Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2021 Sep 5;42(1):576–614. doi: 10.1002/med.21855

The lymphatic vasculature: An active and dynamic player in cancer progression

Sara Rezzola 1, Elena C Sigmund 2, Cornelia Halin 2,, Roberto Ronca 1,
PMCID: PMC9291933  PMID: 34486138

Abstract

The lymphatic vasculature has been widely described and explored for its key functions in fluid homeostasis and in the organization and modulation of the immune response. Besides transporting immune cells, lymphatic vessels play relevant roles in tumor growth and tumor cell dissemination. Cancer cells that have invaded into afferent lymphatics are propagated to tumor‐draining lymph nodes (LNs), which represent an important hub for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites. In recent years many studies have reported new mechanisms by which the lymphatic vasculature affects cancer progression, ranging from induction of lymphangiogenesis to metastatic niche preconditioning or immune modulation. In this review, we provide an up‐to‐date description of lymphatic organization and function in peripheral tissues and in LNs and the changes induced to this system by tumor growth and progression. We will specifically focus on the reported interactions that occur between tumor cells and lymphatic endothelial cells (LECs), as well as on interactions between immune cells and LECs, both in the tumor microenvironment and in tumor‐draining LNs. Moreover, the most recent prognostic and therapeutic implications of lymphatics in cancer will be reported and discussed in light of the new immune‐modulatory roles that have been ascribed to LECs.

Keywords: cancer, immune modulation, lymphangiogenesis, lymphatic vessels, metastasis


Abbreviations

ANG

angiopoietin

COX

cyclooxygenase

DC

dendritic cell

DT

diphtheria toxin

ECM

extracellular matrix

FGF

fibroblast growth factor

FOXC

forkhead transcription factor

FRC

fibroblast reticular cell

HEV

high endothelial venule

HGF

hepatocyte growth factor

HIF

hypoxia‐inducible factor

IDO

indoleamine 2,3‐dioxygenase

IFP

interstitial fluid pressure

LEC

lymphatic endothelial cells

LMC

lymphatic muscle cells

LN

lymph node

lncRNA

long noncoding RNA

LVD

lymphatic vessel density

LYVE

lymphatic vessel endothelial hyaluronan receptor

MDK

midkine

MHC

major histocompatibility complex

miRNA

microRNA

NOS

nitric oxide synthase

NRP

neuropilin

OVA

ovalbumin

PDGF

platelet‐derived growth factor

PDGFR

platelet‐derived growth factor receptor

PDPN

podoplanin

PDT

photodynamic therapy

PD‐1

programmed cell death protein 1

PD‐L1

programmed death ligand 1

PROX

prospero‐related homeobox

S1P

sphingosine‐1‐phosphate

S1P1

sphingosine‐1‐phosphate receptor 1

SCS

subcapsular sinus

SEM

semaphorin

SOX

SRY‐box transcription factor

TAM

tumor associated macrophages

TGF

transforming growth factor

Treg

regulatory T cell

VCAM

vascular cell adhesion molecule

VEGF

vascular endothelial growth factor

VEGFR

vascular endothelial growth factor receptor

YAP

Yes‐associated protein

1. GENERAL STRUCTURE AND FUNCTION OF THE LYMPHATIC SYSTEM

The lymphatic system comprises an extensive network of lymphatic vessels and lymphoid organs and tissues, such as the bone marrow, the spleen, and ~500–600 lymph nodes (LNs). 1 While lymphatic vessels have originally been regarded as passive conduits for fluids and immune cells, it is nowadays clear that this highly specialized vascular network plays vital roles in controlling fluid homeostasis, immune surveillance, and lipid absorption. 2 , 3 Moreover, research on the lymphatic vascular system has been facilitated over the last two decades by the discovery of lymphatic‐specific markers (see Box 1), and in recent years lymphatic vessels have emerged as central players in disease, particularly in the context of cancer and inflammation. 4 , 5 , 6 , 7 , 8

Box 1: LEC‐specific markers.

1.

Although the expression of these molecular markers is not entirely specific to LECs, their use, in combination with pan‐endothelial markers such as VE‐cadherin or PECAM‐1 (CD31), has enabled the unambiguous molecular distinction between blood and lymphatic vasculature in tissues. LEC markers include the transcription factor prospero‐related homeobox 1 (PROX1), which acts as a master regulator factor of LEC identity (see Box 2), 9 , 10 the tyrosine kinase vascular endothelial growth factor (VEGF) receptor 3 (VEGFR3), that binds to the lymphatic growth factor VEGF‐C, 11 , 12 and the mucin‐type glycoprotein podoplanin (PDPN). 13 LECs of initial lymphatic capillaries can be further distinguished from lymphatic collectors by high expression of the lymphatic vessel endothelial hyaluronan receptor 1 (LYVE‐1) 14 , 15 and by high expression of the chemokine (C‐C motif) ligand 21 (CCL21). 16 The latter forms a perilymphatic gradient that attracts CCR7‐expressing dendritic cells and other leukocytes into lymphatic capillaries. 17

Different from the blood vasculature, which is organized in a closed, circular system, the lymphatic vascular system consists of an open, hierarchically organized vascular network, which begins in virtually all vascularized tissues of the body (see Box 2 for information on its developmental origin). Lymphatic vessels initiate in the peripheral tissue as blind‐ended lymphatic capillaries that merge into larger collecting vessels and eventually deliver their content, that is, the lymph, into the bloodstream (Figure 1). Generally, lymphatic vessels that lead toward an LN are designated as afferent lymphatic vessels, while lymphatic collectors leading away from an LN are designated as efferent lymphatic vessels. 18 Considering that LNs are arranged in a sequential manner along the lymphatic vasculature, efferent lymphatic vessels can simultaneously be afferent lymphatic vessels for downstream LNs.

Box 2: Lymphatic vasculature development.

1.

The formation of the lymphatic vasculature occurs in a similar manner in all vertebrates during embryonic development. Lineage tracing experiments suggest that a substantial part of the lymphatic vasculature originates from differentiation of lymphatic progenitor cells from embryonic veins. 19 , 20 However, in recent years, more evidence in support of alternative origins of tissue‐specific lymphatic vessels has emerged. 21 Specifically, in the mesentery and skin, non‐venous single progenitor cells expressing vascular endothelial growth factor (VEGF) receptor 3 (VEGFR3), prospero‐related homeobox 1 (PROX1), and neuropilin 2 (NRP2) were identified to contribute to the formation of these local tissue‐specific lymphatic vessel networks. 22 , 23

In mice, lymphatic development starts around embryonic day (E)9‐9.5, after the blood vascular circulation is established. At this time‐point, SRY‐box transcription factor 18 (Sox18) and orphan nuclear receptor chicken ovalbumin upstream promoter transcription factor (COUP‐TFII) act in concert to induce the expression of PROX1 in a subset of endothelial cells in the wall of the cardinal vein. 19 , 24 , 25 PROX‐1, as a master transcription factor of lymphatic identity, in turn, downregulates blood vessel marker genes and drives expression of VEGFR3 in the lymphatic progenitor cells. 26 PROX1+ progenitor lymphatic endothelial cells (LECs) next elongate, bud off from the cardinal and intersomitic veins around E10, 27 , 28 and start migrating away from the cardinal vein, in a process dependent on a mesodermal cell‐derived gradient of VEGF‐C. Subsequently, LECs upregulate other LEC‐specific markers, such as PDPN 29 and various adhesion molecules that are required to interconnect with other migrating LECs. Around E11.5, progenitor LECs migrate alongside the anterior and posterior axes of the embryo in capillary structures that eventually assemble to form the lymphatic sacs. 27 , 28 At this stage, initial lymphatic vessels start to form and diverge into two types of lymphatic vessels: capillary lymphatic vessels and collecting lymphatic vessels. Analyzing the development of lymphatic collectors in the mesentery, forkhead transcription factor 2 (Foxc2) was shown to be particularly important for the formation and maturation of lymphatic collectors. 30 Specifically, small clusters of cells in newly formed collectors start to upregulate Prox1 and Foxc2 expression around E16, thus initiating lymphatic valve formation via mechanosensitive Connexin37 (Cx37) and calcineurin/NFAT signalling. 31

Figure 1.

Figure 1

Structure of the lymphatic network. (A) The lymphatic network is made up of vessels that originate in peripheral tissues and run through lymph nodes (LNs). Lymphatic vessels eventually converge in the central body region into two lymph ducts (i.e., the thoracic duct and the right lymph duct), which fuse with the blood circulation at the level of the subclavian veins. (B) In peripheral tissues like the skin, lymphatic vessels begin as blind‐ended capillaries, which merge into collecting vessels. Capillaries (upper insert) are surrounded by a thin basement membrane and contain lymphatic endothelial cells (LECs) connected by button‐like junctions. This arrangement generates open flaps, which represent the main entry point for leukocytes. Flap opening is regulated by filaments that connect the flap with the basement membrane. Collecting vessels (lower insert) have a thicker basement membrane and are surrounded by contractile lymphatic muscle cells. LECs forming the collector wall are tightly joined by continuous, zipper‐like cell–cell junctions. Moreover, collectors contain valves to facilitate fluid propagation. Leukocytes within collecting vessels are passively transported with the lymph flow. (C) At the level of the LN, afferent lymphatic collectors connect with the LN subcapsular sinus. The latter surrounds the entire LN parenchyma and is the site of entry for leukocytes arriving via afferent lymphatics. The LN parenchyma is divided into the outer cortex containing the B cell follicles, an inner paracortex containing the T cell area, and HEVs as well as the medulla. The entire parenchyma is interspersed by a network of trabecular and cortical sinuses, which fuse with the medullary sinus located in the region, where the efferent lymphatic vessel exits the LN. Leukocytes exiting from the LN transmigrate through cortical and medullary sinuses to access the efferent lymphatic vessel [Color figure can be viewed at wileyonlinelibrary.com]

1.1. Lymphatic vessels in peripheral tissues

In comparison to capillaries of blood vessels, lymphatic capillaries are larger in diameter (approximately 50–60 µm, as compared to the 5–10 µm wide blood capillaries) 32 , 33 and consist of oak leaf‐shaped lymphatic endothelial cells (LECs), which are interconnected by discontinuous, button‐like junctions. 33 Capillary LECs are loosely connected by anchoring filaments to a discontinuous basement membrane 34 —a setup that prevents lymphatic capillaries from collapsing when tissue fluid pressure increases, for example, during an inflammatory response (Figure 1B). Rather, once the interstitial pressure increases, this causes the opening of the lymphatic “flaps,” which represent gaps of 2–3 µm between overlapping LECs. This, in turn, allows the entry of all components of lymph, that is, interstitial fluid, immune cells, and macromolecules, into the capillary lumen. Capillaries first merge into pre‐collecting vessels, an intermediate vessel type with characteristics of both capillaries and collectors, 35 and subsequently into collectors, which represent the second main type of lymphatic vessels in the body.

Lymphatic collectors transport lymph collected from capillaries and pre‐collectors through a series of sequential LNs, allowing the lymph to eventually be passaged back toward the blood circulation (Figure 1A–C). Different from LECs in capillaries, LECs in lymphatic collectors have an elongated shape and are tightly connected by continuous zipper‐like junctions. Moreover, collecting vessels are surrounded by a continuous basement membrane to prevent leakage of lymph back into the interstitium. 33 , 36 In addition, lymphatic collectors are lined by lymphatic muscle cells (LMCs) that undergo phasic contractions to generate lymphatic flow and propagate lymph. 37 , 38 To ensure unidirectional flow upon contraction, lymphatic collectors contain intraluminal valves that separate vessels into sequential segments, so‐called lymphangions, and prevent backflow from one segment into the previous one. 39 The last steps in the lymph's journey toward the blood vascular system occur through the largest lymphatic vessels in the body, namely the thoracic and lymphatic ducts, which join the blood vascular system at the level of the subclavian veins. 21 , 40

While the above‐described structure of the lymphatic network is characteristic for lymphatics present in the skin 36 or, for example, in the trachea, 33 there are clear differences in the architecture of the lymphatic vessel network in other organs. For example, the lung, 41 the intestine, 42 , 43 the heart, 4 , 44 , 45 and the central nervous system 46 , 47 have lymphatic networks with unique structures and functions. New insights into organ‐specific networks have recently been extensively reviewed elsewhere. 21 , 46 , 48

1.2. Organization of the lymph node and its lymphatic sinus system

LNs provide a confined and highly specialized environment for the initiation of an adaptive immune response, continuous immunosurveillance, and propagation of immune tolerance. LNs can be structurally divided into a cortex, a paracortex, and a medulla. Immune cells derived from peripheral tissues (mostly antigen‐presenting dendritic cells (DCs) and antigen‐experienced T cells), together with soluble antigens and immune mediators reach the LN via afferent lymphatic vessels (Figure 1C). 9 The latter enter into the subcapsular sinus (SCS), which is found beneath the collagen‐rich LN capsule and forms part of a complex sinus system surrounding the LN parenchyma (Figure 1C). The SCS overlays the LN cortex, that is, the outer region in the LN parenchyma, which harbors B cell follicles and interfollicular T cell zones. To enter from the SCS lumen into the LN parenchyma, DCs and activated T cells require the CCR7 chemokine receptor to migrate into the CCL21‐rich T cell zone. 10 By contrast, naive T cells gain access to the LN parenchyma via the medullary sinus system. 10 Besides CCL21, certain DCs reportedly also require the chemokine CCL8 for exit from the SCS. 49 In comparison to the cortex, the paracortex is more deeply located within the LN parenchyma and contains the T cell zones and high endothelial venules (HEVs). The latter represent the entry points for the majority of T and B cells that enter the LN from the blood circulation. 50 Besides a specialized vascular network, the LN also contains a so‐called conduit system formed by extracellular matrix (ECM) components that are ensheathed by fibroblastic reticular cells (FRCs). The conduit system starts from the SCS and serves to transport low‐molecular‐weight macromolecules, including antigens, but also for antibodies (IgM) and virions between different LN compartments. 51 , 52 , 53 The organization of the LN into specialized compartments allows antigen‐presenting cells, tissue‐derived antigens, and soluble immune mediators to efficiently come into close contact with naïve B and T lymphocytes and recirculating central memory T cells. 54 After a few hours of scanning antigen‐presenting cells in the T cell zone for the presence of cognate antigen, most T cells will exit the LN again by exiting via the efferent lymphatic vessel. By contrast, if a T cell encounters its cognate antigen on an antigen‐presenting cell, it will become activated, proliferate, and differentiate, to give rise to millions of antigen‐specific effector cells, which after several days will also leave the LN. 50 , 55 , 56 , 57 Both naïve and antigen‐specific lymphocytes exit the LN parenchyma through blind‐ended lymphatic cortical sinuses that emerge adjacent to HEVs in the paracortex. The cortical sinuses merge into medullary sinuses, which further connect with the efferent lymphatic vessel leaving the LN.

