1
|
Modugno P, Cilla S, Centritto EM, Picone V, Maiorano M, Amatuzio M, Petrilli MP, Fraticelli V, De Filippo CM, Caradonna E, Codispoti FA, Massetti M, Tshomba Y. Autologous Bone Marrow Stem Cells in Patients With Critical Limb Ischaemia not Eligible for Revascularization: A Single Centre Experience. Angiology 2024; 75:865-873. [PMID: 37728082 DOI: 10.1177/00033197231190512] [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: 09/21/2023]
Abstract
We evaluated the use of autologus bone marrow stem cells transplantation in patients with critical limb ischaemia (CLI) not eligible for revascularization. Eighty consecutive patients candidate to amputation were enrolled in a single-centre retrospective study. The primary endpoint was defined as the amputation-free rate from stem cells transplantation. Secondary endpoints were the evaluation of transcutaneous oximetry and its predictive potential for probability of amputation and the evaluation of rest pain. Ankle brachial index, transcutaneous oxygen (TcpO2) and radiological imaging were performed at the enrolment and during the follow-up times. All patients were treated with auto transplant of bone marrow stem cells. Two patients died due to acute renal and acute respiratory failures. 19 patients were amputated from the thigh or leg. In total, 59 of 80 patients intended to thigh amputation saved the limb, preserving the plantar support. TcpO2 was found a predictive metric with an AUC equal to .763, and a threshold for a risk of amputation of 10% and 5% at the values ≤22.7 and ≤26.9 mmHg, respectively. Auto transplant of bone marrow stem cells seems to be a safe and an efficient option for CLI not eligible to revascularizzation.
Collapse
Affiliation(s)
- Pietro Modugno
- Vascular Surgery Unit, Gemelli Molise Hospital, Campobasso, Italy
| | - Savino Cilla
- Medical Physics Unit, Gemelli Molise Hospital, Campobasso, Italy
| | | | - Veronica Picone
- Vascular Surgery Unit, Gemelli Molise Hospital, Campobasso, Italy
| | | | | | | | | | | | - Eugenio Caradonna
- Società italiana di medicina e chirurgia rigenerativa, Caserta, Italy
| | | | - Massimo Massetti
- Cardiac Surgery Unit, Universita' Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Yamume Tshomba
- Vascular Surgery Unit, Universita' Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
2
|
Ueno K, Kurazumi H, Suzuki R, Yanagihara M, Mizoguchi T, Harada T, Morikage N, Hamano K. miR-709 exerts an angiogenic effect through a FGF2 upregulation induced by a GSK3B downregulation. Sci Rep 2024; 14:11372. [PMID: 38762650 PMCID: PMC11102560 DOI: 10.1038/s41598-024-62340-4] [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] [Received: 11/15/2022] [Accepted: 05/15/2024] [Indexed: 05/20/2024] Open
Abstract
The aim of this study was to identify angiogenic microRNAs (miRNAs) that could be used in the treatment of hindlimb ischemic tissues. miRNAs contained in extracellular vesicles (EVs) deriving from the plasma were analyzed in C57BL/6 mice, which have ischemia tolerance, and in BALB/c mice without ischemia tolerance as part of a hindlimb ischemia model; as a result 43 angiogenic miRNA candidates were identified. An aortic ring assay was employed by using femoral arteries isolated from BALC/c mice and EVs containing miRNA; as a result, the angiogenic miRNA candidates were limited to 14. The blood flow recovery was assessed after injecting EVs containing miRNA into BALB/c mice with hindlimb ischemia, and miR-709 was identified as a promising angiogenic miRNA. miR-709-encapsulating EVs were found to increase the expression levels of the fibroblast growth factor 2 (FGF2) mRNA in the thigh tissues of hindlimb ischemia model BALB/c mice. miR-709 was also found to bind to the 3'UTR of glycogen synthase kinase 3 beta (GSK3B) in three places. GSK3B-knockdown human artery-derived endothelial cells were found to express high levels of FGF2, and were characterized by increased cell proliferation. These findings indicate that miR-709 induces an upregulation of FGF2 through the downregulation of GSK3B.
Collapse
Affiliation(s)
- Koji Ueno
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan.
- Division of Advanced Cell Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan.
| | - Hiroshi Kurazumi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Ryo Suzuki
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Masashi Yanagihara
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Takahiro Mizoguchi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Takasuke Harada
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
- Division of Advanced Cell Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
3
|
Husakova J, Echalar B, Kossl J, Palacka K, Fejfarova V, Dubsky M. The Effects of Immunosuppressive Drugs on the Characteristics and Functional Properties of Bone Marrow-Derived Stem Cells Isolated from Patients with Diabetes Mellitus and Peripheral Arterial Disease. Biomedicines 2023; 11:1872. [PMID: 37509511 PMCID: PMC10377428 DOI: 10.3390/biomedicines11071872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/18/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Diabetic patients (DPs) with foot ulcers can receive autologous cell therapy (ACT) as a last therapeutic option. Even DPs who have undergone organ transplantation and are using immunosuppressive (IS) drugs can be treated by ACT. The aim of our study was to analyze the effects of IS drugs on the characteristics of bone marrow-derived stem cells (BM-MSCs). METHODS The cells were isolated from the bone marrow of DPs, cultivated for 14-18 days, and phenotypically characterized using flow cytometry. These precursor cells were cultured in the presence of various IS drugs. The impact of IS drugs on metabolic activity was measured using a WST-1 assay, and the expression of genes for immunoregulatory molecules was detected through RT-PCR. Cell death was analyzed through the use of flow cytometry, and the production of cytokines was determined by ELISA. RESULTS The mononuclear fraction of cultured cells contained mesenchymal stem cells (CD45-CD73+CD90+CD105+), myeloid angiogenic cells (CD45+CD146-), and endothelial colony-forming cells (CD45-CD146+). IS drugs inhibited metabolic activity, the expression of genes for immunoregulatory molecules, the production of cytokines, and the viability of the cells. CONCLUSIONS The results indicate that IS drugs in a dose-dependent manner had a negative impact on the properties of BM-MSCs used to treat ischemic diabetic foot ulcers, and that these drugs could affect the therapeutic potential of BM-MSCs.
Collapse
Affiliation(s)
- Jitka Husakova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
- First Faculty of Medicine, Charles University, 14021 Prague, Czech Republic
| | - Barbora Echalar
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, 14021 Prague, Czech Republic
| | - Jan Kossl
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, 14021 Prague, Czech Republic
| | - Katerina Palacka
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, 14021 Prague, Czech Republic
| | - Vladimira Fejfarova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
| | - Michal Dubsky
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
- First Faculty of Medicine, Charles University, 14021 Prague, Czech Republic
| |
Collapse
|
4
|
Wang SK, Green LA, Gutwein AR, Drucker NA, Babbey CM, Gupta AK, Fajardo A, Motaganahalli RL, Wilson MG, Murphy MP. Ethnic minorities with critical limb ischemia derive equal amputation risk reduction from autologous cell therapy compared with whites. J Vasc Surg 2018; 68:560-566. [PMID: 29503004 DOI: 10.1016/j.jvs.2017.11.088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2017] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Ethnic minorities (nonwhites) with critical limb ischemia (CLI) have historically performed worse compared with whites with regard to major amputation risk reduction and amputation-free survival (AFS) after peripheral vascular intervention. This post hoc analysis was completed to determine whether this precedent also extended to treatment of CLI without a suitable revascularization option with intramuscular injections of concentrated bone marrow aspirate (cBMA). METHODS The treatment arm of the randomized, double-blind, multicenter MarrowStim PAD Kit for the Treatment of Critical Limb Ischemia in Subjects with Severe Peripheral Arterial Disease (MOBILE) trial was stratified by ethnicity and evaluated for demographics, comorbidities, and outcomes. The primary and therapeutic end point was 1-year AFS and major amputation, respectively. Noninferiority analysis was performed with the margin set at historically reported hazard ratios. RESULTS Thirty-seven minority (African American, Hispanic, other) CLI patients (9 placebo, 28 cBMA) with no suitable revascularization option were randomized to cBMA or placebo at a 3:1 ratio during the MOBILE trial. At 1-year follow-up for the treatment group, overall AFS was 80%. Of the 28 minority patients randomized to cBMA intervention, an 89% AFS rate was observed compared with 77% in whites. Specifically, 22 of 24 (92%) African Americans survived amputation free at 1-year follow-up. Noninferiority testing confirmed no difference between whites and the ethnic minority treated with cBMA with respect to major amputation reduction; however, noninferiority could not be confirmed with regard to AFS. No significant differences favoring whites treated with cBMA were noted in the secondary end points of vascular quality of life, limb pain, ankle-brachial index, toe-brachial index, transcutaneous oximetry, and 6-minute walk testing. CONCLUSIONS This post hoc analysis of the MOBILE trial demonstrates noninferiority of cBMA intervention in minorities with no-option CLI for the therapeutic end point of major amputation prevention. cBMA represents a novel treatment paradigm and should be explored for minorities with poor revascularization options who face impending amputation secondary to progressive CLI.
Collapse
Affiliation(s)
- S Keisin Wang
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Linden A Green
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Ashley R Gutwein
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Natalie A Drucker
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Clifford M Babbey
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Alok K Gupta
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Andres Fajardo
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Raghu L Motaganahalli
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Michael G Wilson
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Ind
| | - Michael P Murphy
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind.
| |
Collapse
|
5
|
Sanberg PR, Greene-Zavertnik C, Davis CD. Article Commentary: Cell Transplantation: The Regenerative Medicine Journal. A Biennial Analysis of Publications. Cell Transplant 2017; 12:815-825. [DOI: 10.3727/000000003771000165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC 78, Tampa, FL 33612
| | - Cathryn Greene-Zavertnik
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC 78, Tampa, FL 33612
| | - Cyndy D. Davis
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., MDC 78, Tampa, FL 33612
| |
Collapse
|
6
|
Kim EJ, Seo SG, Shin HS, Lee DJ, Kim JH, Lee DY. Platelet-Derived Growth Factor Receptor-Positive Pericytic Cells of White Adipose Tissue from Critical Limb Ischemia Patients Display Mesenchymal Stem Cell-Like Properties. Clin Orthop Surg 2017; 9:239-248. [PMID: 28567229 PMCID: PMC5435665 DOI: 10.4055/cios.2017.9.2.239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 02/24/2017] [Indexed: 12/25/2022] Open
Abstract
Background The pericytes in the blood vessel wall have recently been identified to be important in regulating vascular formation, stabilization, remodeling, and function. We isolated and identified pericyte-like platelet-derived growth factor receptor beta-positive (PDGFRβ+) cells from the stromal vascular fraction (SVF) of adipose tissue from critical limb ischemia (CLI) patients and investigated their potential as a reliable source of stem cells for cell-based therapy. Methods De-identified subcutaneous fat tissues were harvested after amputation in CLI patients. Freshly isolated SVF cells and culture-expanded adipose-derived stem cells (ADSCs) were quantified using flow cytometry. A matrigel tube formation assay and multi-lineage differentiation were performed to assess pericytic and mesenchymal stem cell (MSC)-like characteristics of PDGFRβ+ ADSCs. Results PDGFRβ+ cells were located in the pericytic area of various sizes of blood vessels and coexpressed mesenchymal stem cell markers. PDGFRβ+ cells in freshly isolated SVF cells expressed a higher level of stem cell markers (CD34 and CXCR4) and mesenchymal markers (CD13, CD44, CD54, and CD90) than PDGFRβ– cells. In vitro expansion of PDGFRβ+ cells resulted in enrichment of the perivascular mesenchymal stem-like (PDGFRβ+/CD90+/CD45–/CD31–) cell fractions. The Matrigel tube formation assay revealed that PDGFRβ+ cells were located in the peritubular area. Conclusions PDGFRβ+ ADSCs cells demonstrated a good multilineage differentiation potential. Pericyte-like PDGFRβ+ cells from the SVF of adipose tissue from CLI patients had MSC-like characteristics and could be amplified by in vitro culture with preservation of their cell characteristics. We believe PDGFRβ+ cells in the SVF of adipose tissue can be used as a reliable source of stem cells even in CLI patients.
