
Mesenchymal stem cell therapy for breast cancer‑related secondary lymphedema (Review)
- Authors:
- Published online on: June 23, 2025 https://doi.org/10.3892/mco.2025.2868
- Article Number: 73
-
Copyright: © Han et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
Secondary lymphedema is a chronic progressive disease characterized by the abnormal accumulation of interstitial fluid and tissue swelling (1). Its core pathological mechanism is due to the structural or functional damage of the lymphatic drainage system. Such damage is often caused by tumor treatments (such as axillary lymph node dissection and radiotherapy for breast cancer), parasitic infections (such as filariasis) or trauma (2). The clinical manifestations include asymmetric swelling of the limbs, fibrosis and recurrent infections. In severe cases, it can progress to lymphangiosarcoma or limb dysfunction, notably impairing the quality of life of patients (3). Among them, breast cancer-related lymphedema (BCRL) accounts for 20-65% of secondary cases (4). Its risk factors include the use of taxane, obesity and radiotherapy to regional lymph nodes at doses exceeding 50 Gy (5-7). A global cancer statistic shows that breast cancer ranks first in the incidence of female malignant tumors (accounting for 24.5% of newly diagnosed cancer cases) (8,9). Reported incidence rates range from 6.7-62.5% depending on surgical and radiotherapy protocols (10,11). However, modern techniques such as sentinel lymph node biopsy and immediate lymphatic reconstruction have reduced the risk to 5-10 and 7.0%, respectively (12,13). BCRL is still a problem that should be solved in the field of cancer rehabilitation.
Currently, the management of BCRL faces a twofold challenge: i) An absence of standardized assessment tools; and ii) limitations of treatment methods. Although the International Society of Lymphology has proposed a staging system based on clinical symptoms (such as the subclinical stage or lymphostatic elephantiasis), it is subjective and unable to quantify the degree of fibrosis (3). While bioimpedance spectroscopy and 3D imaging techniques [such as near-infrared fluorescence lymphatic imaging with 3D reconstruction (14,15), high-resolution volumetric analysis (15) and multiparametric MRI-based radiomics (16,17)] can enhance the sensitivity of early detection, standardized imaging protocols are lacking and are device-dependent, which make it difficult to popularize (3,9). In terms of treatment, conservative therapies (such as compression therapy and manual lymphatic drainage) can relieve symptoms, but their efficacy is limited for advanced fibrosis (18,19). Surgical interventions (such as lymphatic-venous anastomosis and liposuction) can improve the appearance, but they have risks such as damage to residual lymphatic vessels, aesthetic deformities and the formation of unstable scars (20-22). In this context, mesenchymal stem cell (MSC) therapy is a focus of research due to its unique dual mechanisms of tissue repair and immunomodulation (23). MSCs promote lymphangiogenesis by paracrine-secreting factors such as vascular endothelial growth factor C (VEGF-C) and stromal cell-derived factor 1α. Additionally, they inhibit the release of proinflammatory factors such as TNF-α and IL-6, which alleviates fibrosis (24,25).
In the present review, the pathophysiological mechanisms of BCRL, which are the foundation for understanding the disease, were investigated. Subsequently, MSC therapy, with its advantages and disadvantages as a treatment for secondary lymphedema, in the context of BCRL in particular, was evaluated. Finally, the present study summarized and appraised the relevant animal and clinical experiments on various stem cell therapies for secondary lymphedema, including those associated with BCRL. Through this process, the present review aimed to assess the progress made and the problems yet to be solved in this field.
2. Pathophysiological mechanism of BCRL
The physiological function of lymphatic vessels is to facilitate the exchange of materials such as proteins, lipids and water between tissue stroma and blood vessels. Its unique structure enables the transportation of macromolecular proteins, lipids and a notable volume of tissue fluid into lymph fluid, which are typically challenging to transfer through the venous system (26). A number of patients with cancer undergoing radiotherapy and chemotherapy may experience lymphatic vascular insufficiency and impaired drainage due to specific therapeutic agents [for example, doxorubicin decreases the lymphatic pumping frequency (27)] and physical factors [for example, radiation-induces axillary fibrosis at doses >50 Gy (28)]. Additionally, to maintain the normal function of lymphatic vessels, the pressure within the vessels should be kept ≤0 mm H2O, as elevated intraluminal pressure disrupts cytoskeletal dynamics and impairs fluid drainage. This is evidenced by studies on lymphatic biomechanics and pathological conditions such as lymphedema (29-31). Surgical interventions often disrupt axillary lymphatic outflow and cause lymphatic blockage (32), leading to a notable increase in lymphatic pressure. This increased pressure within the lymphatic system affects the transport of proteins and immune cells, resulting in the accumulation of protein-rich fluids in the extracellular matrix and triggering an inflammatory response (Fig. 1).
Previous studies demonstrate an increase in the number of CD4+ T cells and their differentiation into T helper (Th) type 2 (Th2) cells in lymphedema mouse models (33-35). Inflammation-induced CD4+ T cells stimulate the expression of VEGF-C, which drive lymphatic vessel proliferation in tissues (36). After lymphatic damage, M2 macrophages secrete VEGF-C and function as lymphoendothelial progenitor cells. Dendritic cells are activated at the injury site and migrate into lymphatic vessels along the concentration gradient of chemokine C-C motif ligand (CCL)21, which they themselves secrete. Activated dendritic cells then travel to lymph nodes, where they activate Th cells (37). Activated Th cells express surface markers such as cutaneous lymphocyte-associated antigen, C-C chemokine receptor (CCR)4, CCR9 and CCR1. Guided by adhesion molecules and CCLs, activated Th cells infiltrate the injured site and release inflammatory cytokines, including IFN-γ, TNF-α, IL-4 and IL-5, which collectively orchestrate immune responses and tissue remodeling (38-40). A prolonged inflammatory response can lead to tissue remodeling, which is characterized by increased collagen deposition, excessive growth of fat and connective tissue (37). Recruitment of various immune cells, including M2 macrophages, Th2 lymphocytes and neutrophils, promotes fat deposition and fibrosis in lymphedema (41-43). In the chronic phase, there is low-grade inflammation, fibrosis, fat deposition and non-functional lymphangiogenesis, which is characterized by valve defects and systolic dysfunction.
3. Evaluation and grading of BCRL
Lymphography-assisted stem cell therapy represents a novel approach for treating secondary lymphedema, offering benefits such as quantifying the severity of lymphedema and enhancing diagnostic accuracy. A study by Peña Quián et al (44) conducts multiple shallow and deep stem cell injections in the lymphatic vessel trace and groin region of 2 patients with chronic lower limb lymphedema, in order to evaluate the anatomical structure and function of the lymphatic system. The study demonstrates that after stem cell implantation, imaging reveals the presence of new lymphatic branches not observed before treatment, along with thicker radioactive columns. Additionally, patients exhibit an increase in dermal reflux and proximal lymph node activity, suggesting improved lymphatic drainage.
