new research on lymphedema

  • Adolescent and Young Adult Cancer
  • Bile Duct Cancer
  • Bladder Cancer
  • Brain Cancer
  • Breast Cancer
  • Cervical Cancer
  • Childhood Cancer
  • Colorectal Cancer
  • Endometrial Cancer
  • Esophageal Cancer
  • Head and Neck Cancer
  • Kidney Cancer
  • Liver Cancer
  • Lung Cancer
  • Mouth Cancer
  • Mesothelioma
  • Multiple Myeloma
  • Neuroendocrine Tumors
  • Ovarian Cancer
  • Pancreatic Cancer
  • Prostate Cancer
  • Skin Cancer/Melanoma
  • Stomach Cancer
  • Testicular Cancer
  • Throat Cancer
  • Thyroid Cancer
  • Prevention and Screening
  • Diagnosis and Treatment
  • Research and Clinical Trials
  • Survivorship

new research on lymphedema

Request an appointment at Mayo Clinic

new research on lymphedema

A promising new treatment for lymphedema

Share this:.

Share to facebook

By Susan Buckles

Lymphedema is most often associated with cancer treatment, although some cases of lymphedema are congenital. During cancer treatment, lymph nodes may be damaged or removed, and the lymphatic fluid no longer drains. Then lymphedema can develop months or years after cancer treatment.

Research by  Antonio Forte, M.D., Ph.D. , a Mayo Clinic plastic surgeon, seeks to better pinpoint who is most likely to benefit from surgery aimed at regenerating a faulty lymphatic system.

"Without surgery, lymphedema gets progressively worse due to accumulation of the lymphatic fluid in tissue and the chronic changes that causes. The upper and lower extremities can feel heavy, and the affected area is more prone to infections. Wounds take longer to heal, and some people lose dexterity to the point that it's hard to put a shoe on," says Dr. Forte.

Surgical options at Mayo Clinic

Mayo Clinic is a leader in surgical options to reverse the damage from lymphedema. Dr. Forte specializes in lymphovenous bypass, a microsurgery done under powerful microscopes that are magnified 20 to 25 times. Through an incision no larger than a paper cut, the surgery connects tiny lymphatic vessels smaller than a strand of hair to tiny veins, creating a type of detour around the damaged area. The new vessel connections restore the body's ability to drain lymphatic fluids.

"When you remove that fluid, we don't know why, but the body is able to reverse these chronic changes in the tissue. In a way, what we're trying to do is reverse the damage that was done by the accumulation of the lymphatic fluid," says Dr. Forte. "It is a regenerative approach because not only do you restore function, but you actually heal the tissues."

Like a ton of bricks

Rebecca, who asked that her last name not be used, was diagnosed with triple-negative breast cancer in 2009. She prayed she'd live long enough to see her two youngest sons graduate college.

The Savannah, Georgia, resident waged a grueling battle that included chemotherapy, mastectomy and lymph node removal. She emerged victorious and was able to not only attend her sons' graduations, but also their weddings. However, a noxious side effect surfaced four years after her cancer treatment. Lymphedema developed in her right arm, causing swelling and fibrous tissue that progressively got harder.

"I felt as if I was dragging a ton of bricks everywhere I went. I could not go away to visit family without being sure I had all materials and compression garments with me. I had to make a checklist to include everything I needed to care for myself anywhere I went," she says.

Occupational therapy, compression wraps and a disciplined exercise regimen helped control her lymphedema, but it never really went away. A devastating reality set in when she realized that no matter how closely she followed medical advice, the lymphedema would not be cured.

9 was the lucky number

By luck, coincidence or maybe even a miracle, Rebecca discovered a new path to treatment while at the ninth green of the Players Championship at TPC Sawgrass in Ponte Vedra Beach, Florida. She and her family had gathered at the ninth green to watch a golfer sink a putt. Rebecca's right hand had swollen during the day.

"I had left my compression glove at home," she says.

She decided to stand in the shade and rest her hand on a tree to see if the swelling would go down. While standing there, with her right hand above her head resting on an ancient live oak tree, a nearby spectator noticed the compression sleeve.

"She said: 'Excuse me. I see you are a lymphedema patient. Did you know that Mayo Clinic offers lymphovenous bypass surgery?' This woman turned out to be a plastic surgery nurse practitioner at Mayo Clinic in Florida. I had not heard about the surgery until our chance meeting," says Rebecca.

Despite her initial apprehension, preparations for surgery started falling in place. Dr. Forte examined her and found the procedure to be a suitable match, and her insurance approved. One year after lymphovenous bypass surgery, Rebecca says the fluid has drained and her right arm is nearly normal.

"I can now feel the bone in my elbow. I hadn't felt that bone in three years," she says. "This surgery has taken a weight, ball and chain and a ton of bricks off my life. Resentment, fear and worry also have been removed. I truly do not have words to express the depth of gratitude and appreciation I have for Dr. Forte and his nurse practitioner. I weep with private tears of joy and relief."

While surgery relieved Rebecca's symptoms, Dr. Forte says there is a chance it might not work the same for others. However, he believes lymphovenous bypass is a safe and low-risk procedure.

"I love the bypass because the morbidity is so low that there's little downside to the procedure. If lymphovenous surgery doesn't relieve fluid buildup, then all you have to heal from is small incisions similar to paper cuts. If it does work, it's life-changing," he says.

By better understanding how each patient responds to the surgery, Dr. Forte hopes to improve the success rate. He also hopes to learn whether a targeted drug or complementary treatment might lead to quicker recovery.

A version of this story originally appeared on the Mayo Clinic Center for Regenerative Medicine blog .

Learn more about lymphedema .

Find a lymphedema clinical trial at Mayo Clinic .

Mayo Clinic's Center for Individualized Medicine is funding research to identify new biomarkers that could predict which patients are most likely to respond to this regenerative surgery.

Related Posts

new research on lymphedema

New Mayo Clinic research represents a promising step toward identifying biological signals or biomarkers that may aid in early detection.

new research on lymphedema

Study results showed significantly higher survival rates when blinatumomab was added to chemotherapy for people with B-cell acute lymphoblastic leukemia.

new research on lymphedema

Research shows screening protocols fail to catch a notable number of people carrying genetic mutations associated with increased cancer risk.

Study identifies potential drug target for lymphedema

  • Download PDF Copy

The human lymphatic system consists of a vast network of vessels that drain protein-rich fluid in tissues and transport it to lymph nodes. When the machinery goes awry and the lymphatic vessels don't drain properly, the fluid accumulates in the tissues, leading to painful swelling known as lymphedema .

A Cornell-led collaboration built a 3D in-vitro model of a functional human lymphatic vessel that revealed a surprising mechanism that can jam up the necessary drainage: a protein expressed in lymphatic endothelial cells called rho-associated protein kinase 2 (ROCK2).

The researchers demonstrated that by inhibiting ROCK2 they can reverse the effects of lymphedema, creating a potential treatment for a condition that is estimated to affect up to 150 million people worldwide.

The team's paper, "A 3D Biomimetic Model of Lymphatics Reveals Cell-Cell Junction Tightening and Lymphedema Via a Cytokine-Induced ROCK2/JAM-A Complex," published Oct. 2 in Proceedings of the National Academy of Sciences . The paper's lead author is Esak (Isaac) Lee, Meinig Family Investigator in the Life Sciences and assistant professor of biomedical engineering at Cornell Engineering.

Lymphedema has so many patients in the world. Doctors usually suggest you wear a compression garment or do some physical therapy, like massage, to pump out all these fluids from your arms and legs. Unfortunately, there's no FDA-approved drug because we don't understand the mechanism of this disease." Esak (Isaac) Lee, Meinig Family Investigator in the Life Sciences and assistant professor of biomedical engineering at Cornell Engineering

Attempts to locate that mechanism in humans have been thwarted by the fact that the lymphatic system is entangled with the central nervous system, muscle movements, and a raft of other bodily processes.

Working with researchers from Boston University (BU) and Harvard Medical School who were led by co-lead author Christopher Chen of BU, Lee set out to create an in-vitro model – or "lymphatics-on-a-chip" – that could isolate several biological and biophysical factors, such as inflammatory cytokines, ROCK2 signal and interstitial fluid pressure, while mimicking the drainage.

The transparent, thumb-sized device realistically reproduced the "button-like" lymphatic junction – the borderline between the endothelial cells that line the lymphatic vessels – with two hollow channels in 3D collagen. In the first channel, the team seeded these human cells, which were then pressurized by interstitial fluid loaded in the second channel. High fluid pressure opened the junctions of the engineered lymphatics, and the uptake of fluid closed them. The researchers could then measure how much of the fluid drained through the system.

"Demonstrating this button-like junction is so crucial to recapitulate lymphatic function, which has not been successful in vitro before our study," Lee said. "The most important challenge was to make the right conditions: the size of the vessels, the distance between two channels, growth factors used, and the size of interstitial fluid pressure. All these things are really affecting button junctions, so we wanted to make sure they all are realistically mimicking the human biology."

Related Stories

  • Researchers develop new method to study critical type of protein modification process
  • New technique reveals abnormalities in motor neurone disease
  • DeepPBS: A geometric AI model for accurate prediction of protein-DNA binding specificity

The researchers introduced inflammatory cytokines that are known to be expressed in lymphedema patients, such as interleukin-2 and granulocyte-macrophage colony-stimulating factor (GM-CSF), which tighten the lymphatic junctions, leading to fluid buildup and lymphedema.

While these cytokines had been previously known to disrupt blood vessel junctions, the model revealed they were actually tightening the junctions between lymphatic endothelial cells and impeding drainage.

When the researchers inhibited ROCK2, the lymphatic junctions loosened and the blood vessel junctions tightened under inflammation, so that normal fluid drainage could resume.

"There are upwards of 170 to 180 different pan-ROCK inhibitors, but they generally come with serious side effects, such as hypotension, when these ROCK inhibitors block both ROCK1 and ROCK2, two isoforms of ROCK," Lee said. "The side effects of inhibiting ROCK2, however, are minimal, because ROCK2 is more expressed in lymphatic cells than vascular muscle cells in blood vessels, where ROCK1 is highly expressed. This makes it a strong candidate for therapeutics that target lymphatic disease with less vascular toxicity."

Lee's collaborators reproduced the experiment in mice that lose ROCK2 in their lymphatic cells, which showed a dramatic reduction of edema in the animals' tails.

Co-authors include doctoral student Anna Kolarzyk, former postdoctoral researcher Tae Joon Kwak, and researchers from Boston University and Harvard Medical School.

The research was supported by the National Institutes of Health, the National Science Foundation and the Wellcome Leap HOPE (Human Organs Physiology and Engineering) program.

Cornell University

Posted in: Medical Science News | Medical Research News | Medical Condition News

Tags: Blood , Blood Vessel , Blood Vessels , Cell , Central Nervous System , CHIP , Collagen , Cytokine , Cytokines , Edema , Hypotension , in vitro , Inflammation , Interleukin , Interleukin-2 , Kinase , Lymph Nodes , Lymphatic Disease , Lymphatic System , Lymphedema , Macrophage , Medical School , Muscle , Nervous System , OCT , Physical Therapy , Physiology , Protein , Research , Therapeutics , Vascular

Suggested Reading

Mrc1 protein identified as key to maintaining cell identity during division

Cancel reply to comment

  • Trending Stories
  • Latest Interviews
  • Top Health Articles

Dietary patterns like fasting and the Mediterranean diet unlock molecular secrets to healthy aging

How can microdialysis benefit drug development

Ilona Vuist

In this interview, discover how Charles River uses the power of microdialysis for drug development as well as CNS therapeutics.

How can microdialysis benefit drug development

Global and Local Efforts to Take Action Against Hepatitis

Lindsey Hiebert and James Amugsi

In this interview, we explore global and local efforts to combat viral hepatitis with Lindsey Hiebert, Deputy Director of the Coalition for Global Hepatitis Elimination (CGHE), and James Amugsi, a Mandela Washington Fellow and Physician Assistant at Sandema Hospital in Ghana. Together, they provide valuable insights into the challenges, successes, and the importance of partnerships in the fight against hepatitis.

Global and Local Efforts to Take Action Against Hepatitis

Addressing Important Cardiac Biology Questions with Shotgun Top-Down Proteomics

In this interview conducted at Pittcon 2024, we spoke to Professor John Yates about capturing cardiomyocyte cell-to-cell heterogeneity via shotgun top-down proteomics.

Addressing Important Cardiac Biology Questions with Shotgun Top-Down Proteomics

Latest News

Cannabidiol shown to extend lifespan and improve symptoms in Leigh syndrome models

Newsletters you may be interested in

Medical Device

Your AI Powered Scientific Assistant

Hi, I'm Azthena, you can trust me to find commercial scientific answers from News-Medical.net.

A few things you need to know before we start. Please read and accept to continue.

  • Use of “Azthena” is subject to the terms and conditions of use as set out by OpenAI .
  • Content provided on any AZoNetwork sites are subject to the site Terms & Conditions and Privacy Policy .
  • Large Language Models can make mistakes. Consider checking important information.

Great. Ask your question.

Azthena may occasionally provide inaccurate responses. Read the full terms .

While we only use edited and approved content for Azthena answers, it may on occasions provide incorrect responses. Please confirm any data provided with the related suppliers or authors. We do not provide medical advice, if you search for medical information you must always consult a medical professional before acting on any information provided.

Your questions, but not your email details will be shared with OpenAI and retained for 30 days in accordance with their privacy principles.

Please do not ask questions that use sensitive or confidential information.

Read the full Terms & Conditions .

Provide Feedback

new research on lymphedema

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

The Future of Lymphedema: Potential Therapeutic Targets for Treatment

Affiliation.

  • 1 Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 USA.
  • PMID: 37359311
  • PMCID: PMC10233555
  • DOI: 10.1007/s12609-023-00491-5

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: ACE inhibitors; CD4 +; Captopril; Doxycycline; Lymphedema; NSAID; Non-steroidal anti-inflammatory drugs; Pirfenidone; TGFB; TH2 cells; Tacrolimus; Tetracyclines; VEGF-C; Vascular endothelial growth factor C.

© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2023. Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

PubMed Disclaimer

Conflict of interest statement

Conflict of InterestBabak 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.

Similar articles

  • Pharmacological Treatment of Secondary Lymphedema. Brown S, Dayan JH, Coriddi M, Campbell A, Kuonqui K, Shin J, Park HJ, Mehrara BJ, Kataru RP. Brown S, et al. Front Pharmacol. 2022 Jan 25;13:828513. doi: 10.3389/fphar.2022.828513. eCollection 2022. Front Pharmacol. 2022. PMID: 35145417 Free PMC article. Review.
  • Experimental assessment of pro-lymphangiogenic growth factors in the treatment of post-surgical lymphedema following lymphadenectomy. Baker A, Kim H, Semple JL, Dumont D, Shoichet M, Tobbia D, Johnston M. Baker A, et al. Breast Cancer Res. 2010;12(5):R70. doi: 10.1186/bcr2638. Epub 2010 Sep 8. Breast Cancer Res. 2010. PMID: 20825671 Free PMC article.
  • VEGF-C and VEGF-C156S in the pro-lymphangiogenic growth factor therapy of lymphedema: a large animal study. Visuri MT, Honkonen KM, Hartiala P, Tervala TV, Halonen PJ, Junkkari H, Knuutinen N, Ylä-Herttuala S, Alitalo KK, Saarikko AM. Visuri MT, et al. Angiogenesis. 2015 Jul;18(3):313-26. doi: 10.1007/s10456-015-9469-2. Epub 2015 May 28. Angiogenesis. 2015. PMID: 26018927
  • Topical Approach to Delivering Targeted Therapies in Lymphedema Treatment: A Systematic Review. Forte AJ, Boczar D, Huayllani MT, McLaughlin SA, Bagaria S. Forte AJ, et al. Cureus. 2019 Dec 1;11(12):e6269. doi: 10.7759/cureus.6269. Cureus. 2019. PMID: 31815081 Free PMC article. Review.
  • Abnormal lymphatic S1P signaling aggravates lymphatic dysfunction and tissue inflammation. Kim D, Tian W, Wu TT, Xiang M, Vinh R, Chang J, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Kim D, et al. medRxiv [Preprint]. 2023 Jun 12:2023.06.08.23291175. doi: 10.1101/2023.06.08.23291175. medRxiv. 2023. Update in: Circulation. 2023 Oct 17;148(16):1231-1249. doi: 10.1161/CIRCULATIONAHA.123.064181. PMID: 37398237 Free PMC article. Updated. Preprint.
  • Vascular Endothelial Growth Factor C (VEGF-C) Sensitizes Lymphatic Endothelial Cells to Oxidative-Stress-Induced Apoptosis through DNA Damage and Mitochondrial Dysfunction: Implications for Lymphedema. Hossain L, Gomes KP, Yang X, Liu E, Du Toit J, von der Weid PY, Gibson SB. Hossain L, et al. Int J Mol Sci. 2024 Jul 17;25(14):7828. doi: 10.3390/ijms25147828. Int J Mol Sci. 2024. PMID: 39063073 Free PMC article.
  • Molecular pathophysiology of secondary lymphedema. Lee SO, Kim IK. Lee SO, et al. Front Cell Dev Biol. 2024 Jul 8;12:1363811. doi: 10.3389/fcell.2024.1363811. eCollection 2024. Front Cell Dev Biol. 2024. PMID: 39045461 Free PMC article. Review.
  • The Role of Inflammation in Lymphedema: A Narrative Review of Pathogenesis and Opportunities for Therapeutic Intervention. Bowman C, Rockson SG. Bowman C, et al. Int J Mol Sci. 2024 Mar 31;25(7):3907. doi: 10.3390/ijms25073907. Int J Mol Sci. 2024. PMID: 38612716 Free PMC article. Review.
  • Lymphatic System and the Kidney: From Lymphangiogenesis to Renal Inflammation and Fibrosis Development. Stasi E, Sciascia S, Naretto C, Baldovino S, Roccatello D. Stasi E, et al. Int J Mol Sci. 2024 Mar 1;25(5):2853. doi: 10.3390/ijms25052853. Int J Mol Sci. 2024. PMID: 38474100 Free PMC article. Review.
  • Le lymphœdème lié au cancer. Chaput G, Ibrahim M. Chaput G, et al. Can Fam Physician. 2023 Oct;69(10):e206-e210. doi: 10.46747/cfp.6910e206. Can Fam Physician. 2023. PMID: 37833084 Free PMC article. French. No abstract available.
  • 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
  • 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 - PubMed
  • 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
  • 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
  • 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

Publication types

  • Search in MeSH

Related information

Grants and funding.

  • P30 CA008748/CA/NCI NIH HHS/United States
  • R01 CA278599/CA/NCI NIH HHS/United States

LinkOut - more resources

Full text sources.

  • Europe PubMed Central
  • PubMed Central

Research Materials

  • NCI CPTC Antibody Characterization Program

Miscellaneous

  • NCI CPTAC Assay Portal
  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

U.S. flag

An official website of the United States government

Here’s how you know

Official websites use .gov A .gov website belongs to an official government organization in the United States.

Secure .gov websites use HTTPS A lock ( A locked padlock ) or https:// means you’ve safely connected to the .gov website. Share sensitive information only on official, secure websites.

  • Heart-Healthy Living
  • High Blood Pressure
  • Sickle Cell Disease
  • Sleep Apnea
  • Information & Resources on COVID-19
  • The Heart Truth®
  • Learn More Breathe Better®
  • Blood Diseases & Disorders Education Program
  • Publications and Resources
  • Clinical Trials
  • Blood Disorders and Blood Safety
  • Sleep Science and Sleep Disorders
  • Lung Diseases
  • Health Disparities and Inequities
  • Heart and Vascular Diseases
  • Precision Medicine Activities
  • Obesity, Nutrition, and Physical Activity
  • Population and Epidemiology Studies
  • Women’s Health
  • Research Topics
  • All Science A-Z
  • Grants and Training Home
  • Policies and Guidelines
  • Funding Opportunities and Contacts
  • Training and Career Development
  • Email Alerts
  • NHLBI in the Press
  • Research Features
  • Ask a Scientist
  • Past Events
  • Upcoming Events
  • Mission and Strategic Vision
  • Divisions, Offices and Centers
  • Advisory Committees
  • Budget and Legislative Information
  • Jobs and Working at the NHLBI
  • Contact and FAQs
  • NIH Sleep Research Plan
  • News and Events
  • < Back To All News

Advances and challenges in treating lymphedema: A Q and A with Dhruv Singhal, M. D.

Intra Operative photo of Dr. Singhal M.D.

Q: How do problems with the lymphatic system end up causing lymphedema?

A: Normally as blood flows through the body, fluid seeps out of blood vessels and into the body’s tissues. This fluid drains into the lymphatic vessels, then moves to the lymph nodes to be filtered. Eventually the fluid returns to the veins and back into circulation. In lymphedema, however, the lymphatic system fails to keep up with the leaking fluid. That’s because the lymphatic vessels – the places where they’d normally drain – have been damaged. As a result, this fluid builds up and causes swelling, typically in an arm or a leg, and can result in chronic inflammation. Damage to the lymphatic vessels most commonly occurs because of cancer treatment. A surgeon will often remove the lymph nodes near a tumor to prevent the spread of cancer. In that process, lymphatic vessels get severed, and this is what can start the problem.

Q: What do you find most interesting about the lymphatic system?

A: One of the most interesting things about the lymphatic system is its scale, which is remarkably small compared to the blood vascular system (arteries and veins). To give you an idea, the largest artery, the aorta, is about 2 centimeters (cm). So is the largest vein, the inferior vena cava. But the largest lymphatic vessel, the thoracic duct, is only about 2 millimeters (mm), or 10 times smaller. The lymphatic system can almost seem invisible. If you cut yourself and damage an artery or vein, red blood comes out. If you cut 10 lymphatic vessels, you might not even notice, though it would slowly leak clear fluid. Yet the lymphatic system transports as much as 12 liters of fluid a day, whereas the arteries and veins hold only about 5 liters. So really, the lymphatic system is this vast network containing much of the body’s vascular fluid, but often we don’t notice it until something goes wrong.

Q: How is lymphedema treated, and is there a cure?

A: There is currently no cure for lymphedema. The standard treatment is called compression. A patient wears a pressurized sleeve over the arm or leg to compress it, and that helps the fluid drain. The problem is that it’s not feasible to wear these sleeves 24/7. And you can imagine it’s not very comfortable to wear them when you live in places like Florida, or anywhere during the summer. Of course, there are medicines and other approaches that can help. We can also do lymphatic reconstructive surgery, like a lymph node transplant. In that process we take lymph nodes from other parts of the body and transplant them into the affected region. But I think the most promising approach going forward will be prevention.

Q: What kind of prevention do you think is possible, and what will it take to put those approaches into practice broadly?

A: One of the biggest challenges to prevention and treatment of lymphedema is that we don’t fully understand the risk factors. Of four patients undergoing lymph node removal during cancer treatment, only one will develop lymphedema; the risk is about 26-33%. We think part of the answer lies in the small, but different ways lymphatic vessels form in one person to the next. We think this variability may protect some patients from developing lymphedema. A key to this approach will be understanding what normal lymphatic anatomy looks like. If we can accurately map the lymphatic vessels in healthy individuals as well as in patients before and after surgery, we may be able to identify patterns in lymphatic structure that are more likely to lead to lymphedema.

The second major hurdle in treating this disease is the lack of an objective measurement of lymphatic function. One of the biggest things holding back lymphatic care is that there is nothing we can do right now to get a quick assessment of the lymphatic system. If you have chest pain, within an hour we can have sophisticated measurements of heart function. Nothing like that exists right now for measuring lymphatic function. Measuring limb circumference is often used as an indicator of lymphedema, but this can be complicated by other factors and doesn’t give real-time information about lymphatic function. Techniques using radioactive tracers or magnetic resonance imaging (MRI) can be used to measure lymphatic function and structure, but they require specialized instruments and result in poor resolution (ability to see detail). These techniques are also difficult to use at the point of surgery, which would be ideal for determining whether a patient needs immediate intervention or not.

Q: How is your research group tackling these problems, and what do your results mean for lymphedema treatment going forward?

A: Firstly, we think it’s critical to take a step back and try to understand what normal lymphatic anatomy is. I often get asked, “Why not just study the patients who have lymphedema?” But very little has been done to look at lymphatic anatomy in healthy people. Once we’ve done that, we want to know how the anatomy changes in those who do and do not develop lymphedema after lymph nodes have been removed. In those patients who have lymph node damage but don’t develop lymphedema, if we can map how they are able to compensate, perhaps we can come up with a solution for those who do get lymphedema.

We also want to develop better tools and methods for evaluating lymphatic function. We’re working with Dr. Hak Soo Choi in the Gordon Center for Medical Imaging at Massachusetts General Hospital to develop dyes that are specific to the lymphatic system. We could then inject these dyes into the body to see and measure how the dye is being carried away by the lymphatic vessels. This would give us a readout of the efficiency of lymphatic drainage. It would also give us a measurement that we can use to evaluate surgical and medical interventions, and then objectively say which ones improved lymphatic function and which did not.

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 21 October 2021

Primary lymphoedema

  • Pascal Brouillard 1 ,
  • Marlys H. Witte   ORCID: orcid.org/0000-0001-5505-934X 2 ,
  • Robert P. Erickson 3 ,
  • Robert J. Damstra 4 ,
  • Corinne Becker 5 ,
  • Isabelle Quéré   ORCID: orcid.org/0000-0002-1492-9764 6 &
  • Miikka Vikkula   ORCID: orcid.org/0000-0002-6236-338X 1 , 7 , 8  

Nature Reviews Disease Primers volume  7 , Article number:  77 ( 2021 ) Cite this article

3749 Accesses

41 Citations

53 Altmetric

Metrics details

  • Disease genetics
  • Vascular diseases

Lymphoedema is the swelling of one or several parts of the body owing to lymph accumulation in the extracellular space. It is often chronic, worsens if untreated, predisposes to infections and causes an important reduction in quality of life. Primary lymphoedema (PLE) is thought to result from abnormal development and/or functioning of the lymphatic system, can present in isolation or as part of a syndrome, and can be present at birth or develop later in life. Mutations in numerous genes involved in the initial formation of lymphatic vessels (including valves) as well as in the growth and expansion of the lymphatic system and associated pathways have been identified in syndromic and non-syndromic forms of PLE. Thus, the current hypothesis is that most cases of PLE have a genetic origin, although a causative mutation is identified in only about one-third of affected individuals. Diagnosis relies on clinical presentation, imaging of the structure and functionality of the lymphatics, and in genetic analyses. Management aims at reducing or preventing swelling by compression therapy (with manual drainage, exercise and compressive garments) and, in carefully selected cases, by various surgical techniques. Individuals with PLE often have a reduced quality of life owing to the psychosocial and lifelong management burden associated with their chronic condition. Improved understanding of the underlying genetic origins of PLE will translate into more accurate diagnosis and prognosis and personalized treatment.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 1 digital issues and online access to articles

111,21 € per year

only 111,21 € per issue

Buy this article

  • Purchase on SpringerLink
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

new research on lymphedema

Similar content being viewed by others

new research on lymphedema

Lymphatic vessel: origin, heterogeneity, biological functions, and therapeutic targets

new research on lymphedema

Lower extremity lymphatic function predicted by body mass index: a lymphoscintigraphic study of obesity and lipedema

new research on lymphedema

A new severity classification of lower limb secondary lymphedema based on lymphatic pathway defects in an indocyanine green fluorescent lymphography study

Starling, E. H. On the absorption of fluids from the connective tissue spaces. J. Physiol. 19 , 312–326 (1896).

CAS   PubMed   PubMed Central   Google Scholar  

Levick, J. R. & Michel, C. C. Microvascular fluid exchange and the revised Starling principle. Cardiovasc. Res. 87 , 198–210 (2010).

CAS   PubMed   Google Scholar  

Quere, I., Nagot, N. & Vikkula, M. Incidence of cellulitis among children with primary lymphedema. N. Engl. J. Med. 378 , 2047–2048 (2018).

PubMed   Google Scholar  

Moffatt, C. J. et al. Prevalence and risk factors for chronic edema in U.K. community nursing services. Lymphat. Res. Biol. 17 , 147–154 (2019).

PubMed   PubMed Central   Google Scholar  

Moffatt, C. J. et al. Lymphoedema: an underestimated health problem. QJM 96 , 731–738 (2003).

Moffatt, C., Keeley, V. & Quere, I. The concept of chronic edema — a neglected public health issue and an international response: The LIMPRINT Study. Lymphat. Res. Biol. 17 , 121–126 (2019).

Rockson, S. G. & Rivera, K. K. Estimating the population burden of lymphedema. Ann. N. Y. Acad. Sci. 1131 , 147–154 (2008).

Keast, D. H., Despatis, M., Allen, J. O. & Brassard, A. Chronic oedema/lymphoedema: under-recognised and under-treated. Int. Wound J. 12 , 328–333 (2015).

DiSipio, T., Rye, S., Newman, B. & Hayes, S. Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. Lancet Oncol. 14 , 500–515 (2013).

Mortimer, P. S. & Rockson, S. G. New developments in clinical aspects of lymphatic disease. J. Clin. Invest. 124 , 915–921 (2014).

Rockson, S. G., Keeley, V., Kilbreath, S., Szuba, A. & Towers, A. Cancer-associated secondary lymphoedema. Nat. Rev. Dis. Prim. 5 , 22 (2019). This recent detailed review explores various aspects of secondary lymphoedema, which, in developed countries, mostly results from the treatment of cancer as opposed to infection-related secondary lymphoedema in developing countries.

Mercier, G., Pastor, J., Moffatt, C., Franks, P. & Quere, I. LIMPRINT: health-related quality of life in adult patients with chronic edema. Lymphat. Res. Biol. 17 , 163–167 (2019). This large multicentre study prospectively assessed the health-related quality of life of 1,094 adult patients with chronic oedema and underscored a poor disease-specific and generic health-related quality of life.

Lopez, M., Roberson, M. L., Strassle, P. D. & Ogunleye, A. Epidemiology of lymphedema-related admissions in the United States: 2012–2017. Surg. Oncol. 35 , 249–253 (2020).

Biesecker, L. G. et al. A dyadic approach to the delineation of diagnostic entities in clinical genomics. Am. J. Hum. Genet. 108 , 8–15 (2021).

Gordon, K. et al. Update and audit of the St George’s classification algorithm of primary lymphatic anomalies: a clinical and molecular approach to diagnosis. J. Med. Genet. 57 , 653–659 (2020). This review proposes an updated clinical classification algorithm for primary lymphoedema to assist diagnostic workup and patient management, based on age of onset, areas affected by swelling and associated clinical features, in agreement with the International Society for the Study of Vascular Anomalies 2018 classification.

Nonne, M. & Vier Fälle, V. Elephantiasis congenita hereditarian. Arch. für pathologische anatomie und physiologie und für klinische Med. 125 , 189–196 (1891).

Google Scholar  

Milroy, W. F. An undescribed variety of hereditary oedema. N. Y. Med. J. 56 , 505–508 (1892).

Samman, P. D. & White, W. F. The “yellow nail” syndrome. Br. J. Dermatol. 76 , 153–157 (1964).

Meige, H. Distrophie oedemateuse héréditaire. Presse Méd. 6 , 341–343 (1898).

Irrthum, A., Karkkainen, M. J., Devriendt, K., Alitalo, K. & Vikkula, M. Congenital hereditary lymphedema caused by a mutation that inactivates VEGFR3 tyrosine kinase. Am. J. Hum. Genet. 67 , 295–301 (2000).

Karkkainen, M. J. et al. Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema. Nat. Genet. 25 , 153–159 (2000).

Mendola, A. et al. Mutations in the VEGFR3 signaling pathway explain 36% of familial lymphedema. Mol. Syndromol. 4 , 257–266 (2013).

Leppanen, V. M. et al. Characterization of ANGPT2 mutations associated with primary lymphedema. Sci. Transl Med. 12 , eaax8013 (2020). This is the most recent discovery of a novel gene causing primary lymphoedema with functional validation of the mutations in vitro and in a mouse model, underscoring the heterogeneity within genetic causes of PLE .

Iacobas, I. et al. Multidisciplinary guidelines for initial evaluation of complicated lymphatic anomalies-expert opinion consensus. Pediatr. Blood Cancer 67 , e28036 (2020).

Quinn, A. M., Valcarcel, B. N., Makhamreh, M. M., Al-Kouatly, H. B. & Berger, S. I. A systematic review of monogenic etiologies of nonimmune hydrops fetalis. Genet. Med. 23 , 3–12 (2021). This systematic literature review of non-immune hydrops fetalis pinpointed 131 genes with strong evidence for an association with NIHF and 46 genes with emerging evidence, spanning the spectrum of multisystemic syndromes and cardiac, haematological and metabolic disorders.

Smeltzer, D. M., Stickler, G. B. & Schirger, A. Primary lymphedema in children and adolescents: a follow-up study and review. Pediatrics 76 , 206–218 (1985).

Schook, C. C. et al. Primary lymphedema: clinical features and management in 138 pediatric patients. Plast. Reconstr. Surg. 127 , 2419–2431 (2011).

Fastre, E. et al. Splice-site mutations in VEGFC cause loss of function and Nonne-Milroy-like primary lymphedema. Clin. Genet. 94 , 179–181 (2018).

Erickson, R. P. et al. Sex-limited penetrance of lymphedema to females with CELSR1 haploinsufficiency: a second family. Clin. Genet. 96 , 478–482 (2019).

Soo, J. K., Bicanic, T. A., Heenan, S. & Mortimer, P. S. Lymphatic abnormalities demonstrated by lymphoscintigraphy after lower limb cellulitis. Br. J. Dermatol. 158 , 1350–1353 (2008).

Au, A. C. et al. Protein tyrosine phosphatase PTPN14 is a regulator of lymphatic function and choanal development in humans. Am. J. Hum. Genet. 87 , 436–444 (2010).

Hong, S. E. et al. Autosomal recessive lissencephaly with cerebellar hypoplasia is associated with human RELN mutations. Nat. Genet. 26 , 93–96 (2000).

Brice, G. et al. A novel mutation in GJA1 causing oculodentodigital syndrome and primary lymphoedema in a three generation family. Clin. Genet. 84 , 378–381 (2013).

Gumus, E. A rare symptom of a very rare disease: a case report of a oculodentodigital dysplasia with lymphedema. Clin. Dysmorphol. 27 , 91–93 (2018).

Schlogel, M. J. et al. No evidence of locus heterogeneity in familial microcephaly with or without chorioretinopathy, lymphedema, or mental retardation syndrome. Orphanet J. Rare Dis. 10 , 52 (2015).

Commission, E. U. Useful Information on Rare Diseases from EU Perspective . C 151/157–C 151/110 (European Commission 2009).

Park, S. I. et al. Prevalence and epidemiological factors involved in cellulitis in Korean patients with lymphedema. Ann. Rehabil. Med. 40 , 326–333 (2016).

Aslam, A. F., Aslam, A. K., Qamar, M. U. & Levey, R. Primary lymphedema tarda in an 88-year-old African-American male. J. Natl Med. Assoc. 97 , 1031–1035 (2005).

Ibrahim, A. Primary lymphedema tarda. Pan Afr. Med. J. 19 , 16 (2014).

Davey, S. L. et al. The South African multi-disciplinary lymphoedema position statement. Wound Healing South. Afr. 11 , 21–24 (2018).

Julkowska, D. et al. The importance of international collaboration for rare diseases research: a European perspective. Gene Ther. 24 , 562–571 (2017).

