You are on page 1of 10

F o c u s o n va s c u l a r d i s e a s e

review

The lymphatic vasculature in disease


Kari Alitalo

Blood vessels form a closed circulatory system, whereas lymphatic vessels form a one-way conduit for tissue fluid and leukocytes.
In most vertebrates, the main function of lymphatic vessels is to collect excess protein-rich fluid that has extravasated from blood
vessels and transport it back into the blood circulation. Lymphatic vessels have an important immune surveillance function, as
they import various antigens and activated antigen-presenting cells into the lymph nodes and export immune effector cells and
humoral response factors into the blood circulation. Defects in lymphatic function can lead to lymph accumulation in tissues,
dampened immune responses, connective tissue and fat accumulation, and tissue swelling known as lymphedema. This review
© 2011 Nature America, Inc. All rights reserved.

highlights the most recent developments in lymphatic biology and how the lymphatic system contributes to the pathogenesis of
various diseases involving immune and inflammatory responses and its role in disseminating tumor cells.

Structure, function and development of lymphatic vessels Although the blood and lymphatic vascular systems are structurally
Blood plasma is continuously filtered from the arterial side of the related and function in concert, they develop at least partially by separate
capillary bed into the interstitial space, where excess fluid and mac- molecular mechanisms. The blood vasculature is formed by vasculogen-
romolecules drain through the permeable lymphatic endothelial cell esis and angiogenesis, driven chiefly by vascular endothelial growth fac-
junctions into lymphatic vessels (Fig. 1). Lacteal lymphatic vessels inside tor (VEGF), whereas lymphangiogenesis is induced by VEGF-C, which
the intestinal villi absorb dietary lipids. Tissues such as the skin and acts on LECs that differentiate from venous endothelial cells at mid-
mucous membranes, which often encounter foreign antigens, are also gestation8,9. LECs express specific molecules such as prospero-related
particularly rich in lymphatic vessels. Lymphatic capillaries are thin- homeodomain transcription factor (Prox1), vascular endothelial growth
walled vessels of approximately 30–80 mm in diameter, composed of a factor receptor-3 (VEGFR-3), the membrane glycoprotein podoplanin
single layer of oak-leaf–shaped lymphatic endothelial cells (LECs) that and lymphatic vessel hyaluronan receptor-1 (refs. 1,8–10). In embryonic
differ in many ways from blood vascular endothelial cells (BECs)1. In day 9.5 (E9.5) mouse embryos, the transcription factor SOX18 induces
contrast to blood capillaries, lymphatic capillaries lack pericytes, which expression of Prox1, which interacts with the venous nuclear receptor
are connective tissue cells surrounding blood capillaries, and have a COUP-TFII and initiates the LEC differentiation program in a segmen-
discontinuous basement membrane containing portals for cells, such as tal pattern in the anterior cardinal vein11,12 (Fig. 1). The dorsolateral
dendritic cells (DCs), that can migrate into the vessels2. Instead of the sprouting, migration and survival of the first LECs and the formation of
zipper-like endothelial junctions that occur between BECs, LECs have lymph sacs are dependent on paracrine secretion of the VEGFR-3 ligand
discontinuous button-like junctions, allowing fluid and certain leuko- VEGF-C8. The VEGF-C co-receptor neuropilin-2 (NRP-2) and the Eph
cytes to enter into the vessel lumen through flap-like openings that form tyrosine kinase ligand ephrin B2 are required for efficient sprouting of
the primary lymphatic valves3–6. Anchoring filaments attach LECs to lymphatic capillaries13,14. The Notch1-Dll4 signaling pathway is essential
collagen fibers and regulate the valve-like opening into the lymphatic for postnatal lymphatic development15. However, in adult tissues VEGF
vessel lumen (Fig. 1). does not promote efficient lymphatic sprouting unless Notch signaling
Lymph moves from the lymphatic capillary bed into precollector ves- is inhibited, whereas sprouting induced by VEGF-C is not restricted by
sels, which are sparsely covered by smooth muscle cells. The precollector Notch16. In addition, the angiopoietins and their Tie receptors have been
vessels drain into collecting lymphatic vessels, which are characterized implicated in lymphangiogenesis17. Angiopoietin-1 (Ang1) induces lym-
by the presence of a periendothelial smooth muscle cell layer, a basement phatic capillary sprouting in mice, and deletion of Ang2 in developing
membrane, continuous zipper-like interendothelial junctions and bileaf- mouse embryos leads to defective maturation of the collecting lymphatic
let valves (Fig. 1)4,7. The intrinsic contractility of smooth muscle cells, vessels17.
the contraction of surrounding skeletal muscles, and arterial pulsations Recent studies show that the collagen and calcium binding EGF
are necessary for lymph propulsion, whereas the valves prevent lymph domains 1 protein (CCBE1) enhances lymphangiogenesis induced by
backflow. In this respect, the collecting lymphatic vessels resemble small VEGF-C and is also required for the budding and sprouting of lymphan-
veins. gioblasts from the venous endothelium in mouse embryos and for tho-
racic duct formation in zebrafish18,19. Interstitial matrix signals can also
Molecular/Cancer Biology Program, Faculty of Medicine, Institute for Molecular stimulate some VEGFR-3 activation when complexed with collagen and
Medicine Finland and Helsinki University Central Hospital, Biomedicum b1 integrin in LECs20,21 (Fig. 2). The points where the lymphatic circula-
Helsinki, University of Helsinki, Helsinki, Finland. tion separates from blood circulation in the developing mouse embryo
Correspondence should be addressed to K.A. (kari.alitalo@helsinki.fi). (for example, the E11.5 cardinal vein) are sealed by platelet aggregates
that form when blood comes in contact with podoplanin expressed in
Published online 07 November 2011; doi:10.1038/nm.2545 LECs22. This separation requires the hematopoietic signaling proteins

nature medicine volume 17 | number 11 | november 2011 1371


review

Figure 1 The lymphatic vasculature and


molecular mechanisms involved in its
development and growth. (a) Lymphatic
capillaries are derived from venous endothelial
cells. After the arteriovenous differentiation
controlled by Notch and COUP-TFII transcription
factors, Sox18 activates Prox1, which interacts
with COUP-TFII and induces lymphatic
endothelial differentiation9,12, involving elevated
expression of VEGFR-3. VEGF-C then induces the
sprouting of the LECs to generate new vessels8.
VEGF-C and VEGF can also increase the size of
lymphatic vessels by stimulating circumferential
growth. EC, endothelial cell. (b) The formation
of lymphatic valves requires a calcium-induced
signal via phospholipase C-g (PLC-g) and
calmodulin to calcineurin that dephosphorylates
the NFATc1 transcription factor, which enters
the nucleus and induces valve-specific genes
in a complex with FoxC2 (T. Petrova (University
of Lausanne), personal communication)28.
VEGFR-2 and VEGFR-3 and ephrin B2 are
upstream regulators of the pathways necessary
© 2011 Nature America, Inc. All rights reserved.

for valve development32. PDGF-B and collagen


IV (ColIV) production is inhibited simultaneously.
ItgA9, integrin a9; FN-EIIIA, embryonic
form of fibronectin; BM, bone marrow. (c) In
developing lymph nodes, lymphangiogenesis is
first induced when IL-7 and TRANCE stimulate
LTi cells that develop from lymphatic precursor cells (LPCs) under the influence of the chemokine CXCL13 (ref. 78). The LTi cells produce LTa1b2,
which activates VEGF-C expression via the LTb receptor in stromal organizer cells48. In adult lymph nodes, B cell proliferation stimulates VEGF-mediated
lymphangiogenesis62, whereas T cell–derived cytokines restrict lymphangiogenesis by producing interferon-g 63. LN, lymph node.