Collapse
Affiliation(s)
- Eo Jin Kim
- Department of Orthopedic Surgery, Hanil General Hospital, Seoul, Korea
| | - Sang Gyo Seo
- Department of Orthopedic Surgery, Asan Medical Center, Seoul, Korea
| | - Hyuk Soo Shin
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, Korea
| | - Doo Jae Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, Korea
| | - Ji Hye Kim
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, Korea
| | - Dong Yeon Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
7
|
Wang SK, Green LA, Motaganahalli RL, Wilson MG, Fajardo A, Murphy MP. Rationale and design of the MarrowStim PAD Kit for the Treatment of Critical Limb Ischemia in Subjects with Severe Peripheral Arterial Disease (MOBILE) trial investigating autologous bone marrow cell therapy for critical limb ischemia. J Vasc Surg 2017; 65:1850-1857.e2. [PMID: 28390770 DOI: 10.1016/j.jvs.2017.01.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/28/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Critical limb ischemia (CLI) continues to place a significant encumbrance on patients and the health care system as it progresses to limb loss and long-term disability. Traditional methods of revascularization offer a significant benefit; however, for one-third of CLI patients, these surgical options are not technically possible or patency is severely limited by disease burden (deemed "poor-option" for revascularization). In a previous phase I trial, we demonstrated intramuscular injection of concentrated bone marrow aspirate (cBMA) via MarrowStim (Zimmer Biomet, Warsaw, Ind) harvest is safe and may decrease major amputation in patients with CLI unfit for surgical revascularization. Therefore, we describe and rationalize the MarrowStim PAD Kit for the Treatment of Critical Limb Ischemia in Subjects with Severe Peripheral Arterial Disease (MOBILE) trial, a study geared to provide the pivotal proof of efficacy of cBMA in CLI. METHODS MOBILE is a multicenter, randomized, double-blind, placebo-controlled trial designed to assess the efficacy of intramuscular injections of cBMA in promoting amputation-free survival in patients with poor-option CLI. Patients (aged >21 years) with rest pain or tissue loss resulting from advanced peripheral arterial disease, as characterized by ankle-brachial index (<0.6), toe-brachial index (<0.4), or transcutaneous pressure of oxygen (<50 mm Hg), were eligible for inclusion if surgical revascularization was not possible secondary to advanced disease. RESULTS Treatment and 1-year follow-up of 152 patients enrolled in MOBILE are completed. Long-term follow-up is ongoing. Currently, we are in the process of unblinding the initial results for preliminary data analysis. CONCLUSIONS If successful, MOBILE could add definitive, high-quality evidence in support of cBMA for the treatment of poor-option CLI patients and provide an additional modality for patients who face amputation secondary to advanced limb ischemia.
Collapse
Affiliation(s)
- S Keisin Wang
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Linden A Green
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Raghu L Motaganahalli
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Michael G Wilson
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Ind
| | - Andres Fajardo
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Michael P Murphy
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind.
| |
Collapse
|
8
|
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: 4.1] [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
|
9
|
Palmaz JC. Local Endovascular Delivery, Gene Therapy, and Cell Transplantation for Peripheral Arterial Disease. J Endovasc Ther 2016; 11 Suppl 2:II200-206. [PMID: 15760261 DOI: 10.1177/15266028040110s617] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Advances in catheter technology, gene identification, and cell biology may provide novel treatment options for patients with peripheral arterial disease (PAD) who are not candidates for standard revascularization procedures. Animal studies and recent results in human beings suggest that transfer of growth factors or regulatory genes and transplantation of progenitor cells may provide novel therapy options by inducing therapeutic angiogenesis or by inhibiting restenosis. This review will discuss the development of a variety of catheters for localized endovascular delivery, as well as the various cellular and genetic strategies that exist to restore blood flow to ischemic tissue and to reduce neointimal hyperplasia.
Collapse
Affiliation(s)
- Julio C Palmaz
- The University of Texas Health Science Center, San Antonio, Texas 78229, USA.
| |
Collapse
|
10
|
Gupta PK, Krishna M, Chullikana A, Desai S, Murugesan R, Dutta S, Sarkar U, Raju R, Dhar A, Parakh R, Jeyaseelan L, Viswanathan P, Vellotare PK, Seetharam RN, Thej C, Rengasamy M, Balasubramanian S, Majumdar AS. Administration of Adult Human Bone Marrow-Derived, Cultured, Pooled, Allogeneic Mesenchymal Stromal Cells in Critical Limb Ischemia Due to Buerger's Disease: Phase II Study Report Suggests Clinical Efficacy. Stem Cells Transl Med 2016; 6:689-699. [PMID: 28297569 PMCID: PMC5442769 DOI: 10.5966/sctm.2016-0237] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/24/2016] [Indexed: 12/28/2022] Open
Abstract
Critical limb ischemia (CLI) due to Buerger’s disease is a major unmet medical need with a high incidence of morbidity. This phase II, prospective, nonrandomized, open‐label, multicentric, dose‐ranging study was conducted to assess the efficacy and safety of i.m. injection of adult human bone marrow‐derived, cultured, pooled, allogeneic mesenchymal stromal cells (BMMSC) in CLI due to Buerger’s disease. Patients were allocated to three groups: 1 and 2 million cells/kg body weight (36 patients each) and standard of care (SOC) (18 patients). BMMSCs were administered as 40–60 injections in the calf muscle and locally, around the ulcer. Most patients were young (age range, 38–42 years) and ex‐smokers, and all patients had at least one ulcer. Both the primary endpoints—reduction in rest pain (0.3 units per month [SE, 0.13]) and healing of ulcers (11% decrease in size per month [SE, 0.05])—were significantly better in the group receiving 2 million cells/kg body weight than in the SOC arm. Improvement in secondary endpoints, such as ankle brachial pressure index (0.03 [SE, 0.01] unit increase per month) and total walking distance (1.03 [SE, 0.02] times higher per month), were also significant in the group receiving 2 million cells/kg as compared with the SOC arm. Adverse events reported were remotely related or unrelated to BMMSCs. In conclusion, i.m. administration of BMMSC at a dose of 2 million cells/kg showed clinical benefit and may be the best regimen in patients with CLI due to Buerger’s disease. However, further randomized controlled trials are required to confirm the most appropriate dose. Stem Cells Translational Medicine2017;6:689–699
Collapse
Affiliation(s)
| | - Murali Krishna
- Department of Vascular Surgery, Sri Jayadeva Institute of Cardiovascular Sciences, Bangalore, India
| | | | - Sanjay Desai
- Department of Vascular Surgery, MS Ramaiah Medical College & Hospitals, Bangalore, India
| | | | - Santanu Dutta
- Department of Cardiovascular Surgery, Nightingale Hospital, Kolkata, India
| | - Uday Sarkar
- Department of Cardiovascular Surgery, Health Point Hospital, Kolkata, India
| | - Radhakrishnan Raju
- Department of Vascular Surgery, Sri Ramachandra Medical College, Chennai, India
| | - Anita Dhar
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Rajiv Parakh
- Division of Peripheral Vascular and Endovascular Sciences, Medanta‐The Medicity, Gurgaon, Haryana, India
| | | | | | | | | | - Charan Thej
- Stempeutics Research, Bangalore, India
- Manipal University, Manipal, India
| | | | | | | |
Collapse
|
11
|
Bai H, Gao Y, Hoyle DL, Cheng T, Wang ZZ. Suppression of Transforming Growth Factor-β Signaling Delays Cellular Senescence and Preserves the Function of Endothelial Cells Derived from Human Pluripotent Stem Cells. Stem Cells Transl Med 2016; 6:589-600. [PMID: 28191769 PMCID: PMC5442820 DOI: 10.5966/sctm.2016-0089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/09/2016] [Indexed: 12/15/2022] Open
Abstract
Transplantation of vascular cells derived from human pluripotent stem cells (hPSCs) offers an attractive noninvasive method for repairing the ischemic tissues and for preventing the progression of vascular diseases. Here, we found that in a serum‐free condition, the proliferation rate of hPSC‐derived endothelial cells is quickly decreased, accompanied with an increased cellular senescence, resulting in impaired gene expression of endothelial nitric oxide synthase (eNOS) and impaired vessel forming capability in vitro and in vivo. To overcome the limited expansion of hPSC‐derived endothelial cells, we screened small molecules for specific signaling pathways and found that inhibition of transforming growth factor‐β (TGF‐β) signaling significantly retarded cellular senescence and increased a proliferative index of hPSC‐derived endothelial cells. Inhibition of TGF‐β signaling extended the life span of hPSC‐derived endothelial and improved endothelial functions, including vascular network formation on Matrigel, acetylated low‐density lipoprotein uptake, and eNOS expression. Exogenous transforming growth factor‐β1 increased the gene expression of cyclin‐dependent kinase inhibitors, p15Ink4b, p16Ink4a, and p21CIP1, in endothelial cells. Conversely, inhibition of TGF‐β reduced the gene expression of p15Ink4b, p16Ink4a, and p21CIP1. Our findings demonstrate that the senescence of newly generated endothelial cells from hPSCs is mediated by TGF‐β signaling, and manipulation of TGF‐β signaling offers a potential target to prevent vascular aging. Stem Cells Translational Medicine2017;6:589–600
Collapse
Affiliation(s)
- Hao Bai
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yongxing Gao
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dixie L. Hoyle
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Blood Cell Therapy and Technology, Tianjin, People's Republic of China
| | - Zack Z. Wang
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Blood Cell Therapy and Technology, Tianjin, People's Republic of China
| |
Collapse
|
12
|
Liang TW, Jester A, Motaganahalli RL, Wilson MG, G'Sell P, Akingba GA, Fajardo A, Murphy MP. Autologous bone marrow mononuclear cell therapy for critical limb ischemia is effective and durable. J Vasc Surg 2016; 63:1541-5. [PMID: 27021379 DOI: 10.1016/j.jvs.2016.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/13/2016] [Indexed: 10/22/2022]
Abstract
OBJECTIVE We have previously shown that autologous bone marrow mononuclear cell (ABMNC) therapy improves measures of limb perfusion, rest pain, wound healing, and amputation-free survival (AFS) at 1 year in patients with critical limb ischemia (CLI). Long-term durability of ABMNC therapy for CLI remains unknown. The objective of the current study was to evaluate long-term clinical outcomes 5 years after treatment. METHODS Data were retrospectively gathered from a database and via a patient survey and review of medical records of patients previously enrolled in this phase I/II trial. AFS, freedom from major amputation, and freedom from major adverse limb events (MALE) were calculated using the product-limit estimate. The incidence of cardiac, malignant, and other medical events relevant to the safety of cell therapy were tabulated during the time from treatment to follow-up. RESULTS Twenty-one of the 24 patients (88%) who completed the initial 1-year phase I/II trial were available for the 5-year analysis; AFS was 74% (95% confidence interval [CI], 0.53-0.87), freedom from major amputation was 78% (95% CI, 0.58-0.90), and freedom from MALE was 65% (95% CI, 0.45-0.80). Three patients (14%) had major cardiac events. There were no incidences of malignancies or diagnoses of clinically significant proliferative retinopathy. Fifteen patients (71%) report continued improvement in pain-free walking. Nineteen (90%) patients believed that the study was of significant medical value and would participate again. CONCLUSIONS ABMNC therapy provides long-term freedom from AFS, major amputation, and MALE that are comparable with other reports of patients who underwent surgical and endovascular interventions for CLI. Furthermore, no patients developed tumorigenesis or clinically significant retinopathy. Because of the limited number of patients studied, our findings will need to be followed up in a larger phase III trial.
