1
|
Stella GM, Lisini D, Pedrazzoli P, Galli G, Bortolotto C, Melloni G, D’Ambrosio G, Klersy C, Grosso A, Paino F, Tomaselli S, Saracino L, Alessandri G, Pessina A, Grignani E, Rosti V, Corsico AG, Comoli P, Agustoni F. Phase I Clinical Trial on Pleural Mesothelioma Using Neoadjuvant Local Administration of Paclitaxel-Loaded Mesenchymal Stromal Cells (PACLIMES Trial): Study Rationale and Design. Cancers (Basel) 2024; 16:3391. [PMID: 39410011 PMCID: PMC11475395 DOI: 10.3390/cancers16193391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background and rationale. Pleural mesothelioma (PM) is a rare and aggressive neoplasm that originates from the pleural mesothelium and whose onset is mainly linked to exposure to asbestos, which cannot be attacked with truly effective therapies with consequent poor prognosis. The rationale of this study is based on the use of mesenchymal stromal cells (MSCs) as a vehicle for chemotherapy drugs to be injected directly into the pathological site, such as the pleural cavity. Study design. The study involves the use of a conventional chemotherapeutic drug, Paclitaxel (PTX), which is widely used in the treatment of different types of solid tumors, including PM, although some limitations are related to pharmacokinetic aspects. The use of PTX-loaded MSCs to treat PM should provide several potential advantages over the systemically administered drug as reduced toxicity and increased concentration of active drug in the tumor-surrounding context. The PACLIMES trial explores the safety and toxicity of the local administration of Paclimes in chemonaive patients, candidates for pleurectomy. The secondary objective is to find the effective Paclimes dose for subsequent phase II studies and to observe and record the antitumor activity. Future direction. The experimental pre-clinical background and rationale are discussed as well.
Collapse
Affiliation(s)
- Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (P.P.); (G.G.); (A.G.C.); (F.A.)
- Unit of Respiratory Diseases, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.G.); (S.T.); (L.S.)
| | - Daniela Lisini
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Paolo Pedrazzoli
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (P.P.); (G.G.); (A.G.C.); (F.A.)
- Medical Oncology Unit, Oncology and Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulia Galli
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (P.P.); (G.G.); (A.G.C.); (F.A.)
- Medical Oncology Unit, Oncology and Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
- Radiology Unit-Diagnostic Imaging I, Department of Diagnostic Medicine, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulio Melloni
- Unit of Thoracic Surgery, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gioacchino D’Ambrosio
- Pathology Unit, Department of Diagnostical Services and Imaging, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Catherine Klersy
- Biostatistics and Clinical Trial Center, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, 27100 Pavia, Italy;
| | - Amelia Grosso
- Unit of Respiratory Diseases, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.G.); (S.T.); (L.S.)
| | - Francesca Paino
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (F.P.); (G.A.); (A.P.)
| | - Stefano Tomaselli
- Unit of Respiratory Diseases, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.G.); (S.T.); (L.S.)
| | - Laura Saracino
- Unit of Respiratory Diseases, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.G.); (S.T.); (L.S.)
| | - Giulio Alessandri
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (F.P.); (G.A.); (A.P.)
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (F.P.); (G.A.); (A.P.)
| | - Elena Grignani
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Vittorio Rosti
- Phase 1 Clinical Trial Unit and Experimental Therapy, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Angelo Guido Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (P.P.); (G.G.); (A.G.C.); (F.A.)
- Unit of Respiratory Diseases, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.G.); (S.T.); (L.S.)
| | - Patrizia Comoli
- Cell Factory, Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Francesco Agustoni
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (P.P.); (G.G.); (A.G.C.); (F.A.)
- Medical Oncology Unit, Oncology and Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
2
|
Karami Fath M, Bagherzadeh Torbati SM, Saqagandomabadi V, Yousefi Afshar O, Khalilzad M, Abedi S, Moliani A, Daneshdoust D, Barati G. The therapeutic effect of MSCs and their extracellular vesicles on neuroblastoma. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 187:51-60. [PMID: 38373516 DOI: 10.1016/j.pbiomolbio.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/04/2023] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Neuroblastoma is a common inflammatory-related cancer during infancy. Standard treatment modalities including surgical interventions, high-dose chemotherapy, radiotherapy, and immunotherapy are not able to increase survival rate and reduce tumor relapse in high-risk patients. Mesenchymal stem cells (MSCs) are known for their tumor-targeting and immunomodulating properties. MSCs could be engineered to express anticancer agents (i.e., growth factors, cytokines, pro-apoptotic agents) or deliver oncolytic viruses in the tumor microenvironment. As many functions of MSCs are mediated through their secretome, researchers have tried to use extracellular vesicles (EVs) from MSCs for targeted therapy of neuroblastoma. Here, we reviewed the studies to figure out whether the use of MSCs could be worthwhile in neuroblastoma therapy or not. Native MSCs have shown a promoting or inhibiting role in cancers including neuroblastoma. Therefore, MSCs are proposed as a vehicle to deliver anticancer agents such as oncolytic viruses to the neuroblastoma tumor microenvironment. Although modified MSCs or their EVs have been shown to suppress the tumorigenesis of neuroblastoma, further pre-clinical and clinical studies are required to come to a conclusion.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Vahid Saqagandomabadi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | - Mohammad Khalilzad
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Abedi
- Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Afshin Moliani
- Isfahan Medical Students Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Danyal Daneshdoust
- Faculty of Medicine, Babol University of Medical Sciences, Mazandaran, Iran
| | | |
Collapse
|
3
|