The study by Toyserkani et al (45) quantitatively assesses lymphatic drainage by measuring the mean transit time (MTT) in patients with BCRL before and 12 months after injection of adipose-derived regenerative cells. The study notes no notable change in MTT in the lymphedema group before and after treatment, which contradicts the findings of Peña Quián et al (44), the only previous report of lymphography after stem cell therapy. Subsequently, using indocyanine green lymphangiography, the study by Jørgensen et al (46) assesses lymph vessels in real time in 237 patients with BCRL. This approach offers prospective treatment and validates the reliability and safety of this technique for staging BCRL. It provides disease-related information that cannot be obtained by clinical measurements alone (46). Despite facing challenges such as poor spatial resolution and inadequate quantification, lymphatic imaging technology remains the gold standard for assessing the extent of lymphatic dysfunction and is pivotal in determining treatment strategies.
4. MSC therapy for BCRL
MSCs, as pivotal tools in regenerative medicine, vary in biological traits and therapeutic effects by tissue source. In secondary lymphedema management, bone marrow-derived MSCs (BMSCs) and adipose-derived MSCs (ADSCs) demonstrate distinct clinical profiles. Regarding proliferative capacity, BMSCs show notable donor age-dependent limitations in vitro expansion, whereas ADSCs obtained through minimally invasive lipoaspiration maintain superior passage stability (47). Differentiation analyses reveal that BMSCs possess enhanced osteochondral differentiation potential mediated by constitutive activation of the Wnt/β-catenin pathway within their native bone marrow niche (48). By contrast, ADSCs have adipogenic lineage predisposition while demonstrating unique therapeutic value in ischemic tissue repair through robust secretion of angiogenic factors, such as VEGF (49). Safety evaluations indicate a comparable absence of severe adverse events between both cell types; however, the immunogenic risk is notably reduced for ADSCs in allogeneic transplantation due to their human leukocyte antigen-DR isotype-negative phenotype (50).
Over the past decade, there have been a number of reports on MSC-based treatments for lymphedema in both animal studies and clinical trials (Fig. 2; Tables I and II). In the animal studies, lymphedema models are predominantly established using mouse posterior limbs and tails (51-53). Evaluating the treatment outcomes involves immunohistochemical staining methods such as perimeter measurements, depth of skin edema, imaging techniques, anti-CD31 staining for angiogenesis, anti-lymphatic vessel endothelial receptor-1 (LYVE-1) staining for lymphangiogenesis and staining for VEGF receptor expression levels. Additionally, no notable adverse events associated with MSC transplantation were observed in the clinical trials analyzed in the present review.
Application of BMSC therapy
In a randomized controlled trial involving 50 Chinese patients with lymphedema following breast cancer surgery (54), quantitative analysis reveals that BMSC transplantation achieves an improved long-term cure result compared with complex decongestant therapy (CDT). At a 12-month follow-up, the BMSC group demonstrates a 78.5% reduction in mean limb edema volume (vs. 54.5% in CDT) and an 82% decrease in visual analog scale pain scores (vs. 60% in CDT).
In a subsequent mechanistic study by Zhou et al (55) using rabbit models indicates that BMSC transplantation combined with VEGF-C in the treatment of limb lymphedema is more effective compared with stem cells alone. Additionally, the study quantifies the synergistic effect of the BMSC/VEGF-C combination therapy. After 28 days of treatment, the limb volume of the dual treatment group reduces by 58.4±7.1% vs. 38.4±5.7% of the BMSC treatment group alone, and this advantage is maintained at the 6-month follow-up. Furthermore, the study observes an increase in both blood vessel density and lymphatic vessel density in the combined treatment group. These structural enhancements may translate into lymphoscintigraphy, demonstrating accelerated lymphatic clearance, a functional parameter not evaluated in this investigation (55). In a clinical cohort of 40 patients with chronic lymphedema (56), BMSC mononuclear cell therapy achieves a clinically notable ankle circumference reduction after 6 months, which is supported by immunohistochemical evidence of enhanced lymphatic vasculature. Conversely, physical therapy controls reveal no measurable structural improvement and limited functional benefits.
Previously, a number of studies have attempted to apply the arteriovenous ring technique to lymphatic tissue engineering (57-59). Robering et al (60) combine human lymphoendothelial cells (LECs) and BMSCs into the fibrin matrix surrounding the atrioventricular ring, and demonstrate preliminary progress in the culture of human lymphatic blood vessels in rats, providing a possible way to generate transplantable lymphatic blood vessel networks.
Application of ADSCs
A 2013 transcriptomic analysis (61) reveals notable functional divergence in ADSCs from lymphedematous vs. healthy tissues. Lymphedema-associated ADSCs exhibit an 8-fold increase in the peroxisome proliferator-activated receptor-γ expression levels and a 2-fold decrease in VEGF secretion under angiogenic conditions. A study by Dhumale et al (62) also reveals this pathophysiological reprogramming, which confirms the proangiogenesis and parasecretory effects of human ADSCs. The study further indicates that such effects are mainly associated with CD31+ enrichment within this cell population.
In addition to the lymphangiogenic effects of VEGF-C, evidence delineates platelet-mediated regenerative pathways. In a murine tail lymphedema model, ADSCs/platelet-rich plasma (PRP) combination treatment increases the mean surface area of anti-LYVE-1-stained lymphatic vessels at 2 weeks by 4.1±1.0%, nearly a 2-fold increase compared with ADSC only therapy groups (2.5±0.3%). In addition, PRP treatment shows the best results regarding a reduction in wound size and improving wound epithelialization, with less of an increase in wound perfusion (25.1±31 PU vs. 33.8±33 PU in the ADSCs only group) (63). In a subsequent study, Hayashida et al (64) demonstrates that the number of LYVE-1 immunoreactive lymphatic vessels in a lymphedema mouse model undergoing ADSC transplantation notably increases when using indocyanine green lymphatic vessel imaging. The mechanism study by Ogino et al (65) further reveals that after 2 weeks of ADSCs transplantation, the proliferative lymphatic vessel ratio increases by 0.61±0.04%, recovers the orientation of type I collagen fibers from parallel to random and increases the number of type III collagen fibers. A study by Dai et al (66) further divides ADSCs into those expressing podoplanin and those not expressing podoplanin. In a mouse model of limb lymphedema, transplantation of podoplanin-positive ADSCs resulted in a notable generation of lymphatic vessels and remission of lymphedema, compared with both the podoplanin-negative ADSCs and the unsorted ADSC population (the original, heterogeneous cell mixture from which the podoplanin-positive and -negative subpopulations were isolated). The study reveals a large number of lymphangiogenic cytokines such as VEGF-C and D in the podoplanin-positive supernatant, which are absent in the podoplanin-negative and unclassified groups. Furthermore, immunocolocalization reveals that podoplanin-positive cells in lymphatics are LYVE-1 positive.
