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
|
Jiang X, Liu H, Pan T, Gu S, Fang Y, Wei Z, Fang G, Chen B, Jiang J, Shi Y, Liu P, Fu W, Dong Z. Long-Term Outcomes of Peripheral Blood Mononuclear Cells in the Treatment of Angiitis-Induced No-Option Critical Limb-Threatening Ischemia. Front Cardiovasc Med 2021; 8:769472. [PMID: 34938786 PMCID: PMC8687358 DOI: 10.3389/fcvm.2021.769472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Peripheral blood mononuclear cells (PBMNCs) showed encouraging short outcomes in the treatment of angiitis-induced no-option critical limb-threatening ischemia (AICLTI) in the pilot study. This study aimed to demonstrate the long-term outcomes of this treatment. Methods: From May 2014 to December 2018, patients diagnosed with AICLTI and treated by autotransplantation of PBMNCs in our center were enrolled and analyzed. The primary endpoint was major amputation-free survival (MAFS), the secondary endpoints included peak pain-free walking time (PPFWT), Wong-Baker FACES pain rating scale score (WFPRSS), labor recovery, ankle-brachial index (ABI), transcutaneous partial oxygen pressure (TcpO2), and SF-36v2 scores. Results: A total of 58 patients were enrolled. During a minimal follow-up of 36 months, the MAFS was 93.1% and the labor competence restored rate was 62.1%. The WFPRSS was decreased from 8.7 ± 1.6 to 1.6 ± 3.2, and PPFWT was significantly improved from 2.9 ± 4.2 min to 16.6 ± 6.9 min. The quality of life was also significantly improved at each follow-up point. Perfusion evaluating parameters, such as ABI and TcPO2, were also significantly improved. No critical adverse event was observed during the treatment and follow-up period. Conclusions: The treatment of AICLTI by autotransplantation of PBMNCs demonstrated encouraging long-term results. It could not only restore labor competence, improve the quality of life, but also significantly reduce the major amputation rate.
Collapse
Affiliation(s)
- Xiaolang Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Liu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianyue Pan
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyang Gu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Fang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Wei
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gang Fang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Chen
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junhao Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shi
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihui Dong
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Bolaman AZ, Yavasoglu I. Intra-lesionally autologous stem cell application in diabetic foot/ulcer. Transfus Apher Sci 2021; 60:103282. [PMID: 34593331 DOI: 10.1016/j.transci.2021.103282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 12/09/2022]
Abstract
The diabetic fot/ulcer is the cause of high morbidity and mortality in patients with diabetes mellitus (DM). Generally, medical treatment of diabetic foot/ulcer is ineffective and stem cell implantation is an important option in the treatment. Here, we present a 69 years old man admitted to hospital due to a 3 × 4 cm wound in the plantar surface of left foot. Autologous stem cells were applied intralesionally into diabetic ulcers. The lesion shrunken 50 % at the 16th week and there is a wound under the left foot at 32nd week. Intralesionally autologous stem cell application was useful and safe without adverse course in patients with diabetic foot/ulcer.
Collapse
Affiliation(s)
- Ali Zahit Bolaman
- Aydin Adnan Menderes University, Department of Internal Medicine, Aydin, Turkey.
| | - Irfan Yavasoglu
- Aydin Adnan Menderes University, Department of Internal Medicine, Aydin, Turkey
| |
Collapse
|
4
|
Cho H, Blatchley MR, Duh EJ, Gerecht S. Acellular and cellular approaches to improve diabetic wound healing. Adv Drug Deliv Rev 2019; 146:267-288. [PMID: 30075168 DOI: 10.1016/j.addr.2018.07.019] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Chronic diabetic wounds represent a huge socioeconomic burden for both affected individuals and the entire healthcare system. Although the number of available treatment options as well as our understanding of wound healing mechanisms associated with diabetes has vastly improved over the past decades, there still remains a great need for additional therapeutic options. Tissue engineering and regenerative medicine approaches provide great advantages over conventional treatment options, which are mainly aimed at wound closure rather than addressing the underlying pathophysiology of diabetic wounds. Recent advances in biomaterials and stem cell research presented in this review provide novel ways to tackle different molecular and cellular culprits responsible for chronic and nonhealing wounds by delivering therapeutic agents in direct or indirect ways. Careful integration of different approaches presented in the current article could lead to the development of new therapeutic platforms that can address multiple pathophysiologic abnormalities and facilitate wound healing in patients with diabetes.
Collapse
Affiliation(s)
- Hongkwan Cho
- Wilmer Ophthalmologic Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael R Blatchley
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University Baltimore, MD, USA
| | - Elia J Duh
- Wilmer Ophthalmologic Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University Baltimore, MD, USA.
| |
Collapse
|
5
|
Abstract
Objective: We investigated the long-term results of autologous bone marrow mononuclear cells (ABMMNCs) implantation in patients with Buerger’s disease (BD). Methods: Twenty-eight patients (25 males and 3 females) who had BD and critical unilateral limb ischemia were investigated between April 2003 and August 2005. The patients were administered multiple injections of CD34+ and CD45+ positive ABMMNCs into the gastrocnemius muscle, the intermetatarsal region, and the dorsum of the foot (n=26) or forearm (n=2) and saline injection into the contralateral limb. Results: The mean follow-up time was 139.6±10.5 months. No complication related to stem cell therapy was observed during the follow-up. The ankle–brachial pressure index evaluated at 6 months and 120 months was compared to the baseline scores (p<0.001 and p=0.021, respectively). Digital subtraction angiography (DSA) was performed for all patients at baseline, 6 months, and 120 months. The angiographic improvement was 78.5% and 57.1% at 6 and 120 months, respectively. Patients demonstrated a significant improvement in the quality of life parameters at 6 months compared to baseline (p=0.008) and 120 months compared to the baseline (p=0.009). The 10-year amputation-free rate was 96% (95% CI=0.71-1) in ABMMNC-implanted limbs and 93% (95% CI=0.33–0.94) in saline-injected limbs (p=1). Conclusion: Autologous stem cell therapy could be an alternative therapeutic method for BD at long-term follow-up.
Collapse
|
6
|
In-Site Monocyte Implantation in Bone Grafting for Maxillary Atrophy Reconstruction: A Preliminary Observational Proof of Concept Study. IMPLANT DENT 2019; 27:529-541. [PMID: 30239370 DOI: 10.1097/id.0000000000000813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE The main objective of this study, involving 11 patients, is to share our experience on an integrated treatment modality, namely, the use of cellular therapy simultaneously with surgical reconstruction. Published works show that the implantation of monocytes in ischemic tissue enhances healing by providing neo-angiogenesis, a key mechanism in tissue regenerative processes. MATERIALS AND METHODS Our approach included the utilization of autologous monocytes and endothelial precursor cells in the bone graft itself to improve the success rate of the integration of the bone graft and its long-term viability/survival by promoting angiogenesis. We compared the standard regenerative procedures, namely sinus lift grafting performed with xenogeneic particle bone graft and posterior mandible grafting performed with on-lay or in-lay autologous cortical/medullary bone-block graft harvested from the iliac crest, with and without the use of cellular implementation. We evaluated results by both radiological and histological assessment. RESULTS Autologous cortical/medullary bone-block graft had a different response to implementation with monocytes, showing a better osteointegration than expected conversely to the xenogeneic particle bone graft. CONCLUSIONS Monocytes seem to improve autologous bone-block graft according to the "Therapeutic Angiogenesis" concept. Implementation with monocytes does not always improve xenogeneic particle bone graft.
Collapse
|
7
|
Lopes L, Setia O, Aurshina A, Liu S, Hu H, Isaji T, Liu H, Wang T, Ono S, Guo X, Yatsula B, Guo J, Gu Y, Navarro T, Dardik A. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther 2018; 9:188. [PMID: 29996912 PMCID: PMC6042254 DOI: 10.1186/s13287-018-0938-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Diabetic foot ulcer (DFU) is a severe complication of diabetes, preceding most diabetes-related amputations. DFUs require over US$9 billion for yearly treatment and are now a global public health issue. DFU occurs in the setting of ischemia, infection, neuropathy, and metabolic disorders that result in poor wound healing and poor treatment options. Recently, stem cell therapy has emerged as a new interventional strategy to treat DFU and appears to be safe and effective in both preclinical and clinical trials. However, variability in the stem cell type and origin, route and protocol for administration, and concomitant use of angioplasty confound easy interpretation and generalization of the results. METHODS The PubMed, Google Scholar, and EMBASE databases were searched and 89 preclinical and clinical studies were selected for analysis. RESULTS There was divergence between preclinical and clinical studies regarding stem cell type, origin, and delivery techniques. There was heterogeneous preclinical and clinical study design and few randomized clinical trials. Granulocyte-colony stimulating factor was employed in some studies but with differing protocols. Concomitant performance of angioplasty with stem cell therapy showed increased efficiency compared to either therapy alone. CONCLUSIONS Stem cell therapy is an effective treatment for diabetic foot ulcers and is currently used as an alternative to amputation for some patients without other options for revascularization. Concordance between preclinical and clinical studies may help design future randomized clinical trials.