Vignes, S. et al. Primary lymphedema French National Diagnosis and Care Protocol (PNDS; Protocole National de Diagnostic et de Soins). Orphanet J. Rare Dis. 16 , 18 (2021).

Nedergaard, M. & Goldman, S. A. Glymphatic failure as a final common pathway to dementia. Science 370 , 50–56 (2020).

Proulx, S. T. Cerebrospinal fluid outflow: a review of the historical and contemporary evidence for arachnoid villi, perineural routes, and dural lymphatics. Cell Mol. Life Sci. 78 , 2429–2457 (2021).

Witte, M. & Bernas, M. in Rutherford’s Vascular Surgery and Endovascular Therapy (eds Sidaway, A. & Perler, B.) Ch. 10, 105–122 (Elsevier, 2019).

Itkin, M. et al. Research priorities in lymphatic interventions: recommendations from a multidisciplinary research consensus panel. J. Vasc. Interv. Radiol. 32 , 762.e1–762.e7 (2021). This document, by a selected panel of experts in lymphatic medicine from the USA, New Zealand and Korea, identified seven priorities for research in the field, including lymphatic decompression in patients with congestive heart failure, detoxification of thoracic duct lymph in acute illness, development of newer agents for lymphatic imaging, characterization of organ-based lymph composition, and development of lymphatic interventions to treat ascites in liver cirrhosis.

Schwartz, F. R. et al. Lymphatic imaging: current noninvasive and invasive techniques. Semin. Intervent. Radiol. 37 , 237–249 (2020).

Kinmonth, J. B. in The Lymphatic: Disease, Lymphography, and Surgery 114–155 (Edward Arnold, 1972).

Witte, M. H. et al. Structure function relationships in the lymphatic system and implications for cancer biology. Cancer Metastasis Rev. 25 , 159–184 (2006).

Földi, M. & Földi, E. in Földi’s Textbook of Lymphology (eds Földi, M. & Földi, E.) Ch. 2, 135–273 (Urban & Fischer Verlag, 2012).

Executive Committee.The diagnosis and treatment of peripheral lymphedema: 2016 consensus document of the international society of lymphology. Lymphology 49 , 170–184 (2016).

Witte, M. H., Dumont, A. E., Cole, W. R., Witte, C. L. & Kintner, K. Lymph circulation in hepatic cirrhosis: effect of portacaval shunt. Ann. Intern. Med. 70 , 303–310 (1969).

Baish, J. W., Netti, P. A. & Jain, R. K. Transmural coupling of fluid flow in microcirculatory network and interstitium in tumors. Microvasc. Res. 53 , 128–141 (1997).

Michel, C. C., Woodcock, T. E. & Curry, F. E. Understanding and extending the Starling principle. Acta Anaesthesiol. Scand. 64 , 1032–1037 (2020).

Lee, B. B. et al. Diagnosis and treatment of primary lymphedema. Consensus document of the International Union of Phlebology (IUP)-2013. Int. Angiol. 32 , 541–574 (2013).

Baluk, P. et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J. Exp. Med. 204 , 2349–2362 (2007).

Nonomura, K. et al. Mechanically activated ion channel PIEZO1 is required for lymphatic valve formation. Proc. Natl Acad. Sci. USA 115 , 12817–12822 (2018).

Teijeira, A. et al. Lymphatic endothelium forms integrin-engaging 3D structures during DC transit across inflamed lymphatic vessels. J. Invest. Dermatol. 133 , 2276–2285 (2013).

Johnson, L. A. et al. Dendritic cells enter lymph vessels by hyaluronan-mediated docking to the endothelial receptor LYVE-1. Nat. Immunol. 18 , 762–770 (2017).

Executive Committee.The diagnosis and treatment of peripheral lymphedema: 2020 Consensus Document of the International Society of Lymphology. Lymphology 53 , 3–19 (2020). This recent document integrates the broad spectrum of protocols and practices advocated around the world for the diagnosis and treatment of peripheral lymphoedema. It provides a current “Consensus view” of the international community based on various levels of evidence.

Witte, M. H. et al. Phenotypic and genotypic heterogeneity in familial Milroy lymphedema. Lymphology 31 , 145–155 (1998).

Witte, C. L. et al. Advances in imaging of lymph flow disorders. Radiographics 20 , 1697–1719 (2000).

Campisi, C., Boccardo, F., Witte, M. H. & Bernas, M. in Venous and Lymphatic Diseases (eds Dieter, R. S. Jr & Dieter, R. A. III) Ch. 42, 607–629 (McGraw Hill, 2011).

Itkin, M. & Nadolski, G. J. Modern techniques of lymphangiography and interventions: current status and future development. Cardiovasc. Intervent Radiol. 41 , 366–376 (2018).

Sarica, M. et al. Lymphoscintigraphic abnormalities associated with Milroy disease and lymphedema-distichiasis syndrome. Lymphat. Res. Biol. 17 , 610–619 (2019).

Cox, T. C. et al. Imaging of lymphatic dysplasia in Noonan syndrome: case studies and historical atlas. Lymphology 54 , 23–40 (2021).

Kinmonth, J. B. & Wolfe, J. H. Fibrosis in the lymph nodes in primary lymphoedema. Histological and clinical studies in 74 patients with lower-limb oedema. Ann. R. Coll. Surg. Engl. 62 , 344–354 (1980).

Geng, X., Cha, B., Mahamud, M. R. & Srinivasan, R. S. Intraluminal valves: development, function and disease. Dis. Model. Mech. 10 , 1273–1287 (2017).

Gale, N. W. et al. Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1. Dev. Cell 3 , 411–423 (2002).

Kriederman, B. M. et al. FOXC2 haploinsufficient mice are a model for human autosomal dominant lymphedema-distichiasis syndrome. Hum. Mol. Genet. 12 , 1179–1185 (2003).

Dellinger, M. T. & Witte, M. H. Lymphangiogenesis, lymphatic systemomics, and cancer: context, advances and unanswered questions. Clin. Exp. Metastasis 35 , 419–424 (2018).

Northup, K. A., Witte, M. H. & Witte, C. L. Syndromic classification of hereditary lymphedema. Lymphology 36 , 162–189 (2003).

Fang, J. et al. Mutations in FOXC2 (MFH-1), a forkhead family transcription factor, are responsible for the hereditary lymphedema-distichiasis syndrome. Am. J. Hum. Genet. 67 , 1382–1388 (2000).

Brouillard, P., Boon, L. & Vikkula, M. Genetics of lymphatic anomalies. J. Clin. Invest. 124 , 898–904 (2014).

Jones, G. E. & Mansour, S. An approach to familial lymphoedema. Clin. Med. 17 , 552–557 (2017).

Michelini, S. et al. Genetic tests in lymphatic vascular malformations and lymphedema. J. Med. Genet. 55 , 222–232 (2018).

Sabin, F. R. On the origin of the lymphatic system from the veins and the development of the lymph hearts and thoracic duct in the pig. Am. J. Anat. https://doi.org/10.1002/aja.1000010310 (1902). The first description on the origin of development of the lymphatic system: generation of lymphatic sacs from the pre-existing venous system.

Article   Google Scholar  

Hutchinson, G. S. On the development of the jugular lymph sac, of the tributary ulnar lymphatic, and of the thoracic duct from the viewpoint of recent investigations of vertebrate lymphatic ontogeny, together with a consideration of the genetic relations of lymphatic and hemal vascular channels in the embryos of amniotes. Am. J. Anat. 16 , 259–316 (1914).

Schneider, M., Othman-Hassan, K., Christ, B. & Wilting, J. Lymphangioblasts in the avian wing bud. Dev. Dyn. 216 , 311–319 (1999).

Yang, Y. & Oliver, G. Development of the mammalian lymphatic vasculature. J. Clin. Invest. 124 , 888–897 (2014).

Ulvmar, M. H. & Makinen, T. Heterogeneity in the lymphatic vascular system and its origin. Cardiovasc. Res. 111 , 310–321 (2016). This review discusses the heterogeneity observed within the lymphatic system in regard to different organs as well as the functional and molecular specialization of lymphatic endothelial cells and their developmental origin .

Stone, O. A. & Stainier, D. Y. R. Paraxial mesoderm is the major source of lymphatic endothelium. Dev. Cell 50 , 247–255.e3 (2019).

Kaipainen, A. et al. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc. Natl Acad. Sci. USA 92 , 3566–3570 (1995).

Banerji, S. et al. LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. J. Cell Biol. 144 , 789–801 (1999).

Breiteneder-Geleff, S. et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am. J. Pathol. 154 , 385–394 (1999).

Wigle, J. T. & Oliver, G. Prox1 function is required for the development of the murine lymphatic system. Cell 98 , 769–778 (1999). This article describes a key role for the homeobox gene Prox1 , expressed within some venous endothelial cells that, by budding and sprouting, give rise to the lymphatic system. PROX1 is indicated as a specific and required regulator of the development of the lymphatic system.

Johnson, N. C. et al. Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity. Genes Dev. 22 , 3282–3291 (2008).

Ducoli, L. & Detmar, M. Beyond PROX1: transcriptional, epigenetic, and noncoding RNA regulation of lymphatic identity and function. Dev. Cell 56 , 406–426 (2021).

Harada, K. et al. Identification of targets of Prox1 during in vitro vascular differentiation from embryonic stem cells: functional roles of HoxD8 in lymphangiogenesis. J. Cell Sci. 122 , 3923–3930 (2009).

Frye, M. et al. Matrix stiffness controls lymphatic vessel formation through regulation of a GATA2-dependent transcriptional program. Nat. Commun. 9 , 1511 (2018).

Bowles, J. et al. Control of retinoid levels by CYP26B1 is important for lymphatic vascular development in the mouse embryo. Dev. Biol. 386 , 25–33 (2014).

Morooka, N. et al. Polydom is an extracellular matrix protein involved in lymphatic vessel remodeling. Circ. Res. 120 , 1276–1288 (2017).

Brouillard, P. et al. Loss of ADAMTS3 activity causes Hennekam lymphangiectasia-lymphedema syndrome 3. Hum. Mol. Genet. 26 , 4095–4104 (2017).

Jha, S. K. et al. Efficient activation of the lymphangiogenic growth factor VEGF-C requires the C-terminal domain of VEGF-C and the N-terminal domain of CCBE1. Sci. Rep. 7 , 4916 (2017).

Finegold, D. N. et al. Connexin 47 mutations increase risk for secondary lymphedema following breast cancer treatment. Clin. Cancer Res. 18 , 2382–2390 (2012).

Finegold, D. N. et al. HGF and MET mutations in primary and secondary lymphedema. Lymphat. Res. Biol. 6 , 65–68 (2008).

Choi, D. et al. Piezo1 incorporates mechanical force signals into the genetic program that governs lymphatic valve development and maintenance. JCI Insight 4 , e125068 (2019).

PubMed Central   Google Scholar  

Alper, S. L. Genetic diseases of PIEZO1 and PIEZO2 dysfunction. Curr. Top. Membr. 79 , 97–134 (2017).

Petrova, T. V. et al. Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nat. Med. 10 , 974–981 (2004).

Lyons, O. et al. Human venous valve disease caused by mutations in FOXC2 and GJC2. J. Exp. Med. 214 , 2437–2452 (2017).

Kazenwadel, J. et al. GATA2 is required for lymphatic vessel valve development and maintenance. J. Clin. Invest. 125 , 2979–2994 (2015).

Welsh, J. D. et al. Hemodynamic regulation of perivalvular endothelial gene expression prevents deep venous thrombosis. J. Clin. Invest. 129 , 5489–5500 (2019).

Tatin, F. et al. Planar cell polarity protein Celsr1 regulates endothelial adherens junctions and directed cell rearrangements during valve morphogenesis. Dev. Cell 26 , 31–44 (2013).

Gonzalez-Garay, M. L. et al. A novel mutation in CELSR1 is associated with hereditary lymphedema. Vasc. Cell 8 , 1 (2016).

Kanady, J. D., Dellinger, M. T., Munger, S. J., Witte, M. H. & Simon, A. M. Connexin37 and Connexin43 deficiencies in mice disrupt lymphatic valve development and result in lymphatic disorders including lymphedema and chylothorax. Dev. Biol. 354 , 253–266 (2011).

Sabine, A. et al. Mechanotransduction, PROX1, and FOXC2 cooperate to control connexin37 and calcineurin during lymphatic-valve formation. Dev. Cell 22 , 430–445 (2012).

Zhang, F., Zarkada, G., Yi, S. & Eichmann, A. Lymphatic endothelial cell junctions: molecular regulation in physiology and diseases. Front. Physiol. 11 , 509 (2020). This recent review highlights the mechanisms governing specialized lymphatic endothelial cell–cell junctions (button and zipper-like states), which are crucial for the maintenance of lymphatic vessel integrity and proper lymphatic functions.

Ferrell, R. E. et al. GJC2 missense mutations cause human lymphedema. Am. J. Hum. Genet. 86 , 943–948 (2010).

Martin-Almedina, S. et al. EPHB4 kinase-inactivating mutations cause autosomal dominant lymphatic-related hydrops fetalis. J. Clin. Invest. 126 , 3080–3088 (2016).

Choi, D. et al. ORAI1 activates proliferation of lymphatic endothelial cells in response to laminar flow through kruppel-like factors 2 and 4. Circ. Res. 120 , 1426–1439 (2017).

Mustacich, D. J. et al. Digenic inheritance of a FOXC2 mutation and two PIEZO1 mutations underlies congenital lymphedema in a multigeneration family. Am. J. Med. (in the press).

Meens, M. J. et al. Cx47 fine-tunes the handling of serum lipids but is dispensable for lymphatic vascular function. PLoS ONE 12 , e0181476 (2017).

Mustacich, D. J., et al. Abnormal lymphatic phenotype in a crispr mouse model of the human lymphedema-causing connexin47 R260C point mutation. Lymphology (in the press).

Boucher, C. A., Sargent, C. A., Ogata, T. & Affara, N. A. Breakpoint analysis of Turner patients with partial Xp deletions: implications for the lymphoedema gene location. J. Med. Genet. 38 , 591–598 (2001).

Ogata, T., Tyler-Smith, C., Purvis-Smith, S. & Turner, G. Chromosomal localisation of a gene(s) for Turner stigmata on Yp. J. Med. Genet. 30 , 918–922 (1993).

Bardi, F. et al. Is there still a role for nuchal translucency measurement in the changing paradigm of first trimester screening? Prenat. Diagn. 40 , 197–205 (2020).

Hsu, L. Y., Shapiro, L. R., Gertner, M., Lieber, E. & Hirschhorn, K. Trisomy 22: a clinical entity. J. Pediatr. 79 , 12–19 (1971).

Rosenfeld, W. et al. Duplication 3q: severe manifestations in an infant with duplication of a short segment of 3q. Am. J. Med. Genet. 10 , 187–192 (1981).

Greenlee, R., Hoyme, H., Witte, M., Crowe, P. & Witte, C. Developmental disorders of the lymphatic system. Lymphology 26 , 156–168 (1993). This article reviews the chromosomal abnormalities and syndromes associated with lymphatic disorders, with a focus on primary lymphoedema.

Unolt, M. et al. Primary lymphedema and other lymphatic anomalies are associated with 22q11.2 deletion syndrome. Eur. J. Med. Genet. 61 , 411–415 (2018).

Bull, L. N. et al. Mapping of the locus for cholestasis-lymphedema syndrome (Aagenaes syndrome) to a 6.6-cM interval on chromosome 15q. Am. J. Hum. Genet. 67 , 994–999 (2000).

Jha, S. K., Rauniyar, K. & Jeltsch, M. Key molecules in lymphatic development, function, and identification. Ann. Anat. 219 , 25–34 (2018).

Baldwin, M. E. et al. Vascular endothelial growth factor D is dispensable for development of the lymphatic system. Mol. Cell Biol. 25 , 2441–2449 (2005).

Benedito, R. et al. Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling. Nature 484 , 110–114 (2012).

Makinen, T. et al. PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature. Genes Dev. 19 , 397–410 (2005).

Carmeliet, P. & Tessier-Lavigne, M. Common mechanisms of nerve and blood vessel wiring. Nature 436 , 193–200 (2005).

Souma, T. et al. Context-dependent functions of angiopoietin 2 are determined by the endothelial phosphatase VEPTP. Proc. Natl Acad. Sci. USA 115 , 1298–1303 (2018).

Ayadi, A., Suelves, M., Dolle, P. & Wasylyk, B. Net, an Ets ternary complex transcription factor, is expressed in sites of vasculogenesis, angiogenesis, and chondrogenesis during mouse development. Mech. Dev. 102 , 205–208 (2001).

Kajiya, K., Hirakawa, S., Ma, B., Drinnenberg, I. & Detmar, M. Hepatocyte growth factor promotes lymphatic vessel formation and function. EMBO J. 24 , 2885–2895 (2005).

Brouillard, P. et al. Non-hotspot PIK3CA mutations are more frequent in CLOVES than in common or combined lymphatic malformations. Orphanet J. Rare Dis. 16 , 267 (2021).

Schook, C. C. et al. Differential diagnosis of lower extremity enlargement in pediatric patients referred with a diagnosis of lymphedema. Plast. Reconstr. Surg. 127 , 1571–1581 (2011).

Szuba, A., Shin, W. S., Strauss, H. W. & Rockson, S. The third circulation: radionuclide lymphoscintigraphy in the evaluation of lymphedema. J. Nucl. Med. 44 , 43–57 (2003).

Atton, G. et al. The lymphatic phenotype in Turner syndrome: an evaluation of nineteen patients and literature review. Eur. J. Hum. Genet. 23 , 1634–1639 (2015).

Nadarajah, N. et al. A Novel splice-site mutation in VEGFC is associated with congenital primary lymphoedema of Gordon. Int. J. Mol. Sci. 19 , 2259 (2018).

Burnier, P., Niddam, J., Bosc, R., Hersant, B. & Meningaud, J. P. Indocyanine green applications in plastic surgery: a review of the literature. J. Plast. Reconstr. Aesthet. Surg. 70 , 814–827 (2017).

Unno, N. et al. A novel method of measuring human lymphatic pumping using indocyanine green fluorescence lymphography. J. Vasc. Surg. 52 , 946–952 (2010).

Liu, N. F., Yan, Z. X. & Wu, X. F. Classification of lymphatic-system malformations in primary lymphoedema based on MR lymphangiography. Eur. J. Vasc. Endovasc. Surg. 44 , 345–349 (2012).

Biko, D. M. et al. Imaging of central lymphatic abnormalities in Noonan syndrome. Pediatr. Radiol. 49 , 586–592 (2019).

Biko, D. M. et al. Intrahepatic dynamic contrast MR lymphangiography: initial experience with a new technique for the assessment of liver lymphatics. Eur. Radiol. 29 , 5190–5196 (2019).

Dori, Y. Novel lymphatic imaging techniques. Tech. Vasc. Interv. Radiol. 19 , 255–261 (2016).

Kinmonth, J. B., Taylor, G. W., Tracy, G. D. & Marsh, J. D. Primary lymphedema: clinical and lymphangiographic studies of a series of 107 patients in which lower limbs were affected. Br. J. Surg. 45 , 1 (1957).

Rajebi, M. R. et al. Intranodal lymphangiography: feasibility and preliminary experience in children. J. Vasc. Interv. Radiol. 22 , 1300–1305 (2011).

Ho, B., Gordon, K. & Mortimer, P. S. A genetic approach to the classification of primary lymphoedema and lymphatic malformations. Eur. J. Vasc. Endovasc. Surg. 56 , 465–466 (2018).

Dalal, A. et al. Interventions for the prevention of recurrent erysipelas and cellulitis. Cochrane Database Syst. Rev. 6 , CD009758 (2017).

van Karnebeek, C. D. M. et al. The role of the clinician in the multi-omics era: are you ready? J. Inherit. Metab. Dis. 41 , 571–582 (2018).

Damstra, R. J., van Steensel, M. A., Boomsma, J. H., Nelemans, P. & Veraart, J. C. Erysipelas as a sign of subclinical primary lymphoedema: a prospective quantitative scintigraphic study of 40 patients with unilateral erysipelas of the leg. Br. J. Dermatol. 158 , 1210–1215 (2008).

Hayes, S. C. Role of exercise in the prevention and management of lymphedema after breast cancer. Exerc. Sport. Sci. Rev. 38 , 2 (2010).

Hayes, S. C. et al. Exercise for health: a randomized, controlled trial evaluating the impact of a pragmatic, translational exercise intervention on the quality of life, function and treatment-related side effects following breast cancer. Breast Cancer Res. Treat. 137 , 175–186 (2013).

Wirtz, P. & Baumann, F. T. Physical activity, exercise and breast cancer - what is the evidence for rehabilitation, aftercare, and survival? A review. Breast Care 13 , 93–101 (2018).

Dieli-Conwright, C. M. et al. Aerobic and resistance exercise improves physical fitness, bone health, and quality of life in overweight and obese breast cancer survivors: a randomized controlled trial. Breast Cancer Res. 20 , 124 (2018).

Yumuk, V. et al. European guidelines for obesity management in adults. Obes. Facts 8 , 402–424 (2015).

Damstra, R. J. & Halk, A.-B., Dutch Working Group on Lymphoedema.The Dutch lymphoedema guidelines based on the International Classification of Functioning, Disability, and Health and the chronic care model. J. Vasc. Surg. Venous Lymphat. Disord. 5 , 756–765 (2017).

Leysen, L. et al. Risk factors of pain in breast cancer survivors: a systematic review and meta-analysis. Support. Care Cancer 25 , 3607–3643 (2017).

Shahpar, H. et al. Risk factors of lymph edema in breast cancer patients. Int. J. Breast Cancer 2013 , 641818 (2013).

Vieira, R. A. et al. Risk factors for arm lymphedema in a cohort of breast cancer patients followed up for 10 years. Breast Care 11 , 45–50 (2016).

Watt, H., Singh-Grewal, D., Wargon, O. & Adams, S. Paediatric lymphoedema: a retrospective chart review of 86 cases. J. Paediatr. Child. Health 53 , 38–42 (2017).

Badger, C. M., Peacock, J. L. & Mortimer, P. S. A randomized, controlled, parallel-group clinical trial comparing multilayer bandaging followed by hosiery versus hosiery alone in the treatment of patients with lymphedema of the limb. Cancer 88 , 2832–2837 (2000).

O’Donnell, T. F. Jr, Allison, G. M. & Iafrati, M. D. A systematic review of guidelines for lymphedema and the need for contemporary intersocietal guidelines for the management of lymphedema. J. Vasc. Surg. Venous Lymphat. Disord. 8 , 676–684 (2020).

Gloviczki, P. Handbook of Venous Disorders: Guidelines of the American Venous Forum (CRC Press, 2017).

Lymphoedema Framework. Best Practice for the Management of Lymphoedema. International Consensus (MEP Ltd., 2006).

Vreeburg, M. et al. Lymphedema-distichiasis syndrome: a distinct type of primary lymphedema caused by mutations in the FOXC2 gene. Int. J. Dermatol. 47 (Suppl. 1), 52–55 (2008).

Shenoy, V. G., Jawale, S. A., Oak, S. N. & Kulkarni, B. K. Primary lymphedema of the penis: surgical correction by preputial unfurling. Pediatr. Surg. Int. 17 , 169–170 (2001).

Suehiro, K., Morikage, N., Murakami, M., Yamashita, O. & Hamano, K. Primary lymphedema complicated by weeping chylous vesicles in the leg and scrotum: report of a case. Surg. Today 42 , 1100–1103 (2012).

Phillips, J. J. & Gordon, S. J. Conservative management of lymphoedema in children: a systematic review. J. Pediatr. Rehabil. Med. 7 , 361–372 (2014).

Todd, M. Compression in young people living with lymphoedema. Br. J. Nurs. 28 , 908–910 (2019).

Benoughidane, B., Simon, L., Fourgeaud, C. & Vignes, S. Low-stretch bandages to treat primary lower limb lymphoedema: a cohort of 48 children. Br. J. Dermatol. 179 , 1203–1204 (2018).

Vignes, S. & Bellanger, J. Primary intestinal lymphangiectasia (Waldmann’s disease). Orphanet J. Rare Dis. 3 , 5 (2008).

Sarasua, S. M. et al. Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome. Hum. Genet. 133 , 847–859 (2014).

Emberger, J. M., Navarro, M., Dejean, M. & Izarn, P. Deaf-mutism, lymphedema of the lower limbs and hematological abnormalities (acute leukemia, cytopenia) with autosomal dominant transmission. J. Genet. Hum. 27 , 237–245 (1979).

Fuchs, S. et al. Vascular endothelial growth factor (VEGF) levels in short, GH treated children: a distinct pattern of VEGF-C in Noonan syndrome. J. Endocrinol. Invest. 38 , 399–406 (2015).

Ostergaard, P. et al. Mutations in GATA2 cause primary lymphedema associated with a predisposition to acute myeloid leukemia (Emberger syndrome). Nat. Genet. 43 , 929–931 (2011).

Wlodarski, M. W., Collin, M. & Horwitz, M. S. GATA2 deficiency and related myeloid neoplasms. Semin. Hematol. 54 , 81–86 (2017).

Rastogi, N. et al. Successful nonmyeloablative allogeneic stem cell transplant in a child with Emberger syndrome and GATA2 mutation. J. Pediatr. Hematol. Oncol. 40 , e383–e388 (2018).

Ramzan, M. et al. Successful myeloablative matched unrelated donor hematopoietic stem cell transplantation in a young girl with GATA2 deficiency and Emberger syndrome. J. Pediatr. Hematol. Oncol. 39 , 230–232 (2017).

Saida, S. et al. Successful reduced-intensity stem cell transplantation for GATA2 deficiency before progression of advanced MDS. Pediatr. Transpl. 20 , 333–336 (2016).

CAS   Google Scholar  

Lubking, A. et al. Young woman with mild bone marrow dysplasia, GATA2 and ASXL1 mutation treated with allogeneic hematopoietic stem cell transplantation. Leuk. Res. Rep. 4 , 72–75 (2015).

Bishnoi, A. et al. Warty fingers and toes in a child with congenital lymphedema: elephantiasis nostras verrucosa. JAMA Dermatol. 154 , 849–850 (2018).

Perez Botero, J. & Rodriguez, V. Primary lymphedema and viral warts in GATA2 haploinsufficiency. Mayo Clin. Proc. 92 , 482 (2017).

Dorn, J. M. et al. WILD syndrome is GATA2 deficiency: a novel deletion in the GATA2 gene. J. Allergy Clin. Immunol. Pract. 5 , 1149–1152.e1 (2017).

Kreuter, A. et al. A human papillomavirus-associated disease with disseminated warts, depressed cell-mediated immunity, primary lymphedema, and anogenital dysplasia: WILD syndrome. Arch. Dermatol. 144 , 366–372 (2008).

Cusack, C., Fitzgerald, D., Clayton, T. M. & Irvine, A. D. Successful treatment of florid cutaneous warts with intravenous cidofovir in an 11-year-old girl. Pediatr. Dermatol. 25 , 387–389 (2008).

Kreuter, A., Waterboer, T. & Wieland, U. Regression of cutaneous warts in a patient with WILD syndrome following recombinant quadrivalent human papillomavirus vaccination. Arch. Dermatol. 146 , 1196–1197 (2010).

Manevitz-Mendelson, E. et al. Somatic NRAS mutation in patient with generalized lymphatic anomaly. Angiogenesis 21 , 287–298 (2018).

Barclay, S. F. et al. A somatic activating NRAS variant associated with kaposiform lymphangiomatosis. Genet. Med. 21 , 1517–1524 (2019).

Rodriguez-Laguna, L. et al. Somatic activating mutations in PIK3CA cause generalized lymphatic anomaly. J. Exp. Med. 216 , 407–418 (2019).

Foster, J. B. et al. Kaposiform lymphangiomatosis effectively treated with MEK inhibition. EMBO Mol. Med. 12 , e12324 (2020).

Homayun Sepehr, N. et al. KRAS-driven model of Gorham-Stout disease effectively treated with trametinib. JCI Insight https://doi.org/10.1172/jci.insight.149831 (2021).

Article   PubMed   PubMed Central   Google Scholar  

Queisser, A., Seront, E., Boon, L. M. & Vikkula, M. Genetic basis and therapies for vascular anomalies. Circ. Res. 129 , 155–173 (2021). This recent review describes the genetic and pathophysiological discoveries in the field of vascular anomalies and the current status of repurposing of cancer drugs for their targeted management (theranostics).

Makinen, T., Boon, L. M., Vikkula, M. & Alitalo, K. Lymphatic malformations: genetics, mechanisms and therapeutic strategies. Circ. Res. 129 , 136–154 (2021). This recent review portrays the numerous discoveries made on the genetic and pathophysiological bases of lymphatic malformations and understanding of the molecular and cellular mechanisms involved. It also illustrates the fast progress made in the repurposing of small molecule inhibitors developed for oncology for the targeted management of lymphatic malformations.

Li, D. et al. ARAF recurrent mutation causes central conducting lymphatic anomaly treatable with a MEK inhibitor. Nat. Med. 25 , 1116–1122 (2019).

Rockson, S. G. et al. Pilot studies demonstrate the potential benefits of antiinflammatory therapy in human lymphedema. JCI Insight 3 , e123775 (2018). This small clinical trial for lymphoedema suggests the utility of anti-inflammatory therapy with ketoprofen for patients with lymphoedema.

Brorson, H., Svensson, H., Norrgren, K. & Thorsson, O. Liposuction reduces arm lymphedema without significantly altering the already impaired lymph transport. Lymphology 31 , 156–172 (1998). This prospective study on 20 patients with arm lymphoedema after breast cancer treatment showed that liposuction combined with controlled compression therapy is efficacious.

Schaverien, M. V., Munnoch, D. A. & Brorson, H. Liposuction treatment of lymphedema. Semin. Plast. Surg. 32 , 42–47 (2018).

Greene, A. K., Sudduth, C. L. & Taghinia, A. Lymphedema (seminars in pediatric surgery). Semin. Pediatr. Surg. 29 , 150972 (2020). This recent report reviews the preventive, compressive and interventional options, including lympho-venous anastomosis, LNT and liposuction, for the management of lymphoedema.

Brorson, H., Ohlin, K., Olsson, G., Svensson, B. & Svensson, H. Controlled compression and liposuction treatment for lower extremity lymphedema. Lymphology 41 , 52–63 (2008).

Greene, A. K., Slavin, S. A. & Borud, L. Treatment of lower extremity lymphedema with suction-assisted lipectomy. Plast. Reconstr. Surg. 118 , 118e–121e (2006).

Hendrickx, A. A., Damstra, R. J., Krijnen, W. P. & van der Schans, C. P. Improvement of limb volumes after bariatric surgery in nine end-stage primary, secondary, and obesity-induced lymphedema patients: a multiple case report. Lymphat. Res. Biol. https://doi.org/10.1089/lrb.2020.0055 (2021).

Article   PubMed   Google Scholar  

Olszewski, W. L. The treatment of lymphedema of the extremities with microsurgical lympho-venous anastomoses. Int. Angiol. 7 , 312–321 (1988).

Yamamoto, T. et al. Indocyanine green lymphography findings in primary leg lymphedema. Eur. J. Vasc. Endovasc. Surg. 49 , 95–102 (2015).

Hara, H. et al. Indication of lymphaticovenous anastomosis for lower limb primary lymphedema. Plast. Reconstr. Surg. 136 , 883–893 (2015).

Maegawa, J., Mikami, T., Yamamoto, Y., Satake, T. & Kobayashi, S. Types of lymphoscintigraphy and indications for lymphaticovenous anastomosis. Microsurgery 30 , 437–442 (2010).

Dermitas, Y., Ozturk, N., Yapici, O. & Topalan, M. Comparison of primary and secondary lower-extremity lymphedema treated with supramicrosurgical lymphaticovenous anastomosis and lymphaticovenous implantation. J. Reconstr. Microsurg. 26 , 137–143 (2010).

Gennaro, P. et al. Ultramicrosurgery: a new approach to treat primary male genital lymphedema. JPRAS Open 20 , 72–80 (2019).

Becker, C. et al. Surgical treatment of congenital lymphedema. Clin. Plast. Surg. 39 , 377–384 (2012).

Vignes, S., Blanchard, M., Yannoutsos, A. & Arrault, M. Complications of autologous lymph-node transplantation for limb lymphoedema. Eur. J. Vasc. Endovasc. Surg. 45 , 516–520 (2013).

Cheng, M. H., Loh, C. Y. Y. & Lin, C. Y. Outcomes of vascularized lymph node transfer and lymphovenous anastomosis for treatment of primary lymphedema. Plast. Reconstr. Surg. Glob. Open 6 , e2056 (2018).

Rychik, J. et al. Evaluation and management of the child and adult with fontan circulation: a scientific statement from the American Heart Association. Circulation https://doi.org/10.1161/CIR.0000000000000696 (2019).

Itkin, M., Pizarro, C., Radtke, W., Spurrier, E. & Rabinowitz, D. A. Lymphatic management in single-ventricle patients. Semin. Thorac. Cardiovasc. Surg. Pediatr. Card. Surg Annu. 23 , 41–47 (2020).

Schumacher, K. R. et al. Fontan-associated protein-losing enteropathy and plastic bronchitis. J. Pediatr. 166 , 970–977 (2015).

Bamezai, S., Aronberg, R. M., Park, J. M. & Gemmete, J. J. Intranodal lymphangiography and interstitial lymphatic embolization to treat chyluria caused by a lymphatic malformation in a pediatric patient. Pediatr. Radiol. 51 , 1762–1765 (2021).

Yamamoto, M. et al. Intranodal lymphatic embolization for chylocolporrhea caused by chylous reflux syndrome in Noonan syndrome. J. Vasc. Interv. Radiol. 30 , 769–772 (2019).

Itkin, M. et al. Protein-losing enteropathy in patients with congenital heart disease. J. Am. Coll. Cardiol. 69 , 2929–2937 (2017).

Okajima, S. et al. Health-related quality of life and associated factors in patients with primary lymphedema. Jpn. J. Nurs. Sci. 10 , 202–211 (2013).

Herberger, K. et al. Quality of life in patients with primary and secondary lymphedema in the community. Wound Repair. Regen. 25 , 466–473 (2017).

Fu, M. R. et al. Psychosocial impact of lymphedema: a systematic review of literature from 2004 to 2011. Psychooncology 22 , 1466–1484 (2013).

Hanson, C. S. et al. Children and adolescents’ experiences of primary lymphoedema: semistructured interview study. Arch. Dis. Child. 103 , 675–682 (2018).

Stucki, G. & Grimby, G. Applying the ICF in medicine. J. Rehabil. Med. 44 (Suppl.), 5–6 (2004).

Hidding, J. T. et al. Measurement properties of instruments for measuring of lymphedema: systematic review. Phys. Ther. 96 , 1965–1981 (2016).

Viehoff, P. B., Hidding, J. T., Heerkens, Y. F., van Ravensberg, C. D. & Neumann, H. A. Coding of meaningful concepts in lymphedema-specific questionnaires with the ICF. Disabil. Rehabil. 35 , 2105–2112 (2013).

Devoogdt, N. et al. Lymphoedema functioning, disability and health questionnaire for lower limb lymphoedema (Lymph-ICF-LL): reliability and validity. Phys. Ther. 94 , 705–721 (2014).

Klernas, P., Johnsson, A., Horstmann, V., Kristjanson, L. J. & Johansson, K. Lymphedema quality of life inventory (LyQLI)-development and investigation of validity and reliability. Qual. Life Res. 24 , 427–439 (2015).