Wu CZ, Shi ZY, Wu Z, Lin WJ, Chen WB, Jia XW, Xiang SC, Xu HH, Ge QW, Zou KA, Wang X, Chen JL, Wang PE, Yuan WH, Jin HT, Tong PJ. Mid-term outcomes of microfragmented adipose tissue plus arthroscopic surgery for knee osteoarthritis: A randomized, active-control, multicenter clinical trial. World J Stem Cells 2023; 15:1063-1076. [PMID: 38179213 PMCID: PMC10762526 DOI: 10.4252/wjsc.v15.i12.1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most prevalent form of degenerative whole-joint disease. Before the final option of knee replacement, arthroscopic surgery was the most widely used joint-preserving surgical treatment. Emerging regenerative therapies, such as those involving platelet-rich plasma, mesenchymal stem cells, and microfragmented adipose tissue (MFAT), have been pushed to the forefront of treatment to prevent the progression of OA. Currently, MFAT has been successfully applied to treat different types of orthopedic diseases. AIM To assess the efficacy and safety of MFAT with arthroscopic surgery in patients with knee OA (KOA). METHODS A randomized, multicenter study was conducted between June 2017 and November 2022 in 10 hospitals in Zhejiang, China. Overall, 302 patients diagnosed with KOA (Kellgren-Lawrence grades 2-3) were randomized to the MFAT group (n = 151, were administered MFAT following arthroscopic surgery), or the control group (n = 151, were administered hyaluronic acid following arthroscopic surgery). The study outcomes were changes in the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, the visual analog scale (VAS) score, the Lequesne index score, the Whole-Organ Magnetic Resonance Imaging Score (WORMS), and safety over a 24-mo period from baseline. RESULTS The changes in the WOMAC score (including the three subscale scores), VAS pain score, and Lequesne index score at the 24-mo mark were significantly different in the MFAT and control groups, as well as when comparing values at the posttreatment visit and those at baseline (P < 0.001). The MFAT group consistently demonstrated significant decreases in the WOMAC pain scores and VAS scores at all follow-ups compared to the control group (P < 0.05). Furthermore, the WOMAC stiffness score, WOMAC function score, and Lequesne index score differed significantly between the groups at 12 and 24 mo (P < 0.05). However, no significant between-group differences were observed in the WORMS at 24 mo (P = 0.367). No serious adverse events occurred in both groups. CONCLUSION The MFAT injection combined with arthroscopic surgery treatment group showed better mid-term clinical outcomes compared to the control group, suggesting its efficacy as a therapeutic approach for patients with KOA.
Collapse
Affiliation(s)
- Cong-Zi Wu
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zhen-Yu Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Zhen Wu
- Department of Orthopaedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Wen-Jun Lin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang Province, China
| | - Wei-Bo Chen
- Department of Orthopaedic Surgery, Ruian Hospital of Traditional Chinese Medicine, Wenzhou 325299, Zhejiang Province, China
| | - Xue-Wen Jia
- Department of Orthopaedics, Ningbo First Hospital, Ningbo 315010, Zhejiang Province, China
| | - Si-Cheng Xiang
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Hui-Hui Xu
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Qin-Wen Ge
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Kai-Ao Zou
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Xu Wang
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jia-Li Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Ping-Er Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Wen-Hua Yuan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Hong-Ting Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Pei-Jian Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China.
| |
Collapse
|
4
|
Liu M, Lu F, Feng J. Therapeutic potential of adipose tissue derivatives in skin photoaging. Regen Med 2023; 18:869-883. [PMID: 37743749 DOI: 10.2217/rme-2023-0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Photoaging, the primary cause of exogenous skin aging and predominantly caused by ultraviolet radiation, is an essential type of skin aging characterized by chronic skin inflammation. Recent studies have shown that oxidative stress, inflammation, skin barrier homeostasis, collagen denaturation and pigmentation are the main contributors to it. As a composite tissue rich in matrix and vascular components, adipose tissue derivatives have been recently gaining attention as potential therapeutic agents for various human diseases with fat-processing technology upgrades. This review analyzes both 'minimally treated' and 'nonminimally treated' fat derivatives to give an overview of the preclinical and clinical relevance of adipose tissue derivatives for antiphotoaging application, highlighting their good clinical prospects as well as discussing their safety and potential risks.
Collapse
Affiliation(s)
- Meiqi Liu
- Department of Plastic & Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China
| | - Feng Lu
- Department of Plastic & Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China
| | - Jingwei Feng
- Department of Plastic & Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
5
|
Salagean A, Nechifor-Boila A, Bajwa N, Pastorello Y, Slevin M. Micro-Fragmented Adipose Tissue as a Natural Scaffold for Targeted Drug Delivery in Brain Cancer. Int J Mol Sci 2023; 24:11530. [PMID: 37511289 PMCID: PMC10380718 DOI: 10.3390/ijms241411530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Major limitations in the effective treatment of neurological cancer include systemic cytotoxicity of chemotherapy, inaccessibility, and inoperability. The capability to successfully target a drug to the tumor site(s) without incurring serious side effects-especially in the case of aggressive tumors, such as glioblastoma and neuroblastoma-would represent a significant breakthrough in therapy. Orthotopic systems, capable of storing and releasing proteins over a prolonged period at the site of a tumor, that utilize nanoparticles, liposomes, and hydrogels have been proposed. One candidate for drug delivery is Micro-Fragmented Adipose Tissue (MFAT). Easily obtained from the patient by abdominal subcutaneous liposuction (autologous), and with a high content of Mesenchymal Stem Cells (MSCs), mechanically derived nanofat is a natural tissue graft with a structural scaffold organization. It has a well-preserved stromal vascular fraction and a prolonged capacity to secrete anti-tumorigenic concentrations of pre-absorbed chemotherapeutics within extracellular vesicles. This review discusses current evidence supporting the potential of drug-modified MFAT for the treatment of neurological cancer with respect to recent preclinical and in vitro studies. Possible limitations and future perspectives are considered.