1.2.1. Lymphatic endothelial cells of the lymph node

LECs present in LNs have several unique properties in comparison to LECs of peripheral lymphatic vessels. According to recent single‐cell RNA sequencing studies, murine and human LN LECs cluster into different subtypes with differential gene expression: ceiling LECs, floor LECs, (para‐) cortical and medullary LECs. 58 , 59 , 60 , 61 Ceiling LECs and floor LECs line and form the SCS. To facilitate DC and T cell entry, ceiling LECs express the atypical chemokine receptor 4 (ACKR4), a scavenging receptor for the chemokine CCL21 expressed by floor LECs. The resulting CCL21 gradient guides the transmigration of DCs across the SCS to enter the LN parenchyma. 62 Floor LECs are interspersed by a layer of CD169+ macrophages and a few sinus‐resident DCs that capture soluble antigens. 63 , 64 Moreover, floor LECs were shown to directly transcytose macromolecules, such as subcutaneously administered IgG and IgA antibodies, 65 to the LN parenchyma. In addition, floor LECs express plasmalemma vesicle‐associated protein (PLVAP), which forms permeable diaphragm‐like filters (fenestrae) both on their luminal and abluminal side and controls the entry of macromolecules into the LN conduit system. 66 Generally, sinusoidal LN LECs exhibit a high endocytic capacity 67 and can directly present antigens to immune cells. 68 , 69 LN LECs express major histocompatibility complex (MHC) Class I and II molecules, 70 , 71 and further MHC‐II molecules can be acquired from migratory DCs. 72 However, the expression of the typical costimulatory molecules (e.g., B7 family), which are needed for T cell priming, is low or absent in LN LECs in steady state. 68 , 70 , 73 Instead, LN LECs reportedly express co‐inhibitory molecules including programmed death‐ligand 1 (PD‐L1; Figure 2). 70 , 73 Medullary and cortical LECs were additionally shown to contribute to clonal deletion of autoreactive CD8+ T cells by expressing self‐antigens such as melanocyte‐specific tyrosinase, intestinal epithelial protein A33, and intestinal or pancreatic polypeptide (PPY) in an autoimmune regulator (AIRE)‐independent manner. 71 Besides affecting CD8+ T cells, LN LECs have also been implicated in the induction CD4+ T cell anergy. Specifically, LECs were shown to transfer endogenous antigens to anergy‐inducing DCs, 74 or to directly induce CD4+ T cell anergy by peptide‐loaded MHC‐II complexes that they acquired from DCs. 72 A recent study by Nadafi et al. 75 further demonstrated that LN stromal cells, including LECs, can generate antigen‐specific regulatory T cells (Tregs) by conversion of naive CD4+ T cells into Tregs through MHC‐II expression and self‐antigen presentation. 75  Taken together, these observations suggest that LN LECs rather promote tolerance than immune‐stimulatory reactions in steady‐state conditions. On the other hand, a recent study demonstrated that antigen presentation by LN LECs can selectively induce the generation of long‐lived antigen‐specific CD8+ memory T cells that have the ability to rapidly differentiate into effector T cells upon rechallenge. 76 In addition to their direct involvement in antigen presentation, proliferating SCS LECs have been shown to simply capture and “archive” viral antigen during viral infections. Although these antigens were not directly presented to CD8+ T cells by LECs, they persisted in LECs for subsequent transfer to antigen‐presenting cells. In this way, LECs were shown to enhance the effector functions and protective capacity of circulating memory CD8+ T cells. 77 , 78 Finally, LN LECs have also been shown to actively control the egress of lymphocytes from the LN. LN LECs produce sphingosine‐1‐phosphate (S1P), 79 a chemotactic sphingolipid that promotes lymphocyte egress from the LN by binding to S1P receptor 1 (S1P1) expressed on exiting T cells. 80 Mechanisms that determine lymphocyte retention in the LN parenchyma include the downregulation or internalization of S1P1 in a CD69‐dependent manner after MHC molecule engagement and activation. 81 , 82

Figure 2.

Figure 2

Roles of LECs in immune modulation of CD4+ and CD8+ T cells. Antigen presentation of CD4+ and CD8+ T cells in absence of costimulation, combined with programmed cell death protein 1 (PD‐1)/PDL‐1 signaling can have various outcomes on T cell fate and function. Immunosuppressive signals are further enhanced by inducible nitric oxide synthase (NOS)‐ and indoleamine 2,3‐dioxygenase (IDO)‐mediated production of NO and tryptophan metabolites. Of note: with the exception of the PD‐1/PD‐L1 interaction, the depicted functions have primarily been studied in LN LECs 83 , 84 [Color figure can be viewed at wileyonlinelibrary.com]

2. MEDIATORS OF TUMOR‐ASSOCIATED LYMPHANGIOGENESIS

Historically considered as a passive route for tumor cell dissemination, experimental evidence, and clinical studies have shown that the lymphatic vasculature plays an active role in primary tumor dissemination and in the metastasis process (Figure 3). Similar to angiogenesis, lymphangiogenesis is a multistep process in which activated LECs proliferate and migrate in response to specific stimuli to form new vessels. Lymphangiogenesis involves the interaction of different cell types and ECM components and is regulated by a variety of lymphangiogenic growth factors and other mediators, including cell adhesion molecules and noncoding RNAs. The most important factors supporting tumor‐associated lymphangiogenesis will be discussed in the following.

Figure 3.

Figure 3

Schematic illustration of the principal routes of lymphatic metastasis and roles of lymphatic vessels in tumor progression. Lymphangiogenesis is induced by the primary tumor at the site of tumor growth and in draining LNs. Lymphangiogenesis and remodeling in the primary tumor create more surface area for tumor cell–LEC interaction and facilitates lymphatic spread. In the draining LN, it prepares the niche for subsequent colonization with tumor cells. Metastatic foci can occasionally form in transit or—more frequently—in draining LNs. From the LN, metastatic cells can further spread by accessing the systemic circulation via nodal blood vessels, or by entering into efferent lymphatics. The latter may lead to metastasis in subsequent draining LNs and eventually also allows for systemic spread. Besides tumor cells, also antigen‐presenting DCs, T cells, and other leukocytes use lymphatics to migrate to the tumor‐draining LNs, where antitumor immunity may be induced. Lymphatic endothelial cells within both the tumor and in tumor‐draining LNs actively participate in immune modulation, especially in the dampening of antitumor immunity [Color figure can be viewed at wileyonlinelibrary.com]

2.1. Soluble mediators

Tumor‐associated lymphangiogenesis depends on the presence of various soluble mediators (Table 1) mainly produced by cancer cells themselves, but also by macrophages, mast cells, T cells, activated platelets, as well as stromal cells within the tumor microenvironment. 85 , 86 , 87 , 88 , 89 , 90 A selection of the most important soluble lymphangiogenic mediators is provided below.

Table 1.

Soluble mediators involved in tumor lymphangiogenesis

Soluble mediator Category Role References
ACTIVIN A Growth factor Anti‐lymphangiogenic [91]
AM Hormone Pro‐lymphangiogenic [92]
ANGs Growth factor Pro‐lymphangiogenic [ 93 , 94 ]
EGF Growth factor Pro‐lymphangiogenic [95]
EPO Growth factor Pro‐lymphangiogenic [96]
FGF2 Growth factor Pro‐lymphangiogenic [ 97 , 98 ]
HGF Growth factor Pro‐lymphangiogenic [ 99 , 100 , 101 ]
IGF Growth factor Pro‐lymphangiogenic [ 102 , 103 ]
IL6 Cytokine Pro‐lymphangiogenic [ 104 , 105 ]
MDK Growth factor Pro‐lymphangiogenic [106]
OPN Matrix protein Pro‐lymphangiogenic [107]
PDGFs Growth factor Pro‐lymphangiogenic [ 108 , 109 , 110 , 111 ]
S1P Sphingolipid Pro‐lymphangiogenic [ 112 , 113 ]
SLIT2 Matrix protein Pro‐lymphangiogenic [114]
TGFβ Growth factor Pro‐lymphangiogenic [ 115 , 116 , 117 ]
Anti‐lymphangiogenic [ 118 , 119 ]
TNF‐α Cytokine Pro‐lymphangiogenic [ 120 , 121 ]
VEGF‐C Growth factor Pro‐lymphangiogenic [ 122 , 123 , 124 , 125 ]
VEGF‐D Growth factor Pro‐lymphangiogenic [ 126 , 127 ]
WNT1 Secreted glycoprotein Anti‐lymphangiogenic [128]
WNT5B Secreted glycoprotein Pro‐lymphangiogenic [129]

Abbreviations: AM, adrenomedullin; ANG, angiopoietin; EGF, epidermal growth factor; EPO, erythropoietin; FGF2, basic fibroblast growth factor; HGF, hepatocyte growth factor; IGF, insulin‐like growth factor; IL, interleukin; MDK, midkine; OPN, osteopontin; PDGF, platelet‐derived growth factor; S1P, sphingosine 1 phosphate; TGFβ, transforming growth factor β; TNF‐α, tumor necrosis factor α; VEGF, vascular endothelial growth factor; WNT, wingless‐type MMTV integration site family.

2.1.1. Vascular endothelial growth factors

Similar to physiological conditions, the major recognized drivers of tumor lymphangiogenesis are vascular endothelial growth factor (VEGF)‐C and VEGF‐D. Both mainly act through the tyrosine kinase VEGF receptor 3 (VEGFR3) in association with its coreceptor neuropilin 2 (NRP2), which are both expressed on the surface of LECs (see 87, 89, 90 and references therein). VEGF‐C and VEGF‐D are produced as pre‐pro‐polypeptides and undergo a step‐by‐step process of proteolysis to achieve their mature/active structure. 130 Once VEGF‐C and VEGF‐D bind to VEGFR3, this activates protein kinase C/ERK signaling and triggers AKT phosphorylation, thereby promoting LEC migration and proliferation. 131 When assessed in murine experimental models, tumor overexpression of VEGF‐C, VEGF‐D, VEGFR3, and/or NRP2 correlated with increased lymphatic growth and tumor cell metastases to LNs and distant organs. 122 , 123 , 126 , 132 , 133 , 134 , 135 Accordingly, blockade of VEGF/VEGFR signaling using neutralizing anti‐VEGFR3 or anti‐NRP2 monoclonal antibodies, soluble forms of VEGFR3, inhibitors of the receptor kinase activity, or sVEGFR3 gene therapy has been shown to reduce lymphangiogenesis in the primary tumors as well as metastasis to LNs and distant sites. 87 , 90 , 136 , 137

Several mediators have been described to upregulate VEGF‐C and VEGF‐D expression within the tumor microenvironment (see 85 and references therein). A pivotal role in modulating the expression of lymphangiogenic VEGFs is played by the hypoxic tumor environment. 138 Indeed, hypoxia‐inducible factor 1α (HIF‐1α) induces the expression of several growth factors and other molecules stimulating LEC activation, proliferation, and migration. Accordingly, high HIF‐1α levels have been found to correlate with VEGF‐C expression and increased peritumoral lymphangiogenesis in several types of cancers (see 139 and references therein). In addition, the distribution and the expression of lymphangiogenic factors within the tumor mass may be orchestrated by interstitial flow, a transport mechanism that plays a pivotal role in organizing the development of new lymphatic vessels. 140 , 141 Indeed, it has been shown that interstitial fluid flow influenced VEGF‐C spatial and temporal distribution promoting lymphangiogenesis not only inside the tumor mass, but also in the peritumoral normal tissue. 141 , 142 , 143 Accordingly, results obtained in vitro in a model of osteosarcoma cells cultured under high‐pressure conditions revealed an increased expression of VEGF‐C, thus suggesting that fluid flow may directly modulate the production of lymphangiogenic factors. 144

