Abstract
Purpose of Review
This review aims to summarize the current knowledge regarding the pharmacological interventions studied in both experimental and clinical trials for secondary lymphedema.
Recent Findings
Lymphedema is a progressive disease that results in tissue swelling, pain, and functional disability. The most common cause of secondary lymphedema in developed countries is an iatrogenic injury to the lymphatic system during cancer treatment. Despite its high incidence and severe sequelae, lymphedema is usually treated with palliative options such as compression and physical therapy. However, recent studies on the pathophysiology of lymphedema have explored pharmacological treatments in preclinical and early phase clinical trials.
Summary
Many potential treatment options for lymphedema have been explored throughout the past two decades including systemic agents and topical approaches to decrease the potential toxicity of systemic treatment. Treatment strategies including lymphangiogenic factors, anti-inflammatory agents, and anti-fibrotic therapies may be used independently or in conjunction with surgical approaches.
Keywords: Lymphedema, TH2 cells, CD4 + , VEGF-C, TGFB, Non-steroidal anti-inflammatory drugs, NSAID, Doxycycline, Tacrolimus, ACE inhibitors, Captopril, Pirfenidone, Tetracyclines, Vascular endothelial growth factor C
Introduction
Lymphedema is a progressive disease that causes tissue swelling, fluid accumulation, and chronic fibroadipose tissue deposition [1]. Lymphedema can be categorized based on the etiology of the disease. Patients with primary lymphedema have genetic abnormalities of the lymphatic system that manifest as tissue swelling, chylothorax, or chylous ascites in utero, shortly after birth, or at some point in the patient’s life. Secondary lymphedemas are caused by external insults such as infections, radiation, obesity, or iatrogenic injury to the lymphatic system during the course of oncologic surgery [2, 3]. Breast cancer, due to the high prevalence of this disease, is the most common cause of secondary lymphedema in developed countries. Approximately 1 in 3 patients who undergo axillary lymph node dissection (ALND) and 3–5% of the patients treated with sentinel lymph node biopsy (SLND) develop breast cancer–related lymphedema (BCRL) [4–7]. Lymphedema also develops in approximately 1 in 6 patients treated for other solid tumors such as melanoma, gynecological or urological tumors, or sarcomas [8]. Although the estimated number of patients who suffer from lymphedema in the USA is unknown, analysis of the incidence of the disease and cancer survival rates suggest that approximately 1 million American cancer survivors suffer from secondary lymphedema.
Patients with lymphedema have decreased quality of life (QoL) and often complain of pain and functional problems [1, 5, 9–14]. Nearly 40% of patients with lymphedema also develop recurrent infections which, in some cases, can be quite severe resulting in sepsis and hospitalization for intravenous antibiotics. Treatment of lymphedema is time intensive and not always covered by insurance resulting in significant financial toxicity [12, 15, 16]. Despite the high incidence and severe sequelae of lymphedema, the disease is incurable and primarily treated palliatively with compression garments and manual lymphatic drainage [1, 17–20]. Surgical options have been developed in the past decade; however, the success rates of these treatments are variable, and most patients still require compression after surgery [1, 21, 22]. Therefore, developing effective treatment strategies for lymphedema is an important unmet clinical need.
This review aims to summarize the current knowledge regarding the pharmacological interventions in experimental and clinical trials for secondary lymphedema (Table 1) [23–28]. The first section reviews systemic treatments, and the second section summarizes topical approaches.
Table 1.
Type of intervention | Proposed mechanism for lymphedema treatment | Authors, publication year |
---|---|---|
Systemic interventions | ||
VEGF-C | Lymphangiogenesis | Szuba et al., 2002 |
Subcutaneous injection | VEGF-C directly binds VEGFR-3 to activate intracellular signaling pathways that promote LEC growth and survival | |
VEGF-C Naked plasmid technique |
Lymphangiogenesis Transfer of plasmid DNA encoding VEGF-C increases VEGFR-3 expression and activation |
Yoon et al., 2003 |
VEGF-C Viral vectors: Adenovirus ---Lymfactin® Adeno-Associated |
Lymphangiogenesis Adenoviral and adeno-associated gene vectors encoding VEGF-C regenerate lymphatic vessels and preserve lymphatic architecture post VLNT |
Lai et al., 2002 Tammela et al., 2007 Hartiala et al., 2020 |
VEGF-C mRNA vectors |
Lymphangiogenesis Nucleoside-modified mRNA encoding VEGF-C stimulates site-specific lymphatic growth at low dosages |
Pardi et al., 2018 Brown et al., 2020 Szöke et al., 2021 |
Hepatocyte growth factor (HGF) |
Lymphangiogenesis VEGFR-3 independent stimulation of LEC proliferation and migration |
Kajiya et al., 2005 Wong et al., 2021 |
9-cis retinoic acid (RA) |
Lymphangiogenesis Indirect activation of PI3K/Akt pathway via fibroblast receptor signaling to stimulate LEC proliferation |
Choi et al., 2012 Wong et al., 2021 |
Adipose-derived stem cells (ADSCs) |
Lymphangiogenesis ADSCs release a secretome of bioactive factors that reinforce LEC growth and survival |
Hwang et al., 2011 Ahmadzadeh et al., 2020 Yan et al., 2011 |
Ketoprofen (NSAID) |
Anti-inflammatory 5-Lipoxygenase (5-LO) inhibition, leukotriene B4 (LTB4) synthesis |
Nakamura et al., 2009 Tian et al., 2017 Rockson et al., 2018 |
Bestatin (NSAID) |
Anti-inflammatory Leukotriene A4-hydrolyase (LTA4H) inhibition, leukotriene B4 (LTB4) synthesis |
Tian et al., 2017 Cribb et al., 2021 |
Fingolimod (Gilenya®) |
Anti-inflammatory Inhibition of activated CD4 + T cell emigration from LNs impairs TH2 differentiation |
García Nores et al., 2018 |
Neutralizing antibodies |
Anti-inflammatory Th2 inflammatory cytokine blockade (IL-4/IL-13 inhibition) |
Avraham et al., 2013 Savetsky et al., 2015 Mehrara et al., 2021 |
Doxycycline |
Anti-inflammatory Inhibition of Th2 phenotype differentiation, monocyte recruitment, polarization of alternatively activated M |
Debrah et al., 2006 Mand et al., 2012 Furlong-Silva et al., 2021 Brown et al., 2023 |
Anti-transforming growth factor beta-1 (TGF- β1) |
Anti-fibrotic Inhibition of TGF-β1 disrupts fibroblast maturation to myofibroblasts, improving lymphostatic fibrosis |
Meng et al., 2016 Yoon et al., 2020 |
Topical interventions | ||
hADSCs and VEGF-C hydrogel |
Lymphangiogenesis hADSC mediated sustained release of VEGF-C |
Hwang et al., 2011 |
Recombinant human fibroblast growth factor 2 (FGF2) |
Lymphangiogenesis VEGF-C and VEGF-D expression |
Onishi et al., 2014 |
Tacrolimus |
Anti-inflammatory Inhibition of IL-2-mediated CD4 + T cell activation/differentiation |
Gardenier et al., 2017 Gulmark Hansen et al., 2023 |
Pirfenidone |
Anti-fibrotic Inhibition of TGF-β1 |
Baik et al., 2022 |
Captopril |