Collapse
Affiliation(s)
- Alex Salagean
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology, 540142 Târgu Mures, Romania
| | - Adela Nechifor-Boila
- Department of Histology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, 540142 Târgu Mures, Romania
| | - Nosherwan Bajwa
- Department of Neurosurgery, DIAKO Hospital, 24939 Flensburg, Germany
| | - Ylenia Pastorello
- Department of Anatomy and Embryology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, 540142 Târgu Mures, Romania
| | - Mark Slevin
- Center for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science and Technology, 540142 Târgu Mures, Romania
| |
Collapse
|
6
|
Coccè V, Bonelli M, La Monica S, Alfieri R, Doneda L, Martegani E, Alessandri G, Annamaria Lagrasta C, Giannì A, Sordi V, Petrella F, Roncoroni L, Paino F, Pessina A. Mesenchymal stromal cells loaded with Paclitaxel (PacliMES) a potential new therapeutic approach on mesothelioma. Biochem Pharmacol 2023:115678. [PMID: 37399948 DOI: 10.1016/j.bcp.2023.115678] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/05/2023]
Abstract
Malignant pleural mesothelioma is an asbestos-related tumor originating in mesothelial cells of the pleura that poorly responds to chemotherapeutic approaches. Adult mesenchymal stromal cells derived either from bone marrow or from adipose tissue may be considered a good model for cell-based therapy, a treatment which has experienced significant interest in recent years. The present study confirms that Paclitaxel is effective on mesothelioma cell proliferation in 2D and 3D in vitro cultures, and that 80,000 mesenchymal stromal cells loaded with Paclitaxel inhibit tumor growth at a higher extent than Paclitaxel alone. An in vivo approach to treat in situ mesothelioma xenografts using a minimal amount of 106 mesenchymal stromal cells loaded with Paclitaxel showed the same efficacy of a systemic administration of 10 mg/kg of Paclitaxel. These data strongly support drug delivery system by mesenchymal stromal cells as a useful approach against many solid tumors. We look with interest at the favourable opinion recently expressed by the Italian Drug Agency on the procedure for the preparation of mesenchymal stromal cells loaded with Paclitaxel in large-scale bioreactor systems and their storage until clinical use. This new Advanced Medicinal Therapy Product, already approved for a Phase I clinical trial on mesothelioma patients, could pave the way for mesenchymal stromal cells use as drug delivery system on other solid tumors for adjuvant therapy associated with surgery and radiotherapy.
Collapse
Affiliation(s)
- Valentina Coccè
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Mara Bonelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Luisa Doneda
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Eleonora Martegani
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Giulio Alessandri
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | | | - Aldo Giannì
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; Maxillo-Facial and Dental Unit, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Hospital, 20132, Milan, Italy
| | - Francesco Petrella
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; Department of Thoracic Surgery, IRCCS European Institute of Oncology, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Leda Roncoroni
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Francesca Paino
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| |
Collapse
|
7
|
La Monica S, Coccé V, Bonelli M, Alessandri G, Alfieri R, Lagrasta CA, Frati C, Flammini L, Gianni A, Petrella F, Paino F, Pessina A. Micro-fragmented Fat Inhibits the Progression of Human Mesothelioma Xenografts in Mice. Curr Cancer Drug Targets 2023; 23:663-668. [PMID: 36722476 DOI: 10.2174/1568009623666230201092302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Malignant pleural mesothelioma is a pathology with no effective therapy and a poor prognosis. Our previous study demonstrated an in vitro inhibitory effect on mesothelioma cell lines of both the lysate and secretome of adipose tissue-derived Mesenchymal Stromal Cells. The inhibitory activity on tumor growth has been demonstrated also in vivo: five million Mesenchymal Stromal Cells, injected "in situ", produced a significant therapeutic efficacy against MSTO-211H xenograft equivalent to that observed after the systemic administration of paclitaxel. OBJECTIVE The objective of this study is to evaluate the efficacy of low amount (half a million) Mesenchymal Stromal Cells and micro-fragmented adipose tissues (the biological tissue from which the Mesenchymal Stromal Cells were isolated) on mesothelioma cells growth. METHODS Tumor cells growth inhibition was evaluated in vitro and in a xenograft model of mesothelioma. RESULTS The inhibitory effect of micro-fragmented fat from adipose-tissue has been firstly confirmed in vitro on MSTO-211H cell growth. Then the efficacy against the growth of mesothelioma xenografts in mice of both micro-fragmented fat and low amount of Mesenchymal Stromal Cells has been evaluated. Our results confirmed that both Mesenchymal Stromal Cells and micro-fragmented fat, injected "in situ", did not stimulate mesothelioma cell growth. By contrast, micro-fragmented fat produced a significant inhibition of tumor growth and progression, comparable to that observed by the treatment with paclitaxel. Low amount of Mesenchymal Stromal Cells exerted only a little anticancer activity. CONCLUSION Micro-fragmented fat inhibited mesothelioma cell proliferation in vitro and exerted a significant control of the mesothelioma xenograft growth in vivo.