Angst, F., Lehmann, S., Aeschlimann, A., Sandor, P. S. & Wagner, S. Cross-sectional validity and specificity of comprehensive measurement in lymphedema and lipedema of the lower extremity: a comparison of five outcome instruments. Health Qual. Life Outcomes 18 , 245 (2020).

Moffatt, C. J. & Murray, S. G. The experience of children and families with lymphoedema — a journey within a journey. Int. Wound J. 7 , 14–26 (2010).

Moffatt, C. et al. A study to explore the professional conceptualization and challenges of self-management in children and adolescents with lymphedema. Lymphat. Res. Biol. 17 , 221–230 (2019).

Moffatt, C. et al. A study to explore the parental impact and challenges of self-management in children and adolescents suffering with lymphedema. Lymphat. Res. Biol. 17 , 245–252 (2019).

Quere, I. et al. International camps for children with lymphedema and lymphatic anomalies: when education links with psychosocial research. Lymphat. Res. Biol. 19 , 36–40 (2021).

Visser, J., van Geel, M., Cornelissen, A. J. M., van der Hulst, R. & Qiu, S. S. Breast cancer-related lymphedema and genetic predisposition: a systematic review of the literature. Lymphat. Res. Biol. 17 , 288–293 (2019).

Coulie, R. et al. Hypotrichosis-lymphedema-telangiectasia syndrome: Report of ileal atresia associated with a SOX18 de novo pathogenic variant and review of the phenotypic spectrum. Am. J. Med. Genet. A 185 , 2153–2159 (2021).

Kajita, H. et al. Photoacoustic lymphangiography. J. Surg. Oncol. 121 , 48–50 (2020).

Kajita, H. et al. Visualization of lymphatic vessels using photoacoustic imaging. Keio J. Med. https://doi.org/10.2302/kjm.2020-0010-OA (2020).

Shinaoka, A., Yamada, K. & Kimata, Y. in ICG Fluorescence Imaging and Navigation Surgery (eds Kusano, M., Kokudo, N., Toi, M. & Kaibori, M.) 433–442 (Springer 2016).

Hartiala, P. et al. Phase 1 Lymfactin(R) study: short-term safety of combined adenoviral VEGF-C and lymph node transfer treatment for upper extremity lymphedema. J. Plast. Reconstr. Aesthet. Surg. 73 , 1612–1621 (2020).

Heitink, M. V. et al. Lymphedema in Prader-Willi syndrome. Int. J. Dermatol. 47 (Suppl. 1), 42–44 (2008).

Garcia-Cruz, D. et al. Cantu syndrome and lymphoedema. Clin. Dysmorphol. 20 , 32–37 (2011).

Scheuerle, A. E. et al. An additional case of Hennekam lymphangiectasia-lymphedema syndrome caused by loss-of-function mutation in ADAMTS3. Am. J. Med. Genet. A 176 , 2858–2861 (2018).

Alders, M. et al. Mutations in CCBE1 cause generalized lymph vessel dysplasia in humans. Nat. Genet. 41 , 1272–1274 (2009).

Li, D. et al. Pathogenic variant in EPHB4 results in central conducting lymphatic anomaly. Hum. Mol. Genet. 27 , 3233–3245 (2018).

Alders, M. et al. Hennekam syndrome can be caused by FAT4 mutations and be allelic to Van Maldergem syndrome. Hum. Genet. 133 , 1161–1167 (2014).

Doffinger, R. et al. X-linked anhidrotic ectodermal dysplasia with immunodeficiency is caused by impaired NF-kappaB signaling. Nat. Genet. 27 , 277–285 (2001).

Mansour, S. et al. Incontinentia pigmenti in a surviving male is accompanied by hypohidrotic ectodermal dysplasia and recurrent infection. Am. J. Med. Genet. 99 , 172–177 (2001).

Ostergaard, P. et al. Mutations in KIF11 cause autosomal-dominant microcephaly variably associated with congenital lymphedema and chorioretinopathy. Am. J. Hum. Genet. 90 , 356–362 (2012).

Schubbert, S. et al. Germline KRAS mutations cause Noonan syndrome. Nat. Genet. 38 , 331–336 (2006).

Nozawa, A. et al. A somatic activating KRAS variant identified in an affected lesion of a patient with Gorham-Stout disease. J. Hum. Genet. 65 , 995–1001 (2020).

McClelland, J., Burgess, B., Crock, P. & Goel, H. Sotos syndrome: an unusual presentation with intrauterine growth restriction, generalized lymphedema, and intention tremor. Am. J. Med. Genet. A 170A , 1064–1069 (2016).

Foster, A. et al. The phenotype of Sotos syndrome in adulthood: a review of 44 individuals. Am. J. Med. Genet. C. Semin. Med Genet 181 , 502–508 (2019).

Fotiou, E. et al. Novel mutations in PIEZO1 cause an autosomal recessive generalized lymphatic dysplasia with non-immune hydrops fetalis. Nat. Commun. 6 , 8085 (2015).

Lukacs, V. et al. Impaired PIEZO1 function in patients with a novel autosomal recessive congenital lymphatic dysplasia. Nat. Commun. 6 , 8329 (2015).

Yoshida, R., Miyata, M., Nagai, T., Yamazaki, T. & Ogata, T. A 3-bp deletion mutation of PTPN11 in an infant with severe Noonan syndrome including hydrops fetalis and juvenile myelomonocytic leukemia. Am. J. Med. Genet. A 128A , 63–66 (2004).

Croonen, E. A. et al. Prenatal diagnostic testing of the Noonan syndrome genes in fetuses with abnormal ultrasound findings. Eur. J. Hum. Genet. 21 , 936–942 (2013).

Thompson, D. et al. RAF1 variants causing biventricular hypertrophic cardiomyopathy in two preterm infants: further phenotypic delineation and review of literature. Clin. Dysmorphol. 26 , 195–199 (2017).

Burrows, P. E. et al. Lymphatic abnormalities are associated with RASA1 gene mutations in mouse and man. Proc. Natl Acad. Sci. USA 110 , 8621–8626 (2013).

de Wijn, R. S. et al. Phenotypic variability in a family with capillary malformations caused by a mutation in the RASA1 gene. Eur. J. Med. Genet. 55 , 191–195 (2012).

Macmurdo, C. F. et al. RASA1 somatic mutation and variable expressivity in capillary malformation/arteriovenous malformation (CM/AVM) syndrome. Am. J. Med. Genet. A 170 , 1450–1454 (2016).

Gos, M. et al. Contribution of RIT1 mutations to the pathogenesis of Noonan syndrome: four new cases and further evidence of heterogeneity. Am. J. Med. Genet. A 164A , 2310–2316 (2014).

Milosavljevic, D. et al. Two cases of RIT1 associated Noonan syndrome: further delineation of the clinical phenotype and review of the literature. Am. J. Med. Genet. A 170 , 1874–1880 (2016).

Koenighofer, M. et al. Mutations in RIT1 cause Noonan syndrome - additional functional evidence and expanding the clinical phenotype. Clin. Genet. 89 , 359–366 (2016).

Yaoita, M. et al. Spectrum of mutations and genotype-phenotype analysis in Noonan syndrome patients with RIT1 mutations. Hum. Genet. 135 , 209–222 (2016).

Roberts, A. E. et al. Germline gain-of-function mutations in SOS1 cause Noonan syndrome. Nat. Genet. 39 , 70–74 (2007).

Smpokou, P., Tworog-Dube, E., Kucherlapati, R. S. & Roberts, A. E. Medical complications, clinical findings, and educational outcomes in adults with Noonan syndrome. Am. J. Med. Genet. A 158A , 3106–3111 (2012).

Yamamoto, G. L. et al. Rare variants in SOS2 and LZTR1 are associated with Noonan syndrome. J. Med. Genet. 52 , 413–421 (2015).

Cordeddu, V. et al. Activating mutations affecting the Dbl homology domain of SOS2 cause Noonan syndrome. Hum. Mutat. 36 , 1080–1087 (2015).

Lissewski, C. et al. Variants of SOS2 are a rare cause of Noonan syndrome with particular predisposition for lymphatic complications. Eur. J. Hum. Genet. 29 , 51–60 (2021).

Irrthum, A. et al. Mutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia. Am. J. Hum. Genet. 72 , 1470–1478 (2003).

Shamseldin, H. E. et al. Identification of embryonic lethal genes in humans by autozygosity mapping and exome sequencing in consanguineous families. Genome Biol. 16 , 116 (2015).

Abdelrahman, H. A. et al. A recessive truncating variant in thrombospondin-1 domain containing protein 1 gene THSD1 is the underlying cause of nonimmune hydrops fetalis, congenital cardiac defects, and haemangiomas in four patients from a consanguineous family. Am. J. Med. Genet. A 176 , 1996–2003 (2018).

Prato, G. et al. Congenital segmental lymphedema in tuberous sclerosis complex with associated subependymal giant cell astrocytomas treated with Mammalian target of rapamycin inhibitors. J. Child. Neurol. 29 , NP54–NP57 (2014).

Geffrey, A. L., Shinnick, J. E., Staley, B. A., Boronat, S. & Thiele, E. A. Lymphedema in tuberous sclerosis complex. Am. J. Med. Genet. A 164A , 1438–1442 (2014).

Gordon, K. et al. Mutation in vascular endothelial growth factor-C, a ligand for vascular endothelial growth factor receptor-3, is associated with autosomal dominant Milroy-like primary lymphedema. Circ. Res. 112 , 956–960 (2013).

Balboa-Beltran, E. et al. A novel stop mutation in the vascular endothelial growth factor-C gene (VEGFC) results in Milroy-like disease. J. Med. Genet. 51 , 475–478 (2014).

Mukenge, S. et al. Investigation on the role of biallelic variants in VEGF-C found in a patient affected by Milroy-like lymphedema. Mol. Genet. Genomic Med. 8 , e1389 (2020).

Jones, K. L., Schwarze, U., Adam, M. P., Byers, P. H. & Mefford, H. C. A homozygous B3GAT3 mutation causes a severe syndrome with multiple fractures, expanding the phenotype of linkeropathy syndromes. Am. J. Med. Genet. A 167A , 2691–2696 (2015).

Sekiguchi, K. et al. A transient myelodysplastic/myeloproliferative neoplasm in a patient with cardio-facio-cutaneous syndrome and a germline BRAF mutation. Am. J. Med. Genet. A 161A , 2600–2603 (2013).

Joyce, S. et al. The lymphatic phenotype in Noonan and Cardiofaciocutaneous syndrome. Eur. J. Hum. Genet. 24 , 690–696 (2016).

Hanson, H. L. et al. Germline CBL mutation associated with a noonan-like syndrome with primary lymphedema and teratoma associated with acquired uniparental isodisomy of chromosome 11q23. Am. J. Med. Genet. A 164A , 1003–1009 (2014).

Boone, P. M. et al. Biallelic mutation of FBXL7 suggests a novel form of Hennekam syndrome. Am. J. Med. Genet. A 182 , 189–194 (2020).

Michelini, S. et al. Genetic screening in a large cohort of italian patients affected by primary lymphedema using a next generation sequencing (NGS) Approach. Lymphology 49 , 57–72 (2016).

Kawase, K. et al. Nemaline myopathy with KLHL40 mutation presenting as congenital totally locked-in state. Brain Dev. 37 , 887–890 (2015).

Sparks, T. N. et al. Exome sequencing for prenatal diagnosis in nonimmune hydrops fetalis. N. Engl. J. Med. 383 , 1746–1756 (2020).

Ponti, G. et al. Giant elephantiasis neuromatosa in the setting of neurofibromatosis type 1: a case report. Oncol. Lett. 11 , 3709–3714 (2016).

Michelini, S. et al. Segregation analysis of rare NRP1 and NRP2 variants in families with lymphedema. Genes 11 , 1361 (2020).

CAS   PubMed Central   Google Scholar  

Ricci, M. et al. Review of the function of SEMA3A in lymphatic vessel maturation and its potential as a candidate gene for lymphedema: Analysis of three families with rare causative variants. Lymphology 53 , 63–75 (2020).

Gargano, G. et al. Hydrops fetalis in a preterm newborn heterozygous for the c.4A>G SHOC2 mutation. Am. J. Med. Genet. A 164A , 1015–1020 (2014).

Takenouchi, T. et al. Severe craniosynostosis with Noonan syndrome phenotype associated with SHOC2 mutation: clinical evidence of crosslink between FGFR and RAS signaling pathways. Am. J. Med. Genet. A 164A , 2869–2872 (2014).

Michelini, S. et al. TIE1 as a candidate gene for lymphatic malformations with or without lymphedema. Int. J. Mol. Sci. 21 , 6780 (2020).

Lucas, M. & Andrade, Y. Congenital lymphedema with tuberous sclerosis and clinical Hirschsprung disease. Pediatr. Dermatol. 28 , 194–195 (2011).

Klinner, J. et al. Congenital lymphedema as a rare and first symptom of tuberous sclerosis complex. Gene 753 , 144815 (2020).

Hopman, S. M. et al. PTEN hamartoma tumor syndrome and Gorham-Stout phenomenon. Am. J. Med. Genet. A 158A , 1719–1723 (2012).

Scarcella, A., De Lucia, A., Pasquariello, M. B. & Gambardella, P. Early death in two sisters with Hennekam syndrome. Am. J. Med. Genet. 93 , 181–183 (2000).

Greene, A. K., Grant, F. D. & Slavin, S. A. Lower-extremity lymphedema and elevated body-mass index. N. Engl. J. Med. 366 , 2136–2137 (2012).

Burian, E. A. et al. Cellulitis in chronic oedema of the lower leg: an international cross-sectional study. Br. J. Dermatol. 185 , 110–118 (2021).

World Health Organization. Lymphatic filariasis — managing morbidity and preventing disability — an aide-mémoire for national programme managers . Second edition (WHO, 2021).

Zanten, M. et al. A diagnostic dilemma: aetiological diagnosis of lymphoedema patients at an Indian multidisciplinary meeting. J. Lymphoedema 14 , 43–46 (2019).

Mercier, G. et al. Out-of-pocket payments, vertical equity and unmet medical needs in France: A national multicenter prospective study on lymphedema. PLoS ONE 14 , e0216386 (2019).

Download references

Acknowledgements

M.V.’s laboratories were financially supported by the Fonds de la Recherche Scientifique – FNRS Grants T.0026.14 and T.0247.19, the Fund Generet managed by the King Baudouin Foundation (Grant 2018-J1810250-211305), and by la Région wallonne dans le cadre du financement de l’axe stratégique FRFS-WELBIO (WELBIO-CR-2019C-06). M.V. has also received funding from the MSCA-ITN network V. A. Cure No. 814316 and the Lymphatic Malformation Institute, USA. M.H.W. has received research support from the University of Arizona Health Sciences Translational Imaging Program Projects Stimulus (TIPPS) Award and National Institutes of Health NHLBI R25HL108837 for diverse undergraduate research trainees (Luis Luy, Jasmine Jones, Reginald Myles); she is also Secretary-General, International Society of Lymphology, Tucson, AZ, USA, and Zurich, Switzerland. The authors are grateful to Grace Wagner and Juan Ruiz for programmatic assistance and to Liliana Niculescu for expert secretarial assistance.

Author information

Authors and affiliations.

Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium

Pascal Brouillard & Miikka Vikkula

Department of Surgery, Neurosurgery, and Pediatrics, University of Arizona College of Medicine, Tucson, AZ, USA

  • Marlys H. Witte

Department of Pediatrics, University of Arizona College of Medicine, Tucson, AZ, USA

Robert P. Erickson

VASCERN PPL European Reference Centre; Department of Dermatology, Phlebology and Lymphology, Nij Smellinghe Hospital, Drachten, Netherlands

Robert J. Damstra

Lymphoedema Center, Clinique Jouvenet, Paris, France

Corinne Becker

Department of Vascular Medicine, Centre de référence des Maladies Lymphatiques et Vasculaires Rares, Inserm IDESP, CHU Montpellier, Université de Montpellier, Montpellier, France

Isabelle Quéré

VASCERN VASCA European Reference Centre; Center for Vascular Anomalies, Division of Plastic Surgery, University Clinics Saint-Luc, University of Louvain, Brussels, Belgium

Miikka Vikkula

Walloon Excellence in Lifesciences and Biotechnology (WELBIO), de Duve Institute, University of Louvain, Brussels, Belgium

You can also search for this author in PubMed   Google Scholar

Contributions

Introduction (M.V., P.B., M.H.W. and R.P.E.); Epidemiology (M.V., P.B., M.H.W., R.P.E. and I.Q.); Mechanisms/pathophysiology (M.V., P.B., M.H.W., R.P.E. and I.Q.); Diagnosis, screening and prevention (M.V., P.B., M.H.W., R.P.E., R.J.D., C.B. and I.Q.); Management (M.V., P.B., M.H.W., R.P.E., C.B. and I.Q.); Quality of life (M.V., P.B., M.H.W., R.P.E., R.J.D. and I.Q.); Outlook (M.V., P.B., M.H.W., R.P.E. and I.Q.); Overview of Primer (M.V.).

Corresponding author

Correspondence to Miikka Vikkula .

Ethics declarations

Competing interests.

All authors declare no competing interests.

Additional information

Peer review information.

Nature Reviews Disease Primers thanks M.-H. Cheng, T. Yamamoto and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary information, rights and permissions.

Reprints and permissions

About this article

Cite this article.

Brouillard, P., Witte, M.H., Erickson, R.P. et al. Primary lymphoedema. Nat Rev Dis Primers 7 , 77 (2021). https://doi.org/10.1038/s41572-021-00309-7

Download citation

Accepted : 17 September 2021

Published : 21 October 2021

DOI : https://doi.org/10.1038/s41572-021-00309-7

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Lymphatic vessels in the age of cancer immunotherapy.

  • Triantafyllia Karakousi
  • Tenny Mudianto
  • Amanda W. Lund

Nature Reviews Cancer (2024)

Etiology and treatment of cancer-related secondary lymphedema

  • Michael Bernas
  • Sara Al-Ghadban
  • Margaret L. McNeely

Clinical & Experimental Metastasis (2024)

Evaluation of Primary Lymphedema with Intranodal Lymphangiography

  • Charissa Kim
  • Jeffrey Weinstein

CardioVascular and Interventional Radiology (2024)

Cancer metastasis through the lymphatic versus blood vessels

  • Stanley P. Leong

Advances in photoacoustic imaging aided by nano contrast agents: special focus on role of lymphatic system imaging for cancer theranostics

  • Badrinathan Sridharan
  • Hae Gyun Lim

Journal of Nanobiotechnology (2023)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

new research on lymphedema

  • See us on facebook
  • See us on twitter
  • See us on youtube
  • See us on linkedin
  • See us on instagram

Study finds first possible drug treatment for lymphedema

Collaboration between two Stanford labs has resulted in the discovery of a molecular cause for lymphedema and the first possible drug treatment for it.

May 10, 2017 - By Tracie White

Woman standing in front of her garden and home

Tracey Campbell suffers from lymphedema and is participating in a clinical trial of a drug to determine whether it can treat the painful condition. Mark Williams

Tracey Campbell has lived for seven years with lymphedema, a chronic condition that causes unsightly swelling in her left leg.

The disease, which stems from a damaged lymphatic system, can lead to infections, disfigurement, debilitating pain and disability. There is no cure. The only available treatment is to wear compression garments or use massage to suppress the swelling, which can occur throughout the body in some cases. Campbell — who had two quarts of excess water in her left leg by the time she was diagnosed — has for years worn restrictive garments 24 hours a day and has spent an hour each night massaging the lymph fluid out of her leg.

Lymphedema is uncomfortable, exhausting and dangerous if left uncontrolled. As many as 10 million Americans and hundreds of millions of people worldwide suffer from the condition, many from the after-effects of cancer therapy treatments.

“There’s this extra layer of emotional burden,” said Campbell, who added that she has to be constantly vigilant to protect against infection. “All you want to be is normal.”

Now there’s new hope for a possible pharmaceutical treatment for patients like Campbell. A study led by scientists at the Stanford University School of Medicine has uncovered for the first time the molecular mechanism responsible for triggering lymphedema, as well as a drug with the potential for inhibiting that process.

The study was published May 10 in Science Translational Medicine .

“We figured out that the biology behind what has been historically deemed the irreversible process of lymphedema is, in fact, reversible if you can turn the molecular machinery around,” said Stanley Rockson , MD, professor of cardiovascular medicine and the Allan and Tina Neill Professor of Lymphatic Research and Medicine at Stanford. Rockson shares senior authorship of the study with Mark Nicolls , MD, professor of pulmonary and critical care medicine. Stanford research scientists Wen “Amy” Tian, PhD, and Xinguo Jiang, MD, PhD, share lead authorship of the study and are also affiliated with the Veterans Affairs Palo Alto Health Care System .

‘Fundamental new discovery’

“This is a fundamental new discovery,” said Nicolls, who is also a researcher at the VA Palo Alto.

Stanley Rockson

Stanley Rockson

The researchers found that the buildup of lymph fluid is actually an inflammatory response within the tissue of the skin, not merely a “plumbing” problem within the lymphatic system, as previously thought.

Working in the lab, scientists discovered that a naturally occurring inflammatory substance known as leukotriene B4, or LTB4, is elevated in both animal models of lymphedema and in humans with the disease, and that at elevated levels it causes tissue inflammation and impaired lymphatic function.

Further research in mice showed that by using pharmacological agents to target LTB4, scientists were able to induce lymphatic repair and reversal of the disease processes.

“There is currently no drug treatment for lymphedema,” Tian said. Based on results of the study, the drug bestatin, which is not approved for use in the United States but which has been used for decades in Japan to treat cancer, was found to work well as an LTB4 inhibitor, with no side effects, she said.

Based on the research, bestatin (also known as ubenimex), is being tested in a clinical trial that started in May 2016 — known as ULTRA — as a treatment for secondary lymphedema, which occurs because of damage to the lymphatic system from surgery, radiation therapy, trauma or infection. Primary lymphedema, on the other hand, is hereditary. The results of the research pertain to both types.

Rockson is principal investigator for this multisite phase-2 clinical trial.

“The cool thing about this story — which you almost never see — is that a clinical trial testing the therapy has already started before the basic research was even published,” Nicolls said. “This is the first pharmaceutical company-sponsored trial for a medical treatment of lymphedema, a condition that affects millions.”

Nicolls and Tian are co-founders of Eiccose LLC. Eiccose is now part of Eiger BioPharmaceuticals, which gets the drug from Nippon Kayaku in Japan. Eiger is sponsoring the clinical trial. Nicolls and Rockson are both scientific advisers to the company.

Two labs, two diseases

The study, which got underway about four years ago, began somewhat uniquely as a collaboration between two labs that were studying two completely different diseases. At the time, the Nicolls lab, where Tian works, was studying pulmonary hypertension. The Rockson lab was conducting lymphedema research.

Mark Nicolls

Mark Nicolls

The two teams met through SPARK , a Stanford program designed to help scientists translate biomedical research into treatments for patients.

“I was in a privileged position of seeing two faculty conducting important research and recognizing the possible link in causality,” said Kevin Grimes , MD, associate professor of chemical and systems biology and co-founder of SPARK. “It occurred to me that both diseases affected vascular tissues and had strong inflammatory components.”

“He blind-dated us,” Nicolls said. “When Amy Tian and I looked at the data from Stan’s research, Amy said, ‘It looks like it could be the same molecular process.’”

“It was an arranged marriage between us and Stan which worked out great,” Tian said.

At the time, Rockson had begun to suspect that lymphedema was an inflammatory disease. This led to his team’s discovery that the anti-inflammatory drug ketoprofen successfully helped to relieve lymphedema symptoms, although it wasn’t a perfect drug; side effects were a concern, and it remained unclear how the drug worked at the molecular level.

Meanwhile, the Nicolls lab had discovered that LTB4 was part of the cycle of inflammation and injury that keeps pulmonary hypertension progressing. When researchers blocked LTB4 in rats with the disease, their symptoms lessened and blood vessels became less clogged, lowering blood pressure in the lungs.

“When we became aware of Mark’s work, we began to realize that we were both possibly dealing with the activation of steps downstream of the 5-LO [5-lipoxygenase] pathway,” Rockson said . “This became intriguing and formed the basis of our relationship.”

Joining forces

The two teams joined forces to figure out the mechanism that triggered lymphedema, hopefully revealing a target for drug treatment in humans. After determining that ketoprofen was primarily working on the 5-LO pathway, the researchers began blocking the various endpoint pathways after 5-LO activation in mouse models of lymphedema, Rockson said.

“It turned out that, in fact, we were both dealing with the same branch, which is LTB4,” Rockson said.

When all of the sudden one of your limbs begins to swell, you want to understand what the heck is going on.

“So now it became clear we really were dealing with a very similar biological process in two different diseases,” he said. “Because of Mark’s work in pulmonary hypertension, we knew that we had an ideal form of therapy that we could try in lymphedema as well.”

The Nicolls lab had used the drug bestatin, which blocks the enzyme that generates LTB4, to reverse pulmonary hypertension disease processes. When researchers tested bestatin in the mouse lymphedema model, it worked to reverse symptoms of that disease.

“I’m still in awe,” Rockson said. “There are few situations where you take a problem at the bedside, and go into the lab, and then take discoveries back to the bedside. It’s amazingly gratifying.”

Campbell, who is now participating in the double-blinded, placebo-controlled bestatin trial at Stanford, remains hopeful.

“When all of the sudden one of your limbs begins to swell, you want to understand what the heck is going on,” she said. “It’s a tough condition that few people seem to care about, even though millions and millions suffer with it. We’re hoping for something that gives some relief.”

Other Stanford authors are research associate Jeanna Kim; former medical students Adrian Begaye, MD, and Abdullah Feroze, MD; Roham Zamanian , MD, associate professor of medicine and director of the Adult Pulmonary Hypertension Service; Gundeep Dhillon , MD, associate professor of medicine and medical director of the Stanford Lung Transplant Program; and research assistants Eric Shuffle and Allen Tu. Shuffle and Tu are affiliated with both Stanford and the VA Palo Alto.

Researchers at the Georgia Institute of Technology, Virginia Commonwealth University, the University of Michigan Health Systems and the University of Illinois at Chicago are also co-authors.

Eiger BioPharmaceuticals has licensed intellectual property developed by Tian, Rockson, Jiang, Kim and Nicolls involving the targeting of LTB4 for the treatment of lymphedema.

Stanford’s Department of Medicine supported the work.

Tracie White

About Stanford Medicine

Stanford Medicine is an integrated academic health system comprising the Stanford School of Medicine and adult and pediatric health care delivery systems. Together, they harness the full potential of biomedicine through collaborative research, education and clinical care for patients. For more information, please visit med.stanford.edu .

The majestic cell

How the smallest units of life determine our health

Stanford Medicine magazine: Majestic cell

  • Open access
  • Published: 20 November 2021

Cell therapy as a treatment of secondary lymphedema: a systematic review and meta-analysis

  • Hector Lafuente 1   na1 ,
  • Ibon Jaunarena 2 , 3   na1 ,
  • Eukene Ansuategui 4 ,
  • Arantza Lekuona 2 , 3 &
  • Ander Izeta   ORCID: orcid.org/0000-0003-1879-7401 1 , 5  

Stem Cell Research & Therapy volume  12 , Article number:  578 ( 2021 ) Cite this article

5288 Accesses

5 Citations

1 Altmetric

Metrics details

Lymphedema, the accumulation of interstitial fluid caused by poor lymphatic drainage, is a progressive and permanent disease with no curative treatment. Several studies have evaluated cell-based therapies in secondary lymphedema, but no meta-analysis has been performed to assess their efficacy.

We conducted a systematic review and meta-analysis of all available preclinical and clinical studies, with assessment of their quality and risk of bias.

A total of 20 articles using diverse cell types were selected for analysis, including six clinical trials and 14 pre-clinical studies in three species. The meta-analysis showed a positive effect of cell-based therapies on relevant disease outcomes (quantification of edema, density of lymphatic capillaries, evaluation of the lymphatic flow, and tissue fibrosis). No significant publication bias was observed.

Cell-based therapies have the potential to improve secondary lymphedema. The underlying mechanisms remain unclear. Due to relevant heterogeneity between studies, further randomized controlled and blinded studies are required to substantiate the use of these novel therapies in clinical practice.

As knowledge on the diverse lymphatic vasculature roles in health and disease progresses, it increases the lymphatic vessel relevance in understanding the physiopathology of a number of diseases [ 1 ]. Lymphedema is a chronic edema, lasting more than three months, due to the accumulation of interstitial fluid caused by poor lymphatic drainage [ 2 ]. Secondary lymphedema is due to obstruction or infiltration of the lymphatic vessels by tumors, infections (recurrent lymphangitis), obesity, surgery or overload and saturation of the lower limb venous system [ 3 ]. The most frequent cause in undeveloped countries is filariasis, while in developed countries, it is iatrogenic due to radiotherapy or surgery related to the management of malignant neoplasms (breast cancer, malignant melanoma, gyneco-urological cancer) [ 4 ]. Approximately, 30% of women with breast cancer and 20% of melanoma patients who have axillary and inguinal lymph nodes removed, develop lymphedema [ 5 , 6 ].

The accumulation of lymph in the interstitial tissue leads to remodeling of the skin and subcutaneous tissue and the accumulation of fibroadipose tissue [ 7 ]. The chronic form of lymphedema is characterized by swelling, fibrosis, accumulation of adipose tissue and infiltration of immune cells. Clinically, it can be classified into four stages: in stage 0, the condition is considered subclinical; swelling is not present. In stage I, edema is mild; fluid accumulates throughout the day but resolves overnight. In stage II, lymphedema is always present, but varies in severity. Stage III disease is characterized by persistent moderate-to-severe edema in the affected limb [ 8 ].

Lymphedema is a progressive and permanent disease for which there is no curative treatment. The standard treatment is physiotherapy (lymphatic drainage and compression bandaging) [ 9 ], although other treatments used include pharmacotherapy and surgery. More recently, reconstructive microsurgery (lympho-venous anastomosis, lymphatic vessel transplantation and autologous lymph node transplantation) has been proposed as an alternative [ 10 , 11 , 12 ].

Other potential therapies are still in development, e.g., the therapeutic potential of different growth factors, which would facilitate the regrowth of damaged, dysfunctional or obliterated lymphatics, has been investigated [ 13 , 14 ]. Among them, the role of vascular endothelial growth factor VEGF-C as a stimulant of lymphangiogenesis and mediator of lymphatic endothelial cell growth and viability has been studied [ 15 ], as well as fibroblast growth factor-2 and hepatocyte growth factor [ 16 ]. Also, the use of gene therapy via adenovirus, plasmids or even direct application of recombinant VEGF-C has been described to reduce edema in different preclinical models [ 17 , 18 , 19 ]. However, there are currently many unresolved questions, such as the lifespan of recombinant proteins, the time-limited action of gene therapy, as well as the side effects of growth factors on the blood vasculature and on the development of new tumors [ 18 , 20 ].

In the last decade, cell therapy with differentiated or progenitor cells has emerged as a new research target in the therapy of secondary lymphedema [ 21 , 22 ]. Although the cellular pathways through which stem cell therapy could help lymphedema patients are unclear, in vitro studies indicate that stem cells may differentiate into lymphatic endothelial-like cells under in vitro culture conditions and can improve interstitial fluid drainage when injected in vivo [ 13 ]. Stem cells have a wide range of therapeutic effects in terms of anti-inflammation, anti-fibrosis, anti-oxidative stress, as well as promoting the regeneration of different tissues. These properties could promote the regeneration of lymphatic vessels, rebuild lymphatic circulation and successfully treat lymphedema. Currently, several clinical and preclinical studies have evaluated the therapeutic potential of using lymphatic endothelial progenitor cells (LEPCs), embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) or mesenchymal stromal cells (MSCs) in the regeneration of lymphatic vessels. These results suggest that stem cell therapy is feasible and may promote recovery in patients with secondary lymphedema. However, stem cell transplantation has not been fully evaluated for the treatment of secondary lymphedema in clinical settings. In the present study, a meta-analysis of the available data was performed to evaluate the safety and efficacy of stem cell therapy for the treatment of secondary lymphedema.

We conducted a systematic review according to the Cochrane method [ 23 ] and SYRCLE guideline [ 24 ], and the results are reported in accordance with PRISMA guidelines [ 25 ]. The protocol for this review was registered on the International prospective register of systematic reviews website ( https://www.crd.york.ac.uk/prospero/ ) with two separate IDs (CRD42020180348 for preclinical studies and CRD42019130951 for clinical studies).

Search strategy and literature selection

Studies of cell therapy as a treatment of secondary lymphedema were identified from Medline, Web of Science, EMBASE, and The Cochrane library with no language or time restrictions using these search terms: lymphedema, lymphoedema, lymphangiogenesis, lymphatic diseases, lymphatic vessels, lymph nodes, stem cells, stromal cells, mesenchymal stem cells, cell- and tissue-based therapy, cell transplantation, and regenerative medicine. We identified all relevant studies or trials regardless of language or publication status (published, unpublished, in press, and ongoing). Two independent searches were conducted on January 2021, one with the inclusion criteria: pre-clinical studies and all animal models, and the other one with the inclusion criteria: clinical trials and prospective controlled studies in human.

After developing a search strategy for each database and collecting the citations, the search results were combined. The first selection was made using only the title and abstract of the studies. To avoid biases in the selection process, two observers independently screened articles for relevance. The criteria used for the first screening were based on the search components: (SC1) intervention (only cell therapies were included); (SC2) disease of interest (secondary lymphedema); (SC3) type of study (only pre-clinical studies, randomized controlled clinical trials and prospective controlled studies were included. The ex vivo studies, in vitro studies, or in silico studies were not included. Non-intervention studies, no control group, co-intervention studies and studies with other outcomes were not included); and (SC4) publication types (reviews and conference abstracts were not included). Only clearly irrelevant citations were removed. Citations resulting from the first screening underwent a second screening based on the predefined inclusion and exclusion criteria. Throughout the potentially relevant article selection process, the reasons for the removal of citations were documented and reported to facilitate transparency and to independently examine the accuracy of the study removal. Two independent reviewers performed all stages of the review process. Discrepancies were resolved by consensus. The flow diagram of search strategy and literature selection is shown in Fig.  1 for preclinical studies and in Fig.  2 for clinical studies.

figure 1

PRISMA flow diagram of search strategy and literature selection for preclinical studies

figure 2

PRISMA flow diagram of search strategy and literature selection for clinical studies

Assessment of study quality and risk of bias

Quality and risk of bias was assessed for clinical trials by use of Cochrane's risk of bias tool [ 26 ], and for non-randomized studies by use of NewcastleOttawa's risk of bias tool [ 27 ]. For preclinical studies, we used SYRCLE Risk of Bias tool [ 28 ]. Two authors independently assessed the risk of bias of the included studies. A third author was consulted to resolve discrepancies related to risk of bias.

Besides, to overcome the fact that there were too many items classified as “unclear” because of the poor description of details on experimental design and methods, we included three items as other bias: (a) inappropriate influence of funders, (b) mention of randomization at any level, and (c) mention of blinding at any level. For inappropriate influence of funders, “Yes” indicated non-industry source of funding, no funding, or no conflict of interest, “No” indicated the study was funded by industry- or author-mentioned conflict of interests, “unclear” indicated funding source or conflict of interest was not mentioned. For mention of randomization or blinding, “Yes” indicated reported and “No” indicated unreported.

