J Reconstr Microsurg 2021; 37(06): 465-474
DOI: 10.1055/s-0040-1722760
Review Article

Lymphatic Tissue Engineering: A Further Step for Successful Lymphedema Treatment

Andreas Spörlein
1   Department of Hand, Plastic, and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Germany
,
Patrick A. Will
1   Department of Hand, Plastic, and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Germany
,
Katja Kilian
1   Department of Hand, Plastic, and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Germany
,
Emre Gazyakan
1   Department of Hand, Plastic, and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Germany
,
Justin M. Sacks
2   Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University, St. Louis, Missouri
,
Ulrich Kneser
1   Department of Hand, Plastic, and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Germany
,
Christoph Hirche
1   Department of Hand, Plastic, and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Germany
3   Department of Plastic, Hand and Reconstructive Microsurgery, BG Trauma Center Frankfurt, Goethe University Frankfurt, Germany
› Institutsangaben

Abstract

Background Secondary lymphedema, caused by oncologic surgery, radiation, and chemotherapy, is one of the most relevant, nononcological complications affecting cancer survivors. Severe functional deficits can result in impairing quality of life and a societal burden related to increased treatment costs. Often, conservative treatments are not sufficient to alleviate lymphedema or to prevent stage progression of the disease, as they do not address the underlying etiology that is the disruption of lymphatic pathways. In recent years, lymphatic surgery approaches were revolutionized by advances in microsurgical technique. Currently, lymphedema can effectively be treated by procedures such as lymphovenous anastomosis (LVA) and lymph node transfer (LNT). However, not all patients have suitable lymphatic vessels, and lymph node harvesting is associated with risks. In addition, some data have revealed nonresponders to the microsurgical techniques.

Methods A literature review was performed to evaluate the value of lymphatic tissue engineering for plastic surgeons and to give an overview of the achievements, challenges, and goals of the field.

Results While certain challenges exist, including cell harvesting, nutrient supply, biocompatibility, and hydrostatic properties, it is possible and desirable to engineer lymph nodes and lymphatic vessels. The path toward clinical translation is considered more complex for LNTs secondary to the complex microarchitecture and pending final mechanistic clarification, while LVA is more straight forward.

Conclusion Lymphatic tissue engineering has the potential to be the next step for microsurgical treatment of secondary lymphedema. Current and future researches are necessary to optimize this clinical paradigm shift for improved surgical treatment of lymphedema.