Collapse
Affiliation(s)
- Silvia La Monica
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Valentina Coccé
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via della Commenda 10, Milan, 20122, Italy
| | - Mara Bonelli
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Giulio Alessandri
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via della Commenda 10, Milan, 20122, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | | | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Lisa Flammini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, Parma, 43124, Italy
| | - Aldo Gianni
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Francesco Petrella
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via della Commenda 10, Milan, 20122, Italy
| | - Francesca Paino
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via della Commenda 10, Milan, 20122, Italy
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via della Commenda 10, Milan, 20122, Italy
| |
Collapse
|
8
|
Manocha E, Consonni A, Baggi F, Ciusani E, Cocce V, Paino F, Tremolada C, Caruso A, Alessandri G. CD146+ Pericytes Subset Isolated from Human Micro-Fragmented Fat Tissue Display a Strong Interaction with Endothelial Cells: A Potential Cell Target for Therapeutic Angiogenesis. Int J Mol Sci 2022; 23:ijms23105806. [PMID: 35628617 PMCID: PMC9144360 DOI: 10.3390/ijms23105806] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Pericytes (PCs) are mesenchymal stromal cells (MSCs) that function as support cells and play a role in tissue regeneration and, in particular, vascular homeostasis. PCs promote endothelial cells (ECs) survival which is critical for vessel stabilization, maturation, and remodeling. In this study, PCs were isolated from human micro-fragmented adipose tissue (MFAT) obtained from fat lipoaspirate and were characterized as NG2+/PDGFRβ+/CD105+ cells. Here, we tested the fat-derived PCs for the dispensability of the CD146 marker with the aim of better understanding the role of these PC subpopulations on angiogenesis. Cells from both CD146-positive (CD146+) and negative (CD146−) populations were observed to interact with human umbilical vein ECs (HUVECs). In addition, fat-derived PCs were able to induce angiogenesis of ECs in spheroids assay; and conditioned medium (CM) from both PCs and fat tissue itself led to the proliferation of ECs, thereby marking their role in angiogenesis stimulation. However, we found that CD146+ cells were more responsive to PDGF-BB-stimulated migration, adhesion, and angiogenic interaction with ECs, possibly owing to their higher expression of NCAM/CD56 than the corresponding CD146− subpopulation. We conclude that in fat tissue, CD146-expressing cells may represent a more mature pericyte subpopulation that may have higher efficacy in controlling and stimulating vascular regeneration and stabilization than their CD146-negative counterpart.
Collapse
Affiliation(s)
- Ekta Manocha
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.C.); (G.A.)
- Correspondence:
| | - Alessandra Consonni
- Neurology IV—Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (A.C.); (F.B.)
| | - Fulvio Baggi
- Neurology IV—Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (A.C.); (F.B.)
| | - Emilio Ciusani
- Laboratory of Neurological Biochemistry and Neuropharmacology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Valentina Cocce
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (V.C.); (F.P.)
| | - Francesca Paino
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (V.C.); (F.P.)
| | - Carlo Tremolada
- Department of Stem Cells and Regenerative Medicine, Image Institute, 20122 Milan, Italy;
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.C.); (G.A.)
| | - Giulio Alessandri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.C.); (G.A.)
- Department of Stem Cells and Regenerative Medicine, Image Institute, 20122 Milan, Italy;
| |
Collapse
|
9
|
Single-Shot Local Injection of Microfragmented Fat Tissue Loaded with Paclitaxel Induces Potent Growth Inhibition of Hepatocellular Carcinoma in Nude Mice. Cancers (Basel) 2021; 13:cancers13215505. [PMID: 34771667 PMCID: PMC8583409 DOI: 10.3390/cancers13215505] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is poorly beneficiated by intravenous chemotherapy due to inadequate availability of drugs at the tumor site. We previously demonstrated that human micro-fragmented adipose tissue (MFAT) and its devitalized counterpart (DMFAT) could be effective natural scaffolds to deliver Paclitaxel (PTX) to tumors in both in vitro and in vivo tests, affecting cancer growth relapse. Here we tested the efficacy of DMFAT-PTX in a well-established HCC in nude mice. MFAT-PTX and DMFAT-PTX preparations were tested for anti-cancer activity in 2D and 3D assays using Hep-3B tumor cells. The efficacy of DMFAT-PTX was evaluated after a single-shot subcutaneous injection near a Hep-3B growing tumor by assessing tumor volumes, apoptosis rate, and drug pharmacokinetics in an in vivo model. Potent antiproliferative activity was seen in both in vitro 2D and 3D tests. Mice treated with DMFAT-PTX (10 mg/kg) produced potent Hep-3B growth inhibition with 33% complete tumor regressions. All treated animals experienced tumor ulceration at the site of DMFAT-PTX injection, which healed spontaneously. Lowering the drug concentration (5 mg/kg) prevented the formation of ulcers, maintaining statistically significant efficacy. Histology revealed a higher number of apoptotic cancer cells intratumorally, suggesting prolonged presence of PTX that was confirmed by the pharmacokinetic analysis. DMFAT may be a potent and valid new tool for local chemotherapy of HCC in an advanced stage of progression, also suggesting potential effectiveness in other human primary inoperable cancers.
Collapse
|
10
|
Injectable in situ forming hydrogels incorporating dual-nanoparticles for chemo-photothermal therapy of breast cancer cells. Int J Pharm 2021; 600:120510. [PMID: 33766636 DOI: 10.1016/j.ijpharm.2021.120510] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/03/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023]
Abstract
Chemo-photothermal therapy (chemo-PTT) mediated by nanomaterials holds a great potential for cancer treatment. However, the tumor uptake of the systemically administered nanomaterials was recently found to be below 1%. To address this limitation, the development of injectable tridimensional polymeric matrices capable of delivering nanomaterials directly into the tumor site appears to be a promising approach. In this work, an injectable in situ forming ionotropically crosslinked chitosan-based hydrogel co-incorporating IR780 loaded nanoparticles (IR/BPN) and Doxorubicin (DOX) loaded nanoparticles (DOX/TPN) was developed for application in breast cancer chemo-PTT. The produced hydrogels (IR/BPN@Gel and IR/BPN+DOX/TPN@Gel) displayed suitable physicochemical properties and produced a temperature increase of about 9.1 °C upon exposure to Near Infrared (NIR) light. As importantly, the NIR-light exposure also increased the release of DOX from the hydrogel by 1.7-times. In the in vitro studies, the combination of IR/BPN@Gel with NIR light (photothermal therapy) led to a reduction in the viability of breast cancer cells to 35%. On the other hand, the non-irradiated IR/BPN+DOX/TPN@Gel (chemotherapy) only diminished cancer cells' viability to 85%. In contrast, the combined action of IR/BPN+DOX/TPN@Gel and NIR light reduced cancer cells' viability to about 9%, demonstrating its potential for breast cancer chemo-PTT.