Collapse
Affiliation(s)
- Tiffany W Liang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Andrea Jester
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Raghu L Motaganahalli
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Michael G Wilson
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Ind
| | - Patricia G'Sell
- Indiana Center for Vascular Biology and Medicine, Indiana University School of Medicine, Indianapolis, Ind
| | - George A Akingba
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Andres Fajardo
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind
| | - Michael P Murphy
- Division of Vascular Surgery, Indiana University School of Medicine, Indianapolis, Ind.
| |
Collapse
|
13
|
Transcatheter Arterial Infusion of Autologous CD133(+) Cells for Diabetic Peripheral Artery Disease. Stem Cells Int 2016; 2016:6925357. [PMID: 26981134 PMCID: PMC4769775 DOI: 10.1155/2016/6925357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/10/2015] [Accepted: 01/04/2016] [Indexed: 01/06/2023] Open
Abstract
Microvascular lesion in diabetic peripheral arterial disease (PAD) still cannot be resolved by current surgical and interventional technique. Endothelial cells have the therapeutic potential to cure microvascular lesion. To evaluate the efficacy and immune-regulatory impact of intra-arterial infusion of autologous CD133(+) cells, we recruited 53 patients with diabetic PAD (27 of CD133(+) group and 26 of control group). CD133(+) cells enriched from patients' PB-MNCs were reinfused intra-arterially. The ulcer healing followed up till 18 months was 100% (3/3) in CD133(+) group and 60% (3/5) in control group. The amputation rate was 0 (0/27) in CD133(+) group and 11.54% (3/26) in control group. Compared with the control group, TcPO2 and ABI showed obvious improvement at 18 months and significant increasing VEGF and decreasing IL-6 level in the CD133(+) group within 4 weeks. A reducing trend of proangiogenesis and anti-inflammatory regulation function at 4 weeks after the cells infusion was also found. These results indicated that autologous CD133(+) cell treatment can effectively improve the perfusion of morbid limb and exert proangiogenesis and anti-inflammatory immune-regulatory impacts by paracrine on tissue microenvironment. The CD133(+) progenitor cell therapy may be repeated at a fixed interval according to cell life span and immune-regulatory function.
Collapse
|
14
|
Franz RW, Shah KJ, Pin RH, Hankins T, Hartman JF, Wright ML. Autologous bone marrow mononuclear cell implantation therapy is an effective limb salvage strategy for patients with severe peripheral arterial disease. J Vasc Surg 2015; 62:673-80. [PMID: 26304481 DOI: 10.1016/j.jvs.2015.02.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/13/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE This study was conducted to determine if intramuscular and intra-arterial stem cell injections delay or prevent major limb amputations, improve ankle-brachial index measurements, relieve rest pain, and improve ulcer healing. METHODS A prospective case series with interventions occurring between December 2007 and September 2012 and a 3-month minimum follow-up was conducted at an urban tertiary care referral hospital. Patients with severe limb-threatening peripheral arterial disease, without other options for revascularization, were eligible for enrollment. Dual intramuscular and intra-arterial injection of bone marrow mononuclear cells harvested from the iliac crest was performed. Major limb amputation at 3 months was the primary outcome measure. Secondary outcome measures included ankle-brachial index measurements, rest pain, and ulceration healing. Kaplan-Meier survivorship was performed to ascertain overall survivorship of the procedure. RESULTS No complications related to the procedure were reported. Of 49 patients (56 limbs) enrolled, two patients (two limbs) died, but had not undergone major amputation, and five limbs (8.9%) underwent major amputation within the first 3 months. Three-month follow-up evaluations were conducted on the remaining 49 limbs (42 patients). Median postprocedure revised Rutherford and Fontaine classifications were significantly lower compared with median baseline classifications. After 3 months, seven patients (nine limbs) died but had not undergone major amputation, and seven limbs (14.3%) underwent major amputation. At a mean follow-up of 18.2 months, the remaining 33 limbs (29 patients) had not undergone a major amputation. Freedom from major adverse limb events (MALE) was 91.1% (95% confidence interval, 79.9-96.2) at 3 months and 75.6% (95% confidence interval, 59.4-86.1) at 12 months. CONCLUSIONS This procedure was designed to improve limb perfusion in an effort to salvage limbs in patients for whom amputation was the only viable treatment option. The results of this analysis indicate that it is an effective strategy for limb salvage for patients with severe peripheral arterial disease.
Collapse
Affiliation(s)
| | - Kaushal J Shah
- Vascular Surgery, Geisinger - Holy Spirit Health System, Camp Hill, Pa
| | - Richard H Pin
- Vascular and Endovascular Surgery, Southcoast Hospitals Group, Dartmouth, Mass
| | | | - Jodi F Hartman
- Orthopaedic Research & Reporting, Ltd, Westerville, Ohio
| | | |
Collapse
|
15
|
Raval AN, Schmuck EG, Tefera G, Leitzke C, Ark CV, Hei D, Centanni JM, de Silva R, Koch J, Chappell RG, Hematti P. Bilateral administration of autologous CD133+ cells in ambulatory patients with refractory critical limb ischemia: lessons learned from a pilot randomized, double-blind, placebo-controlled trial. Cytotherapy 2014; 16:1720-32. [PMID: 25239491 PMCID: PMC4253573 DOI: 10.1016/j.jcyt.2014.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND AIMS CD133+ cells confer angiogenic potential and may be beneficial for the treatment of critical limb ischemia (CLI). However, patient selection, blinding methods and end points for clinical trials are challenging. We hypothesized that bilateral intramuscular administration of cytokine-mobilized CD133+ cells in ambulatory patients with refractory CLI would be feasible and safe. METHODS In this double-blind, randomized sham-controlled trial, subjects received subcutaneous injections of granulocyte colony-stimulating factor (10 μg/kg per day) for 5 days, followed by leukapheresis, and intramuscular administration of 50-400 million sorted CD133+ cells delivered into both legs. Control subjects received normal saline injections, sham leukapheresis and intramuscular injection of placebo buffered solution. Subjects were followed for 1 year. An aliquot of CD133+ cells was collected from each subject to test for genes associated with cell senescence. RESULTS Seventy subjects were screened, of whom 10 were eligible. Subject enrollment was suspended because of a high rate of mobilization failure in subjects randomly assigned to treatment. Of 10 subjects enrolled (7 randomly assigned to treatment, 3 randomly assigned to control), there were no differences in serious adverse events at 12 months, and blinding was preserved. There were non-significant trends toward improved amputation-free survival, 6-minute walk distance, walking impairment questionnaire and quality of life in subjects randomly assigned to treatment. Successful CD133+ mobilizers expressed fewer senescence-associated genes compared with poor mobilizers. CONCLUSIONS Bilateral administration of autologous CD133+ cells in ambulatory CLI subjects was safe, and blinding was preserved. However, poor mobilization efficiency combined with high CD133+ senescence suggests futility in this approach.
Collapse
Affiliation(s)
- Amish N Raval
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| | - Eric G Schmuck
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Girma Tefera
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cathlyn Leitzke
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cassondra Vander Ark
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Derek Hei
- Waisman Biomanufacturing Facility, Madison, Wisconsin, USA
| | - John M Centanni
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ranil de Silva
- National Heart and Lung Institute, Imperial College London and NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | - Jill Koch
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Richard G Chappell
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Peiman Hematti
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
16
|
Maijub JG, Boyd NL, Dale JR, Hoying JB, Morris ME, Williams SK. Concentration-Dependent Vascularization of Adipose Stromal Vascular Fraction Cells. Cell Transplant 2014; 24:2029-39. [PMID: 25397993 DOI: 10.3727/096368914x685401] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adipose-derived stromal vascular fraction (SVF) cells have been shown to self-associate to form vascular structures under both in vitro and in vivo conditions. The angiogenic (new vessels from existing vessels) and vasculogenic (new vessels through self-assembly) potential of the SVF cell population may provide a cell source for directly treating (i.e., point of care without further cell isolation) ischemic tissues. However the correct dosage of adipose SVF cells required to achieve a functional vasculature has not been established. Accordingly, in vitro and in vivo dose response assays were performed evaluating the SVF cell vasculogenic potential. Serial dilutions of freshly isolated rat adipose SVF cells were plated on growth factor reduced Matrigel and vasculogenesis, assessed as cellular tube-like network assembly, was quantified after 3 days of culture. This in vitro vasculogenesis assay indicated that rat SVF cells reached maximum network length at a concentration of 2.5 × 10(5) cells/ml and network maintained at the higher concentrations tested. The same concentrations of rat and human SVF cells were used to evaluate vasculogenesis in vivo. SVF cells were incorporated into collagen gels and subcutaneously implanted into Rag1 immunodeficient mice. The 3D confocal images of harvested constructs were evaluated to quantify dose dependency of SVF cell vasculogenesis potential. Rat- and human-derived SVF cells yielded a maximum vasculogenic potential at 1 × 10(6) and 4 × 10(6) cells/ml, respectively. No adverse reactions (e.g., toxicity, necrosis, tumor formation) were observed at any concentration tested. In conclusion, the vasculogenic potential of adipose-derived SVF cell populations is dose dependent.
