chin-Ling Hsieh
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Chih-Hao Chen
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Ya-Ming Tseng
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Guey-Yueh Shi
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Hua-Lin Wu
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Cheng-Ho Tsai
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Fang-Yue Lin
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| | - Hung-I Yeh
- From the Departments of Surgery (J.-Y.L.), Internal Medicine (C.-H.S., Y.-J.W., C.-H.T., H.-I.Y.), and Medical Research (C.-H.S., Y.-J.W., T.-Y.T., C.-L.H., C.-H.C., H.-I.Y.), Mackay Memorial Hospital, Mackay Medical College, New Taipei City, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan (C.-H.S., Y.-J.W., T.-Y.T., Y.-M.T., C.-H.T., H.-I.Y.); Graduate Institute of Clinical Medicine, National Taiwan University (J.-Y.L.), Taipei, Taiwan; Department of Biochemistry and
| |
Collapse
|
37
|
Chen XK, Rathbone CR, Walters TJ. Treatment of Tourniquet-Induced Ischemia Reperfusion Injury with Muscle Progenitor Cells. J Surg Res 2011; 170:e65-73. [DOI: 10.1016/j.jss.2011.05.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 05/20/2011] [Accepted: 05/27/2011] [Indexed: 10/18/2022]
|
38
|
Abstract
OBJECTIVES The concept of neovascularization in response to tissue ischemia was recently extended by the finding of postnatal vasculogenesis through circulating endothelial progenitor cells (EPCs). The aim of this study was to assess the role of acute ischemia for EPC mobilization in patients with peripheral arterial occlusive disease (PAOD) and in healthy volunteers. METHODS The number of circulating EPCs was analyzed by flow cytometry in PAOD patients (n = 23) with exercise-induced limb ischemia for up to 72 h after a maximal treadmill test and in healthy volunteers (n = 17) who underwent a 15-min suprasystolic occlusion of one lower extremity to induce limb ischemia. Plasma concentrations of vascular endothelial growth factor, basic fibroblast growth factor, tumor necrosis factor-α, and granulocyte macrophage-colony stimulating factor were determined by ELISA. RESULTS EPCs (CD 34 pos/KDRpos) increased significantly in both PAOD patients from 82 ± 20 to 256 ± 52 (P < 0.05) and healthy volunteers from 144 ± 39 to 590 ± 61 cells per 1 million events (P < 0.05) in response to induced ischemia, with a maximum after 24 h and returned to baseline within 72 h. The relative increase in EPC numbers was significantly lower in patients with PAOD as compared with healthy volunteers (P < 0.05). Plasma levels of vascular endothelial growth factor increased from 27.4 ± 3.1 to 126.4 ± 12 pg/ml in patients with PAOD (P < 0.05) and from 30.7 ± 6.1 to 134.1 ± 12.4 pg/ml in healthy volunteers (P < 0.05). CONCLUSION Both patients with symptomatic PAOD and healthy volunteers respond to a single episode of limb ischemia with a time-dependent increase in circulating EPCs. The increase of EPC numbers in response to ischemia is reduced when vascular disease is present, underlining the reduced vasculogenic potential of patients with PAOD.
Collapse
|
39
|
From bench to bedside: what physicians need to know about endothelial progenitor cells. Am J Med 2011; 124:489-97. [PMID: 21605723 DOI: 10.1016/j.amjmed.2011.01.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 01/31/2011] [Accepted: 01/31/2011] [Indexed: 12/17/2022]
Abstract
Since the discovery of endothelial progenitor cells in 1997, the scientific world has seen their ups and downs. There has been much discussion about the detection methods of endothelial progenitor cells and their diagnostic and predictive value. A lack of standardized methods to define endothelial progenitor cells has led to a number of nomenclatures and measuring methods that are difficult for clinicians to oversee. Therefore, only specialized hematologists and cardiologists were aware of their existence. Now it is time for a change: Most of the controversies have been eliminated by elaborate studies. This review aims to give an overview to the clinically working physician about the measurement, diagnostic potential, predictive value, and therapeutic potential of endothelial progenitor cells.