Collapse
|
11
|
The Potential of Mesenchymal Stromal Cells in Neuroblastoma Therapy for Delivery of Anti-Cancer Agents and Hematopoietic Recovery. J Pers Med 2021; 11:jpm11030161. [PMID: 33668854 PMCID: PMC7996318 DOI: 10.3390/jpm11030161] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common pediatric cancers and a major cause of cancer-related death in infancy. Conventional therapies including high-dose chemotherapy, stem cell transplantation, and immunotherapy approach a limit in the treatment of high-risk neuroblastoma and prevention of relapse. In the last two decades, research unraveled a potential use of mesenchymal stromal cells in tumor therapy, as tumor-selective delivery vehicles for therapeutic compounds and oncolytic viruses and by means of supporting hematopoietic stem cell transplantation. Based on pre-clinical and clinical advances in neuroblastoma and other malignancies, we assess both the strong potential and the associated risks of using mesenchymal stromal cells in the therapy for neuroblastoma. Furthermore, we examine feasibility and safety aspects and discuss future directions for harnessing the advantageous properties of mesenchymal stromal cells for the advancement of therapy success.
Collapse
|
12
|
Zeira O, Ghezzi E, Pettinari L, Re V, Lupi DM, Benali SL, Borgonovo S, Alessandri G, Petrella F, Paroni R, Dei Cas M, Tremolada C, Coccè V, Pessina A. Case Report: Microfragmented Adipose Tissue Drug Delivery in Canine Mesothelioma: A Case Report on Safety, Feasibility, and Clinical Findings. Front Vet Sci 2021; 7:585427. [PMID: 33569396 PMCID: PMC7869746 DOI: 10.3389/fvets.2020.585427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/04/2020] [Indexed: 11/20/2022] Open
Abstract
Mesothelioma is a rare lethal tumor of dogs and humans involving cavities of the body. Dogs are considered a model for new drugs and therapeutic methods since they present spontaneous diseases similar to humans. Microfragmented adipose tissue (MFAT) uploaded by paclitaxel (PTX) is a drug delivery medium providing slow release of chemotherapic drugs. A dog affected by pleural, pericardial, and peritoneal mesothelioma was treated by 17 intracavitary ultrasound-guided injections of MFAT-PTX over 22 months. A long-lasting improvement of general conditions was observed, treatment was well-tolerated, and no toxicity or hypersensitivity was reported. Pharmacokinetic (PK) data indicated low drug localization in the circulatory system and a tendency to enter or remain in the extravascular compartments of the body. Indeed, low levels of free-circulating drugs for a short time produced low toxicity, whereas, a higher intracavitary PTX concentration can have major pharmacological efficacy. To our knowledge, this is the first time that mesothelioma has been treated using such a procedure, and this should be considered as a novel therapeutic approach. The low systemic absorption suggests the possible role of MFAT-PTX for loco-regional/intratumoral therapy also useful in other types of tumors, and further investigation is warranted.
Collapse
Affiliation(s)
- Offer Zeira
- Department of Stem Cells and Regenerative Medicine, San Michele Veterinary Hospital, Tavazzano con Villavesco, Italy
| | - Erica Ghezzi
- Department of Stem Cells and Regenerative Medicine, San Michele Veterinary Hospital, Tavazzano con Villavesco, Italy
| | - Letizia Pettinari
- Department of Stem Cells and Regenerative Medicine, San Michele Veterinary Hospital, Tavazzano con Villavesco, Italy
| | - Valentina Re
- Department of Stem Cells and Regenerative Medicine, San Michele Veterinary Hospital, Tavazzano con Villavesco, Italy
| | - Davide M Lupi
- Department of Stem Cells and Regenerative Medicine, San Michele Veterinary Hospital, Tavazzano con Villavesco, Italy
| | | | | | - Giulio Alessandri
- Department of Cerebrovascular Diseases, Istituto di Ricovero e Cura a Carattere Scientifico, Besta Neurological Institute, Milan, Italy.,Centro di Ricerca Coordinato StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Francesco Petrella
- Department of Stem Cells and Regenerative Medicine, Istituto di Ricovero e Cura a Carattere Scientifico, European Institute of Oncology, Milan, Italy.,Centro di Ricerca Coordinato StaMeTec, Department of Oncology and Emato-Oncology, University of Milan, Milan, Italy
| | - Rita Paroni
- Centro di Ricerca Coordinato StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.,Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Michele Dei Cas
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Carlo Tremolada
- Department of Stem Cells and Regenerative Medicine, Istituto Image, Milan, Italy
| | - Valentina Coccè
- Centro di Ricerca Coordinato StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Augusto Pessina
- Centro di Ricerca Coordinato StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| |
Collapse
|
13
|
Ebadi M, Bullo S, Buskara K, Hussein MZ, Fakurazi S, Pastorin G. Release of a liver anticancer drug, sorafenib from its PVA/LDH- and PEG/LDH-coated iron oxide nanoparticles for drug delivery applications. Sci Rep 2020; 10:21521. [PMID: 33298980 PMCID: PMC7725814 DOI: 10.1038/s41598-020-76504-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022] Open
Abstract
The use of nanocarriers composed of polyethylene glycol- and polyvinyl alcohol-coated vesicles encapsulating active molecules in place of conventional chemotherapy drugs can reduce many of the chemotherapy-associated challenges because of the increased drug concentration at the diseased area in the body. The present study investigated the structure and magnetic properties of iron oxide nanoparticles in the presence of polyvinyl alcohol and polyethylene glycol as the basic surface coating agents. We used superparamagnetic iron oxide nanoparticles (FNPs) as the core and studied their effectiveness when two polymers, namely polyvinyl alcohol (PVA) and polyethylene glycol (PEG), were used as the coating agents together with magnesium-aluminum-layered double hydroxide (MLDH) as the nanocarrier. In addition, the anticancer drug sorafenib (SO), was loaded on MLDH and coated onto the surface of the nanoparticles, to best exploit this nano-drug delivery system for biomedical applications. Samples were prepared by the co-precipitation method, and the resulting formation of the nanoparticles was confirmed by X-ray, FTIR, TEM, SEM, DLS, HPLC, UV-Vis, TGA and VSM. The X-ray diffraction results indicated that all the as-synthesized samples contained highly crystalline and pure Fe3O4. Transmission electron microscopy analysis showed that the shape of FPEGSO-MLDH nanoparticles was generally spherical, with a mean diameter of 17 nm, compared to 19 nm for FPVASO-MLDH. Fourier transform infrared spectroscopy confirmed the presence of nanocarriers with polymer-coating on the surface of iron oxide nanoparticles and the existence of loaded active drug consisting of sorafenib. Thermogravimetric analyses demonstrated the thermal stability of the nanoparticles, which displayed enhanced anticancer effect after coating. Vibrating sample magnetometer (VSM) curves of both produced samples showed superparamagnetic behavior with the high saturation magnetization of 57 emu/g for FPEGSO-MLDH and 49 emu/g for FPVASO-MLDH. The scanning electron microscopy (SEM) images showed a narrow size distribution of both final samples. The SO drug loading and the release behavior from FPEGSO-MLDH and FPVASO-MLDH were assessed by ultraviolet-visible spectroscopy. This evaluation showed around 85% drug release within 72 h, while 74% of sorafenib was released in phosphate buffer solution at pH 4.8. The release profiles of sorafenib from the two designed samples were found to be sustained according to pseudo-second-order kinetics. The cytotoxicity studies confirmed the anti-cancer activity of the coated nanoparticles loaded with SO against liver cancer cells, HepG2. Conversely, the drug delivery system was less toxic than the pure drug towards fibroblast-type 3T3 cells.