Collapse
Affiliation(s)
- John G Maijub
- Cardiovascular Innovation Institute, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | | | | | | |
Collapse
|
17
|
Kudo T, Hosoyama T, Samura M, Katsura S, Nishimoto A, Kugimiya N, Fujii Y, Li TS, Hamano K. Hypoxic preconditioning reinforces cellular functions of autologous peripheral blood-derived cells in rabbit hindlimb ischemia model. Biochem Biophys Res Commun 2014; 444:370-5. [PMID: 24463101 DOI: 10.1016/j.bbrc.2014.01.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 01/15/2014] [Indexed: 11/29/2022]
Abstract
Peripheral blood mononuclear cell (PBMNC) is one of powerful tools for therapeutic angiogenesis in hindlimb ischemia. However, traditional approaches with transplanted PBMNCs show poor therapeutic effects in severe ischemia patients. In this study, we used autograft models to determine whether hypoxic pretreatment effectively enhances the cellular functions of PBMNCs and improves hindlimb ischemia. Rabbit PBMNCs were cultured in the hypoxic condition. After pretreatment, cell adhesion, stress resistance, and expression of angiogenic factor were evaluated in vitro. To examine in vivo effects, we autografted preconditioned PBMNCs into a rabbit hindlimb ischemia model on postoperative day (POD) 7. Preconditioned PBMNCs displayed significantly enhanced functional capacities in resistance to oxidative stress, cell viability, and production of vascular endothelial growth factor. In addition, autologous transplantation of preconditioned PBMNCs significantly induced new vessels and improved limb blood flow. Importantly, preconditioned PBMNCs can accelerate vessel formation despite transplantation on POD 7, whereas untreated PBMNCs showed poor vascularization. Our study demonstrated that hypoxic preconditioning of PBMNCs is a feasible approach for increasing the retention of transplanted cells and enhancing therapeutic angiogenesis in ischemic tissue.
Collapse
Affiliation(s)
- Tomoaki Kudo
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Tohru Hosoyama
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan.
| | - Makoto Samura
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shunsaku Katsura
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Arata Nishimoto
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Naruji Kugimiya
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yasuhiko Fujii
- Department of Blood Transfusion Regeneration and Cell Therapy Center, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Division of Cardiac Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|
18
|
Hernigou P, Homma Y, Flouzat-Lachaniette CH, Poignard A, Chevallier N, Rouard H. Cancer risk is not increased in patients treated for orthopaedic diseases with autologous bone marrow cell concentrate. J Bone Joint Surg Am 2013; 95:2215-21. [PMID: 24352775 DOI: 10.2106/jbjs.m.00261] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND There is concern that regenerative cell-based therapies could result in increased risk of tumor formation. We investigated the long-term risks for systemic and site-specific cancers in patients who had received autologous bone marrow-derived stromal progenitor cells to treat orthopaedic lesions. METHODS A total of 1873 patients were treated from 1990 to 2006 with bone marrow-derived concentrated cells. Patients were monitored for cancer incidence from the date of the first operation (1990) until death, or until December 31, 2011. The mean follow-up time was 12.5 years (range, five to twenty-two years). The average number of colony-forming unit fibroblasts returned to the patients was 483,000 fibroblasts (range, 62,000 to 2,095,000 fibroblasts). The primary outcome was to evaluate with radiographs and/or magnetic resonance imaging the risk of tumorigenesis at the cell therapy treatment sites. The secondary outcome was to evaluate the risk of cancer diagnosed in areas other than the treatment site during the follow-up period. The relative risk of cancer was expressed as the ratio of observed and expected number of cases, that is, the standardized incidence ratio, according to the cancer incidence in the French population. RESULTS No tumor formation was found at the treatment sites on the 7306 magnetic resonance images and 52,430 radiographs among the 1873 patients. Fifty-three cancers were diagnosed in areas other than the treatment site. On the basis of cancer incidence in the general population during the same period, the expected number of cancers was between ninety-seven and 108 for the same age and sex distribution. The range of the standardized incidence ratio for the follow-up period was between 0.49 and 0.54 (95% confidence interval, 0.30 to 0.80). CONCLUSIONS This study found no increased cancer risk in patients after application of autologous cell-based therapy using bone marrow-derived stromal progenitor cells either at the treatment site or elsewhere in the patients after an average follow-up period of 12.5 years.
Collapse
Affiliation(s)
- Philippe Hernigou
- Department of Orthopaedic Surgery (P.H., C.-H.F.-L., and A.P.) and EFS Cell Therapy Facility (N.C. and H.R.), University Paris East, Hospital Henri Mondor, 51 avenue du Marechal de Lattre de Tassigny, 94010 Creteil, France. E-mail address for P. Hernigou:
| | - Yasuhiro Homma
- Juntendo University, Hongo 2-1-1, Bunko-Ku, Tokyo 113-8421, Japan
| | - Charles-Henri Flouzat-Lachaniette
- Department of Orthopaedic Surgery (P.H., C.-H.F.-L., and A.P.) and EFS Cell Therapy Facility (N.C. and H.R.), University Paris East, Hospital Henri Mondor, 51 avenue du Marechal de Lattre de Tassigny, 94010 Creteil, France. E-mail address for P. Hernigou:
| | - Alexandre Poignard
- Department of Orthopaedic Surgery (P.H., C.-H.F.-L., and A.P.) and EFS Cell Therapy Facility (N.C. and H.R.), University Paris East, Hospital Henri Mondor, 51 avenue du Marechal de Lattre de Tassigny, 94010 Creteil, France. E-mail address for P. Hernigou:
| | - Nathalie Chevallier
- Department of Orthopaedic Surgery (P.H., C.-H.F.-L., and A.P.) and EFS Cell Therapy Facility (N.C. and H.R.), University Paris East, Hospital Henri Mondor, 51 avenue du Marechal de Lattre de Tassigny, 94010 Creteil, France. E-mail address for P. Hernigou:
| | - Helene Rouard
- Department of Orthopaedic Surgery (P.H., C.-H.F.-L., and A.P.) and EFS Cell Therapy Facility (N.C. and H.R.), University Paris East, Hospital Henri Mondor, 51 avenue du Marechal de Lattre de Tassigny, 94010 Creteil, France. E-mail address for P. Hernigou:
| |
Collapse
|
19
|
Stem cells in plastic surgery: a review of current clinical and translational applications. Arch Plast Surg 2013; 40:666-75. [PMID: 24286038 PMCID: PMC3840172 DOI: 10.5999/aps.2013.40.6.666] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Stem cells are a unique cell population characterized by self-renewal and cellular differentiation capabilities. These characteristics, among other traits, make them an attractive option for regenerative treatments of tissues defects and for aesthetic procedures in plastic surgery. As research regarding the isolation, culture and behavior of stem cells has progressed, stem cells, particularly adult stem cells, have shown promising results in both translational and clinical applications. METHODS The purpose of this review is to evaluate the applications of stem cells in the plastic surgery literature, with particular focus on the advances and limitations of current stem cell therapies. Different key areas amenable to stem cell therapy are addressed in the literature review; these include regeneration of soft tissue, bone, cartilage, and peripheral nerves, as well as wound healing and skin aging. RESULTS The reviewed studies demonstrate promising results, with favorable outcomes and minimal complications in the cited cases. In particular, adipose tissue derived stem cell (ADSC) transplants appear to provide effective treatment options for bony and soft tissue defects, and non-healing wounds. ADSCs have also been shown to be useful in aesthetic surgery. CONCLUSIONS Further studies involving both the basic and clinical science aspects of stem cell therapies are warranted. In particular, the mechanism of action of stem cells, their interactions with the surrounding microenvironment and their long-term fate require further elucidation. Larger randomized trials are also necessary to demonstrate the continued safety of transplanted stem cells as well as the efficacy of cellular therapies in comparison to the current standards of care.