Collapse
Affiliation(s)
- Lara Lopes
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
- Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ocean Setia
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Afsha Aurshina
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Haidi Hu
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Haiyang Liu
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Tun Wang
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Xiangjiang Guo
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tulio Navarro
- Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
- VA Connecticut Healthcare System, West Haven, CT USA
| |
Collapse
|
8
|
Horie T, Yamazaki S, Hanada S, Kobayashi S, Tsukamoto T, Haruna T, Sakaguchi K, Sakai K, Obara H, Morishita K, Saigo K, Shintani Y, Kubo K, Hoshino J, Oda T, Kaneko E, Nishikido M, Ioji T, Kaneda H, Fukushima M. Outcome From a Randomized Controlled Clinical Trial - Improvement of Peripheral Arterial Disease by Granulocyte Colony-Stimulating Factor-Mobilized Autologous Peripheral-Blood-Mononuclear Cell Transplantation (IMPACT). Circ J 2018; 82:2165-2174. [PMID: 29877199 DOI: 10.1253/circj.cj-17-1220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The clinical usefulness of peripheral blood (PB) mononuclear cell (MNC) transplantation in patients with peripheral arterial disease (PAD), especially in those with mild-to-moderate severity, has not been fully clarified.Methods and Results:A randomized clinical trial was conducted to evaluate the efficacy and safety of granulocyte colony-stimulating factor (G-CSF)-mobilized PBMNC transplantation in patients with PAD (Fontaine stage II-IV and Rutherford category 1-5) caused by arteriosclerosis obliterans or Buerger's disease. The primary endpoint was progression-free survival (PFS). In total, 107 subjects were enrolled. At baseline, Fontaine stage was II/III in 82 patients and IV in 21, and 54 patients were on hemodialysis. A total of 50 patients had intramuscular transplantation of PBMNC combined with standard of care (SOC) (cell therapy group), and 53 received SOC only (control group). PFS tended to be improved in the cell therapy group than in the control group (P=0.07). PFS in Fontaine stage II/III subgroup was significantly better in the cell therapy group than in the control group. Cell therapy-related adverse events were transient and not serious. CONCLUSIONS In this first randomized, large-scale clinical trial of G-CSF-mobilized PBMNC transplantation, the cell therapy was tolerated by a variety of PAD patients. The PBMNC therapy was significantly effective for inhibiting disease progression in mild-to-moderate PAD.
Collapse
Affiliation(s)
| | - Seiji Yamazaki
- Cardiovascular Center, Sapporo Higashi Tokushukai Hospital
| | | | - Shuzo Kobayashi
- Department of Kidney Disease and Transplant Center, Shonan Kamakura General Hospital
| | - Tatsuo Tsukamoto
- Nephrology and Dialysis, The Tazuke Kofukai Medical Research Institute, Kitano Hospital
| | - Tetsuya Haruna
- Cardiovascular Center, The Tazuke Kofukai Medical Research Institute, Kitano Hospital
| | | | - Ken Sakai
- Department of Nephrology, Toho University Faculty of Medicine
| | - Hideaki Obara
- Department of Surgery, Keio University School of Medicine
| | | | - Kenichi Saigo
- Division of Surgery, NHO Chiba-East National Hospital
| | | | - Kohmei Kubo
- Department of Hematology, Aomori Prefectural Central Hospital
| | | | - Teiji Oda
- Department of Thoracic and Cardiovascular Surgery, Shimane University Faculty of Medicine
| | - Eiji Kaneko
- Institute of Education, Tokyo Medical and Dental University
| | | | - Tetsuya Ioji
- Translational Research Informatics Center, Foundation for Biomedical Research and Innovation
| | - Hideaki Kaneda
- Translational Research Informatics Center, Foundation for Biomedical Research and Innovation
| | - Masanori Fukushima
- Translational Research Informatics Center, Foundation for Biomedical Research and Innovation
| | | |
Collapse
|
9
|
Fang Y, Wei Z, Chen B, Pan T, Gu S, Liu P, Guo D, Xu X, Jiang J, Yang J, Shi Z, Zhu T, Shi Y, Liu Y, Dong Z, Fu W. A Five-Year Study of the Efficacy of Purified CD34+ Cell Therapy for Angiitis-Induced No-Option Critical Limb Ischemia. Stem Cells Transl Med 2018; 7:583-590. [PMID: 29709112 PMCID: PMC6090512 DOI: 10.1002/sctm.17-0252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/23/2018] [Indexed: 11/06/2022] Open
Abstract
Angiitis‐induced critical limb ischemia (AICLI) patients constitute a remarkable proportion of no‐option critical limb ischemia (CLI) patients. Stem cell therapy has become an innovative and promising option for no‐option CLI patients. As one of these promising stem cell therapies, purified CD34+ cell transplantation (PuCeT) has shown favorable short‐term results. However, the long‐term efficacy of PuCeT has yet to be reported. This study evaluates the long‐term efficacy of PuCeT in AICLI patients. Twenty‐seven AICLI patients were enrolled from May 2009 to December 2011. Granulocyte colony‐stimulating factor (G‐CSF) and enoxaparin sodium were administered for 5 days. On day 5, CD34+ cell isolation was performed, and cells were transplanted by intramuscular injection. The primary endpoint, major‐amputation‐free survival rate (MAFS), as well as secondary endpoints, such as peak pain‐free walking time (PPFWT) and the Wong‐Baker FACES pain rating scale score (WFPRSS), were routinely evaluated during the 5‐year follow‐up period. The endpoints were as follows: the MAFS was 88.89%; PPFWT increased from 3 ± 3 to 17 ± 6 minutes; WFPRSS decreased from 7 ± 2 to 0.3 ± 1.7; the ulcer healing rate was 85.71%; the recurrence rate was 11.11%; and SF‐36v2 scores were significantly improved at 5 years after PuCeT. The rate of labor recovery 5 years after PuCeT was 65.38%, and no severe adverse effect was observed during the treatment. PuCeT demonstrated long‐term efficacy and durability as a treatment of AICLI not only in achieving limb salvage but also in recovering the labor competence and improving the quality of life of patients. Stem Cells Translational Medicine2018;7:583–590
Collapse
Affiliation(s)
- Yuan Fang
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Zheng Wei
- Department of Hematology, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Bin Chen
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Tianyue Pan
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Shiyang Gu
- Department of Hematology, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Daqiao Guo
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Xin Xu
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Junhao Jiang
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Jue Yang
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Zhenyu Shi
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Ting Zhu
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Yun Shi
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Yifan Liu
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Zhihui Dong
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
10
|
Trignano E, Fallico N, Zingone G, Rubino C, Di Pompeo FS, Campus GV. Combined Treatment of Diabetic Foot Ulcer with Tarsal Tunnel Release and Perilesional Injections of Peripheral Blood Mononuclear Cells. J Am Podiatr Med Assoc 2017; 107:171-174. [PMID: 28394682 DOI: 10.7547/15-098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Emilio Trignano
- Department of Surgery and Microsurgery, University of Sassari, Sassari, Italy
| | - Nefer Fallico
- Department of Plastic Surgery, Sapienza University of Rome, Rome, Italy
| | - Gino Zingone
- Department of Surgery and Microsurgery, University of Sassari, Sassari, Italy
| | - Corrado Rubino
- Department of Plastic Surgery, University of Salerno, Fisciano, Italy
| | | | | |
Collapse
|
11
|
Zhou J, Rogers JH, Lee SH, Sun D, Yao H, Mao JJ, Kong KY. Oral Mucosa Harbors a High Frequency of Endothelial Cells: A Novel Postnatal Cell Source for Angiogenic Regeneration. Stem Cells Dev 2016; 26:91-101. [PMID: 27832737 DOI: 10.1089/scd.2016.0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Endothelial progenitor cells/endothelial cells (EPCs/ECs) have great potential to treat pathological conditions such as cardiac infarction, muscle ischemia, and bone fractures, but isolation of EPC/ECs from existing cell sources is challenging due to their low EC frequency. We have isolated endothelial progenitor (EP)-like cells from rat oral mucosa and characterized their yield, immunophenotype, growth, and in vivo angiogenic potential. The frequency of EP-like cells derived from oral mucosa is thousands of folds higher than EPCs derived from donor-match bone marrow samples. EP-like cells from oral mucosa were positive for EC markers CD31, VE-Cadherin, and VEGFR2. Oral mucosa-derived EP-like cells displayed robust uptake of acetylated low-density lipoprotein and formed stable capillary networks in Matrigel. Subcutaneously implanted oral mucosa-derived EP-like cells anastomosed with host blood vessels, implicating their ability to elicit angiogenesis. Similar to endothelial colony-forming cells, EP-like cells from oral mucosa have a significantly higher proliferative rate than human umbilical vein endothelial cells. These findings identify a putative EPC source that is easily accessible in the oral cavity, potentially from discarded tissue specimens, and yet with robust yield and potency for angiogenesis in tissue and organ regeneration.