Publikationsverlauf

Eingereicht: 16. Juni 2020

Angenommen: 19. November 2020

Artikel online veröffentlicht:
31. Januar 2021

© 2021. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Beaulac SM, McNair LA, Scott TE, LaMorte WW, Kavanah MT. Lymphedema and quality of life in survivors of early-stage breast cancer. Arch Surg 2002; 137 (11) 1253-1257
  • 2 Shih Y-CT, Xu Y, Cormier JN. et al. Incidence, treatment costs, and complications of lymphedema after breast cancer among women of working age: a 2-year follow-up study. J Clin Oncol 2009; 27 (12) 2007-2014
  • 3 Ridner SH, Deng J, Fu MR. et al. Symptom burden and infection occurrence among individuals with extremity lymphedema. Lymphology 2012; 45 (03) 113-123
  • 4 Rockson SG, Rivera KK. Estimating the population burden of lymphedema. Ann N Y Acad Sci 2008; 1131: 147-154
  • 5 Zou L, Liu FH, Shen PP. et al. The incidence and risk factors of related lymphedema for breast cancer survivors post-operation: a 2-year follow-up prospective cohort study. Breast Cancer 2018; 25 (03) 309-314
  • 6 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 (06) 500-515
  • 7 Hirche C, Engel H, Seidenstuecker K. et al. Lympho-reconstructive microsurgery for secondary lymphedema: Consensus of the German-Speaking Society for Microsurgery of Peripheral Nerves and Vessels (DAM) on indication, diagnostic and therapy by lymphovenous anastomosis (LVA) and vascularized lymph node transfer (VLNT). Handchir Mikrochir Plast Chir 2019; 51 (06) 424-433
  • 8 Deng J, Ridner SH, Dietrich MS. et al. Prevalence of secondary lymphedema in patients with head and neck cancer. J Pain Symptom Manage 2012; 43 (02) 244-252
  • 9 Ridner SH, Dietrich MS, Niermann K, Cmelak A, Mannion K, Murphy B. A prospective study of the lymphedema and fibrosis continuum in patients with head and neck cancer. Lymphat Res Biol 2016; 14 (04) 198-205
  • 10 Smith BG, Hutcheson KA, Little LG. et al. Lymphedema outcomes in patients with head and neck cancer. Otolaryngol Head Neck Surg 2015; 152 (02) 284-291
  • 11 Cormier JN, Askew RL, Mungovan KS, Xing Y, Ross MI, Armer JM. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer 2010; 116 (22) 5138-5149
  • 12 Hayes SC, Janda M, Ward LC. et al. Lymphedema following gynecological cancer: Results from a prospective, longitudinal cohort study on prevalence, incidence and risk factors. Gynecol Oncol 2017; 146 (03) 623-629
  • 13 Mehrara BJ, Greene AK. Lymphedema and obesity: is there a link?. Plast Reconstr Surg 2014; 134 (01) 154e-160e
  • 14 Schmitz KH, Ahmed RL, Troxel A. et al. Weight lifting in women with breast-cancer-related lymphedema. N Engl J Med 2009; 361 (07) 664-673
  • 15 Andersen L, Højris I, Erlandsen M, Andersen J. Treatment of breast-cancer-related lymphedema with or without manual lymphatic drainage--a randomized study. Acta Oncol 2000; 39 (03) 399-405
  • 16 Kung TA, Champaneria MC, Maki JH, Neligan PC. Current concepts in the surgical management of lymphedema. Plast Reconstr Surg 2017; 139 (04) 1003e-1013e
  • 17 Dellon AL, Hoopes JE. The Charles procedure for primary lymphedema. Long-term clinical results. Plast Reconstr Surg 1977; 60 (04) 589-595
  • 18 Brorson H. Liposuction in lymphedema treatment. J Reconstr Microsurg 2016; 32 (01) 56-65
  • 19 Basta MN, Gao LL, Wu LC. Operative treatment of peripheral lymphedema: a systematic meta-analysis of the efficacy and safety of lymphovenous microsurgery and tissue transplantation. Plast Reconstr Surg 2014; 133 (04) 905-913