Anti-fibrotic ACE inhibitor—inhibition of intracellular TGF-β1 signaling pathways |
Brown et al., 2023 |
Abbreviations: VEGF vascular endothelial growth factor, VEGFR vascular endothelial growth factor receptor, LEC lymphatic endothelial cell, DNA deoxyribonucleic acid, VLNT vascularized lymph node transplant, mRNA messenger ribonucleic acid, IL interleukin, hADSCs human adipose-derived stem cells
Systemic Treatments for Secondary Lymphedema
Lymphangiogenic Factors
Vascular endothelial growth factor C (VEGF-C) is a high-affinity ligand for the tyrosine kinase receptor VEGFR3. VEGFR3 is expressed primarily by lymphatic endothelial cells (LECs) and regulates important functions such as lymphatic vessel sprouting, LEC proliferation, migration, differentiation, and expression of endothelial nitric oxide synthase (eNOS) [1, 29–36]. Delivery of VEGF-C using an adenoviral vector to mice with primary lymphedema resulting from a heterozygous inactivating Vegfr-3 mutation increases lymphangiogenesis and decreases tissue swelling [37]. These findings were supported by studies in a rabbit ear model of secondary lymphedema in which injection of recombinant VEGF-C into the surgical bed increased lymphangiogenesis, restored normal lymphatic function, and decreased the severity of ear swelling compared with control animals [38–40]. Other groups incorporated recombinant VEGF-C in nanofibrillar collagen scaffolds to support collateral lymphatic formation across the obstructed area (BioBridge™) [41]. Animals treated with the device had decreased extracellular fluid accumulation 3 months after surgery. In follow-up animal studies, treatment with BioBridge resulted in increased collateral lymphatic formation toward other lymph node drainage basins as well as decreased edema 4 months after surgery [42]. Using the BioBridge in patients treated with vascularized lymph node transplantation (VLNT) or lymphovenous bypass (LVB) improved lymphatic drainage and decreased swelling [43]. Another retrospective study of 29 patients treated with BioBridge and VLNT also showed an increase in the number of lymphatic collectors and a decrease of dermal backflow 1 year after surgery [44].
Due to the high costs of recombinant VEGF-C, gene therapy was explored to increase VEGF-C expression. Yoon et al. utilized a naked plasmid technique to transfer plasmid DNA encoding human VEGF-C to a rabbit ear model. This study yielded similar results to treatment with recombinant VEGF-C injection decreased dermal thickening and improvement in lymphatic pumping [45]. VEGF-C gene therapy successfully increased the expression of lymphangiogenic markers like VEGFR-3, improving lymphatic vessel growth and lymphatic drainage [45, 46].
Viral vectors for VEGF delivery were also developed by several groups. These included a VEGFR3-specific VEGF-C isoform (VEGF-C156S) delivered using an adeno-associated viral (AAV) vector enabling long-term delivery [47, 48]. AAV-VEGF-C administration to the site of surgical injury increased lymphatic capillary regrowth and lymph vessel maturation in animal models [49, 50]. In humans, the efficacy of Lymfactin®, an investigational adenoviral type-5-based gene therapy vector encoding expression of human VEGF-C, has been studied in phase I and II clinical trials in conjunction with VLNT treatment [51–53]. No adverse events were reported at 24-month follow-up in a phase I clinical trial of 15 breast cancer–related upper limb lymphedema (BCRL) patients (NCT02994771). In the higher dose group, a 46% reduction in excess arm volume was demonstrated at 12 months after surgery alongside significant improvement of quality of life scores [53]. However, the results of a phase II double-blind, randomized, placebo-controlled, multicenter clinical trial (NCT03658967) were inconclusive, and development of the drug was stopped [52].
The use of mRNA therapeutics is an evolving area of gene delivery therapy that has gained increased interest since the COVID-19 pandemic [54, 55]. Nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNPs) encoding VEGF-C stimulates lymphatic growth in a mouse lymphedema model [56]. A single injection of low-dose VEGF-C mRNA-LNPs resulted in sustained VEGF-C levels for as long as 60 days after treatment, stimulated site-specific lymphatic growth, restored lymphatic function, and reversed clinical signs of lymphedema [56].
Other lymphangiogenic factors such as hepatocyte growth factor (HGF) and retinoic acid agonist 9-cis retinoic acid (9-cRA) also have therapeutic potential in preclinical models of lymphedema [57–61]. Treatment of cultured lymphatic endothelial cells (LECs) with HGF increases proliferation, migration, and lymphatic tubule formation, independent of the VEGFR-3 pathway [62]. 9-cRA, a derivative of vitamin A, stimulates LEC differentiation, migration, and collateral lymphatic formation in part by activating fibroblast receptor signaling and by downregulating expression of cell cycle inhibitors P27 and p57 [60, 61, 63–66].
Anti-inflammatory Agents
Non-steroidal Anti-inflammatory Drugs (NSAID)
Rockson and colleagues demonstrated the important role of inflammation in the pathophysiology of lymphedema and showed that ketoprofen, a non-steroidal anti-inflammatory (NSAID) drug, decreases inflammation, improves dermal-epidermal architecture, decreases swelling, and increases collateral lymphatic formation in the mouse tail model [67, 68]. Ketoprofen also significantly decreased expression of inflammatory cytokines (TNF-α and MCP-1) and increased expression of VEGF-C. Ketoprofen was used in a clinical trial (NCT02257970) in patients with primary or secondary lymphedema of the upper or lower extremities [69]. In the early phases of the study, 21 patients with lymphedema were enrolled in an open-label trial in which 75 mg ketoprofen was orally administered 3 times daily for a duration of 4 months [69]. Treatment with ketoprofen resulted in dermal thickness and collagen deposition and decreased perivascular inflammation compared to pretreatment skin biopsies. No changes in limb volume or bioimpedance were noted. The authors then conducted a randomized placebo-controlled trial with 34 patients (16 treated with ketoprofen for 4 months vs. 18 placebo controls) with upper/lower extremity lymphedema. These studies also showed improved skin changes (e.g., dermal thickness) and decreased expression of systemic inflammatory markers such as granulocyte colony-stimulating factor (G-CSF). However, as with the non-randomized trial, no significant changes in limb volume or bioimpedance were noted.
Follow-up studies by Rockson’s group found that the beneficial effects of ketoprofen are attributed to its inhibition of the 5-lipoxygenase pathway (5-LOX) metabolite, leukotriene B4 (LTB4) [70]. These findings led to another randomized clinical trial (NCT02700529) with bestatin (Ubenimex), a selective LTB4 antagonist. A cohort of 146 lower extremity lymphedema patients were treated three times daily for 6 months with bestatin. The results of this trial were inconclusive, and larger trials are planned [71].