Collapse
Affiliation(s)
- Jian Zhou
- 1 Center for Craniofacial Regeneration, Columbia University Medical Center , New York, New York.,2 Department of General Dentistry, Capital Medical University School of Stomatology , Beijing, China
| | - Jason H Rogers
- 3 Department of Internal Medicine and the Cancer Research and Treatment Center, University of New Mexico Health Science Center , Albuquerque, New Mexico
| | - Scott H Lee
- 4 Pratt School of Engineering, Duke University , Durham, North Carolina
| | - DongMing Sun
- 5 W. M. Keck Center for Collaborative Neuroscience, Rutgers University , New Brunswick, New Jersey
| | - Hai Yao
- 6 Clemson-MUSC Bioengineering Program , Department of Craniofacial Biology, Charleston, South Carolina
| | - Jeremy J Mao
- 1 Center for Craniofacial Regeneration, Columbia University Medical Center , New York, New York
| | - Kimi Y Kong
- 1 Center for Craniofacial Regeneration, Columbia University Medical Center , New York, New York.,7 Hematology/Oncology Division, Department of Medicine, University of Florida , Gainesville, Florida
| |
Collapse
|
12
|
Kobayashi S. [CKD and Progression of PAD.]. NIHON NAIKA GAKKAI ZASSHI. THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE 2016; 105:842-849. [PMID: 29182836 DOI: 10.2169/naika.105.842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
|
13
|
Spaltro G, Straino S, Gambini E, Bassetti B, Persico L, Zoli S, Zanobini M, Capogrossi MC, Spirito R, Quarti C, Pompilio G. Characterization of the Pall Celeris system as a point-of-care device for therapeutic angiogenesis. Cytotherapy 2015; 17:1302-13. [PMID: 26038175 DOI: 10.1016/j.jcyt.2015.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 03/23/2015] [Accepted: 04/14/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND AIMS The Pall Celeris system is a filtration-based point-of-care device designed to obtain a high concentrate of peripheral blood total nucleated cells (PB-TNCs). We have characterized the Pall Celeris-derived TNCs for their in vitro and in vivo angiogenic potency. METHODS PB-TNCs isolated from healthy donors were characterized through the use of flow cytometry and functional assays, aiming to assess migratory capacity, ability to form capillary-like structures, endothelial trans-differentiation and paracrine factor secretion. In a hind limb ischemia mouse model, we evaluated perfusion immediately and 7 days after surgery, along with capillary, arteriole and regenerative fiber density and local bio-distribution. RESULTS Human PB-TNCs isolated by use of the Pall Celeris filtration system were shown to secrete a panel of angiogenic factors and migrate in response to vascular endothelial growth factor and stromal-derived factor-1 stimuli. Moreover, after injection in a mouse model of hind limb ischemia, PB-TNCs induced neovascularization by increasing capillary, arteriole and regenerative fiber numbers, with human cells detected in murine tissue up to 7 days after ischemia. CONCLUSIONS The Pall Celeris system may represent a novel, effective and reliable point-of-care device to obtain a PB-derived cell product with adequate potency for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Gabriella Spaltro
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| | - Stefania Straino
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, IRCCS, Rome, Italy; Explora Biotech srl, Rome, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Luca Persico
- DIEC-Dipartimento di Economia, Università degli Studi di Genova, Genoa, Italy
| | - Stefano Zoli
- Department of Cardiovascular Surgery, University of Milan, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Marco Zanobini
- Department of Cardiovascular Surgery, University of Milan, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Maurizio C Capogrossi
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, IRCCS, Rome, Italy
| | - Rita Spirito
- Department of Cardiovascular Surgery, University of Milan, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
14
|
De Angelis B, Gentile P, Orlandi F, Bocchini I, Di Pasquali C, Agovino A, Gizzi C, Patrizi F, Scioli MG, Orlandi A, Cervelli V. Limb rescue: a new autologous-peripheral blood mononuclear cells technology in critical limb ischemia and chronic ulcers. Tissue Eng Part C Methods 2015; 21:423-35. [PMID: 25341088 DOI: 10.1089/ten.tec.2014.0245] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Therapeutic angiogenesis by autologous-peripheral blood mononuclear cells (A-PBMNC) implantation has been shown to be a safe and effective treatment for critical limb ischemia (CLI). We herein report our investigation of the long-term efficacy of implantation of A-PBMNC produced by selective filtration to treat patients with CLI, for which surgical bypass and/or percutaneous transluminal angioplasty are not possible. MATERIALS AND METHODS This is a prospective, and not a randomized, study based on a treated group who did not respond to conventional therapy (n=43) when implanted with A-PBMNC cells versus a historically matched control group. Patients of both groups were suffering from CLI Fontaine scale IV with chronic ulcers and various accompanying conditions (diabetes, heart disease, kidney failure, etc.). Treated patients were implanted with 12 mL of A-PBMNC, 0.2-0.3 mL for each bolus, collected by selective filtration from 120 mL of peripheral blood in the ischemic area of the limbs. Patients were not mobilized by granulocyte colony-stimulating factor, and the A-PBMNC treatment was repeated for a maximum of three times. RESULTS The A-PBMNC-treated group showed a statistically significant improvement of limb rescue of 95.3% versus 52.2% of the control group (p<0.001), and the result had been maintained for 2 years. The A-PBMNC group also showed reduction in pain at rest, increased maximum walking distance, and healing of the wound, which led to an overall improvement in the quality of life. Post-treatment radiological studies showed an improvement of vascularization with the formation of new collateral and by histological findings. Within 2 years of follow-up, none of the patients whom we treated showed any major or systemic adverse effects. CONCLUSION The local injection of A-PBMNC showed striking early and long-term effects together with a favorable safety profile, significantly decreasing the risk of amputation. Our results are comparable with published data obtained by injection of bone marrow mononuclear cells, but with a lot less invasive approach. Moreover the intraoperative selective filtration system we used is fast, safe, not operator dependent, and easy to use in a sterile operating theatre. This system aims to produce fresh A-PBMNC as a valuable treatment option, particularly for those difficult patients who cannot undergo revascularization.
Collapse
Affiliation(s)
- Barbara De Angelis
- 1 Department of Plastic and Reconstructive Surgery, University of Rome Tor Vergata , Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Preclinical and clinical evidence for stem cell therapies as treatment for diabetic wounds. Drug Discov Today 2015; 20:703-17. [PMID: 25603421 DOI: 10.1016/j.drudis.2015.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 12/10/2014] [Accepted: 01/12/2015] [Indexed: 01/07/2023]
Abstract
Diabetic wounds remain a global unsolved problem and the cost of diabetes-related amputations and diabetic wound treatment is approximately US$3 billion and US$9 billion per year, respectively. Diabetic foot ulcers (DFUs) occur in 15% of all patients with diabetes and precede 84% of all diabetes-related lower leg amputations. Currently, there is no satisfying treatment for these hard-to-heal-wounds. However, as we discuss here, experimental preclinical evidence for the successful use of adult stem cell therapies for diabetic wounds gives new hope for the development of effective treatments for use in the clinic.
Collapse
|
16
|
Teraa M, Sprengers RW, Schutgens REG, Slaper-Cortenbach ICM, van der Graaf Y, Algra A, van der Tweel I, Doevendans PA, Mali WPTM, Moll FL, Verhaar MC. Effect of repetitive intra-arterial infusion of bone marrow mononuclear cells in patients with no-option limb ischemia: the randomized, double-blind, placebo-controlled Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation (JUVENTAS) trial. Circulation 2015; 131:851-60. [PMID: 25567765 DOI: 10.1161/circulationaha.114.012913] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Patients with severe limb ischemia may not be eligible for conventional therapeutic interventions. Pioneering clinical trials suggest that bone marrow-derived cell therapy enhances neovascularization, improves tissue perfusion, and prevents amputation. The objective of this trial was to determine whether repetitive intra-arterial infusion of bone marrow mononuclear cells (BMMNCs) in patients with severe, nonrevascularizable limb ischemia can prevent major amputation. METHODS AND RESULTS The Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation (JUVENTAS) trial is a randomized, double-blind, placebo-controlled clinical trial in 160 patients with severe, nonrevascularizable limb ischemia. Patients were randomly assigned to repetitive (3 times; 3-week interval) intra-arterial infusion of BMMNC or placebo. No significant differences were observed for the primary outcome, ie, major amputation at 6 months, with major amputation rates of 19% in the BMMNC versus 13% in the placebo group (relative risk, 1.46; 95% confidence interval, 0.62-3.42). The safety outcome (all-cause mortality, occurrence of malignancy, or hospitalization due to infection) was not significantly different between the groups (relative risk, 1.46; 95% confidence interval, 0.63-3.38), neither was all-cause mortality at 6 months with 5% versus 6% (relative risk, 0.78; 95% confidence interval, 0.22-2.80). Secondary outcomes quality of life, rest pain, ankle-brachial index, and transcutaneous oxygen pressure improved during follow-up, but there were no significant differences between the groups. CONCLUSIONS Repetitive intra-arterial infusion of autologous BMMNCs into the common femoral artery did not reduce major amputation rates in patients with severe, nonrevascularizable limb ischemia in comparison with placebo. The general improvement in secondary outcomes during follow-up in both the BMMNC and the placebo group, as well, underlines the essential role for placebo-controlled design of future trials. CLINICAL TRIAL REGISTRATION URL http://www.clinicaltrials.gov. Unique identifier: NCT00371371.