Thus far, VEGF‐C and/or VEGF‐D levels were found upregulated in several human tumors (see 85, 90 and references therein). Moreover, in the majority of clinical studies high VEGF‐C and VEGF‐D expression was found to correlate with increased tumoral lymphangiogenesis, lymphatic metastasis, and reduced patient survival. 90 , 122 , 124 , 145 , 146 , 147 Other studies, however, did not confirm this correlation, 148 , 149 which suggests that additional biological mechanisms may be involved in the regulation of lymphatic metastasis, such as the presence of other growth factors able to directly modulate lymphangiogenesis. For example, VEGF‐A, one of the main mediators involved in blood vessel formation and angiogenesis, also promotes the proliferation and migration of LECs in vitro. 150 , 151 In a cutaneous squamous cell carcinoma model, overexpression of VEGF‐A was found to strongly induce tumor lymphangiogenesis and LN metastasis. 152 Accordingly, inhibition of VEGF‐A/VEGFR1 and VEGF‐A/VEGFR2 signaling pathways with antiangiogenic compounds inhibited VEGF‐A‐mediated lymphangiogenesis and sentinel LN metastasis in xenograft mouse models. 153 , 154

2.1.2. Platelet‐derived growth factor

Platelet‐derived growth factor B (PDGF‐BB) directly stimulates lymphangiogenesis and vessel remodeling through the binding to PDGF receptor α (PDGFRα) and PDGFRβ 108 , 109 expressed on LEC surface. Even though in human cancers PDGF‐BB overexpression is often associated with VEGF‐C upregulation, 110 PDGF‐BB activity on LECs seems to be independent of VEGF‐C signaling, since treatment with an anti‐VEGFR3 antibody did not affect its pro‐lymphangiogenic activity. 108 , 111 Interestingly, surface expression of PDGFRβ by LECs is dependent on the transcription factor prospero‐related homeobox 1 (PROX1). 155

2.1.3. Hepatocyte growth factor

Hepatocyte growth factor (HGF) may stimulate lymphangiogenesis either directly or through indirect mechanisms of action. HGF was found to promote lymphangiogenesis in vivo 99 , 100 by interacting with its cognate receptor c‐Met expressed in tumor‐associated lymphatics. 156 In line with this evidence, it has been recently reported that the pro‐lymphangiogenic SRY‐box transcription factor 18 (SOX18) 157 promoted tumor progression by activating HGF and epidermal growth factor pathways in a clear cell renal cell carcinoma mouse model. 158 On the other hand, HGF may indirectly exert its lymphangiogenic activity by cooperating with VEGF‐C 159 or via induction of VEGF‐C/VEGF‐D expression. 101

2.1.4. Fibroblast growth factor

The pro‐tumoral/proangiogenic basic fibroblast growth factor (bFGF/FGF2) 160 modulates lymphangiogenesis by binding to FGF receptor 3 (FGFR3) expressed by LECs. 161 The interaction between FGF2 and the lymphatic vessel endothelial hyaluronan receptor 1 (LYVE‐1) on the LEC surface appears to play a fundamental role in mediating FGF2‐dependent lymphangiogenic activity, possibly by participating in FGF2 internalization. 97 Relevant to tumor progression, blockade of FGF2 by a neutralizing antibody reduced the density of lymphatic vessels in a xenograft model of lung cancer. 162 In addition to the direct stimulation of LEC‐expressed FGFR3 described above, FGF2 may also cooperate with VEGF‐C and foster tumor lymphangiogenesis and metastasis through FGFR1/VEGFR3‐dependent pathways. 98

2.1.5. Angiopoietins

Angiopoietins (ANGs) participate in lymphangiogenesis through binding to LEC‐expressed TIE receptors. 163 Overexpression of ANG1 and ANG2 in malignant pancreatic β cells was found to result in augmented lymphatic vasculature in the tumor mass. 93 Moreover, inhibition of ANG2 impaired tumor lymphangiogenesis and affected LN and lung metastasis formation in human lung carcinoma xenograft mouse models. 164 These results are in agreement with clinical evidence showing that in patients affected by papillary thyroid carcinoma ANG1 overexpression correlated with lymphovascular invasion and LN metastasis. 94 Notwithstanding, ANG1 expression has been found to correlate with decreased risk of LN metastasis in early stage cervical cancer patients, 165 suggesting that ANG signaling may exert different functions in different tumor settings.

2.1.6. Transforming growth factor β

The effect of transforming growth factor β (TGFβ) on tumor lymphangiogenesis is still controversial. Depending on the experimental tumor model and the involvement of other mediators, TGFβ may exert both pro‐ or anti‐lymphangiogenic activities. For instance, TGFβ increased the expression of VEGF‐C by cooperating with sine oculis homeobox homolog 1 (SIX1), a transcription factor whose overexpression correlates with poor clinical prognosis in numerous malignancies, 115 and enhanced the expression of lymphangiogenic genes in KRAS‐mutated pancreatic carcinoma models. 116 Recent work by Evans et al. 117 on a triple‐negative breast cancer model found that TGFβ1 produced and released by tumor‐associated macrophages (TAMs) adherent to LECs activated RhoA cascade signaling in LECs, inducing cell contraction in vitro and increasing permeability in vivo. Conversely, TGFβ may also exert inhibitory effects on the lymphatic vasculature. Specifically, TGFβ was found to inhibit the expression of collagen and calcium‐binding EGF domain‐1 (CCBE1), 118 a molecule involved in the proteolytic activation and maturation of VEGF‐C. 166 Accordingly, it has been shown that the blockade of endogenous TGFβ signaling resulted in enhanced VEGF‐C induced lymphangiogenesis in xenograft mouse models. 119

2.1.7. S1P

S1P is a sphingolipid involved in various physiological and pathological conditions. 112 During cancer progression, S1P reportedly stimulates tumor cell migration and invasion, inflammatory cell recruitment, angiogenesis, and lymphangiogenesis. 167 In accordance, inhibition of S1P production was found to reduce lymphangiogenesis in a murine model of breast cancer. 113 Even though S1P alone is not able to stimulate LEC sprouting in vitro, the combination of S1P, FGF2, and VEGF‐A was found to induce a strong lymphangiogenic response, which could be exploited for the screening of inhibitory compounds in vitro and in vivo. 168

2.2. Other mediators

In addition to the above‐described soluble mediators, also other molecules, such as adhesion molecules, microRNAs (miRNAs), and long noncoding RNAs (lncRNAs), have been reported to play a relevant role in tumor lymphangiogenesis.

2.2.1. Integrins

Several integrins have been described to orchestrate angiogenesis during cancer progression. 169 , 170 Comparably little is known about how integrins participate in tumor lymphangiogenesis, the majority of reports focusing on the role of β1 integrin (also known as CD29). 170 For instance, activation of β1 integrin signaling by semaphorin 7a (SEM7A), a glycophosphatidylinositol membrane‐anchored protein frequently overexpressed in mammary tumors, stimulated lymphangiogenesis and tumor cell spreading in a breast cancer model. 171 In addition, α4β1 integrin was found to be upregulated in spontaneous and experimental tumor‐associated LECs, where its activity was crucial for VEGF‐C‐mediated lymphangiogenic stimulation both in vitro and in vivo. 172 Accordingly, antagonists of α4β1 were shown to inhibit tumor lymphangiogenesis and LN metastasis formation in experimental murine tumor models. 172 Finally, β1 integrin expression by the mucin‐type glycoprotein podoplanin (PDPN)+ TAMs was recently found to play a key role in mediating TAM chemotactic migration and adhesion to LECs, thereby contributing to TAM‐mediated lymphangiogenesis. 173 Along this line, a similar role has also been ascribed to LEC‐expressed integrin β4, which was shown to retain a subpopulation of TAMs on the LEC surface, thereby supporting the growth of tumor lymphatic vessels. 117

2.2.2. Other adhesion molecules

Besides integrins, other cell adhesion molecules reportedly play a role in tumor lymphangiogenesis. For instance, loss of neural cell adhesion molecule (CD56) in a transgenic mouse model of pancreatic β cell carcinogenesis was associated with VEGF‐C and VEGF‐D upregulation and with increased tumor lymphangiogenesis and LN metastasis. 174 A further example is melanoma cell adhesion molecule (CD146), a transmembrane protein upregulated in almost all cancer types. CD146 was found to function as cell surface receptor for a variety of different ligands, including VEGF‐C. VEGF‐C‐mediated activation of CD146 resulted in lymphatic vessel sprouting via p38 and ERK pathways. 175 In addition, CD146 can interact also with PDGFRβ, contributing to vessel stabilization (as reviewed in 176).

2.2.3. Noncoding RNAs

In the last years, considerable attention has been paid to miRNA and lncRNA role in tumor progression, including their impact on lymphangiogenesis and lymphatic metastasis. Several miRNAs/lncRNAs have been found to be expressed in LECs and to promote or inhibit lymphatic vessel formation in different types of cancer (recently reviewed in 177). A detailed list of these miRNAs and lncRNAs, and their roles in tumor lymphangiogenesis is included in Table 2 and Table 3, respectively.

Table 2.

miRNAs involved in tumor lymphangiogenesis

miRNA miRNA tumor levels Lymphangiogenesis‐related miRNA target Tumor model References
miR‐182‐5p Downregulated VEGF‐C Colon cancer [178]
miR‐186 Chondrosarcoma [179]
miR‐624‐3p [180]
miR‐381 [181]
miR‐27b [ 182 , 183 ]
miR‐507 [184]
miR‐195‐3p Oral squamous cell carcinoma [185]
miR‐300 [186]
miR‐128 Non‐small cell lung cancer [187]
miR‐206 Downregulated KRAS Pancreatic ductal adenocarcinoma [188]
ANXA2
VEGF‐C
miR‐503‐5p Downregulated VEGF‐A Colon cancer [189]
miR‐126 Oral squamous cell carcinoma [190]
miR‐4306 Downregulated SIX1 Breast cancer [191]
CDC42
VEGF‐A
miR‐486‐5p Downregulated NRP2 Colorectal carcinoma [192]
miR‐93 Downregulated ANG2 Lung adenocarcinoma [193]
miR‐129‐5p Downregulated ZIC2 Nasopharyngeal carcinoma [194]
miR‐7 Downregulated NF‐κB Gastric cancer [195]
miR‐526b Upregulated CPEB2A Breast cancer [145]
miR‐655 PTEN
miR‐19a Upregulated TSP‐1 Colorectal cancer [196]
miR‐155 Upregulated BRG1 Natural Killer/T cell lymphoma [197]
miR‐221‐3p Upregulated VASH1 Cervical squamous cell carcinoma [198]
miR‐548k Upregulated ADAMTS1 Esophageal squamous cell carcinoma [199]

Abbreviations: ADAMTS1, a disintegrin and metalloproteinase with thrombospondin motifs 1; ANG2, angiopoietin 2; ANXA2, annexin A2; BRG1, Brahma‐related gene 1; CDC42, cell division control protein 42; CPEB2A, cytosolic polyadenylation element‐binding 2A; HIF‐2α, hypoxia‐induced factor 2α; KRAS, Kirsten rat sarcoma virus; NF‐κB, nuclear factor kappa‐light‐chain‐enhancer of activated B cells; NRP2, neuropilin 2; PTEN, phosphatase and tensin homolog; SIX1, sineoculis homeobox homolog 1; TSP‐1, thrombospondin‐1; VASH1, vasohibin‐1; VEGF, vascular endothelial growth factor; ZIC2, zinc finger protein ZIC2.

Table 3.

lncRNAs involved in tumor lymphangiogenesis

lncRNA lncRNA tumor levels Effect on cancer lymphangiogenesis Tumor model References
ANRIL Upregulated Increased lymphangiogenesis Colorectal cancer [200]
ASLNC07322 Upregulated Sponging of miR‐128‐3p and upregulation of VEGF‐C Metastatic colon cancer [201]
BLACAT2 Upregulated Upregulation of VEGF‐C Bladder cancer [202]
HNF1A‐AS1 Upregulated Sponging of miR‐30b‐3p and upregulation of PI3K/AKT signaling pathways Gastric cancer [203]
HUMT Upregulated FOXK1 activation and upregulation of VEGF‐C Triple‐negative breast cancer [204]
LNMAT1 Upregulated Upregulation of MCP1 and macrophage recruitment Bladder cancer [205]
LNMAT2 Upregulated Upregulation of PROX1 Bladder cancer [206]

Abbreviations: AKT, protein kinase B; ANRIL, antisense noncoding RNA in the INK4 locus; BLACAT2, bladder cancer‐associated transcript 2; FOXK1, forkhead box K1; HNF1A‐AS1, hepatocyte nuclear factor 1 homeobox A – antisense RNA 1; HUMT, highly upregulated in metastatic triple‐negative breast cancer; LNMAT1, lymph node metastasis associated transcript 1; LNMAT2, lymph node metastasis associated transcript 2; MCP1, monocyte chemoattractant protein 1; PI3K, phosphoinositide 3‐kinase; VEGF, vascular endothelial growth factor.