Collapse
Affiliation(s)
- Mona Ebadi
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Selangor, Malaysia
| | - Saifullah Bullo
- Department of Linguistic and Human Sciences, Begum Nusrat Bhutto Women University, Sukkur, Sindh, 65200, Pakistan
| | - Kalaivani Buskara
- Laboratory of Vaccine and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohd Zobir Hussein
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Selangor, Malaysia.
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Laboratory of Vaccine and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| |
Collapse
|
14
|
Liang T, Wen D, Zhong X, Jiang L, Zhu JJ, Gu Z. Therapeutic potential of adipose tissue. Sci Bull (Beijing) 2020; 65:1702-1704. [PMID: 36659240 DOI: 10.1016/j.scib.2020.06.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Tingxizi Liang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Di Wen
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Xintong Zhong
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Liping Jiang
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
15
|
Li Q, Zhao F, Li Z, Duan X, Cheng J, Zhang J, Fu X, Zhang J, Shao Z, Guo Q, Hu X, Ao Y. Autologous Fractionated Adipose Tissue as a Natural Biomaterial and Novel One-Step Stem Cell Therapy for Repairing Articular Cartilage Defects. Front Cell Dev Biol 2020; 8:694. [PMID: 32903809 PMCID: PMC7438948 DOI: 10.3389/fcell.2020.00694] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022] Open
Abstract
Articular cartilage damage remains a tough challenge for clinicians. Stem cells have emerged promising biologics in regenerative medicine. Previous research has widely demonstrated that adipose-derived mesenchymal stem cells (ADSCs) can promote cartilage repair due to their multipotency. However, enzymatic isolation and monolayer expansion of ADSCs decrease their differentiation potential and limit their clinical application. Here, a novel adipose tissue-derived product, extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel), was obtained by simple mechanical shifting and centrifugation to separate the fat oil and concentrate the effective constituents. This study aimed to evaluate the therapeutic effect of this natural biomaterial on the repair of articular cartilage defects. Scanning electron microscopy showed that the fibrous structure in the ECM/SVF-gel was preserved. ADSCs sprouted from the ECM/SVF-gel were characterized by their ability of differentiation into chondrocytes, osteoblasts, and adipocytes. In a rabbit model, critical-sized cartilage defects (diameter, 4 mm; depth, 1.5 mm) were created and treated with microfracture (MF) or a combination of autologous ECM/SVF-gel injection. The knee joints were evaluated at 6 and 12 weeks through magnetic resonance imaging, macroscopic observation, histology, and immunohistochemistry. The International Cartilage Repair Society score and histological score were significantly higher in the ECM/SVF-gel group than those in the MF-treated group. The ECM/SVF-gel distinctly improved cartilage regeneration, integration with surrounding normal cartilage, and the expression of hyaline cartilage marker, type II collagen, in comparison with the MF treatment alone. Overall, the ready-to-use ECM/SVF-gel is a promising therapeutic strategy to facilitate articular cartilage regeneration. Moreover, due to the simple, time-sparing, cost-effective, enzyme-free, and minimally invasive preparation process, this gel provides a valuable alternative to stem cell-based therapy for clinical translation.
Collapse
Affiliation(s)
- Qi Li
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Fengyuan Zhao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Zong Li
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Xiaoning Duan
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Jin Cheng
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Jiahao Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Xin Fu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Jiying Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Zhenxing Shao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Qinwei Guo
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Yingfang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| |
Collapse
|
16
|
Badekila AK, Kini S, Jaiswal AK. Fabrication techniques of biomimetic scaffolds in three-dimensional cell culture: A review. J Cell Physiol 2020; 236:741-762. [PMID: 32657458 DOI: 10.1002/jcp.29935] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022]
Abstract
In the last four decades, several researchers worldwide have routinely and meticulously exercised cell culture experiments in two-dimensional (2D) platforms. Using traditionally existing 2D models, the therapeutic efficacy of drugs has been inappropriately validated due to the failure in generating the precise therapeutic response. Fortunately, a 3D model addresses the foregoing limitations by recapitulating the in vivo environment. In this context, one has to contemplate the design of an appropriate scaffold for favoring the organization of cell microenvironment. Instituting pertinent model on the platter will pave way for a precise mimicking of in vivo conditions. It is because animal cells in scaffolds oblige spontaneous formation of 3D colonies that molecularly, phenotypically, and histologically resemble the native environment. The 3D culture provides insight into the biochemical aspects of cell-cell communication, plasticity, cell division, cytoskeletal reorganization, signaling mechanisms, differentiation, and cell death. Focusing on these criteria, this paper discusses in detail, the diversification of polymeric scaffolds based on their available resources. The paper also reviews the well-founded and latest techniques of scaffold fabrication, and their applications pertaining to tissue engineering, drug screening, and tumor model development.