Tetracyclines
Tetracycline antibiotics (e.g., doxycycline) have been used to treat patients with chronic filarial secondary lymphedema and decrease limb and tissue swelling independent of their antibiotic effects [72–76,77••]. A double-blind, placebo-controlled trial in Ghana evaluated lymphedema outcomes in patients with bancroftian filariasis who were treated with a 6-week regimen of 200 mg/day doxycycline and anti-parasitic therapy vs. anti-parasitic treatments alone (ISRCTN 14,757) [73]. Lymphedema outcomes were measured at 12- and 24-month time points and showed decreased serum levels of VEGF-C and soluble VEGFR3 levels, as improved skin texture and skin crease character. These findings prompted a larger randomized study with 3 treatments arms to compare the efficacy of 6-week courses of amoxicillin (1000 mg/day), doxycycline (200 mg/day), or placebo in 164 patients with mild-to-moderate filarial lymphedema (ISRCTN 90,861,344) [74]. At 12- and 24-month follow-up intervals, 44% of doxycycline patients had decreased skin thickness and swelling compared to only 3.2% and 5.6% of amoxicillin and placebo groups, respectively. Based on these results, the authors reached a consensus recommending a 6-week course of doxycycline treatment once a year or every other year for patients with mild-to-moderate filarial lymphedema [74]. Recent findings of a retrospective analysis by our group in 17 patients with breast cancer–related lymphedema (BCRL) showed improvements in patient-reported quality of life following treatment with a 6-week course of doxycycline (200 mg/day) [78]. However, in contrast to the studies in filariasis, we found no differences in limb volume or bioimpedance suggesting that larger studies may be needed.
Neutralizing Antibodies
Neutralizing antibodies have revolutionized treatment of chronic inflammatory disorders by causing targeted reductions in inflammatory responses and resultant decreased systemic toxicity. Our lab has previously shown that Th2 immune responses play a key role in the pathophysiology of lymphedema [79–81]. Based on these findings, we conducted a phase I open-label clinical trial (NCT02494206) evaluating the efficacy of anti-IL4/IL13 neutralizing antibodies in the management of unilateral BCRL. Nine women with stage I/II BCRL were treated with once-monthly injections of QBX258, a drug consisting of humanized monoclonal antibodies against IL-4 (VAK296) and IL-13 (QAX576) for 4 months [82••]. QBX258 treatment was safe with most reported adverse events being minor and self-limited in nature. Anti-IL4/IL13 neutralizing antibody treatment resulted in improved histologic appearance of lymphedematous tissues, reduced skin stiffness, and improved QoL outcome measures. Therapy with compound QBX258 significantly attenuated keratinocyte hyperplasia, mast cell infiltration, and Th2-related cytokine expression within the skin. However, we found no significant improvements in arm volumes or bioimpedance. Future, larger studies possibly with more targeted anti-Th2 therapies are needed and planned.
Topical Treatments for Secondary Lymphedema
Lymphangiogenic Factors
Topical formulations of recombinant human VEGF-C have been developed to ensure sustained release and to avoid direct injection [38]. Hwang and colleagues used a gelatin hydrogel containing VEGF-C at the site of tissue injury in a lymphedema mouse model [83]. Mice treated with both implanted human adipose-derived stem cells (hADSCs) and VEGF-C hydrogel had decreased dermal edema and improved lymphatic regeneration compared to the controls treated with just hADSCs or VEGF-C alone.
Fibroblast growth factor 2 (FGF2) promotes lymphangiogenesis via induction of VEGF-C and VEGF-D [84]. Onishi et al. used topical basic fibroblast growth factor (bFGF) to treat secondary lymphedema in a rat tail model [85]. Treatment with topical bFGF increased expression of VEGF-C/D, increased lymphatic vessel density, decreased tail swelling, and improved lymphatic function.
Anti-inflammatory Agents
Tacrolimus
Tacrolimus is a macrolide calcineurin inhibitor with anti-T-cell activity. FDA-approved for treating cutaneous inflammatory conditions, topical tacrolimus administration inhibits CD4+ cell proliferation and differentiation by inhibiting IL-2 [86–94]. Topical administration of tacrolimus decreases inflammation, Th2 cytokines, fibroadipose tissue deposition, swelling, and improves lymphatic function in mouse models of lymphedema [95]. Topical application of tacrolimus, unlike oral administration, did not result in significant systemic absorption and had no significant systemic anti-inflammatory effects [91]. A recent open-label, single-arm, phase II trial (NCT04541290) demonstrated that a 6-month treatment course with topical tacrolimus significantly improved limb volumes, bioimpedance scores, and quality of life scores in eighteen women with BCRL. However, assessment of lymphatic function using ICG lymphography was inconclusive [96].
Anti-fibrotic Therapies
Dermal fibrosis is a histological hallmark of lymphedema, and the degree of fibrosis positively correlates with the severity of lymphedema [97–99]. Progressive fibrosis results in lymphatic obstruction perpetuating the cycle of fluid accumulation, inflammation, and fibrosis. Transforming growth factor beta-1 (TGF‐β1) is a key regulator of fibrosis in a variety of organ systems [99, 101, 102,100••, ]. Using a rat hindlimb model of secondary lymphedema, Sano et al. found that the TGF‐β1/Smad signaling pathway is responsible for the early to late changes of fibrosis that is seen in lymphedema patients. We have shown that the expression of TGF-B1 and its downstream mediators is markedly increased in skin biopsies collected from patients with stage II/III BCRL [100••]. Other studies have shown that inhibiting TGF‐β1 activity with a small-molecule inhibitor improves radiation-induced fibrosis and lymphatic dysfunction [103]. Blockade of TGF‐β1 signaling using neutralizing antibodies decreases fibrosis, increases collateral lymphatic formation, and decreases the pathophysiology of lymphedema in preclinical mouse models [102, 104, 105]. TGF‐β1 in lymphatic fluid increases stiffness and proliferation of fibroblasts, LECs, and lymphatic smooth muscle cells (LSMCs). Topical application of pirfenidone (PFD), a small‐molecule inhibitor approved by the US Food and Drug Administration (FDA) for the treatment of idiopathic pulmonary fibrosis, decreases TGF‐β1 signaling, fibrosis, and the pathophysiologic findings of lymphedema in mouse models.
TGF-B1 activity in cardiac, renal, and hepatic fibrosis is modulated by tissue activity of tissue-specific renin-angiotensin system [106–115]. Expression of angiotensin-converting enzyme in these tissues drives the conversion of angiotensin I (Ang I) to angiotensin II (Ang II). AngII is a key regulator of fibrosis by modulating intracellular TGF-B1 activity and downstream signaling. ACE inhibitors or Ang II receptor antagonists inhibit fibrosis by decreasing TGF-β1 activity in cardiac, renal, and hepatic models of fibrosis [106–115]. We have recently shown that the expression of ACE and AngII is increased in clinical lymphedema biopsy specimens [116]. Topical captopril treatment in mouse models of lymphedema resulted in decreased fibrosis, inhibition of intracellular TGF-β1 signaling pathways, decreased inflammation, and decreased swelling.