3. LYMPHATICS IN THE PRIMARY TUMOR

In the early stages of tumor progression, the lymphatic network undergoes active modifications, including the formation of novel lymphatics from pre‐existing ones, both inside and around the tumor mass, and the dilation of vessel lumen, thus facilitating cancer cell entrance into the lymphatic circulation. 7 , 90 These changes contribute to a substantial remodeling of vessels, resulting in an increased flow rate, and thus also facilitate the spreading of cancer cells to tumor‐draining LNs. 90 , 207 , 208 In addition, emerging pieces of literature underline that newly formed lymphatic vessels play key, and sometimes contradictory roles in the anti‐ and pro‐tumor immune responses taking place in the tumor microenvironment. These roles will be introduced in the following.

3.1. Cancer cell‐lymphatic interplay in the primary tumor

In response to the lymphangiogenic stimuli produced during tumor growth, lymphatics undergo a dynamic process of vascular remodeling (Figure 3). There are few transcriptional studies that have thus far been published on tumor‐derived lymphatic collectors, 207 tumor‐associated LECs expanded in vitro, 209 and recently on LECs directly isolated from the primary tumor of an orthotopic triple‐negative breast cancer. 210  Taken together, these studies revealed the modulation of genes and pathways involved in cell migration, inflammation, and immune‐related mechanisms in tumor LECs when compared to control LECs. These findings strongly suggest that cancer lymphatic vessels possess a specific molecular profile reflecting the plasticity of LECs and their capacity to adapt in response to tumor microenvironment. The exact mechanisms underlying the lymphatic invasion of tumor cells have not been fully elucidated. The interplay between tumor cells and the lymphatic system occurs at different levels as outlined in the following.

3.1.1. Tumor cell attraction and entrance into lymphatics

Even though the exact mechanisms of cancer cell entrance and dissemination through lymphatics have not yet been fully elucidated, the capacity of tumor cells to exploit immune‐specific chemokine signaling pathways has become evident (recently reviewed in 88). For example, the CCL21/CCR7 signaling, that normally mediates the trafficking of immune cells through CCL21+ lymphatic vessels, has been described to promote lymphogenous spreading of CCR7+ cancer cells. 211 , 212 CCL1, on the other hand, appears to be primarily expressed by SCS LECs and to specifically facilitate the entry of CCR8+ metastatic tumor cells to the tumor‐draining LN. Its receptor CCR8 is expressed in human and murine metastatic melanoma. In murine melanoma, treatment with a CCR8 antagonist was able to reduce LN metastasis, resulting in the retention of tumor cells in collecting vessels at the junction with the subcapsular sinus. 213 In addition, other chemokines expressed by LECs, such as CXCL10, CXCL12, CXCL1, and CCL5, were found to induce migration of CXCR3‐, CXCR4‐, CXCR2‐, or CCR5‐ expressing tumor cells and to be involved in cancer cell invasion and LN metastasis (reviewed in 88). On the other hand, chemokines not only mediate the attraction of tumor cells toward lymphatics, but may also act in an opposite manner. Specifically, the chemokines CXCL5, CXCL12, and CCL27 or CCL28 produced by tumor cells were found to induce a directed migration in LECs expressing the respective chemokine receptors (CXCR2, CXCR4, and CCR10), thereby contributing to lymphangiogenesis and metastasis in vivo (see 88, 214 and references therein).

In addition to the role of integrins in lymphangiogenesis (see Section 2.2), cancer cell‐expressed integrins are also involved in tumor cell metastasis. For example, the trans‐activation of β1 integrin, expressed on cancer cells, by SEM7A, expressed on neighboring cells, was found to promote invasion into tumor lymphatics. 171 In line with these results, inhibition of the α4β1 ligand vascular cell adhesion molecule 1 (VCAM‐1) decreased lymphatic vessel permeability and tumor cell intravasation. 210

3.1.2. Changes in lymphatic vessel integrity

In the primary tumor, formation of lymphatic vessels may occur both within and/or around the tumor mass. Since most of the intratumoral vessels appear collapsed and squeezed due to the high interstitial fluid pressure (IFP) and the solid pressure exerted by tumor cells, these vessels are thought to be functionally compromised. 215 , 216 , 217 By contrast, peritumoral vessels reportedly represent the principal route of escape for cancer cells from the primary tumor. 218 , 219 , 220 Different mechanisms mediate vessel dilation, alter endothelial integrity, and facilitate the entry of cancer cells into peritumoral lymphatics. For instance, enlargement of peritumoral capillaries and collecting lymphatic vessels has been shown to be influenced by prostaglandin stimulation of LECs. 221 Prostaglandins may be secreted by LECs themselves as a consequence of VEGF‐D stimulation, 207 or by TAMs through VEGF‐C‐mediated upregulation of cyclooxygenase 2 (COX2). 222 Alteration/destruction of lymphatic endothelium has been reported in the presence of tumor‐derived arachidonic acid metabolite 12S‐HETE, produced by 15‐lipoxygenase‐1 (ALOX15) catalysis, which favored endothelial cell retraction and transiently reduced VE‐cadherin expression. 223 When compared to normal dermal LECs, LECs isolated from a T‐241/VEGF‐C fibrosarcoma mouse model presented an altered expression of molecules involved in the maintenance of the integrity of cell junctions and the sub‐endothelial matrix. 209 Moreover, it has been shown that high levels of lymphangiogenic signaling by VEGF‐C resulted in altered LMC coverage of lymphatic collectors 224 and reduced the integrity of endothelial cell junctions, 122 contributing to an increase of lymphatic permeability.

3.1.3. Fluid flow into/through tumor lymphatics

Tumor cell intravasation and delivery to the LN are also dependent on fluid flow. 225 At the primary tumor site a high IFP is observed, as a consequence of the uncontrolled flow and of the abnormal permeability of pathological/cancer blood vessels. 226 In addition, the intratumoral lymphatics are often collapsed and unable to properly clear the accumulated fluids. 227 As a consequence, fluid drains from the center to the tumor periphery, 225 thus creating an IFP gradient that promotes cancer cell invasion and/or ameboid migration toward the peritumoral lymphatic vessels. 228 , 229 It has been reported that chemokines produced and secreted by tumor cells may distribute along the IFP gradient and regulate autologous chemotaxis of chemokine receptor‐expressing tumor cells toward the lymphatics. 230 Interestingly, the IFP may also affect tumor macrophages inducing their polarization to an M2 phenotype. 231 Polarized macrophages, in turn, orchestrate the chemotactic migration and the escape of tumor cells through the lymphatic drainage. 231 , 232

Compared to the blood circulation, once entered in the lymphatic vasculature, circulating tumor cells experience less shear stress and lower flow velocities what favor their survival and arrest within lymphatics or in the LN SCS (as reviewed in 225). Interestingly, in vitro evidence have shown that lymphatic‐like, but not vascular‐like, fluid shear stress may activate Yes‐associated protein 1 (YAP1) and a transcriptional co‐activator with PDZ‐binding motif (TAZ) signaling pathways in circulating tumor cells, resulting in increased cancer cell migration and proliferation, respectively. 233 , 234 Whether shear stress is able to orchestrate such intracellular signaling also in vivo remains to be determined.

3.1.4. Adaptation of tumor cells to survive in lymphatics and LNs

Once they have entered into the circulation, cancer cells are exposed to biological and mechanical stress that might complicate their journey to target organs and result in cell death. This process has been widely explored in the case of blood circulation, where the association with blood components such as platelets, may confer mechanical protection to circulating tumor cells and mechano‐adaptive molecular mechanisms—that is, rearrangement of cytoskeleton via RhoA/actomyosin activation in response to fluid shear stress—contribute to cancer cell dissemination. 235 , 236 Little experimental evidence is available concerning the resistance or cancer cell trafficking within lymphatic vessels. The lower lymph flow velocities result in reduced fluid shear stress in lymphatic vessels (less than 1 dyne/cm2) in comparison to blood vessels (where it can rise up to 1000 dynes/cm2 in turbulent vessels), 225 , 237 what should favor tumor cell survival. 225 In addition to a more favorable mechanical context, it has recently been reported that the exposure of melanoma cells to the lymph environment, characterized by low levels of free iron and high levels of glutathione and oleic acid, reduced oxidative stress and ferroptosis in circulating tumor cells and explained their ability to survive and give rise to subsequent metastasis. 238 Additionally, S1P produced by LECs has been hypothesized to support the survival of tumor cells once they have entered the lymphatic circulation. 167

Similar to blood circulation, cohesive migration of tumor cells has also been described in lymphatic vessels. For example, experimental evidence has shown that breast cancer cells initially entered lymphatic vessels as single cells and later were found as cell aggregates within the vessel. 239 Giampieri et al. 240  demonstrated that TGFβ1 can mediate this switch of cancer cells from cohesive to single‐cell motility, and that blockade of TGFβ signaling prevented single‐cell movement, but not collective one, in vivo and that these cell clusters were capable of lymphatic invasion. Interestingly, it has been suggested that the perturbation/blockage of the free flow in the lymphatic system by tumor cell clusters may lead to abnormal flow stagnation, favoring the accumulation and the growth of tumor cells at lymph vessel junctions. 241 , 242

Finally, it has been shown that metastatic tumor cells may require a metabolic rewiring to adapt and colonize LNs. Indeed, in a murine model, melanoma cells were found to upregulate genes of the fatty acid oxidation (FAO) pathway and to shift their metabolism in this direction via the activation of the transcriptional factor YAP. 243 In the fatty acid‐rich LN microenvironment, metastasizing cells activated this YAP‐dependent metabolic pathway and maintained it through autocrine production of YAP‐activating bile acids. Notably, this activation of YAP was observed also in a cohort of LN‐metastatic melanoma patients where this adaptive behavior of tumor cells correlated with further dissemination and reduced survival. 243

3.1.5. Contribution of lymphatics to cancer stem cell niche

In the primary tumor, the interaction between cancer cells and stroma generates an active microenvironment where tumor cells with stem‐like traits reside. This “cancer stem cell niche” is believed to support cancer progression, resistance, and outgrowth of metastasis. 244 Indeed, cancer stem cells have a greater potential in inducing angiogenesis and lymphangiogenesis, and can favor the formation of new blood and lymphatic vessels through the production of different factors/mediators, as reported for stem cell‐like glioma, 245 glioblastoma, 246 and serous ovarian cancer. 247 Moreover, in this context a population of cancer stem cells can directly enter the flow or undergo epithelial‐to‐mesenchymal transition acquiring a motile phenotype which eventually results in the metastasis of tumor cells. Finally, it is worth mentioning that, beyond the primary tumor, LECs may promote the formation of a lymphovascular niche in the LN as a milieu for the recruitment and maintenance of cancer stem‐like cells, which represent the most resistant and durable candidates for metastasis and tumor relapse. In this regard, CD133+ tumor cells were found associated with tumor lymphatic vessels in metastatic LNs and organs, and it has been shown that LECs promote the migration of a CD133+/CXCR4+ cell subset to target organs producing CXCL12. 248

3.2. Immunomodulation in the tumor microenvironment

Lymphatic vessels have been described as active players in the creation and maintenance of the immunosuppressive tumor microenvironment (Figures 2 and 3). This may at a first glance appear counterintuitive, considering that tumor‐associated lymphatic vessels naturally have essential functions for the initiation of antitumor immune responses, by transporting free antigen or tumor antigen‐bearing DCs to the tumor‐draining LN. The requirement of lymphatic vessels for mounting antitumor immune responses has been described in several tumor studies using animal models with impaired lymphatic vessel growth or lymphatic vessel dysfunction. 249 , 250 , 251 For example, intradermal implantation of B16F10 melanoma into K14‐VEGFR3‐Ig mice, which are devoid of lymphatic vessels in the dermis, resulted in less distant (lung) metastasis, a marked reduction in leukocyte infiltration, and an impaired antitumor immunity in response to dermal vaccine delivery. 251 Moreover, a recent study performed in mice lymphatic‐specific expression of the Diphtheria toxin (DT) receptor, what allows for a DTmediated depletion of LECs, found that local ablation of lymphatic vessels around subcutaneously implanted melanoma and breast cancer tumors resulted in increased peritumoral edema, enhanced inflammatory cell accumulation, increased tumor PD‐L1 expression, and decreased accumulation of cytotoxic T cells in the tumor microenvironment. 250 In a glioblastoma setting, Song et al. 252 recently described that ectopic expression of VEGF‐C enhanced CD8+ T cell priming in the draining cervical LNs and promoted intratumoral accumulation of CD8+ T cells and rapid cancer clearance, presumably due to enhanced lymphatic drainage of antigens to the LNs. 252  Taken together, these observations suggest that peritumoral lymphatic vessels, in principle can contribute to inhibition of tumor growth by facilitating the egress of activated immune cells from the tumor to initiate anti‐immune response and by modulating the tumor environment. However, at the same time, numerous reports have demonstrated that an increased tumor‐associated lymphatic vascular density correlates with poorer outcomes in many cancer types. 7 , 90 , 123 This apparent discrepancy between pro‐immune functions of tumor‐associated lymphatic vessels and clinical outcomes highlights the dual role of the lymphatic vasculature during tumor progression. While CCR7/CCL21‐dependent migration through lymphatic vessels allows immune cell trafficking from the tumor to the tumor‐draining LN and priming of CD8+ cytotoxic T lymphocyte responses, this pathway can, on the other hand, also be hijacked by cancer cells to enhance LN metastasis. Previous studies have established that CCR7‐expressing cancer cells can utilize VEGF‐C‐mediated induction of local CCL21 expression in tumor‐associated lymphatic vessels to escape from the primary tumor to the tumor‐draining LN. 212 , 253