Collapse
|
40
|
Capobianco S, Chennamaneni V, Mittal M, Zhang N, Zhang C. Endothelial progenitor cells as factors in neovascularization and endothelial repair. World J Cardiol 2010; 2:411-20. [PMID: 21191542 PMCID: PMC3011136 DOI: 10.4330/wjc.v2.i12.411] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/20/2010] [Accepted: 09/27/2010] [Indexed: 02/06/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous population of cells that are provided by the bone marrow and other adult tissue in both animals and humans. They express both hematopoietic and endothelial surface markers, which challenge the classic dogma that the presumed differentiation of cells into angioblasts and subsequent endothelial and vascular differentiation occurred exclusively in embryonic development. This breakthrough stimulated research to understand the mechanism(s) underlying their physiologic function to allow development of new therapeutic options. One focus has been on their ability to form new vessels in injured tissues, and another has been on their ability to repair endothelial damage and restore both monolayer integrity and endothelial function in denuded vessels. Moreover, measures of their density have been shown to be a better predictor of cardiovascular events, both in healthy and coronary artery disease populations than the classical tools used in the clinic to evaluate the risk stratification. In the present paper we review the effects of EPCs on revascularization and endothelial repair in animal models and human studies, in an attempt to better understand their function, which may lead to potential advancement in clinical management.
Collapse
Affiliation(s)
- Stefano Capobianco
- Stefano Capobianco, Department of Cardiology, Gaetano Rummo Hospital, Via Dell'Angelo 1, 82100 Benevento, Italy
| | | | | | | | | |
Collapse
|
41
|
Lawall H, Bramlage P, Amann B. Treatment of peripheral arterial disease using stem and progenitor cell therapy. J Vasc Surg 2010; 53:445-53. [PMID: 21030198 DOI: 10.1016/j.jvs.2010.08.060] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 08/18/2010] [Accepted: 08/21/2010] [Indexed: 10/18/2022]
Abstract
Peripheral arterial disease (PAD) is a highly prevalent atherosclerotic syndrome associated with significant morbidity and mortality. PAD is most commonly caused by atherosclerosis obliterans (ASO) and thromboangiitis obliterans (TAO), and can lead to claudication and critical limb ischemia (CLI), often resulting in a need for major amputation and subsequent death. Standard treatment for such severe cases of PAD is surgical or endovascular revascularization. However, up to 30% of patients are not candidates for such interventions, due to high operative risk or unfavorable vascular involvement. Therefore, new strategies are needed to offer these patients a viable therapeutic option. Bone-marrow derived stem and progenitor cells have been identified as a potential new therapeutic option to induce angiogenesis. These findings prompted clinical researchers to explore the feasibility of cell therapies in patients with peripheral and coronary artery disease in several small trials. Clinical benefits were reported from these trials including improvement of ankle-brachial index (ABI), transcutaneous partial pressure of oxygen (TcO(2)), reduction of pain, and decreased need for amputation. Nonetheless, large randomized, placebo-controlled, double-blind studies are necessary and currently ongoing to provide stronger safety and efficacy data on cell therapy. Current literature is supportive of intramuscular bone marrow cell administration as a relatively safe, feasible, and possibly effective therapy for patients with PAD who are not subjects for conventional revascularization.