An overall score was calculated by adding all the items scores as yes equals one, while no and not applicable equal zero. A score was given for every paper to classify them as poor, fair, or good conducted studies, where a score from 0 to 5 was considered poor, 6–9 as fair, and 10–14 as good.

Data extraction

For clinical studies, details about the study design, cell type, primary outcome assessment, follow-up time and results were extracted.

Data on animal model characteristics (animal species, total sample size, total groups, number of animals in control group, number of animals in intervention group), lymphedema model (tail, hindlimb, etc.), cell administration characteristics (cell type, source, as well as administration route, dose, timing and anatomical site of intervention), and primary outcome measures (evaluation of the lymphatic flow, quantification of edema, density of lymphatic capillaries and tissue fibrosis) were extracted.

For included articles, all independent comparisons were identified. Replications were also collected separately. Information on primary outcome was extracted from both text and graphs, when raw data or mean/median/incidence, SD/SE were reported or recalculated. In several studies, the results were adapted to be able to be analyzed with the rest of the studies. Gsys 2.4.6. software (Hokkaido University Nuclear Reaction Data Centre) was used to obtain values from graphs. When the number of animals was reported as a range, the lowest group size was collected. When no clear data could be extracted, the report was excluded from further meta-analysis.

Statistical analyses

Quantitative analysis was conducted using Review Manager (RevMan) version 5.3 software (The Nordic Cochrane Centre, The Cochrane Collaboration, Copenhagen, Denmark). Treatment effects were first calculated separately for each study outcome. For all analyses, a random effect, inverse variance model was used to calculate standardized mean differences (SMD) and 95% confidence intervals (CI). Because most animal experiments use fewer than ten animals per group, we used Hedge's G effect sizes (which is based on Cohen’s D but includes a correction factor for small sample size bias) for SMD analyses. The effect of heterogeneity (I 2 ) was used to measure the degree of inconsistency across pooled studies due to variability rather than chance, with larger values indicative of high heterogeneity (0–25% is considered to reflect very low heterogeneity; 25–50% reflects low heterogeneity; 50–75% reflects moderate heterogeneity; > 75% reflects high heterogeneity). Considering the anticipated heterogeneity, random effects models were used to conducted meta-analysis. Mean effect size, 95% confidence intervals (95% CI), significance, weight and forest plots were analyzed by the inverse variance method and the standard mean differences. The possibility of publication bias was assessed by analyzing funnel plot asymmetry (with trim-and-fill). The trim-and-fill method provides an estimate of the number of missing studies, and also provides an estimated intervention effect ‘adjusted’ for the publication bias (based on the filled studies). Finally, to explore sources of heterogeneity, stratified meta-analysis and meta-regression were performed.

A total of 20 articles were selected for analysis. Six of these were clinical studies [ 29 , 30 , 31 , 32 , 33 , 34 ], including a randomized clinical trial, three nonrandomized clinical trials and two prospective controlled studies (Table 1 ). A case report and an observational study were excluded from the analysis. Five of them studied the effect of cell therapy on the upper limb, while the other studied lower limb edema. Mesenchymal stromal cells (MSCs) of different origins were used: three studies used bone marrow-derived MSCs (BM-MSCs) and the remaining three used adipose-derived MSCs (ADSCs). The follow‐up period ranged from 3 to 12 months.

Fourteen animal studies [ 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 ] were included in the analysis (Table 2 ). These studies included three different animal models (mouse, rat and rabbit). In murine models, tail, hind limb, back skin flap, or lymph node transplantation was used. In rabbit models, hind limb was used. The cell types used included stem or progenitor cells (BM-MSCs, ADSCs, muscle‐derived stem cells and multipotent progenitor cells) and differentiated cells (lymphatic endothelial cells and T reg cells), and the number of cells used ranged from 10 4 to 10 7 . The follow‐up period ranged from 14 days to 6 months.

Assessment of study quality and risk of bias

The study design, including details of the method of randomization of subjects to treatment groups, criteria for eligibility in the study, blinding, method of assessing the outcome, and handling of protocol deviations are important features defining study quality. Due to the high risk of bias (data not shown) and the fact that only one of the human studies included was a properly blinded randomized controlled trial, a meta-analysis was not performed for clinical studies.

None of the pre-clinical studies had published protocols nor were registered with CAMARADES (University of Edinburgh, Scotland). Therefore, the selective outcome reporting item on the SYRCLE tool was scored as “unclear.” There was insufficient information reported for many of the remaining nine questions which were scored as “unclear.” Several studies reported any randomization, although details were not given. 50% reported any blinding, either of investigators, animal handlers or outcome assessors. Overall, all studies had significant risks of bias according to the SYRCLE tool (Fig.  3 ), but these were not sufficiently remarkable as to be excluded from any analyses. Only two studies (Conrad et al. and Zhou et al.) did not report sample size for control and intervention groups, and thus those studies were not included in the meta-analysis.

figure 3

Assessment of bias in 14 animal studies using the SYRCLE risk of bias tool

Meta-analysis and effect evaluation

Meta-analysis was performed for outcomes that had data in at least three studies. The outcomes analyzed were: quantification of edema, density of lymphatic capillaries, evaluation of the lymphatic flow, and tissue fibrosis.

Quantification of edema

Eleven studies were included to investigate the effect of cell therapy treatment on edema reduction in secondary lymphedema. The pooled estimate showed a significant decrease in edema (SMD 3.18; 95% CI 1.798, 4.567 ( p  < 0.001); however, between-study heterogeneity was very high ( I 2  = 92%; Fig.  4 ). Subgroup analysis as a function of the animal model used did not reduce heterogeneity. Subgrouping as a function of cell type indicated a similar reduction in edema with stem or progenitor cell treatment than differentiated cell treatment, with no evidence of heterogeneity in this subgroup (Table 3 ). Random effect meta-regression analysis was applied to estimate functional relationship of effect size on follow-up time. The regression coefficient was -0.02, and it was statistically insignificant ( p  > 0.05). These results indicated that the effect of follow-up time on the effect size was insignificant. Consistently, a linear relationship was not found (Fig.  5 ).

Density of lymphatic capillaries

figure 4

Forest plot of the effects of cell therapy on the edema reduction

figure 5

Meta-regression analysis of follow-up time on effect of the cell therapy on the edema reduction

Ten studies were included to investigate the effect of the cell therapy treatment on the lymphatic regeneration in secondary lymphedema. The overall pooled analysis showed a significant increase in lymphatic vessel density in experimental group versus control group (SMD 6.35; 95% CI 4.115, 8.581; p  = 0.00). However, the test for heterogeneity was significant ( I 2  = 93%; Fig.  6 ). Subgroup analysis as a function of animal model did not show differences between groups and did not reduce heterogeneity. Analysis as a function of cell type could not be carried out due to the small number of studies (Table 3 ). Random effect meta-regression analysis was applied and a regression coefficient of 0.03 was found, which was statistically insignificant ( p  > 0.05). These results indicated that follow-up time does not explain the heterogeneity found between the studies (Fig.  7 ).

Evaluation of the lymphatic flow

figure 6

Forest plot of the effects of cell therapy on the lymphatic regeneration

figure 7

Meta-regression analysis of follow-up time on effect of the cell therapy on the lymphatic regeneration

Four studies were included to investigate the effect of the cell therapy treatment on the lymphatic perfusion restoration in secondary lymphedema. The pooled estimate suggested a significant improvement of lymphatic perfusion (SMD 2.49; 95% CI 0.583, 4.394 ( p  = 0.01); I 2  = 88%, Fig.  8 ). Due to the limited availability of data, it was not possible to conduct subgroup analyses. Using random effects meta-regression analysis, the regression coefficient was -0.04, which was not statistically significant ( p  > 0.05). The results indicated that follow-up time does not explain the heterogeneity between studies (Fig.  9 ).

Tissue fibrosis

figure 8

Forest plot of the effects of cell therapy on the lymphatic perfusion restoration

figure 9

Meta-regression analysis of follow-up time on effect of the cell therapy on lymphatic perfusion restoration

Only three studies were included to investigate the effect of cell therapy treatments on the fibrosis reduction in secondary lymphedema. The analysis of the effect size showed a significant reduction in the fibrosis (SMD 4.39; 95% CI 1.439, 7.352 ( p  < 0.01); I 2  = 82%, Fig.  10 ). Subgroup analysis was not carried out due to the small number of studies included. The regression coefficient was found to be -0.19 and statistically insignificant ( p  > 0.05) using random effect meta-regression analysis. The study's heterogeneity was not explained by the follow-up period, according to the findings (Fig.  11 ).

figure 10

Forest plot of the effects of cell therapy on the fibrosis reduction

figure 11

Meta-regression analysis of follow-up time on effect of the cell therapy on the fibrosis reduction

Publication Bias

The publication bias evaluation (Funnel plots) for the meta-analysis of lymphatic regeneration (ten studies) is shown in Fig.  12 . After adjusting for missing studies, we found that the point estimate of the overall effect size continued to show a positive effect in favor of cell therapy (SMD 5.65 [CI 95% 2.48–8.83]). No significant publication bias was observed for edema reduction, lymphatic perfusion restoration and fibrosis reduction. This confirms that if there were a publication bias, the effect of cell therapy on secondary lymphedema would not be modified.

figure 12

Funnel plot (with trim-and-fill) analysis of the cell therapy on the lymphatic regeneration

In the present study, we performed a systematic review and meta-analysis to evaluate the safety and efficacy of stem cell therapy for the treatment of secondary lymphedema, both in preclinical and clinical studies. We found that cell therapy proved to generate a robust beneficial effect in animal models of secondary lymphedema. Although several in vitro and in vivo studies have reported beneficial effects of cell therapy against secondary lymphedema [ 21 , 49 , 50 ], a formal meta-analysis that assesses the regenerative activity of cell therapy in animal models of secondary lymphedema had not been performed.

Animal studies are critical for understanding disease processes and assessing the safety and effectiveness of treatments. Animal trials, however, are inherently heterogeneous, even more than clinical trials. Understanding sources of heterogeneity and their influence on effect size is critical to successfully translating preclinical findings to human diseases [ 51 ].

No animal model mimics perfectly the pathophysiology of human lymphedema [ 52 ], mainly because animals present higher regenerative capacity and it is difficult to classify the severity of edema [ 49 ]. There are also significant differences between models [ 52 ]. Although the tail model yields more consistent results than the hindlimb model, lymphedema resolves naturally over time, thus confounding results of additional interventions [ 53 ]. Of note, the current lack of standardization in study design and outcome measures make it hard to compare preclinical results. Despite the experimental heterogeneity of available studies, insight from animal models has shed light on the molecular mechanisms underlying lymphedema, e.g., lymphangiogenesis [ 54 ], fibrosis [ 55 ] and inflammation [ 56 , 57 ].

Regarding the human studies, only six studies were identified and included for the analysis, and since only one of them is a randomized clinical trial, it was not possible to perform the meta-analysis. Furthermore, the difference in the follow-up period between the studies did not allow us to confirm the observed effect of cell therapy on secondary lymphedema. However, it should be noted that the current human studies showed promising results of BM-MSCs [ 29 , 30 , 33 ] and ADSCs [ 31 , 32 , 34 ] in terms of reduction in edema, relief of symptoms, and an improved quality-of-life. Although no adverse effects related to cancer have been observed, the potential risk of cancer recurrence of using stem cells in the treatment of secondary lymphedema should be studied. A recently published Phase I study has found no evidence of breast cancer recurrence at 4-year follow up [ 58 ]. However, to further substantiate this relevant safety concern, a greater number of patients must be followed up longer-term in randomized clinical studies to formally rule out any contribution of stem cell transplants to cancer recurrence.

In the preclinical studies included in the review, different cell types have been tested for secondary lymphedema. In all cases, stem/progenitor cells have shown promise in halting lymphedema progression, sometimes even reverse the pathological process. However, the underlying mechanisms are not clear. It is speculated that stem cells may differentiate into lymphatic endothelial progenitor cells that in turn generate new lymphatics, or secrete cytokines to induce lymphangiogenesis [ 59 ]. Several studies have combined cell therapy with growth factors, such as VEGF‐C [ 36 ] and PRP [ 41 ] which are thought to costimulate lymphangiogenesis. Co-transplantation with lymphatic endothelial cells (LECs) [ 40 ] may guide differentiation of stem cells to LEPCs. Combination of cell therapy with lymph node transfer [ 44 ] improved both lymphangiogenesis and lymphatic flow. Of course, immune modulation could be another cell-based approach to tackle this disease. Gousopoulos et al. have shown that treatment with T reg cells reversed major hallmarks of lymphedema, such as edema, inflammation, and fibrosis [ 43 ]. Cell-based therapies seem thus to improve lymphedema's outcomes (edema reduction, lymphatic regeneration, lymphatic perfusion restoration, and fibrosis reduction), and the effect is seen across multiple species (mouse, rat, and rabbit), so that translation of these novel therapies to humans seems to be warranted.

The main limitations of this study are (i) the significant methodological differences between studies, especially the animal model used, the number of infused cells and timing of follow-up; (ii) small sample sizes and small study dataset for the meta-analysis, with most studies having no pre-published protocols or sample size estimations; (iii) the included studies had moderate or unknown bias risks, mainly due to poor reporting detail, and (iv) lack of operator blindness and randomization. These limitations emphasize the importance of applying more rigor to reporting standards and publishing in vivo experimental protocols [ 60 ].

Conclusions

Cell-based therapies have the potential to improve secondary lymphedema through their effects on the edema, lymphangiogenesis and fibrosis. The underlying mechanisms remain unclear. Due to relevant heterogeneity between studies, further randomized controlled and blinded studies are required to substantiate the use of these novel therapies in clinical practice.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

Adipose-derived stem cells

Bone marrow-derived mesenchymal stem cells

Embryonic stem cells

Induced pluripotent stem cells

Effect of heterogeneity

Lymphatic endothelial cells

Lymphatic endothelial progenitor cells

Mesenchymal stromal cells

Standardized mean differences

95% Confidence intervals

Oliver G, Kipnis J, Randolph GJ, Harvey NL. The lymphatic vasculature in the 21 st century: novel functional roles in homeostasis and disease. Cell. 2020;182(2):270–96.

PubMed   PubMed Central   CAS   Google Scholar  

Manrique OJ, Bustos SS, Ciudad P, Adabi K, Chen WF, Forte AJ, et al. Overview of lymphedema for physicians and other clinicians: a review of fundamental concepts. Mayo Clinic Proc. 2020; in press. https://doi.org/10.1016/j.mayocp.2020.01.006 .

Article   Google Scholar  

Brix B, Sery O, Onorato A, Ure C, Roessler A, Goswami N. Biology of lymphedema. Biology. 2021;10(4):261. https://doi.org/10.3390/biology10040261 .

Article   PubMed   PubMed Central   Google Scholar  

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):5138–49.

PubMed   Google Scholar  

McLaughlin SA, Wright MJ, Morris KT, Giron GL, Sampson MR, Brockway JP, et al. Prevalence of lymphedema in women with breast cancer 5 years after sentinel lymph node biopsy or axillary dissection: objective measurements. J Clin Oncol. 2008;26(32):5213–9.

PubMed   PubMed Central   Google Scholar  

Williams AF, Franks PJ, Moffatt CJ. Lymphoedema: estimating the size of the problem. Palliat Med. 2005;19(4):300–13.

Stritt S, Koltowska K, Makinen T. Homeostatic maintenance of the lymphatic vasculature. Trends Mol Med. 2021;27(10):955–70.

PubMed   CAS   Google Scholar  

Morgan CL, Lee BB. Classification and staging of lymphedema. Lymphedema: diagnosis and treatment. London: Springer London; 2008. p. 21–30.

Google Scholar  

Brandao ML, Soares H, Andrade MDA, Faria A, Pires RS. Efficacy of complex decongestive therapy for lymphedema of the lower limbs: a systematic review. J Vasc Bras. 2020;19:e20190074.

Tang JB, Landin L, Cavadas PC, Thione A, Chen J, Pons G, et al. Unique techniques or approaches in microvascular and microlymphatic surgery. Clin Plast Surg. 2020;47(4):649–61.

Chen K, Sinelnikov MY, Shchedrina MA, Mu L, Lu P. Surgical management of postmastectomy lymphedema and review of the literature. Ann Plastic Surg. 2021;86(3S Suppl 2):S173–6.

CAS   Google Scholar  

Gasteratos K, Morsi-Yeroyannis A, Vlachopoulos NC, Spyropoulou GA, Del Corral G, Chaiyasate K. Microsurgical techniques in the treatment of breast cancer-related lymphedema: a systematic review of efficacy and patient outcomes. Breast Cancer (Tokyo, Japan). 2021;28(5):1002–15.

Dayan JH, Ly CL, Kataru RP, Mehrara BJ. Lymphedema: pathogenesis and novel therapies. Annu Rev Med. 2018;69:263–76.

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.

Cheung L, Han J, Beilhack A, Joshi S, Wilburn P, Dua A, et al. An experimental model for the study of lymphedema and its response to therapeutic lymphangiogenesis. BioDrugs. 2006;20(6):363–70.

Saito Y, Nakagami H, Morishita R, Takami Y, Kikuchi Y, Hayashi H, et al. Transfection of human hepatocyte growth factor gene ameliorates secondary lymphedema via promotion of lymphangiogenesis. Circulation. 2006;114(11):1177–84.

Enholm B, Karpanen T, Jeltsch M, Kubo H, Stenback F, Prevo R, et al. Adenoviral expression of vascular endothelial growth factor-C induces lymphangiogenesis in the skin. Circ Res. 2001;88(6):623–9.

Saaristo A, Veikkola T, Tammela T, Enholm B, Karkkainen MJ, Pajusola K, et al. Lymphangiogenic gene therapy with minimal blood vascular side effects. J Exp Med. 2002;196(6):719–30.

Szuba A, Skobe M, Karkkainen MJ, Shin WS, Beynet DP, Rockson NB, et al. Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEB J. 2002;16(14):1985–7.

Goldman J, Le TX, Skobe M, Swartz MA. Overexpression of VEGF-C causes transient lymphatic hyperplasia but not increased lymphangiogenesis in regenerating skin. Circ Res. 2005;96(11):1193–9.

Chen K, Sinelnikov MY, Reshetov IV, Timashev P, Gu Y, Mu L, et al. Therapeutic potential of mesenchymal stem cells for postmastectomy lymphedema: a literature review. Clin Transl Sci. 2021;14(1):54–61.

Hu LR, Pan J. Adipose-derived stem cell therapy shows promising results for secondary lymphedema. World J Stem Cells. 2020;12(7):612–20.

Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA. Cochrane handbook for systematic reviews of interventions version 6.2 (updated February 2021). Cochrane; 2021. Available from www.training.cochrane.org/handbook .

Hooijmans CR, Wever KE, de Vries RBM. SYRCLEs starting guide for systematic reviews of preclinical animal interventions studies; 2016. Available from: www.radboudumc.nl/getmedia/4b1cbcb8-d9b6-45d5-b9fe-c92e43ab1dd4/SYRCLE-starting-guide-tool.aspx .

Moher D, Liberati A, Tetzlaff J, Altman DG, Grup P. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Int J Surg. 2010;8(5):336–41.

Sterne JAC, Savovic J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:I4898.

Wells GA, Shea B, O’Connell D, Peterson J, Welch V, Losos M, Tugwell P. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. 2013. Available from: http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp .

Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14:43.

Hou C, Wu X, 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. 2008;38(10):670–4.

Maldonado GEM, Perez CAA, Covarrubias EEA, Cabriales SAM, Leyva LA, Perez JCJ, et al. Autologous stem cells for the treatment of post-mastectomy lymphedema: A pilot study. Cytotherapy. 2011;13(10):1249–55.

Toyserkani NM, Jensen CH, Sheikh SP, Sorensen JA. Cell-assisted lipotransfer using autologous adipose-derived stromal cells for alleviation of breast cancer-related lymphedema. Stem Cells Transl Med. 2016;5(7):857–9.

Toyserkani NM, Jensen CH, Andersen DC, Sheikh SP, Sorensen JA. Treatment of breast cancer-related lymphedema with adipose-derived regenerative cells and fat grafts: a feasibility and safety study. Stem Cells Transl Med. 2017;6(8):1666–72.

Ismail AM, Abdou SM, Abdelnaby AY, Hamdy MA, El Saka AA, 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. 2018;16(3):270–7.

Toyserkani NM, Jensen CH, Tabatabaeifar S, Jorgensen MG, Hvidsten S, Simonsen JA, et al. Adipose-derived regenerative cells and fat grafting for treating breast cancer-related lymphedema: lymphoscintigraphic evaluation with 1 year of follow-up. J Plast Reconstr Aesthetic Surg. 2019;72(1):71–7.

Conrad C, Niess H, Huss R, Huber S, von Luettichau I, Nelson PJ, et al. Multipotent mesenchymal stem cells acquire a lymphendothelial phenotype and enhance lymphatic regeneration in vivo. Circulation. 2009;119(2):281–9.

Hwang JH, Kim IG, Piao S, Lee DS, Lee TS, Ra JC, et al. Therapeutic lymphangiogenesis using stem cell and VEGF-C hydrogel. Biomaterials. 2011;32(19):4415–23.

Zhou H, Wang M, Hou C, Jin X, 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. 2011;41(7):841–6.

Shimizu Y, Shibata R, Shintani S, Ishii M, Murohara T. Therapeutic lymphangiogenesis with implantation of adipose-derived regenerative cells. J Am Heart Assoc. 2012;1(4):e000877.

Park HS, Jung IM, Choi GH, Hahn S, Yoo YS, Lee T. Modification of a rodent hindlimb model of secondary lymphedema: Surgical radicality versus radiotherapeutic ablation. BioMed research international. 2013;2013 (no pagination)(208912).

Kawai Y, Shiomi H, Abe H, Naka S, Kurumi Y, Tani T. Cell transplantation therapy for a rat model of secondary lymphedema. J Surg Res. 2014;189(1):184–91.

Ackermann M, Wettstein R, Senaldi C, Kalbermatten DF, Konerding MA, Raffoul W, et al. Impact of platelet rich plasma and adipose stem cells on lymphangiogenesis in a murine tail lymphedema model. Microvasc Res. 2015;102:78–85.

Yoshida S, Hamuy R, Hamada Y, Yoshimoto H, Hirano A, Akita S. Adipose-derived stem cell transplantation for therapeutic lymphangiogenesis in a mouse secondary lymphedema model. Regen Med. 2015;10(5):549–62.

Gousopoulos E, Proulx ST, Bachmann SB, Scholl J, Dionyssiou D, Demiri E, et al. Regulatory T cell transfer ameliorates lymphedema and promotes lymphatic vessel function. JCI insight. 2016;1(16):e89081.

Hayashida K, Yoshida S, Yoshimoto H, Fujioka M, Saijo H, Migita K, et al. 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. 2017;139(3):639–51.

Beerens M, Aranguren XL, Hendrickx B, Dheedene W, Dresselaers T, Himmelreich U, et al. Multipotent adult progenitor cells support lymphatic regeneration at multiple anatomical levels during wound healing and lymphedema. Sci Rep. 2018;8(1):3852.

Bucan A, Dhumale P, Jorgensen MG, Dalaei F, Wiinholt A, Hansen CR, et al. Comparison between stromal vascular fraction and adipose derived stem cells in a mouse lymphedema model. J Plast Surg Hand Surg. 2020;54(5):302–11.

Dai T, Jiang Z, Cui C, Sun Y, Lu B, Li H, et al. The roles of podoplanin-positive/podoplanin-negative cells from adipose-derived stem cells in lymphatic regeneration. Plast Reconstr Surg. 2020;145(2):420–31.

Ogino R, Hayashida K, Yamakawa S, Morita E. Adipose-derived stem cells promote intussusceptive lymphangiogenesis by restricting dermal fibrosis in irradiated tissue of mice. Int J Mol Sci. 2020;21(11):3885. https://doi.org/10.3390/ijms21113885 .

Article   PubMed Central   CAS   Google Scholar  

Chen CE, Chiang NJ, Perng CK, Ma H, Lin CH. Review of preclinical and clinical studies of using cell-based therapy for secondary lymphedema. J Surg Oncol. 2020;121(1):109–20.

Li ZJ, Yang E, Li YZ, Liang ZY, Huang JZ, Yu NZ, et al. Application and prospect of adipose stem cell transplantation in treating lymphedema. World J Stem Cells. 2020;12(7):676–87.

Vesterinen HM, Sena ES, Egan KJ, Hirst TC, Churolov L, Currie GL, et al. Meta-analysis of data from animal studies: a practical guide. J Neurosci Methods. 2014;221:92–102.

Hadrian R, Palmes D. Animal models of secondary lymphedema: new approaches in the search for therapeutic options. Lymphat Res Biol. 2017;15(1):2–16.

Frueh FS, Gousopoulos E, Rezaeian F, Menger MD, Lindenblatt N, Giovanoli P. Animal models in surgical lymphedema research–a systematic review. J Surg Res. 2016;200(1):208–20.

Lee YJ. Cell fate determination of lymphatic endothelial cells. Int J Mol Sci. 2020;21(13):4790. https://doi.org/10.3390/ijms21134790 .

Zhou C, Su W, Han H, Li N, Ma G, Cui L. Mouse tail models of secondary lymphedema: fibrosis gradually worsens and is irreversible. Int J Clin Exp Pathol. 2020;13(1):54–64.

Kataru RP, Baik JE, Park HJ, Wiser I, Rehal S, Shin JY, et al. Regulation of immune function by the lymphatic system in lymphedema. Front Immunol. 2019;10:470.

Yuan Y, Arcucci V, Levy SM, Achen MG. Modulation of immunity by lymphatic dysfunction in lymphedema. Front Immunol. 2019;10:76.

Jorgensen MG, Toyserkani NM, Jensen CH, Andersen DC, Sheikh SP, Sorensen 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. 2021;10(6):844–54.

Religa P, Cao R, Bjorndahl M, Zhou Z, Zhu Z, Cao Y. Presence of bone marrow-derived circulating progenitor endothelial cells in the newly formed lymphatic vessels. Blood. 2005;106(13):4184–90.

Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, et al. The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. BMJ Open Sci. 2020;4(1):e100115.

Download references

Acknowledgements

Not applicable.

This work was supported by grants from the Department of Health of the Basque Government (2020111004, 2020333021, 2019222008, 2018222032 and 2017222004), Diputación Foral de Gipuzkoa, Instituto de Salud Carlos III (PI19/01621), cofunded by the European Union (European Regional Development Fund/ European Science Foundation, Investing in your future) and the Department of Economic Development of the Basque Government (Elkartek program).

Author information

Hector Lafuente and Ibon Jaunarena have contributed equally to this work

Authors and Affiliations

Tissue Engineering Group, Biodonostia Health Research Institute, 20014, San Sebastián, Spain

Hector Lafuente & Ander Izeta

Gynecology Oncology Unit, Donostia University Hospital, 20014, San Sebastián, Spain

Ibon Jaunarena & Arantza Lekuona

Obstetrics and Gynaecology Group, Biodonostia Health Research Institute, 20014, San Sebastián, Spain

Clinical Epidemiology Group, Biodonostia Health Research Institute, 20014, San Sebastián, Spain

Eukene Ansuategui

School of Engineering, Tecnun-University of Navarra, 20009, San Sebastián, Spain

Ander Izeta

You can also search for this author in PubMed   Google Scholar

Contributions

EA and AL developed a search strategy for each database and collected the citations; HL and IJ performed the assessment of study quality and risk of bias, the data extraction and statistical analysis; HL and AI were major contributors in writing the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ander Izeta .

Ethics declarations

Ethics approval and consent to participate, consent for publication, competing interests.

The authors declare that they have no competing interest.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article.

Lafuente, H., Jaunarena, I., Ansuategui, E. et al. Cell therapy as a treatment of secondary lymphedema: a systematic review and meta-analysis. Stem Cell Res Ther 12 , 578 (2021). https://doi.org/10.1186/s13287-021-02632-y

Download citation

Received : 15 September 2021

Accepted : 16 October 2021

Published : 20 November 2021

DOI : https://doi.org/10.1186/s13287-021-02632-y

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Lymphatic vasculature
  • Regeneration
  • Regenerative medicine
  • Systematic review
  • Meta-analysis

Stem Cell Research & Therapy

ISSN: 1757-6512

  • Submission enquiries: Access here and click Contact Us
  • General enquiries: [email protected]

new research on lymphedema

Switch language:

CTA

Koya’s NPCD device proves more effective than APCDs in lymphedema

Patients using the Dayspring system saw a greater reduction in limb volume and experienced improved quality of life compared to APCDs.

  • Share on Linkedin
  • Share on Facebook

new research on lymphedema

Koya Medical has shared data highlighting that its non-pneumatic compression device (NPCD) demonstrated better efficacy and improved quality of life in patients with lower extremity lymphedema.

The NPCD device, dubbed Dayspring, is a non-pneumatic treatment that provides continuous, static compression to manage lymphedema. The system comes with a mesh garment made from Koya’s patented Flexframe technology, a rechargeable controller, and a mobile app.

Go deeper with GlobalData

ReportsLogo

LOA and PTSR Model - Camidanlumab Tesirine in Myelodysplastic Syndrome

Loa and ptsr model - camidanlumab tesirine in myeloproliferative di..., data insights.

The gold standard of business intelligence.

Find out more

Data from the TEAYS study (NCT05507346), published in the Journal of Vascular Surgery , found that Dayspring outperformed advanced pneumatic compression device (APCD) – the standard of care for treating lymphedema – in several areas. Patients using Dayspring saw a significantly greater reduction in limb volume (369.9mL versus 83.1mL) and experienced improved quality of life, with a mean lymphedema quality of life questionnaire (LYMQOL) score increase of 1.01 compared to 0.17 for APCD.

According to US-based company Koya, TEAYS is the eighth study investigating Dayspring since its Food and Drug Administration (FDA) clearance in May 2021. The study enrolled 71 patients with confirmed lower extremity lymphedema, comparing Dayspring with APCDs.

The gold standard for lymphedema treatment, APCDs use air-filled chambers to deliver intermittent pneumatic pressure in cycles, compared to the continuous, static compression provided by NPCDs. This mechanism actively pumps fluid away from the limb. However, NPCDs may be more comfortable and easier to use compared to APCDs, promoting better long-term adherence in some patients. Data from this study demonstrated that Dayspring showed superior adherence of 81%, compared to 56% with APCD.

The TEAYS study’s lead investigator and associate professor of surgery at the University of Tennessee Health Science Center Michael Barfield said: “The introduction of Dayspring represents a clinically differentiated and therapeutically distinct advancement in the treatment of lower extremity lymphedema.

How well do you really know your competitors?

Access the most comprehensive Company Profiles on the market, powered by GlobalData. Save hours of research. Gain competitive edge.

new research on lymphedema

Your download email will arrive shortly

Not ready to buy yet? Download a free sample

We are confident about the unique quality of our Company Profiles. However, we want you to make the most beneficial decision for your business, so we offer a free sample that you can download by submitting the below form

“Patients participating in the TEAYS study expressed a strong preference for the Dayspring treatment over advanced pneumatic compression devices. The ability for patients to remain mobile while receiving effective treatment is a game-changer in improving adherence and overall quality of life.” 

In November 2023, Koya secured $30m in financing led by healthcare investment company OrbiMed, to support the expansion and strategic growth of Dayspring. This followed on from a February 2022 Series B funding round where Koya secured $26m, led by 3×5 Partners.

Sign up for our daily news round-up!

Give your business an edge with our leading industry insights.

Upcoming Event

12th annual clinical trial supply nordics 2024.

This is the unmissable event that the pharmaceutical biotechnology and medical device communities need to come together and discuss strategies...

More Relevant

 alt=

Boehringer and CDR-Life report data from geographic atrophy treatment trial

Vesper bio reports data from phase i frontotemporal dementia treatment trial, nice approves new osteoporosis treatment for high-risk patients in the uk, nh theraguix advances phase ii brain metastases treatment trial, sign up to the newsletter: in brief, your corporate email address, i would also like to subscribe to:.

I consent to Verdict Media Limited collecting my details provided via this form in accordance with Privacy Policy

Thank you for subscribing

View all newsletters from across the GlobalData Media network.

new research on lymphedema

Current diagnostic and quantitative techniques in the field of lymphedema management: a critical review

  • Open access
  • Published: 05 September 2024
  • Volume 41 , article number  241 , ( 2024 )

Cite this article

You have full access to this open access article

new research on lymphedema

  • Mary Vargo   ORCID: orcid.org/0000-0002-6286-6876 1 ,
  • Melissa Aldrich   ORCID: orcid.org/0000-0002-8114-7607 2 ,
  • Paula Donahue   ORCID: orcid.org/0000-0003-4656-6539 3 ,
  • Emily Iker   ORCID: orcid.org/0000-0002-6882-1089 4 ,
  • Louise Koelmeyer   ORCID: orcid.org/0000-0002-2736-2330 5 ,
  • Rachelle Crescenzi   ORCID: orcid.org/0000-0003-1008-4570 6 &
  • Andrea Cheville   ORCID: orcid.org/0000-0001-7668-6115 7  

Lymphedema evaluation entails multifaceted considerations for which options continue to evolve and emerge. This paper provides a critical review of the current status of diagnostic and quantitative measures for lymphedema, from traditional and novel bedside assessment tools for volumetric and fluid assessment, to advanced imaging modalities. Modalities are contrasted with regard to empirical support and feasibility of clinical implementation. The manuscript proposes a grid framework for comparing the ability of each modality to quantify specific lymphedema characteristics, including distribution, dysmorphism, tissue composition and fluid content, lymphatic anatomy and function, metaplasia, clinical symptoms, and quality of life and function. This review additionally applies a similar framework approach to consider how well assessment tools support important clinical needs, including: (1) screening, (2) diagnosis and differential diagnosis, (3) individualization of treatment, and (4) monitoring treatment response. The framework highlights which clinical needs are served by an abundance of assessment tools and identifies others that have problematically few. The framework clarifies which tools have greater or lesser empirical support. The framework is designed to assist stakeholders in selecting appropriate diagnostic and surveillance modalities, gauging levels of confidence when applying tools to specific clinical needs, elucidating overarching patterns of diagnostic and quantitative strengths and weaknesses, and informing future investigation.

Explore related subjects

  • Medical Imaging

Avoid common mistakes on your manuscript.

Introduction

Lymphedema diagnostic methods comprise an area of vigorous current research, with multifaceted priorities including early detection, establishing diagnosis and pathophysiology, localization, guiding intervention, and surveillance of severity and response to therapy [ 1 ]. Clinical use requires weighing empirical standards including favorable reliability, validity, diagnostic accuracy, precision, and sensitivity, as well as meeting feasibility demands related to cost, time, space, level of training, level of personnel, least possible risk, and patient acceptance. In the setting of cancer-related lymphedema, increasing rigor continues to be applied to a prospective surveillance and early intervention model of care. Additionally, in patients with cancer diagnoses, differential diagnostic distinctions may arise between benign versus malignant lymphedema, between lymphedema and neuromusculoskeletal or soft tissue conditions producing confounding locoregional symptoms, and between lymphedema and other etiologies of limb swelling or enlargement, especially venous disorders, lipedema, and systemic comorbidities.