Besides favoring tumor cell metastases, increasing evidence also suggests that tumor‐associated lymphatic vessels adopt an immunosuppressive phenotype characterized by the expression of immune‐inhibitory molecules (Figure 2). The establishment of an immunosuppressive environment in the primary tumor is a known mechanism of tumor escape, as it can cause T cell dysfunction and exhaustion. 254 , 255 PD‐L1, a checkpoint inhibitor, was one of the immunosuppressive molecules upregulated in tumor‐associated lymphatic vessels. 256 , 257 PD‐L1 blockade on LECs was shown to increase antigen‐specific T cell activation in vitro, suggesting that LEC‐presented PD‐L1 can dampen CD8+ T cell responses in the tumor microenvironment. 257 Lane et al. 256 concluded that antigen‐specific CD8+ T cells were the drivers of PD‐L1 expression on LECs in an interferon‐γ‐dependent manner in a mouse model of melanoma. This, in turn, caused reduced CD8+ T cell accumulation at the primary tumor side, indicative of a negative feedback loop. 256 Besides PD‐L1, also the inducible nitric oxide synthase (iNOS) and indoleamine 2,3‐dioxygenase (IDO) were upregulated in tumor‐associated lymphatic vessels of metastatic LNs. 83 iNOS catalyzes the production of NO, which is known to have suppressive functions on T cell proliferation, 84 while IDO is a rate‐limiting enzyme involved in tryptophan degradation, resulting in metabolites that are involved in Treg differentiation and inhibit effector T cell proliferation. 258 Thus, it appears that tumor‐associated LECs contribute to the local immunosuppressive tumor environment, which limits the action of tumor‐infiltrating lymphocytes and causes an exhausted or tolerogenic phenotype (Figure 2). Altogether these pleiotropic and contradictory roles played by the lymphatics, by at the same time facilitating LN‐tumor trafficking as well as immune modulation, open new questions regarding the suitability to target lymphatic vessels or lymphangiogenesis for preventing tumor dissemination.

4. LYMPH NODE METASTASES

LNs represent the first site of metastasis in several solid tumors (Figure 3). This crucial step of initial dissemination from primary tumor requires a complex reorganization and the activation of several adaptive pathways both in the lymphatics and in the tumor cells. It has been reported that in tumor‐draining LNs a number of alterations in the lymphatic vessels and in the vasculature occur even before the arrival of cancer cells. Indeed, tumor cells rewire the lymphatics and the tumor‐draining LNs in many ways to cause the remodeling, the expansion of the lymphatic vascular network, and the formation of pre‐metastatic niches. 89 , 259 In the cancer settings, the enlargement of tumor‐draining LNs goes along with increased recruitment or proliferation of lymphocytes, significant expansion of stromal cells, including the remodeling and proliferative expansion of the lymphatic network. 86 , 87 , 89 , 90 , 260

4.1. Lymphatics in the lymph node pre‐metastatic niche

The formation of the metastatic niche in the tumor‐draining LN has been reported to involve different pro‐lymphangiogenic stimuli. Tumors release a plethora of soluble factors, as well as extracellular vesicles 261 , 262 or circulating tumor cells that enter and traffic through the blood and the lymphatic vessels. 225 These tumor‐derived “messengers” trigger lymphangiogenesis in the LN as a key mechanism to create new lymphatics in sentinel LNs and favor metastatic spread. 263 Canonical pro‐lymphangiogenic factors such as VEGF‐C and VEGF‐A have been widely investigated and implicated in LN lymphangiogenesis. In a mouse model of chemically‐induced skin carcinogenesis, specific skin‐restricted overexpression of VEGF‐A was associated with increased primary tumor growth, proliferation of VEGFR2+ tumor‐associated lymphatic vessels, and also with LN lymphangiogenesis accompanied by metastasis in draining and distant LNs. 99 In a similar model, skin‐specific overexpression of VEGF‐C induced a significant expansion of lymphatic networks in the sentinel LN and increased the impact of LN and distant metastases, with no significant effect on primary tumor growth. 264

Interestingly, it has been shown in different tumor models that VEGF‐C activates PI3K in LECs and this promoted the remodeling of the lymphatic network in the draining LN, and the activation of the integrin α4β1, thus facilitating the binding of VCAM‐1+ metastatic tumor cells. 265 Tumor‐induced lymphangiogenesis in metastasis‐free sentinel LNs has also been reported from murine melanoma models after footpad 266 and ear‐sponge implantation. 267 Moreover, in a murine model of nasopharyngeal carcinoma, the establishment of metastasis in the sentinel LN was preceded by enrichment of blood and lymph vessels, and the expansion of the lymph sinuses correlated with the weight of the primary tumor. 268 More recently, using a transgenic mouse model where a Vegfr3‐driven reporter revealed sites of LEC activation (Vegfr3Luc nu/nu mice), Olmeda et al. 106 have confirmed that increased lymphatic vessel density (LVD) in the LN is required to obtain efficient nodal metastasis. In this model, regardless of the genetic alterations (i.e., BRAF, NRAS, PTEN, and p53 mutations) or of VEGF‐C levels, subcutaneously grafted human melanoma cell lines caused LN and visceral dissemination only after a detectable local, distal, or systemic activation of the lymphatic network. 106

In addition to VEGF family members, other mediators have been implicated in LN lymphangiogenesis. For example, erythropoietin was found to exert a potent lymphangiogenic effect facilitating LN lymphangiogenesis and nodal metastases in murine models of breast cancer and melanoma. 96 In addition, in a murine model of lung carcinoma metastasis, the preparation of a pre‐metastatic niche in the LN was triggered by DCs via activation of the COX‐2‐derived prostaglandin E2 that induced CXCL12 in the subcapsular regions of the LN. 269 Recently, the heparin‐binding factor midkine (MDK), thus far implicated in the acquisition of critical hallmarks of cancer such as cell growth, survival, metastasis, migration, and angiogenesis, 270 has been identified as a mediator of LN lymphangiogenesis. 106 MDK is produced by melanoma cells (and other tumor types) as a secreted factor or as an exosome cargo and is able to activate the mTOR pathway in LECs, 106 a well‐characterized player in lymphangiogenesis. 271 Interestingly, MDK accumulated in the lymphatic vessels of LNs and of visceral organs before tumor colonization in murine melanoma, and this was observed also in a cohort of melanoma patients. 106

4.1.1. Tumor‐induced gene expression changes in LN LECs

Transcriptomics studies have shown that LECs adapt their gene expression signature in presence of the primary tumor—not only at the site of tumor growth, but also in tumor‐draining LNs. Indeed, RNA sequencing of tumor‐draining LNs of breast and melanoma models revealed that LEC sprouting and proliferative pathways are activated as early as 4 days after primary tumor implantation and before tumor cell seeding to the LN. In agreement with the presence of an active lymphangiogenic process, genes responsible for cell division, immune modulation, and cell adhesion were upregulated. Moreover, strong alterations in the expression levels of genes involved in cell–cell and cell–matrix adhesion were observed. 260 In particular, integrin αIIb was found to be significantly overexpressed in terms of mRNA and protein levels in activated LECs of tumor‐draining LNs, and to co‐localize with fibrinogen, which is increased and accumulated around lymphatic sinuses in these LNs. These observations, and the fact that integrin αIIb mediates adhesion of LECs to fibrinogen in vitro, 260 suggest that fibrinogen deposition via integrin αIIb might be one mechanism driving the formation of the pre‐metastatic niche in draining LNs. In addition, other adhesive mediators have been found to be differentially expressed. For instance, the downregulation of Jam3, which encodes for the junctional adhesion molecule C (JAM‐C), resulted in reduced vessel permeability and leukocyte trafficking in the LN. 260

4.2. Evasion of immune surveillance in the LN

Besides serving as a route for tumor dissemination to the draining LN and further systemic metastasis, lymphatic vessels were shown to actively propagate immune tolerance not only in the primary tumor but also in the tumor‐draining LN. Before infiltrating the tumor and exerting an antitumor response, antigen‐specific CD8+ T cells need to be activated in the tumor‐draining LN by antigen‐presenting cells such as conventional DCs that present tumor‐derived antigen to antigen‐specific T cells. However, tumor cells can often evade immune recognition. This is on one hand due to typically diminished antigenicity of transformed cells compared to, for example, pathogens encountered in the context of an infection. Tumor cells also frequently lose MHC expression, rendering them invisible to antigen‐specific cytotoxic T lymphocytes. Moreover, in contrast to infections that are also recognized by the innate immune system through pathogen‐associated molecular patterns (PAMPs), growing tumors induce less activation of innate immunity. 272 , 273 As a consequence, tumor‐antigen presenting DCs, that travel to the tumor‐draining LN, often fail to express costimulatory molecules required for T cell activation. 274 In the worst case, this might not only prevent the mounting of an antitumor immune response but cause tolerance against the tumor antigens, by inducing the generation of Tregs or T cell dysfunction (anergy and exhaustion). In uninflamed, steady‐state conditions, LN represents an immunosuppressive environment maintained by LN stromal cells such as FRCs, blood endothelial cells, and LECs. 275 Stromal cells, including LECs, produce immunosuppressive molecules that can profoundly affect the survival, fate, and activation of naïve T cells and other lymphocyte subsets to prevent autoimmune reactions. Examples of such molecules are the immune checkpoint molecules PD‐L1, NOS2/NO, which can promote the induction of Tregs from CD4+ CD25 T cell and inhibit T cell proliferation, 75 , 84 , 276 or IDO, which inhibits naïve T cell proliferation. 277 Moreover, FRCs and LECs in steady‐state LNs have been shown to present peripheral tissue antigens and induce T cell anergy or clonal deletion in a programmed cell death protein 1 (PD‐1)/PD‐L1 dependent manner or alternatively by induction of Tregs (Figure 2). 68 , 70 , 71 , 73 , 75 At this point, there are only a few experimental studies that have closely investigated the immunological consequences of tumor antigen uptake by LN LECs during tumor progression, and whether antigen presentation by LN LECs also exerts immunogenic or exclusively immunosuppressive activity. In support of the latter, Lund et al. 69 reported that in a B16F10 melanoma model expressing the foreign antigen ovalbumin (OVA), LECs in the tumor‐draining LN were found to cross‐present OVA on MHC Class I molecules. Subsequently, cocultured OVA‐specific CD8+ T cells with tumor antigen‐presenting LN LECs caused abnormal proliferation and apoptosis of CD8+ T cells ex vivo, suggesting that LN LECs might actively suppress the activation of tumor‐specific CD8+ T cells in tumor‐draining LNs. Besides directly presenting antigen, it is perceivable that LN LECs could also impact adaptive immunity by taking up tumor‐derived antigen and passing it on to other antigen‐presenting cells, in analogy to the antigen archiving function described for medullary sinus LECs in the context of viral infection and vaccination. 77 , 78 Specifically, it was shown that LECs “pass on” antigen to cross‐presenting DCs, which induce potent CD8+ T cells responses. It is interesting to note that in the tumor context, similar mechanisms of antigen transfer have been reported for migratory and resident DCs. While migratory DCs were shown to capture and transport tumor antigen‐containing vesicles to draining LNs, the induction of antitumor cytotoxic T cell responses depended on vesicle transfer and Ag presentation by resident DCs. 278 , 279 Future studies are expected to shed more light on the mechanisms of antigen presentation by LECs or antigen transfer between LECs and other antigen‐presenting cells in tumor‐draining LNs.

4.3. Further dissemination from tumor‐draining LNs

Long distance and visceral metastases represent the real life‐threatening aspects of metastatic cancer. Formation of these types of metastases is always preceded by extravasation of cancer cells from the blood circulation into the target organ, demonstrating the importance of blood vessels in this process. Nevertheless, in most solid tumors the process of lymphatic metastasis precedes dissemination by blood vessels. Lymphatic metastasis is driven by multiple mechanisms, and a recent report suggests that cells that metastasize through the lymphatics are different from those spreading through the blood circulation. 280

Metastatic cells from the tumor‐draining LN can further colonize distal nodes or be a source of cancer cells that give rise to distant metastases (Figure 3). Entry of metastatic cells into the blood circulation may either occur by dissemination through efferent lymphatic vessels and the thoracic ducts into the subclavian vein or, as recently demonstrated, directly via the LN blood vasculature. 281 , 282 The latter possibility of direct lymph‐to‐blood vessel passage has been hypothesized for a long time until recent experimental evidence. Brown et al. 281 demonstrated that intra‐lymphatic microinfusion of metastatic murine breast cancer cells into the SCS of popliteal LNs resulted in rapid infiltration of the LN parenchyma, invasion of blood vessels, and consequent seeding to the lungs, with no involvement of the thoracic duct. Interestingly, after entering the SCS, tumor cells were limited in their migration to the medullary sinus, and this prevented or slowed down further dissemination via the efferent lymphatics. In this avascular milieu tumor cells activated invasive programs and took contact with and wrapped around HEVs, which represent the main sites of leukocytes extravasation from blood into the LN. 281 Pereira et al., 282 on the other hand, performed photoconversion experiments and established that, after orthotopic implantation of melanoma and breast cancer cells and sentinel LN colonization, the long‐distance lung metastases where composed preferentially (around 70%) by cancer cells derived from the LN. Even though it remains to be established if tumor cell entry into nodal blood vessels also occurs in patients, these findings reveal that LN‐resident tumor cells represent great contributors to the systemic dissemination of neoplastic cells.