Collapse
Affiliation(s)
- Holger Lawall
- SRH-Klinikum Karlsbad-Langensteinbach, Angiology/Diabetology, Guttmannnstraβe 1, Karlsbad, Germany.
| | | | | |
Collapse
|
42
|
Rationale and design of the JUVENTAS trial for repeated intra-arterial infusion of autologous bone marrow-derived mononuclear cells in patients with critical limb ischemia. J Vasc Surg 2010; 51:1564-8. [PMID: 20488328 DOI: 10.1016/j.jvs.2010.02.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 02/04/2010] [Accepted: 02/06/2010] [Indexed: 11/24/2022]
Abstract
Critical limb ischemia (CLI) continues to form a substantial burden on Western healthcare. Many patients still face amputation as a last treatment option. Autologous bone marrow (BM)-derived cell administration has emerged as a potential new treatment, but proof for sustainable clinical effects of BM-derived cell therapy in CLI is still lacking. The JUVENTAS (reJUVenating ENdothelial progenitor cells via Transcutaneous intra-Arterial Supplementation) trial is the first randomized, placebo-controlled, double-blinded clinical trial on repeated intra-arterial BM mononuclear cell (MNC) infusion in 110 to 160 CLI patients, designed to provide definite proof for the efficacy of stem cell therapy. Primary outcome is the incidence of major amputation at 6 months. Inclusion of patients is well underway. If BM-MNC cells therapy is beneficial, it could become a novel treatment to prevent amputation in patients with CLI.
Collapse
|
43
|
Abstract
Critical limb ischemia (CLI), defined as chronic ischemic rest pain, ulcers, or gangrene attributable to objectively proven arterial occlusive disease, is the most advanced form of peripheral arterial disease. Traditionally, open surgical bypass was the only effective treatment strategy for limb revascularization in this patient population. However, during the past decade, the introduction and evolution of endovascular procedures have significantly increased treatment options. In a certain subset of patients for whom either surgical or endovascular revascularization may not be appropriate, primary amputation remains a third treatment option. Definitive high-level evidence on which to base treatment decisions, with an emphasis on clinical and cost effectiveness, is still lacking. Treatment decisions in CLI are individualized, based on life expectancy, functional status, anatomy of the arterial occlusive disease, and surgical risk. For patients with aortoiliac disease, endovascular therapy has become first-line therapy for all but the most severe patterns of occlusion, and aortofemoral bypass surgery is a highly effective and durable treatment for the latter group. For infrainguinal disease, the available data suggest that surgical bypass with vein is the preferred therapy for CLI patients likely to survive 2 years or more, and for those with long segment occlusions or severe infrapopliteal disease who have an acceptable surgical risk. Endovascular therapy may be preferred in patients with reduced life expectancy, those who lack usable vein for bypass or who are at elevated risk for operation, and those with less severe arterial occlusions. Patients with unreconstructable disease, extensive necrosis involving weight-bearing areas, nonambulatory status, or other severe comorbidities may be considered for primary amputation or palliative measures.
Collapse
Affiliation(s)
- Andres Schanzer
- University of Massachusetts-Memorial Medical Center, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Michael S. Conte
- Division of Vascular and Endovascular Surgery, Heart and Vascular Center, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA 94143 USA
| |
Collapse
|
44
|
Lasala GP, Silva JA, Gardner PA, Minguell JJ. Combination Stem Cell Therapy for the Treatment of Severe Limb Ischemia: Safety and Efficacy Analysis. Angiology 2010; 61:551-6. [DOI: 10.1177/0003319710364213] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The infusion of a source of endothelial progenitors (EPCs) to limb ischemia (LB) patients has been used to increase angiogenesis. Because the formation of new blood vessels involves, in addition to EPCs, other cells and angiogenic regulators, we postulate that a combination cell therapy including EPCs and mesenchymal stem cells (a source of pericytes progenitors and angiogenic regulators) may represent a preferential stimuli for the development of blood vessels. In this phase I clinical trial, patients with LI were infused with a cell product consisting of autologous bone marrow-derived mononuclear and mesenchymal stem cells. After 10 2 months of follow-up, efficacy assessment demonstrated improvements in walking time, ankle brachial pressure, and quality of life. Concomitantly, angiographic and 99mTc-TF perfusion scintigraphy scores confirmed increased perfusion in the treated limbs. These results show that the use of a combination cell therapy is safe, feasible, and appears effective in patients with LI.