Collapse
Affiliation(s)
- Martin Teraa
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Ralf W Sprengers
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Roger E G Schutgens
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Ineke C M Slaper-Cortenbach
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Yolanda van der Graaf
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Ale Algra
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Ingeborg van der Tweel
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Pieter A Doevendans
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Willem P Th M Mali
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Frans L Moll
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands
| | - Marianne C Verhaar
- From Department of Nephrology & Hypertension (M.T., M.C.V.), Department of Vascular Surgery (M.T., F.L.M.), Department of Radiology (R.W.S., W.P.Th.M.M.), Van Creveldkliniek/Department of Hematology (R.E.G.S.), Cell Therapy Facility/ Department of Clinical Pharmacy (I.C.M.S.-C.), Julius Center for Health Sciences and Primary Care (Y.v.d.G., A.A., I.v.d.T.), Brain Center Rudolf Magnus, Department of Neurology and Neurosurgery (A.A.), and Department of Cardiology (P.A.D.), University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
17
|
Lee KB, Kim DI. Clinical application of stem cells for therapeutic angiogenesis in patients with peripheral arterial disease. Int J Stem Cells 2014; 2:11-7. [PMID: 24855515 DOI: 10.15283/ijsc.2009.2.1.11] [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] [Accepted: 04/08/2009] [Indexed: 12/16/2022] Open
Abstract
Peripheral arterial disease (PAD) may ultimately cause to the loss of the affected limb due to gangrene or infection. Some patients with PAD may have severe coexisting diseases and diffuse involvement of their distal arteries, and so they are poor candidates for revascularization procedures. Angiogenesis has recently been suggested to be a new emerging treatment strategy for patients with PAD. Angiogenesis is defined as the sprouting of new capillaries from pre-existing vascular structures; this process plays a major role in the development of collateral vessels in an ischemic limb. Yet, the exact mechanism of angiogenesis is currently poorly understood. It has been established that angiogenesis is initiated by hypoxia and it requires various pro-angiogenic factors such as vascular endothelial growth factor. Therapeutic angiogenesis is aimed at enhancing natural angiogenesis by the administration of the cells or genes that can trigger angiogenesis and this can lead to pain relief and wound healing by the development of collateral vessels. Most of the recent clinical trials have reported that stem cell therapy for promoting angiogenesis in patients with PAD improves the ischemic symptoms and enhances wound healing. However, there are several limitations to approve a standard treatment for PAD such as small sample size in several prevous studies, their diverse inclusion criteria and the lack of standard assessment methods for the safety and outcome. Therefore, multicenter, large-scale and randomized controlled studies are needed to prove the safety and efficacy of the clinically applying stem cells for therapeutic angiogenesis in patients with PAD.
Collapse
Affiliation(s)
- Kyung-Bok Lee
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Cell transplantation therapy for a rat model of secondary lymphedema. J Surg Res 2014; 189:184-91. [DOI: 10.1016/j.jss.2013.11.1116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/24/2013] [Accepted: 11/27/2013] [Indexed: 12/21/2022]
|
19
|
Pelosi E, Castelli G, Testa U. Endothelial progenitors. Blood Cells Mol Dis 2014; 52:186-94. [DOI: 10.1016/j.bcmd.2013.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 11/13/2013] [Accepted: 11/13/2013] [Indexed: 12/31/2022]
|
20
|
Shalaby SY, Blume P, Sumpio BE. New modalities in the chronic ischemic diabetic foot management. Clin Podiatr Med Surg 2014; 31:27-42. [PMID: 24296016 DOI: 10.1016/j.cpm.2013.09.009] [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] [Indexed: 10/26/2022]
Abstract
The diabetic population is increasing worldwide at a staggering rate. Diabetic foot ulcers are a major contributor to nontraumatic lower limb amputations and peripheral arterial disease is one of main contributing pathophysiologic causes of diabetic ulcers. The dire need to reduce complication and wound healing recovery period of the chronic ischemic diabetic foot (CIDF) is indispensable to limb salvage and improvement of quality of life of patients with CIDF. This article discusses newer modalities that have been proposed to improve CIDF efficiently, safely, and effectively either alone or as adjuvants to conventional therapy.
Collapse
Affiliation(s)
- Sherif Y Shalaby
- Department of Vascular Surgery, Yale University School of Medicine, 310 Cedar Street, New Haven, CT 06510, USA
| | | | | |
Collapse
|
21
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
22
|
Cruciani M, Lipsky BA, Mengoli C, de Lalla F. Granulocyte-colony stimulating factors as adjunctive therapy for diabetic foot infections. Cochrane Database Syst Rev 2013:CD006810. [PMID: 23955465 DOI: 10.1002/14651858.cd006810.pub3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Granulocyte-colony stimulating factor (G-CSF) increases the release of neutrophil endothelial progenitor cells from the bone marrow and improves neutrophil functions, which are often impaired in people with diabetes. OBJECTIVES To examine the effects of adjunctive G-CSF compared with placebo or no growth factor added to usual care on rates of infection, cure and wound healing in people with diabetes who have a foot infection. SEARCH METHODS In March 2013, for this second update, we searched the Cochrane Wounds Group Specialised Register (searched 14 March 2013); the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2013, Issue 2); Ovid MEDLINE (1948 to March Week 1 2013); Ovid EMBASE (1974 to 2013 March 13); Ovid MEDLINE (In-Process march 13,2013); and EBSCO CINAHL (1982 to 28 February 2013). SELECTION CRITERIA Randomised controlled trials (RCTs) that evaluated the effect of adding G-CSF to usual care in people with a diabetic foot infection. DATA COLLECTION AND ANALYSIS Three review authors independently assessed trial eligibility, methodological quality and extracted data. We reported risk ratio (RR) or, for continuous outcomes, mean differences (MD), with 95% confidence intervals (CI). In the case of low or no heterogeneity we pooled studies using a fixed-effect model. MAIN RESULTS We identified and included five eligible trials with a total of 167 patients. The investigators administered various G-CSF preparations, at different doses and for different durations of time. Adding G-CSF did not significantly affect the likelihood of resolution of infection or wound healing, but it was associated with a significantly reduced likelihood of lower extremity surgical interventions (RR 0.38; 95 % CI 0.21 to 0.70), including amputation (RR 0.41; 95 % CI 0.18 to 0.95). Moreover, providing G-CSF reduced the duration of hospital stay (MD -1.40 days; 95% CI -2.27 to -0.53 days), but did not significantly affect the duration of systemic antibiotic therapy (MD -0.27 days; 95% CI -1.30 to 0.77 days). AUTHORS' CONCLUSIONS The available evidence is limited, but suggests that adjunctive G-CSF treatment in people with a diabetic foot infection, including infected ulcers, does not appear to increase the likelihood of resolution of infection or healing of the foot ulcer. However, it does appear to reduce the need for surgical interventions, especially amputations, and the duration of hospitalisation. Clinicians might consider adding G-CSF to the usual treatment of diabetic foot infections, especially in patients with a limb-threatening infection, but it is not clear which patients might benefit.
Collapse
Affiliation(s)
- Mario Cruciani
- Center of Community Medicine and Infectious Diseases Service, ULSS 20 Verona, Via Germania, 20, Verona, Italy, 37135
| | | | | | | |
Collapse
|
23
|
Kawazoe T, Kim HH. Tissue augmentation by white blood cell-containing platelet-rich plasma. Cell Transplant 2012; 21:601-7. [PMID: 22793069 DOI: 10.3727/096368911x605538] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Platelet-rich plasma (PRP) is a matrix of fibrin and platelets that releases cytokines that are important in wound healing. PRP is produced from the patient's blood and therefore has less risk of allergic reaction and infection. We have obtained PRP with an enhanced white blood cell component (W-PRP) by optimizing the centrifugal separation of PRP from plasma. Here we show that injection of W-PRP into the auricle of nude mice gave greater tissue augmentation compared to PRP. Further augmentation occurred when bFGF was added to W-PRP, and there was a significant increase in the number of α-smooth muscle actin-positive cells in mice treated with W-PRP+bFGF. Our results suggest that W-PRP may have value in cosmetic surgery aimed at rejuvenation of wrinkled and sagging skin. W-PRP injection constitutes a new concept in cell transplantation, in which cells required for tissue regeneration are induced by cytokines released from the transplanted cells.