Syk and Slp-76, which are activated in platelets by the transmembrane regulation. The identification of mutant genes in hereditary human
podoplanin receptor Clec2 (refs. 22,23). After their formation, lymphatic lymphedemas has pinpointed crucial pathways during the development
vessels mature and remodel into a branched network that covers the skin of the lymphovascular system1. For example, the most common form
and most internal organs of the body. The development of a hierarchy of of primary lymphedema, lymphedema-distichiasis, is caused by muta-
vessels resembling blood capillaries and veins enables the specialization tions in FOXC2, which encodes a transcription factor that interacts with
of the absorptive functions of the lymphatic capillaries and the transport the calcineurin-NFATc1 signal transduction pathway during normal
and immune functions of the collecting vessels. development of the collecting vessels and valves28 (Fig. 1). Analyses
of Foxc2-mutant mice and individuals with lymphedema-distichiasis
Analyzing lymphatic malfunction in lymphedema have indicated that the underlying cause for the lymphatic failure in
Techniques for direct imaging of the lymphatic system have devel- lymphedema-distichiasis is defective lymphatic valve development and
oped significantly over the last twenty years, allowing new insights to aberrant recruitment of periendothelial smooth muscle cells and basal
be derived into the pathogenesis of lymphatic-related disorders such lamina components to lymphatic capillaries. Downstream of Foxc2,
as lymphedema. Lymphoscintigraphy is routinely used for diagnostic specific connexins are expressed in the developing lymphatic vessels,
imaging of lymphatics and involves injecting radioactive colloid par- including the valve areas, and defective connexins are associated with
ticles intradermally or intraparenchymally, allowing the visualization lymphatic disorders in mice and man29,30. Interestingly, both mice
of their accumulation in the lymphatic plexus and sentinel lymph with Foxc2 mutations as well as humans with lymphedema-distichiasis
nodes. However, this technique is not well suited for imaging lymph also show venous valve insufficiency in addition to lymphedema31,32.
flow. Instead, optical imaging with near-infrared fluorescence dyes such Although the development of venous valves is initiated by the expression
as indocyanine green has a higher sensitivity that permits quantitative of ephrin B2, an arterial marker, these valves later express several genes
imaging of lymph flow, especially when stabilized liposomal formula- that are key regulators of lymphangiogenesis, indicating that there are
tions of the dye are used to prevent toxicity24,25. Other promising new similar morphogenetic mechanisms for valve development in veins and
approaches are also under development, for example optical frequency- lymphatic vessels32.
domain imaging, which overcomes the limitations of fluorescent trac- Another example is provided by analysis of Milroy’s disease
ers such as their tissue penetration and extravasation and is also suited (Supplementary Fig. 1), which is characterized by early-onset congeni-
for high-resolution imaging of tumor microvasculature26. Ultrasound tal lymphedema. This disease commonly involves heterozygous mis-
array-based real-time photoacoustic microscopy enables noninvasive sense mutations that inactivate the kinase activity of VEGFR3, which
high-speed three-dimensional imaging of sentinel lymph nodes27. acts upstream of the FoxC2-NFATc1 pathway (Fig. 1)1,7. In a mouse
Recent molecular genetic studies of lymphangiogenic growth factors, model of Milroy’s disease, the resulting defective lymphangiogenesis
receptors and signaling pathways have also illuminated the pathogen- can be rescued by therapeutic delivery of excess VEGF-C, which gen-
esis of lymphedema, allowing the identification of candidate targets for erates functional lymphatic capillaries33. Genes so far implicated in
lymphedema treatment. These studies have led to a better understand- various lymphedema syndromes have been listed in Table 1(refs. 1,34).
ing of the molecular mechanisms of lymphatic vessel development and The molecular mechanisms of pathogenesis behind several of these

1372 volume 17 | number 11 | november 2011 nature medicine


review

conditions are still poorly understood. In addition, lymphedema may These promising findings could provide a basis for clinical trials combin-
be a component of other rare syndromes, such as Turner’s syndrome, ing autologous transplantation and VEGF-C therapy in patients with,
Noonan’s syndrome and cholestasis-lymphedema syndrome1. for example, postoperative lymphatic insufficiency.
Proteolytic processing of VEGF-C and VEGF-D generates short forms
Rational treatment of secondary lymphedema of these growth factors that bind and activate VEGFR-2, which potently
Lymphangiogenic growth factor therapy may be most appropriate to promotes angiogenesis (Fig. 2). These forms can be used for increas-
treat common forms of secondary lymphedema after lymphatic ves- ing blood and lymphatic vessel size and density in mouse skeletal and
sel damage by surgery, infections or radiation therapy. For example, cardiac muscle43,44. Recent structural studies have indicated that the
one of the most common causes of secondary lymphedema is radical short forms of VEGF-C and VEGF-D have an extended aminoterminal
axillary lymph node dissection during breast cancer surgery. Twenty to alpha helix that is not present in VEGF45,46. Furthermore, one of the two
thirty percent of these patients develop lymphedema of the arm on the VEGF-D isoforms was found to bind VEGFR-2 but not VEGFR-3 (ref.
same side of the body as the lymph node dissection, and patients with 47). Accordingly, this form with a truncated N-terminal helix induced
a high peripheral blood vascular filtration rate seem to be predisposed angiogenesis but not lymphangiogenesis in skeletal muscle, making it a
to this complication35,36. Preclinical studies of lymphedema treatment candidate molecule for proangiogenic therapy in tissue ischemia.
have employed transfer of the gene encoding VEGF-C (or VEGF-D) via
adenoviruses, adeno-associated viruses or naked plasmids or adminis- Lymphatic vessels and immune function
tered VEGF-C protein, which all stimulate the formation of new lym- The collection of interstitial fluid and associated antigens by lymphatic
phatic capillaries and, after an initial increase in lymph extravasation, vessels permits downstream immune monitoring by lymph nodes. In
reduce edema7. In mice, VEGF-C therapy of damaged collecting lymph mouse embryonic development, lymph node induction is initiated at
vessels has been shown to lead to lymphatic capillary growth, which was E12.5 along large veins by lymphoid tissue inducer (LTi) cells of hema-
© 2011 Nature America, Inc. All rights reserved.

followed by intrinsic remodeling, differentiation and maturation into topoietic origin. The tumor necrosis factor family cytokine TRANCE
functional vessels that had normal zipper-like endothelial cell-cell junc- and interleukin 7 (IL-7) induce the LTi cells to produce lymphotoxin
tions and intraluminal valves and were covered by smooth muscle cells37. (LT)-a1b2, which binds the LTb receptor of stromal organizer cells and
Combining VEGF-C therapy with lymph node transplantation in a stimulates production of chemokines, adhesion receptors and cytokines
mouse model of limb lymphedema showed that the growth factor–trans- that attract naive lymphocytes into the developing lymph node (Fig. 1).
duced lymph nodes formed both afferent and efferent connections with Activation of the LTb receptor further induces VEGF-C production in
the preexisting lymphatic vessel network and could even trap metastatic stromal organizer cells, recruiting lymphatic vessels toward the nascent
tumor cells37. In agreement with earlier findings indicating that a con- lymph node48. However, extranodal lymphatic vessel development is
tinuous influx of lymph is required for the maintenance of organized not driven via the LTb receptor pathway, nor does lack of lymph sacs
lymph nodes, control-treated lymph nodes regressed, and their follicular prevent the early stages of lymph node development49.
structure was replaced by adipose and fibrotic tissue37,38. Similar strat- The lymphatic vessels are the principal conduit for soluble antigens
egies have also been used to repair damaged lymphatic networks in a and antigen-presenting cells from peripheral tissues to the lymph nodes
large animal model. In pigs, VEGF-C or VEGF-D therapy effectively for the mounting of immune responses. They also help to clear other
restored lymphatic vasculature to the site of surgical damage and greatly types of leukocytes from sites of resolving inflammation. The LECs
increased the structural preservation and functionality of the transferred of afferent lymphatic vessels attract activated DCs, T cells and B cells
lymph nodes (Supplementary Fig. 2)38. Both growth factors increased expressing the chemokine receptors CCR7 and CCR10 by secreting their
lymphatic drainage when analyzed two months postoperatively. ligands CCL21 (also known as secondary lymphoid chemokine) and
A number of surgical approaches to lymphedema have also been CCL27 (precollector vessels), respectively50,51. Leukocyte trafficking in
reported that are based on small numbers of patients, use nonstandard- the lymphatic vessels is also regulated by LEC products such as com-
ized or inconsistent measurement techniques and lack long-term follow- mon lymphatic endothelial and vascular receptor-1, which mediates
up39. Although lymph node transfer without growth factor therapy has migration of T and B lymphocytes to the draining lymph nodes, and
been shown to provide at least some benefit for humans with lymph- sphingosine-1-phosphate (S1P), which stimulates egress of B and T cells
edema40,41, autologous lymph nodes incorporate into existing lymphatic from lymph nodes52,53. S1P also regulates the maturation of lymphatic
vasculature at a low frequency. However, the new findings in mouse and vessel button and zipper-like junctions either directly or by lymphocyte-
pig models described above suggest that the transfer of chains of lymph mediated mechanisms52.
nodes from another location in the patient could be combined with Antigen-presenting cells such as DCs and small soluble antigens in
VEGF-C treatment to form lymphatic microvascular anastomoses37,42. afferent lymphatic vessels can reach the lymph node subcapsular sinus. A

Table 1 Genes identified behind various lymphedema syndromes1,34


Gene Disease name (OMIM number) Findings
FLT4 (VEGFR-3) Hereditary lymphedema IA (OMIM 153100, AD) Congenital lymphedema
GJC2 Hereditary lymphedema IC (OMIM 613480, AD) Lymphedema of the extremities, onset at <15 years of age
FOXC2 Lymphedema-distichiasis syndrome (OMIM 153400) Lymphedema of mainly lower limbs, triple row of eyelashes, varicose
veins
CCBE1 Hennekam lymphangiectasia-lymphedema syndrome (OMIM 235510, AR) Lymphedema of the extremities, intestinal lymphangiectasia, mental
retardation
SOX18 Hypotrichosis-lymphedema-telangiectasia syndrome (OMIM 607823, AD) Lymphedema, alopecia, teleangiectasias
PTPN14 Lymphedema-choanal atresia syndrome (OMIM 608911, AR) Lymphedema of lower limbs in children, lack of nasal airways
GATA2 Emberger’s syndrome (OMIM 614038, AD) Lymphedema of lower extremities and genitalia, immune dysfunction,
cutaneous warts, deafness
AD, autosomal dominant; AR, autosomal recessive.

nature medicine volume 17 | number 11 | november 2011 1373


review

a S S
CT VHD NT b D1
S S
SS SS
?
S S S S VEGF-C/D VEGF-C
CCBE1
Ab
D2
PlGF VEGF Inhibiting
VEGF-B Collagen Ang1–4 ligand
binding

VEGFR-2
D3

D4

NRP-1 CLP24 Ephrin B2 NRP-2 Integrin Dimerization


b1 inhibiting
PDZ
Src allosteric Ab

VEGFR-1 VEGFR-2 Internalization VEGFR-3 Tie1 Tie2 D5


vesicular trafficking
© 2011 Nature America, Inc. All rights reserved.