Conclusions
Many potential treatment options for lymphedema have been explored throughout the past two decades including systemic agents and topical approaches to decrease the potential toxicity of systemic treatment. Treatment strategies including lymphangiogenic factors, anti-inflammatory agents, and anti-fibrotic therapies may be used independently or in conjunction with current surgical approaches. Larger studies with better methodological design and standardized outcome measures are needed to define the role of these strategies in the clinical setting.
Funding
This research was supported in part by the NIH through R01 HL111130 awarded to Babak J. Mehrara, MD, T32 CA009501 (stipend for A.C.), the Cancer Center Support Grant P30 CA008748 that supports the research infrastructure at Memorial Sloan Kettering Cancer Center. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Declarations
Conflict of Interest
Babak J. Mehrara, MD, is the recipient of investigator-initiated research grants from PureTech and Regeneron and has received royalty payments from PureTech; he also has served as a consultant for Pfizer Corp.
Joseph H. Dayan, MD, is a paid consultant for the Stryker Corporation, has intellectual property rights with Elucida Oncology and equity interest in Welwaze Medical, LLC, and has a royalty agreement with Springer Publishers for Multimodal Management of Upper and Lower Extremity Lymphedema. All other authors report no potential conflicts of interest.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Stav Brown, Email: brownrs@mskcc.org.
Adana C. Campbell, Email: campbea5@mskcc.org
Kevin Kuonqui, Email: kuonquik@mskcc.org.
Ananta Sarker, Email: sarkera@mskcc.org.
Hyeung Ju Park, Email: parkh@mskcc.org.
Jinyeon Shin, Email: shinj@mskcc.org.
Raghu P. Kataru, Email: katarur@mskcc.org
Michelle Coriddi, Email: coriddim@mskcc.org.
Joseph H. Dayan, Email: dayanj@mskcc.org
Babak J. Mehrara, Email: mehrarab@mskcc.org
References
Papers of particular interest, published recently, have been highlighted as: •• Of major importance
- 1.Dayan JH, Ly CL, Kataru RP, Mehrara BJ. Lymphedema: pathogenesis and novel therapies. Annu Rev Med. 2018;69:263–276. doi: 10.1146/annurev-med-060116-022900. [DOI] [PubMed] [Google Scholar]
- 2.Greene AK, Goss JA. Diagnosis and staging of lymphedema. Semin Plast Surg. 2018;32(1):12–16. doi: 10.1055/s-0038-1635117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rockson SG, Rivera KK. Estimating the population burden of lymphedema. Ann N Y Acad Sci. 2008;1131:147–154. doi: 10.1196/annals.1413.014. [DOI] [PubMed] [Google Scholar]
- 4.DiSipio T, Rye S, Newman B, Hayes S. Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. Lancet Oncol. 2013;14(6):500–515. doi: 10.1016/s1470-2045(13)70076-7. [DOI] [PubMed] [Google Scholar]
- 5.Warren AG, Brorson H, Borud LJ, Slavin SA. Lymphedema: a comprehensive review. Ann Plast Surg. 2007;59(4):464–472. doi: 10.1097/01.sap.0000257149.42922.7e. [DOI] [PubMed] [Google Scholar]
- 6.Rockson SG. Lymphedema after breast cancer treatment. N Engl J Med. 2018;379(20):1937–1944. doi: 10.1056/NEJMcp1803290. [DOI] [PubMed] [Google Scholar]
- 7.Salinas-Huertas S, Luzardo-González A, Vázquez-Gallego S, et al. Risk factors for lymphedema after breast surgery: a prospective cohort study in the era of sentinel lymph node biopsy. Breast Dis. 2021. 10.3233/bd-210043. [DOI] [PubMed]
- 8.Cormier JN, Askew RL, Mungovan KS, Xing Y, Ross MI, Armer JM. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer. 2010;116(22):5138–5149. doi: 10.1002/cncr.25458. [DOI] [PubMed] [Google Scholar]
- 9.Beaulac SM, McNair LA, Scott TE, LaMorte WW, Kavanah MT. Lymphedema and quality of life in survivors of early-stage breast cancer. Arch Surg. 2002;137(11):1253–1257. doi: 10.1001/archsurg.137.11.1253. [DOI] [PubMed] [Google Scholar]
- 10.Heiney SP, McWayne J, Cunningham JE, et al. Quality of life and lymphedema following breast cancer. Lymphology. 2007;40(4):177–184. [PubMed] [Google Scholar]
- 11.Pusic AL, Cemal Y, Albornoz C, et al. Quality of life among breast cancer patients with lymphedema: a systematic review of patient-reported outcome instruments and outcomes. J Cancer Surviv. 2013;7(1):83–92. doi: 10.1007/s11764-012-0247-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Shih YC, Xu Y, Cormier JN, et al. Incidence, treatment costs, and complications of lymphedema after breast cancer among women of working age: a 2-year follow-up study. J Clin Oncol. 2009;27(12):2007–2014. doi: 10.1200/JCO.2008.18.3517. [DOI] [PubMed] [Google Scholar]
- 13.Taghian NR, Miller CL, Jammallo LS, O’Toole J, Skolny MN. Lymphedema following breast cancer treatment and impact on quality of life: a review. Crit Rev Oncol Hematol. 2014;92(3):227–234. doi: 10.1016/j.critrevonc.2014.06.004. [DOI] [PubMed] [Google Scholar]
- 14.Yuan Y, Arcucci V, Levy SM, Achen MG. Modulation of immunity by lymphatic dysfunction in lymphedema. Front Immunol. 2019;10:76. doi: 10.3389/fimmu.2019.00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Armer J, Porock D. Self-management of fatigue among women with lymphedema. Lymphology. 2002;35(Suppl):208–213. [Google Scholar]
- 16.De Vrieze T, Nevelsteen I, Thomis S, et al. What are the economic burden and costs associated with the treatment of breast cancer-related lymphoedema? A systematic review. Support Care Cancer. 2020;28(2):439–449. doi: 10.1007/s00520-019-05101-8. [DOI] [PubMed] [Google Scholar]
- 17.Barufi S, Pereira de Godoy HJ, Pereira de Godoy JM, Guerreiro Godoy MF. Exercising and compression mechanism in the treatment of lymphedema. Cureus. 2021;13(7):e16121. doi: 10.7759/cureus.16121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Uzkeser H, Karatay S, Erdemci B, Koc M, Senel K. Efficacy of manual lymphatic drainage and intermittent pneumatic compression pump use in the treatment of lymphedema after mastectomy: a randomized controlled trial. Breast Cancer (Tokyo, Japan) 2015;22(3):300–307. doi: 10.1007/s12282-013-0481-3. [DOI] [PubMed] [Google Scholar]