Most of the existing diagnostic literature for lymphedema emphasizes breast cancer-related and upper quadrant lymphedema, especially regarding traditional or common measures such as tape measurement, water displacement and bioimpedance spectroscopy [ 2 ]. Lower extremity lymphovenous insufficiency is also extremely common, yet not as well studied [ 3 ]. Given the importance of lymphedema in the vascular space, three American vascular societies collectively published an expert opinion consensus on lymphedema diagnosis and treatment which suggests agreement from the experts with diagnostic work-up and risk factors for lymphedema, though highlights the high degree of variability regarding lymphedema treatment and practice patterns necessitating further investigation [ 4 ]. Furthermore, the International Society of Lymphology 2020 guidelines highlight the complexity and vast array of lymphedema clinical contexts, which “impact individualized patient care” [ 5 ]. Additionally, lymphedema has a striking 90% incidence in the survivors of head and neck cancer, however is still drastically under-diagnosed and treated from a recent publication utilizing a large US healthcare claims repository database [ 6 ]. Thus, there is a critical need for understanding and incorporating diagnostic and quantitative techniques into routine clinical practice which spans a significant and diverse population of individuals living with lymphedema.

Regarding detection protocols, overriding themes which have emerged in the literature include the importance of obtaining a baseline preoperative or pretreatment measurement whenever possible [ 4 , 7 , 8 , 9 ], and also to employ a consistent measurement modality in surveillance over time, rather than using different methodologies interchangeably [ 2 , 10 ]. None of the diagnostic methods have perfect accuracy and therefore there can be value in clustering observations from different available perspectives [ 2 ]. A 2020 systematic review of guidelines for detection of lymphedema noted overall limited evidence quality and could not make definitive conclusions, but found common themes including importance of clinical evaluation, use of duplex ultrasound to assess tissue thickness and fibrosis, computed tomography (CT) or magnetic resonance imaging (MRI) to rule out neoplasm, lymphoscintigraphy when needed to establish diagnosis, and measurement strategies including circumferences, perometry, and bioimpedance [ 3 ]. There is increasing advocacy for universal guidelines, especially for breast cancer-related lymphedema [ 11 ], incorporating symptoms, clinical examination and objective measurement, and also that all at-risk patients receive regular screening and education [ 12 ].

This paper explores the spectrum of traditional through emerging modalities, with the goal of evaluating practicable strategies by type of clinical scenario. The emphasis is on peripheral lymphedema, especially cancer-related lymphedema. Aims include (1) facilitation of best decision-making for modality choice in clinical and research applications, and (2) allowing overarching patterns to be identified, including strengths and gaps in current literature with regard to identified lymphedema assessment priorities.

A critical review was conducted by an experienced group of clinicians and researchers in the field of lymphedema and cancer rehabilitation, who identified modalities utilized for lymphedema diagnosis and analysis, and also identified core dimensions or purposes of lymphedema evaluation. The group was comprised of three physiatrists (AC, EI, MV), one physical therapist (PD), one occupational therapist (LK), one immunologist (MA), and one biochemist/molecular biophysicist (RC). The work of the group was conducted over a one-year period including independent efforts, six virtual meetings, and interspersed electronic communications. Preliminary content was presented at the Lymphedema Summit, sponsored by the Lymphology Association of North America (LANA) and the American Cancer Society (ACS), on October 7, 2023. Virtual meetings focused on the format and focus of this work, and, especially, on consensus with regard to the modalities themselves and on the integrative frameworks for core dimensions of lymphedema evaluation.

Via standard literature review, each of the modalities was briefly summarized with regard to their major capabilities and limitations, empirical and feasibility characteristics, as well as clinical versus research applicability. Primary authorship of each modality section was accorded with consideration of the diverse expertise of the authors, for content areas including history and physical examination (MV), tape measurement (MV), water displacement (MV), perometry (PD), bioimpedance (LK), tissue dielectric constant (PD), tonometry (PD), three-dimensional imaging (LK), patient reported outcomes (AC), lymphoscintigraphy (EI), ultrasound (EI), CT (EI), near-infrared fluorescence (MA), and MRI (RC). These summaries incorporated literature search as well as the expertise and experience of the authors. The literature search was broad and included primary studies as well as existing summative materials such as reviews and practice guidelines. An electronic mailbox was kept to which all authors had access, which included these documents, and other resources such as images and presentation materials. Each of the summaries was reviewed by all of the other authors and areas of controversy resolved via group communication.

Subsequently, via expert consensus process, five separate grids were generated with regards to the utility of each modality for assessing lymphedema characteristics, as well as for clinical dimensions of screening, diagnosis, individualizing treatment, and monitoring treatment response. Within each of these five categories, critical subcategories were identified through group consensus, and the utility of each modality for each subcategory was assigned a score of strong (S), indeterminate (I), unknown (U) and not useful or not applicable (N). A strong (S) rating was applied when the modality has an excellent favorable evidence basis as pertains to the specific subdomain, or, alternatively, a “Strong” rating could also be applied when at least 3 of 4 of the following four criteria were met:

Clinical practice-based acceptance, track record.

Some published evidence to support use.

Expert consensus.

Compelling theoretical basis.

If less than three of the above criteria were met but there remained a compelling rationale for use, an “intermediate” (I) rating would be assigned, such that at least 2 criteria were met or one criterion was strongly met. Feasibility was not used as a separate criterion, as this can vary with availability and accessibility of diagnostic modalities in individual treatment settings. However, feasibility considerations are generally implied within the clinical practice-based acceptance criterion. In situations of contextual variability, interpretation leaned towards the contemporary application of the modality (e.g., bioimpedance for early detection of breast cancer-related lymphedema). Furthermore, areas were highlighted that are especially relevant for research applications or a particularly active focus of current investigation.

The noninvasive office-based measures, including history and physical (H&P), patient reported outcome measures (PROMs), tape measure, perometry, water displacement, bioimpedance, tissue dielectric constant (TDC), tonometry, and three-dimensional (3D) camera, were placed on the left side of the charts and the more advanced imaging measures, including lymphoscintigraphy, ultrasound (US), indocyanine green lymphography (ICG), CT, and MRI, on the right side of the charts. It is recognized that ICG and ultrasound may also be performed in office, however they were considered advanced for the purpose of this review given their current less-frequent application by direct-treating lymphedema clinicians.

Lymphedema Assessment Modalities at Bedside or Office

History and physical examination.

Clinical H&P encompasses numerous domains including symptoms, time course, underlying risk factors, such as infectious history and other medical comorbidities, mobility, positioning, family and social history, cutaneous and soft tissue effects, motor-sensory and musculoskeletal abnormalities, and body weight. A varied taxonomy of descriptive terms may apply to skin and soft tissue characteristics, including basic characteristics as pitting, firmness, fibrosis, thickening, or erythema, to more elaborate descriptors including, but not limited to hyperkeratosis, lipodermatosclerosis, papillomatosis, nodular fibrosis, lymphedema rubra, phlebolymphedema, lymphangiomas (blisters containing lymph fluid), sausage digits, and elephantiasis verrucosa nostra [ 13 , 14 , 15 , 16 ]. Associated observations may include evidence of cellulitis, lymphorrhea, or wounds [ 5 ]. Palpation is employed to assess fibrosis, including skin and deeper tissue mobility, and extent of pitting, but reliable grading criteria for fibrosis are needed [ 17 ]. In the lower extremities, Stemmer’s sign (inability to pinch a skinfold at the dorsum of the second toe) is a classic observation [ 13 , 18 ], and individuals with history of axillary lymph node dissection may exhibit axillary cording especially early in the posttreatment course.

Various staging or grading paradigms exist, most notably the International society of lymphology (ISL) system ranging from subclinical lymphedema to lymphostatic elephantiasis, and which contains morphologic as well as size criteria [ 5 , 19 ]. The ISL stages include subclinical (0 or Ia), early accumulation with pitting (I), increased fat and fibrosis infiltration (II) and lymphostatic elephantiasis (III). Other paradigms have also been employed. The Campisi scale delineates 5 stages including, in order of severity, initial/irregular edema, persistent lymphedema, persistent lymphedema with lymphangitis, fibrolymphedema, and elephantiasis [ 13 , 20 ]. The National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) 5.0 (Grades 1–3), emphasizes soft tissue and skin characteristics, and in the severe category this scale incorporates limitation of self care as a criterion [ 21 ]. An earlier version, NCI-CTCAE 3.0 incorporates multiple lymphedema-related contexts including limb, head and neck, trunk/genital, lymphocele, cording and lymphedema-related fibrosis [ 22 ]. While not specific to lymphedema, the Pitting Edema Scale (1–4) has also been applied to lymphedema evaluation though reliability is likely limited [ 23 ].

Despite its ubiquity and importance to clinical care, studies are lacking with regard to accuracy and intra/interrater reliability of the physical examination for either clinical or research settings [ 4 ]. Lymphedema staging has value as a conceptual shorthand, and high feasibility to be incorporated into office visits. Limitations of clinical staging include imprecision regarding severity, that features of more than one stage may be present in the same individual, and that mixed etiologies of swelling may be present.

Bedside and office measures: general statement

An understanding of how to use and interpret the results of the bedside or office-based tools for surveillance and monitoring can greatly enhance the earliest diagnosis and treatment of lymphedema as well as give objective measures of change related to therapeutic intervention. A word of caution: no one device to-date can adequately measure all elements of therapeutic change given the heterogeneity of lymphedema. Furthermore, utility may break down in the absence of a reliable benchmark for comparison, such as a baseline measurement or unaffected contralateral limb. This requires a clinician to contextualize clinical interpretation.

Volume measurement

Volume measurement is an extension of the physical examination that can be undertaken with varying levels of technological sophistication. While parameters for detection exist, none are universally agreed upon, but volumetric thresholds of 3%, 5% or 10% change from baseline or in comparison to a normal contralateral side have been employed [ 5 , 24 ], as well as absolute value of 150-200 ml difference in volume [ 25 ]. ISL 2020 guidelines define severity as minimal (> 5 < 20% increase in limb volume), moderate (20–40% increase), or severe (> 40% increase), with some clinics preferring to use > 5–10% as minimal and > 10– < 20% as mild. The variability in limb volumes between different individuals limits utility of using absolute volume change as a severity parameter for breast cancer-related lymphedema [ 24 , 26 ]. Good reliability and validity have been found with numerous techniques including the traditional objective measures of water volumetry and tape measurement, as well as with more advanced approaches including perometry and (the nonvolumetric technique of) bioimpedance spectroscopy [ 27 ]. While most research has been conducted for breast cancer-related lymphedema, these principles apply to all types of lymphedema, including patients at risk for secondary lymphedema of the lower limbs who have had central operations with nodal sampling or resection.

Tape measurement

Tape measure-derived circumferential and volumetric measurements remain the most common clinical method [ 5 , 10 , 13 , 28 , 29 ] as well as the most extensively researched with regard to its reliability, validity and diagnostic accuracy [ 2 , 27 , 30 , 31 ]. Tape measurement has been employed in multiple trials, but there is potential for interrater variability, and likely low sensitivity for subclinical disease [ 24 ].

A difference or change of 2 cm or more at any one measurement site has been described as significant, (though of questionable reliability, especially in the setting of higher limb girths)[ 19 ] versus a combined difference of 5 cm or more summed over 5 sites along the limb [ 17 ]. Volumetric conversions may be performed by fulstrum or truncated cone calculations, with the former considered more accurate and less likely to overestimate volumes [ 31 ]. Formulaic adjustments also exist which account for dynamic factors such as body weight [ 32 ].

There is not universal agreement on measurement paradigms between fixed intervals or landmark-based. Taylor et al., examining the arms of breast cancer survivors with lymphedema, found volumes calculated from anatomic landmarks to be reliable, valid, and more accurate than those obtained from circumferential measurements based on distance from fingertips [ 30 ]. Sun et al. concurred with these findings, in a volumetric study of breast cancer patients assessed serially postoperatively, comparing landmark versus fixed-interval derived tape measurements to perometry as the gold standard. In the Sun study, the landmark-based measurements had better sensitivity and specificity than the fixed interval measures (93.1%/63.1% overall for landmark-based and 81.9%/16% for 4 cm intervals) though the landmark technique underestimated upper arm volumes and had only 63.2–66.7% sensitivity for the lower (5–10%) relative volume change subcategory [ 33 ].

Tape measurement and water displacement are highly correlated but can yield different values, with between 5% and 15–19% variation reported, so these techniques cannot be used interchangeably [ 30 , 31 ]. While reported diagnostic thresholds for treatment initiation vary, a clinical practice guideline on upper quadrant lymphedema yielded Grade B recommendations for tape measure use including volume ratio of 1.04 from the unaffected limb, volume differential > / = 200 ml between sides, and 5% or greater increase from baseline, but recommended against using a single site 2 cm difference as a diagnostic criterion [ 2 ].

Tape measurement has excellent feasibility regarding expense and accessibility. Concerns include the need for standardized protocols, training, and time to complete and compute measurements. Tape measurement is most applicable for limb rather than axial lymphedema, and like most volumetric measures it does not capture tissue composition.

Water displacement

Water displacement tanks for volumetric assessment have a limited role in most clinical settings due to feasibility barriers, though can be advantageous in research settings due to high degree of precision, with upper limb standard error of measurement of 3.6% compared to 6.6% for tape measurement [ 27 ]. However, error may be introduced if the limb is not submerged to a consistent level [ 30 ].

While water displacement is valid, accurate and reliable, disadvantages include being cumbersome, non-portable, requiring space, time and with risk of cross contamination [ 10 , 30 , 34 ]. Additionally, water displacement indicates the totality of volume but does not provide geometric detail on focal areas of enlargement. Measurement of lymphedema involvement near the root of the limb may be limited [ 5 ]. The Clinical Practice Guideline developed by the Oncology Section of the American Physical Therapy Association gave water displacement a Grade B recommendation, with criteria of > 200 ml or > 10% interlimb difference [ 2 ].

Perometry provides a quick measure of limb circumference measures and calculated summed volume using an infrared scanner frame that slides over the body region in seconds. The device measures arms, legs, torso, hands and feet depending on the perometer model. The advantages of using perometry are reliability of repeat measurements, rapid measurements with ease of repeating measurements and ability to evaluate limb circumference at 0.5 cm increments. Additionally, measures can be used for compression garment fitting, though some additional measurements may be necessary with custom garment fitting. In breast cancer-related lymphedema, perometry has good correlation with circumferential measures and good correction with bioimpedance spectroscopy [ 35 , 36 ]. Disadvantages include potential awkward patient positioning, expense of device, not easily transported requiring designated space, and limited centers with access to the equipment mostly found in research centers [ 35 , 37 ]. Additionally, there is difficulty with hand and foot measurements and positioning can be problematic for some patients to get accurate reproducible arm and leg volume results where consistency in measurement technique needs to be maintained for repeated measures over time [ 35 , 38 ]. Although considered a reliable measure of limb size and offers potential earlier detection of swelling compared to tape measure and palpation, as with other volume and circumference measurements, perometry is unable to discern extracellular fluid accumulation directly, rather volume changes could be related to muscle and fat changes as well within the area of interest. Therefore, similar to other devices of water displacement and tape measurements, perometry is not recommended as the sole method to measure and monitor lymphedema.

Bioimpedance

Bioimpedance technology is a method that measures biological impedance at different frequencies permitting clinicians to assess fluid compartments and body composition. This potentially provides clinicians a variety of information to help with making a clinical diagnosis of lymphedema and tailoring patient care treatment. Multiple frequency bioelectrical impedance analysis (MFBIA) and bioimpedance spectroscopy (BIS) provide a more comprehensive analysis compared with single frequency bioelectrical impedance analysis (SFBIA) by obtaining bioimpedance data at several different frequencies (MFBIA at different points from 1 to 1000 kHz and BIS continually from 0 to 1000 kHz) where BIS includes the crucial 0 kHz frequency [ 39 ] as part of its assessment. Impedance comprises resistance and reactance, indicating opposition from body fluids and cell membranes, respectively [ 40 ]. The inverse relationship between impedance and tissue fluid volume is a key principle [ 41 ]. The frequency of the current determines what is being measured, with zero frequency current unable to penetrate cell membranes [ 42 ]. Additionally, bioimpedance measures provide body composition data and phase angle data where a growing body of research is promising to inform oncology clinicians on changes to lean body mass and a prognostic factor in the advanced cancer setting, respectively [ 43 , 44 ].

Bioimpedance devices gage resistance to electrical current flow, especially in the extracellular fluid compartment at low frequencies. More specifically for bioimpedance spectroscopy (BIS) devices, an “impedance ratio” methodology is utilized for assessing unilateral arm or leglymphedema. This involves comparing the resistance at 0 kHz in the affected/at-risk limb to that in the unaffected limb, expressed as a ratio [ 45 ]. Alternatively, this ratio can be linearized into an L-Dex score, where abnormal values indicate deviations from the normal range (− 10 to + 10 L-Dex units) and a change exceeding 6.5 L-Dex units from baseline signifies subclinical lymphedema [ 46 ]. While this is currently a manufacturer-specific paradigm, similar scoring systems exist for other available bioimpedance devices. BIS is recognized for its non invasive effectiveness in measuring extracellular fluid and detecting subclinical changes indicative of lymphedema onset [ 42 , 47 ]. It offers high sensitivity, standardized cut-off measurements, and excellent inter-observer variability [ 48 , 49 ]. Moreover, BIS can assess intra and extracellular fluid as well as total body water [ 50 ], and is now eligible for insurance reimbursement in the United States.

Stand-on devices are available for BIS and MFBIA which incorporate stainless steel contact electrodes within hand and foot plates, with values graphically displayed over time. At this time, BIS has a larger body of research, as well as greater clinical penetration, however the extent to which BIS or MFBIA may be more advantageous than the other is unknown. Although BIS and MFBIA offer fluid and body composition measurements and are relatively quick to use, there is an expense to obtaining the technology which may also be on a subscription basis, and the information obtained in the severely obese population remains questionable [ 51 ].

Tissue dielectric constant—percent water content

Tissue dielectric constant (TDC) are focal measures of skin-to-fat water providing a quantified measure of tissue water within the skin and subcutis to aid a clinician’s assessment of local edema detection [ 52 ]. Advantages to TDC is its portable and focal assessment of superficial tissue water content for suspicious tissue allowing for comparison to contralateral focal body region. Focal measurements can be obtained on any skin region where newer devices provide user feedback on force and have the option of calculating the percent water ratio to “spot” scan for impairment. This portable tool provides rapid localized information on superficial water content on any skin region including torso, breast, top of hand and foot, and head and neck. TDC readings can be converted to percentage water content (PWC) for ease with comparing involved or at-risk with contralateral body regions [ 53 ]. Growing evidence reveals its use in early detection of breast lymphedema and the use of percent water content [ 52 , 53 , 54 , 55 , 56 , 57 ]. Disadvantages include the analysis of fluid is superficial to the skin and subdermis, depth assessment varies with the probe used typically ranging from 0.5 to 5.0 mm, focal measurement does not allow for efficient whole limb surveillance for edema, normative values appear to vary across body regions and are sparsely reported to-date, and evidence is lacking in how measures may change with advancing stages of lymphedema. Though this device also cannot be used without clinical examination, it offers promising use for focal swelling and comparison of contralateral sites on the trunk, breast, head, neck, feet and hands to provide clinicians an objective noninvasive measure of superficial fluid levels.

Tissue tonometry evaluates the resistance to pressure exerted on the skin providing an output of tissue induration in Newtons (N). Measurements assess local tissue areas and newer devices provide user feedback on speed and pressure for measurement accuracy. Some models require repeat measures and automatically compute the measurement average. The local skin measurement takes less than one minute to assess for an experienced clinician using the tool. Advantages to tonometry device are its ability to provide a measurement of tissue firmness and its portability and inter reliability [ 58 ]. Disadvantages are that measures may be influenced by ambient fluctuations, there are challenges with repeat measurement, device expense, limited literature on normative and impaired values, and uncertainty as to how the measures will vary across the stages of lymphedema [ 35 , 58 ], as fibrotic areas may soften with treatment while overly soft and edematous regions may harden with treatment. Research suggests potential for tonometry to inform on tissue fibrosis and treatment efficacy [ 59 , 60 , 61 , 62 ]. Further research is essential for a more in-depth understanding of this technology’s utility toward clinical assessment and treatment allocation in lymphedema.

Three-dimensional (3D) imaging

In the setting of lymphedema, 3D imaging technology advancements offer fast, noninvasive anthropometric body measurements of volume and circumference for baseline and longitudinal uses. The technology may involve scanners or a camera sensor attachment to a device with software subscription to obtain the images, in which currently limb and torso measurements are the most commonly measured. There is also the option for measurements of the hands, feet along with potential for the head and neck, necessitating further research to optimize the options across all body regions for clinical use. The 3D imaging outputs typically involve a silhouette of the body image captured providing total volume and circumferential measurements currently up to every 4 mm along the torso and limbs (arms and legs). The speed of obtaining circumferential and volumetric measures through 3D imaging offer a fast and portable opportunity for clinics to obtain a substantial amount of anthropometric data illustrating the change of volume and circumference over time [ 63 , 64 ]. Though this technology is efficient and reliable for certain body regions to use for the trained user, ongoing research is necessary to optimize model fitting and measurement extraction for entire body regions. Compared to the tape measure, the technology offers more promise with speed and interrater accuracy, yet this technology is more costly potentially involving a software subscription. Similar to perometry, 3D imaging currently cannot provide proficient data for “tight” circumferential measurements needed in particular regions when fitting for custom compression garments.

Patient reported outcome measures

Patient reported outcome measures (PROMs) assess latent constructs such as symptoms, quality of life (QoL), and self-efficacy that could not be otherwise quantified. PROMs typically measure defining, yet subjective, dimensions of a patient’s experience, and may be condition-specific or generic. In the case of lymphedema-specific PROMs, tools have been developed and validated to evaluate the impact of lymphedema on the physical, psychoemotional, functional, and social aspects of a patient’s life [ 65 , 66 , 67 , 68 , 69 , 70 ]. Most lymphedema-specific PROMs have been characterized with respect to both their psychometric (e.g., Cronbach’s alpha, intraclass correlation coefficients, etc.) and general measurement properties (e.g., reliability, responsiveness, validity, etc.) [ 71 , 72 ]. Access to performance information becomes critical when selecting contextually appropriate PROMs for clinical and investigative applications, since instruments may perform inconsistently across subgroups defined by lymphedema location, severity, and etiology, as well as in the presence of comorbid conditions like obesity and organ failure [ 73 ].

A subset of lymphedema-specific PROMs has been evaluated as screening tools to detect lymphedema. Separate instruments are used to screen for upper and lower extremity lymphedema. Upper extremity instruments include the Lymphedema Breast Cancer Questionnaire (area under the curve, or AUC, not reported) [ 74 , 75 ], and a telephone screening questionnaire developed by Norman et al. [ 76 ](sensitivity 0.96 and specificity 0.75). Lower extremity instruments include the Lower Extremity Lymphedema (LEL) Screening Questionnaire (AUC of 0.92) [ 73 ] and the Gynecological Cancer Lymphedema Questionnaire (GCLQ) (AUC of 0.95) [ 77 ].

Generic, or condition agnostic, PROMs have been shown to have acceptable to excellent measurement properties in the assessment of function and QoL among patients with lymphedema [ 78 , 79 ] . Use of generic PROMs allows for cross population comparisons and use of PROM data collected by other clinical disciplines. Additionally, generic PROMs have been developed and validated for administration via computerized adaptive testing (CAT) to increase precision and efficiency. At present, only two lymphedema-specific PROMs are available for CAT administration [ 80 , 81 ].

As lymphedema assessment tools PROMs offer salient benefits including low cost, ease of repeated longitudinal assessment, simple scoring and interpretation, availability in diverse languages, remote administration, and non-necessity of specialized training or personnel for high fidelity assessment. PROMs offer the only standardized means of quantifying the impact of lymphedema most pertinent to the patient, its treatment, and sequelae on a patient’s lived experience. However, PROMs are also subject to important limitations including respondent burden, inconsistent correlation with objective measures of lymphedema severity [ 82 ], and biases such as social acceptability, central tendency, end aversion, among others. Additionally, they may perform inconsistently across patient subgroups defined by sociodemographic and clinical characteristics.

Advanced imaging measures

Lymphoscintigraphy.

Lymphoscintigraphy has been considered the primary investigation to confirm the diagnosis of lymphedema, visualizing the functional status of the lymphatic system and guiding the management of lymphedema patients [ 83 ]. Lymphoscintigraphy is accurate and does not risk allergic reactions to contrast dye [ 84 , 85 ]. Lymphoscintigraphy has a sensitivity of 97% and a specificity of 100% [ 86 ]. The test requires injecting technetium Tc99m labeled antimony sulfur or albumin into the affected extremity’s second interdigital web space. Sequential images of bilateral extremities are performed in 20 min, and 3 h [ 87 ]. Lymphatic dysfunction is diagnosed if lymphoscintigraphy exhibits delayed transit time of the radiolabeled colloid to the regional lymph nodes, dermal backflow, asymmetric node uptake, and/or formation of collateral lymphatic channels [ 88 , 89 ] (Fig. 1 ). Lymphoscintigraphic study of breast cancer-related lymphedema has found excellent intra-rater reliability (ICC = 0.946) and interrater reliability (ICC = 0.846) [ 90 ]. Limitations include expense, planar images with limited spatial resolution (unless combined with CT), fuzzy or grainy images, radioactivity exposure and disposal issues, and time required for testing.

A combined scintigraphic and CT technique, SPECT-CT, allows functional visualization of altered lymphatic flow in three dimensions as opposed to lymphoscintigraphy which is planar, and SPECT-CT simultaneously demonstrates lymphatic stigmatae per CT imaging as above. While found to have some advantages in accuracy of lymphoscintigraphic staging of lymphedema compared to lymphoscintigraphy alone [ 91 ], it is costly and not routinely used.

figure 1

Lymphoscintigraphy for assessment of lymphatic function. a Lower extremity lymphedema showing pooling of lymphatic fluid in left calf and decreased left pelvic lymph node activity. b Postmastectomy lymphedema with pooling in distal left upper limb and markedly reduced axillary lymph node activity

Because the physical examination is primarily subjective and does not represent subcutaneous tissue distribution and lymphedema tissue alteration, use of ultrasound has been advocated to enhance lymphedema staging [ 92 ]. (Fig. 2 ) A noninvasive tool, ultrasound offers visualization of the skin and subcutaneous tissue changes in extremities with chronic lymphedema, caused by changes in the extracellular matrix, such as connective tissue hypertrophy, fat accumulation resulting from both fat hypertrophy and an increased number of adipocytes, and interstitial protein-rich fluid accumulation [ 93 ]. The observation of these changes in various parts of the extremity may further elucidate the severity and extent of the disease.

Skin thickness, subcutaneous tissue thickness, and subcutaneous echogenicity all show a significant positive correlation with the ISL stage [ 94 ]. Ultrasound studies report interrater reliability, accuracy, and variability with some bias in small sample size groups.

In a study of ultrahigh frequency ultrasound of upper and lower limbs affected by secondary lymphedema, Bianchi et al. found a high correlation between the ultrasound and histological findings when used prospectively for preoperative and intraoperative analysis for lymphovenous anastomosis [ 95 , 96 , 97 ].

Unlike most forms of imaging, ultrasound can be administered at point of care, though user expertise is required, along with investment in the device.

figure 2

Ultrasound to distinguish normal, lymphedema and lipedema. Lymphedema is associated with increased dermal thickness and with subcutaneous tissue hyperechogenicity, whereas lipedema is associated with increased thickness and hypoechogenicity of the subcutaneous fat (dermis marked in upper arrows, subcutaneous tissues in lower arrows)

Computed tomography

CT scans are essential in the evaluation of the causes of limb swelling. CT imaging can provide objective volume measurements and information about the structural characteristics of subcutaneous tissue in lymphedema [ 98 ]. CT scans demonstrate the alterations in epidermal and subcutaneous tissue, but can also detect neoplasms obstructing the lymphatics causing secondary lymphedema [ 99 ]. MRI and CT are useful tools for measuring both the volume of subcutaneous tissue and structural changes. CT imaging has been shown to have high sensitivity (93%) and specificity (100%) in confirming the diagnosis of lymphedema. CT imaging is not routinely applied to evaluate lymphedema because of the associated high cost and radiation exposure.

The subcutaneous tissue of lymphedema exhibits a pathognomonic honeycomb distribution of edema due to fibrotic tissue and fluid surrounding the accumulation of fatty tissue; the skin is thickened [ 99 , 100 , 101 ]. CT also can display the size and the number of lymph nodes, which helps define the type of primary lymphedema.

Lymphatic imaging with near-infrared (NIR) fluorescence

Two very similar methods for imaging shallow lymphatics use near-infrared fluorescence: Indocyanine Green Lymphography (ICG-L) and Near-InfraRed Fluorescence Lymphatic Imaging (NIRF-LI). Both ICG-L and NIRF-LI use intradermally injected ICG as a contrast dye to allow visualization of dermal lymphatics and lymph nodes to depths of 1 cm (ICG-L) or 3–4 cm (NIRF-LI) [ 102 , 103 ]. Slightly different optics technology also allows NIRF-LI to image lymphatic pumping in near-real time for extended periods of time [ 104 ]. Both technologies take advantage of the fact that light in the near-infrared (NIR) portion of the light spectrum (wavelength 600–800 nm) is not readily absorbed by melanin, water, hemoglobin, or other components of human tissue. For example, if you shine a flashlight at the base of a finger, most light wavelengths will be absorbed, but NIR wavelengths will penetrate the skin between your fingers and appear red. When injected intradermally, ICG binds to intracellular albumin and other large proteins that are selectively taken up by lymphatic, but not arterial or venular, capillaries. Dim NIR laser light shining on skin excites ICG, so that shallow lymphatic vessel anatomy (and pumping, with NIRF-LI) are visible.

NIR tools can provide information as to which collector vessels are used, as well as identify anatomical areas where lymph is stagnant, appearing as “dermal backflow (Fig.  3 ) Dermal backflow never appears in healthy limbs, but is almost always present with subclinical and clinical lymphedema [ 105 ]. In healthy lymphatics, ICG bound to large proteins is readily transported from interstitial spaces through primary capillaries directly into lymphatic collectors. Dermal backflow in patients with lymphedema, readily detected by ICG-L, appears as cloudiness, tiny tortuous vessels clusters, or punctate bright spots within cloudy areas. Such images represent lymph that is stagnating in interstitial spaces, primary capillaries, or small pre-collector vessels.

figure 3

Breast Cancer-Related Lymphedema seen by Near-Infrared Fluorescence Lymphatic Imaging (NIRF-LI). Example images of (left) healthy lymphatics with well defined, linear lymphatic structure and contractile function, and (right) diseased lymphatics with fluorescent network of tortuous lymphatic vessels and dermal backflow

These imaging modalities can be used as tools for (1) early diagnosis of lymphedema, (2) objective assessment of lymphedema conservative and surgical treatments, and (3) discernment of lymphedema in a variety of clinical contexts, not limited to post-surgical lymphedema. ICG-L and NIRF-LI can detect failing lymphatic function well before clinically recognized lymphedema can be diagnosed. Of note, lymphatic dysfunction can be missed using clinical swelling as a criterion, thus delaying treatment. For example, Fig.  4 shows NIRF-LI images of breast cancer patients clearly displaying dermal backflow, yet arm swelling is below 5% (and even negative) [ 106 ]. Such patients are assumed to not have lymphedema, and are not treated when the lymphatics may be most amenable to favorable outcomes, before irreversible tissue changes, such as skin fibrosis and subdermal adipose accumulation, are established [ 107 ]. Importantly, a recent prospective longitudinal study of breast cancer patients showed that NIRF-LI detected dermal backflow 8–23 months before clinical lymphedema was diagnosed by arm swelling [ 108 ]. NIRF-LI has shown that manual lymphatic drainage and pneumatic compression therapy directly improve lymph movement, and a study of reparative lymphatic microsurgeries is underway [ 109 , 110 , 111 , 112 ]. Such studies demonstrating physiologic benefits of these treatment methods have provided evidence to medical insurers in support of their reimbursement. NIRF-LI has detected dermal backflow in breast cancer patients who have yet to receive mastectomy or lumpectomy with axillary lymph node dissection, suggesting effects of the cancer itself, [ 108 ] of neoadjuvant chemotherapy [ 113 ] and/or effects of inflammation (which has been shown to systemically halt lymph pumping) [ 114 ], as risk factors for lymphedema. Breast cancer patients who receive immediate lymphatic repair at the time of axillary lymph node dissection (the lymphatic microsurgical preventative healing approach, or LYMPHA technique) are reported to have lower incidence of lymphedema [ 115 ]. Longitudinal NIR imaging could track efficacy of LYMPHA, to determine if lymphatic pumping is always restored with lymphatic-vein anastomosis.

figure 4

Breast Cancer-Related Lymphedema seen by Near-Infrared Fluorescence Lymphatic Imaging (NIRF-LI) in the setting of normal arm volumes. Dermal backflow (top images) is evident in three different breast cancer patients’ affected arms. White-light (bottom) images were also collected with a white-light camera mounted on the imager arm. Relative volume changes (RVC) for all three patients were negative, well below 5-10% used to clinically diagnose breast cancer-related lymphedema

A major limitation of ICG-L and NIRF-LI is the inability to show deep lymphatic vessels, such as the cisterna chyli/thoracic duct, although NIRF-LI can detect pulsing in collector vessels that are 3–4 cm deep, if there is no dermal backflow overlaid [ 103 ]. Nonetheless, in several cases, NIRF-LI has detected dermal backflow in patients with normal lymphoscintigraphy results, allowing early treatment for extremity lymphedema [ 102 ].

Magnetic resonance imaging (MRI) is a biomedical imaging modality with versatile image contrast that can be used to visualize anatomy at high spatial resolution, quantify tissue molecular composition, and probe vascular morphology and function in the lymphatic network comprised of vessels, nodes, and organs. MRI methods are clinically feasible to perform anatomical imaging at 3D high spatial resolution (typically 0.5–1 mm 2 in-plane) by standard T 1 - and T 2 -weighted MRI. Anatomical imaging in lymphedema can be used to measure the limb’s volume and soft tissue remodeling such as skin or subcutaneous adipose tissue thickening, for instance (Fig.  5 ). MRI contrast can also be sensitized to tissue molecular content in lymphedema. Dixon MRI separately visualizes water- and fat-dominant tissue composition in a single acquisition, and can quantify tissue water and fat-fraction asymmetries to inform disease severity and location [ 116 , 117 ]. MR lymphangiography with or without exogenous contrast agents is also clinically feasible with standard pulse sequences available on most hospital scanners, although further standardization of protocols and tracers is needed [ 118 ]. Non-tracer MR lymphangiography hyper-intense signal patterns in subcutaneous edema and tissue sodium content by sodium MRI are being investigated to inform individualized physical therapy strategies for edema mobilization [ 119 , 120 ]. While current cost and accessibility limit the widespread use of MRI in clinical lymphedema management, its high spatial resolution, noninvasive molecular specificity, and adaptability to functional lymphatic imaging offer particular advantages and clinical use scenarios [ 121 ].

figure 5

Noninvasive MRI methods for structural and physiologic lymphedema assessment. Noninvasive MRI methods provide structural and physiologic imaging measures for upper-extremity lymphedema (top row) and lower-extremity lymphedema (bottom row). A Optimized image contrast at 3T MRI provided lymph node anatomical imaging sensitive to the afferent and efferent vessel and lymph node substructures. Structural MRI of soft tissue anatomy demonstrates tissue remodeling in disease such as skin and adipose tissue thickening, and fibrosis deposition. B MR lymphangiography visualizes deep lymphatic vessel morphology, such as in the thoracic duct anterior to the spinal cord. Edema is also visualized as hyper-intense signal on non-tracer MR lymphangiography to localize dependent edema and response to manual therapies. C Tissue composition in lymphedema can be quantified with 3T MR relaxometry (e.g. T 2 relaxation time map in upper-extremity unilateral lymphedema), and Dixon fat-fraction mapping. These measures indicate edematous and heterogeneous tissue in lymphedema, which responds to manual lymphatic drainage therapy. D Molecular imaging methods relevant for lymphedema include CEST-MRI of proteins (e.g. magnetization transfer ratio, MTR of amide proteins) and sodium MRI of endogenous tissue sodium content (mmol/L). Molecular imaging is being investigated for early biomarkers of edema formation and objective, localized measures of lymphatic disease severity. Together, MRI is a versatile modality to investigate lymphatic physiology and disease with high potential for clinical translation.