Collapse
|
45
|
Kumar AHS, Caplice NM. Clinical potential of adult vascular progenitor cells. Arterioscler Thromb Vasc Biol 2010; 30:1080-7. [PMID: 20453166 DOI: 10.1161/atvbaha.109.198895] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cell therapy to treat vascular and cardiovascular diseases has evolved over the past decade with improved understanding of progenitor cell mobilization, recruitment, and differentiation. The beneficial effects seen in several preclinical studies have prompted translation of adult vascular progenitor therapy to clinical trials. To date, progenitor cells isolated from bone marrow and peripheral blood have been tested in the context of acute myocardial infarction and chronic ischemic cardiomyopathy, with moderate benefit. This therapeutic effect occurs despite a relatively small number of injected progenitor cells and short-term residence in the target zone. Thus, indirect benefits, such as paracrine factors released from these cells, have been suggested as significant contributors to therapeutic efficacy. Several additional vascular progenitors of endothelial, smooth muscle, mesenchymal, and cardiac origin have been identified that may contribute to vasculogenesis. Indeed, a unifying paradigm for the most effective cell therapy strategies to date appears to be robust support of angiogenesis. Here we discuss a number of progenitor cells that currently show potential as cardiovascular therapeutics, either singly or in combination. We look at emerging cell types and disease targets that may be exploited for therapeutic benefit and future strategies that may maximize clinical efficacy.
Collapse
Affiliation(s)
- Arun H S Kumar
- Centre for Research in Vascular Biology, Biosciences Institute, University College Cork, Cork, Ireland
| | | |
Collapse
|
46
|
Varu VN, Hogg ME, Kibbe MR. Critical limb ischemia. J Vasc Surg 2010; 51:230-41. [PMID: 20117502 DOI: 10.1016/j.jvs.2009.08.073] [Citation(s) in RCA: 235] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 08/16/2009] [Indexed: 11/30/2022]
Abstract
Critical limb ischemia (CLI) continues to be a significantly morbid disease process for the aging population. Rigid guidelines for the management of patients with CLI are inappropriate due to the complexities that are involved in optimally treating these patients. A thin line exists in the decision process between medical management vs surgical management by revascularization or amputation, and the perception of "success" in this patient population is evolving. This review explores these issues and examines the challenges the treating physician will face when managing the care of patients with CLI. The epidemiology and natural history of CLI is discussed, along with the pathophysiology of the disease process. A review of the literature in regards to the different treatment modalities is presented to help the physician optimize therapy for patients with CLI. New scoring systems to help predict outcomes in patients with CLI undergoing revascularization or amputation are discussed, and an overview of the current status of patient-oriented outcomes is provided. Finally, we briefly examine emerging therapies for the treatment of CLI and provide an algorithm to help guide the practicing physician on how to approach the critically ischemic limb with regard to the complicated issues surrounding these patients.
Collapse
Affiliation(s)
- Vinit N Varu
- Division of Vascular Surgery, Northwestern University, Chicago, Ill 60611, USA
| | | | | |
Collapse
|
47
|
Sneider EB, Nowicki PT, Messina LM. Regenerative medicine in the treatment of peripheral arterial disease. J Cell Biochem 2010; 108:753-61. [PMID: 19711369 DOI: 10.1002/jcb.22315] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The last decade has witnessed a dramatic increase in the mechanistic understanding of angiogenesis and arteriogenesis, the two processes by which the body responds to obstruction of large conduit arteries. This knowledge has been translated into novel therapeutic approaches to the treatment of peripheral arterial disease, a condition characterized by progressive narrowing of lower extremity arteries and heretofore solely amenable to surgical revascularization. Clinical trials of molecular, genetic, and cell-based treatments for peripheral artery obstruction have generally provided encouraging results.