Collapse
Affiliation(s)
- Takeshi Kawazoe
- Department of Plastic Reconstructive and Aesthetic Surgery, Kijunkai, Yoshikawa Hospital, Sakyo-ku, Kyoto, Japan.
| | | |
Collapse
|
24
|
Botti C, Maione C, Coppola A, Sica V, Cobellis G. Autologous bone marrow cell therapy for peripheral arterial disease. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2012; 5:5-14. [PMID: 24198534 PMCID: PMC3781761 DOI: 10.2147/sccaa.s28121] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inadequate blood supply to tissues caused by obstruction of arterioles and/or capillaries results in ischemic injuries – these injuries can range from mild (eg, leg ischemia) to severe conditions (eg, myocardial infarction, stroke). Surgical and/or endovascular procedures provide cutting-edge treatment for patients with vascular disorders; however, a high percentage of patients are currently not treatable, owing to high operative risk or unfavorable vascular involvement. Therapeutic angiogenesis has recently emerged as a promising new therapy, promoting the formation of new blood vessels by the introduction of bone marrow–derived stem and progenitor cells. These cells participate in the development of new blood vessels, the enlargement of existing blood vessels, and sprouting new capillaries from existing blood vessels, providing evidence of the therapeutic utility of these cells in ischemic tissues. In this review, the authors describe peripheral arterial disease, an ischemic condition affecting the lower extremities, summarizing different aspects of vascular regeneration and discussing which and how stem cells restore the blood flow. The authors also present an overview of encouraging results from early-phase clinical trials using stem cells to treat peripheral arterial disease. The authors believe that additional research initiatives should be undertaken to better identify the nature of stem cells and that an intensive cooperation between laboratory and clinical investigators is needed to optimize the design of cell therapy trials and to maximize their scientific rigor. Only this will allow the results of these investigations to develop best clinical practices. Additionally, although a number of stem cell therapies exist, many treatments are performed outside international and national regulations and many clinical trials have been not registered on databases such as ClinicalTrials.gov or EudraCT. Therefore, more rigorous clinical trials are required to confirm the first hopeful results and to address the challenging issues.
Collapse
Affiliation(s)
- C Botti
- Department of General Pathology, Second University of Naples, Naples, Italy
| | | | | | | | | |
Collapse
|
25
|
Klepanec A, Mistrik M, Altaner C, Valachovicova M, Olejarova I, Slysko R, Balazs T, Urlandova T, Hladikova D, Liska B, Tomka J, Vulev I, Madaric J. No Difference in Intra-Arterial and Intramuscular Delivery of Autologous Bone Marrow Cells in Patients with Advanced Critical Limb Ischemia. Cell Transplant 2012; 21:1909-18. [DOI: 10.3727/096368912x636948] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stem cell therapy has been proposed to be an alternative therapy in patients with critical limb ischemia (CLI), not eligible for endovascular or surgical revascularization. We compared the therapeutic effects of intramuscular (IM) and intra-arterial (IA) delivery of bone marrow cells (BMCs) and investigated the factors associated with therapeutic benefits. Forty-one patients (mean age, 66 ± 10 years; 35 males) with advanced CLI (Rutherford category, 5 and 6) not eligible for revascularization were randomized to treatment with 40 ml BMCs using local IM ( n = 21) or selective IA infusion ( n = 20). Primary endpoints were limb salvage and wound healing. Secondary endpoints were changes in transcutaneous oxygen pressure (tcpO2), quality-of-life questionnaire (EQ5D), ankle–brachial index (ABI), and pain scale (0–10). Patients with limb salvage and wound healing were considered to be responders to BMC therapy. At 6-month follow-up, overall limb salvage was 73% (27/37) and 10 subjects underwent major amputation. Four patients died unrelated to stem cell therapy. There was significant improvement in tcpO2 (15 ± 10 to 29 ± 13 mmHg, p < 0.001), pain scale (4.4 ± 2.6 to 0.9 ± 1.4, p < 0.001), and EQ5D (51 ± 15 to 70 ± 13, p < 0.001) and a significant decrease in the Rutherford category of CLI (5.0 ± 0.2 to 4.3 ± 1.6, p < 0.01). There were no differences among functional parameters in patients undergoing IM versus IA delivery. Responders ( n = 27) were characterized by higher CD34+ cell counts in the bone marrow concentrate (CD34+ 29 ± 15×106 vs. 17 ± 12×106, p < 0.05) despite a similar number of total nucleated cells (4.3 ± 1.4×109 vs. 4.1 ± 1.2×109, p = 0.66) and by a lower level of C-reactive protein (18 ± 28 vs. 100 ± 96 mg/L, p < 0.05) as well as serum leukocytes (8.3 ± 2.1×109/L vs. 12.3 ± 4.5×109/L, p < 0.05) as compared with nonresponders (10 patients). Both IM and IA delivery of autologous stem cells are effective therapeutic strategies in patients with CLI. A higher concentration of CD34+ cells and a lower degree of inflammation are associated with better clinical therapeutic responses.
Collapse
Affiliation(s)
- Andrej Klepanec
- Slovak Medical University, Bratislava, Slovakia
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Martin Mistrik
- Clinic of Haematology and Transfusiology, Faculty Hospital, Bratislava, Slovakia
| | - Cestmir Altaner
- Institute of Experimental Oncology, Slovak Academy of Science, Bratislava, Slovakia
| | | | | | - Roman Slysko
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Tibor Balazs
- National Cardiovascular Institute, Bratislava, Slovakia
| | | | | | | | - Jan Tomka
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Ivan Vulev
- Slovak Medical University, Bratislava, Slovakia
- National Cardiovascular Institute, Bratislava, Slovakia
| | - Juraj Madaric
- Slovak Medical University, Bratislava, Slovakia
- National Cardiovascular Institute, Bratislava, Slovakia
| |
Collapse
|
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
|
Barclay GR, Tura O, Samuel K, Hadoke PW, Mills NL, Newby DE, Turner ML. Systematic assessment in an animal model of the angiogenic potential of different human cell sources for therapeutic revascularization. Stem Cell Res Ther 2012; 3:23. [PMID: 22759659 PMCID: PMC3580461 DOI: 10.1186/scrt114] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 07/03/2012] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Endothelial progenitor cells (EPC) capable of initiating or augmenting vascular growth were recently identified within the small population of CD34-expressing cells that circulate in human peripheral blood and which are considered hematopoietic progenitor cells (HPC). Soon thereafter human HPC began to be used in clinical trials as putative sources of EPC for therapeutic vascular regeneration, especially in myocardial and critical limb ischemias. However, unlike HPC where hematopoietic efficacy is related quantitatively to CD34+ cell numbers implanted, there has been no consensus on how to measure EPC or how to assess cellular graft potency for vascular regeneration. We employed an animal model of spontaneous neovascularization to simultaneously determine whether human cells incorporate into new vessels and to quantify the effect of different putative angiogenic cells on vascularization in terms of number of vessels generated. We systematically compared competence for therapeutic angiogenesis in different sources of human cells with putative angiogenic potential, to begin to provide some rationale for optimising cell procurement for this therapy. METHODS Human cells employed were mononuclear cells from normal peripheral blood and HPC-rich cell sources (umbilical cord blood, mobilized peripheral blood, bone marrow), CD34+ enriched or depleted subsets of these, and outgrowth cell populations from these. An established sponge implant angiogenesis model was adapted to determine the effects of different human cells on vascularization of implants in immunodeficient mice. Angiogenesis was quantified by vessel density and species of origin by immunohistochemistry. RESULTS CD34+ cells from mobilized peripheral blood or umbilical cord blood HPC were the only cells to promote new vessel growth, but did not incorporate into vessels. Only endothelial outgrowth cells (EOC) incorporated into vessels, but these did not promote vessel growth. CONCLUSIONS These studies indicate that, since EPC are very rare, any benefit seen in clinical trials of HPC in therapeutic vascular regeneration is predominantly mediated by indirect proangiogenic effects rather than through direct incorporation of any rare EPC contained within these sources. It should be possible to produce autologous EOC for therapeutic use, and evaluate the effect of EPC distinct from, or in synergy with, the proangiogenic effects of HPC therapies.
Collapse
|
28
|
Jiang XY, Lu DB, Chen B. Progress in stem cell therapy for the diabetic foot. Diabetes Res Clin Pract 2012; 97:43-50. [PMID: 22221581 DOI: 10.1016/j.diabres.2011.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/05/2011] [Accepted: 12/08/2011] [Indexed: 12/20/2022]
Abstract
The diabetic foot is a common and severe complication of diabetes comprising a group of lesions including vasculopathy, neuropathy, tissue damage and infection. Vasculopathy due to ischemia is a major contributor to the pathogenesis, natural history and outcome of the diabetic foot. Despite conventional revascularization interventions including angioplasty, stenting, atherectomy and bypass grafts to vessels, a high incidence of amputation persists. The need to develop alternative therapeutic options is compelling; stem cell therapy aims to increase revascularization and alleviate limb ischemia or improve wound healing by stimulating new blood vessel formation, and brings new hope for the treatment of the diabetic foot.
Collapse
Affiliation(s)
- Xiao-Yan Jiang
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | |
Collapse
|
29
|
Surgical Therapy of End-Stage Heart Failure: Understanding Cell-Mediated Mechanisms Interacting with Myocardial Damage. Int J Artif Organs 2011; 34:529-45. [DOI: 10.5301/ijao.5000004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2011] [Indexed: 01/19/2023]
Abstract
Worldwide, cardiovascular disease results in an estimated 14.3 million deaths per year, giving rise to an increased demand for alternative and advanced treatment. Current approaches include medical management, cardiac transplantation, device therapy, and, most recently, stem cell therapy. Research into cell-based therapies has shown this option to be a promising alternative to the conventional methods. In contrast to early trials, modern approaches now attempt to isolate specific stem cells, as well as increase their numbers by means of amplifying in a culture environment. The method of delivery has also been improved to minimize the risk of micro-infarcts and embolization, which were often observed after the use of coronary catheterization. The latest approach entails direct, surgical, transepicardial injection of the stem cell mixture, as well as the use of tissue-engineered meshes consisting of embedded progenitor cells.