This evidence further underlies the fact that LN metastases are not the final step of the metastatic process but just on stepping stone in systemic cancer spread. This has also been revealed by clonal genetic evolution studies of primary tumors, LNs, and distant metastases in human colorectal cancer, revealing a scenario where LN metastases and distant metastases often share a common origin. 283 In this complex panorama, also the metastatic sites may actively contribute to further expand systemic colonization. Indeed, lymphangiogenesis also occurs in metastases and strongly contributes to further dissemination to other organs generating an intra‐metastasis spreading, as reported in prostate cancer patients. 284 This has been shown in a transgenic model where inducible lung overexpression of VEGF‐C was accompanied by increased density of lymphatic vessels at metastatic sites as well as higher number of metastasis not only in the lungs, but also in other organs. 134 Importantly, despite the central role of LN metastases in terms of prognosis, various clinical trials have demonstrated that removal of regional LNs has no impact on the survival rate in different types of metastatic cancers, including melanoma. 285 , 286 These observations confirm that the tumor cells present in regional LNs at the time of resection are not the only ones responsible for the overall tumor diffusion in the body. In fact, further diffusion to distant LNs, intravasation into blood vessels, or micro‐colonization of distant organs might have already occurred and secondary lesions might appear at distant sites after a short or long lifespan.

5. PROGNOSTIC VALUE OF TUMOR‐INFILTRATING LYMPHATIC VESSELS

Even though the architecture and functionality of tumor‐infiltrating lymphatics may vary depending on tumor type, an increasing body of evidence indicates that a strong correlation may exist between tumor lymphangiogenesis and patient outcomes. Indeed, various parameters, such as overexpression of lymphangiogenic factors and their receptors, intratumoral and peritumoral LVD, enlargement, and remodeling of lymphatics, or tumor cell invasion of lymphatic vessels, have been associated with metastasis occurrence and reduced overall survival in different cancers (see 86, 87, 90 and references therein). For example, metastatic cutaneous melanomas were characterized by higher intratumoral and peritumoral LVD when compared to non‐metastatic tumors. 287 This is in line with the observation that the overexpression of VEGF‐C and ANG2 correlated with a poorer prognosis in melanoma patients. 288 , 289 In addition, a multivariate risk analysis conducted on primary tumors and sentinel LN biopsies from 45 melanoma patients revealed that the lymphatic vascular area of primary melanomas was an extremely sensitive and specific prognostic marker for the occurrence of tumor‐draining LN metastasis and could predict the metastatic phenotype more accurately than measurements of tumor thickness measuring. 290 Similar findings have been reported for head and neck, breast, lung, colorectal, and bladder carcinomas, where the peritumoral and/or intratumoral LVD have been found to associate with LN metastasis and shorter disease‐free/overall survival in the majority of the clinical studies (see 86, 90 and references therein).

Interestingly, the link between lymphangiogenesis and patient prognosis has also been reported in tumor‐draining LN and distant metastases. 134 , 263 For example, a study conducted on 65 breast cancer patients showed that the proliferation index of LECs in tumor‐draining LN metastases significantly correlated with the presence of metastasis‐positive non‐sentinel LN and further metastatic spread. 291 In addition, a retrospective study of 266 melanoma patients with lung metastases showed that the lymphatic area and the LVD around tumor metastases correlated with a poorer prognosis and a reduced survival. 134 Finally, it was recently reported that lymphatic exudates collected from metastatic melanoma patients that underwent lymphadenectomy were highly enriched in cancer biomarkers (e.g., extracellular vesicles containing tumor‐derived proteins and miRNAs). 259 , 261 These results may open a new field of research based on tumor‐draining lymph fluids as “liquid biopsy” to be utilized for the discovery of novel biomarkers, as well as for the tumor staging and patient outcome prediction, including risk of relapse.

6. THERAPEUTIC PERSPECTIVES

As already discussed, lymphatic vessels mediate the spread of tumor cells to draining LNs and more distant sites in the body and at the same time may serve as reservoir for in‐transit metastatic cells. Moreover, they are emerging as important players in tumoral immunosuppression. Colonization of LNs by tumor cells represents a recognized prognostic factor of tumor aggressiveness and patient survival. 87 , 90 , 286 , 292 , 293 , 294 , 295 However, even though the presence of LN metastasis correlates with poorer outcomes, removal of LNs does not always appear to be beneficial. 296 , 297 , 298 Nevertheless, given the multiple and key roles of lymphatics in cancer progression, a significant interest exists to therapeutically target lymphangiogenesis or to exploit the lymphatic vasculature for drug delivery.

6.1. Lymphangiogenesis as a therapeutic target

Direct targeting of the lymphatic vasculature could allow to interfere with tumor cell dissemination to draining LNs and beyond, but might also represent a strategy for modulating tumoral immunosuppression. Considering that VEGFR3 has been identified as the main driver of lymphangiogenesis, it is not surprising that the majority of studies and drug development strategies have thus far focused on targeting the VEGF‐C/VEGF‐D/VEGFR3 axis. In fact, this is somewhat similar to the field of tumor angiogenesis, where numerous drugs targeting the VEGF‐A/VEGFR1/VEGFR2 axis have been developed and approved over the last two decades. Well‐known examples of the latter are monoclonal antibodies directed against VEGF‐A (i.e., bevacizumab and ranibizumab) or VEGFR2 (i.e., ramuricumab), as well as other tyrosine kinase inhibitors (i.e., axitinib, tivozanib, sunitinib, pazopanib, cabozantinib, and cediranib) targeting also VEGF receptors. 299 , 300 Likewise, drugs that more specifically target components of the VEGF‐C/VEGFR3 signaling pathway are being evaluated for clinical development (reviewed in 86, 301; Figure 4). However, thus far, only a small number of antibodies, receptor traps, and small molecules that specifically target VEGF‐C or VEGFR3 have entered clinical development. 302 , 303 A first Phase I clinical trial that assessed the tolerability of a humanized IgG1 antibody blocking VEGFR3 (LY3022856/IMC‐3C5, developed by Eli Lilly and Company), was conducted in patients with advanced solid tumors and colorectal cancer. The trial was completed in 2014 with the conclusion that the treatment was well tolerated but had limited antitumor activity. 302 Similarly, results from a Phase I clinical study evaluating the effects of a human VEGF‐C neutralizing monoclonal antibody (VGX‐100), developed by Circadian Technologies) in patients with advanced solid tumors found this treatment to be well tolerated. 303 A Phase I study (NCT01514123) is still ongoing with the aim to evaluate the tolerability and possible synergisms of VGX‐100 in combination with bevacizumab in adult subjects with advanced or metastatic solid tumors. However, for all investigational drugs, apparently, no follow‐up or Phase II studies have been initiated so far, what may reflect the poor primary outcome on tumor growth and the need for additional insights on the actual impact of targeting lymphangiogenesis in advanced solid tumors. On the other hand, several molecules that inhibit signaling pathways involved in, but not exclusively occurring in lymphangiogenesis, such ANG1/ANG2, c‐Met, or HGF, are currently under clinical development as anticancer treatments (reviewed in 86). The underrepresentation of drugs in clinical development that specifically targets lymphangiogenesis in cancer might be due to several reasons. On one hand, this might be caused by the only relatively recent recognition of the importance of lymphatic vessels in tumor dissemination, combined with the generally lengthy duration of drug development. On the other hand, despite overwhelming evidence of the involvement of lymphatic vessels in cancer spread, experience from clinical studies with VEGF‐A/VEGFR1/VEGFR2‐targeting drugs might suggest that the benefit of such monotherapies could fall short of the expectations raised by the preclinical studies. Bevacizumab, for example, turned out to have a modest impact on patient survival when given as a monotherapy and is therefore mostly used in combination therapies. 304 , 305 , 306 Furthermore, in contrast to antiangiogenic drugs, which reduce the growth of tumor lesions, anti‐lymphangiogenic treatment is expected to primarily impact tumor cell dissemination and overall survival, that is, parameters that typically can only be evaluated in lengthy and Phase II/III clinical trials involving high patient numbers.

Figure 4.

Figure 4

Drug targets in the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) signaling pathways. Graphic depiction of the currently approved drugs or drugs in development that target either VEGF‐A/VEGFR2 or VEGF‐C/VEGF‐D/VEGFR3 signaling for inhibition of (lymph)angiogenesis. Drug formats comprise monoclonal antibodies, receptor tyrosine kinase (RTKs) inhibitors, and receptor traps, which represent the extracellular portions of the receptors fused to an antibody Fc moiety [Color figure can be viewed at wileyonlinelibrary.com]

It is worth mentioning that, besides anti‐lymphangiogenic drugs, also other approaches have been proposed in the attempt to “hit” the lymphatic vasculature. For example, since LECs proliferate much less compared to tumor cells or other cells in the tumor microenvironment, they have been proposed to be insensitive to radiation therapy. 307 Combining radiotherapy with anti‐lymphangiogenic drugs might therefore act synergistically to reduce the risk of lymphatic metastasis. 242 A potential caveat, however, is that radiation reportedly induces secondary dysfunctions of lymphatics, as the vessels lose their contractile capacity and ability to drain fluids when the surrounding irradiated tissue becomes fibrotic. 308 Combination of anti‐lymphangiogenic drugs and radiotherapy might therefore be particularly problematic in patients with post‐surgery lymphedema.

Another treatment that may display synergism with anti‐lymphangiogenic therapy is photodynamic therapy (PDT). PDT is based in the uptake of photosensitizer molecules which, upon excitation by light of a particular wavelength, react with oxygen and generate reactive oxygen species in target tissues causing cell death. 309 To date, PDT has been clinically approved for the treatment of a variety of solid cancers (e.g., lung, bladder, skin, and pancreatic tumors). 310 Moreover, blood vascular‐targeted PDT has been approved in Europe for the treatment of low‐risk prostate cancer patients 311 and a number of Phase I/II/III clinical trials are ongoing to evaluate its efficacy for urologic, prostatic, and esophagogastric cancers (reviewed in 312). Interestingly, the potential of lymphatic‐targeted PDT is currently under preclinical evaluation and is based on the use of intra‐ or peritumoral injection of phototoxic compounds, such as verteporfin, that under liposomal formulation preferentially accumulate in lymphatics, and once activated react with the surrounding tissues, destroy lymphatic vessels and, if present, tumor cells within them (i.e., in transit lymphatic metastases). 313 , 314 , 315 However, even though the combination of PDT with anti‐lymphangiogenic drugs may have the advantage to prevent the regeneration of novel lymphatic vessels, 314 both approaches reportedly suppress the induction of the antitumor immune response, by inhibiting the immunologic communication—, that is, DC trafficking and antigen drainage—with the draining LN. 316

Adding to these difficulties in development might be the emerging complex and the dual role that lymphatic vessels appear to play in tumor progression/dissemination. Although lymphangiogenesis is generally associated with cancer spread and poorer patient prognosis, 86 , 87 , 90 , 317 recent evidence indicates that the presence of intratumoral and peritumoral lymphatic vessels and (high) VEGF‐C expression at the same time may be beneficial, by enhancing the efficacy of cancer immunotherapy. Specifically, a study by Fankhauser et al. 149 reported that VEGF‐C expression and high LVD correlated with a good cancer immunotherapy response. Although the study initially found that in murine melanoma models, blocking VEGFR3 signaling was associated with decreased infiltration of immunosuppressive Tregs, unexpectedly, VEGF‐C signaling also potentiated the responsiveness of melanoma to immunotherapy. This effect was attributed to increased recruitment and local activation of CCR7+ T cells in lymphangiogenic (VEGF‐C expressing B16‐OVA/VC) tumors. Similarly, in human metastatic melanoma patients, serum VEGF‐C levels correlated with T cell activation and expansion after peptide vaccination and clinical response to checkpoint blockade. 149 Along the same line, Bordry et al. 83 reported that increased tumor LVD positively correlated with an immunosuppressive phenotype and increased CD8+ T cell infiltration in patients with cutaneous melanoma. Taken together, it is likely that the success of therapies targeting tumor‐associated lymphangiogenesis might depend on several factors including the tumor type, stage, and possibly the combination with immunotherapy. Therefore, it might be necessary to evaluate the potential benefits and risks of anti‐lymphangiogenic drugs, depending on defined prognostic criteria.