Collapse
Affiliation(s)
- Anjana K Badekila
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Sudarshan Kini
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Amit K Jaiswal
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
17
|
Tran PHL, Tran TTD. Current Designs and Developments of Fucoidan-based Formulations for Cancer Therapy. Curr Drug Metab 2020; 20:933-941. [PMID: 31589118 DOI: 10.2174/1389200220666191007154723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/28/2019] [Accepted: 09/04/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND Natural nanostructure materials have been involved in antitumor drug delivery systems due to their biocompatibility, biodegradation, and bioactive properties. METHODS These materials have contributed to advanced drug delivery systems in the roles of both bioactive compounds and delivery nanocarriers. Fucoidan, a valuable ocean material used in drug delivery systems, has been exploited in research on cancer and a variety of other diseases. RESULTS Although the uniqueness, structure, properties, and health benefits of fucoidan have been mentioned in various prominent reviews, current developments and designs of fucoidan-based formulations still need to be assessed to further develop an effective anticancer therapy. In this review, current important formulations using fucoidan as a functional material and as an anticancer agent will be discussed. This article will also provide a brief principle of the methods that incorporate functional nanostructure materials in formulations exploiting fucoidan. CONCLUSION Current research and future perspectives on the use of fucoidan in anticancer therapy will advance innovative and important products for clinical uses.
Collapse
Affiliation(s)
| | - Thao T D Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam.,Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
18
|
Zhang Z, Du Y, Liu T, Wong KH, Chen T. Systematic acute and subchronic toxicity evaluation of polysaccharide-protein complex-functionalized selenium nanoparticles with anticancer potency. Biomater Sci 2020; 7:5112-5123. [PMID: 31573569 DOI: 10.1039/c9bm01104h] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Functionalized selenium nanoparticles (SeNPs) have demonstrated potential for applications in cancer chemotherapy, radio-sensitization, nephroprotection and drug delivery. However, their clinical application requires further systemic safety evaluation. Therefore, in this study, we examine the systematic acute and subchronic toxicity of polysaccharide-protein complex coated SeNPs (PTR-SeNPs). These particles exhibited a low oral acute toxicity (higher LD50) in SPF grade ICR mice and SD rats, and the evaluation of subchronic toxicity demonstrated that the no observed effect level (NOAEL) of the PTR-SeNPs was less than 200 μg Se per kg BW per day, which is about 30 times the tolerable upper intake levels of Se in the human body. In addition, we also found that, under a safe dose (0.75-7.5 mg kg-1), the oral administration of PTR-SeNPs dramatically inhibited the growth of cancer in a tumor-bearing nude mouse model, and the results of the histological analysis indicated that PTR-SeNPs did not significantly damage the major organs, including the liver, spleen, heart, kidneys and lungs. Moreover, the induction of caspase activation and mitochondrial dysfunction was the major anticancer action mechanism of PTR-SeNPs. Taken together, the results of this study provide a simple approach for the facile and large-scale manufacturing of SeNPs with reduced toxicity and enhanced anticancer activity through the regulation of the surface properties of SeNPs. Furthermore, this study generates evidence for the future exploration and translational application of these materials through oral administration in nanomedicine and nutritional sciences.
Collapse
Affiliation(s)
- Zehang Zhang
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | | | | | | | | |
Collapse
|
19
|
Gupta AA, Kheur S, Arakeri G, Thirumal Raj A, Badhe RV, Patil S, Rao Us V, Patil S, Gomez RS, Thomson P, Brennan PA. Efficacy of scaffold-mediated localized chemotherapy in cancer: A systematic review of current research. J Oral Pathol Med 2020; 49:375-385. [PMID: 31975526 DOI: 10.1111/jop.12994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/29/2019] [Accepted: 01/23/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To assess the efficacy of scaffold-mediated localized chemotherapy in cancer. METHODS Databases including PubMed, Cochrane Library, and SCOPUS were searched for articles reporting the use of scaffold-mediated localized drug delivery in cancer. Essential data including scaffold fabrication material and methods, drug dosage and release duration and its effect on the cancer cells were extracted. RESULTS 15 articles out of 60 screened, fulfilled the eligibility criteria. Among the 15 studies, 5 studies included only cell lines and 2 studies were on mouse models, while 8 studies involved a combination of cell lines and mouse models. Scaffold materials included both synthetic polymers such as poly-lactide, polycaprolactone and natural materials including d-periosteum and human micro-fragmented adipose tissueA wide number of other variables included the fabrication procedure, drugs used, and the methods used to assess the effects on cancer. As a result, it was not possible to make any direct comparison of the efficacy of the therapeutic strategy used in each of these studies. CONCLUSION Irrespective of the many variables, a common consensus in all the included studies was that scaffold mediated localized drug delivery effectively reduced cancer cell viability by increasing drug bioavailability to the target tissue, while its localized effect reduced the risk of systemic toxicity.