In a randomized controlled trial by Jørgensen et al (67), 34 patients that survived breast cancer with secondary lymphedema were enrolled, with 10 patients (71.4%) completing a 4-year follow-up after receiving ADSCs therapy combined with axillary scar release. In the study, ADSCs are isolated from autologous abdominal or thigh liposuction aspirates, with a mean cell count of 2.2x105 cells/ml per injection. Quantitative assessments reveal that 60% of treated patients achieve ≥50% reduction in self-reported lymphedema-related symptoms measured by the disabilities of the arm, shoulder and hand questionnaire, with notable improvements in upper extremity function scores. Additionally, 6 patients (60%) down staged the lymphedema treatment on their own initiative. The 4-year survival analysis demonstrates no evidence of locoregional recurrence, with only 2 cases of distant metastasis unrelated to treatment. These findings suggest that ADSC-based interventions provide clinically meaningful symptomatic relief and functional improvement in patients with post-mastectomy lymphedema, warranting further large-scale randomized trials with standardized outcome metrics.
Recent studies in extracellular vesicle (EV) therapeutics demonstrate the superior efficacy of ADSC-derived EVs compared with conventional therapeutic approaches [such as the transplantation of intact ADSCs and standard decongestive therapy (including manual lymphatic drainage, compression bandaging and exercise)] in promoting lymphatic regeneration and reducing edema volume in secondary lymphedema management (68-70). The potential oncogenic risk of EV treatment is reduced compared with direct stem cell transplantation, as ADSC-EVs lack cellular replication capacity and exhibit targeted delivery of therapeutic cargo without promoting tumor proliferation or metastasis in preclinical models (70-72). The previous study reveals that ADSCs-EV can promote the proliferation and migration of LECs and enhance lymphatic formation. In murine models, EV treatment achieves a 65.1±4.5% limb volume reduction at 4 weeks post treatment, with an increase in the total number of LYVE-1-positive lymphatic vessels (25.3±5.2% vs. 16.1±2.8% in controls) (70). Mechanistically, an analysis of mRNA expression levels reveals a 2-fold enrichment of lymphangiogenic markers such as VEGF-C, prospero-related homeobox 1 (Prox1), LYVE-1 and podoplanin, in an EV treatment group compared with a PBS treatment group (73-75).
Other cell-based treatments
In addition to BMSCs and ADSCs, other cell populations associated with stem cells have also been considered for the treatment of secondary lymphedema.
A study by Kawai et al (76) tried to treat secondary lymphedema with human LECs transplantation and achieved certain results. LECs extracted from the normal dermis of patients with breast cancer were transplanted into a rat model of caudal lymphedema. While this local injection did not ensure the long-term survival of LECs, in the short term, the skin of the rat tail in the podoplanin-positive purified LECs transplantation group became thinner, and evidence of lymphatic vessel regeneration from the rats was observed. A study by Deng et al (77) demonstrates that lentiviral-mediated Prox1 overexpression in human ADSCs enables stable differentiation into lymphoendothelial-like cells. By transfecting human ADSCs with Prox1-encoding vectors, sustained lymphatic differentiation was established, which was confirmed via flow cytometry, reverse transcription-quantitative-PCR, protein quantification and immunofluorescence analyses, notably increasing the protein levels of podoplanin, LYVE-1 and VEGFR3 (2 weeks post-differentiation). This genetic modification maintained lymphoendothelial characteristics while bypassing transient differentiation patterns observed in non-engineered counterparts.
In addition to the application of LECs, BMSCs also expand regulatory T cells (Tregs) in vitro and in vivo studies (72,78,79), and Treg induction may serve a positive role in stem cell transplantation in the treatment of lymphedema (80). Furthermore, intense inflammatory response and immune cell infiltration serve an important role in the pathogenesis of secondary lymphedema. A study by Gousopoulos et al (81) quantifies this axis in murine lymphedema models and reveals that a depletion of CD4+ T cells (shown via anti-CD4 antibodies) results in a notable reduction of the tissue area that is covered by lymphatic vessels. Furthermore, IL-2/anti-IL-2 complex-induced Treg expansion achieves edema reduction with a decrease in the CD45+, CD206+ and CD68+ infiltrates. Adoptive transfer of splenic Tregs decreases dermal TGF-β1, reduces collagen deposition (shown with Sirius Red staining) without altering VEGF-C levels and increases the fluorescence intensity of the lymphatic-specific tracer (82-84).
Additionally, other studies investigate the utilization of autologous peripheral blood stem cell transplantation as a treatment for primary lymphedema (85,86). A clinical study involving 10 patients reveal that this treatment can improve edema symptoms to an extent (85); however, further high-quality clinical studies are required to verify its effectiveness and scope of application.
5. Discussion
BCRL is predominantly induced by radical surgery, radiotherapy and cytotoxic chemotherapy, and manifests in 20% of patients with upper extremity, thoracic or breast involvement (87). The emerging therapeutic potential of MSCs in degenerative, autoimmune and genetic disorders has extended to lymphedema management. BMSCs demonstrate therapeutic efficacy through paracrine-mediated lymphangiogenic properties that facilitate damaged network reconstruction. However, their clinical implementation is constrained by invasive harvesting procedures and age-dependent proliferative decline (88-90). By contrast, ADSCs exhibit superior proangiogenic capabilities via elevated VEGF secretion, coupled with minimally invasive harvesting procedures, demonstrating enhanced ischemic tissue revascularization and lymphatic remodeling in preclinical models. However, the adipogenic tendency of ADSCs may increase the risk of post-transplant fat deposition (91). The present review suggests that the selection of the MSCs source is optimized according to BCRL heterogeneity and pathological stage (acute edema/chronic fibrosis), and that the synergistic effects of multi-source MSCs combination therapies is investigated.