- 19.Keith L, Rowsemitt C, Richards LG. Lifestyle modification group for lymphedema and obesity results in significant health outcomes. Am J Lifestyle Med. 2020;14(4):420–428. doi: 10.1177/1559827617742108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Schmitz KH, Troxel AB, Dean LT, et al. Effect of home-based exercise and weight loss programs on breast cancer-related lymphedema outcomes among overweight breast cancer survivors: the WISER survivor randomized clinical trial. JAMA Oncol. 2019;5(11):1605–1613. doi: 10.1001/jamaoncol.2019.2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kayıran O, De La Cruz C, Tane K, Soran A. Lymphedema: from diagnosis to treatment. Turk J Surg. 2017;33(2):51–57. doi: 10.5152/turkjsurg.2017.3870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Carl HM, Walia G, Bello R, et al. Systematic review of the surgical treatment of extremity lymphedema. J Reconstr Microsurg. 2017;33(6):412–425. doi: 10.1055/s-0037-1599100. [DOI] [PubMed] [Google Scholar]
- 23.Forte AJ, Boczar D, Huayllani MT, Lu X, McLaughlin SA. Pharmacotherapy agents in lymphedema treatment: a systematic review. Cureus. 2019;11(12):e6300. doi: 10.7759/cureus.6300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Walker J, Tanna S, Roake J, Lyons O. A systematic review of pharmacologic and cell-based therapies for treatment of lymphedema (2010–2021). J Vasc Surg Venous Lymphatic Disord. 2021. 10.1016/j.jvsv.2021.09.004. [DOI] [PubMed]
- 25.Badger C, Seers K, Preston N, Mortimer P. Antibiotics / anti-inflammatories for reducing acute inflammatory episodes in lymphoedema of the limbs. Cochrane Database Syst Rev. 2004;2:Cd003143. doi: 10.1002/14651858.CD003143.pub2. [DOI] [PubMed] [Google Scholar]
- 26.Forte AJ, Boczar D, Huayllani MT, McLaughlin SA, Bagaria S. Topical approach to delivering targeted therapies in lymphedema treatment: a systematic review. Cureus. 2019;11(12):e6269. doi: 10.7759/cureus.6269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Forte AJ, Boczar D, Huayllani MT, McLaughlin SA, Bagaria S. Use of gene transfer vectors in lymphedema treatment: a systematic review. Cureus. 2019;11(10):e5887. doi: 10.7759/cureus.5887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Forte AJ, Boczar D, Huayllani MT, Cinotto GJ, McLaughlin S. Targeted therapies in surgical treatment of lymphedema: a systematic review. Cureus. 2019;11(8):e5397. doi: 10.7759/cureus.5397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Secker GA, Harvey NL. Regulation of VEGFR signalling in lymphatic vascular development and disease: an update. Int J Mol Sci. 2021;22(14). 10.3390/ijms22147760. [DOI] [PMC free article] [PubMed]
- 30.Lahdenranta J, Hagendoorn J, Padera TP, et al. Endothelial nitric oxide synthase mediates lymphangiogenesis and lymphatic metastasis. Can Res. 2009;69(7):2801–2808. doi: 10.1158/0008-5472.Can-08-4051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mäkinen T, Jussila L, Veikkola T, et al. Inhibition of lymphangiogenesis with resulting lymphedema in transgenic mice expressing soluble VEGF receptor-3. Nat Med. 2001;7(2):199–205. doi: 10.1038/84651. [DOI] [PubMed] [Google Scholar]
- 32.Pytowski B, Goldman J, Persaud K, et al. Complete and specific inhibition of adult lymphatic regeneration by a novel VEGFR-3 neutralizing antibody. J Natl Cancer Inst. 2005;97(1):14–21. doi: 10.1093/jnci/dji003. [DOI] [PubMed] [Google Scholar]
- 33.Veikkola T, Jussila L, Makinen T, et al. Signalling via vascular endothelial growth factor receptor-3 is sufficient for lymphangiogenesis in transgenic mice. EMBO J. 2001;20(6):1223–1231. doi: 10.1093/emboj/20.6.1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Oh SJ, Jeltsch MM, Birkenhäger R, et al. VEGF and VEGF-C: specific induction of angiogenesis and lymphangiogenesis in the differentiated avian chorioallantoic membrane. Dev Biol. 1997;188(1):96–109. doi: 10.1006/dbio.1997.8639. [DOI] [PubMed] [Google Scholar]
- 35.Mäkinen T, Veikkola T, Mustjoki S, et al. Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3. EMBO J. 2001;20(17):4762–4773. doi: 10.1093/emboj/20.17.4762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zarkada G, Heinolainen K, Makinen T, Kubota Y, Alitalo K. VEGFR3 does not sustain retinal angiogenesis without VEGFR2. Proc Natl Acad Sci USA. 2015;112(3):761–766. doi: 10.1073/pnas.1423278112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Karkkainen MJ, Saaristo A, Jussila L, et al. A model for gene therapy of human hereditary lymphedema. Proc Natl Acad Sci USA. 2001;98(22):12677–12682. doi: 10.1073/pnas.221449198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Szuba A, Skobe M, Karkkainen MJ, et al. Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEB J. 2002;16(14):1985–1987. doi: 10.1096/fj.02-0401fje. [DOI] [PubMed] [Google Scholar]
- 39.Cheung L, Han J, Beilhack A, et al. An experimental model for the study of lymphedema and its response to therapeutic lymphangiogenesis. BioDrugs. 2006;20(6):363–370. doi: 10.2165/00063030-200620060-00007. [DOI] [PubMed] [Google Scholar]
- 40.Jin DP, An A, Liu J, Nakamura K, Rockson SG. Therapeutic responses to exogenous VEGF-C administration in experimental lymphedema: immunohistochemical and molecular characterization. Lymphat Res Biol. 2009;7(1):47–57. doi: 10.1089/lrb.2009.0002. [DOI] [PubMed] [Google Scholar]
- 41.Hadamitzky C, Zaitseva TS, Bazalova-Carter M, et al. Aligned nanofibrillar collagen scaffolds - guiding lymphangiogenesis for treatment of acquired lymphedema. Biomaterials. 2016;102:259–267. doi: 10.1016/j.biomaterials.2016.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nguyen D, Zaitseva TS, Zhou A, et al. Lymphatic regeneration after implantation of aligned nanofibrillar collagen scaffolds: preliminary preclinical and clinical results. J Surg Oncol. 2021. 10.1002/jso.26679. [DOI] [PubMed]
- 43.Danielle H Rochlin MP, Dung H Nguyen. Nanofibrillar collagen scaffold implantation enhances lymphatic regeneration in conjunction with lymphatic venous anastomosis or vascularized lymph node transfer. 2019 WSLS Free Paper 003. 2019.