Collapse
|
20
|
Shimamura M, Nakagami H, Koriyama H, Morishita R. Gene therapy and cell-based therapies for therapeutic angiogenesis in peripheral artery disease. BIOMED RESEARCH INTERNATIONAL 2013; 2013:186215. [PMID: 24294599 PMCID: PMC3835886 DOI: 10.1155/2013/186215] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/09/2013] [Indexed: 02/02/2023]
Abstract
Gene therapy and cell-based therapy have emerged as novel therapies to promote therapeutic angiogenesis in critical limb ischemia (CLI) caused by peripheral artery disease (PAD). Although researchers initially focused on gene therapy using proangiogenic factors, such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), and hepatocyte growth factors (HGF), cell therapy using bone marrow mononuclear cells (BMMNCs), mesenchymal stem cells (BMMSCs), G-CSF-mobilized peripheral blood mononuclear cells (M-PBMNCs), and endothelial progenitor cells (EPCs) have also been extensively studied. Based on the elaborate studies and favorable results of basic research, some clinical phase I/II trials have been performed, and the results demonstrate the safety of these approaches and their potential for symptomatic improvement in CLI. However, the phase 3 clinical trials have thus far been limited to gene therapy using the HGF gene. Further studies using well-designed larger placebo-controlled and long-term randomized control trials (RCTs) will clarify the effectiveness of gene therapy and cell-based therapy for the treatment of CLI. Furthermore, the development of efficient gene transfer systems and effective methods for keeping transplanted cells healthy will make these novel therapies more effective and ease the symptoms of CLI.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, 2-1 Yamadaoka, Suita 565-0817, Osaka, Japan
| | - Hironori Nakagami
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, 2-1 Yamadaoka, Suita 565-0817, Osaka, Japan
| | - Hiroshi Koriyama
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, and Hamamatsu University School of Medicine, 2-1 Yamadaoka, Suita 565-0817, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
21
|
Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, Krishnamurthy S, Anthony N, Pherwani A, Majumdar AS. A double blind randomized placebo controlled phase I/II study assessing the safety and efficacy of allogeneic bone marrow derived mesenchymal stem cell in critical limb ischemia. J Transl Med 2013; 11:143. [PMID: 23758736 PMCID: PMC3688296 DOI: 10.1186/1479-5876-11-143] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 05/27/2013] [Indexed: 12/15/2022] Open
Abstract
Background Peripheral vascular disease of the lower extremities comprises a clinical spectrum that extends from no symptoms to presentation with critical limb ischemia (CLI). Bone marrow derived Mesenchymal Stem Cells (BM- MSCs) may ameliorate the consequences of CLI due to their combinatorial potential for inducing angiogenesis and immunomodulatory environment in situ. The primary objective was to determine the safety of BM- MSCs in patients with CLI. Methods Prospective, double blind randomized placebo controlled multi-center study was conducted in patients with established CLI as per Rutherford classification in category II-4, III-5, or III-6 with infra-inguinal arterial occlusive disease and were not suitable for or had failed revascularization treatment. The primary end point was incidence of treatment – related adverse events (AE). Exploratory efficacy end points were improvement in rest pain, increase in Ankle Brachial Pressure Index (ABPI), ankle pressure, healing of ulcers, and amputation rates. Twenty patients (BM-MSC: Placebo = 1:1) were administered with allogeneic BM-MSCs at a dose of 2 million cells/kg or placebo (PlasmaLyte A) at the gastrocnemius muscle of the ischemic limb. Results Improvement was observed in the rest pain scores in both the arms. Significant increase in ABPI and ankle pressure was seen in BM-MSC arm compared to the placebo group. Incidence of AEs in the BM-MSC arm was 13 vs. 45 in the placebo arm where as serious adverse events (SAE) were similar in both the arms (5 in BM-MSC and 4 in the placebo group). SAEs resulted in death, infected gangrene, amputations in these patients. It was observed that the SAEs were related to disease progression and not related to stem cells. Conclusion BM-MSCs are safe when injected IM at a dose of 2 million cells/kg body weight. Few efficacy parameters such as ABPI and ankle pressure showed positive trend warranting further studies. Trial registration NIH website (http://www.clinicaltrials.gov/ct2/show/NCT00883870)
Collapse
Affiliation(s)
- Pawan K Gupta
- Stempeutics Research Pvt Ltd, Akshay Tech Park, No, 72 & 73, 2nd Floor, EPIP Zone, Phase I-Area, Whitefield, Bangalore 560066, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Minamimura A, Ichioka S, Sano H, Sekiya N. Comparison of collagen matrix treatment impregnated with platelet rich plasma vs bone marrow. J Plast Surg Hand Surg 2013; 48:15-20. [DOI: 10.3109/2000656x.2013.793193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
23
|
Nishimoto S, Kawai K, Tsumano T, Fukuda K, Fujiwara T, Kakibuchi M. Impacts of bone marrow aspirate and peripheral blood derived platelet-rich plasma on the wound healing in chronic ischaemic limb. J Plast Surg Hand Surg 2013; 47:169-74. [PMID: 23621096 DOI: 10.3109/2000656x.2012.752739] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Platelet rich plasma (PRP) has attracted attention as a safe and cost-effective source of growth factors that stimulate cells to regenerate tissue. Bone marrow cells are also estimated as an effective material for treating chronic ulcers. With the same technique to concentrate PRP from peripheral blood, bone marrow aspirate was processed and marrow cells were concentrated as well as platelets. Impact of PRP derived from bone marrow aspirate (bm-PRP) and that from peripheral blood (pb-PRP) on wound healing of persistent ischaemic rabbits' limbs were observed. Full thickness skin defects were made on the thighs, which had been treated to be persistent ischaemic status 3 weeks previously. Saline, pb-PRP, and bm-PRP were injected into the wound floor, respectively. Skin defected areas on ischaemic limbs were significantly wider than those on non-ischaemic limbs. bm-PRP injected wounds showed a significantly smaller skin defect area compared with pb-PRP and ischaemic-saline wounds at all time points. Fluorescently dyed cells of bm-PRP, injected into the wounds, could be traced 4 weeks after, whereas those of pb-PRP could be traced no more than 2 weeks. Wound healing on an ischaemic limb was accelerated with bm-PRP, whereas pb-PRP could not show any significance from saline. This difference can be attributed to the kind of cells contained in the PRPs. Injection of bm-PRP is a good candidate for treating wounds on ischaemic limbs.
Collapse
Affiliation(s)
- Soh Nishimoto
- Department of Plastic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8131, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Intra-arterial Allogeneic Mesenchymal Stem Cells for Critical Limb Ischemia are Safe and Efficacious: Report of a Phase I Study. World J Surg 2013; 37:915-22. [DOI: 10.1007/s00268-012-1892-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Brenes RA, Bear M, Jadlowiec C, Goodwin M, Hashim P, Protack CD, Ziegler KR, Li X, Model LS, Lv W, Collins MJ, Dardik A. Cell-based interventions for therapeutic angiogenesis: review of potential cell sources. Vascular 2012; 20:360-8. [PMID: 23086985 DOI: 10.1258/vasc.2011.201205] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Alternative therapies are currently being developed to treat patients with chronic limb ischemia who are unable to be revascularized in order to avoid amputation. Cell-based therapy using mononuclear cells is gaining attention as many clinical trials are currently underway. We review cell differentiation along with the different potential cell sources for use in therapeutic angiogenesis.
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
|
26
|
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: 75] [Impact Index Per Article: 6.3] [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
|
27
|
Ouma GO, Jonas RA, Usman MHU, Mohler ER. Targets and delivery methods for therapeutic angiogenesis in peripheral artery disease. Vasc Med 2012; 17:174-92. [PMID: 22496126 PMCID: PMC3760002 DOI: 10.1177/1358863x12438270] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Therapeutic angiogenesis utilizing genetic and cellular modalities in the treatment of arterial obstructive diseases continues to evolve. This is, in part, because the mechanism of vasculogenesis, angiogenesis, and arteriogenesis (the three processes by which the body responds to obstruction of large conduit arteries) is a complex process that is still under investigation. To date, the majority of human trials utilizing molecular, genetic, and cellular modalities for therapeutic angiogenesis in the treatment of peripheral artery disease (PAD) have not shown efficacy. Consequently, the current available knowledge is yet to be translated into novel therapeutic approaches for the treatment of PAD. The aim of this review is to discuss relevant scientific and clinical advances in therapeutic angiogenesis and their potential application in the treatment of ischemic diseases of the peripheral arteries. Additionally, this review article discusses past and recent developments, such as some unconventional approaches that have the potential to be applied as therapeutic targets. The article also includes advances in the delivery of genetic, cellular, and bioactive endothelial growth factors.
Collapse
Affiliation(s)
- Geoffrey O Ouma
- Department of Medicine, Cardiovascular Division, Vascular Medicine Section, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | |
Collapse
|
28
|
Kirana S, Stratmann B, Prante C, Prohaska W, Koerperich H, Lammers D, Gastens MH, Quast T, Negrean M, Stirban OA, Nandrean SG, Götting C, Minartz P, Kleesiek K, Tschoepe D. Autologous stem cell therapy in the treatment of limb ischaemia induced chronic tissue ulcers of diabetic foot patients. Int J Clin Pract 2012; 66:384-93. [PMID: 22284892 DOI: 10.1111/j.1742-1241.2011.02886.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIM Despite improvements in surgical revascularisation, limitations like anatomical factors or atherosclerosis limit the success of revascularisation in diabetic patients with critical limb ischaemia. Stem cells were shown to improve microcirculation in published studies. The aim of this study was to evaluate safety, feasibility and efficacy of transplantation of bone marrow derived cellular products regarding improvement in microcirculation and lowering of amputation rate. METHODS Bone marrow mononuclear cells (BMCs) in comparison with expanded bone marrow cells enriched in CD90+ cells ('tissue repair cells', TRCs) were used in the treatment of diabetic ulcers to induce revascularisation. Diabetic foot patients with critical limb ischaemia without option for surgical or interventional revascularisation were eligible. Parameters examined were ABI, TcPO(2) , reactive hyperaemia and angiographic imaging before and after therapy. RESULTS Of 30 patients included in this trial, 24 were randomised to receive either BMCs or TRCs. The high number of drop-outs in the control group (4 of 6) led to exclusion from evaluation. A total of 22 patients entered treatment; one patient in the TRC group and two in the BMC group did not show wound healing during follow up, one patient in each treatment group died before reaching the end of the study; one after having achieved wound healing (BMC group), the other one without having achieved wound healing (TRC group). Thus, 18 patients showed wound healing after 45 weeks. The total number of applicated cells was 3.8 times lower in the TRC group, but TRC patients received significantly higher amounts of CD90+ cells. Improvement in microvascularisation was detected in some, but not all patients by angiography, TcPO(2) improved significantly compared with baseline in both therapy groups. CONCLUSION The transplantation of BMCs as well as TRCs proved to be safe and feasible. Improvements of microcirculation and complete wound healing were observed in the transplant groups.
Collapse
Affiliation(s)
- S Kirana
- Diabetes Centre, Heart and Diabetes Centre NRW, Ruhr University Bochum, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
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: 4.4] [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
|
30
|
Diabetic impairment of C-kit bone marrow stem cells involves the disorders of inflammatory factors, cell adhesion and extracellular matrix molecules. PLoS One 2011; 6:e25543. [PMID: 21984919 PMCID: PMC3184966 DOI: 10.1371/journal.pone.0025543] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 09/05/2011] [Indexed: 12/21/2022] Open
Abstract
Bone marrow stem cells from diabetes mellitus patients exhibit functional impairment, but the relative molecular mechanisms responsible for this impairment are poorly understood. We investigated the mechanisms responsible for diabetes-related functional impairment of bone marrow stem cells by extensively screening the expression levels of inflammatory factors, cell cycle regulating molecules, extracellular matrix molecules and adhesion molecules. Bone marrow cells were collected from type 2 diabetic (db/db) and healthy control (db/m+) mice, and c-kit+ stem cells were purified (purity>85%) for experiments. Compared with the healthy control mice, diabetic mice had significantly fewer c-kit+ stem cells, and these cells had a lower potency of endothelial differentiation; however, the production of the angiogenic growth factor VEGF did not differ between groups. A pathway-focused array showed that the c-kit+ stem cells from diabetic mice had up-regulated expression levels of many inflammatory factors, including Tlr4, Cxcl9, Il9, Tgfb1, Il4, and Tnfsf5, but no obvious change in the expression levels of cell cycle molecules. Interestingly, diabetes-related alterations of the extracellular matrix and adhesion molecules were varied; Pecam, Mmp10, Lamc1, Itgb7, Mmp9, and Timp4 were up-regulated, but Col11a1, Fn1, Admts2, and Itgav were down-regulated. Some of these changes were also confirmed at the protein level by flow cytometry analysis. In conclusion, c-kit+ bone marrow stem cells from diabetic mice exhibited an extensive enhancement of inflammatory factors and disorders of the extracellular matrix and adhesion molecules. Further intervention studies are required to determine the precise role of each molecule in the diabetes-related functional impairment of c-kit+ bone marrow stem cells.