Collapse
Affiliation(s)
- Archana A Gupta
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Pune, India
| | - Supriya Kheur
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Pune, India
| | - Gururaj Arakeri
- Department of Maxillofacial Surgery, Navodaya Dental College and Hospital, Raichur, India
| | - A Thirumal Raj
- Department of Oral Pathology and Microbiology, Sri Venkateswara Dental College and Hospital, Chennai, India
| | - Ravindra V Badhe
- Department of Pharmaceutical Sciences and Research, Dr. D.Y. Patil College of Pharmacy, Pune, India
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Jazan, Saudi Arabia
| | - Vishal Rao Us
- Department of Head and Neck Surgical Oncology & Robotic Surgery, HCG Cancer Hospital, Bangaluru, India
| | - Shekhar Patil
- Department of Medical Oncology, HCG Cancer Hospital, Bangaluru, India
| | - Ricardo S Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Peter Thomson
- Department of Oral & Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Prince Philip Dental Hospital, Sai Ying Pun, Hong Kong
| | - Peter A Brennan
- Department of Oral & Maxillofacial Surgery, Queen Alexandra Hospital, Portsmouth, UK
| |
Collapse
|
20
|
Yang J, Shi Z, Liu R, Wu Y, Zhang X. Combined-therapeutic strategies synergistically potentiate glioblastoma multiforme treatment via nanotechnology. Theranostics 2020; 10:3223-3239. [PMID: 32194864 PMCID: PMC7053190 DOI: 10.7150/thno.40298] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and devastating brain tumor characterized by poor prognosis and high rates of recurrence. Numerous therapeutic strategies and delivery systems are developed to prolong the survival time. They exhibit enhanced therapeutic effects in animal models, whereas few of them is applied in clinical trials. Taking into account the drug-resistance and high recurrence of GBM, combined-therapeutic strategies are exploited to maximize therapeutic efficacy. The combined therapies demonstrate superior results than those of single therapies against GBM. The co-therapeutic agents, the timing of therapeutic strategies and the delivery systems greatly affect the overall outcomes. Herein, the current advances in combined therapies for glioblastoma via systemic administration are exhibited in this review. And we will discuss the pros and cons of these combined-therapeutic strategies via nanotechnology, and provide the guidance for developing rational delivery systems to optimize treatments against GBM and other malignancies in central nervous system.
Collapse
|
21
|
Liu Y, Khan AR, Du X, Zhai Y, Tan H, Zhai G. Progress in the polymer-paclitaxel conjugate. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
22
|
Hudetz D, Borić I, Rod E, Jeleč Ž, Kunovac B, Polašek O, Vrdoljak T, Plečko M, Skelin A, Polančec D, Zenić L, Primorac D. Early results of intra-articular micro-fragmented lipoaspirate treatment in patients with late stages knee osteoarthritis: a prospective study. Croat Med J 2019. [PMID: 31187950 PMCID: PMC6563172 DOI: 10.3325/cmj.2019.60.227] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim To analyze clinical and functional effects of intra-articular injection of autologous micro-fragmented lipoaspirate (MLA) in patients with late stage knee osteoarthritis (KOA). Secondary aims included classifying cell types contributing to the treatment effect, performing detailed MRI-based classification of KOA, and elucidating the predictors for functional outcomes. Methods This prospective, non-randomized study was conducted from June 2016 to February 2018 and enrolled 20 patients with late stage symptomatic KOA (Kellgren Lawrence grade III, n = 4; and IV, n = 16) who received an intra-articular injection of autologous MLA in the index knee joint. At baseline radiological KOA grade and MRI were assessed in order to classify the morphology of KOA changes. Stromal vascular fraction cells obtained from MLA samples were stained with antibodies specific for cell surface markers. Patients were evaluated at baseline and 12-months after treatment with visual analog scale (VAS), Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and Knee Injury and Osteoarthritis Outcome Score (KOOS). Results Three patients (15%) received a total knee replacement and were not followed up completely. Seventeen patients (85%) showed a substantial pattern of KOOS and WOMAC improvement, significant in all accounts. KOOS score improved from 46 to 176% when compared with baseline, WOMAC decreased from 40 to 45%, while VAS rating decreased from 54% to 82% (all P values were <0.001). MLA contained endothelial progenitor cells, pericytes, and supra-adventitial adipose stromal cells as most abundant cell phenotypes. Conclusion This study is among the first to show a positive effect of MLA on patients with late stages KOA. ISRCTN registration ID: ISRCTN13337022.
Collapse
Affiliation(s)
- Damir Hudetz
- Damir Hudetz, St Catherine Specialty Hospital, Bracak 8, 49210 Zabok, Croatia,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bari E, Ferrarotti I, Torre ML, Corsico AG, Perteghella S. Mesenchymal stem/stromal cell secretome for lung regeneration: The long way through "pharmaceuticalization" for the best formulation. J Control Release 2019; 309:11-24. [PMID: 31326462 DOI: 10.1016/j.jconrel.2019.07.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022]
Abstract
Pulmonary acute and chronic diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and pulmonary hypertension, are considered to be major health issues worldwide. Cellular therapies with Mesenchymal Stem Cells (MSCs) offer a new therapeutic approach for chronic and acute lung diseases related to their anti-inflammatory, immunomodulatory, regenerative, pro-angiogenic and anti-fibrotic properties. Such therapeutic effects can be attributed to MSC-secretome, made of free soluble proteins and extracellular vesicles (EVs). This review summarizes the recent findings related to the efficacy and safety of MSC-derived products in pre-clinical models of lung diseases, pointing out the biologically active substances contained into MSC-secretome and their mechanisms involved in tissue regeneration. A perspective view is then provided about the missing steps required for the secretome "pharmaceuticalization" into a high quality, safe and effective medicinal product, as well as the formulation strategies required for EV non-invasive route of administration, such as inhalation.
Collapse
Affiliation(s)
- Elia Bari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia, Italy
| | - Ilaria Ferrarotti
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia, Italy; PharmaExceed srl, 27100 Pavia, Italy.
| | - Angelo Guido Corsico
- Center for Diagnosis of Inherited Alpha1-antitrypsin Deficiency, Dept of Internal Medicine and Therapeutics, Pneumology Unit IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy; PharmaExceed srl, 27100 Pavia, Italy
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, Pavia, Italy; PharmaExceed srl, 27100 Pavia, Italy
| |
Collapse
|