- 44.Nguyen DH, Zhou A, Posternak V, Rochlin DH. Nanofibrillar collagen scaffold enhances edema reduction and formation of new lymphatic collectors after lymphedema surgery. Plast Reconstr Surg. 2021. 10.1097/prs.0000000000008590. [DOI] [PubMed]
- 45.Yoon YS, Murayama T, Gravereaux E, et al. VEGF-C gene therapy augments postnatal lymphangiogenesis and ameliorates secondary lymphedema. J Clin Invest. 2003;111(5):717–725. doi: 10.1172/jci15830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Liu Y, Fang Y, Dong P, et al. Effect of vascular endothelial growth factor C (VEGF-C) gene transfer in rat model of secondary lymphedema. Vascul Pharmacol. 2008;49(1):44–50. doi: 10.1016/j.vph.2008.05.003. [DOI] [PubMed] [Google Scholar]
- 47.Saaristo A, Veikkola T, Tammela T, et al. Lymphangiogenic gene therapy with minimal blood vascular side effects. J Exp Med. 2002;196(6):719–730. doi: 10.1084/jem.20020587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lai CM, Lai YK, Rakoczy PE. Adenovirus and adeno-associated virus vectors. DNA Cell Biol. 2002;21(12):895–913. doi: 10.1089/104454902762053855. [DOI] [PubMed] [Google Scholar]
- 49.Tammela T, Saaristo A, Holopainen T, et al. Therapeutic differentiation and maturation of lymphatic vessels after lymph node dissection and transplantation. Nat Med. 2007;13(12):1458–1466. doi: 10.1038/nm1689. [DOI] [PubMed] [Google Scholar]
- 50.Lähteenvuo M, Honkonen K, Tervala T, et al. Growth factor therapy and autologous lymph node transfer in lymphedema. Circulation. 2011;123(6):613–620. doi: 10.1161/circulationaha.110.965384. [DOI] [PubMed] [Google Scholar]
- 51.Hartiala P, Suominen S, Suominen E, et al. Phase 1 Lymfactin(Ⓡ) study: short-term safety of combined adenoviral VEGF-C and lymph node transfer treatment for upper extremity lymphedema. J Plast Reconstr Aesthetic Surg. 2020;73(9):1612–1621. doi: 10.1016/j.bjps.2020.05.009. [DOI] [PubMed] [Google Scholar]
- 52.Hartiala P, Lahdenperä O, Vuolanto A, Saarikko A. Abstract OT1-06-01: Lymfactin, an investigational adenoviral gene therapy expressing VEGF-C, is currently studied in a double-blind, randomized, placebo-controlled, multicenter, phase 2 clinical study in patients suffering from breast cancer associated secondary lymphedema (BCAL) Cancer Res. 2020;80(4 Supplement):OT1-06-01-OT1-06-01. doi: 10.1158/1538-7445.Sabcs19-ot1-06-01. [DOI] [Google Scholar]
- 53.Leppäpuska IM, Hartiala P, Suominen S, et al. Phase 1 Lymfactin® study: 24-month efficacy and safety results of combined adenoviral VEGF-C and lymph node transfer treatment for upper extremity lymphedema. J Plast Reconstr Aesthet Surg. 2022;75(11):3938–3945. doi: 10.1016/j.bjps.2022.08.011. [DOI] [PubMed] [Google Scholar]
- 54.Brown S, Brown T, Cederna PS, Rohrich RJ. The race for a COVID-19 vaccine: current trials, novel technologies, and future directions. Plast Reconstr Surg Global Open. 2020;8(10):e3206. doi: 10.1097/gox.0000000000003206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261–279. doi: 10.1038/nrd.2017.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Szőke D, Kovács G, Kemecsei É, et al. Nucleoside-modified VEGFC mRNA induces organ-specific lymphatic growth and reverses experimental lymphedema. Nat Commun. 2021;12(1):3460. doi: 10.1038/s41467-021-23546-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kajiya K, Hirakawa S, Ma B, Drinnenberg I, Detmar M. Hepatocyte growth factor promotes lymphatic vessel formation and function. Embo j. 2005;24(16):2885–2895. doi: 10.1038/sj.emboj.7600763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Saito Y, Nakagami H, Morishita R, et al. Transfection of human hepatocyte growth factor gene ameliorates secondary lymphedema via promotion of lymphangiogenesis. Circulation. 2006;114(11):1177–1184. doi: 10.1161/circulationaha.105.602953. [DOI] [PubMed] [Google Scholar]
- 59.Cao R, Björndahl MA, Gallego MI, et al. Hepatocyte growth factor is a lymphangiogenic factor with an indirect mechanism of action. Blood. 2006;107(9):3531–3536. doi: 10.1182/blood-2005-06-2538. [DOI] [PubMed] [Google Scholar]
- 60.Choi I, Lee S, Kyoung Chung H, et al. 9-cis retinoic acid promotes lymphangiogenesis and enhances lymphatic vessel regeneration: therapeutic implications of 9-cis retinoic acid for secondary lymphedema. Circulation. 2012;125(7):872–882. doi: 10.1161/circulationaha.111.030296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wong A. Role of therapeutic lymphangiogenesis in the prevention and treatment of lymphedema. Boston Lymphatic Symposium (unpublished) 2021.