Collapse
|
31
|
Subrammaniyan R, Amalorpavanathan J, Shankar R, Rajkumar M, Baskar S, Manjunath SR, Senthilkumar R, Murugan P, Srinivasan VR, Abraham S. Application of autologous bone marrow mononuclear cells in six patients with advanced chronic critical limb ischemia as a result of diabetes: our experience. Cytotherapy 2011; 13:993-9. [PMID: 21671823 DOI: 10.3109/14653249.2011.579961] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND AIMS Previous clinical studies have reported that the injection of bone marrow (BM)-derived mononuclear cells (MNC) results in improvement in symptoms and healing of ulcers in patients with critical limb ischemia (CLI) up to stage IV of Fontaine's classification. However, most patients with Fontaine stage IV CLI limbs had to undergo amputation even after stem cell therapy. We report on six patients, who had poorly controlled diabetes with extensive ulceration and gangrene of limbs because of Fontaine stage IV CLI and had been advised amputation elsewhere, who underwent injection of autologous BM MNC. METHODS In all six patients, BM was aspirated and the isolated MNC from the BM were injected intralesionally at various sites of the ulcer and its surroundings after necessary debridement. The patients were followed up at regular intervals for at least 6 months. RESULTS At the end of the 6-month follow-up, the lower limb pain and ulcers had improved significantly in all patients. The mean toe-brachial index had increased from 0.26 to 0.36. One patient died a month after therapy because of causes unrelated to the procedure. Limb salvage was possible in the remaining five patients and they had a pain-free walking distance of 100 m within 6 months. CONCLUSIONS Limb salvage was possible in all six diabetic patients with Fontaine stage IV CLI following autologous BM MNC injection. The procedure was safe without any adverse outcomes.
Collapse
|
32
|
Franz RW, Shah KJ, Johnson JD, Pin RH, Parks AM, Hankins T, Hartman JF, Wright ML. Short- to mid-term results using autologous bone-marrow mononuclear cell implantation therapy as a limb salvage procedure in patients with severe peripheral arterial disease. Vasc Endovascular Surg 2011; 45:398-406. [PMID: 21669864 DOI: 10.1177/1538574411405545] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Short- to mid-term results of a prospective study evaluating dual intramuscular and intra-arterial autologous bone-marrow mononuclear cell (BM-MNC) implantation for the treatment of patients with severe peripheral arterial occlusive disease (PAD) in whom amputation was considered the only viable treatment option are presented. Ankle-brachial indices (ABIs), rest pain, and ulcer healing were assessed at 3 months. Success was defined as improvement in ABI measurements; absence of rest pain; absence of ulcers; and absence of major limb amputations. Twenty patients (21 limbs) have been enrolled. Three-month follow-up evaluation accounting included 18 patients (19 limbs). Four (22.2%) major and 2 (11.1%) minor amputations were performed within 3 months postoperatively. With 17 (94.4%) of 18 limbs demonstrating at least one criterion for success and major amputation avoided in 14 (77.8%) of 18 limbs at the 3-month evaluation, this specific BM-MNC implantation technique is an effective limb salvage strategy for patients with severe PAD.
Collapse
Affiliation(s)
- Randall W Franz
- Grant Medical Center, Vascular and Vein Center, Columbus, OH, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Autologous bone marrow mononuclear cell therapy is safe and promotes amputation-free survival in patients with critical limb ischemia. J Vasc Surg 2011; 53:1565-74.e1. [PMID: 21514773 DOI: 10.1016/j.jvs.2011.01.074] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 01/27/2011] [Accepted: 01/28/2011] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The purpose of this Phase I open label nonrandomized trial was to assess the safety and efficacy of autologous bone marrow mononuclear cell (ABMNC) therapy in promoting amputation-free survival (AFS) in patients with critical limb ischemia (CLI). METHODS Between September 2005 and March 2009, 29 patients (30 limbs), with a median age of 66 years (range, 23-84 years; 14 male, 15 female) with CLI were enrolled. Twenty-one limbs presented with rest pain (RP), six with RP and ulceration, and three with ulcer only. All patients were not candidates for surgical bypass due to absence of a patent artery below the knee and/or endovascular approaches to improving perfusion was not possible as determined by an independent vascular surgeon. Patients were treated with an average dose of 1.7 ± 0.7 × 10(9) ABMNC injected intramuscularly in the index limb distal to the anterior tibial tuberosity. The primary safety end point was accumulation of serious adverse events, and the primary efficacy end point was AFS at 1 year. Secondary end points at 12 weeks posttreatment were changes in first toe pressure (FTP), toe-brachial index (TBI), ankle-brachial index (ABI), and transcutaneous oxygen measurements (TcPO(2)). Perfusion of the index limb was measured with positron emission tomography-computed tomography (PET-CT) with intra-arterial infusion of H(2)O(15). RP, using a 10-cm visual analogue scale, quality of life using the VascuQuol questionnaire, and ulcer healing were assessed at each follow-up interval. Subpopulations of endothelial progenitor cells were quantified prior to ABMNC administration using immunocytochemistry and fluorescent-activated cell sorting. RESULTS There were two serious adverse events; however, there were no procedure-related deaths. Amputation-free survival at 1 year was 86.3%. There was a significant increase in FTP (10.2 ± 6.2 mm Hg; P = .02) and TBI (0.10 ± 0.05;P = .02) and a trend in improvement in ABI (0.08 ± 0.04; P = .73). Perfusion index by PET-CT H(2)O(15) increased by 19.3 ± 3.1, and RP decreased significantly by 2.2 ± 0.6 cm (P = .02). The VascuQol questionnaire demonstrated significant improvement in quality of life, and three of nine ulcers (33%) healed completely. KDR(+) but not CD34(+) or CD133(+) subpopulations of ABMNC were associated with improvement in limb perfusion. CONCLUSION This Phase I study has demonstrated safety, and the AFS rates suggest efficacy of ABMNC in promoting limb salvage in "no option" CLI. Based on these results, we plan to test the concept that ABMNCs improve AFS at 1 year in a Phase III randomized, double-blinded, multicenter trial.
Collapse
|
34
|
Idei N, Soga J, Hata T, Fujii Y, Fujimura N, Mikami S, Maruhashi T, Nishioka K, Hidaka T, Kihara Y, Chowdhury M, Noma K, Taguchi A, Chayama K, Sueda T, Higashi Y. Autologous bone-marrow mononuclear cell implantation reduces long-term major amputation risk in patients with critical limb ischemia: a comparison of atherosclerotic peripheral arterial disease and Buerger disease. Circ Cardiovasc Interv 2011; 4:15-25. [PMID: 21205941 DOI: 10.1161/circinterventions.110.955724] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Bone-marrow mononuclear cell (BM-MNC) implantation improves ischemic symptoms in patients with critical limb ischemia (CLI). The purpose of this study was to evaluate long-term clinical outcomes after autologous BM-MNC implantation in patients with CLI. METHODS AND RESULTS We assessed long-term clinical outcomes after BM-MNC implantation in 51 patients with CLI, including 25 patients with peripheral arterial disease (PAD) and 26 patients with Buerger disease. Forty-six CLI patients who had no BM-MNC implantation served as control subjects. Median follow-up period was 4.8 years. The 4-year amputation-free rates after BM-MNC implantation were 48% in PAD patients and 95% in Buerger disease, and they were 0% in control PAD patients and 6% in control Buerger disease. The 4-year overall survival rates after BM-MNC implantation were 76% in PAD patients and 100% in Buerger disease, and they were 67% in control PAD patients and 100% in control Buerger disease. Multivariable Cox proportional hazards analysis revealed that BM-MNC implantation correlated with prevention of major amputation and that hemodialysis and diabetes mellitus correlated with major amputation. In Buerger disease, ankle brachial pressure index and transcutaneous oxygen pressure were significantly increased after 1 month and remained high during 3-year follow-up. However, in patients with PAD, ankle brachial pressure index and transcutaneous oxygen pressure significantly increased after 1 month and gradually decreased during 3-year follow-up and returned to baseline levels. CONCLUSIONS These findings suggest that BM-MNC implantation is safe and effective in patients with CLI, especially in patients with Buerger disease. Clinical Trial Registration- URL: http://home.hiroshima-u.ac.jp/angio/. Unique identifier: 001769.
Collapse
Affiliation(s)
- Naomi Idei
- Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Therapeutic angiogenesis by autologous bone marrow cell implantation together with allogeneic cultured dermal substitute for intractable ulcers in critical limb ischaemia. J Plast Reconstr Aesthet Surg 2010; 63:1875-82. [DOI: 10.1016/j.bjps.2009.11.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Revised: 09/27/2009] [Accepted: 11/26/2009] [Indexed: 10/20/2022]
|
36
|
Zafarghandi MR, Ravari H, Aghdami N, Namiri M, Moazzami K, Taghiabadi E, Fazel A, Pournasr B, Farrokhi A, Sharifian RA, Salimi J, Moini M, Baharvand H. Safety and efficacy of granulocyte–colony-stimulating factor administration following autologous intramuscular implantation of bone marrow mononuclear cells: a randomized controlled trial in patients with advanced lower limb ischemia. Cytotherapy 2010; 12:783-91. [DOI: 10.3109/14653240903518163] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Onodera R, Teramukai S, Tanaka S, Kojima S, Horie T, Matoba S, Murohara T, Matsubara H, Fukushima M. Bone marrow mononuclear cells versus G-CSF-mobilized peripheral blood mononuclear cells for treatment of lower limb ASO: pooled analysis for long-term prognosis. Bone Marrow Transplant 2010; 46:278-84. [DOI: 10.1038/bmt.2010.110] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
Lara-Hernandez R, Lozano-Vilardell P, Blanes P, Torreguitart-Mirada N, Galmés A, Besalduch J. Safety and efficacy of therapeutic angiogenesis as a novel treatment in patients with critical limb ischemia. Ann Vasc Surg 2010; 24:287-94. [PMID: 20142004 DOI: 10.1016/j.avsg.2009.10.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 09/23/2009] [Accepted: 10/05/2009] [Indexed: 11/28/2022]
Abstract
BACKGROUND In some patients with critical limb ischemia (CLI) the possibility of revascularizing treatment does not exist. In this case therapeutic angiogenesis (TA) using autologous endothelial progenitor cell (EPC) transplantation could be an alternative. The objective of our study was to evaluate the safety and efficacy of TA using EPC. METHODS Twenty-eight patients with CLI who were not candidates for surgical or endovascular revascularization were included in a prospective study. To mobilize EPCs from the bone marrow, granulocyte colony-stimulating growth factor was injected subcutaneously at doses of 5 microg/kg/day for 5 days. Apheresis was performed, obtaining 50 mL of blood with a high rate of EPCs (CD34(+) and CD133(+) cells were counted). EPCs were implanted in the ischemic limb by intramuscular injections. Primary end points were the safety and feasibility of the procedure and limb salvage rate for amputation at 12 months. Other variables studied were improvement in rest pain, healing of ulcers, ankle-brachial pressure index (ABI), and digital plethysmography. All procedures were done pretreatment and every 3 months for a year on average. Postransplantation arteriography was done in selected cases. RESULTS No adverse effects were observed. Mean follow-up was 14 months. Before treatment, mean basal ABI was 0.35+/-0.2 and at 18 months postimplantation, 0.72+/-0.51 (p=0.009). There was a mean decrease of five points in pain scale: basal 8.7+/-1, after TA 3.8+/-2.9 (p=0.01). Seven patients required major amputation. Kaplan-Meier analysis revealed a limb salvage rate of 74.4% after 1 year. CONCLUSION Implantation of EPCs in CLI is a safe alternative, improves tissue perfusion, and obtains high amputation-free rates. Nevertheless, this is a small cohort and results should be tested with long randomized trials.