While MSCs exhibit therapeutic potential in autoimmune diseases and chronic inflammation through their immunomodulatory properties and paracrine effects, their clinical application presents a complex duality of advantages and risks that requires examination. A prominent limitation is their unpredictable differentiation behavior (92). As a study by Yoon et al (93) demonstrates, transplanting MSCs may form ectopic tissues due to microenvironmental cues, where BMSCs without immunophenotypic purification (for example, those that lack CD105⁺, CD90⁺ or CD73⁺) cause myocardial calcification instead of functional repair. Furthermore, their dual role in oncology is also concerning. While MSCs can inhibit tumors via immune modulation (94), they may paradoxically promote tumor progression by suppressing natural killer/CD8+ T cell activity, polarizing Th2 responses and stimulating angiogenesis (95). Such contradictions highlight the need for further mechanistic studies to reconcile the regenerative benefits of MSCs with their pathological risks.
Current animal models for BCRL successfully mimic localized swelling but fail to recapitulate the inflammatory tumor microenvironment, which obscures the risks associated with MSC therapies. Cancer exerts an influence on the immune response through the release of various factors, such as cytokines and chemokines, and these factors have the capacity to modify the capability of the immune system to recognize and eliminate cancer cells (96). Limitations in animal lifespans and experimental durations restrict long-term safety evaluations, especially for delayed tumorigenic effects. By contrast, clinical trials observe no MSC-triggered cancer recurrence or metastasis in cases with BCRL. ADSCs, prioritized for their availability, ethical compliance and trophic factor release, show clinical potential. However, their inherent tumor tropism and unaddressed chronic safety issues emphasize the necessity to balance regenerative benefits against oncological hazards during therapeutic development.
Although MSCs demonstrate therapeutic efficacy in clinical trials through tissue regeneration and immunomodulation, their clinical translation remains hindered by unresolved biological risks such as uncontrolled differentiation and unclear long-term safety (97,98). While MSC-derived exosomes show reduced oncogenic concerns compared with whole-cell therapies, several trials have yielded favorable clinical outcomes, showcasing both safety and efficacy (99-101). Previous studies highlight MSCs transplantation safety in short-term applications, yet gaps in the understanding of chronic inflammatory responses, genomic instability and tumor microenvironment interactions remain. To advance MSC-based therapies, further studies are imperative, in which direct comparisons of their regenerative benefits against latent pathological risks should be prioritized.
6. Conclusions
MSC therapies exhibit promise in ameliorating BCRL by addressing edema, fostering lymphangiogenesis and mitigating fibrosis. However, due to the absence of a universally recognized or standardized treatment regimen for BCRL, additional clinical studies with larger sample sizes and extended follow-up periods are required to further investigate this prospective therapeutic modality.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
YZ, JC and FL designed, guided and modified the present study and manuscript. SH conducted the study, wrote the manuscript and collected and collated the data. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Grada AA and Phillips TJ: Lymphedema: Pathophysiology and clinical manifestations. J Am Acad Dermatol. 77:1009–1020. 2017.PubMed/NCBI View Article : Google Scholar | |
Hu LR and Pan J: Adipose-derived stem cell therapy shows promising results for secondary lymphedema. World J Stem Cells. 12:612–620. 2020.PubMed/NCBI View Article : Google Scholar | |
Vargo M, Aldrich M, Donahue P, Iker E, Koelmeyer L, Crescenzi R and Cheville A: Current diagnostic and quantitative techniques in the field of lymphedema management: A critical review. Med Oncol. 41(241)2024.PubMed/NCBI View Article : Google Scholar | |
McLaughlin SA, Brunelle CL and Taghian A: Breast cancer-related lymphedema: Risk factors, screening, management, and the impact of locoregional treatment. J Clin Oncol. 38:2341–2350. 2020.PubMed/NCBI View Article : Google Scholar | |
Aguilera-Eguía RA, Seron P, Gutiérrez-Arias R and Zaror C: Which physical therapy intervention is most effective in reducing secondary lymphoedema associated with breast cancer? Protocol for a systematic review and network meta-analysis. BMJ Open. 12(e065045)2022.PubMed/NCBI View Article : Google Scholar | |
Zhang H, Wang L, Chen Y, Wu Q, Chen G, Shen X, Wang Q, Yan Y, Yu Y, Zhong Y, et al: Outcomes of novel coronavirus disease 2019 (COVID-19) infection in 107 patients with cancer from Wuhan, China. Cancer. 126:4023–4031. 2020.PubMed/NCBI View Article : Google Scholar | |
Jariwala P and Kaur N: A descriptive study on prevalence of arm/shoulder problems and its impact on quality of life in breast cancer survivors. Indian J Cancer. 58:201–206. 2021.PubMed/NCBI View Article : Google Scholar | |
Xia C, Dong X, Li H, Cao M, Sun D, He S, Yang F, Yan X, Zhang S, Li N and Chen W: Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J (Engl). 135:584–590. 2022.PubMed/NCBI View Article : Google Scholar | |
Hasenoehrl T, Palma S, Ramazanova D, Kölbl H, Dorner TE, Keilani M and Crevenna R: Resistance exercise and breast cancer-related lymphedema-a systematic review update and meta-analysis. Support Care Cancer. 28:3593–3603. 2020.PubMed/NCBI View Article : Google Scholar | |
DiSipio T, Rye S, Newman B and Hayes S: Incidence of unilateral arm lymphoedema after breast cancer: A systematic review and meta-analysis. Lancet Oncol. 14:500–515. 2013.PubMed/NCBI View Article : Google Scholar | |