MRI offers exciting possibilities as a research tool to advance lymphedema care. Examples of advancements in lymphatic MRI include MR relaxometry of lymphatic fluid, lymph nodes, and soft tissue in lymphedema that is fundamental to optimizing anatomical image contrast across the lymphatic system [ 122 , 123 , 124 ]. MR lymphangiography has advanced in specificity for lymphatics by combining T 1 and T 2 shortening agents with techniques like DARC-MRL and contrast-enhanced MRL [ 125 ]. Dynamic lymphangiography analysis tools will be needed to translate these techniques into a clinical setting. Physiologic imaging is exploring MRI quantification of tissue protein, sodium, and fat composition in lymphedematous tissue to investigate the impact of lymphatic dysfunction on limb physiology and potentially image-based personalized risk factors for developing lymphedema or lipedema [ 126 , 127 , 128 ]. As novel therapeutics are developed for lymphedema, MRI modalities could likely play a significant role as objective, sensitive outcome measures of therapeutic mechanism and response to therapy. MRI research opportunities remain at all translational research levels to improve the specificity and objectivity of lymphatic disease assessment.

Dysmorphism: Shape and contour deviations. Metaplasia: Altered tissue integrity that is not on malignant trajectory.

Lymphedema evaluation: tables and summaries.

No individual method is effective for the entire range of lymphedema subdomains Domains Assessed by Specific Modalities (Table 1 ), with the fewest options available for dysmorphism and metaplasia. H&P, PROM’s, MRI and CT show the strongest multidimensional capabilities, with ultrasound and ICG also promising. The various methods available to assess volume are largely unidimensional, however some measures (perometry, 3D camera, and to some extent tape measure) do give detail about contour. Technologies such as bioimpedance and TDC, which assess fluid content, are becoming increasingly available, but these are limited in their ability to delineate other tissue composition factors such as fibrosis or fat, which require use of advanced measures including ultrasound, CT and MRI. Measures which are both clinically feasible and have good discrimination for soft tissue factors beyond extracellular fluid assessment are a priority for future work. Clinically feasible measures of lymphatic function are also a gap area.

(Table 2 ) This category relates mainly to clinical situations of individuals at risk for lymphedema, most commonly following cancer treatment. Screening constitutes an especially active focus of current lymphedema research. Furthermore, from a clinical viewpoint, mechanisms for early detection of cancer-related lymphedema are increasingly prioritized for integration into routine care [ 129 ]. The need to reconcile the dual priorities of optimizing both quality and feasibility, which are pertinent to all dimensions of lymphedema measurement, reaches primacy with screening, due to population health-level need for repeated measurements over time in a large number of patients.

Studies suggest that the sensitivity of bioimpedance is in the 70–76% range for cancer-related lymphedema when advanced measures including lymphoscintigraphy [ 48 ], or ICG [ 130 , 131 ] are used as reference standards. Bioimpedance has also exhibited favorable specificity in some studies. [ 132 , 134 ]. The international multicenter PREVENT randomized controlled trial data has found fewer patients progressing to clinically evident lymphedema when bioimpedance has been integrated into screening processes for breast cancer-related lymphedema [ 132 ]. Studies comparing bioimpedance to perometry have shown anywhere from modest [ 133 ] to good [ 134 , 135 , 136 ] correlation between the two modalities. Dylke et al. [ 48 ] found slightly better sensitivity and specificity with perometry (81%/96%) compared to bioimpedance spectroscopy (76%/93%), using lymphoscintigraphy as the reference standard, in the context of mild to moderate upper limb lymphedema. On the other hand, Czerniec et al. [ 136 ], while finding high concordance between bioimpedance spectroscopy and perometry, found significantly higher interlimb ratios with bioimpedance spectroscopy than perometry in individuals with clinically identified mild to severe lymphedema, and this pattern held true with both whole-arm and segmental analyses.

Overall, fluid-detection measures are becoming increasingly utilized for screening, based on favorable accuracy characteristics found in the breast cancer population, as well as feasibility, especially for Individualizing Treatment (Table 3 ). bioimpedance. However, this issue is not an entirely decided matter. Volumetric measurement techniques continue to be employed in screening research trials, and they also remain relevant based on common use. On the favorable side, volumetric measures exhibit value as complementary information, but on the challenging side, they can be difficult to upscale for frequent repeated measurements. Tape measurement prevails in most lymphedema practices and is particularly advantageous in stressed resource settings. Tape measurement can also be adapted for self-monitoring. Perometry has particularly good sensitivity to small volume change. 3D imaging via phone or tablet shows strong future potential given its promising convenience. PROM’s also show some favorable characteristics and, while not a stand-alone assessment, offer complementary information. In summary the use of a comprehensive multi-modal assessment is recommended with clinical evaluation and symptom reporting also important.

Despite favorable aspects of bioimpedance, current technology has limits, especially with regards to detecting axial (i.e., head and neck, trunk, breast, chest wall, genitalia) lymphedema, and for evaluation of other focality (such as dorsum feet or hands). Measures that are useful for axial lymphedema, such as ultrasound and TDC, are not yet in wide use for this purpose, and have their own feasibility considerations. While bioimpedance utilization has expanded, implementation challenges remain due to cost, as, despite its FDA approval for BIS in the United States, gaps remain in insurance coverage.

Diagnosis (Table 4 ) pertains to a range of clinical situations not limited to cancer-related lymphedema and may entail distinguishing lymphedema from other causes of swelling or soft tissue changes, and/or establishing the underlying disease causing lymphedema. Regarding lymphedema diagnosis, a noteworthy observation is that despite multiple available methodologies, no individual modality outperforms H&P, especially among the measurement tools available in office and at the bedside. H&P provides advantages of myriad components as well as overall pattern recognition. However, H&P can have limits diagnostically, especially in medically complex patients or those with mixed etiologies. Depending on context, distinguishing exact contributors may or may not make a difference in management, and H&P, including the crucial history portion, helps to identify when more testing is needed. ICG, MRI and lymphoscintigraphy do have areas of strength and can be considered when H&P alone provides insufficient objective and diagnostic confidence in distinguishing lymphedema from other etiologies, identifying earliest potential onset of lymphedema, and determining affected versus unaffected areas. Longitudinal assessment and/or utilizing more than one type of modality can also be helpful in verifying diagnosis in difficult or elusive cases.

The individualizing treatment (Table 4 ) category shows some areas of strength, especially with regard to distinguishing degree of involvement in affected territories, and informing treatment, with H&P, ultrasound, ICG, MRI and lymphoscintigraphy showing strongest characteristics, and some advantages also seen for tape measure, perometry, bioimpedance, and TDC. Limitations remain, especially in guiding follow up logistical needs, as well as in stratifying risk for adverse outcomes, especially as risks may be affected by changes in treatments. Therefore, while useful tools exist, optimizing their integration into clinical care paradigms is a priority.

The majority of modalities are highly limited, except for Monitoring Treatment Response (Table 5 ). the domain of detecting volumetric changes, which shows several effective options. Of these, perometry and water displacement have the highest precision to capture change over time, though tape measure is commonly used based on feasibility. Bioimpedance has defined criteria for intervention, and favorable utility for surveillance, though the emphasis of the bioimpedance literature to-date has been towards lymphedema identification and screening rather than on its reliability in chronic situations and clinically relevant improvements during rehabilitation. Other subdomains show a lack of abundant options, especially for tissue composition and metaplasia.

Several concurrent approaches are layered within this work. First, a comprehensive framework has been generated of assessment dimensions and subcategories of lymphedema diagnosis and quantification, which assists with a practical understanding of the wide-ranging considerations pertinent to this diverse topic. Second the S/I/U/N ratings are assigned, employing defined criteria with emphasis on the evidence basis. Third, the grid format allows identification of overarching patterns, such as (1) the strength and weakness of particular modalities versus others within-dimensions of lymphedema evaluation (diagnosis, screening, individualizing treatment, monitoring treatment response), (2) the lymphedema characteristics which can be most confidently assessed (i.e., volume, tissue composition, symptoms, etc.), and (3) the lymphedema evaluation dimensions which overall have the strongest or weakest evidence foundation.

The overriding purpose of the above process is to produce a point-in-time snapshot of lymphedema diagnostic methods to update clinicians in their selection process of situationally appropriate modalities, to apply to their patient care as well as program development. Accomplishing this goal requires the dual priorities of highlighting traditional methods of continuing relevance, as well as areas of exciting progress. The substantial remaining gaps are also accentuated.

While specific clinical context ultimately guides decision-making, patterns seen among the various forms of lymphedema evaluation are noteworthy. Of the lymphedema domains assessed by specific modalities (as seen in Table  1 ), the overall highest performing modalities across the spectrum of domains, with the greatest number of strong ratings, are MRI, H&P, CT, and PROMs; the lymphedema domains most likely to have strongly rated assessment methods are volume and fluid content. Of the various dimensions of lymphedema assessment (Tables 2 – 5 ), individualization of treatment attains the highest frequency of strong ratings (45%), followed by screening (24%), diagnosis (20%), and monitoring treatment response (15%). Across the 20 subcategories within these four tables of lymphedema dimensions, H&P demonstrates the highest number of strong ratings (11), followed by ICG (10), MRI (8), lymphoscintigraphy (7), bioimpedance (6), PROMs (5), ultrasound (5), tape measure (5), perometry (3), TDC (3), CT (3), 3D camera (2), water displacement (1), and tonometry (0). When modalities receiving either strong or intermediate ratings are considered, across these 20 subcategories, the values become, in order of frequency, H&P (15), MRI (14), ultrasound (12), ICG (11), tape measure (10), perometry (9), bioimpedance (9), TDC (9), lymphoscintigraphy (8),CT (8), PROMs (7), 3D camera (7), water displacement (5), and tonometry (3).

Dysmorphism: Shape and contour deviations.

Metaplasia: Altered tissue integrity that is not on malignant trajectory.

Of note, in considering diagnostic modalities, we elected not to include simple lymphography, which has become less favored due risks of lymphatic vessel cannulation and to potential for lymphatic damage with radioopaque contrast; however, lymphography may be especially useful, for deep/central lymphatic system imaging (thoracic duct, cisterna chyli), and is considered when other advanced imaging approaches are not available [ 137 , 138 ]. In the United States, ultrasound, ICG-L, MRI/MRL, CT or lymphoscintigraphy are more commonly used. But even the use of lymphoscintigraphy, considered the gold standard to diagnose lymphedema, is only used for ~ 6.7–9.5% of cancer-related lymphedema patients [ 139 ].

Clinical vs research considerations

No single type of measure is applicable to all aspects of clinical and research integration. For routine clinical use, modalities which are feasibly integrated into office or bedside settings will be favored, with radiologic imaging measures applied more selectively. Water displacement, while cumbersome for routine clinical use, has been advocated for research given its high precision, as has perometry. PROMs show favorable characteristics for use in clinical settings, including that they capture factors such as symptoms and function which are missed by other tools and provide clinicians with patient-specific impairments to tailor personalized care. But patient burden must be considered with PROMs, as well as clinic workflow and ability to clinically incorporate the information, therefore PROM tools need to be chosen very intentionally to assess target domains as efficiently as possible. In the research setting, use of more expanded PROM item banks may be appropriate. Bedside or clinic options which constitute a particularly strong focus of current research include bioimpedance, TDC, and 3D camera. Tools such as US and ICG have not yet enjoyed widespread clinical application in diagnosis, but ultrasound is one of the stronger noninvasive, portable modalities for evaluating soft tissues, and has potential for expanded role at the bedside. Additionally, ICG is beginning to show application beyond surgical planning, such as prognostication and possible role in guiding tailored lymphedema therapy [ 137 ] MRI is a multifaceted and evolving topic in current lymphedema research that will likely have a range of future applications albeit an inherent limitation for patients not compatible for MRI use. More specific qualities can also drive selection, for example the utility of MRI and CT in characterizing lymphedema involvement at the level of deep structures.

In clinical care, cost effectiveness remains a crucial component of feasibility. However, from another vantage point, lymphedema as a diagnostic entity has been widely considered as underrecognized and undertreated. Furthermore, the role of the full range of diagnostic options should be weighed for appropriate best practice. Challenging circumstances may require the incremental precision obtainable with overlapping or hierarchical approaches. The cost benefit of additional testing will likely vary based on context. Lipedema is an example of a clinical situation where additional testing may promote cost effectiveness, sparing costly lymphedema treatment.

An imaging toolbox that utilizes several different modalities could allow optimal understanding of lymphatic dysfunction but may be costly and present accessibility difficulties. For example, to present figures at the time of this writing, ICG-L imagers are commercially available, but they can cost hundreds of thousands of US dollars, and ICG costs $110 per vial in the United States. The billing code used by reparative microsurgeons for ICG-L prior to lymphovenous bypass and/or vascularized lymph node transplant reimburses at ~ $700. ICG-L is used in Europe for assessing lymphedema treatment efficacy, but there may currently be patent issues for such use in the US. NIRF-LI is only available for research use at present. Comparatively, lymphoscintigraphy averages $1500, and MRL may cost $900. Cost effectiveness studies are needed, with an imaging protocol streamlined to maximize the cost/benefit ratio.

Current and emerging themes

Simultaneous with judicious use of emerging technologies, traditional evaluation methods including clinical history and physical examination and other office-based measures remain highly relevant mainstays of comprehensive assessment for theexperienced clinician, who can order and interpret correctly the various modalities described.

Specific practice settings, especially whether high, medium or low in resources will influence feasibility in translation of available techniques.

Need exists for improved bedside measures of soft tissue characteristics, including tissue composition (moving beyond fluid content), dysmorphism and metaplasia.

Bioimpedance and PROMs may be particularly valuable for early detection and surveillance of lymphedema.

TDC is showing strong potential for detecting and assessing focal lymphedema.

3D camera is another emerging tool, for which more research is needed, with potential advantages including speed (compared to tape measurement) and feasibility factors (compared to perometry). 3D camera is also showing favorable spatial resolution (4 mm).

Use of ultrasound holds promise for more widespread use in evaluating tissue characteristics, especially for focal lymphedema, and has potential to serve as a bridge between bedside availability and traditional diagnostic imaging.

ICG and MRI to-date have been employed mainly in specialized situations such as presurgical evaluations and research, however potential clinical applicability has far-reaching implications, including guaging the effectiveness of therapy approaches on a physiological level.

Additional research studies focusing on correlation between advanced modalities (such as MRI, ICG or lymphoscintigraphy), and bedside tools such as bioimpedance, TDC, and perometry are encouraged to better inform clinical practice.

Imaging techniques such as MRI have a role in development of new therapies such as mechanistic or functional based treatment, even if not easily translatable into routine clinical care.

Application of artificial intelligence (AI) has potential to assist with interpretability of lymphedema diagnostic testing. However, a need exists for more standardized acquisitions with techniques to allow AI systems to be more reliable.

Limitations

In formulating the above charts and assigning specific ratings, the authors acknowledge inherent interpretative limitations due to (1) heterogeneous clinical contexts, including patient mix, physical, financial and workforce resources of various care environments, and models of care, and (2) varying technologies within some of the modality categories. Because of this contextual variance, our expert panel employed “current usual circumstances” as a best-fit guiding principle in gauging a particular modality, and also assumed that end-user clinicians or researchers were appropriately trained and experienced in use of the particular modality. However, even with these assumptions, the authors emphasize that the focus for the reader is most appropriately placed on patterns of observations, rather than considering any one particular rating as definitive for all situations.

Additionally, there have been some limitations in focus, most notably that this work has emphasized peripheral lymphedema, rather than the central lymphatic system. The lymphatic system is highly heterogeneous with peripheral and central aspects. Internally, the lymphatic networks will vary according to whether the topic is hepatic, intestinal, cardiac, and so forth. [ 137 ] Central lymphedema evaluation may center around evaluating for structural disruptions such as fistula or leakage [ 140 ], whereas peripheral etiologies are more likely to relate to lymphatic congestion [ 137 ], with the latter being implicit in this paper. Head and neck lymphedema may include external or internal components, or both. [ 141 ] In the brain, a combination of glyphatic system (parenchymal) and lymphatic (meningeal) function has been found [ 142 ].

Various other limitations are present. Much of the literature examining office-based modalities is problematic with regard to reference standards, obscuring the ability to interpret sensitivity, specificity and other aspects of empirical data. The literature overall is skewed towards breast cancer-related lymphedema; while this emphasis is understandable given its prevalence and the potential to intervene early, other clinical contexts, including lower limb etiologies, merit more attention. Some important related aspects are beyond the scope of this paper, including surgical planning, surveillance time-course models, and clinical aspects of risk factor evaluation. Laboratory medicine aspects of lymphedema such as genetic, biomarker, or inflammatory aspects of lymphedema are not explored. While cost factors are given consideration in our discussions of the modalities, detailed cost effectiveness analyses are beyond the current scope of available evidence Lastly, while we attempted to highlight areas particularly relevant to research, we acknowledge and even emphasize that virtually all of the domains and modalities assessed in this paper are relevant to lymphedema research.

Conclusions

Lymphedema diagnostic and quantitative methods encompass an increasing array of modalities, which are pertinent to various lymphedema characteristics, and applicable to clinical and research aims, including dimensions of diagnosis, screening, individualization of patient care, and monitoring treatment response. This paper provides guidance to clinicians in (1) affirming the role of traditional measures when appropriate, and (2) identifying instances when integrating emerging technologies and imaging modalities can enhance patient care.

Data availability

No datasets were generated or analysed during the current study.

O’Donnell TF Jr, Rasmussen JC, Sevick-Muraca EM. New diagnostic modalities in the evaluation of lymphedema. J Vasc Surg Venous Lymphat Disord. 2017;5(2):261–73. https://doi.org/10.1016/j.jvsv.2016.10.083 .

Article   PubMed   PubMed Central   Google Scholar  

Levenhagen K, Davies C, Perdomo M, Ryans K, Gilchrist L. Diagnosis of upper quadrant lymphedema secondary to cancer: clinical practice guideline from the oncology section of the American physical therapy association. Phys Ther. 2017;97(7):729–45. https://doi.org/10.1093/ptj/pzx050 .

O’Donnell TF Jr, Allison GM, Iafrati MD. A systematic review of guidelines for lymphedema and the need for contemporary intersocietal guidelines for the management of lymphedema. J Vasc Surg Venous Lymphat Disord. 2020;8(4):676–84. https://doi.org/10.1016/j.jvsv.2020.03.006 .

Article   PubMed   Google Scholar  

Lurie F, Malgor RD, Carman T, Dean SM, Iafrati MD, Khilnani NM, Labropoulos N, Maldonado TS, Mortimer P, O’Donnell TF Jr, Raffetto JD, Rockson SG, Gasparis AP. The American venous forum, American vein and lymphatic society and the society for vascular medicine expert opinion consensus on lymphedema diagnosis and treatment. Phlebology. 2022;37(4):252–66. https://doi.org/10.1177/02683555211053532 .

Executive Committee of the International Society of Lymphology. The diagnosis and treatment of peripheral lymphedema: 2020 consensus document of the international society of lymphology. Lymphology. 2020;53(1):3–19.

Google Scholar  

Stubblefield MD, Weycker D. Under recognition and treatment of lymphedema in head and neck cancer survivors—a database study. Support Care Cancer. 2023;31:229. https://doi.org/10.1007/s00520-023-07698-3 .

Russo S, Walker JL, Carlson JW, Carter J, Ward LC, Covens A, Tanner EJ 3rd, Armer JM, Ridner S, Hayes S, Taghian AG, Brunelle C, Lopez-Acevedo M, Davidson BA, Schaverien MV, Ghamande SA, Bernas M, Cheville AL, Yost KJ, Schmitz K, Coyle B, Zucker J, Enserro D, Pugh S, Paskett ED, Ford L, McCaskill-Stevens W. Standardization of lower extremity quantitative lymphedema measurements and associated patient-reported outcomes in gynecologic cancers. Gynecol Oncol. 2021;160(2):625–32. https://doi.org/10.1016/j.ygyno.2020.10.026 .

Havens LM, Brunelle CL, Gillespie TC, Bernstein M, Bucci LK, Kassamani YW, Taghian AG. Use of technology to facilitate a prospective surveillance program for breast cancer-related lymphedema at the Massachusetts general hospital. Mhealth. 2021;7:11. https://doi.org/10.21037/mhealth-19-218.PMID:33634194;PMCID:PMC7882272 .

Sun F, Skolny MN, Swaroop MN, Rawal B, Catalano PJ, Brunelle CL, Miller CL, Taghian AG. The need for preoperative baseline arm measurement to accurately quantify breast cancer-related lymphedema. Breast Cancer Res Treat. 2016;157(2):229–40. https://doi.org/10.1007/s10549-016-3821- .

Article   CAS   PubMed   Google Scholar  

Armer JM, Hulett JM, Bernas M, Ostby P, Stewart BR, Cormier JN. Best practice guidelines in assessment, risk reduction, management, and surveillance for post-breast cancer lymphedema. Curr Breast Cancer Rep. 2013;5(2):134–44. https://doi.org/10.1007/s12609-013-0105-0 .

Kassamani YW, Brunelle CL, Gillespie TC, Bernstein MC, Bucci LK, Nassif T, Taghian AG. Diagnostic criteria for breast cancer-related lymphedema of the upper extremity: the need for universal agreement. Ann Surg Oncol. 2022;29(2):989–1002. https://doi.org/10.1245/s10434-021-10645-3 .

Brunelle CL, Taghian AG. ASO author reflections: breast cancer-related lymphedema-a suggested clinical pathway for diagnosis. Ann Surg Oncol. 2022;29(2):1003–4. https://doi.org/10.1245/s10434-021-10702-x .

Pappalardo M, Starnoni M, Franceschini G, Baccarani A, De Santis G. Breast cancer-related lymphedema: recent updates on diagnosis, severity and available treatments. J Pers Med. 2021;11(5):402. https://doi.org/10.3390/jpm11050402 .

Farrow W. Phlebolymphedema-a common underdiagnosed and undertreated problem in the wound care clinic. J Am Col Certif Wound Spec. 2010;2(1):14–23. https://doi.org/10.1016/j.jcws.2010.04.004 .

Sleigh BC, Manna B. Lymphedema. In: StatPearls. Treasure Island (FL): StatPearls Publishing. 2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK537239/ . Accessed 6 Mar 2023.

Fife CE, Farrow W, Hebert AA, Armer NC, Stewart BR, Cormier JN, Armer JM. Skin and wound care in lymphedema patients: a taxonomy, primer, and literature review. Adv Skin Wound Care. 2017;30(7):305–18. https://doi.org/10.1097/01.ASW.0000520501.23702.82 .

Johnson KC, Kennedy AG, Henry SM. Clinical measurements of lymphedema. Lymphat Res Biol. 2014;12(4):216–21. https://doi.org/10.1089/lrb.2014.0019 .

Greene AK, Goss JA. Diagnosis and staging of lymphedema. Semin Plast Surg. 2018;32(1):12–6. https://doi.org/10.1055/s-0038-1635117 .

Cheville AL, McGarvey CL, Petrek JA, Russo SA, Thiadens SR, Taylor ME. The grading of lymphedema in oncology clinical trials. Semin Radiat Oncol. 2003;13(3):214–25. https://doi.org/10.1016/S1053-4296(03)00038-9 .

Campisi C, Davini D, Bellini C, Taddei G, Villa G, Fulcheri E, Zilli A, Da Rin E, Eretta C, Boccardo F. Lymphatic microsurgery for the treatment of lymphedema. Microsurgery. 2006;26:65–9.

CTCAE 5.0. https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5x11.pdf . Accessed 10 May 2024.

CTCAE 3.0. https://ctep.cancer.gov/protocolDevelopment/electronic_applications/docs/ctcaev3.pdf . Accessed 10 May 2024.

Sanderson J, Tuttle N, Box R, Reul-Hirche HM, Laakso EL. The pitting test; an investigation of an unstandardized assessment of lymphedema. Lymphology. 2015;48(4):175–83.

CAS   PubMed   Google Scholar  

Sayegh HE, Asdourian MS, Swaroop MN, Brunelle CL, Skolny MN, Salama L, Taghian AG. Diagnostic methods, risk factors, prevention, and management of breast cancer-related lymphedema: past, present, and future directions. Curr Breast Cancer Rep. 2017;9(2):111–21. https://doi.org/10.1007/s12609-017-0237-8 .

Article   CAS   PubMed   PubMed Central   Google Scholar  

McLaughlin SA, Wright MJ, Morris KT, et al. Prevalence of lymphedema in women with breast cancer 5 years after sentinel lymph node biopsy or axillary dissection: objective measurements. J Clin Oncol. 2008;26:5213–9.

Ancukiewicz M, Miller CL, Skolny MN, O’Toole J, Warren LE, Jammallo LS, Specht MC, Taghian AG. Comparison of relative versus absolute arm size change as criteria for quantifying breast cancer-related lymphedema: the flaws in current studies and need for universal methodology. Breast Cancer Res Treat. 2012;135(1):145–52. https://doi.org/10.1007/s10549-012-2111-8 .

Hidding JT, Viehoff PB, Beurskens CH, van Laarhoven HW, Nijhuis-van der Sanden MW, van der Wees PJ. Measurement properties of instruments for measuring of lymphedema: systematic review. Phys Ther. 2016;96(12):1965–81. https://doi.org/10.2522/ptj.20150412 .

Dixon JB, Weiler MJ. Bridging the divide between pathogenesis and detection in lymphedema. Semin Cell Dev Biol. 2015;38:75–82. https://doi.org/10.1016/j.semcdb.2014.12.003 .

Tandra P, Kallam A, Krishnamurthy J. Identification and management of lymphedema in patients with breast cancer. J Oncol Pract. 2019;15(5):255–62. https://doi.org/10.1200/JOP.18.00141 .

Taylor R, Jayasinghe UW, Koelmeyer L, Ung O, Boyages J. Reliability and validity of arm volume measurements for assessment of lymphedema. Phys Ther. 2006;86:205–14.

Sander AP, Hajer NM, Hemenway K, Miller AC. Upper-extremity volume measurements in women with lymphedema: a comparison of measurements obtained via water displacement with geometrically determined volume. Phys Ther. 2002;82(12):1201–12.

Miller CL, Specht MC, Horick N, Skolny MN, Jammallo LS, O’Toole J, Taghian AG. A novel, validated method to quantify breast cancer-related lymphedema (BCRL) following bilateral breast surgery. Lymphology. 2013;46(2):64–74.

Sun F, Hall A, Tighe MP, Brunelle CL, Sayegh HE, Gillespie TC, Daniell KM, Taghian AG. Perometry versus simulated circumferential tape measurement for the detection of breast cancer-related lymphedema. Breast Cancer Res Treat. 2018;172(1):83–91. https://doi.org/10.1007/s10549-018-4902-z .

Megens AM, Harris SR, Kim-Sing C, McKenzie DC. Measurement of upper extremity volume in women after axillary dissection for breast cancer. Arch Phys Med Rehabil. 2001;82:1639–44.

Perdomo M, Levenhagen K, Davies C, Ryans K. Assessment measures of secondary lymphedema in breast cancer survivors. Rehabil Oncol. 2014;32(1):22–35.

Article   Google Scholar  

Sharkey AR, King SW, Kuo RY, Bickerton SB, Ramsden AJ, Furniss D. Measuring limb volume: accuracy and reliability of tape measurement versus perometer measurement. Lymphat Res Biol. 2018;16(2):182–6. https://doi.org/10.1089/lrb.2017.0039 .

Armer JM. The problem of post-breast cancer lymphedema: impact and measurement issues. Cancer Invest. 2005;23(1):76–83.

Lee MJ, Boland RA, Czerniec S, Kilbreath SL. Reliability and concurrent validity of the perometer for measuring hand volume in women with and without lymphedema. Lymphat Res Biol. 2011;9(1):13–8. https://doi.org/10.1089/lrb.2010.0021 .

Thomas B, Cornish BH, Ward LC. Bioelectrical impedance analysis for measurement of body fluid volumes a review. J Clin Eng. 1992;17:505–10.

York SL, Ward LC, Czerniec S, Lee MJ, Refshauge KM, Kilbreath SL. Single frequency versus bioimpedance spectroscopy for the assessment of lymphedema. Breast Cancer Res Treat. 2009;117(1):177–82.

Ward LC. Inter-instrument comparison of bioimpedance spectroscopic analysers. Open Med Device J. 2009;1:3–10.

Cornish B. Bioimpedance analysis: scientific background. Lymphat Res Biol. 2006;4(1):47–50.

Price KL, Earthman CP. Update on body composition tools in clinical settings: computed tomography, ultrasound, and bioimpedance applications for assessment and monitoring. Eur J Clin Nutr. 2019;73:187–93. https://doi.org/10.1038/s41430-018-0360-2 .

Hui D, Dev R, Pimental L, Park M, Cerana MA, Liu D, Bruera E. Association between multi-frequency phase angle and survival in patients with advanced cancer. J Pain Symptom Manage. 2017;53(3):571–7. https://doi.org/10.1016/j.jpainsymman.2016.09.016 .

Ward LC. Bioelectrical impedance spectrometry for the assessment of lymphoedema: principles and practice. In: Greene AK, Slavin SA, Brorson H, editors. Lymphedema. Switzerland: Springer International Publishing; 2015. p. 123–32.

Chapter   Google Scholar  

Fu MR, Cleland CM, Guth AA, Kayal M, Haber J, Cartwright F, Kleinman R, Kang Y, Scagliola J, Axelrod D. L-dex ratio in detecting breast cancer-related lymphedema: reliability, sensitivity, and specificity. Lymphology. 2013;46(2):85–96.

CAS   PubMed   PubMed Central   Google Scholar  

Soran A, Ozmen T, McGuire KP, Diego EJ, McAuliffe PF, Bonaventura M, Ahrendt GM, DeGore L, Johnson R. The importance of detection of subclinical lymphedema for the prevention of breast cancer-related clinical lymphedema after axillary lymph node dissection; a prospective observational study. Lymphat Res Biol. 2014;12(4):289–94.

Dylke ES, Schembri GP, Bailey DL, Bailey E, Ward LC, Refshauge K, Beith J, Black D, Kilbreath SL. Diagnosis of upper limb lymphedema: development of an evidence-based approach. Acta Oncol. 2016;55:1477–83. https://doi.org/10.1080/0284186X.2016.1191668 .

Kilbreath SL, Refshauge KM, Beith JM, Ward LC, Ung OA, Dylke ES, French JR, Yee J, Koelmeyer L, Gaitatzis K. Risk factors for lymphoedema in women with breast cancer: a large prospective cohort. Breast. 2016;28:29–36.

Tattersall J. Bioimpedance analysis in dialysis: state of the art and what we can expect. Blood Purif. 2009;27(1):70–4.

Sullivan PA, Still CD, Jamieson ST, Dixon CB, Irving BA, Andreacci JL. Evaluation of multi-frequency bioelectrical impedance analysis for the assessment of body composition in individuals with obesity. Obes Sci Pract. 2019;5(2):141–7.

Mayrovitz HN, Weingrad DN, Davey S. Tissue dielectric constant (TDC) measurements as a means of characterizing localized tissue water in arms of women with and without breast cancer treatment related lymphedema. Lymphology. 2014;47(3):142–50.

Jane Wigg J, Pugh S, Phillips N. The use of tissue dielectric constant in the assessment of lymphoedema. British Journal of Community Nursing, LTA: Compendium of best practice. 2023; S14-S8.

Mayrovitz HN, Somarriba C, Weingrad DN. Breast tissue dielectric constant as a potential breast edema assessment parameter. Lymphat Res Biol. 2022;20(1):33–8. https://doi.org/10.1089/lrb.2020.0137 .

Mayrovitz HN. Tissue dielectric constant of the lower leg as an index of skin water: temporal variations. Cureus. 2022;14(7):e26506. https://doi.org/10.7759/cureus.26506.PMID:35923478;PMCID:PMC9339369 .

Mayrovitz HN, Weingrad DN, Lopez L. Assessing localized skin-to-fat water in arms of women with breast cancer via tissue dielectric constant measurements in pre- and post-surgery patients. Ann Surg Oncol. 2015;22(5):1483–9. https://doi.org/10.1245/s10434-014-4185-5 .

Mazor M, Smoot BJ, Mastick J, Mausisa G, Paul SM, Kober KM, Elboim C, Singh K, Conley YP, Mickevicius G, Field J, Hutchison H, Miaskowski C. Assessment of local tissue water in the arms and trunk of breast cancer survivors with and without upper extremity lymphoedema. Clin Physiol Funct Imaging. 2019;39(1):57–64. https://doi.org/10.1111/cpf.12541 .

Chen Y-W, Tsai H-J, Hung H-C, Tsauo J-Y. Reliability study of measurements for lymphedema in breast cancer patients. Am J Phys Med Rehabil. 2008;87(1):33–8.

Moseley A, Piller N. Reliability of bioimpedance spectroscopy and tonometry after breast conserving cancer treatment. Lymphat Res Biol. 2008;6:85–7.

Piller N, Keeley V, Ryan TJ, Hayes S, Ridner S. Early detection—a strategy to reduce risk and severity? J Lymphoedema. 2009;4:89–95.

Liu NF, Olszewski W. Use of tonometry to assess lower extremity lymphedema. Lymphology. 1992;25:155–8.

Kar SK, Kar PK, Mania J. Tissue tonometry: a useful tool for assessing filarial lymphedema. Lymphology. 1992;25:55–61.

Binkley JM, Weiler MJ, Frank N, Bober L, Dixon JB, Stratford PW. Assessing arm volume in people during and after treatment for breast cancer: reliability and convergent validity of the LymphaTech System. Phys Ther. 2020;100:457–67. https://doi.org/10.1093/ptj/pzz175 .

Rumbo-Rodríguez L, Sánchez-SanSegundo M, Ferrer-Cascales R, García-D’Urso N, Hurtado-Sánchez JA, Zaragoza-Martí A. Comparison of body scanner and manual anthropometric measurements of body shape: a systematic review. Int J Environ Res Public Health. 2021;18:6213.

Beelen LM, van Dishoeck AM, Tsangaris E, et al. Patient-reported outcome measures in lymphedema: a systematic review and COSMIN analysis. Ann Surg Oncol. 2021;28(3):1656–68. https://doi.org/10.1245/s10434-020-09346-0 .

Eidenberger M. Patient-reported outcome measures with secondary lower limb lymphedemas: a systematic review. J Adv Pract Oncol. 2021;12(2):174–87. https://doi.org/10.6004/jadpro.2021.12.2.5 .

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. https://doi.org/10.1007/s11764-012-0247-5 .

Cemal Y, Jewell S, Albornoz CR, Pusic A, Mehrara BJ. Systematic review of quality of life and patient reported outcomes in patients with oncologic related lower extremity lymphedema. Lymphat Res Biol. 2013;11(1):14–9. https://doi.org/10.1089/lrb.2012.0015 .