Collapse
Affiliation(s)
- Erica B Sneider
- Department of Surgery, Division of Vascular Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | |
Collapse
|
48
|
Germani A, Di Campli C, Pompilio G, Biglioli P, Capogrossi MC. Regenerative therapy in peripheral artery disease. Cardiovasc Ther 2010; 27:289-304. [PMID: 19903190 DOI: 10.1111/j.1755-5922.2009.00105.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Patients with peripheral artery disease (PAD) and critical limb ischemia are the main candidates for limb amputations and have a poor life expectancy. Frequently, these patients are not eligible for either surgical or percutaneous interventions aimed at mechanical revascularization. Therefore, new strategies need to be identified to offer these patients a viable therapeutic option. Gene and cell therapy hold great promise for the treatment of peripheral vascular diseases because, in animal models, local delivery of growth factors and endothelial progenitor cells result in new blood vessel formation and regeneration of ischemic tissues. In this article, are reviewed phase I and phase II gene, and cell therapy clinical trials in patients with PAD.
Collapse
|
49
|
Bone marrow-derived cells are differentially involved in pathological and physiological retinal angiogenesis in mice. Biochem Biophys Res Commun 2009; 391:1268-73. [PMID: 20006575 DOI: 10.1016/j.bbrc.2009.12.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 12/09/2009] [Indexed: 11/20/2022]
Abstract
PURPOSE Bone marrow-derived cells have been shown to play roles in angiogenesis. Although these cells have been shown to promote angiogenesis, it is not yet clear whether these cells affect all types of angiogenesis. This study investigated the involvement of bone marrow-derived cells in pathological and physiological angiogenesis in the murine retina. MATERIALS AND METHODS The oxygen-induced retinopathy (OIR) model was used as a retinal angiogenesis model in newborn mice. To block the influence of bone marrow-derived cells, the mice were irradiated with a 4-Gy dose of radiation from a (137)Cs source. Irradiation was performed in four different conditions with radio dense 2-cm thick lead disks; (1) H group, the head were covered with these discs to protect the eyes from radiation; (2) A group, all of the body was covered with these discs; (3) N group, mice were completely unshielded; (4) C group, mice were put in the irradiator but were not irradiated. On P17, the retinal areas showing pathological and physiological retinal angiogenesis were measured and compared to the retinas of nonirradiated mice. RESULTS Although irradiation induced leukocyte depletion, it did not affect the number of other cell types or body weight. Retinal nonperfusion areas were significantly larger in irradiated mice than in control mice (P<0.05), indicating that physiological angiogenesis was impaired. However, the formation of tuft-like angiogenesis processes was more prominent in the irradiated mice (P<0.05), indicating that pathological angiogenesis was intact. CONCLUSIONS Bone marrow-derived cells seem to be differentially involved in the formation of physiological and pathological retinal vessels. Pathological angiogenesis in the murine retina does not require functional bone marrow-derived cells, but these cells are important for the formation of physiological vessels. Our results add a new insight into the pathology of retinal angiogenesis and bolster the hypothesis that bone marrow cells are involved in the pathology or severity of retinal angiogenic diseases.
Collapse
|
50
|
Abstract
The therapeutic potential of 'adult' or at least non-embryonic stem cells and their progeny has developed gradually over the past half century as a consequence of the wealth of knowledge derived from stem cell research. Translational research coupled with clinical trials and derived from basic research has led the way to the clinic. This commenced with the use of haematopoietic stem cell transplantation (HSCT), to treat haematological malignancies, to be followed by the most recent clinical trials to treat a variety of coronary and peripheral artery diseases. Stem cells and their progeny isolated from bone marrow or blood appear to exert an ameliorating effect in certain vascular disorders. Although promising, some of these treatments remain controversial and further research and, where indicated, appropriately powered trials are required to confirm the safety and determine the efficacy of these novel therapies.
Collapse
Affiliation(s)
- E Martin-Rendon
- Stem Cell Research Laboratory, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, UK.
| | | | | |
Collapse
|