- 62.Palmieri M, Di Sarno L, Tommasi A, et al. MET somatic activating mutations are responsible for lymphovenous malformation and can be identified using cell-free DNA next generation sequencing liquid biopsy. J Vasc Surg Venous Lymphat Disord. 2021;9(3):740–744. doi: 10.1016/j.jvsv.2020.07.015. [DOI] [PubMed] [Google Scholar]
- 63.Bramos A, Perrault D, Yang S, Jung E, Hong YK, Wong AK. Prevention of postsurgical lymphedema by 9-cis retinoic acid. Ann Surg. 2016;264(2):353–361. doi: 10.1097/sla.0000000000001525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Perrault DP, Lee GK, Park SY, et al. Small peptide modulation of fibroblast growth factor receptor 3-dependent postnatal lymphangiogenesis. Lymphat Res Biol. 2019;17(1):19–29. doi: 10.1089/lrb.2018.0035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jin M, Yu Y, Qi H, et al. A novel FGFR3-binding peptide inhibits FGFR3 signaling and reverses the lethal phenotype of mice mimicking human thanatophoric dysplasia. Hum Mol Genet. 2012;21(26):5443–5455. doi: 10.1093/hmg/dds390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Daneshgaran G, Paik CB, Cooper MN, et al. Prevention of postsurgical lymphedema via immediate delivery of sustained-release 9-cis retinoic acid to the lymphedenectomy site. J Surg Oncol. 2020;121(1):100–108. doi: 10.1002/jso.25587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Tabibiazar R, Cheung L, Han J, et al. Inflammatory manifestations of experimental lymphatic insufficiency. PLoS Med. 2006;3(7):e254. doi: 10.1371/journal.pmed.0030254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nakamura K, Radhakrishnan K, Wong YM, Rockson SG. Anti-inflammatory pharmacotherapy with ketoprofen ameliorates experimental lymphatic vascular insufficiency in mice. PLoS One. 2009;4(12):e8380. doi: 10.1371/journal.pone.0008380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Rockson SG, Tian W, Jiang X, et al. Pilot studies demonstrate the potential benefits of antiinflammatory therapy in human lymphedema. JCI Insight. 2018;3(20). 10.1172/jci.insight.123775. [DOI] [PMC free article] [PubMed]
- 70.Tian W, Rockson SG, Jiang X, et al. Leukotriene B(4) antagonism ameliorates experimental lymphedema. Sci Transl Med. 2017;9(389). 10.1126/scitranslmed.aal3920. [DOI] [PubMed]
- 71.Ubenimex in adult patients with lymphedema of the lower limb (ULTRA). . July 16, 2021. https://ClinicalTrials.gov/show/NCT02700529
- 72.Sapadin AN, Fleischmajer R. Tetracyclines: nonantibiotic properties and their clinical implications. J Am Acad Dermatol. 2006;54(2):258–265. doi: 10.1016/j.jaad.2005.10.004. [DOI] [PubMed] [Google Scholar]
- 73.Debrah AY, Mand S, Specht S, et al. Doxycycline reduces plasma VEGF-C/sVEGFR-3 and improves pathology in lymphatic filariasis. PLoS Pathog. 2006;2(9):e92. doi: 10.1371/journal.ppat.0020092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mand S, Debrah AY, Klarmann U, et al. Doxycycline improves filarial lymphedema independent of active filarial infection: a randomized controlled trial. Clin Infect Dis. 2012;55(5):621–630. doi: 10.1093/cid/cis486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bandi C, McCall JW, Genchi C, Corona S, Venco L, Sacchi L. Effects of tetracycline on the filarial worms Brugia pahangi and Dirofilaria immitis and their bacterial endosymbionts Wolbachia. Int J Parasitol. 1999;29(2):357–364. doi: 10.1016/s0020-7519(98)00200-8. [DOI] [PubMed] [Google Scholar]
- 76.Hoerauf A, Mand S, Fischer K, et al. Doxycycline as a novel strategy against bancroftian filariasis-depletion of Wolbachia endosymbionts from Wuchereria bancrofti and stop of microfilaria production. Med Microbiol Immunol. 2003;192(4):211–216. doi: 10.1007/s00430-002-0174-6. [DOI] [PubMed] [Google Scholar]
- 77.•• Furlong-Silva J, Cross SD, Marriott AE, et al. Tetracyclines improve experimental lymphatic filariasis pathology by disrupting interleukin-4 receptor-mediated lymphangiogenesis. J Clin Investig. 2021;131(5). 10.1172/jci140853. Findings from this study suggest that tetracyclines decrease limb and tissue swelling independent of their antibiotic effects. [DOI] [PMC free article] [PubMed]
- 78.Brown S, Dayan JH, Coriddi M, McGrath L, Kataru RP, Mehrara BJ. Doxycycline for the treatment of breast cancer-related lymphedema. Front Pharmacol. 2022;13:1028926. doi: 10.3389/fphar.2022.1028926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Garcia Nores GD, Ly CL, Cuzzone DA, et al. CD4(+) T cells are activated in regional lymph nodes and migrate to skin to initiate lymphedema. Nat Commun. 2018;9(1):1970. doi: 10.1038/s41467-018-04418-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zampell JC, Yan A, Elhadad S, Avraham T, Weitman E, Mehrara BJ. CD4(+) cells regulate fibrosis and lymphangiogenesis in response to lymphatic fluid stasis. PLoS One. 2012;7(11):e49940. doi: 10.1371/journal.pone.0049940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Avraham T, Zampell JC, Yan A, et al. Th2 differentiation is necessary for soft tissue fibrosis and lymphatic dysfunction resulting from lymphedema. FASEB J. 2013;27(3):1114–1126. doi: 10.1096/fj.12-222695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.•• Mehrara BJ, Park HJ, Kataru RP, et al. Pilot study of anti-Th2 immunotherapy for the treatment of breast cancer-related upper extremity lymphedema. Biology. 2021;10(9):934. Findings from this study suggest that anti-IL4/IL13 neutralizing antibody treatment is a promising avenue for the treatment of lymphedema. [DOI] [PMC free article] [PubMed]
- 83.Hwang JH, Kim IG, Lee JY, et al. Therapeutic lymphangiogenesis using stem cell and VEGF-C hydrogel. Biomaterials. 2011;32(19):4415–4423. doi: 10.1016/j.biomaterials.2011.02.051. [DOI] [PubMed] [Google Scholar]
- 84.Javerzat S, Auguste P, Bikfalvi A. The role of fibroblast growth factors in vascular development. Trends Mol Med. 2002;8(10):483–489. doi: 10.1016/s1471-4914(02)02394-8. [DOI] [PubMed] [Google Scholar]
- 85.Onishi T, Nishizuka T, Kurahashi T, et al. Topical bFGF improves secondary lymphedema through lymphangiogenesis in a rat tail model. Plast Reconstr Surg Glob Open. 2014;2(8):e196. doi: 10.1097/gox.0000000000000154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Clipstone NA, Crabtree GR. Identification of calcineurin as a key signalling enzyme in T-lymphocyte activation. Nature. 1992;357(6380):695–697. doi: 10.1038/357695a0. [DOI] [PubMed] [Google Scholar]
- 87.Liao W, Lin JX, Leonard WJ. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity. 2013;38(1):13–25. doi: 10.1016/j.immuni.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Rautajoki KJ, Kylaniemi MK, Raghav SK, Rao K, Lahesmaa R. An insight into molecular mechanisms of human T helper cell differentiation. Ann Med. 2008;40(5):322–335. doi: 10.1080/07853890802068582. [DOI] [PubMed] [Google Scholar]
- 89.Smith KA. Interleukin-2: inception, impact, and implications. Science (New York, NY) 1988;240(4856):1169–1176. doi: 10.1126/science.3131876. [DOI] [PubMed] [Google Scholar]
- 90.Chow CW, Rincón M, Davis RJ. Requirement for transcription factor NFAT in interleukin-2 expression. Mol Cell Biol. 1999;19(3):2300–2307. doi: 10.1128/mcb.19.3.2300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.US prescribing information: Protopic (Tacrolimus). https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/050777s018lbl.pdf.