Collapse
Affiliation(s)
- R Lara-Hernandez
- Vascular Surgery Department, Hospital Universitario Son Dureta, Palma de Mallorca, Baleares, Spain.
| | | | | | | | | | | |
Collapse
|
39
|
Sauerbier S, Stricker A, Kuschnierz J, Bühler F, Oshima T, Xavier SP, Schmelzeisen R, Gutwald R. In VivoComparison of Hard Tissue Regeneration with Human Mesenchymal Stem Cells Processed with Either the FICOLL Method or the BMAC Method. Tissue Eng Part C Methods 2010; 16:215-23. [DOI: 10.1089/ten.tec.2009.0269] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Sebastian Sauerbier
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Andres Stricker
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Jens Kuschnierz
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Felicia Bühler
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Toshiyuki Oshima
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
- Faculty of Health Sciences, Institute for International Cooperation, Okayama, Japan
| | - Samuel Porfirio Xavier
- Department of Oral Maxillofacial Surgery and Periodontology, Faculty of Dentistry of University of Sao Paulo, Ribeirao Preto, Brazil
| | - Rainer Schmelzeisen
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Ralf Gutwald
- Department of Oral and Craniomaxillofacial Surgery, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
40
|
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: 2.2] [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
|
41
|
Intraoperative adjunctive stem cell treatment in patients with critical limb ischemia using a novel point-of-care device. Ann Vasc Surg 2009; 24:367-72. [PMID: 19896796 DOI: 10.1016/j.avsg.2009.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Revised: 03/22/2009] [Accepted: 07/27/2009] [Indexed: 11/20/2022]
Abstract
INTRODUCTION In a prospective trial we tested whether adjunctive intraoperative stem cell treatment in patients with critical limb ischemia (CLI) can be performed safely in combination with bypass surgery and/or interventional treatment. The end point of our study was the safety and integrity of a novel point-of-care system used in patients with CLI. METHODS We included only patients with CLI and tissue loss according to Rutherford categories 4-6. The Harvest Bone Marrow Aspirate Concentrate System consists of an automated, microprocessor-controlled dedicated centrifuge with decanting capability and the accessory BMAC Pack for processing a patient's bone marrow aspirate (BMA). The centrifuge is portable and enables BMA to be rapidly processed in the operating room to provide an autologous concentrate of nucleated cells for immediate injection. The surgeon aspirated 120 ml BMA from the iliac crest. RESULTS Eight consecutive patients were treated according to the study protocol. The mean follow-up period was 9.2 months (range 2-18). Stem cells were always injected during the final revascularization attempt. One minor amputation and two major amputations were required. In five of eight patients there was a discrete increase in the ankle-brachial index post-stem cell treatment. The dose of stem cells after centrifugation was 17.2 (range 13.8-54.2)x10E6 CD34-positive cells and 7.8 (range 1.8-35.9)x10E6 CD133-positive cells. The injected dose of VEGFR-2-coexpressing stem cells was 0.5-5.7x10E4. CONCLUSION We were able to show that the buffy coat preparation using a point-of-care system is a simple and fast method to enrich stem cells from BMAs. This automated system gives high recovery rates and good reproducibility.
Collapse
|
42
|
Franz RW, Parks A, Shah KJ, Hankins T, Hartman JF, Wright ML. Use of autologous bone marrow mononuclear cell implantation therapy as a limb salvage procedure in patients with severe peripheral arterial disease. J Vasc Surg 2009; 50:1378-90. [PMID: 19837539 DOI: 10.1016/j.jvs.2009.07.113] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 07/24/2009] [Accepted: 07/30/2009] [Indexed: 11/26/2022]
Abstract
BACKGROUND Few options other than amputation exist for some patients with peripheral arterial occlusive disease (PAD) and severe anatomical limitations. METHODS This prospective study presents short-term results of dual intramuscular and intra-arterial autologous bone marrow mononuclear cell (BM-MNC) implantation for the treatment of patients with severe PAD in whom amputation was considered the only viable treatment option. Baseline, two-week, and three-month evaluations were conducted. Ankle brachial indices (ABI) were calculated for both the dorsal pedis and the posterior tibial arteries. Rest pain and ulcer healing also were assessed. Success was defined as meeting the following four criteria: improvement in ABI measurements; relief of rest pain; ulcer healing, if applicable; and absence of major limb amputations. Patients not undergoing major limb amputations continued to be monitored for subsequent procedures. RESULTS Nine patients for whom limb amputation was recommended underwent this procedure. The study population was comprised of five females and four males, with a mean age of 61.7 years. Eight (88.9%) patients had rest pain. Seven (77.8%) patients also had diabetes. Non-healing ulcers were present in eight (88.9%) cases. After the procedure, non-significant improvements of 0.12 and 0.08 in ABI were observed for the dorsalis pedis and posterior tibial ankle arteries, respectively. Three (33.3%) major amputations subsequently were performed, including a below-knee amputation 4.1 weeks after the BM-MNC implantation and two above-knee amputations at 5.4 and 11.0 weeks after the procedure. The six (66.7%) patients who did not have major amputations demonstrated improvement in symptom severity three months after the procedure, as evidenced by alleviation of rest pain and improvements by at least one level in Rutherford and Fontaine classifications, and have not required amputations at a mean follow-up of 7.8 months. Complete wound healing was achieved within three months in all patients who had ulcers prior to BM-MNC implantation and for whom amputation was not required. This specific BM-MNC implantation technique was fully successful in three (33.3%) patients, as major amputation was avoided and the other applicable criteria were met. Five (55.6%) additional patients demonstrated success in at least one of the four criteria. CONCLUSIONS With eight (88.9%) of nine patients showing some level of improvement and amputation avoided in six (66.7%) patients, these short-term results indicate the use of BM-MNC implantation as a means of limb salvage therapy for patients with severe PAD shows promise in postponing or avoiding amputation in a patient population currently presented with few alternatives to amputation.
Collapse
Affiliation(s)
- Randall W Franz
- Vascular and Vein Center at Grant Medical Center, Columbus, Ohio 43215, USA.
| | | | | | | | | | | |
Collapse
|
43
|
van Tongeren RB, Hamming JF, le Cessie S, van Erkel AR, van Bockel JH. Limited value of digital subtraction angiography in the evaluation of cell-based therapy in patients with limb ischemia. Int J Cardiovasc Imaging 2009; 26:19-25. [PMID: 19757148 DOI: 10.1007/s10554-009-9507-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 08/29/2009] [Indexed: 01/11/2023]
Abstract
Cell-based therapy has been proposed as a novel strategy for patients with severe peripheral arterial disease by stimulating vascular growth. In clinical studies of this therapy, the emphasis has been on demonstrating recovery of clinical parameters, rather than on evaluation of blood flow recovery. Angiography is still the gold standard for the assessment of lower leg arteries. Therefore, we studied the usefulness of angiography in the evaluation of cell-based therapy. Sixteen patients with critical leg ischemia (ischemic rest pain or ulcers), or persistent (>12 months) profound disabling claudication were unilaterally treated with autologous bone marrow-derived mononuclear cells. Pre- and 6 months post-treatment digital subtraction angiographies (DSA) were assessed and compared in a blinded fashion twice by a panel of seven vascular surgeons and interventional radiologists. Inter- and intraobserver variability on qualitative (poor/moderate/rich) and semi-quantitative (increase/no difference/decrease) assessment of collateral circulation were evaluated. Agreement was expressed inter- and intraclass correlation coefficients (CC). Inter- and intraobserver agreement was moderate for the qualitative grading of collateral extent (CC = 0.46 and 0.60, respectively). Agreement was moderate (inter-CC = 0.60) to good (intra-CC = 0.73) for comparing pre- and post-treatment DSA. Clinical response was based on limb salvage, pain-free walking distance, ankle-brachial pressure index and pain scores. No difference was observed in the extent of collateral circulation between pre- and post treatment DSA after separate analysis of clinical responding and non-responding patients (P = 0.92). DSA is not a suited modality for the evaluation of therapeutic angiogenesis.
Collapse
Affiliation(s)
- Robert B van Tongeren
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
44
|
Prather WR, Toren A, Meiron M, Ofir R, Tschope C, Horwitz EM. The role of placental-derived adherent stromal cell (PLX-PAD) in the treatment of critical limb ischemia. Cytotherapy 2009; 11:427-34. [PMID: 19526389 DOI: 10.1080/14653240902849762] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) are spindle-shaped plastic-adherent cells isolated from bone marrow (BM), adipose tissue and other organs, including the placenta. Autologous BM-derived MSC have been studied in animals with experimentally induced critical limb ischemia (CLI) as a model of end-stage peripheral vascular disease. While demonstrating therapeutic benefit, the use of these cells is limited by the need to surgically extract BM and the fear of thrombosis secondary to the use of granulocyte-colony-stimulating factor (G-CSF) to mobilize the cells. METHODS We studied the use of placental-derived adherent stromal cells (ASC) in a standard limb ischemia model of male Balb/c mice. These placental-derived cells, termed PLX-PAD, share the adherence and marker expression of BM-derived MSC but lack their differentiation potential. PLX-PAD are isolated from human placenta following a Caesarean section and cultured in a bioreactor, termed the PluriX System. The PluriX System provides a three-dimensional (3-D) microenvironment that enables the large-scale growth of these cells. PLX-PAD are stable adhesive cells that can be expanded in vitro without the loss of phenotype and without showing signs of karyotypic changes. RESULTS The intramuscular (i.m.) administration of PLX-PAD in our model significantly improved blood flow (BF) (P=0.0008), increased capillary density (P=0.021), reduced oxidative stress (P=0.034) and reduced endothelial damage (P=0.004), while increasing limb function versus the administration of a phosphate-buffered saline (PBS) control vehicle in the affected limb. CONCLUSIONS Allogeneic placental-derived ASC may provide an off-the-shelf supply of therapeutic cells that would need no histocompatible tissue matching and be potentially less expensive and considerably more convenient than BM or adipose-derived MSC.