Dessources K, Aviki E and Leitao MM Jr: Lower extremity lymphedema in patients with gynecologic malignancies. Int J Gynecol Cancer. 30:252–260. 2020.PubMed/NCBI View Article : Google Scholar | |
Bruno C, Cesta CE, Hjellvik V, Ulrichsen SP, Bjørk MH, Esen B, Gillies MB, Gissler M, Havard A, Karlstad Ø, et al: Corrigendum to Antipsychotic use during pregnancy and risk of specific neurodevelopmental disorders and learning difficulties in children: A multinational cohort study [eClinicalMedicine 70 (2024) 102531/DOI: 10.1016/j.eclinm.2024.102531]. EClinicalMedicine 81: 103139, 2025. | |
Hassan AM, Fisher CS and Hassanein AH: ASO author reflections: navigating the nuances of lymphedema prevention with immediate lymphatic reconstruction. Ann Surg Oncol: Apr 20, 2025 (Epub ahead of print). | |
Bouhali S, Merchant F, Karni RJ, Gutierrez C and Rasmussen JC: 3D rendering and analysis of dermal backflow as an early indicator of cancer-acquired lymphedema using RGB-D and near-infrared fluorescence lymphatic imaging. Proc SPIE 12930, Medical Imaging 2024: Clinical and Biomedical Imaging, 1293008, 2024. | |
Frueh FS, Körbel C, Gassert L, Müller A, Gousopoulos E, Lindenblatt N, Giovanoli P, Laschke MW and Menger MD: High-resolution 3D volumetry versus conventional measuring techniques for the assessment of experimental lymphedema in the mouse hindlimb. Sci Rep. 6(34673)2016.PubMed/NCBI View Article : Google Scholar | |
Wang N, Liao C, Cao X, Nishimura M, Brackenier YWE, Yurt M, Gao M, Abraham D, Alkan C, Iyer SS, et al: Spherical echo-planar time-resolved imaging (sEPTI) for rapid 3D quantitative T2* and susceptibility imaging. Magn Reson Med. 93:121–137. 2025.PubMed/NCBI View Article : Google Scholar | |
Xie K, Jiang H, Chen X, Ning Y, Yu Q, Lv F, Liu R, Zhou Y, Xu L, Yue Q and Peng J: Multiparameter MRI-based model integrating radiomics and deep learning for preoperative staging of laryngeal squamous cell carcinoma. Sci Rep. 15(16239)2025.PubMed/NCBI View Article : Google Scholar | |
Rogan S, Taeymans J, Luginbuehl H, Aebi M, Mahnig S and Gebruers N: Therapy modalities to reduce lymphoedema in female breast cancer patients: A systematic review and meta-analysis. Breast Cancer Res Treat. 159:1–14. 2016.PubMed/NCBI View Article : Google Scholar | |
Gao Y, Ma T, Han M, Yu M and Wang X, Lv Y and Wang X: Effects of acupuncture and moxibustion on breast cancer-related lymphedema: A systematic review and meta-analysis of randomized controlled trials. Integr Cancer Ther. 20(15347354211044107)2021.PubMed/NCBI View Article : Google Scholar | |
Keeley V: Advances in understanding and management of lymphoedema (cancer, primary). Curr Opin Support Palliat Care. 11:355–360. 2017.PubMed/NCBI View Article : Google Scholar | |
Raju A and Chang DW: Vascularized lymph node transfer for treatment of lymphedema: A comprehensive literature review. Ann Surg. 261:1013–1023. 2015.PubMed/NCBI View Article : Google Scholar | |
No authors listed. Successful mesenchymal stem cell treatment of leg ulcers complicated by Behcet disease: A case report and literature review: Erratum. Medicine (Baltimore). 97(e0670)2018.PubMed/NCBI View Article : Google Scholar | |
Toyserkani NM, Christensen ML, Sheikh SP and Sørensen JA: Stem cells show promising results for lymphoedema treatment-a literature review. J Plast Surg Hand Surg. 49:65–71. 2015.PubMed/NCBI View Article : Google Scholar | |
Das M, Mayilsamy K, Mohapatra SS and Mohapatra S: Mesenchymal stem cell therapy for the treatment of traumatic brain injury: Progress and prospects. Rev Neurosci. 30:839–855. 2019.PubMed/NCBI View Article : Google Scholar | |
Pittenger MF, Discher DE, Péault BM, Phinney DG, Hare JM and Caplan AI: Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen Med. 4(22)2019.PubMed/NCBI View Article : Google Scholar | |
Jensen MR, Simonsen L, Karlsmark T and Bülow J: Microvascular filtration is increased in the forearms of patients with breast cancer-related lymphedema. J Appl Physiol (1985). 114:19–27. 2013.PubMed/NCBI View Article : Google Scholar | |
Castilla DM, Liu ZJ, Tian R, Li Y, Livingstone AS and Velazquez OC: A novel autologous cell-based therapy to promote diabetic wound healing. Ann Surg. 256:560–572. 2012.PubMed/NCBI View Article : Google Scholar | |
Wariss BR, de Souza Abrahão K, de Aguiar SS, Bergmann A and Thuler LCS: Effectiveness of four inflammatory markers in predicting prognosis in 2374 women with breast cancer. Maturitas. 101:51–56. 2017.PubMed/NCBI View Article : Google Scholar | |
Cong C, Rao C, Ma Z, Yu M, He Y, He Y, Hao Z, Li C, Lou H and Gao D: ‘Nano-lymphatic’ photocatalytic water-splitting for relieving tumor interstitial fluid pressure and achieving hydrodynamic therapy†. Mater Horiz. 7:3266–3274. 2020. | |
Zhuang T, Lei Y, Chang JJ, Zhou YP, Li Y, Li YX, Yang YF, Chen MH, Meng T, Fu SM, et al: A2AR-mediated lymphangiogenesis via VEGFR2 signaling prevents salt-sensitive hypertension. Eur Heart J. 44:2730–2742. 2023.PubMed/NCBI View Article : Google Scholar | |
Schoofs H, Daubel N, Schnabellehner S, Grönloh MLB, Palacios Martínez S, Halme A, Marks AM, Jeansson M, Barcos S, Brakebusch C, et al: Dynamic cytoskeletal regulation of cell shape supports resilience of lymphatic endothelium. Nature. 641:465–475. 2025.PubMed/NCBI View Article : Google Scholar | |
Chen CE, Chiang NJ, Perng CK, Ma H and Lin CH: Review of preclinical and clinical studies of using cell-based therapy for secondary lymphedema. J Surg Oncol. 121:109–120. 2020.PubMed/NCBI View Article : Google Scholar | |
Avraham T, Zampell JC, Yan A, Elhadad S, Weitman ES, Rockson SG, Bromberg J and Mehrara BJ: Th2 differentiation is necessary for soft tissue fibrosis and lymphatic dysfunction resulting from lymphedema. FASEB J. 27:1114–1126. 2013.PubMed/NCBI View Article : Google Scholar | |