- 92.Ruzicka T, Bieber T, Schöpf E, et al. A short-term trial of tacrolimus ointment for atopic dermatitis. European Tacrolimus Multicenter Atopic Dermatitis Study Group. N Engl J Med. 1997;337(12):816–21. doi: 10.1056/nejm199709183371203. [DOI] [PubMed] [Google Scholar]
- 93.Wang C, Lin A. Efficacy of topical calcineurin inhibitors in psoriasis. J Cutan Med Surg. 2014;18(1):8–14. doi: 10.2310/7750.2013.13059. [DOI] [PubMed] [Google Scholar]
- 94.Mancuso G, Berdondini RM. Localized scleroderma: response to occlusive treatment with tacrolimus ointment. Br J Dermatol. 2005;152(1):180–182. doi: 10.1111/j.1365-2133.2004.06318.x. [DOI] [PubMed] [Google Scholar]
- 95.Gardenier JC, Kataru RP, Hespe GE, et al. Topical tacrolimus for the treatment of secondary lymphedema. Nat Commun. 2017;8:14345. doi: 10.1038/ncomms14345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gulmark Hansen FC, Jørgensen MG, Sørensen JA. Treatment of breast cancer-related lymphedema with topical tacrolimus: a prospective, open-label, single-arm, phase II pilot trial. J Breast Cancer. 2023;26(1):46–59. doi: 10.4048/jbc.2023.26.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kataru RP, Wiser I, Baik JE, et al. Fibrosis and secondary lymphedema: chicken or egg? Transl Res. 2019;209:68–76. doi: 10.1016/j.trsl.2019.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mihara M, Hara H, Hayashi Y, et al. Pathological steps of cancer-related lymphedema: histological changes in the collecting lymphatic vessels after lymphadenectomy. PloS One. 2012;7(7):e41126. doi: 10.1371/journal.pone.0041126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Avraham T, Daluvoy S, Zampell J, et al. Blockade of transforming growth factor-beta1 accelerates lymphatic regeneration during wound repair. Am J Pathol. 2010;177(6):3202–3214. doi: 10.2353/ajpath.2010.100594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.•• Baik JE, Park HJ, Kataru RP, et al. TGF-β1 mediates pathologic changes of secondary lymphedema by promoting fibrosis and inflammation. Clin Transl Med. 2022;12(6):e758. 10.1002/ctm2.758. Findings from this study suggest that expression of TGF-B1 and its downstream mediators is markedly increased in lymphedema, suggesting that this is an important potential therapeutic target for lymphedema. [DOI] [PMC free article] [PubMed]
- 101.Brown S, Nores GDG, Sarker A, et al. Topical captopril: a promising treatment for secondary lymphedema. Transl Res. 2023. 10.1016/j.trsl.2023.01.005. [DOI] [PMC free article] [PubMed]
- 102.Sano M, Hirakawa S, Suzuki M, et al. Potential role of transforming growth factor-beta 1/Smad signaling in secondary lymphedema after cancer surgery. Cancer Sci. 2020;111(7):2620–2634. doi: 10.1111/cas.14457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Avraham T, Yan A, Zampell JC, et al. Radiation therapy causes loss of dermal lymphatic vessels and interferes with lymphatic function by TGF-beta1-mediated tissue fibrosis. Am J Physiol Cell Physiol. 2010;299(3):C589–605. doi: 10.1152/ajpcell.00535.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Clavin NW, Avraham T, Fernandez J, et al. TGF-beta1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol. 2008;295(5):H2113–H2127. doi: 10.1152/ajpheart.00879.2008. [DOI] [PubMed] [Google Scholar]
- 105.Yoon SH, Kim KY, Wang Z, et al. EW-7197, a transforming growth factor-beta type I receptor kinase inhibitor, ameliorates acquired lymphedema in a mouse tail model. Lymphat Res Biol. 2020;18(5):433–438. doi: 10.1089/lrb.2018.0070. [DOI] [PubMed] [Google Scholar]
- 106.Zhang Y, Shao L, Ma A, et al. Telmisartan delays myocardial fibrosis in rats with hypertensive left ventricular hypertrophy by TGF-β1/Smad signal pathway. Hypertension Res. 2014;37(1):43–49. doi: 10.1038/hr.2013.119. [DOI] [PubMed] [Google Scholar]
- 107.Jonsson JR, Clouston AD, Ando Y, et al. Angiotensin-converting enzyme inhibition attenuates the progression of rat hepatic fibrosis. Gastroenterology. 2001;121(1):148–155. doi: 10.1053/gast.2001.25480. [DOI] [PubMed] [Google Scholar]
- 108.Chen K, Wei Y, Sharp GC, Braley-Mullen H. Inhibition of TGFbeta1 by anti-TGFbeta1 antibody or lisinopril reduces thyroid fibrosis in granulomatous experimental autoimmune thyroiditis. J Immunol (Baltimore, Md : 1950) 2002;169(11):6530–8. doi: 10.4049/jimmunol.169.11.6530. [DOI] [PubMed] [Google Scholar]
- 109.Sun N, Zhai L, Li H, Shi LH, Yao Z, Zhang B. Angiotensin-converting enzyme inhibitor (ACEI)-mediated amelioration in renal fibrosis involves suppression of mast cell degranulation. Kidney Blood Press Res. 2016;41(1):108–118. doi: 10.1159/000368549. [DOI] [PubMed] [Google Scholar]
- 110.Fang QQ, Wang XF, Zhao WY, et al. Angiotensin-converting enzyme inhibitor reduces scar formation by inhibiting both canonical and noncanonical TGF-β1 pathways. Sci Reports. 2018;8(1):3332. doi: 10.1038/s41598-018-21600-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Töx U, Steffen HM. Impact of inhibitors of the renin-angiotensin-aldosterone system on liver fibrosis and portal hypertension. Curr Med Chem. 2006;13(30):3649–3661. doi: 10.2174/092986706779026138. [DOI] [PubMed] [Google Scholar]
- 112.Morrissey JJ, Ishidoya S, McCracken R, Klahr S. The effect of ACE inhibitors on the expression of matrix genes and the role of p53 and p21 (WAF1) in experimental renal fibrosis. Kidney Int Suppl. 1996;54:S83–S87. [PubMed] [Google Scholar]
- 113.Wengrower D, Zanninelli G, Pappo O, et al. Prevention of fibrosis in experimental colitis by captopril: the role of tgf-beta1. Inflamm Bowel Dis. 2004;10(5):536–545. doi: 10.1097/00054725-200409000-00007. [DOI] [PubMed] [Google Scholar]
- 114.Cohen EP, Fish BL, Moulder JE. The renin-angiotensin system in experimental radiation nephropathy. J Lab Clin Med. 2002;139(4):251–257. doi: 10.1067/mlc.2002.122279. [DOI] [PubMed] [Google Scholar]
- 115.Hye Khan MA, Fish B, Wahl G, et al. Epoxyeicosatrienoic acid analogue mitigates kidney injury in a rat model of radiation nephropathy. Clin Sci (London, England : 1979) 2016;130(8):587–99. doi: 10.1042/cs20150778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ardekani GS, Aghaie S, Nemati MH, Handjani F, Kasraee B. Treatment of a postburn keloid scar with topical captopril: report of the first case. Plast Reconstr Surg. 2009;123(3):112e–113e. doi: 10.1097/PRS.0b013e31819a34db. [DOI] [PubMed] [Google Scholar]