Lymphangiogenesis Endothelial survival,


Angiogenesis
Arteriogenesis Angiogenesis vessel stabilization

Figure 2 Lymphangiogenic growth factor–endothelial receptor interactions and binding sites of blocking antibodies. (a) Schematic view of the endothelial
cell–specific growth factors (VEGF, VEGF-B, placental growth factor, VEGF-C, VEGF-D, Ang1, Ang2 and Ang3/4) and receptor tyrosine kinases (VEGFR-1,
VEGFR-2, VEGFR-3 and Tie1 and Tie2)120. Lymphangiogenic signals (in color) are transduced by VEGF-C and VEGF-D, which undergo stepwise proteolytic
activation and bind VEGFR-2 and VEGFR-3. The further functions of the VEGF-C propeptides are unknown and indicated by a question mark. VEGF binds
VEGFR-1 and VEGFR-2. NRP-2, ephrin B2 growth factor and claudin-like protein-24 (CLP24) 121 contain C-terminal PDZ binding domains, which in
NRP-2 and ephrin B2 are involved in the internalization and vesicular trafficking of the receptors. The lymphangiogenic factor CCBE1 binds pericellular
matrix components, such as some collagens19. Collagen can also transduce signals via integrins, VEGFR-3 and Src to LECs20. (b) A structural model of the
VEGF-C–VEGFR-2 complex (in color), as determined by X-ray and small-angle X-ray scattering studies 45,46. In the model, the crystal structure of VEGF-C
in complex with VEGFR-2 domains 2 and 3 are shown in orange and in blue, respectively, in combination with the related Kit receptor domains 1, 4 and 5
in gray, suggesting that VEGFR-2 domains 4 or 5 or both may contribute additional interactions for dimerization upon ligand-induced activation. VEGFR-2
dimerization and activation involves homotypic interactions between the membrane proximal immunoglobulin-like domains 7 (ref. 122). Antibodies (Ab)
that block ligand activation of the receptor interact either with the ligand binding site or allosterically interfere with the region involved in the formation of
active receptor homo- or heterodimers95 (see Fig. 4 for details).

system of conduits that extend into the follicles mediates delivery of small VEGF produced by activated follicular B cells, whereas T cell–derived
soluble antigens to cognate B cells and follicular DCs54. Larger antigens interferon-g signals restrict lymphangiogenesis62,63 (Fig. 1).
are captured by cortical DCs and subcapsular sinus macrophages for Lymphangiogenesis is also associated with chronic inflammation,
translocation, B cell encounter and transfer of opsonized antigen along for example in psoriasis or rheumatoid arthritis64. Furthermore, lym-
macrophage processes to initiate early T cell priming events within the phatic vessel dysfunction and lymphangiogenesis may be involved in
first four hours after antigenic challenge55,56. It is possible that cytoplas- Crohn’s disease, where inflammatory granulomas and ‘fat wrapping’
mic microparticles released by various cells are also transported in lymph around the intestinal lymphatic structures may result from a failure of
and signal to remote lymph nodes57. Importantly, recent studies have the lymphatic vessels to transport inflammatory cells and lipids65. In
indicated that lymph node–resident stromal cells and LECs can induce kidney transplants, CCL21 produced by host-derived lymphatic ves-
tolerance to host peripheral tissue antigens by direct antigen cross-pre- sels attracts CCR7-expressing DCs66, which elicit primary alloantigen
sentation to antigen-specific CD8+ T cells, leading to their deletion58. It is recognition events in the lymph nodes that drain the graft (Fig. 3). In
still unclear how important this mechanism is overall in the maintenance contrast, blocking lymphangiogenic signals mediated by VEGFR-3
of peripheral tolerance, but its failure could contribute, for example, to reduces CCL21 expression in allograft LECs and suppresses adaptive
the chronic inflammation that develops in individuals with lymphedema. immune responses toward heart transplants in a rat model67. Inhibition
of lymphangiogenesis also promotes corneal and pancreatic endocrine
Lymphatic vessels in inflammation islet allograft survival68,69. Thus, whereas abundant lymphatic vessels
Striking changes in lymphatic vessels are associated with acute tissue provide resolution of the inflammatory infiltrate and reduce signs of
inflammation, such as that in bacterial infection. Proinflammatory inflammation in peripheral tissues, the blocking of lymphangiogenesis
cytokines and bacterial lipopolysaccharide induce VEGF-C expression and associated DC migration provide an anti-inflammatory mechanism
in several cell types via the downstream nuclear factor-kB pathway3,59. for the prevention of alloimmunization. The therapeutic concept that
High levels of lymphangiogenic factors are also produced by macro- emerges from these studies suggests that a lymphangiogenic growth
phages and granulocytes in inflamed tissue, and blocking these factors factor such as VEGF-C would clearly be beneficial in dampening tis-
suppresses lymphangiogenesis and reactive inflammation of the lymph sue inflammation associated with lymphedema, whereas inhibition of
node, lymphadenitis3. Lymphangiogenesis in inflammatory settings lymphangiogenesis in the context of heterologous tissue transplantation
facilitates the resolution of tissue edema and promotes macrophage and would be beneficial by dampening the development of a full-blown
DC mobilization3,60,61. Lymph node lymphangiogenesis is stimulated by acquired immune response toward the graft.

1374 volume 17 | number 11 | november 2011 nature medicine


review

Interestingly, because the lymphatic basement membrane has pre- Organ transplantation
formed holes and the lymphatic endothelium has flaps, lymphatic entry
of DCs may not require proteolysis or integrins2,70. Sessile DCs located Soluble
antigen
along blood vessels are mobilized by inflammation, move direction-
ally toward lymphatics, contact CCL21 deposits and migrate through Lymphangiogenesis APC
connective tissue portals and endothelial flaps into the lumen6. Inside CCR7+
VEGF-C VEGF-C
VEG VEGFR3+
lymphatic vessels, DCs extend lamellipodia to crawl along lymphatic
Macrophage
endothelium, and, finally, in the collecting lymphatic vessels they start
T cell
drifting freely along the fluid flow6. DC migration can be promoted by CCL21
VEGF-C, increased transmural fluid flow, delayed type hypersensitiv-
ity reactions and tumor necrosis factor, all of which upregulate CCL21
Blood
Blood vessels Lymphatic
secretion, as well as the ICAM-1 leukocyte adhesion receptor by the and
nd circulation
an vessels
LECs71–73. CCL21 binds heparan sulfate, which seems essential for
Spleen
DC intravasation; podoplanin and collagen type IV may be involved
as well6,74. However, although ICAM-1 contributes to lymphatic trans-
migration in at least some inflammatory processes73, inflamed LECs
were reported to suppress DC maturation in an ICAM-1–dependent
manner, perhaps to prevent undesired (auto)immune responses during
inflammatory processes75. Additionally, D6, a decoy receptor, which
Lymph
ph n
nodes
is expressed in LECs, scavenges inflammatory CC chemokines and is
© 2011 Nature America, Inc. All rights reserved.