Collapse
|
30
|
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
|
31
|
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
|
32
|
Abstract
INTRODUCTION AND BACKGROUND Peripheral vascular disease is the leading cause of limb ischemia (LI). LI is manifested by claudication, ischemic rest pain, ulcers or gangrene. It is the result of peripheral arterial disease due to atherosclerosis. Over the last decade, several centers around the world have initiated clinical trials utilizing stem cells as a treatment for this disease. SOURCES OF DATA Published medical literature, clinical trials announced in clinical trials.gov and TCA cellular therapy experience. AREAS OF AGREEMENT There is general agreement that stem cells are useful for LI. AREAS OF CONTROVERSY These arise from the type of cells, dose, route of administration and methods to evaluate efficacy. GROWING POINTS Growing evidence suggests that bone marrow derived-mesenchymal stem cells are as good as or superior to mononuclear cells, and a combination of both cell types may be even better. AREAS TIMELY FOR DEVELOPING RESEARCH Based on current trials and publications, several promising biological products could become part of the therapeutic arsenal for LI. This may include combinations of more than one type of adult/induced pluripotent stem cells/embryonic stem cells, use of stem cells with growth factors or extracellular matrix molecules.
Collapse
|
33
|
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]
|
34
|
Rationale and design of the JUVENTAS trial for repeated intra-arterial infusion of autologous bone marrow-derived mononuclear cells in patients with critical limb ischemia. J Vasc Surg 2010; 51:1564-8. [PMID: 20488328 DOI: 10.1016/j.jvs.2010.02.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 02/04/2010] [Accepted: 02/06/2010] [Indexed: 11/24/2022]
Abstract
Critical limb ischemia (CLI) continues to form a substantial burden on Western healthcare. Many patients still face amputation as a last treatment option. Autologous bone marrow (BM)-derived cell administration has emerged as a potential new treatment, but proof for sustainable clinical effects of BM-derived cell therapy in CLI is still lacking. The JUVENTAS (reJUVenating ENdothelial progenitor cells via Transcutaneous intra-Arterial Supplementation) trial is the first randomized, placebo-controlled, double-blinded clinical trial on repeated intra-arterial BM mononuclear cell (MNC) infusion in 110 to 160 CLI patients, designed to provide definite proof for the efficacy of stem cell therapy. Primary outcome is the incidence of major amputation at 6 months. Inclusion of patients is well underway. If BM-MNC cells therapy is beneficial, it could become a novel treatment to prevent amputation in patients with CLI.
Collapse
|
35
|
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]
|
36
|
Abstract
The therapeutic potential of 'adult' or at least non-embryonic stem cells and their progeny has developed gradually over the past half century as a consequence of the wealth of knowledge derived from stem cell research. Translational research coupled with clinical trials and derived from basic research has led the way to the clinic. This commenced with the use of haematopoietic stem cell transplantation (HSCT), to treat haematological malignancies, to be followed by the most recent clinical trials to treat a variety of coronary and peripheral artery diseases. Stem cells and their progeny isolated from bone marrow or blood appear to exert an ameliorating effect in certain vascular disorders. Although promising, some of these treatments remain controversial and further research and, where indicated, appropriately powered trials are required to confirm the safety and determine the efficacy of these novel therapies.
Collapse
Affiliation(s)
- E Martin-Rendon
- Stem Cell Research Laboratory, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, UK.
| | | | | |
Collapse
|
37
|
Autologous stem cell therapy for peripheral arterial disease meta-analysis and systematic review of the literature. Atherosclerosis 2009; 209:10-7. [PMID: 19740466 DOI: 10.1016/j.atherosclerosis.2009.08.033] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 08/05/2009] [Accepted: 08/17/2009] [Indexed: 11/21/2022]
Abstract
BACKGROUND Peripheral arterial disease (PAD) is a common cause of disability and mortality. Up to one third of patients are not susceptible to traditional revascularization and may benefit from stem cell therapies. OBJECTIVE In this meta-analysis, we sought to determine whether autologous cell therapy is effective in the treatment of PAD. METHODS We searched the English literature in Medline, Excerpta Medica and the Cochrane database for trials of autologous cell therapy in patients with PAD published before 31 January 2009. We included controlled and non-controlled, randomized and non-randomized trials using autologous bone marrow or granulocyte colony stimulating factor (G-CSF) mobilized peripheral blood cells to treat PAD. We also collected data from trials of G-CSF monotherapy, as a control treatment. RESULTS In a meta-analysis of 37 trials, autologous cell therapy was effective in improving surrogate indexes of ischemia, subjective symptoms and hard endpoints (ulcer healing and amputation). On the contrary, G-CSF monotherapy was not associated with significant improvement in the same endpoints. Patients with thromboangiitis obliterans showed some larger benefits than patients with atherosclerotic PAD. The intramuscular route of administration and the use of bone marrow cells seemed somehow more effective than intrarterial administration and the use of mobilized peripheral blood cells. The procedures were well tolerated and generally safe. CONCLUSION This meta-analysis indicates that intramuscular autologous bone marrow cell therapy is a feasible, relatively safe and potentially effective therapeutic strategy for PAD patients, who are not candidate for traditional revascularization. Larger, placebo-controlled, randomized multicenter trials need to be planned and conducted to confirm these findings.
Collapse
|
38
|
Horie T, Onodera R, Akamastu M, Ichikawa Y, Hoshino J, Kaneko E, Iwashita C, Ishida A, Tsukamoto T, Teramukai S, Fukushima M, Kawamura A. Long-term clinical outcomes for patients with lower limb ischemia implanted with G-CSF-mobilized autologous peripheral blood mononuclear cells. Atherosclerosis 2009; 208:461-6. [PMID: 19720375 DOI: 10.1016/j.atherosclerosis.2009.07.050] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Revised: 07/08/2009] [Accepted: 07/23/2009] [Indexed: 11/30/2022]
Abstract
BACKGROUND Many studies have described the clinical effects of treating critical limb ischemia with granulocyte colony-stimulating factor-mobilized autologous peripheral blood mononuclear cells (M-PBMNC); however, there are no long-term data available on survival, limb salvage, or prognostic factors. METHODS To investigate the long-term clinical outcomes of M-PBMNC implantation, we reviewed data for 162 consecutive patients with limb ischemia who were treated with M-PBMNC implantation at 6 hospitals between 2001 and 2006. A subset of 123 patients with homogenous clinical profiles was selected for prognostic factor analysis. RESULTS Of the 162 patients, 50 died during the follow-up period. The median follow-up time for surviving patients was 26.4 months. The 2-year survival rate was 65% for the 140 patients with arteriosclerosis obliterans (ASO), and 100% for the 11, 4 and 7 patients with thromboangiitis obliterans (TAO), diabetic gangrene (DG) and connective tissue disease (CTD), respectively. The 1-year amputation-free rates for ASO, TAO, DG and CTD were 70%, 79%, 75% and 83%, respectively. Common serious adverse events included heart failure (15 cases), myocardial infarction (15 cases), serious infection (13 cases), stroke (10 cases), and malignant tumor (9 cases). Significant negative prognostic factors associated with overall survival were ischemic heart disease and collection of a small number of CD34-positive cells. Factors associated with time-to-amputation and amputation-free survival were a combination of Fontaine classification and lower limb gangrene, and history of dialysis. CONCLUSIONS Collection of a small number of CD34-positive cells and ischemic heart disease were associated with a reduction in overall survival.