Nishioka T, Katayama KI, Kumegawa S, Isono K, Baba T, Tsujimoto H, Yamada G, Inoue N and Asamura S: Increased infiltration of CD4+ T cell in the complement deficient lymphedema model. BMC Immunol. 24(42)2023.PubMed/NCBI View Article : Google Scholar | |
Uemura K, Katayama KI, Nishioka T, Watanabe H, Yamada G, Inoue N and Asamura S: Dynamics of immune cell infiltration and fibroblast-derived IL-33/ST2 axis induction in a mouse model of post-surgical lymphedema. Int J Mol Sci. 26(1371)2025.PubMed/NCBI View Article : Google Scholar | |
Ogata F, Fujiu K, Matsumoto S, Nakayama Y, Shibata M, Oike Y, Koshima I, Watabe T, Nagai R and Manabe I: Excess lymphangiogenesis cooperatively induced by macrophages and CD4(+) T cells drives the pathogenesis of lymphedema. J Invest Dermatol. 136:706–714. 2016.PubMed/NCBI View Article : Google Scholar | |
Ogino R, Yokooji T, Hayashida M, Suda S, Yamakawa S and Hayashida K: Emerging anti-inflammatory pharmacotherapy and cell-based therapy for lymphedema. Int J Mol Sci. 23(7614)2022.PubMed/NCBI View Article : Google Scholar | |
Higgins ET, Busse WW, Esnault S, Christian BT, Klaus DR, Bach JC, Frye CJ and Rosenkranz MA: Fueling the fire in the lung-brain axis: The salience network connects allergen-provoked TH17 responses to psychological stress in asthma. Brain Behav Immun. 128:276–288. 2025.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print). | |
De Castro V, Abdellaoui O, Dehecq B, Ndao B, Mercier-Letondal P, Dauvé A, Garnache-Ottou F, Adotévi O, Loyon R and Godet Y: Characterization of the aryl hydrocarbon receptor as a potential candidate to improve cancer T cell therapies. Cancer Immunol Immunother. 74(200)2025.PubMed/NCBI View Article : Google Scholar | |
Li H, Wu Y, Song XH, Li CX, Cai Y and Chen C: Expression of Th1 and Th2 cytokines in serum of patients with lupus nephritis. Mod Prev Med. 40 746:2013. | |
Zhu Q, Yang H, Altaf F, Wu N, Hu Y, Su L, Li J, Liu J, Wang G, Igbiriki DG, et al: SOCS8 deficiency models MAFLD-like progression in the zebrafish gut-liver axis. Water Biology and Security. Elsevier, pp100414, 2025. | |
Lee SO and Kim IK: Molecular pathophysiology of secondary lymphedema. Front Cell Dev Biol. 12(1363811)2024.PubMed/NCBI View Article : Google Scholar | |
Duhon BH, Phan TT, Taylor SL, Crescenzi RL and Rutkowski JM: Current mechanistic understandings of lymphedema and lipedema: Tales of fluid, fat, and fibrosis. Int J Mol Sci. 23(6621)2022.PubMed/NCBI View Article : Google Scholar | |
Peña Quián Y, Hernández Ramirez P, Batista Cuellar JF, Perera Pintado A and Coca Pérez MA: Lymphoscintigraphy for the assessment of autologous stem cell implantation in chronic lymphedema. Clin Nucl Med. 40:217–219. 2015.PubMed/NCBI View Article : Google Scholar | |
Toyserkani NM, Jensen CH, Tabatabaeifar S, Jørgensen MG, Hvidsten S, Simonsen JA, Andersen DC, Sheikh SP and Sørensen JA: Adipose-derived regenerative cells and fat grafting for treating breast cancer-related lymphedema: Lymphoscintigraphic evaluation with 1 year of follow-up. J Plast Reconstr Aesthet Surg. 72:71–77. 2019.PubMed/NCBI View Article : Google Scholar | |
Jørgensen MG, Toyserkani NM, Hansen FCG, Thomsen JB and Sørensen JA: Prospective validation of indocyanine green lymphangiography staging of breast cancer-related lymphedema. Cancers (Basel). 13(1540)2021.PubMed/NCBI View Article : Google Scholar | |
Kern S, Eichler H, Stoeve J, Klüter H and Bieback K: Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 24:1294–1301. 2006.PubMed/NCBI View Article : Google Scholar | |
Yu SJ, Kim HJ, Lee ES, Park CG, Cho SJ and Jeon SH: β-catenin accumulation is associated with increased expression of nanog protein and predicts maintenance of MSC self-renewal. Cell Transplant. 26:365–377. 2017.PubMed/NCBI View Article : Google Scholar | |
Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P and Hedrick MH: Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 13:4279–4295. 2002.PubMed/NCBI View Article : Google Scholar | |
Le Blanc K and Ringdén O: Immunomodulation by mesenchymal stem cells and clinical experience. J Intern Med. 262:509–525. 2007.PubMed/NCBI View Article : Google Scholar | |
Hassanein AH, Sinha M, Neumann CR, Mohan G, Khan I and Sen CK: A murine tail lymphedema model. J Vis Exp. (10.3791/61848)2021.PubMed/NCBI View Article : Google Scholar | |
Arruda G, Ariga S, de Lima TM, Souza HP and Andrade M: A modified mouse-tail lymphedema model. Lymphology. 53:29–37. 2020.PubMed/NCBI | |
Yu J and Guo W: Modern medical progress of peripheral lymphedema treated by integrated traditional Chinese and western medicine. Adv Clin Med. 12:4228–4234. 2022. | |
Hou C, Wu X and Jin X: Autologous bone marrow stromal cells transplantation for the treatment of secondary arm lymphedema: A prospective controlled study in patients with breast cancer related lymphedema. Jpn J Clin Oncol. 38:670–674. 2008.PubMed/NCBI View Article : Google Scholar | |
Zhou H, Wang M, Hou C, Jin X and Wu X: Exogenous VEGF-C augments the efficacy of therapeutic lymphangiogenesis induced by allogenic bone marrow stromal cells in a rabbit model of limb secondary lymphedema. Jpn J Clin Oncol. 41:841–846. 2011.PubMed/NCBI View Article : Google Scholar | |
Ismail AM, Abdou SM, Abdelnaby AY, Hamdy MA, El Saka AA and Gawaly A: Stem cell therapy using bone marrow-derived mononuclear cells in treatment of lower limb lymphedema: A randomized controlled clinical trial. Lymphat Res Biol. 16:270–277. 2018.PubMed/NCBI View Article : Google Scholar | |
Weigand A, Beier JP, Arkudas A, Al-Abboodi M, Polykandriotis E, Horch RE and Boos AM: The arteriovenous (AV) loop in a small animal model to study angiogenesis and vascularized tissue engineering. J Vis Exp. (54676)2016.PubMed/NCBI View Article : Google Scholar | |