Russo S, Walker JL, Carlson JW, et al. Standardization of lower extremity quantitative lymphedema measurements and associated patient-reported outcomes in gynecologic cancers. Gynecol Oncol. 2021;160(2):625–32. https://doi.org/10.1016/j.ygyno.2020.10.026 .

Coriddi M, Dayan J, Sobti N, et al. Systematic review of patient-reported outcomes following surgical treatment of lymphedema. Cancers (Basel). 2020. https://doi.org/10.3390/cancers12030565 .

Klassen AF, Tsangaris E, Kaur MN, et al. Development and psychometric validation of a patient-reported outcome measure for arm lymphedema: the LYMPH-Q upper extremity module. Ann Surg Oncol. 2021;28(9):5166–82. https://doi.org/10.1245/s10434-021-09887-y .

Williams AE, Rapport F, Russell IT, Hutchings HA. Psychometric development of the upper limb lymphedema quality of life questionnaire demonstrated the patient-reported outcome measure to be a robust measure for breast cancer-related lymphedema. J Clin Epidemiol. 2018;100:61–70. https://doi.org/10.1016/j.jclinepi.2018.04.014 .

Yost KJ, Cheville AL, Weaver AL, Al Hilli M, Dowdy SC. Development and validation of a self-report lower-extremity lymphedema screening questionnaire in women. Phys Ther. 2013;93(5):694–703. https://doi.org/10.2522/ptj.20120088 .

Armer JM, Radina ME, Porock D, Culbertson SD. Predicting breast cancer-related lymphedema using self-reported symptoms. Nurs Res. 2003;52(6):370–9. https://doi.org/10.1097/00006199-200311000-00004 .

Armer JM, Stewart BR. A comparison of four diagnostic criteria for lymphedema in a post-breast cancer population. Lymphat Res Biol. 2005;3(4):208–17. https://doi.org/10.1089/lrb.2005.3.208 .

Norman SA, Miller LT, Erikson HB, Norman MF, McCorkle R. Development and validation of a telephone questionnaire to characterize lymphedema in women treated for breast cancer. Phys Ther. 2001;81(6):1192–205.

Carter J, Raviv L, Appollo K, Baser RE, Iasonos A, Barakat RR. A pilot study using the gynecologic cancer lymphedema questionnaire (GCLQ) as a clinical care tool to identify lower extremity lymphedema in gynecologic cancer survivors. Gynecol Oncol. 2010;117(2):317–23. https://doi.org/10.1016/j.ygyno.2010.01.022 .

Benz T, Lehmann S, Sandor PS, Angst F. Relationship between subjectively-rated and objectively-tested physical function across six different medical diagnoses. J Rehabil Med. 2023. https://doi.org/10.2340/jrm.v55.9383 .

Xu C, Schaverien MV, Christensen JM, Sidey-Gibbons CJ. Efficient and precise ultra-QuickDASH scale measuring lymphedema impact developed using computerized adaptive testing. Qual Life Res. 2022;31(3):917–25. https://doi.org/10.1007/s11136-021-02979-y .

Deutscher D, Hayes D, Cook KF, et al. Upper quadrant edema patient-reported outcome measure is reliable, valid, and efficient for patients with lymphatic and venous disorders. Phys Ther. 2021. https://doi.org/10.1093/ptj/pzab219 .

Deutscher D, Kallen MA, Hayes D, et al. Lower quadrant edema patient-reported outcome measure is reliable, valid, and efficient for patients with lymphatic and venous disorders. Phys Ther. 2023. https://doi.org/10.1093/ptj/pzad083 .

Xu C, Christensen JM, Haykal T, Asaad M, Sidey-Gibbons C, Schaverien M. Measurement properties of the lymphedema life impact scale. Lymphat Res Biol. 2022;20(4):425–34. https://doi.org/10.1089/lrb.2021.0051 .

Greene AK, Slavin SA. What is the best imaging modality to confirm the diagnosis of lymphedema? In: Weinzweig J, editor. Plastic surgery secrets plus. 2nd ed. Amsterdam: Elsevier; 2010. p. 630–5.

Weber E, Aglianò M, Bertelli E, Gabriele G, Gennaro P, Barone V. Lymphatic collecting vessels in health and disease: a review of histopathological modifications in lymphedema. Lymphat Res Biol. 2022;20(5):468–77. https://doi.org/10.1089/lrb.2021.0090 .

Committee E. The Diagnosis and treatment of peripheral lymphedema: 2016 consensus document of the international society of lymphology. Lymphology. 2016;49(4):170–84.

Pappalardo M, Cheng MH. Lymphoscintigraphy for the diagnosis of extremity lymphedema: current controversies regarding protocol, interpretation, and clinical application. J Surg Oncol. 2020;121(1):37–47. https://doi.org/10.1002/jso.25526 .

Pappalardo M, Cheng MH. A new lymphoscintigraphy staging for unilateral extremity lymphedema: validation and correlation between nuclear images and clinical findings. Plast Reconstr Surg Glob Open. 2018;6(9S):74–5.

Article   PubMed Central   Google Scholar  

Dylke ES, McEntee MF, Schembri GP, Brennan PC, Bailey E, Ward LC, Kilbreath SL. Reliability of a radiological grading system for dermal backflow in lymphoscintigraphy imaging. Acad Radiol. 2013;20(6):758–63.

Oh A, Kajita H, Imanishi N, Sakuma H, Takatsume Y, Okabe K, Aiso S, Kishi K. Three-dimensional analysis of dermal backflow in cancer-related lymphedema using photoacoustic lymphangiography. Arch Plast Surg. 2022;49(01):99–107.

Villa G, Campisi CC, Ryan M, Boccardo F, Di Summa P, Frascio M, Sambuceti G, Campisi C. Procedural recommendations for lymphoscintigraphy in the diagnosis of peripheral lymphedema: the Genoa protocol. Nucl Med Mol Imaging. 2019;53:47–56.

Yoon HJ, Woo KJ, Kim JY, Kang SY, Moon BS, Kim BS. The added value of SPECT/CT lymphoscintigraphy in the initial assessment of secondary extremity lymphedema patients. Sci Rep. 2023;13(1):19494. https://doi.org/10.1038/s41598-023-44471-2 .

Tassenoy A, De Strijcker D, Adriaenssens N, Lievens P. The use of noninvasive imaging techniques in the assessment of secondary lymphedema tissue changes as part of staging lymphedema. Lymphat Res Biol. 2016;14(3):127–33.

Ricci V, Ricci C, Gervasoni F, Giulio C, Farì G, Andreoli A, Özçakar L. From physical to ultrasound examination in lymphedema: a novel dynamic approach. J Ultrasound. 2022. https://doi.org/10.1007/s40477-021-00633-4 .

Iker E, Mayfield CK, Gould DJ, Patel KM. Characterizing lower extremity lymphedema and lipedema with cutaneous ultrasonography and an objective computer-assisted measurement of dermal echogenicity. Lymphat Res Biol. 2019;17(5):525–30.

Forte AJ, Huayllani MT, Boczar D, Cinotto G, McLaughlin SA. Ultrasound elastography use in lower extremity lymphedema: a systematic review of the literature. Cureus. 2019. https://doi.org/10.7759/cureus.5578 .

Bianchi A, Visconti G, Hayashi A, Santoro A, Longo V, Salgarello M. Ultra-high frequency ultrasound imaging of lymphatic channels correlates with their histological features: a step forward in lymphatic surgery. J Plast Reconstr Aesthet Surg. 2020;73(9):1622–9.

Naouri M, Samimi M, Atlan M, Perrodeau E, Vallin C, Zakine G, Vaillant L, Machet L. High-resolution cutaneous ultrasonography to differentiate lipoedema from lymphoedema. Br J Dermatol. 2010;163(2):296–301.

Monnin-Delhom ED, Gallix BP, Archard C, Bruel JM, Janbon C. High resolution unenhanced computed tomography in patients with swollen legs. Lymphology. 2002;35(3):121–8.

Lee DG, Lee S, Kim KT. Computed tomography-based quantitative analysis of fibrotic changes in skin and subcutaneous tissue in lower extremity lymphedema following gynecologic cancer surgery. Lymphat Res Biol. 2022;20(5):488–95.

Shin SU, Lee W, Park EA, Shin CI, Chung JW, Park JH. Comparison of characteristic CT findings of lymphedema, cellulitis, and generalized edema in lower leg swelling. Int J Cardiovasc Imaging. 2013;29:135–43.

Yoo JS, Chung SH, Lim MC, Kim YJ, Kim KG, Hwang JH, Kim YH. Computed tomography-based quantitative assessment of lower extremity lymphedema following treatment for gynecologic cancer. J Gynecol Oncol. 2017;28(2):e18. https://doi.org/10.3802/jgo.2017.28.e18 .

Mihara M, Hara H, Araki J, Kikuchi K, Narushima M, Yamamoto T, Iida T, Yoshimatsu H, Murai N, Mitsui K, Okitsu T, Koshima I. Indocyanine green (ICG) lymphography is superior to lymphoscintigraphy for diagnostic imaging of early lymphedema of the upper limbs. PLoS ONE. 2012;7:e38182. https://doi.org/10.1371/journal.pone.0038182 .

Rasmussen JC, Tan I-C, Marshall MV, Adams KE, Kwon S, Fife CE, Maus EA, Smith LA, Covington KR, Sevick-Muraca EM. Human lymphatic architecture and dynamic transport imaged using near-infrared fluorescence. Transl Oncol. 2010;3:362–72. https://doi.org/10.1593/tlo/10190 .

Zhu B, Rasmussen JC, Sevick-Muraca EM. A matter of collection and detection for intraoperative and noninvasive near-infrared fluorescence molecular imaging: to see or not to see? Med Phy. 2014;41:022105.

Abbaci M, Conversano A, De Leeuw F, Laplace-Builhé C, Mazouni C. Near-infrared fluorescence imaging for the prevention and management of breast cancer-related lymphedema: a systematic review. Eur J Surg Oncol. 2019;45:1778–86. https://doi.org/10.1016/j.ejso.2019.06.009 .

Aldrich MB, Rasmussen JC, Fife CE, Shaitelman SF, Sevick-Muraca EM. The development and treatment of lymphatic dysfunction in cancer patients and survivors. Cancers. 2020;12:2280. https://doi.org/10.3390/cancers120822808 .

Stout Gergich NL, Pfalzer LA, McGarvey C, Springer B, Gerber LH, Soballe P. Preoperative assessment enables the early diagnosis and successful treatment of lymphedema. Cancer. 2008;112(12):2809–19. https://doi.org/10.1002/cncr.23494 .

Aldrich MB, Rasmussen JC, DeSnyder SM, Woodward WA, Chan W, Sevick-Muraca EM, Mittendorf EA, Smith DB, Stauder MC, Strom EA, Perkins GH, Hoffman KE, Mitchell MP, Barcenas CH, Isales LE, Shaitelman SF. Prediction of breast cancer-related lymphedema by dermal backflow detected with near-infrared fluorescence lymphatic imaging. Breast Cancer Res Treat. 2022;195:33–41. https://doi.org/10.1007/s10549-022-06667-4 .

Stolarz AJ, Sarimollaoglu M, Marecki JC, Fletcher TW, Galanzha EI, Rhee SW, Zharov VP, Klimberg VS, Rusch NJ. Doxorubicin activates ryanodine receptors in rat lymphatic muscle cells to attenuate rhythmic contractions and lymph flow. J Pharmacol Exp Ther. 2019;371(2):278–89. https://doi.org/10.1124/jpet.119.257592 .

Aldrich MB, Sevick-Muraca EM. Cytokines are systemic effectors of lymphatic function in acute inflammation. Cytokine. 2013;64(1):362–9. https://doi.org/10.1016/j.cyto.2013.05.015 .

Tan I-C, Maus EA, Rasmussen JC, Marshall MV, Adams KE, Fife CE, Smith LA, Chan W, Sevick-Muraca EM. Assessment of lymphatic contractile function following manual lymphatic drainage using near-infrared fluorescence imaging. Arch Phys Med Rehabil. 2011;92:756-764.e1. https://doi.org/10.1016/j.apmr.2010.12.027 .

Adams KE, Rasmussen JC, Darne C, Tan I-C, Aldrich MB, Marshall MV, Fife CE, Maus EA, Smith LA, Guilloid R, Hoy S, Sevick-Muraca EM. Direct evidence of lymphatic function improvement after advanced pneumatic compression device treatment of lymphedema. Biomed Opt Express. 2010;1:114–25.

Aldrich MB, Gross D, Morrow JR, Fife CE, Rasmussen JC. Effect of pneumatic compression therapy on lymph movement in lymphedema-affected extremities, as assessed by near-infrared fluorescence lymphatic imaging. J Innov Opt Health Sci. 2017;10:1650049. https://doi.org/10.1142/S1793545816500498 .

Belgrado JP, Vandermeeren L, Vankerckhove S, Valsamis JB, Malloizel-Delaunay J, Moraine JJ, Liebens F. Near-infrared fluorescence lymphatic imaging to reconsider occlusion pressure of superficial lymphatic collectors in upper extremities of healthy volunteers. Lymphat Res Biol. 2016;14:70–7.

Lipman K, Luan A, Stone K, Wapnir I, Karin M, Nguyen D. Lymphatic microsurgical preventive healing approach (LYMPHA) for lymphedema prevention after axillary lymph node dissection—a single institution experience and feasibility of technique. J Clin Med. 2022;11:92.

Article   CAS   Google Scholar  

Borri M, Gordon KD, Hughes JC, Scurr ED, Koh DM, Leach MO, Mortimer PS, Schmidt MA. Magnetic resonance imaging-based assessment of breast cancer-related lymphoedema tissue composition. Invest Radiol. 2017;52(9):554–61.

Crescenzi R, Donahue PMC, Garza M, Lee CA, Patel NJ, Gonzalez V, Jones RS, Donahue MJ. Elevated magnetic resonance imaging measures of adipose tissue deposition in women with breast cancer treatment-related lymphedema. Breast Cancer Res Treat. 2022;191(1):115–24.

Mills M, van Zanten M, Borri M, Mortimer PS, Gordon K, Ostergaard P, Howe FA. Systematic review of magnetic resonance lymphangiography from a technical perspective. J Magn Reson Imaging. 2021;53(6):1766–90.

Donahue PMC, Crescenzi R, Petersen KJ, Garza M, Patel N, Lee C, Chen SC, Donahue MJ. Physical therapy in women with early stage lipedema: potential impact of multimodal manual therapy, compression, exercise, and education interventions. Lymphat Res Biol. 2021. https://doi.org/10.1089/lrb.2021.0039 .

Crescenzi R, Donahue PMC, Hartley KG, Desai AA, Scott AO, Braxton V, Mahany H, Lants SK, Donahue MJ. Lymphedema evaluation using noninvasive 3T MR lymphangiography. J Magn Reson Imaging. 2017;46(5):1349–60. https://doi.org/10.1002/jmri.25670 .

Salehi BP, Sibley RC, Friedman R, Kim G, Singhal D, Loening AM, Tsai LL. MRI of lymphedema. J Magn Reson Imaging. 2023;57(4):977–91.

Rane S, Donahue PMC, Towse T, Ridner S, Chappell M, Jordi J, Gore J, Donahue MJ. Clinical feasibility of noninvasive visualization of lymphatic flow with principles of spin labeling MR imaging: implications for lymphedema assessment. Radiology. 2013;269(3):893–902. https://doi.org/10.1148/radiol.13120145 .

Crescenzi R, Donahue PM, Braxton VG, Scott AO, Mahany HB, Lants SK, Donahue MJ. 3.0 T relaxation time measurements of human lymph nodes in adults with and without lymphatic insufficiency: Implications for magnetic resonance lymphatic imaging. NMR Biomed. 2018;31(12):e4009.

Donahue PM, Crescenzi R, Scott AO, Braxton V, Desai A, Smith SA, Jordi J, Meszoely IM, Grau AM, Kauffmann RM. Bilateral changes in deep tissue environment after manual lymphatic drainage in patients with breast cancer treatment-related lymphedema. Lymphat Res Biol. 2017;15(1):45–56.

Maki JH, Neligan PC, Briller N, Mitsumori LM, Wilson GJ. Dark blood magnetic resonance lymphangiography using dual-agent relaxivity contrast (DARC-MRL): a novel method combining gadolinium and iron contrast agents. Curr Probl Diagn Radiol. 2016;45(3):174–9.

Crescenzi R, Donahue PMC, Mahany H, Lants SK, Donahue MJ. CEST MRI quantification procedures for breast cancer treatment-related lymphedema therapy evaluation. Magn Reson Med. 2020;83(5):1760–73.

Crescenzi R, Donahue PMC, Petersen KJ, Garza M, Patel N, Lee C, Beckman JA, Donahue MJ. Upper and lower extremity measurement of tissue sodium and fat content in patients with lipedema. Obesity (Silver Spring). 2020;28(5):907–15.

Crescenzi R, Marton A, Donahue PMC, Mahany HB, Lants SK, Wang P, Beckman JA, Donahue MJ, Titze J. Tissue sodium content is elevated in the skin and subcutaneous adipose tissue in women with lipedema. Obesity (Silver Spring). 2018;26(2):310–7.

Munoz DC, Virk SS, Oladeru OT, et al. Practical approach to establishing a lymphedema screening program: tips and tricks. Curr Breast Cancer Rep. 2023;15(3):242–51. https://doi.org/10.1007/s12609-023-00501-6 .

Ward LC, Thompson B, Gaitatzis K, Koelmeyer LA. Comparison of volume measurements and bioimpedance spectroscopy using a stand-on device for assessment of unilateral breast cancer-related lymphedema. Eur J Breast Health. 2024;20(2):141–8. https://doi.org/10.4274/ejbh.galenos.2024.2023-12-8.PMID:38571690;PMCID:PMC10985575 .

Qin ES, Bowen MJ, Chen WF. Diagnostic accuracy of bioimpedance spectroscopy in patients with lymphedema: a retrospective cohort analysis. J Plast Reconstr Aesthet Surg. 2018;71(7):1041–50. https://doi.org/10.1016/j.bjps.2018.02.012 .

Ridner SH, Dietrich MS, Boyages J, Koelmeyer L, Elder E, Hughes TM, French J, Ngui N, Hsu J, Abramson VG, Moore A. A comparison of bioimpedance spectroscopy or tape measure triggered compression intervention in chronic breast cancer lymphedema prevention. Lymphat Res Biol. 2022;20(6):618–28.

Bundred NJ, Stockton C, Keeley V, Riches K, Ashcroft L, Evans A, Skene A, Purushotham A, Bramley M, Hodgkiss T, Investigators of BEA PLACE studies. Comparison of multi-frequency bioimpedance with perometry for the early detection and intervention of lymphoedema after axillary node clearance for breast cancer. Breast Cancer Res Treat. 2015;151(1):121–9. https://doi.org/10.1007/s10549-015-3357-8 .

Ward LC, Czerniec S, Kilbreath SL. Operational equivalence of bioimpedance indices and perometry for the assessment of unilateral arm lymphedema. Lymphat Res Biol. 2009;7(2):81–5. https://doi.org/10.1089/lrb.2008.1027 .

Coroneos CJ, Wong FC, DeSnyder SM, Shaitelman SF, Schaverien MV. Correlation of L-dex bioimpedance spectroscopy with limb volume and lymphatic function in lymphedema. Lymphat Res Biol. 2019;17(3):301–7. https://doi.org/10.1089/lrb.2018.0028 .

Czerniec SA, Ward LC, Lee MJ, et al. Segmental measurement of breast cancer-related arm lymphoedema using perometry and bioimpedance spectroscopy. Support Care Cancer. 2011;19:703–10. https://doi.org/10.1007/s00520-010-0896-8 .

Koelmeyer L, Thompson B, Mackie H, Blackwell R, Heydon-White A, Moloney E, Gaitatzis K, Boyages J, Suami H. Personalizing conservative lymphedema management using ICG-guided manual lymphatic drainage. Lymphat Res Biol. 2021;19(1):56–65.

Benjamin J, O’Leary C, Hur S, Gurevich A, Klein WM, Itkin M. Imaging and interventions for lymphatic and lymphatic-related disorders. Radiology. 2023;307(3):e220231. https://doi.org/10.1148/radiol.220231 .

Moon T, O’Donnell TF, Weycker D, Iafrati M. Lymphoscintigraphy is frequently recommended but seldom used in a “real world setting.” J Vasc Surg Venous Lymphat Disord. 2024;12(2):101738. https://doi.org/10.1016/j.jvsv.2023.101738 .

Guermazi A, Brice P, Hennequin C, Sarfati E. Lymphography: an old technique retains its usefulness. Radiographics. 2003;23(6):1541–58. https://doi.org/10.1148/rg.236035704 .

Fadhil M, Singh R, Havas T, Jacobson I. Systematic review of head and neck lymphedema assessment. Head Neck. 2022;44(10):2301–15. https://doi.org/10.1002/hed.27136 .

van Schaik CJ, Boer LL, Draaisma JMT, van der Vleuten CJM, Janssen JJ, Fütterer JJ, Schultze Kool LJ, Klein WM. The lymphatic system throughout history: From hieroglyphic translations to state of the art radiological techniques. Clin Anat. 2022;35(6):701–10. https://doi.org/10.1002/ca.23867 .

Download references

Acknowledgements

This research article was prepared for the 2023 Lymphedema Summit: Forward Momentum; Future Steps in Lymphedema Management. This Summit was sponsored by the American Cancer Society, the Lymphology Association of North America, Washington University School of Medicine in St. Louis, and the Stryker Corporation.

Open Access funding enabled and organized by CAUL and its Member Institutions. Lymphology Association of North America,Washington University in St. Louis,Stryker Corporation,American Cancer Society

Author information

Authors and affiliations.

Department of Physical Medicine and Rehabilitation, MetroHealth Rehabilitation Institute, Case Western Reserve University, Cleveland, OH, USA

Center for Molecular Imaging, UTHealth, Houston, TX, USA

Melissa Aldrich

Department of Physical Medicine and Rehabilitation, Vanderbilt University Medical Center, Nashville, TN, USA

Paula Donahue

Lymphedema Center, Santa Monica, CA, USA

Australian Lymphoedema Education, Research & Treatment (ALERT) Program, Macquarie University, Sydney, Australia

Louise Koelmeyer

Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA

Rachelle Crescenzi

Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA

Andrea Cheville

You can also search for this author in PubMed   Google Scholar

Contributions

All authors made substantial contributions to the conception, methodology, literature search, analysis, synthesis and consensus of information, and manuscript writing. PD assisted in identifying multiple members in the formation of this group. AC provided the concept and initial content for the clinical integration tables, and all authors substantially contributed successive iterations of these tables. EI prepared Figs.  1 , 2 , MA prepared Figs.  3 , 4 , and RC prepared Fig.  5 . PD and LK provided depth of expertise on emerging office-based tools, and EI, MA and RC on the imaging modalities. MV provided administrative oversight and overall synthesis of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Louise Koelmeyer .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ .

Reprints and permissions

About this article

Vargo, M., Aldrich, M., Donahue, P. et al. Current diagnostic and quantitative techniques in the field of lymphedema management: a critical review. Med Oncol 41 , 241 (2024). https://doi.org/10.1007/s12032-024-02472-9

Download citation

Received : 31 May 2024

Accepted : 02 August 2024

Published : 05 September 2024

DOI : https://doi.org/10.1007/s12032-024-02472-9

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Measurement

Advertisement

  • Find a journal
  • Publish with us
  • Track your research

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Turk J Surg
  • v.33(2); 2017

Logo of turkjsurgery

Lymphedema: From diagnosis to treatment

Oğuz kayıran.

1 Division of Breast Surgery and Lymphedema Program, Magee-Womens Hospital of University of Pittsburgh Medical Center, Pittsburgh, USA

2 Department of Plastic and Reconstructive Surgery, Baltalimani Hospital, İstanbul, Turkey

Carolyn De La Cruz

3 Department of Plastic and Reconstructive Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA

Atilla Soran

Lymphedema is a chronic and progressive disorder resulting from impaired lymphatic system function. In developed countries, upper extremity lymphedema is mainly the consequence of breast cancer surgery in which axillary lymph node dissection and radiation alter upper extremity lymphatic flow.

Diagnosis of lymphedema is made clinically. Nevertheless, there are numerous diagnostic tools available for disease staging. Recently, a new technology namely magnetic resonance lymphangiography has emerged in the medical field to assist in both diagnosis and management.

There are non-surgical and surgical treatment options available. Non-surgical methods are always the first-line treatment; however, surgical options can be explored in appropriate patients. Recent studies focus on the prevention of lymphedema using surgical techniques utilizing axillary reverse mapping to delineate arm lymphatics from axillary lymphatics.

Finding the most suitable technique for each type of lymphedema with variable stages is one of the most complicated decisions for practitioners. More studies are needed to reveal the exact biology of lymphedema to ensure complete understanding of the disease and improve outcomes.

INTRODUCTION

Lymphedema (LE) is the accumulation of protein-rich fluid in tissues. The impaired function of lymph vessels interrupts the drainage of lymphatic system that is a part of the circulatory system just like the arterial and venous structures. Lymph vessels remove excess fluid from tissues and transport it back to the circulation. In addition, the maturation of immune cells takes place in the lymphatic system; thus, it constitutes one of the most critical defense mechanisms throughout the body. Lymph capillaries are located in the dermis, woven like a cobweb, then drain to lymphatic vessels in the subcutaneous plane and ultimately to the deeper system and the thoracic duct.

Lymphedema can either be primary or secondary. Regardless of the etiology, it is clinically characterized with chronic swelling, localized pain, atrophic skin changes and secondary infections ( 1 ). However, the main devastating aspect of LE is the appearance of the affected limb that causes psychological morbidity. Primary LE is related to developmental abnormalities of the lymphatic system whereas secondary LE is attributed to the impairment of lymphatic vessels due to an acquired condition such as trauma, tumor, surgery or infection ( Table 1 ). In developing countries, secondary LE is mainly due to infections-infestations influencing lymphatic channels. On the other hand, in developed countries, secondary LE occurs most commonly after surgical removal of lymph nodes for cancer treatment ( 2 ). Breast cancer is the most common cancer among women in the world and Breast Cancer Related Lymphedema (BCRL) occurs more often than any other type of LE ( 3 ). This review will focus on BCRL.

The etiology of lymphedema

PrimarySecondary
CongenitalTrauma
Milroy diseaseTumor
Lymphedema praecoxSurgery
Lymphedema tardaInfection-infestation
Post-venous thrombosis

Breast Cancer Related Lymphedema is detected in 7–77% of patients who undergo axillary lymph node dissection (ALND) due to transection of lymph vessels as depicted in selected studies ( 4 ). Sentinel lymph node biopsy (SLNB) significantly reduces this risk to 3–7% ( 5 , 6 ). This incidence is based on multiple factors such as extent of disease, treatment modality (i.e. radiotherapy), and duration of follow-up ( 6 , 7 ). In addition to these, a study revealed that patients with occupations that require extra upper extremity activity had the worst stage and grade LE clinically ( 3 ). In another study, factors associated with the development of BCRL included occupation, infection, and increased BMI. Immediate reconstruction of the breast was not found as a risk factor for BCRL ( 8 ). A recent study reported a risk assessment tool using these BCRL predictors (RATE-L), which included significant predictors such as post-mastectomy radiation, age above 65 years, and axillary dissection ( 9 ).

Although there is no unique sign or criteria for defining LE, the diagnosis is usually made clinically by thorough evaluation and physical examination ( 2 , 3 , 10 ). Family history is important in the diagnosis of primary LE. The main symptoms are chronic swelling, progressive atrophic skin changes, and recurrent infections. It is important to identify whether the swelling is transient or persistent. In one study, it was reported that one-third of initial swelling attacks were transient ( 10 ). Since effective treatment of LE can be established in early stages, accurate and timely diagnosis is crucial ( 11 ). History of trauma or surgery must be addressed clearly. Physical examination consists of volume and shape discrepancies, and skin changes among the extremities. Figure 1 outlines the alternatives in the diagnosis of LE.

An external file that holds a picture, illustration, etc.
Object name is tjs-33-2-51-g01.jpg

A scheme for the available options in the diagnosis of lymphedema

BIS: bioimpedence spectroscopy; MRI: magnetic resonance imaging; NIRF: near infra-red fluorescence imaging; PDE: photo dynamic eye (PDE; Hamamatsu Photonics K.K., Hamamatsu, Japan); USG: ultrasonography; CT: computed tomography

Circumferential (>2 cm) and/or volume (>200 mL) differences between the affected and non-affected extremity can be performed to confirm the diagnosis ( 2 ). Volume can be measured by tape, water displacement or perometry (Perometer; Perosystems, Wuppertal, Germany) ( 12 ). Tape measurements require formula calculations; therefore, it is recommended that the measurements should be performed by the same person at defined intervals ( 12 ). It is mainly preferred on head and neck lymphedema follow-ups. Water displacement is an accurate method that is the gold-standard for volume assessment, especially on extremities ( 12 ); however, it is not used in daily practice because it doesn’t delineate the affected area. If there is an open wound, it is not feasible to use this technique. Perometry is a computer-based study that calculates the volume of the affected limb via infra-red optical electronic scanner and can demonstrate small changes, but is expensive ( 2 ).

Non-invasive measurements (tonometry, bioimpedence spectroscopy) and imaging techniques (lymphoscintigraphy, ultrasonography, computed tomography, and magnetic resonance imaging) may aid in the detection of LE. The principal mechanism of a tissue tonometer is to evaluate tissue resistance by applying compression. Skin pliability and fibrosis can be measured with a tonometer. It gives a good idea about how changes occur during LE treatment. Tissue dielectric constant and tonometry can measure skin texture and resistance ( 12 – 14 ). Ultrasonography, computed tomography and magnetic resonance imaging can show the presence of extra fluid within tissues ( 12 ).

Bioimpedence spectroscopy (BIS) is a new diagnostic tool to diagnose LE. It is a technique that assesses the extracellular fluid compartment before visible changes have settled ( 15 ). BIS mainly focuses on changes in electrical conductance of extracellular fluid. Since it depends on water content of the study area, advanced and fibrotic edema detected in late-stage LE may not be diagnosed properly by BIS ( 12 ). In other words, BIS is reliable in early-stage BCRL. A prospective observational study demonstrated the impact of L-Dex® (L-Dex; Impedimed, Brisbane, Australia) measurements in identification of subclinical BCRL and its use in routine clinical practice ( 16 ). L-Dex® is the score representing extracellular fluid ratio of the affected limb to the unaffected limb, and is sensitive in predicting the onset of LE up to 10 months prior to clinical diagnosis ( 15 ).

Lymphoscintigraphy is a nuclear medicine study and demonstrates slow or absent lymph flow usually in later stages of LE ( 12 ). Technetium 99m sulfur colloid is injected intradermally and the transit time to lymph node basins can be measured; however, subdermal lymphatics cannot be assessed. A new technique for imaging lymph vessels is Near Infra-Red Fluorescence Imaging (NIRF) by using indocyanine green. This dynamic test allows visualization of superficial lymphatic flow and functioning lymphatic vessels; thus, finds abnormalities at early stages. It can be used to stage the severity of disease and for preoperative-intraoperative planning ( 17 , 18 ) ( Figure 2 ). Lymphography is another entity where radio-opaque material is directly injected into peripheral lymph vessels. This technique is rarely done due to the risk of damaging lymph vessels.

An external file that holds a picture, illustration, etc.
Object name is tjs-33-2-51-g02.jpg

Near Infra-Red Fluorescence imaging (NIRF) for preoperative-intraoperative planning

Magnetic resonance lymphangiography is a new entity that involves the injection of Gadolinium into the hand or foot to clarify the course of lymphatics. Gadolinium can also get into the venous system making the interpretation of lymphatic channels difficult ( 19 ). Magnetic resonance venogram and ferumoxytole (Feraheme; Advanced Magnetics, Cambridge, MA, USA) are used to help to differentiate lymphatics from veins ( 19 , 20 ). With the advent of magnetic resonance lymphangiography, the severity of LE can be delineated while the anatomy of the lymphatic channels and the status of the soft tissues can also be depicted ( 20 ) ( Figure 3 ). We suggested an algorithm for the management of patients with LE, by using Magnetic resonance lymphangiography and LE stage in this review ( Figure 4 ).

An external file that holds a picture, illustration, etc.
Object name is tjs-33-2-51-g03.jpg

a, b. (a) Magnetic resonance lymphangiography. Three irregular tubular structures extending from the dorsal aspect of the wrist to the lateral/dorsal aspect of the right forearm are compatible with enlarged lymphatics. These vessels are subcutaneous and measure up to 3–4 mm in caliber. In the lateral and ventral aspect of the mid-portion of the forearm (in proximity to the expected location of two lymphovenuler anastomosis), there seems to be communication between these lymphatics and small venules, branches of the basilic and ventral branches of the cephalic vein. There is minimal dermal backflow in the lateral aspect of the mid-portion of the right forearm. (b) Lymphoscintigraphy. Abnormally delayed lymphatic transit and dermal backflow are identified in the left forearm

An external file that holds a picture, illustration, etc.
Object name is tjs-33-2-51-g04.jpg

An algorithm for the treatment of lymphedema according to magnetic resonance lymphangiography

LVA: lymphaticovenular bypass; VLNT: vascularized lymph node transfer

The International Society of Lymphology classified LE into 4 stages to overcome multiple classification schemes and obtain a consensus ( 3 ) ( Table 2 ). In addition, Campisi et al. ( 21 ) proposed a staging system consisting of 3 stages, especially for elders.

Staging of lymphedema adapted from The International Society of Lymphology

StageDescriptionCharacteristics
0LatentSome damage to lymphatics; No visible edema yet
1Spontaneously reversible, acute phasePitting edema; reversible with elevation of the arm. Usually, upon waking in the morning, the limb(s) or affected area is normal or almost normal size
2Spontaneously irreversible, chronic phaseSpongy consistency and is “non-pitting,” Fibrosis found in Stage 2 lymphedema marks the beginning of the hardening of the limbs and increasing size
3Elephantiasis; irreversible, end-stageIrreversible and usually the limb(s) is/are very large. The tissue is hard (fibrotic) and unresponsive; consider debulking surgey at this stage

The management of LE consists of accurate diagnosis, successful classification and patient education. Unfortunately, there is no absolute cure for LE. On the other hand, effective treatment is available. Two main modalities include non-surgical and surgical options ( Table 3 ). The mainstays of non-surgical LE treatment modalities are complete decongestive therapy (CDT), compression therapy, advanced pneumatic compression pumps and exercise. These treatments are effective mainly in early-stage LE ( 2 ). There is a global trend for surgical intervention and surgical techniques including physiological and reductive methods.

Treatment options in lymphedema

Non-surgical treatmentsSurgical treatments
Complete decongestive therapyReductive techniques
 Manual lymph drainage Direct excision
 Compression therapy Liposuction
 ExercisePhysiological techniques
 Skin care Lymphatico-lymphatic by-pass
 Compression garments Lymphatico-venous by-pass
Advanced pneumatic compression therapy Lymph node transfer

Laser therapy

Non-Surgical Treatments

Patient education is both crucial and mandatory ( 22 ). Self-care and risk-reductive practices, self-lymph drainage, skin care, proper alignment of bandages and garments, good nutrition, exercise and weight control are the basics prior to LE treatment ( 12 ).