thus capable of reducing the inflammatory response in various organs76. Figure 3 A schematic model of lymphatic vessel function in organ
Although lymph nodes are highly plastic organs, complete lymph transplantation. Inflammatory cells such as macrophages in organ
nodes cannot form de novo postnatally. However, ectopic, ‘tertiary’ transplants can produce high levels of VEGF-C66. The resulting lymphatic
vessel activation is associated with enhanced secretion of the CCL21 ligand
lymphoid organs consisting of clonally expanding B cell follicles, T
of the chemokine receptor CCR7, improved flow of lymph that contains
cell compartments and high endothelial venules can be found at sites soluble antigens, and mobilization of activated antigen-presenting cells
of chronic inflammation, even in atherosclerosis, and their formation (APCs) that express CCR7; some APCs also express VEGFR-3 (ref. 67). APCs
seems to involve many of the same chemokines that operate in lymph migrate to lymph vessels, then to lymph nodes, and further, via the blood
node development77,78. LTa can promote inflammation-associated lym- vessels, to the spleen to activate T cells for immune rejection of the graft,
phangiogenesis, and CD11c-positive DCs are crucial in the induction of being subsequently delivered to the graft via the blood vessels. The blocking
of lymphangiogenesis, which has so far not shown adverse side effects in
lymphangiogenesis in the tertiary lymphoid structures79,80.
normal tissue, inhibits acute graft rejection in the context of heart, corneal
and pancreatic islet cell transplantation survival67–69.
Tissue inflammation links lymphatic vessels to obesity and
cardiovascular disease
A functional link has emerged between lymphatic malfunction and arteries adjacent to small blood vessels, called the vasa vasorum, which
the pathogenesis of obesity, atherosclerosis and cardiovascular disease, are expanded in atherosclerotic plaques77,88. Mice lacking apolipopro-
which are leading causes of disability and mortality in developed coun- tein E have not only atherosclerotic plaques and tertiary lymphoid tissue
tries. Lymphatic vessels are important in lipid metabolism, as intestinal in the adventitia of arteries but also dysfunctional lymphatic vessels77,89.
lymphatics take up dietary lipids in the form of chylomicrons, large Lymphatic vessels could perhaps provide a protective pathway for lipid
lipoprotein particles, to transport them to the bloodstream. Lymph and inflammatory cell efflux from the arterial wall, which could oppose
nodes and collecting lymphatic vessels are commonly embedded in the development of atherosclerotic plaques. Further studies should
subcutaneous or visceral fat, and the perinodal adipose tissue undergoes reveal whether manipulation of lymphangiogenesis can provide a means
dynamic changes in response to low-level inflammatory processes81, to slow down fat deposition and tissue inflammation associated with the
which are known to contribute to obesity-associated metabolic disease development of atherosclerotic lesions.
involving insulin resistance, type 2 diabetes and cardiovascular disease. Inhibition of the VEGF-C receptor VEGFR-3 acts as a lymphatic
A link between lymphatic dysfunction, inflammation and adipogenesis vessel–targeted immunomodulatory therapy for the arteriosclerosis that
is evident from the development of ectopic subcutaneous adipose tis- develops in a cardiac allograft model67. The heart itself has lymphatic
sue in edematous sites in individuals with chronic lymphedema82, but vessels, but virtually nothing is known about their role in the healthy
the molecular mechanisms of these changes are poorly known. Mice or failing heart. Cardiac contraction generates considerable transmural
with a heterozygous Prox1-inactivating mutation were found to have pressures that are predicted to result in cardiac or pericardial edema if
leaky lymphatic vessels and to develop obesity and inflammation with the lymphatic vessels do not remove the excess fluid. However, it remains
features associated with late-onset obesity in humans, such as higher to be seen whether increased lymphatic function improves myocardial
levels of leptin and insulin and fatty liver83. Furthermore, mice with performance in, for example, infarcted or dilated heart, in which the
a heterozygous mutation in Vegfr3 (so-called Chy mice) had lymph- wall tension is severely elevated.
edema and chronic tissue inflammation in association with abnormal fat Recent results have shown that hypertension induced by salt overload
accumulation33,84. Conversely, therapeutic lymphangiogenesis induced leads to inflammatory cell recruitment and increased lymphangiogenic
by VEGF-C alleviated tissue inflammation and edema in the mouse growth factor production in the skin90. These studies have suggested
models33,85. a VEGF-C–mediated mechanism in the skin that maintains normal
Lymphatic vessels are also found at sites of atherosclerosis, which blood pressure in states of interstitial salt accumulation. An excess of
is associated with inflammation and lipid accumulation in arterial salt accumulated in the skin without associated water retention recruits
walls86,87. Arterial smooth muscle cells constitutively produce lymphan- macrophages, which are induced to secrete VEGF-C. Selective blockade
giogenic factors, and lymphatic vessels are present in the adventitia of of VEGF-C and VEGFR-3–mediated signals resulted in salt-sensitive

nature medicine volume 17 | number 11 | november 2011 1375


review

gramming of primary LECs to mesenchymal, invasive ‘spindle’ cells via


VEGF-C/D trap
activation of the Notch signal transduction pathway92,93. Moreover, a
component of the KSHV capsid has been shown to activate VEGFR-3
VEGF trap
and induce endothelial proliferation and migration94. Inhibition of
Ab VEGFR-3 with a combination of the two blocking antibodies schemati-
VEGF Ab VEGF-C Ab Pb Ang2
cally illustrated in Figure 4 was effective in reducing (lymph)angiogenic
sprouting and growth of KSHV-infected LECs in three-dimensional cul-
ture, suggesting that this approach could provide a potential treatment
Ab option for Kaposi’s sarcoma95.
Lymphangioleiomyomatosis (LAM) is characterized by the infiltra-
Ab*
tion of abnormal, smooth muscle–like LAM cells through the pulmo-
VEGF/Ang2 nary interstitium, perivascular spaces and the lymphatics of young
trap females, leading to cystic structures that obstruct airways and lead to
lymphatic disruption with effusion of fluid containing lymph and lipids,
finally resulting in respiratory failure. In about one-third of the cases,
TKI LAM is associated with mutations in the tuberous sclerosis tumor sup-
pressors TSC1 or TSC2 involved in mTOR signaling and also with the
formation of renal angiomyolipomas (tumors made up of LAM cells,
adipose tissue and poorly developed blood vessels)96. Although some
VEGFR-1 VEGFR-2 VEGFR-3 Tie1 Tie2 patients with LAM benefit from the mTOR inhibitor rapamycin97, pul-
© 2011 Nature America, Inc. All rights reserved.

monary transplantation is needed for treatment of progressive disease.


High, often diagnostic levels of VEGF-D are secreted by LAM cells that
Angiogenesis Lymphangiogenesis Angiogenesis
frequently become covered by LECs and invade the lymphatic system
Figure 4 Inhibitors of VEGF, VEGF-C, Ang2 and their receptors. The blocking and lungs98. Although an animal model of LAM is lacking, VEGFR-3–
antibodies (Ab) against VEGF (bevacizumab) and VEGF-C and against the blocking antibodies, which are currently in phase 1 clinical trials (http://
ligand-binding parts of VEGFR-2 (Ramucirumab) and VEGFR-3 (http://
clinicaltrials.gov/ct2/show/NCT01288989), may provide another agent
clinicaltrials.gov/ct2/show/NCT01288989) are shown in red. The antibodies
marked Ab* are allosteric inhibitors of active VEGFR dimer formation95,123. for LAM treatment.
The VEGF trap (aflibercept) contains the ligand-binding immunoglobulin
homology domain 2 (Ig2) of VEGFR-1 fused to the Ig3 domain of VEGFR-2 Lymphatic metastasis and its inhibition
and further to the Fc part of human IgG (http://clinicaltrials.gov/ct2/show/ The lymphatic vessels also provide a route for tumor cells to metastasize.
NCT00561470?term=aflibercept&phase=2&rank=4). The VEGF-C/VEGF-D Solid tumors have high interstitial pressure, which may at least partly
trap consists of a fusion of the Ig1–3 domains of VEGFR-3 with the IgG Fc
have a role in enabling tumor cells to enter lymphatic vessels in the
domain42. The VEGF/Ang2 trap fuses the Ig1 and Ig2 domains plus the EGF
homology domains of Tie2 with the Ig2 domain of VEGFR-1 and further with
tumor periphery and to metastasize99. When injected into tumor tissue,
the IgG Fc domain124. The peptibody (Pb) against Ang2 is a fusion of an tracer compounds are readily taken up by the lymphatic vessels that
inhibitory Ang2 binding peptide with the IgG Fc domain. Ang2 inhibitors and drain the tumor, providing the means for sentinel lymph node mapping
VEGF inhibitors together result in increased reduction of tumor vascularity for tumor staging, for example in breast cancer and melanoma42,100.
and growth125,126. VEGF-B, PlGF and VEGFR-1 are outside of the scope of However, growth of tumor cells after their arrival in the marginal sinus of
this review. TKI, tyrosine kinase inhibitor. the sentinel lymph node may impede lymph flow and reroute the lymph
flow around the blocked lymph node, thus compromising the mapping
hypertension despite increased expression of endothelial nitric oxide of sentinel nodes24,100,101. Despite these considerations, lymph node
synthetase90. These findings suggest that macrophages are extrarenal metastasis continues to be used as an important parameter in tumor
regulators of electrolyte, volume and blood pressure homeostasis via a staging and therapeutic decision-making.
unique regulatory feedback control system involving lymphangiogenic Growth in the lymph node microenvironment may select tumor cells
factors. for increased metastatic potential. Current cancer treatments comprise
surgical removal of the primary tumor and metastatic lymph nodes fol-
Lymphovascular tumors lowed by various types of adjuvant therapy. A tumor-positive sentinel
A number of tumor types can form in the lymphatic tissues, and lymph node biopsy frequently leads to complete surgical resection of
researchers are just beginning to understand the pathogenesis of these lymph nodes. However, several large clinical studies have failed to con-
tumors. Studies investigating lymphovascular tumors have highlighted firm a survival advantage despite the previously held view that axillary
that manipulation of lymphangiogenesis and angiogenesis may be of lymph node dissection and irradiation improve the overall survival of
potential therapeutic benefit for lymphatic tumors. Lymphangiosarcoma individuals with breast carcinoma whose sentinel lymph node biopsy
is a malignant tumor that occurs in long-standing cases of primary or contains tumor cells102,103. Overall, the studies suggest that there is
secondary lymphedema, but its molecular pathogenesis is unclear. LECs tumor type–specific variation between the contribution of lymphatic
may also give rise to Kaposi’s sarcoma, which is an angiogenic tumor vessels and the formation of distant metastases.
common especially in the HIV-infected populations of sub-Saharan Recent progress in tumor imaging, high-throughput analysis of
Africa. Kaposi’s sarcoma is caused by infection with a gammaherpes- genomic aberrations and clonal single-cell analysis has provided new
virus (KSHV) that has previously been shown to induce reprogramming insights into early tumor cell dissemination, and tumor cell dormancy
of BECs to LECs (and vice versa)91. Correspondingly, global transcrip- versus metastasis outgrowth104,105. Considerable heterogeneity is evi-
tomic analysis has demonstrated that the gene expression signature dent when the primary tumor and different lymphatic and distant organ
in the Kaposi’s sarcoma tumors resembles more that of LECs than of metastases are compared106,107. This has enabled the drawing of phylo-
BECs. More recently, KSHV was shown to induce transcriptional repro- genetic trees across metastases, which show the organ-specific branches