Collapse
Affiliation(s)
- Takashi Horie
- Department of Surgery, Sapporo Hokuyu Hospital, Higashisapporo 6-6, Shiroishi-ku, Sapporo 003-0006, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cruciani M, Lipsky BA, Mengoli C, de Lalla F. Granulocyte-colony stimulating factors as adjunctive therapy for diabetic foot infections. Cochrane Database Syst Rev 2009:CD006810. [PMID: 19588405 DOI: 10.1002/14651858.cd006810.pub2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND G-CSF increases the release of neutrophil endothelial progenitor cells from the bone marrow, and improves neutrophil functions, which are often impaired in people with diabetes. OBJECTIVES To examine the effects of adjunctive G-CSF compared with placebo or no growth factor added to usual care on rates of infection, cure and wound healing in people with diabetes who have a foot infection. SEARCH STRATEGY We searched the Cochrane Wounds Group Specialised Register (Searched 16/3/09); the Cochrane Central Register of Controlled Trials (The Cochrane Library, issue 1 2009); Ovid MEDLINE (1950 to March Week 1 2009); Ovid EMBASE (1980 to 2009 Week 11); EBSCO CINAHL (1982 to March Week 2 2009); LookSmart's Find Articles (January 1990 to January 2008); conference proceedings and references lists in the included studies. SELECTION CRITERIA Randomised controlled trials (RCTs) that evaluated the effect of adding G-CSF to usual care in people with a diabetic foot infection. DATA COLLECTION AND ANALYSIS Three review authors independently assessed trial eligibility, methodological quality and extracted data. Relative risk (RR), or for continuous outcomes, mean differences (MD), with 95% confidence intervals (CI) were reported. In the case of low or no heterogeneity studies were pooled using a fixed-effect model. MAIN RESULTS We identified and included five eligible trials with a total of 167 patients. The investigators administered various G-CSF preparations, at different doses and for different durations of time. Adding G-CSF did not significantly affect the likelihood of resolution of infection or wound healing, but it was associated with a significantly reduced likelihood of lower extremity surgical interventions (RR 0.37; 95 % CI 0.20 to 0.68), including amputation (RR 0.41; 95 % CI 0.18 to 0.95). Moreover, providing G-CSF reduced the duration of hospital stay (MD, -1.40 days; 95 % CI, -2.27 to -0.53 days), but did not significantly affect the duration of systemic antibiotic therapy (MD, -0.27 days; 95 % CI, -1.30 to 0.77 days). AUTHORS' CONCLUSIONS The available evidence is limited, but suggests that adjunctive G-CSF treatment in people with a diabetic foot infection, including infected ulcers, does not appear to increase the likelihood of resolution of infection or healing of the foot ulcer. However, it does appear to reduce the need for surgical interventions, especially amputations, and the duration of hospitalisation. Clinicians might consider adding G-CSF to the usual treatment of diabetic foot infections, especially in patients with a limb-threatening infection, but it is not clear which patients might benefit.
Collapse
Affiliation(s)
- Mario Cruciani
- Center of Preventive Medicine & HIV Outpatient Clinic, G. Fracastoro Hospital, San Bonifacio, Via Germania, 20, Verona, Italy, 37135
| | | | | | | |
Collapse
|
40
|
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
|
41
|
Lara-Hernández R, Lozano-Vilardell P, Cordobés-Gual J. [Novel therapies of non-revascularizing peripheral arterial occlusive disease: therapeutic angiogenesis]. Med Clin (Barc) 2009; 131:665-9. [PMID: 19087795 DOI: 10.1157/13128727] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Critical limb ischemia is the end stage of peripheral arterial occlusive disease, with a deep impact in patient's quality of life. In some patients, there is no revascularizing treatment options, that determines major limb amputation in a high percentage of patients, not only for uncontrolled limb pain but also for complications of the trophic lesions. In the last years, several studies have shown the possibility to increase the perfusion in the ischemic tissue, by recombinant proteins, gene therapy or cellular therapy, all of them known as therapeutic angiogenesis. Several good results have been published but the way of treatment, doses and possible adverse effects still lack definitive conclusions. Randomized comparative studies should be carried out to determine the best treatment option.
Collapse
Affiliation(s)
- Raúl Lara-Hernández
- Servicio de Angiología y Cirugía Vascular. Hospital Universitario Arnau de Vilanova. Institut de Recerca Biomédica de Catalunya. Lleida. España.
| | | | | |
Collapse
|
42
|
Boda Z, Udvardy M, Rázsó K, Farkas K, Tóth J, Jámbor L, Oláh Z, Ilonczai P, Szarvas M, Kappelmayer J, Veréb Z, Rajnavölgyi E. Stem cell therapy: a promising and prospective approach in the treatment of patients with severe Buerger's disease. Clin Appl Thromb Hemost 2008; 15:552-60. [PMID: 18818231 DOI: 10.1177/1076029608319882] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
No effective blood-flow enhancement therapies are available for patients with severe peripheral arterial disease (SPAD), thus amputation remains the only option for relief of rest pain or gangrene. Autologous bone marrow-derived stem cell therapy (ABMSCT) is an emerging modality to induce angiogenesis from endothelial progenitors. A total of 5 patients with SPAD were treated by ABMSCT using isolated CD34+ cells with characterized phenotype administered by intramuscular injections. The follow-up before and 1, 3, 6, 9, and 12 months after ABMSCT was based on clinical (rest pain, walking distance without pain, nonhealing ulcers, ankle-brachial index [ABI]) and laboratory (angiography, duplex and laser ultrasonography, TcPO(2)) parameters. Significant improvement of pain and walking distance was observed in all patients. Nonhealing ulcers disappeared in 3 patients and became smaller and thinner in 1 patient. The average of ABI improved significantly on the treated limb but did not change on the contralateral limb. New collaterals were detected by angiography in 3 patients, but duplex ultrasonography detected improvement in one patient only. Laser ultrasonography showed a mild significant change, TcPO(2) values improved mainly on the foot. Severe adverse events were not observed. We conclude that ABMSCT with isolated CD34+ cells is safe, effective, and results in sustained clinical benefit for patients with SPAD.
Collapse
Affiliation(s)
- Zoltan Boda
- 2nd Department of Medicine, University of Debrecen, Debrecen, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Han Y, Yan L, Han G, Zhou X, Hong L, Yin Z, Zhang X, Wang S, Wang J, Sun A, Liu Z, Xie H, Wu K, Ding J, Fan D. Controlled trials in hepatitis B virus-related decompensate liver cirrhosis: peripheral blood monocyte transplant versus granulocyte-colony-stimulating factor mobilization therapy. Cytotherapy 2008; 10:390-6. [PMID: 18574771 DOI: 10.1080/14653240802129901] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Liver cirrhosis represents the end stage of chronic liver injury. Currently, liver transplantation provides the only definite cure but it is beset with many problems, including lack of donors and risk of rejection. Stem cell therapy is very attractive in this setting because it has the potential to help tissue regeneration. In this study, we aimed to investigate the therapeutic effect of peripheral blood monocyte cell (PBMC) transplantation in decompensated liver cirrhosis. METHODS A total of 40 subjects (31 men and nine females, age range 21-71 years) was recruited to two groups. Group 1 received granulocyte-colony-stimulating factor (G-CSF) mobilization, PBMC collection by leukapheresis and PBMC transplant therapy. Group 2 received G-CSF mobilization for 4 days. At baseline and 6 months after treatment, liver function of the two groups was monitored by blood examination and ultrasonagraphy. RESULTS Both groups gained significant improvement in liver synthetic function, such as serum albumin and prothrombin time, from baseline to 6 months after treatment (P<0.01). However, there was no significant difference in alanine aminotransferase, aspartate aminotransferase and total bilirubin in both groups (P>0.05). Compared with group 2, a significantly improved liver function was observed in group 1, including elevated serum albumin level and a decreased CTP score (P<0.05). No major adverse effects were noted. DISCUSSION Autologous PBMC transplantation could be considered as a novel and alternative treatment for patients with decompensated liver cirrhosis.
Collapse
Affiliation(s)
- Y Han
- Department of Gastroenterology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Aranguren XL, Verfaillie CM, Luttun A. Emerging hurdles in stem cell therapy for peripheral vascular disease. J Mol Med (Berl) 2008; 87:3-16. [PMID: 18712330 DOI: 10.1007/s00109-008-0394-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 06/27/2008] [Accepted: 07/17/2008] [Indexed: 12/01/2022]
Abstract
Peripheral vascular disease (PVD) is a growing medical problem in Western societies and presents itself mainly in two different clinical forms. Intermittent claudication is an early moderate manifestation, while patients with critical limb ischemia suffer from severe muscle tissue loss or ulcers and are at high risk for limb amputation. Unfortunately, many patients cannot be helped with currently available surgical or endovascular revascularization procedures because of the complex anatomy of the vascular occlusion and/or the presence of other risk factors. Noninvasive stem cell therapy has been proposed as an alternative for such patients. Although pioneering clinical experience with stem cell-related therapy seems promising, it is too early for general clinical use of this technique, since many questions remain unanswered. Indeed, while questions about safety, dose, and administration route/timing/frequency are the first ones to be addressed when designing a stem cell-based clinical approach, there is accumulating evidence from recent (pre-)clinical studies that other issues may also be at stake. For instance, the choice of stem cells to be used and its precise mechanism of action, the need/possibility for concurrent tissue regeneration in case of irreversible tissue loss, the differentiation degree and specific vascular identity of the transplanted cells, and the long-term survival of engrafted cells in the absence of a normal supportive tissue environment should be well considered. Here, rather than presenting a comprehensive and extensive overview on the current literature on stem/progenitor cells and revascularization, we highlight some of the outstanding issues emerging from the recent (pre-)clinical literature that may codetermine the successful application of stem cells in a wide range of PVD patients in the future.