Boos AM, Loew JS, Weigand A, Deschler G, Klumpp D, Arkudas A, Bleiziffer O, Gulle H, Kneser U, Horch RE and Beier JP: Engineering axially vascularized bone in the sheep arteriovenous-loop model. J Tissue Eng Regen Med. 7:654–664. 2013.PubMed/NCBI View Article : Google Scholar | |
Weigand A, Horch RE, Boos AM, Beier JP and Arkudas A: The arteriovenous loop: Engineering of axially vascularized tissue. Eur Surg Res. 59:286–299. 2018.PubMed/NCBI View Article : Google Scholar | |
Robering JW, Al-Abboodi M, Titzmann A, Horn I, Beier JP, Horch RE, Kengelbach-Weigand A and Boos AM: Tissue engineering of lymphatic vasculature in the arteriovenous loop model of the rat. Tissue Eng Part A. 27:129–141. 2021.PubMed/NCBI View Article : Google Scholar | |
Levi B, Glotzbach JP, Sorkin M, Hyun J, Januszyk M, Wan DC, Li S, Nelson ER, Longaker MT and Gurtner GC: Molecular analysis and differentiation capacity of adipose-derived stem cells from lymphedema tissue. Plast Reconstr Surg. 132:580–589. 2013.PubMed/NCBI View Article : Google Scholar | |
Dhumale P, Nielsen JV, Hansen ACS, Burton M, Beck HC, Jørgensen MG, Toyserkani NM, Haahr MK, Hansen ST, Lund L, et al: CD31 defines a subpopulation of human adipose-derived regenerative cells with potent angiogenic effects. Sci Rep. 13(14401)2023.PubMed/NCBI View Article : Google Scholar | |
Ackermann M, Wettstein R, Senaldi C, Kalbermatten DF, Konerding MA, Raffoul W and Erba P: Impact of platelet rich plasma and adipose stem cells on lymphangiogenesis in a murine tail lymphedema model. Microvasc Res. 102:78–85. 2015.PubMed/NCBI View Article : Google Scholar | |
Hayashida K, Yoshida S, Yoshimoto H, Fujioka M, Saijo H, Migita K, Kumaya M and Akita S: Adipose-derived stem cells and vascularized lymph node transfers successfully treat mouse hindlimb secondary lymphedema by early reconnection of the lymphatic system and lymphangiogenesis. Plast Reconstr Surg. 139:639–651. 2017.PubMed/NCBI View Article : Google Scholar | |
Ogino R, Hayashida K, Yamakawa S and Morita E: Adipose-derived stem cells promote intussusceptive lymphangiogenesis by restricting dermal fibrosis in irradiated tissue of mice. Int J Mol Sci. 21(3885)2020.PubMed/NCBI View Article : Google Scholar | |
Dai T, Jiang Z, Cui C, Sun Y, Lu B, Li H, Cao W, Chen B, Li S and Guo L: The roles of podoplanin-positive/podoplanin-negative cells from adipose-derived stem cells in lymphatic regeneration. Plast Reconstr Surg. 145:420–431. 2020.PubMed/NCBI View Article : Google Scholar | |
Jørgensen MG, Toyserkani NM, Jensen CH, Andersen DC, Sheikh SP and Sørensen JA: Adipose-derived regenerative cells and lipotransfer in alleviating breast cancer-related lymphedema: An open-label phase I trial with 4 years of follow-up. Stem Cells Transl Med. 10:844–854. 2021.PubMed/NCBI View Article : Google Scholar | |
Yang S, Sun Y and Yan C: Recent advances in the use of extracellular vesicles from adipose-derived stem cells for regenerative medical therapeutics. J Nanobiotechnology. 22(316)2024.PubMed/NCBI View Article : Google Scholar | |
Kasseroller RG and Brenner E: Effectiveness of manual lymphatic drainage in intensive phase I therapy of breast cancer-related lymphedema-a retrospective analysis. Support Care Cancer. 32(5)2023.PubMed/NCBI View Article : Google Scholar | |
Tashiro K, Yoshioka Y and Ochiya T: Extracellular vesicles from adipose-derived stem cells relieve extremity lymphedema in mouse models. Plast Reconstr Surg. 152:1011–1021. 2023.PubMed/NCBI View Article : Google Scholar | |
Cheng X, Henick BS and Cheng K: Anticancer therapy targeting cancer-derived extracellular vesicles. ACS Nano. 18:6748–6765. 2024.PubMed/NCBI View Article : Google Scholar | |
Yuan Z, Zhu Z, Zhu F, Ding F, Wang Y, Wang X, Luo X, Yang J, Liu F and Sun D: Impact of human adipose tissue-derived stem cells on dermatofibrosarcoma protuberans cells in an indirect co-culture: An in vitro study. Stem Cell Res Ther. 12(440)2021.PubMed/NCBI View Article : Google Scholar | |
Diao X, Guo C, Zheng H, Zhao K, Luo Y, An M, Lin Y, Chen J, Li Y, Li Y, et al: SUMOylation-triggered ALIX activation modulates extracellular vesicles circTLCD4-RWDD3 to promote lymphatic metastasis of non-small cell lung cancer. Signal Transduct Target Ther. 8(426)2023.PubMed/NCBI View Article : Google Scholar | |
Li Y, Zheng H, Luo Y, Lin Y, An M, Kong Y, Zhao Y, Yin Y, Ai L, Huang J and Chen C: An HGF-dependent positive feedback loop between bladder cancer cells and fibroblasts mediates lymphangiogenesis and lymphatic metastasis. Cancer Commun (Lond). 43:1289–1311. 2023.PubMed/NCBI View Article : Google Scholar | |
Zhang HF, Wang YL, Tan YZ, Wang HJ, Tao P and Zhou P: Enhancement of cardiac lymphangiogenesis by transplantation of CD34+VEGFR-3+ endothelial progenitor cells and sustained release of VEGF-C. Basic Res Cardiol. 114(43)2019.PubMed/NCBI View Article : Google Scholar | |
Kawai Y, Shiomi H, Abe H, Naka S, Kurumi Y and Tani T: Cell transplantation therapy for a rat model of secondary lymphedema. J Surg Res. 189:184–191. 2014.PubMed/NCBI View Article : Google Scholar | |
Deng J, Dai T, Sun Y, Zhang Q, Jiang Z, Li S and Cao W: Overexpression of Prox1 induces the differentiation of human adipose-derived stem cells into lymphatic endothelial-like cells in vitro. Cell Reprogram. 19:54–63. 2017.PubMed/NCBI View Article : Google Scholar | |
Ou HX, Guo BB, Liu Q, Li YK, Yang Z, Feng WJ and Mo ZC: Regulatory T cells as a new therapeutic target for atherosclerosis. Acta Pharmacol Sin. 39:1249–1258. 2018.PubMed/NCBI View Article : Google Scholar | |
Chen DB: Experimental study of bone marrow mesenchymal stem cells (BMSCs) promoting hematopoietic reconstruction and immune regulation POST-HSCT. Fujian Medical University, 2022. | |
Salek Farrokhi A, Zarnani AH, Rezaei Kahmini F and Moazzeni SM: Mesenchymal stem cells induce expansion of regulatory T cells in abortion-prone mice. Reproduction. 161:477–487. 2021.PubMed/NCBI View Article : Google Scholar | |