1376 volume 17 | number 11 | november 2011 nature medicine


review

Figure 5 Mechanisms contributing to lymphatic


metastasis. (a) Top left, the normal lymph node
a b Tumor Sentinel LN
(LN) has been stained with antibodies detecting B LN Metastasis in LN VEGF-C, VEGF, Ang2↑ LN lymphangiogenesis
cells (red), T cells (green) and lymphatic sinusoid Tolerance induction?
VEGF, VEGF-C↑
(white)63. Top right, the eosin-stained (blue) LN Lymphangiogenesis
Lipoxygenase↑
Angiogenesis Collecting
shows metastatic foci in gray. Bottom, increased CCL21↑
CCL21↑
lymphatic vessels
VEGF-C and D secretion by tumor cells or tumor- Increase in size
associated inflammatory cells induces tumor Increased flow
lymphangiogenesis, hyperplasia of collecting Mø
LTi
lymphatic vessels and tumor cell translocation Treg Distant
Circumferential
into lymphatic vessels and lymph nodes10,42,64. hyperplasia LN
(b) Aberrant expression of chemokine receptors Mø
such as CCR7 may increase tumor cell migration
toward lymphatic vessels that produce its
Angiogenesis and
ligand CCL21 (refs. 50,108). Tumors may also tumor growth
stimulate LN lymphangiogenesis (sinusoidal Inhibitors:
hyperplasia) 64,112 in the draining (sentinel) Anti–VEGF
Anti–VEGFR-2
lymph node and invade the generated vessels Anti–VEGFR-3
in the lymph nodes by a lipoxygenase-mediated VEGF trap
VEGF-C/VEGF-D trap
mechanism that contributes to further metastasis
TKI
to secondary lymph nodes112. VEGF and Ang2 Tumor Intralymphatic Anti–Ang-2
can also contribute to these lymphangiogenic lymphangiogenesis tumor growth Ang2 peptibody
VEGF/Ang2 trap
changes, whereas macrophages (MØ) are recruited
© 2011 Nature America, Inc. All rights reserved.

Lymphangiogenesis
to the tumor stroma. VEGF-C also upregulates
Metastasis
lymph flow and CCL21 expression, which has
been shown to lead to recruitment of CCR7-
expressing immune cells, including regulatory
T cells (Treg) and LTi cells, resulting in a shift of the host immune response from immunogenic to tolerogenic, which facilitates tumor progression 108. Some of
the tumor-reactive CD8+ T cells may also be eliminated in the sentinel lymph node111.

and genetic heterogeneity among metastasis-initiating cells and indicate lymphatic vessels into postsentinel lymph nodes of the same chain (Fig.
that driver mutations may be required for metastasis development107. In 5). In human mammary carcinomas and their matching axillary lymph
different tumor types, the density of tumor-associated lymphatic vessels nodes, intrametastatic lymphatic vessels, lipoxygenase expression and
varies and correlates with the incidence of lymph node metastasis and bulk tumor cell invasion into these vessels highly correlates with the
poor prognosis10,42,64. However, it is not clear to what extent lymph formation of postsentinel metastasis112.
node metastases promote spreading of the tumor cells by expanding the Inhibition of lymphangiogenesis with a soluble form of VEGFR-3 or
tumor and serving as a launch pad for further systemic metastasis105. monoclonal antibodies that block this receptor inhibits 50–70% of tumor
Intratumoral lymphatic vessels have been considered to be poorly metastasis to lymph nodes in mouse and rat models42,105. VEGFR-3 sig-
functional as a result of high intratumoral pressure, whereas lymphatic naling can also contribute to angiogenesis in tumors where the receptor
vessels in the tumor periphery are functional99. Because of the lower is expressed on both tumor blood and lymphatic vessels113. The involve-
mechanical stress inside peritumoral lymphatic vessels, they may pro- ment of VEGFR-3 in the growth of both blood vessels and lymphatics
vide a more favorable environment for metastasis dissemination than makes this receptor a promising candidate for cancer therapy, as it is pos-
blood vessels. In addition, LECs secrete ‘lymphangiocrine’ chemokines, sible that, by blocking this receptor, a dual effect on lymphangiogenesis
such as CCL21, that can promote tumor cells expressing the cognate and angiogenesis may be obtained. Consistent with this, monoclonal
receptor CCR7 to migrate toward the lymphatic vessels. Another antibodies that block the binding of VEGF-C and VEGF-D to VEGFR-3
important chemokine produced by LECs is stromal-derived factor-1 can suppress angiogenesis, and antibodies against VEGFR-3 and
(also known as CXCL12), which binds CXCR4 on tumor cells, promot- VEGFR-2 in combination resulted in additive inhibition of lymphangio-
ing a lymphatic microenvironment that supports tumor growth108,109. genesis114, angiogenesis and tumor growth113. Furthermore, the efficacy
Although tumor cells can invade lymphatic vessels, it has become evi- of angiogenesis inhibition was increased through the simultaneous use
dent that tumors overexpressing either VEGF-C or VEGF-D in tumor of an antibody that blocks growth factor binding to VEGFR and another
cells or stromal cells, such as tumor-associated macrophages, induce antibody that allosterically inhibits the formation of kinase-activated
sprouting of lymphatic vessels, enlargement of collecting lymph vessels VEGFR dimers95. Also, NRP-2–blocking antibodies have recently been
and lymph node lymphangiogenesis (also known as sinusoidal hyper- shown to reduce tumor lymphangiogenesis and lymph node metastasis,
plasia) in the draining lymph nodes42,64,110. Such tumors are particularly at least partly by suppressing LEC migration13,115. However, the safety
prone to developing lymph node metastases10,42,64. When overexpressed, of a treatment targeting VEGFR-2 and VEGFR-3 needs to be carefully
several other growth factors, such as VEGF, fibroblast growth factors, addressed as, for example, hematopoietic reconstitution after irradiation
platelet-derived growth factor-B (PDGF-B), hepatocyte growth factor for bone marrow depletion may require VEGFR-2 and VEGFR-3 expres-
and insulin-like growth factor-1, can also induce lymphangiogenesis sion in the bone marrow sinusoidal endothelium116. Further validation
and metastasis, presumably via more indirect pathways, such as inflam- of the efficacy and safety of antibody combinations in preclinical models
matory cell recruitment and VEGF-C induction10,42. It has been sug- could lead to inhibitors that block both tumor angiogenesis and lymph
gested that the tumor-draining lymph nodes are capable of promoting node metastasis in patients with cancer.
immune tolerance toward tumor antigens by antigen cross-presentation Despite complete removal of the primary tumor, approximately one-
to CD8+ T cells that can lead to the destruction of the tumor antigen- fifth of individuals with melanoma develop local metastasis, termed
specific T cells110. Furthermore, although metastases form initially in, satellite metastasis or in-transit metastasis, after surgery117, and the
for example, axillary sentinel lymph nodes, they progress via connecting lymphatic system may contribute to this. Indeed, clinical studies have

nature medicine volume 17 | number 11 | november 2011 1377


review

suggested that tumor-draining collecting lymphatic vessels host tumor ACKNOWLEDGMENTS


cells. We have recently shown that photodynamic therapy with the ben- I am grateful to M. Bry, L. Eklund, S. Jalkanen, D. Kerjaschki, G.Y. Koh,
V.-M. Leppänen, T. Mäkinen, A. Nykänen, M. Öhman, P. Ojala, P. Saharinen,
zoporphyrin derivative verteporfin can be used to destroy lymphatic
M. Swartz and T. Tammela for useful discussions of the topics of this review.
vessels and small tumor nodules inside them that have the capacity to The lymph node images in Figure 5a were kindly provided by D. Kerjaschki
develop into secondary tumors118. In addition, the photodynamic ther- and by G.Y. Koh, and draft figures were produced by H. Schmidt. The studies in
apy in combination with blocking of the VEGFR-3 pathway prevented my laboratory are currently supported by the Academy of Finland, the Finnish
metastasis of mouse melanoma cells as well as subsequent relapse118. Cancer Organisations, the Association for International Cancer Research, the
Sigrid Juselius Foundation, Seventh Framework Program of the European Union
These studies show considerable potential for the development of clini- (ERC Advanced Grant), Finnish Foundation for Cardiovascular Research and
cal antimetastasis therapy. Biocentrum Finland. I apologize to the many authors whose important work could
not be cited because of space restrictions and the focus on the recent developments;
Clinical translation of lymphatic biology older references appear in the many excellent reviews that have been cited.
The essential role of the lymphatic vessels in a multitude of biological COMPETING FINANCIAL INTERESTS
functions such as in the maintenance of tissue fluid balance, immuno- The author declares competing financial interests: details accompany the full-text
surveillance and transport of dietary fats, and as a gateway for tumor HTML version of the paper at http://www.nature.com/naturemedicine/.
metastasis, make them excellent targets for drug development in vari- Published online at http://www.nature.com/naturemedicine/.
ous diseases. Through the discovery of lymphangiogenic factors and Reprints and permissions information is available online at http://www.nature.com/
studies of their signal transduction pathways, it has become possible to reprints/index.html.
stimulate or inhibit lymphatic vessel formation in adult tissues. This has
provided new possibilities for the treatment of secondary lymphedema 1. Schulte-Merker, S., Sabine, A. & Petrova, T.V. Lymphatic vascular morphogenesis in
development, physiology, and disease. J. Cell Biol. 193, 607–618 (2011).
in humans and for the inhibition of lymphatic metastasis with the benefit
© 2011 Nature America, Inc. All rights reserved.