Complete Decongestive Therapy

Complete Decongestive Therapy (CDT) is considered the gold-standard treatment method in the management of LE and includes two phases: reductive (phase 1) and maintenance (phase 2) ( 23 ). CDT is a good option in decreasing LE volume and includes manual lymph drainage, compression therapy, physical exercise, skin care as self-management, followed by wearing compression garments ( 23 , 24 ). Although it is safe and effective in most patients, it is expensive, time-consuming and needs certified therapists. In addition, patient compliance to long-term CDT is challenging. Nevertheless, CDT can be individualized with modifications until the lymphedematous volume reduction has been maximized.

Manual lymph drainage (MLD)

MLD is a hands-on technique and differs from standard massage by orienting the lymphedematous fluid to proper functioning lymphatics ( 24 ).

Compression Therapy

Compression therapy includes effective gradient compression with tubular bandaging on the affected limb ( 25 ). Short-stretch bandages provide low “resting pressure” when the patient is at rest and “working pressure” which allows muscle contractions to direct interstitial fluid flow ( 23 , 25 ). These bandages also reduce fibrosis in the skin ( 25 ). Compression garments are different from compression bandages and are preferred in long-term treatment.

Specific exercise is beneficial for LE patients ( 12 ). It is recommended that compression bandages or garments should be worn during activity ( 12 ). Patients with LE or people at-risk for LE are encouraged to exercise. A meta-analysis showed that active exercising reduces edema volume in BCRL ( 26 ). A recent pilot study demonstrated that yoga has beneficial effects on an individuals’ posture and strength ( 27 ).

Establishing proper hygiene is important for patients with LE. Low pH moisturizers are recommended to overcome skin cracking and drying, in order to prevent entrance of microorganisms ( 12 ).

Compression Garments

Initial control of LE can be achieved with the use of compression bandages. Long term control is obtained with compression garments ( 12 ). The type of the garment depends on the body part. Patients should have several garments to alternate and ensure the proper pressure and hygienic control. Accurate fitted garments are essential. Some patients require additional coverage night-or-day to control or reduce LE ( 12 ).

Advanced New Generation Pneumatic Compression Therapy

Advanced Pneumatic Compression (APC) therapy can be used as an adjunct to CDT either in early or late phases ( 12 , 28 ). It mimics the pump effect of muscular contraction on lymphatic system ( 2 ). Ranging between 35 and 180 mm-Hg, pump pressures are adjusted to mostly 20–60 mm-Hg ( 2 , 12 ). The pressure must be individualized in order to prevent skin damage during application. APC therapy was found beneficial in reducing LE, whereas compression sleeves prevented additional swelling without influencing volume reduction ( 2 ).

Laser Therapy

A number of randomized trials have reported that Low-Level Laser Therapy (LLLT) improved measurable physical parameters as well as subjective pain scores ( 29 ). LLLT increases lymphatic drainage by stimulating the formation of new lymph vessels, by improving lymphatic motricity, and by preventing formation of fibrotic tissue ( 30 ). Usually, LLLT is used in combination with CDT. Most studies did not report any adverse events to participants, although one study stated development of cellulitis in LLLT patients as an adverse event ( 31 ). Its causal relationship to LLLT was unknown.

SURGICAL TREATMENTS

Reductive techniques, direct excision.

These techniques include removal of lymphedematous tissue. Previously described methods such as the Charles procedure include complete removal of all subcutaneous tissue and skin grafting ( 32 ). This method, although effective at volume reduction, can be quite disfiguring. It also can require blood transfusions and lengthy wound healing. Another technique used in the past involved buried dermal flaps with variable success ( 33 ). Direct excision techniques may involve full-thickness skin grafting (FTSG) or vacuum-assisted closure therapy ( 2 ). In extreme cases, these techniques allow for improvement in quality of life.

Liposuction

Surgical debulking of the affected extremity using liposuction has been shown to be very effective at reducing the volume to near normal ( 34 ). This technique has been used in both congenital and acquired LE. It has also been used in cases of lipedema. It has been reported that liposuction technique provides long standing reduction in extremity size as compared to the normal side ( 35 ). This technique requires patient compliance with compression therapy to prevent regression. Patients considering this technique should undergo pre-operative conservative management with no pitting edema ( 34 ). It has been shown to be effective both in the upper and lower extremity, although it is more effective in the upper extremity. It is known that adipose tissue functions as a crucial organ and a cytokine-activated cell in LE ( 36 ). The removal of adipose tissue using liposuction does not affect the already decreased lymph transport system in LE ( 34 ). Moreover, a significant improvement was detected in skin blood flow and quality of life after liposuction ( 37 , 38 ). Its complications include infection, skin necrosis and recurrence.

PHYSIOLOGIC TECHNIQUES

Lymphatic venous anastomosis, lymphatico-lymphatic by-pass, and lymph node transfer can be listed as physiologic methods. Many of these methods use recent developments in technology to assist in identifying lymphatic channels and lymph nodes ( 2 , 39 ).

Lymphatic Venous Anastomosis (LVA) or Bypass

LVA was first described in an animal model with several human studies to follow ( 40 , 35 ). This technique involves the creation of connections between the lymphatic system and the venous system in the distal or proximal extremity. Superficial or deep lymphatics are anastomosed with neighboring veins. Fluorescence is used to help identify the lymphatic system and an operating microscope is used to assist in microsurgery ( 41 ). Single or multiple LVA’s have been reported by different authors using differing surgical sites ( 39 , 42 – 44 ). Supermicrosurgery (anastomosis less than 0.8 mm vessels) is used in this technique, in which lymphatic vessels and adjacent venules are anastomosed, mostly in an end-to-end fashion ( 39 , 43 ) ( Figure 5 ). Variations on the configuration of anastomosis type were described in several studies with variable success rates ( 45 , 46 ). Studies have reported improvement in patients both subjectively and objectively. In general, LVA’s have been shown to be a safe technique for the management of LE ( 39 , 43 ).

An external file that holds a picture, illustration, etc.
Object name is tjs-33-2-51-g05.jpg

Lymphovenous anastomosis. Please note supermicrosurgery is used to establish such an anastomosis

Vascularized Lymph Node Transfer

Vascularized lymph node transfer was first introduced in animal models ( 47 ). It has recently been applied to humans with gaining popularity. There are different options for lymph node transfer, namely the location of the donor and the recipient sites. Options for lymph node harvest include the lateral thoracic region, groin, submental region, supraclavicular region and intraabdominal lymph nodes ( 44 , 48 , 49 ). Each donor site has its particular anatomic advantages and disadvantages, and contains varying number of lymph nodes ranging from 1–10. The lymph nodes can be harvested together with a portion of the skin if necessary. These operations require microsurgical skills to perform an arterial and venous anastomosis to provide blood supply to the transferred tissue. The results of lymph node transfer are quite promising and have been shown to provide both objective and subjective improvements ( 35 ).

One consideration for lymph node transfer is the concern for possible LE at the donor site ( 50 ). Reverse lymph node mapping, originally described as a technique to refine axillary dissection, can be used to minimize lymph node harvest related morbidity ( 51 ). It allows differential identification of nodes which drain the neighboring extremity in addition to the ones that are included in the tissue to be removed. Reverse lymph node mapping involves the use of the photodynamic eye, Technetium and ICG dye. Using this technique, the surgeon can be reassured that no lymph nodes are removed that drain the extremity ( 51 ). Clinically, the benefit of lymph node transfer is to restore immunologic function to the extremity and improve fluid drainage. However, the action mechanism of lymph node transfers is poorly understood. The transferred nodes have been shown to be active in a number of studies. One proposed mechanism suggests that the new lymph nodes act as “pumps” which filter the surrounding fluid ( 52 ). The best site for lymph node implantation is currently unknown. In some cases, the nodes have been placed distally whereas in others they were implanted proximally ( 52 ). Well-designed controlled prospective studies are needed to clarify if the suggested functional surgical methods are beneficial in the long-term.

A variety of lymph node transfers includes a tissue portion to be transferred with lymph nodes. Such examples include transferring the lower abdominal tissue in case of total breast anatomical reconstruction (TBAR) and latissimus dorsi flaps with lymph nodes. In such cases, both the breast and the lymph nodes are reconstructed ( 44 , 50 , 53 ). Other types of flaps such as those harvested intraabdominally carry lymphatic tissue from the omentum ( 49 ). These can be harvested either by open surgery or laparoscopically.

Intraoperative Considerations

The nodal status defines one of the most important prognostic factors in breast cancer. However, although necessary, axillary dissection may compromise the lymphatic system thus contributing to the development of LE.

Recently, the lymphatic drainage of the arm and breast tissue were studied and it was found that preserving arm lymphatics during SLNB and/or ALND via a new concept called axillary reverse mapping (ARM) may reduce the risk of BCRL ( 4 , 54 , 55 ). ARM is based on the hypothesis that drainage of arm lymphatics differs from that of breast lymphatics ( 4 , 54 ). However, it was shown that lymphatic interconnections exist in the axilla between arm and breast lymphatics ( 56 ). The technique of mapping the arm and breast lymphatics is comprised of radioisotope injection to the breast and blue dye injection to the upper arm ( Figure 6 ). The lymphatic pathways and interconnections are determined. When a crossover is identified, blue nodes should be removed ( 55 ). ARM reduced BCRL when compared with conventional breast cancer surgeries, nevertheless, randomized controlled studies are needed ( 55 ).

An external file that holds a picture, illustration, etc.
Object name is tjs-33-2-51-g06.jpg

The mapping of axillary lymphatics. A radioisotope is injected into the breast, whereas blue-dye is administered subcutaneously to the upper arm. The lymphatic flow of the breast and arm is separately revealed

Breast Cancer Related Lymphedema is a devastating disease affecting millions of women. Its treatment is aimed at curing the disease and reducing recurrence rate. However, treatment methods create both physical and psychological morbidity to the patients. BCRL influences daily activities and affects patient self-esteem in various ways. Modern surgical and non-surgical techniques offer numerous methods for the patients to overcome BCRL. In the future, we hope to ensure 100% success in the control or elimination of BCRL. Until then, the exact biology, pathogenesis of lymphatic system diseases and the treatment options require further research to be able to understand this devastating disease.

Peer-review: Externally peer-reviewed.

Author Contributions: Concept - O.K., C.D.L.A., K.T., A.T.; Design - O.K., C.D.L.A., K.T., A.T.; Supervision - O.K., C.D.L.A., K.T., A.T.; Resource - O.K., C.D.L.A.,, K.T., A.T.; Materials - O.K., C.D.L.A., K.T., A.T.; Data Collection and/or Processing - O.K., C.D.L.A., K.T., A.T.; Analysis and/or Interpretation - O.K., C.D.L.A., K.T., A.T.; Literature Search - O.K., C.D.L.A., K.T., A.T.; Writing Manuscript - O.K., C.D.L.A., K.T., A.T.; Critical Reviews - O.K., C.D.L.A., K.T., A.T.

Conflict of Interest: No conflict of interest was declared by the authors.

Financial Disclosure: The authors declared that this study has received no financial support.

Health | New treatment could be “game-changing tool” in…

Share this:.

  • Click to share on Facebook (Opens in new window)
  • Click to share on Reddit (Opens in new window)
  • Click to share on Twitter (Opens in new window)

Digital Replica Edition

  • Latest Headlines
  • Environment
  • Transportation
  • News Obituaries

Health | New treatment could be “game-changing tool” in fight against Alzheimer’s, CSU research finds

Combination of drugs targets two brain proteins critical in neuroinflammation involved in brain aging and alzheimer’s.

Elizabeth Hernandez - Staff portraits in The Denver Post studio on October 5, 2022. (Photo by Eric Lutzens/The Denver Post)

The combination of drugs targets two brain proteins critical in neuroinflammation, which is involved in brain aging and Alzheimer’s, according to a study published in July in the Journal of Neuroinflammation featuring CSU researchers.

Results from the study show this medicine could become “a game-changing tool” against Alzheimer’s, researchers said.

“There are no effective treatments right now,” said Devin Wahl, a CSU postdoctoral fellow, who co-authored the study. “We have treatments that can manage symptoms, but we don’t have any that can stop the disease. We want to try to identify novel treatments that may be effective to slow, or even reduce, the effects of Alzheimer’s disease.”

This cocktail of medicines could also improve memory in aging adults, the study found, and, potentially, reverse cognitive decline.

The research came out of a partnership between CSU faculty member Tom LaRocca’s Healthspan Biology Lab and Colorado-based biotech company Sachi Bio.

“This is a novel and effective treatment to improve memory in mice,” said Prashant Nagpal, who co-founded Sachi Bio with his wife, Anushree Chatterjee. “A very important finding that we saw in this study is that you can reverse some cognitive decline. We are hoping to take this to human clinical trials next year.”

The mice behavioral tests measured memory and grip strength because grip strength and muscle function are closely linked to brain function, researchers said.

“If we can target what comes before Alzheimer’s disease, which is what this drug is meant to do, that will give people more treatment options, especially earlier in life,” Wahl said.

By next year or 2026, Nagpal hopes there will be a more conclusive data set including human trials.

“We’ve all been touched by seeing older parents and family members just being a shadow of themselves,” Nagpal said. “It’s just heartbreaking. It may seem like just a glimmer of hope, but can you latch onto it and just, you know, go for it?”

Sign up for our weekly newsletter to get health news sent straight to your inbox.

  • Report an Error
  • Submit a News Tip

More in Health

By MONIKA PRONCZUK DAKAR, Senegal (AP) — The Africa Center for Disease Control and Prevention and the World Health Organization launched on Friday a continent-wide response plan to the outbreak of mpox, three weeks after WHO declared outbreaks in 12 African countries a global emergency. The estimated budget for the six-month plan is almost $600 […]

WHO and Africa CDC launch a response plan to the mpox outbreak

Exterior of the Center for Disease ...

Environment | Colorado gets $225,000 from CDC to measure lead, PFA exposure

Parents and neighbors are concerned about how increased pollutants would impact people's health, especially schoolchildren who play outside at Dupont Elementary School.

Environment | Adams 14 district, parents at Dupont Elementary plan to fight gasoline storage expansion near school

Recipients can receive up to $20 in matching funds per transaction, for a maximum of $60 per month.

Health | Coloradans using food assistance to buy produce can receive bonus matching funds

  • News & Media
  • Chemical Biology
  • Computational Biology
  • Ecosystem Science
  • Cancer Biology
  • Exposure Science & Pathogen Biology
  • Metabolic Inflammatory Diseases
  • Advanced Metabolomics
  • Mass Spectrometry-Based Measurement Technologies
  • Spatial and Single-Cell Proteomics
  • Structural Biology
  • Biofuels & Bioproducts
  • Human Microbiome
  • Soil Microbiome
  • Synthetic Biology
  • Computational Chemistry
  • Chemical Separations
  • Chemical Physics
  • Atmospheric Aerosols
  • Human-Earth System Interactions
  • Modeling Earth Systems
  • Coastal Science
  • Plant Science
  • Subsurface Science
  • Terrestrial Aquatics
  • Materials in Extreme Environments
  • Precision Materials by Design
  • Science of Interfaces
  • Friction Stir Welding & Processing
  • Dark Matter
  • Flavor Physics
  • Fusion Energy Science
  • Neutrino Physics
  • Quantum Information Sciences
  • Emergency Response
  • AGM Program
  • Tools and Capabilities
  • Grid Architecture
  • Grid Cybersecurity
  • Grid Energy Storage
  • Earth System Modeling
  • Energy System Modeling
  • Transmission
  • Distribution
  • Appliance and Equipment Standards
  • Building Energy Codes
  • Advanced Building Controls
  • Advanced Lighting
  • Building-Grid Integration
  • Building and Grid Modeling
  • Commercial Buildings
  • Federal Performance Optimization
  • Resilience and Security
  • Grid Resilience and Decarbonization
  • Building America Solution Center
  • Energy Efficient Technology Integration
  • Home Energy Score
  • Electrochemical Energy Storage
  • Flexible Loads and Generation
  • Grid Integration, Controls, and Architecture
  • Regulation, Policy, and Valuation
  • Science Supporting Energy Storage
  • Chemical Energy Storage
  • Waste Processing
  • Radiation Measurement
  • Environmental Remediation
  • Subsurface Energy Systems
  • Carbon Capture
  • Carbon Storage
  • Carbon Utilization
  • Advanced Hydrocarbon Conversion
  • Fuel Cycle Research
  • Advanced Reactors
  • Reactor Operations
  • Reactor Licensing
  • Solar Energy
  • Wind Resource Characterization
  • Wildlife and Wind
  • Community Values and Ocean Co-Use
  • Wind Systems Integration
  • Wind Data Management
  • Distributed Wind
  • Energy Equity & Health
  • Environmental Monitoring for Marine Energy
  • Marine Biofouling and Corrosion
  • Marine Energy Resource Characterization
  • Testing for Marine Energy
  • The Blue Economy
  • Environmental Performance of Hydropower
  • Hydropower Cybersecurity and Digitalization
  • Hydropower and the Electric Grid
  • Materials Science for Hydropower
  • Pumped Storage Hydropower
  • Water + Hydropower Planning
  • Grid Integration of Renewable Energy
  • Geothermal Energy
  • Algal Biofuels
  • Aviation Biofuels
  • Waste-to-Energy and Products
  • Hydrogen & Fuel Cells
  • Emission Control
  • Energy-Efficient Mobility Systems
  • Lightweight Materials
  • Vehicle Electrification
  • Vehicle Grid Integration
  • Contraband Detection
  • Pathogen Science & Detection
  • Explosives Detection
  • Threat-Agnostic Biodefense
  • Discovery and Insight
  • Proactive Defense
  • Trusted Systems
  • Nuclear Material Science
  • Radiological & Nuclear Detection
  • Nuclear Forensics
  • Ultra-Sensitive Nuclear Measurements
  • Nuclear Explosion Monitoring
  • Global Nuclear & Radiological Security
  • Disaster Recovery
  • Global Collaborations
  • Legislative and Regulatory Analysis
  • Technical Training
  • Additive Manufacturing
  • Deployed Technologies
  • Rapid Prototyping
  • Systems Engineering
  • 5G Security
  • RF Signal Detection & Exploitation
  • Climate Security
  • Internet of Things
  • Maritime Security
  • Millimeter Wave
  • Artificial Intelligence
  • Graph and Data Analytics
  • Software Engineering
  • Computational Mathematics & Statistics
  • High-Performance Computing
  • Adaptive Autonomous Systems
  • Visual Analytics
  • Lab Objectives
  • Publications & Reports
  • Featured Research
  • Diversity, Equity, Inclusion & Accessibility
  • Lab Leadership
  • Lab Fellows
  • Staff Accomplishments
  • Undergraduate Students
  • Graduate Students
  • Post-graduate Students
  • University Faculty
  • University Partnerships
  • K-12 Educators and Students
  • STEM Workforce Development
  • STEM Outreach
  • Meet the Team
  • Internships
  • Regional Impact
  • Philanthropy
  • Volunteering
  • Available Technologies
  • Industry Partnerships
  • Licensing & Technology Transfer
  • Entrepreneurial Leave
  • Visual Intellectual Property Search (VIPS)
  • Atmospheric Radiation Measurement User Facility
  • Electricity Infrastructure Operations Center
  • Energy Sciences Center
  • Environmental Molecular Sciences Laboratory
  • Grid Storage Launchpad
  • Institute for Integrated Catalysis
  • Interdiction Technology and Integration Laboratory
  • PNNL Portland Research Center
  • PNNL Seattle Research Center
  • PNNL-Sequim (Marine and Coastal Research)
  • Radiochemical Processing Laboratory
  • Shallow Underground Laboratory

Research Vessel Resilience Charts Course to the Future of Marine Research

Dedication of the new hybrid-electric vessel was celebrated by laboratory, state, tribal and federal officials 

The RV Resilience vessel drives by PNNL Sequim campus.

RV Resilience, the Department of Energy’s new hybrid electric marine research vessel. 

(Photo by Eric Francavilla | Pacific Northwest National Laboratory)

SEQUIM, Wash.—Officials gathered at the Sequim campus of the Department of Energy’s Pacific Northwest National Laboratory today to dedicate DOE’s first hybrid-electric research vessel, RV Resilience.

The event marks the start of a new era of marine energy research at  PNNL-Sequim , part of DOE’s Office of Science national laboratory system and Resilience’s new home port. Speakers at the dedication included U.S. Sen. Maria Cantwell, U.S. Rep. Derek Kilmer, Washington State Rep. Steve Tharinger and representatives from DOE and PNNL.

“DOE is focused on clean energy solutions. The RV Resilience enables us to accelerate the development and deployment of novel marine energy technologies from testing at the bench scale to early demonstration under real ocean operating conditions,” said Geri Richmond, DOE’s undersecretary for science and innovation. “Demand for these technologies is likely to increase in the coming years, unlocking opportunities for ocean science and maritime industries equipped to explore new applications for marine energy research that will help power the blue economy.”

Richmond’s chief of staff, Ariel Marshall, spoke on her behalf at the dedication ceremony.

PNNL Director Steve Ashby speaks at podium to dedicate the RV Resilience.

The 50-foot research vessel will allow researchers to transport and install large equipment in Sequim Bay, such as demonstration-scale marine energy devices. These devices will help accelerate marine energy testing and support new partnerships with industry developers. In addition to reducing carbon emissions, the hybrid-electric vessel is nearly silent when operated in fully electric mode. This minimizes noise pollution for marine wildlife and enables more sensitive acoustic measurements during research operations. 

“RV Resilience represents DOE’s and PNNL’s commitment to demonstrating how innovative approaches, like the design and construction of this unique hybrid vessel, can advance the nation’s quest for clean energy,” said Laboratory Director Steve Ashby.

The RV Resilience can operate on diesel engines or in a completely electric mode using onboard battery banks. These batteries can be charged with the diesel engines, at any marina or through a rapid charging station at the PNNL-Sequim dock.

The RV Resilience was made possible with support from DOE’s Office of Energy Efficiency and Renewable Energy and its Water Power Technologies Office. It will be managed and operated by researchers at PNNL-Sequim—a regional hub for marine energy research, development and testing—and enables research operations in nearby Sequim Bay. 

“We’re exploring the potential of marine energy by conducting world-leading coastal and oceanographic science and research,” said Alejandro Moreno, associate principal deputy assistant secretary for EERE. “This new hybrid research vessel enables that work with fewer emissions and less impact on the ocean’s wildlife."

The RV Resilience is the latest in a series of investments that will grow PNNL’s capabilities in marine technology research to continue advancing renewable energy, climate resilience and national security. These planned investments include a pre-permitted marine testing site, an underwater cabled array connecting at-sea devices to shore facilities and an onshore microgrid and battery storage system. 

Combined with the RV Resilience and a host of new onshore laboratory facilities, these capabilities will enable PNNL-Sequim to support DOE’s marine energy mission, including supporting the development of offshore wind and tidal energy, as well as marine carbon dioxide removal. 

RV Resilience was built by Snow & Company in Seattle, Wash.  To learn more about vessel specifications, visit PNNL’s website . 

Pacific Northwest National Laboratory draws on its distinguishing strengths in chemistry , Earth sciences , biology and data science to advance scientific knowledge and address challenges in sustainable energy and national security . Founded in 1965, PNNL is operated by Battelle for the Department of Energy’s Office of Science, which is the single largest supporter of basic research in the physical sciences in the United States. DOE’s Office of Science is working to address some of the most pressing challenges of our time. For more information, visit https://www.energy.gov/science/ . For more information on PNNL, visit PNNL's News Center . Follow us on Twitter , Facebook , LinkedIn and Instagram .

Published: September 5, 2024

Research topics

Wellesley team’s new research on anesthesia unlocks important clues about the nature of consciousness

From left: Professor Mike Wiest and Wellesley College students Jess Lopes, Emeline Gaunce, and Derin Timuçin.

  • Research and innovation
  • Share to Facebook
  • Share to Twitter

For decades, one of the most fundamental and vexing questions in neuroscience has been: what is the physical basis of consciousness in the brain? Most researchers favor classical models, based on classical physics, while a minority have argued that consciousness must be quantum in nature, and that its brain basis is a collective quantum vibration of “microtubule” proteins inside neurons.

New research by Wellesley College professor Mike Wiest and a group of Wellesley College undergraduate students has yielded important experimental results relevant to this debate, by examining how anesthesia affects the brain. Wiest and his research team found that when they gave rats a drug that binds to microtubules, it took the rats significantly longer to fall unconscious under an anesthetic gas. The research team’s microtubule-binding drug interfered with the anesthetic action, thus supporting the idea that the anesthetic acts on microtubules to cause unconsciousness.

“Since we don’t know of another (i.e,. classical) way that anesthetic binding to microtubules would generally reduce brain activity and cause unconsciousness,” Wiest says, “this finding supports the quantum model of consciousness.”

It’s hard to overstate the significance of the classical/quantum debate about consciousness, says Wiest, an associate professor of neuroscience at Wellesley. “When it becomes accepted that the mind is a quantum phenomenon, we will have entered a new era in our understanding of what we are,” he says. The new approach “would lead to improved understanding of how anesthesia works, and it would shape our thinking about a wide variety of related questions, such as whether coma patients or non-human animals are conscious, how mysterious drugs like lithium modulate conscious experience to stabilize mood, how diseases like Alzheimer’s or schizophrenia affect perception and memory, and so on.”

More broadly, a quantum understanding of consciousness “gives us a world picture in which we can be connected to the universe in a more natural and holistic way,” Wiest says. Wiest plans to pursue future research in this field, and hopes to explain and explore the quantum consciousness theory in a book for a general audience.

Wellesley students who co-authored the paper with Wiest are Sana Khan ’25, Yixiang Huang ’25, Derin Timucin ’27, Shantelle Bailey ’24, Sophia Lee ’23, Jessica Lopes ’26, Emeline Gaunce ’26, Jasmine Mosberger ’25, Michelle Zhan ’24, Bothina Abdelrahman ’26 and Xiran Zeng ’27. Published September 1 in eNeuro , the Wellesley study demonstrates that anesthesia works by binding to microtubules inside neurons, thus providing important evidence for a quantum theory of consciousness while reviving a focus on microtubules in anesthesia.

eNeuro is the Society for Neuroscience's open-access journal.

IMAGES

  1. Ongoing Research Aims to Develop Standards for Lymphedema Therapy

    new research on lymphedema

  2. Before & After Results

    new research on lymphedema

  3. Understanding Lymphedema

    new research on lymphedema

  4. Lymphedema Treatment Act

    new research on lymphedema

  5. About Lymphedema

    new research on lymphedema

  6. Lymphedema

    new research on lymphedema

VIDEO

  1. Research Investigating New Treatments to Managing Lymphoedema

  2. Lymphedema Treatment after Breast Cancer

  3. Celebrate the 2nd annual World Lymphedema Day

  4. Future of Lymphedema Treatment Unveiled!

  5. Chronic Disease " Lymphedema "

  6. Lower Extremity Lymphedema & Nonmelanoma Skin Cancers

COMMENTS

  1. A promising new treatment for lymphedema

    Research by Antonio Forte, M.D., Ph.D., a Mayo Clinic plastic surgeon, seeks to better pinpoint who is most likely to benefit from surgery aimed at regenerating a faulty lymphatic system. "Without surgery, lymphedema gets progressively worse due to accumulation of the lymphatic fluid in tissue and the chronic changes that causes.

  2. Advances in Lymphedema

    Lymphedema is a common, complex, and inexplicably underappreciated human disease. Despite a history of relative neglect by health care providers and by governmental health care agencies, the last decade has seen an explosive growth of insights into, and approaches to, the problem of human lymphedema. The current review highlights the significant advances that have occurred in the investigative ...

  3. Lymph vessels on a chip reveal possible new drug target for lymphedema

    By Eva Frederick. Scientists have identified a new potential drug target for lymphedema, a painful swelling condition often caused by surgery or radiation treatments. SPL/Science Source. WASHINGTON, D.C. — There's no real cure for lymphedema, a painful accumulation of fluid in the arms and legs that afflicts more than 150 million people ...

  4. Advances in lymphedema: An under-recognized disease with a hopeful

    Increased investigation has demonstrated the key role of inflammation and resultant fibrosis and adipose deposition leading to the clinical sequelae and associated reduction in quality of life with lymphedema. New imaging techniques including magnetic resonance imaging (MRI), indocyanine green lymphography, and high-frequency ultrasound offer ...

  5. Study identifies potential drug target for lymphedema

    New research identifies key protein complex in cellular quality control; The researchers introduced inflammatory cytokines that are known to be expressed in lymphedema patients, such as ...

  6. The Future of Lymphedema: Potential Therapeutic Targets for Treatment

    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 ...

  7. The Future of Lymphedema: Potential Therapeutic Targets for ...

    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 ...

  8. Advances and challenges in treating lymphedema: A Q and A with Dhruv

    Lymphedema is a potentially debilitating condition affecting more than 1 million people in the U.S. each year. In research funded in part by the National Heart, Lung, and Blood Institute (NHLBI), Dhruv Singhal, M.D., director of Lymphatic Surgery at Beth Israel Deaconess Medical Center in Boston, seeks to understand the causes of lymphedema and what puts some people at higher risk for getting it.

  9. Nanotechnology could treat lymphedema

    Nanotechnology could treat lymphedema. When lymphatic vessels fail, typically their ability to pump out the fluid is compromised. Researchers have now developed a new treatment using nanoparticles ...

  10. Primary lymphoedema

    Taija Mäkinen. Nature Cardiovascular Research (2023) Lymphoedema is the swelling of one or several parts of the body owing to lymph accumulation in the extracellular space. It is often chronic ...

  11. Avoiding the Swell: Advances in Lymphedema Prevention, Detection, and

    Lymphedema is a feared complication of cancer treatments that negatively affects survivors' quality of life. The true incidence of lymphedema is difficult to determine given its long latency period. As the number of survivors of cancer is increasing in the United States, lymphedema is poised to be a major health concern. The most noteworthy risk factor for lymphedema is comprehensive lymph ...

  12. Cell therapy as a treatment of secondary lymphedema: a systematic

    Ten studies were included to investigate the effect of the cell therapy treatment on the lymphatic regeneration in secondary lymphedema. The overall pooled analysis showed a significant increase in lymphatic vessel density in experimental group versus control group (SMD 6.35; 95% CI 4.115, 8.581; p = 0.00).

  13. Study finds first possible drug treatment for lymphedema

    Based on the research, bestatin (also known as ubenimex), is being tested in a clinical trial that started in May 2016 — known as ULTRA — as a treatment for secondary lymphedema, which occurs because of damage to the lymphatic system from surgery, radiation therapy, trauma or infection. Primary lymphedema, on the other hand, is hereditary.

  14. Cell therapy as a treatment of secondary lymphedema: a systematic

    Lymphedema, the accumulation of interstitial fluid caused by poor lymphatic drainage, is a progressive and permanent disease with no curative treatment. Several studies have evaluated cell-based therapies in secondary lymphedema, but no meta-analysis has been performed to assess their efficacy. We conducted a systematic review and meta-analysis of all available preclinical and clinical studies ...

  15. The Future of Lymphedema: Potential Therapeutic Targets for ...

    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 ...

  16. Koya's NPCD device proves more effective than APCDs in lymphedema

    Data from the TEAYS study (NCT05507346), published in the Journal of Vascular Surgery, found that Dayspring outperformed advanced pneumatic compression device (APCD) - the standard of care for treating lymphedema - in several areas.Patients using Dayspring saw a significantly greater reduction in limb volume (369.9mL versus 83.1mL) and experienced improved quality of life, with a mean ...

  17. Anti-inflammatory drug effective for treating lymphedema symptoms

    This new treatment doesn't cure lymphedema, but our studies show it has the capacity to make the illness more livable, more workable." ... Further research in mice showed that using ketoprofen to target LTB4 induced lymphatic repair and reversed the disease processes. This indicated that perhaps other therapies could reverse the negative ...

  18. Overview of Lymphedema for Physicians and Other Clinicians: A Review of

    Lymphedema has historically been underrated in clinical practice, education, and scholarship to the detriment of many patients with this chronic, debilitating condition. The mechanical insufficiency of the lymphatic system causes the abnormal accumulation of protein-rich fluid in the interstitium, which triggers a cascade of adverse consequences such as fat deposition and fibrosis. As the ...

  19. Mayo Clinic study suggests lower extremity lymphedema is a risk factor

    ROCHESTER, Minn. — New research from Mayo Clinic finds that patients with lymphedema —swelling of the limbs caused by the accumulation of protein-rich fluids of the immune system — had twice the risk of developing skin cancer than patients without lymphedema.. Patients with lymphedema had a significantly higher frequency of developing basal cell carcinomas and squamous cell carcinomas on ...

  20. Lymphedema: New Concepts in Diagnosis and Treatment

    Purpose of Review This review's aim is to coalesce current evidence regarding the lymphatic system and lymphedema, bridge the gap from traditional approaches to emerging concepts, and present simple strategies that can be immediately implemented into everyday clinical practice. Recent Findings New evidence of the microcirculation redefines all edema as lymphedema and having the potential to ...

  21. Current diagnostic and quantitative techniques in the field of

    Lymphedema evaluation entails multifaceted considerations for which options continue to evolve and emerge. This paper provides a critical review of the current status of diagnostic and quantitative measures for lymphedema, from traditional and novel bedside assessment tools for volumetric and fluid assessment, to advanced imaging modalities. Modalities are contrasted with regard to empirical ...

  22. Biomarker for lipedema, other lymphatic diseases discovered

    November 23, 2020 - By Tracie White. Stanley Rockson co-authored a paper that identified a biomarker for lipedema, a little understood fat disorder that is often confused with obesity. Steve Fisch. Throughout her life, Kristy has been told she was obese and needed to lose weight. She was teased and tormented for looking fat.

  23. Lymphedema: From diagnosis to treatment

    Abstract. Lymphedema is a chronic and progressive disorder resulting from impaired lymphatic system function. In developed countries, upper extremity lymphedema is mainly the consequence of breast cancer surgery in which axillary lymph node dissection and radiation alter upper extremity lymphatic flow. Diagnosis of lymphedema is made clinically.

  24. Living with Lymphedema and Lymphatic Disease

    154 West 14th Street, 2nd Floor, New York, NY 10011 The Lymphatic Education & Research Network is a tax-exempt 501(c)3 nonprofit organization. Our Identification ...

  25. Lymphedema after Breast Cancer Treatment

    Breast cancer-related lymphedema is the most common form of lymphedema in the United States and other developed nations. •. Axillary lymph-node dissection and adjuvant radiation therapy are ...

  26. Alzheimer's: New CSU treatment could be "game-changing tool"

    New treatment could be "game-changing tool" in fight against Alzheimer's, CSU research finds Combination of drugs targets two brain proteins critical in neuroinflammation involved in brain ...

  27. Research Vessel Resilience Charts Course to the Future of Marine

    "This new hybrid research vessel enables that work with fewer emissions and less impact on the ocean's wildlife." The RV Resilience is the latest in a series of investments that will grow PNNL's capabilities in marine technology research to continue advancing renewable energy, climate resilience and national security. ...

  28. Funding awarded for research on bagworms, bubonic plague, ancient

    Bloch, Porto and their colleagues will develop and use new artificial intelligence software to analyze these fossils and search for hidden patterns. They'll use these data to determine how functional diversity — which measures the range of ways organisms use resources in their environment — was altered in mammal communities during the ...

  29. Wellesley

    New research by Wellesley College professor Mike Wiest and a group of Wellesley College undergraduate students has yielded important experimental results relevant to this debate, by examining how anesthesia affects the brain. Wiest and his research team found that when they gave rats a drug that binds to microtubules, it took the rats ...