  • 20 Markkula SP, Leung N, Allen VB, Furniss D. Surgical interventions for the prevention or treatment of lymphoedema after breast cancer treatment. Cochrane Database Syst Rev 2019; 2: CD011433
  • 21 Chang DW, Suami H, Skoracki R. A prospective analysis of 100 consecutive lymphovenous bypass cases for treatment of extremity lymphedema. Plast Reconstr Surg 2013; 132 (05) 1305-1314
  • 22 Yang JC-S, Wu S-C, Lin W-C, Chiang M-H, Chiang P-L, Hsieh C-H. Supermicrosurgical lymphaticovenous anastomosis as alternative treatment option for moderate-to-severe lower limb lymphedema. J Am Coll Surg 2020; 230 (02) 216-227
  • 23 Tourani SS, Taylor GI, Ashton MW. Long-term patency of lymphovenous anastomoses: a systematic review. Plast Reconstr Surg 2016; 138 (02) 492-498
  • 24 Dionyssiou D, Demiri E, Tsimponis A. et al. A randomized control study of treating secondary stage II breast cancer-related lymphoedema with free lymph node transfer. Breast Cancer Res Treat 2016; 156 (01) 73-79
  • 25 Scaglioni MF, Arvanitakis M, Chen Y-C, Giovanoli P, Chia-Shen Yang J, Chang EI. Comprehensive review of vascularized lymph node transfers for lymphedema: outcomes and complications. Microsurgery 2018; 38 (02) 222-229
  • 26 Cheng M-H, Chen S-C, Henry SL, Tan BK, Lin MC, Huang J-J. Vascularized groin lymph node flap transfer for postmastectomy upper limb lymphedema: flap anatomy, recipient sites, and outcomes. Plast Reconstr Surg 2013; 131 (06) 1286-1298
  • 27 Saaristo AM, Niemi TS, Viitanen TP, Tervala TV, Hartiala P, Suominen EA. Microvascular breast reconstruction and lymph node transfer for postmastectomy lymphedema patients. Ann Surg 2012; 255 (03) 468-473
  • 28 Pons G, Masia J, Loschi P, Nardulli ML, Duch J. A case of donor-site lymphoedema after lymph node-superficial circumflex iliac artery perforator flap transfer. J Plast Reconstr Aesthet Surg 2014; 67 (01) 119-123
  • 29 Coriddi M, Skoracki R, Eiferman D. Vascularized jejunal mesenteric lymph node transfer for treatment of extremity lymphedema. Microsurgery 2017; 37 (02) 177-178
  • 30 Di Taranto G, Chen S, Elia R. et al. Free gastroepiploic lymph nodes and omentum flap for treatment of lower limb ulcers in severe lymphedema: Killing two birds with one stone. J Surg Oncol 2019
  • 31 Baumeister RGH, Mayo W, Notohamiprodjo M, Wallmichrath J, Springer S, Frick A. Microsurgical lymphatic vessel transplantation. J Reconstr Microsurg 2016; 32 (01) 34-41
  • 32 Sabin FR. 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 1902; 1: 367-389
  • 33 Pepper MS, Skobe M. Lymphatic endothelium: morphological, molecular and functional properties. J Cell Biol 2003; 163 (02) 209-213
  • 34 Suami H, Scaglioni MF. Anatomy of the lymphatic system and the lymphosome concept with reference to lymphedema. Semin Plast Surg 2018; 32 (01) 5-11
  • 35 Podgrabinska S, Braun P, Velasco P, Kloos B, Pepper MS, Skobe M. Molecular characterization of lymphatic endothelial cells. Proc Natl Acad Sci U S A 2002; 99 (25) 16069-16074
  • 36 Hu X, Jiang Z, Liu N. A novel approach for harvesting lymphatic endothelial cells from human foreskin dermis. Lymphat Res Biol 2006; 4 (04) 191-198
  • 37 Jiang Z, Hu X, Kretlow JD, Liu N. Harvesting and cryopreservation of lymphatic endothelial cells for lymphatic tissue engineering. Cryobiology 2010; 60 (02) 177-183
  • 38 Marino D, Luginbühl J, Scola S, Meuli M, Reichmann E. Bioengineering dermo-epidermal skin grafts with blood and lymphatic capillaries. Sci Transl Med 2014; 6 (221) 221ra14