Collapse
Affiliation(s)
- Xabier L Aranguren
- Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven (KULeuven), Campus Gasthuisberg, Herestraat 49, 3000, Leuven, Belgium
| | | | | |
Collapse
|
45
|
Boda Z, Udvardy M, Farkas K, Tóth J, Jámbor L, Soltész P, Rázsó K, Oláh Z, Ilonczai P, Szarvas M, Litauszky K, Hunyadi J, Sipos T, Kappelmayer J, Veréb Z, Rajnavölgyi E. [Autologous bone marrow-derived stem cell therapy in patients with severe peripheral arterial disorder]. Orv Hetil 2008; 149:531-40. [PMID: 18343769 DOI: 10.1556/oh.2008.28125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Amputation is the only current option for relief of rest pain or gangrene in patients with severe peripheral arterial disease. Up to now, no effective blood-flow enhancement therapies are available. Autologous bone marrow-derived stem cell transplantation is an arising therapy modality with an option of building new blood vessels through endothelial stem and/or progenitor cells. PATIENTS AND METHODS Five patients with severe peripheral arterial disorder were treated by autologous bone marrow-derived stem cell therapy. CD34+, CD133+ and CD45+/- cell number and ratio were determined. CD34+ cells were isolated by magnetic separation and collected into a 10 ml sample. The cell suspension was administered by local intramuscular injections (0.5-1.0 ml injections in the musculus gastrocnemius). The follow-up (before; 1, 3, 6, 9 and 12 months after the autologous bone marrow-derived stem cell therapy) based on clinical (rest pain, walking distance without pain, changes of non-healing ischaemic ulcers, ankle-brachial index) and laboratory (angiography, Color- and Laser-Doppler scan, measurement of transcutaneous oxygen tension and endothelial function test) parameters was documented and analyzed. RESULTS Improvement of pain and walking distance was observed in all five cases. In three cases the non-healing ischaemic ulcers disappeared, in one other case they became smaller and thinner, and in one case no change was realized. The average of ankle-brachial index improved significantly (before: 0.41, twelve months after: 0.83). New collaterals were detected by angiography in three patients, but duplex ultrasonography detected improvement in one patient only. Before and 1, 6 and 12 months after stem cell therapy the transcutaneous oxygen tension changed on the foot from 18.80/16.78/23.83/37.50 mmHg, and on the calf from 36.66/31.25/45.00/37.30 mmHg. The macro- and microcirculation parameters did not show improvement after 1 month, however, after 3, 6, 9 and 12 months improved parameters were recorded. Severe adverse events were not observed. In one case elevated level of serum creatinine phosphokinase, and in another case a mild form of vasculitis were detected. CONCLUSION autologous bone marrow-derived stem cell therapy with isolated CD34+ cells is effective, safe and results in sustained clinical benefit for patients with severe peripheral arterial disease.
Collapse
Affiliation(s)
- Zoltán Boda
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum, Altalános Orvostudományi Kar II. Belgyógyászati Klinika, Haemostasis Tanszék Debrecen Nagyerdei krt. 98. 4012.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yan L, Han Y, Wang J, Liu J, He Y, Wang H, Fan D. Peripheral blood monocytes from the decompensated liver cirrhosis could migrate into nude mouse liver with human hepatocyte-markers expression. Biochem Biophys Res Commun 2008; 371:635-8. [PMID: 18435910 DOI: 10.1016/j.bbrc.2008.04.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2008] [Accepted: 04/09/2008] [Indexed: 12/21/2022]
Abstract
Adult stem cells provide a promising alternative for the treatment of decompensated liver cirrhosis. Our previous study showed that peripheral blood monocytes (PBMCs) from decompensated liver cirrhosis could differentiate into hepatocyte-like cells in vitro. We here aimed to investigate the differential potential of the PBMCs under liver environment. PBMCs were isolated from one cirrhotic patient, who was mobilized by the recombinant human granulocyte colony stimulating factor for consecutive 3days, and then PBMCs were transplanted into nude mice by tail vein after labeled with PKH26-GL. The location of the transplanted PBMCs was identified by PKH26-GL staining and PRINS for human SRY gene. The expressions of human hepatocyte-markers were detected by immunohistochemistry, RT-PCR, and Western blot analysis. Our results demonstrated that PBMCs from decompensated liver cirrhosis could migrate into the liver of nude mice with human hepatocyte-markers expression, indicating that autologous PBMCs transplantation might be one alternative therapy for the decompensated liver cirrhosis.
Collapse
Affiliation(s)
- Li Yan
- Department of Gastroenterology, Xijing Hospital, The Fourth Military Medical University, 17 Changle Western Road, Xi'an, Shaanxi Province 710032, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Progenitor Cell Therapy in Patients With Critical Limb Ischemia Without Surgical Options. Ann Surg 2008; 247:411-20. [DOI: 10.1097/sla.0b013e318153fdcb] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
48
|
Wester T, Jørgensen JJ, Stranden E, Sandbæk G, Tjønnfjord G, Bay D, Kollerøs D, Kroese AJ, Brinchmann JE. Treatment with Autologous Bone Marrow Mononuclear Cells in Patients with Critical Lower Limb Ischaemia. A Pilot Study. Scand J Surg 2008; 97:56-62. [DOI: 10.1177/145749690809700108] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background and Aims: Treatment with autologous, bone marrow mononuclear stem cells has shown effects in patients with chronic limb ischaemia in one randomized clinical study. The aim of the study was to test the potential effect of stem cell treatment in a strict defined group of patients with stable critical limb ischaemia (CLI). Design: A prospective, combined-centre pilot study. Material: Eight patients with CLI of the lower extremities, and without any other treatment options. Methods: Bone marrow cells were harvested from the patient's iliac crest and, after separation, injected into the calf muscles of the affected leg. Outcome was evaluated by digital subtraction angiography (DSA), visual analogue scale (VAS) and several non-invasive circulatory physiological tests. Results: There were no complications from the procedures. Two patients were amputated two months after cell injection. Five patients reported pain relief after four months. Five patients could be evaluated at eight months. According to VAS and physiological tests, they were all either stable or showed improvement. Conclusion: This method seems to be a safe option for treating patients with CLI. Inclusion of patients took a long time, mainly because many patients with CLI are offered endovascular treatment in our institution. While symptomatic improvement was found in individual patients, larger trials are required to investigate efficacy. This will probably require multi-centre participation.
Collapse
Affiliation(s)
- T. Wester
- Department of Vascular Surgery, Aker University Hospital
| | | | - E. Stranden
- Department of Vascular Diagnosis and Research, Aker University Hospital
| | - G. Sandbæk
- Department of Radiology, Oslo Vascular Centre, Aker University Hospital
| | - G. Tjønnfjord
- Department of Immunology, Rikshospitalet-Radiumhospitalet Medical Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - D. Bay
- Department of Radiology, Oslo Vascular Centre, Aker University Hospital
| | - D. Kollerøs
- Department of Anaesthesiology, Aker University Hospital
| | - A. J. Kroese
- Department of Vascular Surgery, Aker University Hospital
| | - J. E. Brinchmann
- Department of Immunology, Rikshospitalet-Radiumhospitalet Medical Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
49
|
Yip HK, Chang LT, Sun CK, Youssef AA, Sheu JJ, Wang CJ. Shock Wave Therapy Applied to Rat Bone Marrow-Derived Mononuclear Cells Enhances Formation of Cells Stained Positive for CD31 and Vascular Endothelial Growth Factor. Circ J 2008; 72:150-6. [DOI: 10.1253/circj.72.150] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Hon-Kan Yip
- Department of Cardiology, Chang Gung Memorial Hospital Hospital-Kaohsiung Medical Center, Chang Gung University, College of Medicine
| | - Li-Teh Chang
- Basic Science, Nursing Department, Meiho Institute of Technology
| | - Cheuk-Kwan Sun
- Department of General Surgery, Chang Gung Memorial Hospital Hospital-Kaohsiung Medical Center, Chang Gung University, College of Medicine
| | | | - Jiunn-Jye Sheu
- Department of Cardiovascular Surgery, Chang Gung Memorial Hospital Hospital-Kaohsiung Medical Center, Chang Gung University, College of Medicine
| | - Ching-Jen Wang
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital Hospital-Kaohsiung Medical Center, Chang Gung University, College of Medicine
| |
Collapse
|
50
|
Yan L, Han Y, Wang J, Liu J, Hong L, Fan D. Peripheral blood monocytes from patients with HBV related decompensated liver cirrhosis can differentiate into functional hepatocytes. Am J Hematol 2007; 82:949-54. [PMID: 17724706 DOI: 10.1002/ajh.21030] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Peripheral blood monocytes (PBMCs) have the potential to differentiate into various progenitor cells. Here we have investigated the differentiation potential of PBMCs derived from patients with HBV related decompensated liver cirrhosis into hepatocyte-like cells. In our clinical trial, the PBMCs from 2 patients were mobilized by the recombinant human granulocyte colony stimulating factor, followed by leukapheresis and transplantation of PBMCs. PBMCs, induced by recombinant human hepatocyte growth factors, were identified by the expression of hepatocyte markers and specific biological functions with biochemical assays in vitro. Patients showed a lasting clinical amelioration for more than one year after transplantation, and hepatocyte-like cells were identified by expressing liver specific genes, synthesizing albumin, urea, aspirate transaminase, and glycogen, which were all similar to the human normal hepatic cell line QZG. Our results clearly demonstrated that mobilized PBMCs from patients with HBV related decompensated liver cirrhosis could differentiate into functional hepatocyte-like cells, indicating the possibility of autologous cell transplantation for treating patients with HBV related decompensated liver cirrhosis.
Collapse
Affiliation(s)
- Li Yan
- Department of Gastroenterology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | |
Collapse
|