Collapse
|
45
|
Gutwald R, Haberstroh J, Kuschnierz J, Kister C, Lysek DA, Maglione M, Xavier SP, Oshima T, Schmelzeisen R, Sauerbier S. Mesenchymal stem cells and inorganic bovine bone mineral in sinus augmentation: comparison with augmentation by autologous bone in adult sheep. Br J Oral Maxillofac Surg 2009; 48:285-90. [PMID: 19665265 DOI: 10.1016/j.bjoms.2009.06.226] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2009] [Indexed: 02/06/2023]
Abstract
Our aim was to compare the osteogenic potential of mononuclear cells harvested from the iliac crest combined with bovine bone mineral (BBM) (experimental group) with that of autogenous cancellous bone alone (control group). We studied bilateral augmentations of the sinus floor in 6 adult sheep. BBM and mononuclear cells (MNC) were mixed and placed into one side and autogenous bone in the other side. Animals were killed after 8 and 16 weeks. Sites of augmentation were analysed radiographically and histologically. The mean (SD) augmentation volume was 3.0 (1.0) cm(3) and 2.7 (0.3) cm(3) after 8 and 16 weeks in the test group, and 2.8 (0.3) cm(3) (8 weeks) and 2.8 (1.2) cm(3) (16 weeks) in the control group, respectively. After 8 weeks, histomorphometric analysis showed 24 (3)% BBM, and 19 (11)% of newly formed bone in the test group. The control group had 20 (13%) of newly formed bone. Specimens after 16 weeks showed 29 (12%) of newly formed bone and 19 (3%) BBM in the test group. The amount of newly formed bone in the control group was 16 (6%). The results show that mononuclear cells, including mesenchymal stem cells, in combination with BBM as the biomaterial, have the potential to form bone.
Collapse
Affiliation(s)
- Ralf Gutwald
- Universitätsklinik für Zahn-, Mund- und Kieferheilkunde, Abteilung Klinik und Poliklinik für Mund-, Kiefer- und Gesichtschirurgie, Hugstetter Str. 55, D-79106 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Nikolic B, Faintuch S, Goldberg SN, Kuo MD, Cardella JF. Stem Cell Therapy: A Primer for Interventionalists and Imagers. J Vasc Interv Radiol 2009; 20:999-1012. [DOI: 10.1016/j.jvir.2009.04.075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 04/24/2009] [Accepted: 04/28/2009] [Indexed: 02/06/2023] Open
|
47
|
Kubo M, Li TS, Kamota T, Ohshima M, Qin SL, Hamano K. Increased expression of CXCR4 and integrin alphaM in hypoxia-preconditioned cells contributes to improved cell retention and angiogenic potency. J Cell Physiol 2009; 220:508-14. [PMID: 19415696 DOI: 10.1002/jcp.21803] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cell-based angiogenesis is a promising method for the treatment of ischemic diseases, but the poor retention of implanted cells in targeted tissues is a major drawback. We tested whether hypoxic preconditioning increased retention and angiogenic potency of implanted cells in ischemic tissue. Hypoxic preconditioning of mouse peripheral blood mononuclear cells (PBMNCs) was done with 24 h of culture under 2% O(2). Normoxia-cultured PBMNCs were used as a control. Hypoxic preconditioning increased the adhesion capacity of the PBMNCs. Moreover, the expression of integrin alphaM and CXCR4 was significantly higher in the hypoxia-preconditioned PBMNCs than in the normoxia-cultured PBMNCs. Interestingly, the expression of intercellular adhesion molecule-1 (ICAM-1), a ligand of integrin alphaM, and stromal cell-derived factor-1 (SDF-1), a chemokine for CXCR4, were remarkably increased in the ischemic hindlimbs. The retention of the hypoxia-preconditioned PBMNCs was significantly higher than that of the normoxia-cultured PBMNCs, 3 days after their intramuscular implantation into ischemic hindlimbs. We also noted better blood flow in the ischemic hindlimbs implanted with the hypoxia-preconditioned PBMNCs than in those implanted with the normoxia-cultured PBMNCs, 14 days after treatment. Furthermore, antibody neutralization of integrin alphaM and CXCR4 abolished completely the increased cell retention and angiogenic potency of the hypoxia-preconditioned PBMNCs after implantation into the ischemic hindlimbs. These results indicate that hypoxic preconditioning of implanted cells is a feasible method of enhancing therapeutic angiogenesis by increasing their retention.
Collapse
Affiliation(s)
- Masayuki Kubo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
48
|
An update on therapeutic angiogenesis for peripheral vascular disease. Ann Vasc Surg 2009; 24:258-68. [PMID: 19467829 DOI: 10.1016/j.avsg.2008.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 12/11/2008] [Accepted: 12/21/2008] [Indexed: 12/22/2022]
Abstract
BACKGROUND We reviewed the issue of stem cells and therapeutic angiogenesis in the treatment of peripheral vascular disease. METHODS MEDLINE (1997-2008) with the following search terms: "stem cell therapy," "endothelial progenitor cells," "peripheral blood mononuclear cells," and "peripheral vascular disease." Relevant published papers involving the above search terms, preclinical studies, and clinical trials using stem cells and progenitors for the treatment of peripheral occlusive vascular disease were included. RESULTS Transplantation of bone marrow-derived progenitor cells or peripheral blood mononuclear cells promotes tissue angiogenesis, as has already been explored in preclinical studies; angiogenesis can also be sustained using genetic, protein therapeutic approaches. Engineered scaffolding with stem cells is a further strategy, which is still in its infancy. The treatment of patients with severe peripheral arterial disease is generally reported as a series of case reports; all studies generally show an improvement in clinical symptoms, e.g., rest pain and pain-free walking time, as well as transcutaneous oxygen pressure, without any important adverse reactions. The few clinical trials also report similar encouraging results. All the studies have their shortcomings, including absence of control groups and objective evaluation of the results of treatments as well as short-term follow-up. CONCLUSION Promoting angiogenesis using genetic, protein, stem cell-based therapies is a promising option for the treatment of peripheral vascular disease when unresponsive to medical and surgical therapy.
Collapse
|
49
|
Amann B, Luedemann C, Ratei R, Schmidt-Lucke JA. Autologous bone marrow cell transplantation increases leg perfusion and reduces amputations in patients with advanced critical limb ischemia due to peripheral artery disease. Cell Transplant 2009; 18:371-80. [PMID: 19500466 DOI: 10.3727/096368909788534942] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bone marrow cell transplantation has been shown to induce angiogenesis and thus improve ischemic artery disease. This study evaluates the effects of intramuscular bone marrow cell transplantation in patients with limb-threatening critical limb ischemia with a very high risk for major amputation. After failed or impossible operative and/or interventional revascularization and after unsuccessful maximum conservative therapy, 51 patients with impending major amputation due to severe critical limb ischemia had autologous bone marrow cells (BMC) transplanted into the ischemic leg. Patients 1-12 received Ficoll-isolated bone marrow mononuclear cells (total cell number 1.1 +/- 1.1 x 10(9)), patients 13-51 received point of care isolated bone marrow total nucleated cells (3.0 +/- 1.7 x 10(9)). Limb salvage was 59% at 6 months and 53% at last follow-up (mean 411 +/- 261 days, range 175-1186). Perfusion measured with ankle-brachial index (ABI) and transcutaneous oxygen tension (tcpO(2)) at baseline and after 6 months increased in patients with consecutive limb salvage (ABI 0.33 +/- 0.18 to 0.46 +/- 0.15, tcpO(2) 12 +/- 12 to 25 +/- 15 mmHg) and did not change in patients eventually undergoing major amputation. No difference in clinical outcome between the isolation methods were seen. Clinically most important, patients with limb salvage improved from a mean Rutherford category of 4.9 at baseline to 3.3 at 6 months (p = 0.0001). Analgesics consumption was reduced by 62%. Total walking distance improved in nonamputees from zero to 40 m. Three severe periprocedural adverse events resolved without sequelae, and no unexpected long-term adverse events occurred. In no-option patients with end-stage critical limb ischemia due to peripheral artery disease, bone marrow cell transplantation is a safe procedure that can improve leg perfusion sufficiently to reduce major amputations and permit durable limb salvage.
Collapse
Affiliation(s)
- Berthold Amann
- Department of Medicine, Franziskuskrankenhaus, Berlin Vascular Center, Berlin, Germany.
| | | | | | | |
Collapse
|
50
|
Ohshima M, Li TS, Kubo M, Qin SL, Hamano K. Antioxidant therapy attenuates diabetes-related impairment of bone marrow stem cells. Circ J 2008; 73:162-6. [PMID: 19023156 DOI: 10.1253/circj.cj-08-0123] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Bone marrow cells from humans and animals with diabetes exhibit decreased angiogenic potency, thought to be related to oxidative stress, so the present study investigated if antioxidant therapy would attenuate the diabetes-related impairment. METHODS AND RESULTS Diabetic mice were given antioxidant therapy, as a daily subcutaneous injection of superoxide dismutase-mimic (10 mg kg(-1) day(-1)). Diabetic and healthy mice given a vehicle treatment were used as the control. After 4 weeks of treatment, bone marrow mononuclear cells (BM-MNCs) were collected for analysis and the endothelial progenitor cells in BM-MNCs were evaluated by flow cytometry. The intracellular reactive oxygen species (ROS) levels in BM-MNCs were measured using 6-carboxy-2'7'-dichlorodihydrofluorescein diacetate. Endothelial differentiation from the BM-MNCs was estimated by immunostaining with VE-cadherin 7 days after culture. BM-MNCs from the control diabetic mice had fewer Flk-1/CD34 double-positive progenitor cells and higher intracellular ROS levels, with lower potency of endothelial differentiation than BM-MNCs from the healthy mice. Antioxidant therapy decreased the intracellular ROS level in BM-MNCs from that in the diabetic mice significantly (P<0.05), but increased significantly the percentage of endothelial progenitor cells (P<0.05) and their potency of differentiation into endothelial cells (P<0.05). CONCLUSIONS Antioxidant therapy attenuated the diabetes-related impairment of BM-MNCs by reducing oxidative stress.
Collapse
Affiliation(s)
- Mako Ohshima
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | | | | | | | | |
Collapse
|