Gousopoulos E, Proulx ST, Bachmann SB, Scholl J, Dionyssiou D, Demiri E, Halin C, Dieterich LC and Detmar M: Regulatory T cell transfer ameliorates lymphedema and promotes lymphatic vessel function. JCI Insight. 1(e89081)2016.PubMed/NCBI View Article : Google Scholar | |
Choi G, Na H, Kuen DS, Kim BS and Chung Y: Autocrine TGF-β1 maintains the stability of Foxp3+ regulatory T cells via IL-12Rβ2 downregulation. Biomolecules. 10(819)2020.PubMed/NCBI View Article : Google Scholar | |
Christofi P, Pantazi C, Psatha N, Sakellari I, Yannaki E and Papadopoulou A: Promises and pitfalls of next-generation treg adoptive immunotherapy. Cancers (Basel). 15(5877)2023.PubMed/NCBI View Article : Google Scholar | |
Mao LL, Yuan H, Wang WW, Wang YJ, Yang MF, Sun BL, Zhang ZY and Yang XY: Adoptive regulatory T-cell therapy attenuates perihematomal inflammation in a mouse model of experimental intracerebral hemorrhage. Cell Mol Neurobiol. 37:919–929. 2017.PubMed/NCBI View Article : Google Scholar | |
Ehyaeeghodraty V, Molavi B, Nikbakht M, Malek Mohammadi A, Mohammadi S, Ehyaeeghodraty N, Fallahi B, Mousavi SA, Vaezi M and Sefidbakht S: Effects of mobilized peripheral blood stem cells on treatment of primary lower extremity lymphedema. J Vasc Surg Venous Lymphat Disord. 8:445–451. 2020.PubMed/NCBI View Article : Google Scholar | |
Białobrzeska M, Stępniewski J, Martyniak A, Szuba A and Dulak J: Generation of human induced pluripotent stem cell line from peripheral blood of patient with lymphedema-distichiasis syndrome. Stem Cell Res. 85(103693)2025.PubMed/NCBI View Article : Google Scholar | |
Ren Y, Kebede MA, Ogunleye AA, Emerson MA, Evenson KR, Carey LA, Hayes SC and Troester MA: Burden of lymphedema in long-term breast cancer survivors by race and age. Cancer. 128:4119–4128. 2022.PubMed/NCBI View Article : Google Scholar | |
Huang Y, Luo L, Xu Y, Li J, Wu Z, Zhao C, Wen J, Jiang P, Zhu H, Wang L, et al: UHRF1-mediated epigenetic reprogramming regulates glycolysis to promote progression of B-cell acute lymphoblastic leukemia. Cell Death Dis. 16(351)2025.PubMed/NCBI View Article : Google Scholar | |
Deng Y, Lin A, Lai C, He W, Li J, Zhang N, Huang S, Tong L, Lai Y, Huo Y and Xu J: Combined inhibition of importin-β and PBR enhances osteogenic differentiation of BMSCs by reducing nuclear accumulation of glucocorticoid receptor and promoting its mitochondrial translocation. J Steroid Biochem Mol Biol. 250(106731)2025.PubMed/NCBI View Article : Google Scholar | |
Liu Y, Xu W, Liu G, Ma L and Li Z: Therapeutic efficacy of autologous bone marrow mesenchymal stem cell transplantation in patients with spinal cord injury: A meta-analysis. EFORT Open Rev. 10:309–315. 2025.PubMed/NCBI View Article : Google Scholar | |
Xiang Q, Xu F, Li Y, Liu X, Chen Q, Huang J, Yu N, Zeng Z, Yuan M, Zhang Q, et al: Transcriptome analysis and functional identification of adipose-derived mesenchymal stem cells in secondary lymphedema. Gland Surg. 9:558–574. 2020.PubMed/NCBI View Article : Google Scholar | |
Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, Armstrong L, Djonov V, Lako M and Stojkovic M: Ethical and safety issues of stem cell-based therapy. Int J Med Sci. 15:36–45. 2018.PubMed/NCBI View Article : Google Scholar | |
Yoon YS, Park JS, Tkebuchava T, Luedeman C and Losordo DW: Unexpected severe calcification after transplantation of bone marrow cells in acute myocardial infarction. Circulation. 109:3154–3157. 2004.PubMed/NCBI View Article : Google Scholar | |
Lan T, Luo M and Wei X: Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol. 14(195)2021.PubMed/NCBI View Article : Google Scholar | |
Ljujic B, Milovanovic M, Volarevic V, Murray B, Bugarski D, Przyborski S, Arsenijevic N, Lukic ML and Stojkovic M: Human mesenchymal stem cells creating an immunosuppressive environment and promote breast cancer in mice. Sci Rep. 3(2298)2013.PubMed/NCBI View Article : Google Scholar | |
Marcella S, Braile M, Grimaldi AM, Soricelli A and Smaldone G: Exploring thymic stromal lymphopoietin in the breast cancer microenvironment: A preliminary study. Oncol Lett. 29(182)2025.PubMed/NCBI View Article : Google Scholar | |
Zang L, Li Y, Hao H, Liu J, Cheng Y, Li B, Yin Y, Zhang Q, Gao F, Wang H, et al: Efficacy and safety of umbilical cord-derived mesenchymal stem cells in Chinese adults with type 2 diabetes: A single-center, double-blinded, randomized, placebo-controlled phase II trial. Stem Cell Res Ther. 13(180)2022.PubMed/NCBI View Article : Google Scholar | |
Astori G, Amati E, Bambi F, Bernardi M, Chieregato K, Schäfer R, Sella S and Rodeghiero F: Platelet lysate as a substitute for animal serum for the ex-vivo expansion of mesenchymal stem/stromal cells: Present and future. Stem Cell Res Ther. 7(93)2016.PubMed/NCBI View Article : Google Scholar | |
Thongsit A, Oontawee S, Siriarchavatana P, Rodprasert W, Somparn P, Na Nan D, Osathanon T, Egusa H and Sawangmake C: Scalable production of anti-inflammatory exosomes from three-dimensional cultures of canine adipose-derived mesenchymal stem cells: Production, stability, bioactivity, and safety assessment. BMC Vet Res. 21(81)2025.PubMed/NCBI View Article : Google Scholar | |
Xie X, Song Q, Dai C, Cui S, Tang R, Li S, Chang J, Li P, Wang J, Li J, et al: Clinical safety and efficacy of allogenic human adipose mesenchymal stromal cells-derived exosomes in patients with mild to moderate Alzheimer's disease: A phase I/II clinical trial. Gen Psychiatr. 36(e101143)2023.PubMed/NCBI View Article : Google Scholar | |
Chu M, Wang H, Bian L, Huang J, Wu D, Zhang R, Fei F, Chen Y and Xia J: Nebulization therapy with umbilical cord mesenchymal stem cell-derived exosomes for COVID-19 pneumonia. Stem Cell Rev Rep. 18:2152–2163. 2022.PubMed/NCBI View Article : Google Scholar |