2. Pflicke, H. & Sixt, M. Preformed portals facilitate dendritic cell entry into afferent
of additional inhibition of tumor angiogenesis. lymphatic vessels. J. Exp. Med. 206, 2925–2935 (2009).
The first inhibitor of lymphangiogenesis, the VEGFR-3–targeting 3. Baluk, P. et al. Pathogenesis of persistent lymphatic vessel hyperplasia in chronic
airway inflammation. J. Clin. Invest. 115, 247–257 (2005).
monoclonal antibody IMC-3C5, has recently entered phase 1 clinical 4. Dejana, E., Tournier-Lasserve, E. & Weinstein, B.M. The control of vascular integrity
trials for patients with advanced solid tumors that are refractory to stan- by endothelial cell junctions: molecular basis and pathological implications. Dev.
Cell 16, 209–221 (2009).
dard therapy or for which no standard therapy is available. The animal 5. Pfeiffer, F. et al. Distinct molecular composition of blood and lymphatic vascular
data predict that clinical studies using VEGFR-3–blocking antibodies endothelial cell junctions establishes specific functional barriers within the peripheral
should provide benefit as antiangiogenic and antimetastatic agents. lymph node. Eur. J. Immunol. 38, 2142–2155 (2008).
6. Tal, O., et al. DC mobilization from the skin requires docking to immobilized CCL21
Further trials using lymphangiogenesis blockers seem equally straight- on lymphatic endothelium and intralymphatic crawling. J. Exp. Med. 208, 2141–
forward for the treatment of Kaposi’s sarcoma or LAM, and they may 2153 (2011).
7. Norrmén, C., Tammela, T., Petrova, T.V. & Alitalo, K. Biological basis of therapeutic
aid in prolonging graft survival in, for example, corneal transplantation. lymphangiogenesis. Circulation 123, 1335–1351 (2011).
These inhibitors may also be useful in treating dry eye disease118. 8. Karkkainen, M.J. et al. Vascular endothelial growth factor C is required for sprouting
A clinical trial using VEGF-C growth factor therapy is imminent of the first lymphatic vessels from embryonic veins. Nat. Immunol. 5, 74–80 (2004).
9. Wigle, J.T. & Oliver, G. Prox1 function is required for the development of the murine
for the regeneration of lymphatic vessels in the treatment of secondary lymphatic system. Cell 98, 769–778 (1999).
lymphedema. The ability of VEGF-C to regulate the growth of lym- 10. Albrecht, I. & Christofori, G. Molecular mechanisms of lymphangiogenesis in develop-
ment and cancer. Int. J. Dev. Biol. 55, 483–494 (2011).
phatic vessels should facilitate successful lymph node transfer when 11. François, M. et al. Sox18 induces development of the lymphatic vasculature in mice.
combined transplant and VEGF-C therapy is used to treat lymphedema. Nature 456, 643–647 (2008).
This strategy should result in more durable lymph node transplants, 12. Srinivasan, R.S. et al. The nuclear hormone receptor Coup-TFII is required for the
initiation and early maintenance of Prox1 expression in lymphatic endothelial cells.
reducing the need for pressure bandages, which are sometimes painful. Genes Dev. 24, 696–707 (2010).
In addition, broader applications of VEGF-C and VEGF-D may become 13. Xu, Y. et al. Neuropilin-2 mediates VEGF-C–induced lymphatic sprouting together
possible in regenerative medicine and tissue engineering for patients with VEGFR3. J. Cell Biol. 188, 115–130 (2010).
14. Mäkinen, T. et al. PDZ interaction site in ephrinB2 is required for the remodeling of
with tissue defects, including those with lymphatic vessel damage due lymphatic vasculature. Genes Dev. 19, 397–410 (2005).
to skin burn injuries. 15. Niessen, K. et al. The Notch1-Dll4 signaling pathway regulates mouse postnatal
lymphatic development. Blood 118, 1989–1997 (2011).
Knowledge of the genetics of inherited syndromes involving the lym- 16. Zheng, W. et al. Notch restricts lymphatic vessel sprouting induced by vascular
phatic vessels has provided insight into the pathogenesis of lymphatic endothelial growth factor. Blood 118, 1154–1162 (2011).
vascular diseases. There is now increased attention on the mechanisms 17. Augustin, H.G., Koh, G.Y., Thurston, G. & Alitalo, K. Control of vascular morphogen-
esis and homeostasis through the angiopoietin-Tie system. Nat. Rev. Mol. Cell Biol.
coupling lymphatic vessels to immune regulation, host defense to infec- 10, 165–177 (2009).
tions, allo- and autoimmune responses and perhaps also obesity, meta- 18. Hogan, B.M. et al. Ccbe1 is required for embryonic lymphangiogenesis and venous
sprouting. Nat. Genet. 41, 396–398 (2009).
bolic disease and atherosclerosis. Such research should result in the 19. Bos, F.L. et al. CCBE1 Is Essential for Mammalian lymphatic vascular development
design of new therapies for these conditions. Improved knowledge on and enhances the lymphangiogenic effect of vascular endothelial growth factor-C in
lymphatic vascular biology could aid also the development of better vac- vivo. Circ. Res. 109, 486–491 (2011).
20. Galvagni, F. et al. Endothelial cell adhesion to the extracellular matrix induces c-Src–
cination strategies. Furthermore, new questions are continuously emerg- dependent VEGFR-3 phosphorylation without the activation of the receptor intrinsic
ing from the findings obtained from preclinical studies. For instance, do kinase activity. Circ. Res. 106, 1839–1848 (2010).
the persistently enlarged lymphatic vessels in airway submucosa after 21. Tammela, T., et al. VEGFR-3 controls tip to stalk conversion at vessel fusion sites by
reinforcing Notch signalling. Nat. Cell Biol. 13, 1202–1213 (2011).
respiratory infections provide more efficient secondary responses upon 22. Uhrin, P. et al. Novel function for blood platelets and podoplanin in developmental
re-infection, or do they predispose to inflammatory diseases3? What are separation of blood and lymphatic circulation. Blood 115, 3997–4005 (2010).
23. Bertozzi, C.C. et al. Platelets regulate lymphatic vascular development through CLEC-
the pathogenenetic mechanisms behind intestinal lymphangiectasias 2-SLP-76 signaling. Blood 116, 661–670 (2010).
associated with protein-losing enteropathy? Answering such questions 24. Proulx, S.T. et al. Quantitative imaging of lymphatic function with liposomal indo-
should provide additional clinical concepts for the therapeutic targeting cyanine green. Cancer Res. 70, 7053–7062 (2010).
25. Rasmussen, J.C., Tan, I.C., Marshall, M.V., Fife, C.E. & Sevick-Muraca, E.M.
of the lymphatic system in various human diseases. Lymphatic imaging in humans with near-infrared fluorescence. Curr. Opin.
Biotechnol. 20, 74–82 (2009).
Note: Supplementary information is available on the Nature Medicine website. 26. Vakoc, B.J. et al. Three-dimensional microscopy of the tumor microenvironment in