  • 39 Conrad C, Niess H, Huss R. et al. Multipotent mesenchymal stem cells acquire a lymphendothelial phenotype and enhance lymphatic regeneration in vivo. Circulation 2009; 119 (02) 281-289
  • 40 Yang Y, Yang J-T, Chen X-H. et al. Construction of tissue-engineered lymphatic vessel using human adipose derived stem cells differentiated lymphatic endothelial like cells and decellularized arterial scaffold: A preliminary study. Biotechnol Appl Biochem 2018; 65 (03) 428-434
  • 41 Tammela T, Alitalo K. Lymphangiogenesis: molecular mechanisms and future promise. Cell 2010; 140 (04) 460-476
  • 42 Kaipainen A, Korhonen J, Mustonen T. et al. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc Natl Acad Sci U S A 1995; 92 (08) 3566-3570
  • 43 Mellor RH, Hubert CE, Stanton AWB. et al. Lymphatic dysfunction, not aplasia, underlies Milroy disease. Microcirculation 2010; 17 (04) 281-296
  • 44 Tervala TV, Hartiala P, Tammela T. et al. Growth factor therapy and lymph node graft for lymphedema. J Surg Res 2015; 196 (01) 200-207
  • 45 Honkonen KM, Visuri MT, Tervala TV. et al. Lymph node transfer and perinodal lymphatic growth factor treatment for lymphedema. Ann Surg 2013; 257 (05) 961-967
  • 46 Visuri MT, Honkonen KM, Hartiala P. et al. VEGF-C and VEGF-C156S in the pro-lymphangiogenic growth factor therapy of lymphedema: a large animal study. Angiogenesis 2015; 18 (03) 313-326
  • 47 Yoon YS, Murayama T, Gravereaux E. et al. VEGF-C gene therapy augments postnatal lymphangiogenesis and ameliorates secondary lymphedema. J Clin Invest 2003; 111 (05) 717-725
  • 48 Skobe M, Hawighorst T, Jackson DG. et al. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat Med 2001; 7 (02) 192-198
  • 49 Kärpänen T, Heckman CA, Keskitalo S. et al. Functional interaction of VEGF-C and VEGF-D with neuropilin receptors. FASEB J 2006; 20 (09) 1462-1472
  • 50 Byzova TV, Goldman CK, Pampori N. et al. A mechanism for modulation of cellular responses to VEGF: activation of the integrins. Mol Cell 2000; 6 (04) 851-860
  • 51 Hartiala P, Saarikko AM. Lymphangiogenesis and lymphangiogenic growth factors. J Reconstr Microsurg 2016; 32 (01) 10-15
  • 52 Sonnenberg E, Meyer D, Weidner KM, Birchmeier C. Scatter factor/hepatocyte growth factor and its receptor, the c-met tyrosine kinase, can mediate a signal exchange between mesenchyme and epithelia during mouse development. J Cell Biol 1993; 123 (01) 223-235
  • 53 Gibot L, Galbraith T, Kloos B. et al. Cell-based approach for 3D reconstruction of lymphatic capillaries in vitro reveals distinct functions of HGF and VEGF-C in lymphangiogenesis. Biomaterials 2016; 78: 129-139
  • 54 Avraham T, Daluvoy S, Zampell J. et al. Blockade of transforming growth factor-β1 accelerates lymphatic regeneration during wound repair. Am J Pathol 2010; 177 (06) 3202-3214
  • 55 Johnson NC, Dillard ME, Baluk P. et al. Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity. Genes Dev 2008; 22 (23) 3282-3291
  • 56 Petrova TV, Karpanen T, Norrmén C. et al. Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nat Med 2004; 10 (09) 974-981
  • 57 Strassburg S, Torio-Padron N, Finkenzeller G, Frankenschmidt A, Stark GB. Adipose-derived stem cells support lymphangiogenic parameters in vitro. J Cell Biochem 2016; 117 (11) 2620-2629
  • 58 Knezevic L, Schaupper M, Mühleder S. et al. Engineering blood and lymphatic microvascular networks in fibrin matrices. Front Bioeng Biotechnol 2017; 5: 25