6.2. Lymphatic vessels as drug delivery routes

Most chemotherapeutic drugs are administered by the oral or intravenous route. Typically, these drugs display poor tumor uptake and distribute to and accumulate in normal organs and tissues, leading to substantial side effects. In addition, due to their small size, they are rapidly cleared from tissues by reabsorption into blood vessels, rather than entering lymphatic vessels. 318 Therefore, it is a unique challenge to achieve sufficient accumulation and retention of these types of drugs at the tumor or metastatic site and/or in lymphoid tissues. 133 , 319 , 320 , 321 In the case of LN metastases or in‐transit metastases, one option for reaching higher therapeutic drug concentrations at the site of disease could consist of using the lymphatic vasculature as a delivery route. Due to the unique structure of initial lymphatic capillaries, uptake of compounds from the injection site in the interstitium into lymphatics as compared to the blood vessels is dependent on several factors, such as the size of the molecule/particle, as well as its charge or hydrophobicity. 85 Lymphatic uptake and LN accumulation are most efficient for macromolecules, including large peptides and proteins, ranging from 20 to 50 nm and for particles between 10 and 100 nm. 318 , 322 Particles larger than 100 nm cannot easily diffuse through the interstitium, and most of them remain at the injection site until being cleared by phagocytes. 323 By contrast, molecules smaller than 5–10 nm, such as most low molecular weight drug molecules, can cross blood vessel wall and therefore directly enter the blood circulation. 324 , 325

Novel approaches aiming at delivering subcutaneously, intradermally, or intramuscularly injected low molecular weight therapeutics via lymphatic vessels to draining LNs take advantage of associating or packaging these drugs in or with macromolecular carriers, such as nanoparticles, capsules, polymers, micelles, liposomes, and dendrimers (extensively reviewed in 85, 326, 327). As an example, liposomes containing doxorubicin have been tested in clinical trials of patients with gastric carcinoma. In this study, injection of liposomal‐doxorubicin into the gastric submucosa allowed to reach significantly higher concentrations of doxorubicin in the primary and secondary draining LNs in comparison to intravenously injected liposomal–doxorubicin or free doxorubicin injected both intravenously or also into the gastric submucosa. 328 Similarly, subcutaneous injection of depot forming cancer vaccines (i.e., specific tumor antigens and adjuvant) exploit lymphatic delivery to LNs, and might be further enhanced by direct lymphatic targeting, local modulation of lymphatic drainage, or by recruitment of antigen‐presenting cells. 327 An alternative, recently published approach made use of microneedles to enhance the lymphatic uptake and delivery of a CTLA‐4 blocking antibody (i.e., a checkpoint inhibitor) to the tumor‐draining LN in a murine orthotopic mammary carcinoma model. 329 Compared to systemic (intravenous) treatment with the checkpoint inhibitor, the intradermal delivery to the tumor‐draining LN resulted in more effective tumor growth inhibition, increased tumor‐infiltrating lymphocytes, and decreased metastasis, presumably due to a better targeting of the tumor‐draining LN and consequently more efficient activation of tumor‐specific T cells. 329 The therapeutic benefits of targeting immune checkpoint inhibitors to tumor‐draining LNs was recently confirmed by another study and extended to the use of PD‐1 inhibitors. 330

Likewise, but less efficaciously than subcutaneous administration, intramuscular drug injection was demonstrated to increase lymphatic uptake of liposome‐entrapped methotrexate 331 and mitomycin C conjugated with dextran. 332 As a further refinement of this principle, another recent study described the use of sonoporation using acoustic liposomes and ultrasound as a method to enhance the uptake of intra‐lymphatically delivered doxorubicin to metastasis‐bearing LNs growing in the SCS. In this case, intralymphatic delivery and release of doxorubicin were shown to induce tumor necrosis and prevented further tumor invasion into the LN parenchyma. 333 Another strategy would be to exploit peptides and molecules that bind to receptors expressed by (tumor‐associated) lymphatic vessels to achieve lymphatic trafficking/homing. An interesting example is the synthetic nonapeptide Ly‐1P, which was shown to target tumor cells, tumor‐associated lymphatics, and macrophages. 334 , 335 Ly‐1P has been synthetically linked/conjugated to polymeric micelles, 336 PEGylated, doxorubicin‐containing liposomes, 337 or nanoparticles. 338 Subcutaneous injection of PEGylated, doxorubicin‐containing liposomes, for example, resulted in increased accumulation of LyP‐1‐conjugated PEGylated liposomes compared to unconjugated liposomes in metastatic LNs and suppressed LN metastasis in vivo. 337 , 339 However as Ly‐P1 also exhibits cytotoxicity toward tumor cells and gets internalized by tumor cells as well as LECs, safety, and efficacy of Ly‐P1 would need to be further evaluated. 335 , 336 Moreover, a general drawback of the above‐mentioned approaches might be that they will only allow to target metastatic disease in LNs draining tissues that are accessible by injection from outside the body, such as via the skin. Future studies will be required to elucidate which of these experimental approaches can be successfully translated to the clinics.

7. CONCLUSIONS

Over the last decades, it has become clear that the lymphatic vasculature plays crucial roles both in the maintenance of tissue homeostasis as well as in disease. In addition to its contribution to immune‐related and inflammatory disorders, an overwhelming scientific literature now documents the importance of lymphatic vessels in cancer biology and its relevance for the prognosis of cancer patients. While the lymphatic vasculature undoubtedly represents an active and dynamic player in cancer progression, it is also clear that further investigations will be required to increase our understanding of the complex interplay occurring among cancer cells, LECs, and the immune system, and for ultimately translating these new insights into therapies.

In the era of “immuno‐oncology”, more and more studies are reporting the key role of the lymphatic vasculature in modulating the antitumor immune response. More knowledge of both the tumor cell‐ LEC as well as the LEC‐immune cell crosstalk will help to better understand the two‐fold role that lymphatics appear to play in either promoting or dampening tumor spread. Already now it is apparent that no “one‐fits all” explanation will be found, but that the role of lymphatics will likely depend on various aspects, such as the tumor type (e.g., aggressiveness, mutation load) and tissue localization. It is clear that LECs respond to the tumor environment, yet only a few studies have thus far explored this cellular plasticity in the tumor context. In view of recent advances in single‐cell sequencing, it is likely that we will soon know much more about tumor‐specific LEC signatures. This knowledge will be instrumental for the identification of new prognostic markers as well as potential therapeutic targets. In addition, it will provide a better understanding of the lymphatic‐tumor cell crosstalk and how it evolves during the different phases of cancer onset, progression, and dissemination.

Considering that lymphangiogenesis or immune‐modulation by lymphatics appears to occur both locally in the tumor and in tumor‐draining LNs, it is at present not possible to unambiguously assign the relative importance of a process taking place at either site for tumor progression. The availability of tools to specifically target lymphatic vessels in a particular tissue or in LNs, for example, with specific antibodies or mouse models allowing for genetic deletion of a gene in lymphatics at either site, would allow for great advances in this field. It is likely that new insights, for example, gained from emerging transcriptomics analyses into organ‐specific differences in the lymphatic vasculature, 21 will open up new opportunities in the future.

From a therapeutic perspective, it is expected and desirable that more anti‐lymphangiogenic approaches will soon reach the stage of clinical development. However, it remains to be carefully evaluated where and when anti‐lymphangiogenic therapy can be applied to “mechanically” reduce the dissemination of tumor cells, without potentially impacting the endogenous antitumor immune response or the efficacy of immunotherapy. Also, in this regard it is likely that a case‐by‐case evaluation of the tumor type and disease burden will be required, considering not only the lymphatic involvement, but also immune‐related parameters like tumor immunogenicity and responsiveness to tumor immunotherapy. Last but not least, further studies investigating how lymphatics may be used for the delivery of therapeutic drugs to metastatic LNs or sites of antitumor immune induction will likely reveal new avenues for exploiting the lymphatic vasculature in oncology.

ACKNOWLEDGMENTS

Roberto Ronca is supported by Associazione Italiana per la Ricerca sul Cancro (AIRC IG 2019 – ID: 23151). Sara Rezzola is supported by a Fondazione Umberto Veronesi fellowship. Cornelia Halin and Elena C. Sigmund gratefully acknowledge funding from ETH Zurich and from the Swiss National Fund Sinergia program (CRSII3_160719/1). Open access funding provided by Universita degli Studi di Brescia within the CRUI‐CARE Agreement.

Biographies

Sara Rezzola is a Postdoctoral Researcher at the Department of Molecular and Translational Medicine of the University of Brescia (Italy), where she obtained her PhD in Cellular and Molecular Biotechnologies in 2015. Her research activity focuses on the role of angiogenesis in different pathological settings, including cancer and ocular neovascular disorders.

Elena C. Sigmund is a Postdoctoral Researcher at the Institute of Pharmaceutical Sciences at ETH Zurich (Switzerland), where she obtained her PhD in 2020. Her research activity focuses on the role of atypical chemokine receptors in lymphatic development and function as well as immune cell migration.

Cornelia Halin is an Associate Professor of Pharmaceutical Immunology at the Institute of Pharmaceutical Sciences of ETH Zurich (Switzerland). Her research interests lie at the crossroads of immunology and vascular biology. The main aims of her work are to elucidate basic mechanisms of leukocyte migration through afferent lymphatic vessels and to identify new therapeutic targets for the treatment of disorders that involve the lymphatic vasculature.

Roberto Ronca is an Associate Professor in General Pathology and Immunology at the Department of Molecular and Translational Medicine of the University of Brescia (Italy). After graduating in Biology in 1999 at the University of Padua (Italy), he obtained his PhD in Medical Biotechnologies at the University of Brescia. His research expertise is focused on the study of tumor–stroma interactions, including aspects of angiogenesis, vascular biology, and the development of therapeutic approaches with anti‐cancer and anti‐angiogenetic properties.

Rezzola S, Sigmund EC, Halin C, Ronca R. The lymphatic vasculature: An active and dynamic player in cancer progression. Med Res Rev. 2022;42:576‐614. 10.1002/med.21855

Sara Rezzola and Elena C. Sigmund contributed equally to this study.

Contributor Information

Cornelia Halin, Email: cornelia.halin@pharma.ethz.ch.

Roberto Ronca, Email: roberto.ronca@unibs.it.