1378 volume 17 | number 11 | november 2011 nature medicine


review

vivo using optical frequency domain imaging. Nat. Med. 15, 1219–1223 (2009). 60. Kataru, R.P. et al. Critical role of CD11b+ macrophages and VEGF in inflamma-
27. Song, L., Maslov, K., Shung, K.K. & Wang, L.V. Ultrasound-array–based real-time tory lymphangiogenesis, antigen clearance and inflammation resolution. Blood 113,
photoacoustic microscopy of human pulsatile dynamics in vivo. J. Biomed. Opt. 15, 5650–5659 (2009).
021303 (2010). 61. Huggenberger, R. et al. An important role of lymphatic vessel activation in limiting
28. Norrmén, C. et al. FOXC2 controls formation and maturation of lymphatic collecting acute inflammation. Blood 117, 4667–4678 (2011).
vessels through cooperation with NFATc1. J. Cell Biol. 185, 439–457 (2009). 62. Angeli, V. et al. B cell–driven lymphangiogenesis in inflamed lymph nodes enhances
29. Kanady, J.D., Dellinger, M.T., Munger, S.J., Witte, M.H. & Simon, A.M. Connexin37 dendritic cell mobilization. Immunity 24, 203–215 (2006).
and Connexin43 deficiencies in mice disrupt lymphatic valve development and 63. Kataru, R.P. et al. T lymphocytes negatively regulate lymph node lymphatic vessel
result in lymphatic disorders including lymphedema and chylothorax. Dev. Biol. formation. Immunity 34, 96–107 (2011).
354, 253–266 (2011). 64. Cueni, L.N. & Detmar, M. The lymphatic system in health and disease. Lymphat.
30. Ferrell, R.E. et al. GJC2 missense mutations cause human lymphedema. Am. J. Hum. Res. Biol. 6, 109–122 (2008).
Genet. 86, 943–948 (2010). 65. von der Weid, P.Y., Rehal, S. & Ferraz, J.G. Role of the lymphatic system in the
31. Mellor, R.H. et al. Mutations in FOXC2 are strongly associated with primary valve pathogenesis of Crohn’s disease. Curr. Opin. Gastroenterol. 27, 335–341 (2011).
failure in veins of the lower limb. Circulation 115, 1912–1920 (2007). 66. Kerjaschki, D. et al. Lymphatic endothelial progenitor cells contribute to de novo
32. Bazigou, E. et al. Genes regulating lymphangiogenesis control venous valve formation lymphangiogenesis in human renal transplants. Nat. Med. 12, 230–234 (2006).
and maintenance in mice. J. Clin. Invest. 121, 2984–2992 (2011). 67. Nykänen, A.I. et al. Targeting lymphatic vessel activation and CCL21 production by
33. Karkkainen, M.J. et al. A model for gene therapy of human hereditary lymphedema. vascular endothelial growth factor receptor-3 inhibition has novel immunomodulatory
Proc. Natl. Acad. Sci. USA 98, 12677–12682 (2001). and antiarteriosclerotic effects in cardiac allografts. Circulation 121, 1413–1422
34. Ostergaard, P., et al. Mutations in GATA2 cause primary lymphedema associated with (2010).
a predisposition to acute myeloid leukemia (Emberger syndrome). Nat. Genet. 43, 68. Albuquerque, R.J. et al. Alternatively spliced vascular endothelial growth factor
929–931 (2011). receptor-2 is an essential endogenous inhibitor of lymphatic vessel growth. Nat.
35. Stanton, A.W., Modi, S., Mellor, R.H., Levick, J.R. & Mortimer, P.S. Recent advances Med. 15, 1023–1030 (2009).
in breast cancer-related lymphedema of the arm: lymphatic pump failure and pre- 69. Yin, N. et al. Targeting lymphangiogenesis after islet transplantation prolongs islet
disposing factors. Lymphat. Res. Biol. 7, 29–45 (2009). allograft survival. Transplantation 92, 25–30 (2011).
36. McLaughlin, S.A. et al. Prevalence of lymphedema in women with breast cancer 5 70. Lämmermann, T. et al. Rapid leukocyte migration by integrin-independent flowing
years after sentinel lymph node biopsy or axillary dissection: objective measurements. and squeezing. Nature 453, 51–55 (2008).
J. Clin. Oncol. 26, 5213–5219 (2008). 71. Miteva, D.O. et al. Transmural flow modulates cell and fluid transport functions of
© 2011 Nature America, Inc. All rights reserved.

37. Tammela, T. et al. Therapeutic differentiation and maturation of lymphatic vessels lymphatic endothelium. Circ. Res. 106, 920–931 (2010).
after lymph node dissection and transplantation. Nat. Med. 13, 1458–1466 (2007). 72. Schumann, K. et al. Immobilized chemokine fields and soluble chemokine gradients
38. Lähteenvuo, M. et al. Growth factor therapy and autologous lymph node transfer in cooperatively shape migration patterns of dendritic cells. Immunity 32, 703–713
lymphedema. Circulation 123, 613–620 (2011). (2010).
39. Cormier, J.N., Rourke, L., Crosby, M., Chang, D. & Armer, J. The surgical treatment 73. Johnson, L.A. & Jackson, D.G. Inflammation-induced secretion of CCL21 in lym-
of lymphedema: a systematic review of the contemporary literature (2004–2010). phatic endothelium is a key regulator of integrin-mediated dendritic cell transmigra-
Ann. Surg. Oncol. published online, doi:10.1245/s10434-011-2017-4 (24 August tion. Int. Immunol. 22, 839–849 (2010).
2011). 74. Bao, X. et al. Endothelial heparan sulfate controls chemokine presentation in recruit-
40. Saaristo, A.M. et al. Microvascular breast reconstruction and lymph node transfer ment of lymphocytes and dendritic cells to lymph nodes. Immunity 33, 817–829
for postmastectomy lymphedema patients. Ann. Surg. (in the press). (2010).
41. Becker, C., Assouad, J., Riquet, M. & Hidden, G. Postmastectomy lymphedema: 75. Podgrabinska, S. et al. Inflamed lymphatic endothelium suppresses dendritic cell
long-term results following microsurgical lymph node transplantation. Ann. Surg. maturation and function via Mac-1/ICAM-1–dependent mechanism. J. Immunol.
243, 313–315 (2006). 183, 1767–1779 (2009).
42. Tammela, T. & Alitalo, K. Lymphangiogenesis: Molecular mechanisms and future 76. Vetrano, S. et al. The lymphatic system controls intestinal inflammation and inflam-
promise. Cell 140, 460–476 (2010). mation-associated colon cancer through the chemokine decoy receptor D6. Gut 59,
43. Rissanen, T.T. et al. VEGF-D is the strongest angiogenic and lymphangiogenic effec- 197–206 (2010).
tor among VEGFs delivered into skeletal muscle via adenoviruses. Circ. Res. 92, 77. Gräbner, R. et al. Lymphotoxin b receptor signaling promotes tertiary lymphoid organ-
1098–1106 (2003). ogenesis in the aorta adventitia of aged Apoe-/- mice. J. Exp. Med. 206, 233–248
44. Anisimov, A. et al. Activated forms of VEGF-C and VEGF-D provide improved vascular (2009).
function in skeletal muscle. Circ. Res. 104, 1302–1312 (2009). 78. van de Pavert, S.A. & Mebius, R.E. New insights into the development of lymphoid
45. Leppänen, V.M. et al. Structural determinants of growth factor binding and specificity tissues. Nat. Rev. Immunol. 10, 664–674 (2010).
by VEGF receptor 2. Proc. Natl. Acad. Sci. USA 107, 2425–2430 (2010). 79. Muniz, L.R., Pacer, M.E., Lira, S.A. & Furtado, G.C. A critical role for dendritic cells
46. Kisko, K. et al. Structural analysis of vascular endothelial growth factor receptor-2/ in the formation of lymphatic vessels within tertiary lymphoid structures. J. Immunol.
ligand complexes by small-angle X-ray solution scattering. FASEB J. 25, 2980–2986 187, 828–834 (2011).
(2011). 80. Mounzer, R.H. et al. Lymphotoxin-a contributes to lymphangiogenesis. Blood 116,
47. Leppänen, V.M. et al. Structural determinants of vascular endothelial growth factor-D 2173–2182 (2010).
receptor binding and specificity. Blood 117, 1507–1515 (2011). 81. Harvey, N.L. The link between lymphatic function and adipose biology. Ann. NY Acad.
48. Vondenhoff, M.F. et al. LTbetaR signaling induces cytokine expression and up-regu- Sci. 1131, 82–88 (2008).
lates lymphangiogenic factors in lymph node anlagen. J. Immunol. 182, 5439–5445 82. Dixon, J.B. Lymphatic lipid transport: sewer or subway? Trends Endocrinol. Metab.
(2009). 21, 480–487 (2010).
49. Vondenhoff, M.F. et al. Lymph sacs are not required for the initiation of lymph node 83. Harvey, N.L. et al. Lymphatic vascular defects promoted by Prox1 haploinsufficiency
formation. Development 136, 29–34 (2009). cause adult-onset obesity. Nat. Genet. 37, 1072–1081 (2005).
50. Förster, R., Davalos-Misslitz, A.C. & Rot, A. CCR7 and its ligands: balancing immunity 84. Rutkowski, J.M. et al. Dermal collagen and lipid deposition correlate with tissue
and tolerance. Nat. Rev. Immunol. 8, 362–371 (2008). swelling and hydraulic conductivity in murine primary lymphedema. Am. J. Pathol.
51. Wick, N. et al. Lymphatic precollectors contain a novel, specialized subpopulation of 176, 1122–1129 (2010).
podoplanin low, CCL27-expressing lymphatic endothelial cells. Am. J. Pathol. 173, 85. Szuba, A. et al. Therapeutic lymphangiogenesis with human recombinant VEGF-C.
1202–1209 (2008). FASEB J. 16, 1985–1987 (2002).
52. Pham, T.H. et al. Lymphatic endothelial cell sphingosine kinase activity is required 86. Libby, P., Ridker, P.M. & Hansson, G.K. Inflammation in atherosclerosis: from patho-
for lymphocyte egress and lymphatic patterning. J. Exp. Med. 207, 17–27 (2010). physiology to practice. J. Am. Coll. Cardiol. 54, 2129–2138 (2009).
53. Karikoski, M. et al. Clever-1/Stabilin-1 regulates lymphocyte migration within 87. Kholová, I. et al. Lymphatic vasculature is increased in heart valves, ischaemic and
lymphatics and leukocyte entrance to sites of inflammation. Eur. J. Immunol. 39, inflamed hearts and in cholesterol-rich and calcified atherosclerotic lesions. Eur. J.
3477–3487 (2009). Clin. Invest. 41, 487–497 (2011).
54. Roozendaal, R. et al. Conduits mediate transport of low-molecular-weight antigen to 88. Nakano, T. et al. Angiogenesis and lymphangiogenesis and expression of lymphangio-
lymph node follicles. Immunity 30, 264–276 (2009). genic factors in the atherosclerotic intima of human coronary arteries. Hum. Pathol.
55. Alvarez, D., Vollmann, E.H. & von Andrian, U.H. Mechanisms and consequences of 36, 330–340 (2005).
dendritic cell migration. Immunity 29, 325–342 (2008). 89. Lim, H.Y. et al. Hypercholesterolemic mice exhibit lymphatic vessel dysfunction and
56. Phan, T.G., Green, J.A., Gray, E.E., Xu, Y. & Cyster, J.G. Immune complex relay by degeneration. Am. J. Pathol. 175, 1328–1337 (2009).
subcapsular sinus macrophages and noncognate B cells drives antibody affinity 90. Machnik, A. et al. Macrophages regulate salt-dependent volume and blood pressure
maturation. Nat. Immunol. 10, 786–793 (2009). by a vascular endothelial growth factor-C–dependent buffering mechanism. Nat.
57. Kunder, C.A. et al. Mast cell-derived particles deliver peripheral signals to remote Med. 15, 545–552 (2009).
lymph nodes. J. Exp. Med. 206, 2455–2467 (2009). 91. Wang, H.W. et al. Kaposi sarcoma herpesvirus-induced cellular reprogramming con-
58. Cohen, J.N. et al. Lymph node–resident lymphatic endothelial cells mediate periph- tributes to the lymphatic endothelial gene expression in Kaposi sarcoma. Nat. Genet.
eral tolerance via Aire-independent direct antigen presentation. J. Exp. Med. 207, 36, 687–693 (2004).
681–688 (2010). 92. Cheng, F. et al. Virus-induced Notch-MT1-MMP axis leads to lymphatic endothelial-
59. Kang, S. et al. Toll-like receptor 4 in lymphatic endothelial cells contributes to LPS- to-mesenchymal transition. Cell Host. Microbe (in the press).
induced lymphangiogenesis by chemotactic recruitment of macrophages. Blood 113, 93. Liu, R. et al. KSHV-induced notch components render endothelial and mural cell
2605–2613 (2009). characteristics and cell survival. Blood 115, 887–895 (2010).