  • 59 Rohringer S, Hofbauer P, Schneider KH. et al. Mechanisms of vasculogenesis in 3D fibrin matrices mediated by the interaction of adipose-derived stem cells and endothelial cells. Angiogenesis 2014; 17 (04) 921-933
  • 60 Yan A, Avraham T, Zampell JC, Haviv YS, Weitman E, Mehrara BJ. Adipose-derived stem cells promote lymphangiogenesis in response to VEGF-C stimulation or TGF-β1 inhibition. Future Oncol 2011; 7 (12) 1457-1473
  • 61 Zhu J, Marchant RE. Design properties of hydrogel tissue-engineering scaffolds. Expert Rev Med Devices 2011; 8 (05) 607-626
  • 62 Chan KLS, Khankhel AH, Thompson RL. et al. Crosslinking of collagen scaffolds promotes blood and lymphatic vascular stability. J Biomed Mater Res A 2014; 102 (09) 3186-3195
  • 63 Helm C-LE, Zisch A, Swartz MA. Engineered blood and lymphatic capillaries in 3-D VEGF-fibrin-collagen matrices with interstitial flow. Biotechnol Bioeng 2007; 96 (01) 167-176
  • 64 Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 2005; 15 (05) 378-386
  • 65 Kanapathy M, Kalaskar D, Mosahebi A, Seifalian AM. Development of a tissue-engineered lymphatic graft using nanocomposite polymer for the treatment of secondary lymphedema. Artif Organs 2016; 40 (03) E1-E11
  • 66 Dai Tt, Jiang Zh, Li Sl. et al. Reconstruction of lymph vessel by lymphatic endothelial cells combined with polyglycolic acid scaffolds: a pilot study. J Biotechnol 2010; 150 (01) 182-189
  • 67 Zhang WJ, Liu W, Cui L, Cao Y. Tissue engineering of blood vessel. J Cell Mol Med 2007; 11 (05) 945-957
  • 68 Schaner PJ, Martin ND, Tulenko TN. et al. Decellularized vein as a potential scaffold for vascular tissue engineering. J Vasc Surg 2004; 40 (01) 146-153
  • 69 Nakamura M, Arai K, Mimura T. et al. Engineering of artificial lymph node. In: Watanabe T, Takahama Y. eds. Synthetic Immunology. Tokyo, Japan: Springer; 2016: 181-200
  • 70 Randolph GJ, Ivanov S, Zinselmeyer BH, Scallan JP. The lymphatic system: integral roles in immunity. Annu Rev Immunol 2017; 35: 31-52
  • 71 Willard-Mack CL. Normal structure, function, and histology of lymph nodes. Toxicol Pathol 2006; 34 (05) 409-424
  • 72 Lüllmann-Rauch R, Asan E. Taschenlehrbuch Histologie. 5th ed.. Stuttgart, Germany: Thieme Verlag; 2015
  • 73 Rouwkema J, Rivron NC, van Blitterswijk CA. Vascularization in tissue engineering. Trends Biotechnol 2008; 26 (08) 434-441
  • 74 Kobayashi Y, Kato K, Watanabe T. Synthesis of functional artificial lymphoid tissues. Discov Med 2011; 12 (65) 351-362
  • 75 Suematsu S, Watanabe T. Generation of a synthetic lymphoid tissue-like organoid in mice. Nat Biotechnol 2004; 22 (12) 1539-1545
  • 76 Okamoto N, Chihara R, Shimizu C, Nishimoto S, Watanabe T. Artificial lymph nodes induce potent secondary immune responses in naive and immunodeficient mice. J Clin Invest 2007; 117 (04) 997-1007
  • 77 Cuzzone DA, Albano NJ, Aschen SZ, Ghanta S, Mehrara BJ. Decellularized lymph nodes as scaffolds for tissue engineered lymph nodes. Lymphat Res Biol 2015; 13 (03) 186-194
  • 78 Richardson TP, Peters MC, Ennett AB, Mooney DJ. Polymeric system for dual growth factor delivery. Nat Biotechnol 2001; 19 (11) 1029-1034
  • 79 Grikscheit TC, Sala FG, Ogilvie J. et al. Tissue-engineered spleen protects against overwhelming pneumococcal sepsis in a rodent model. J Surg Res 2008; 149 (02) 214-218
  • 80 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 (01) 208-220
  • 81 Stacker SA, Williams SP, Karnezis T, Shayan R, Fox SB, Achen MG. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat Rev Cancer 2014; 14 (03) 159-172