nature medicine volume 17 | number 11 | november 2011 1379


review

94. Zhang, X. et al. Kaposi’s sarcoma–associated herpesvirus activation of vascular endo- 111. Contassot, E., Preynat-Seauve, O., French, L. & Huard, B. Lymph node tumor metas-
thelial growth factor receptor 3 alters endothelial function and enhances infection. tases: more susceptible than primary tumors to CD8+ T cell immune destruction.
J. Biol. Chem. 280, 26216–26224 (2005). Trends Immunol. 30, 569–573 (2009).
95. Tvorogov, D. et al. Effective suppression of vascular network formation by combination 112. Kerjaschki, D. et al. Lipoxygenase mediates invasion of intrametastatic lymphatic
of antibodies blocking VEGFR ligand binding and receptor dimerization. Cancer Cell vessels and propagates lymph node metastasis of human mammary carcinoma xeno-
18, 630–640 (2010). grafts in mouse. J. Clin. Invest. 121, 2000–2012 (2011).
96. Harari, S., Torre, O. & Moss, J. Lymphangioleiomyomatosis: what do we know and 113. Tammela, T. et al. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular
what are we looking for? Eur. Respir. Rev. 20, 34–44 (2011). network formation. Nature 454, 656–660 (2008).
97. McCormack, F.X. et al. Efficacy and safety of sirolimus in lymphangioleiomyomatosis. 114. Roberts, N. et al. Inhibition of VEGFR-3 activation with the antagonistic antibody
N. Engl. J. Med. 364, 1595–1606 (2011). more potently suppresses lymph node and distant metastases than inactivation of
98. Seyama, K. et al. Vascular endothelial growth factor-D is increased in serum of VEGFR-2. Cancer Res. 66, 2650–2657 (2006).
patients with lymphangioleiomyomatosis. Lymphat. Res. Biol. 4, 143–152 (2006). 115. Caunt, M. et al. Blocking neuropilin-2 function inhibits tumor cell metastasis. Cancer
99. Fukumura, D., Duda, D.G., Munn, L.L. & Jain, R.K. Tumor microvasculature and Cell 13, 331–342 (2008).
microenvironment: novel insights through intravital imaging in pre-clinical models. 116. Hooper, A.T. et al. Engraftment and reconstitution of hematopoiesis is dependent
Microcirculation 17, 206–225 (2010). on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 4,
100. Mumprecht, V. et al. In vivo imaging of inflammation- and tumor-induced lymph 263–274 (2009).
node lymphangiogenesis by immuno-positron emission tomography. Cancer Res. 70, 117. Zbytek, B. et al. Current concepts of metastasis in melanoma. Expert Rev. Dermatol.
8842–8851 (2010). 3, 569–585 (2008).
101. Leijte, J.A., van der Ploeg, I.M., Valdes Olmos, R.A., Nieweg, O.E. & Horenblas, S. 118. Tammela, T. et al. Photodynamic ablation of lymphatic vessels and intralymphatic
Visualization of tumor blockage and rerouting of lymphatic drainage in penile cancer cancer cells prevents metastasis. Sci. Transl. Med. 3, 69ra11 (2011).
patients by use of SPECT/CT. J. Nucl. Med. 50, 364–367 (2009). 119. Goyal, S., Chauhan, S.K. & Dana, R. Blockade of prolymphangiogenic vascular
102. Giuliano, A.E., et al. Association of occult metastases in sentinel lymph nodes and endothelial growth factor C in dry eye disease. Arch. Ophthalmol. published online,
bone marrow with survival among women with early-stage invasive breast cancer. doi:10.1001/archophthalmol.2011.266 (12 September 2011).
J. Am. Med. Assoc. 306, 385–393 (2011). 120. Koch, S., Tugues, S., Li, X., Gualandi, L. & Claesson-Welsh, L. Signal transduction
103. Louis-Sylvestre, C. et al. Axillary treatment in conservative management of operable by vascular endothelial growth factor receptors. Biochem. J. 437, 169–183 (2011).
breast cancer: dissection or radiotherapy? Results of a randomized study with 15 121. Saharinen, P. et al. Claudin-like protein 24 interacts with the VEGFR-2 and VEGFR-3
years of follow-up. J. Clin. Oncol. 22, 97–101 (2004). pathways and regulates lymphatic vessel development. Genes Dev. 24, 875–880
© 2011 Nature America, Inc. All rights reserved.

104. Chaffer, C.L. & Weinberg, R.A. A perspective on cancer cell metastasis. Science 331, (2010).
1559–1564 (2011). 122. Yang, Y., Xie, P., Opatowsky, Y. & Schlessinger, J. Direct contacts between extracel-
105. Sleeman, J.P., Nazarenko, I. & Thiele, W. Do all roads lead to Rome? Routes to lular membrane-proximal domains are required for VEGF receptor activation and cell
metastasis development. Int. J. Cancer 128, 2511–2526 (2011). signaling. Proc. Natl. Acad. Sci. USA 107, 1906–1911 (2010).
106. Stoecklein, N.H. & Klein, C.A. Genetic disparity between primary tumours, dissemi- 123. Kendrew, J. et al. An antibody targeted to VEGFR-2 Ig domains 4–7 inhibits VEGFR-2
nated tumour cells and manifest metastasis. Int. J. Cancer 126, 589–598 (2010). activation and VEGFR-2–dependent angiogenesis without affecting ligand binding.
107. Campbell, P.J. et al. The patterns and dynamics of genomic instability in metastatic Mol. Cancer Ther. 10, 770–783 (2011).
pancreatic cancer. Nature 467, 1109–1113 (2010). 124. Koh, Y.J. et al. Double antiangiogenic protein, DAAP, targeting VEGF-A and angio-
108. Shields, J.D., Kourtis, I.C., Tomei, A.A., Roberts, J.M. & Swartz, M.A. Induction of poietins in tumor angiogenesis, metastasis and vascular leakage. Cancer Cell 18,
lymphoidlike stroma and immune escape by tumors that express the chemokine 171–184 (2010).
CCL21. Science 328, 749–752 (2010). 125. Hashizume, H. et al. Complementary actions of inhibitors of angiopoietin-2 and VEGF
109. Kim, M. et al. CXCR4 signaling regulates metastasis of chemoresistant melanoma on tumor angiogenesis and growth. Cancer Res. 70, 2213–2223 (2010).
cells by a lymphatic metastatic niche. Cancer Res. 70, 10411–10421 (2010). 126. Brown, J.L. et al. A human monoclonal anti-ANG2 antibody leads to broad antitumor
110. Madsen, C.D. & Sahai, E. Cancer dissemination—lessons from leukocytes. Dev. Cell activity in combination with VEGF inhibitors and chemotherapy agents in preclinical
19, 13–26 (2010). models. Mol. Cancer Ther. 9, 145–156 (2010).

1380 volume 17 | number 11 | november 2011 nature medicine

You might also like