REFERENCES

  • 1. Cupedo T, Coles MC, Veiga‐Fernandes H. Development and structure of lymph nodes in humans and mice. Dev Biol Peripher Lymphoid Organs. 2011:1‐177. [Google Scholar]
  • 2. Aspelund A, Robciuc MR, Karaman S, Makinen T, Alitalo K. Lymphatic system in cardiovascular medicine. Circ Res. 2016;118(3):515‐530. [DOI] [PubMed] [Google Scholar]
  • 3. Ulvmar MH, Makinen T. Heterogeneity in the lymphatic vascular system and its origin. Cardiovasc Res. 2016;111(4):310‐321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. 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):270‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Schwager S, Detmar M. Inflammation and lymphatic function. Front Immunol. 2019;10:308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Randolph GJ, Ivanov S, Zinselmeyer BH, Scallan JP. The lymphatic system: integral roles in immunity. Annu Rev Immunol. 2017;35:31‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Garnier L, Gkountidi AO, Hugues S. Tumor‐associated lymphatic vessel features and immunomodulatory functions. Front Immunol. 2019;10:720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Escobedo N, Oliver G. The lymphatic vasculature: its role in adipose metabolism and obesity. Cell Metab. 2017;26(4):598‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Schineis P, Runge P, Halin C. Cellular traffic through afferent lymphatic vessels. Vascul Pharmacol. 2019;112:31‐41. [DOI] [PubMed] [Google Scholar]
  • 10. Braun A, Worbs T, Moschovakis GL, et al. Afferent lymph‐derived T cells and DCs use different chemokine receptor CCR7‐dependent routes for entry into the lymph node and intranodal migration. Nat Immunol. 2011;12(9):879‐887. [DOI] [PubMed] [Google Scholar]
  • 11. Kukk E, Lymboussaki A, Taira S, et al. VEGF‐C receptor binding and pattern of expression with VEGFR‐3 suggests a role in lymphatic vascular development. Development. 1996;122(12):3829‐3837. [DOI] [PubMed] [Google Scholar]
  • 12. 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 USA. 1995;92(8):3566‐3570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Breiteneder‐Geleff S, Soleiman A, Kowalski H, et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 1999;154(2):385‐394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Mäkinen T, Adams RH, Bailey J, et al. PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature. Genes Dev. 2005;19(3):397‐410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Banerji S, Ni J, Wang SX, et al. LYVE‐1, a new homologue of the CD44 glycoprotein, is a lymph‐specific receptor for hyaluronan. J Cell Biol. 1999;144(4):789‐801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Kriehuber E, Breiteneder‐Geleff S, Groeger M, et al. Isolation and characterization of dermal lymphatic and blood endothelial cells reveal stable and functionally specialized cell lineages. J Exp Med. 2001;194(6):797‐808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Weber M, Hauschild R, Schwarz J, et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science. 2013;339(6117):328‐332. [DOI] [PubMed] [Google Scholar]
  • 18. Forster R, Braun A, Worbs T. Lymph node homing of T cells and dendritic cells via afferent lymphatics. Trends Immunol. 2012;33(6):271‐280. [DOI] [PubMed] [Google Scholar]
  • 19. Srinivasan RS, Dillard ME, Lagutin OV, et al. Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature. Genes Dev. 2007;21(19):2422‐2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Yaniv K, Isogai S, Castranova D, Dye L, Hitomi J, Weinstein BM. Live imaging of lymphatic development in the zebrafish. Nat Med. 2006;12(6):711‐716. [DOI] [PubMed] [Google Scholar]
  • 21. Petrova TV, Koh GY. Organ‐specific lymphatic vasculature: from development to pathophysiology. J Exp Med. 2018;215(1):35‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Stanczuk L, Martinez‐Corral I, Ulvmar MH, et al. cKit lineage hemogenic endothelium‐derived cells contribute to mesenteric lymphatic vessels. Cell Rep. 2015;10(10):1708‐1721. [DOI] [PubMed] [Google Scholar]
  • 23. Martinez‐Corral I, Ulvmar MH, Stanczuk L, et al. Nonvenous origin of dermal lymphatic vasculature. Circ Res. 2015;116(10):1649‐1654. [DOI] [PubMed] [Google Scholar]
  • 24. François M, Caprini A, Hosking B, et al. Sox18 induces development of the lymphatic vasculature in mice. Nature. 2008;456(7222):643‐647. [DOI] [PubMed] [Google Scholar]
  • 25. Srinivasan RS, Geng X, Yang Y, et al. The nuclear hormone receptor Coup‐TFII is required for the initiation and early maintenance of Prox1 expression in lymphatic endothelial cells. Genes Dev. 2010;24(7):696‐707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Karkkainen MJ, Haiko P, Sainio K, et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol. 2004;5(1):74‐80. [DOI] [PubMed] [Google Scholar]
  • 27. Hägerling R, Pollmann C, Andreas M, et al. A novel multistep mechanism for initial lymphangiogenesis in mouse embryos based on ultramicroscopy. EMBO J. 2013;32(5):629‐644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Yang Y, García‐Verdugo JM, Soriano‐Navarro M, et al. Lymphatic endothelial progenitors bud from the cardinal vein and intersomitic vessels in mammalian embryos. Blood. 2012;120(11):2340‐2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. François M, Short K, Secker GA, et al. Segmental territories along the cardinal veins generate lymph sacs via a ballooning mechanism during embryonic lymphangiogenesis in mice. Dev Biol. 2012;364(2):89‐98. [DOI] [PubMed] [Google Scholar]
  • 30. Norrmén C, Ivanov KI, Cheng J, et al. FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1. J Cell Biol. 2009;185(3):439‐457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Sabine A, Agalarov Y, Maby‐El Hajjami H, et al. Mechanotransduction, PROX1, and FOXC2 cooperate to control connexin37 and calcineurin during lymphatic‐valve formation. Dev Cell. 2012;22(2):430‐445. [DOI] [PubMed] [Google Scholar]
  • 32. 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(9):974‐981. [DOI] [PubMed] [Google Scholar]
  • 33. Baluk P, Fuxe J, Hashizume H, et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J Exp Med. 2007;204(10):2349‐2362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Leak LV, Burke JF. Ultrastructural studies on the lymphatic anchoring filaments. J Cell Biol. 1968;36(1):129‐149. [PMC free article] [PubMed] [Google Scholar]
  • 35. Lutter S, Xie S, Tatin F, Makinen T. Smooth muscle‐endothelial cell communication activates Reelin signaling and regulates lymphatic vessel formation. J Cell Biol. 2012;197(6):837‐849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Pflicke H, Sixt M. Preformed portals facilitate dendritic cell entry into afferent lymphatic vessels. J Exp Med. 2009;206(13):2925‐2935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. von der Weid PY, Zawieja DC. Lymphatic smooth muscle: the motor unit of lymph drainage. Int J Biochem Cell Biol. 2004;36(7):1147‐1153. [DOI] [PubMed] [Google Scholar]
  • 38. Scallan JP, Zawieja SD, Castorena‐Gonzalez JA, Davis MJ. Lymphatic pumping: mechanics, mechanisms and malfunction. J Physiol. 2016;594(20):5749‐5768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Lauweryns JM, Boussauw L. The ultrastructure of lymphatic valves in the adult rabbit lung. Z Zellforsch Mikrosk Anat. 1973;143(2):149‐168. [DOI] [PubMed] [Google Scholar]
  • 40. Schulte‐Merker S, Sabine A, Petrova TV. Lymphatic vascular morphogenesis in development, physiology, and disease. J Cell Biol. 2011;193(4):607‐618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Reed HO, Wang L, Sonett J, et al. Lymphatic impairment leads to pulmonary tertiary lymphoid organ formation and alveolar damage. J Clin Invest. 2019;129(6):2514‐2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Bernier‐Latmani J, Petrova TV. Intestinal lymphatic vasculature: structure, mechanisms and functions. Nat Rev Gastroenterol Hepatol. 2017;14(9):510‐526. [DOI] [PubMed] [Google Scholar]
  • 43. Bernier‐Latmani J, Cisarovsky C, Demir CS, et al. DLL4 promotes continuous adult intestinal lacteal regeneration and dietary fat transport. J Clin Invest. 2015;125(12):4572‐4586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Bradham RR, Parker EF, Barrington BA, Jr , Webb CM, Stallworth JM. The cardiac lymphatics. Ann Surg. 1970;171(6):899‐902 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Brakenhielm E, Alitalo K. Cardiac lymphatics in health and disease. Nat Rev Cardiol. 2019;16(1):56‐68. [DOI] [PubMed] [Google Scholar]
  • 46. Petrova TV, Koh GY. Biological functions of lymphatic vessels. Science. 2020;369(6500). [DOI] [PubMed] [Google Scholar]
  • 47. Ma Q, Ries M, Decker Y, et al. Rapid lymphatic efflux limits cerebrospinal fluid flow to the brain. Acta Neuropathol. 2019;137(1):151‐165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Wong BW, Zecchin A, Garcia‐Caballero M, Carmeliet P. Emerging concepts in organ‐specific lymphatic vessels and metabolic regulation of lymphatic development. Dev Cell. 2018;45(3):289‐301. [DOI] [PubMed] [Google Scholar]
  • 49. Sokol CL, Camire RB, Jones MC, Luster AD. The chemokine receptor CCR8 promotes the migration of dendritic cells into the lymph node parenchyma to initiate the allergic immune response. Immunity. 2018;49(3):449‐463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Girard JP, Moussion C, Forster R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol. 2012;12(11):762‐773. [DOI] [PubMed] [Google Scholar]
  • 51. Reynoso GV, Weisberg AS, Shannon JP, et al. Lymph node conduits transport virions for rapid T cell activation. Nat Immunol. 2019;20(5):602‐612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Sixt M, Kanazawa N, Selg M, et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity. 2005;22(1):19‐29. [DOI] [PubMed] [Google Scholar]
  • 53. Thierry GR, Kuka M, De Giovanni M, et al. The conduit system exports locally secreted IgM from lymph nodes. J Exp Med. 2018;215(12):2972‐2983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Leon B, Lund FE. Compartmentalization of dendritic cell and T‐cell interactions in the lymph node: anatomy of T‐cell fate decisions. Immunol Rev. 2019;289(1):84‐100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Breart B, Bousso P. S1P1 downregulation tailors CD8(+) T‐cell residence time in lymph nodes to the strength of the antigenic stimulation. Eur J Immunol. 2016;46(12):2730‐2736. [DOI] [PubMed] [Google Scholar]
  • 56. Benechet AP, Menon M, Xu D, et al. T cell‐intrinsic S1PR1 regulates endogenous effector T‐cell egress dynamics from lymph nodes during infection. Proc Natl Acad Sci USA. 2016;113(8):2182‐2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Qi H, Kastenmuller W, Germain RN. Spatiotemporal basis of innate and adaptive immunity in secondary lymphoid tissue. Annu Rev Cell Dev Biol. 2014;30:141‐167. [DOI] [PubMed] [Google Scholar]
  • 58. Fujimoto N, He Y, D'Addio M, Tacconi C, Detmar M, Dieterich LC. Single‐cell mapping reveals new markers and functions of lymphatic endothelial cells in lymph nodes. PLOS Biol. 2020;18(4):e3000704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Xiang M, Grosso RA, Takeda A, et al. A single‐cell transcriptional roadmap of the mouse and human lymph node lymphatic vasculature. Front Cardiovasc Med. 2020;7:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Takeda A, Hollmén M, Dermadi D, et al. Single‐cell survey of human lymphatics unveils marked endothelial cell heterogeneity and mechanisms of homing for neutrophils. Immunity. 2019;51(3):561‐572. [DOI] [PubMed] [Google Scholar]
  • 61. Jalkanen S, Salmi M. Lymphatic endothelial cells of the lymph node. Nat Rev Immunol. 2020;20(9):566‐578. [DOI] [PubMed] [Google Scholar]
  • 62. Ulvmar MH, Werth K, Braun A, et al. The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes. Nat Immunol. 2014;15(7):623‐630. [DOI] [PubMed] [Google Scholar]
  • 63. Gray EE, Cyster JG. Lymph node macrophages. J Innate Immun. 2012;4(5–6):424‐436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Gerner MY, Torabi‐Parizi P, Germain RN. Strategically localized dendritic cells promote rapid T cell responses to lymph‐borne particulate antigens. Immunity. 2015;42(1):172‐185. [DOI] [PubMed] [Google Scholar]
  • 65. Kähäri L, Fair‐Mäkelä R, Auvinen K, et al. Transcytosis route mediates rapid delivery of intact antibodies to draining lymph nodes. J Clin Invest. 2019;129(8):3086‐3102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Rantakari P, Auvinen K, Jäppinen N, et al. The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes. Nat Immunol. 2015;16(4):386‐396. [DOI] [PubMed] [Google Scholar]
  • 67. Farr AG, Cho Y, De, Bruyn PP. The structure of the sinus wall of the lymph node relative to its endocytic properties and transmural cell passage. Am J Anat. 1980;157(3):265‐284. [DOI] [PubMed] [Google Scholar]
  • 68. Hirosue S, Vokali E, Raghavan VR, et al. Steady‐state antigen scavenging, cross‐presentation, and CD8+ T cell priming: a new role for lymphatic endothelial cells. J Immunol. 2014;192(11):5002‐5011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Lund AW, Duraes FV, Hirosue S, et al. VEGF‐C promotes immune tolerance in B16 melanomas and cross‐presentation of tumor antigen by lymph node lymphatics. Cell Rep. 2012;1(3):191‐199. [DOI] [PubMed] [Google Scholar]
  • 70. Fletcher AL, Lukacs‐Kornek V, Reynoso ED, et al. Lymph node fibroblastic reticular cells directly present peripheral tissue antigen under steady‐state and inflammatory conditions. J Exp Med. 2010;207(4):689‐697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Cohen JN, Guidi CJ, Tewalt EF, et al. Lymph node‐resident lymphatic endothelial cells mediate peripheral tolerance via Aire‐independent direct antigen presentation. J Exp Med. 2010;207(4):681‐688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Dubrot J, Duraes FV, Potin L, et al. Lymph node stromal cells acquire peptide‐MHCII complexes from dendritic cells and induce antigen‐specific CD4(+) T cell tolerance. J Exp Med. 2014;211(6):1153‐1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Tewalt EF, Cohen JN, Rouhani SJ, et al. Lymphatic endothelial cells induce tolerance via PD‐L1 and lack of costimulation leading to high‐level PD‐1 expression on CD8 T cells. Blood. 2012;120(24):4772‐4782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Rouhani SJ, Eccles JD, Riccardi P, et al. Roles of lymphatic endothelial cells expressing peripheral tissue antigens in CD4 T‐cell tolerance induction. Nat Commun. 2015;6:6771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Nadafi R, Gago de Graça C, Keuning ED, et al. Lymph node stromal cells generate antigen‐specific regulatory T cells and control autoreactive T and B cell responses. Cell Rep. 2020;30(12):4110‐4123. [DOI] [PubMed] [Google Scholar]
  • 76. Vokali E, Yu SS, Hirosue S, et al. Lymphatic endothelial cells prime naive CD8(+) T cells into memory cells under steady‐state conditions. Nat Commun. 2020;11(1):538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Kedl RM, Lindsay RS, Finlon JM, Lucas ED, Friedman RS, Tamburini BAJ. Migratory dendritic cells acquire and present lymphatic endothelial cell‐archived antigens during lymph node contraction. Nat Commun. 2017;8(1):2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Tamburini BA, Burchill MA, Kedl RM. Antigen capture and archiving by lymphatic endothelial cells following vaccination or viral infection. Nat Commun. 2014;5:3989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Pham TH, Baluk P, Xu Y, et al. Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning. J Exp Med. 2010;207(1):17‐27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Schwab SR, Pereira JP, Matloubian M, Xu Y, Huang Y, Cyster JG. Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science. 2005;309(5741):1735‐1739. [DOI] [PubMed] [Google Scholar]
  • 81. Cyster JG, Schwab SR. Sphingosine‐1‐phosphate and lymphocyte egress from lymphoid organs. Annu Rev Immunol. 2012;30:69‐94. [DOI] [PubMed] [Google Scholar]
  • 82. Matloubian M, Lo CG, Cinamon G, et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427(6972):355‐360. [DOI] [PubMed] [Google Scholar]
  • 83. Bordry N, Broggi MAS, de Jonge K, et al. Lymphatic vessel density is associated with CD8(+) T cell infiltration and immunosuppressive factors in human melanoma. Oncoimmunology. 2018;7(8):e1462878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Lukacs‐Kornek V, Malhotra D, Fletcher AL, et al. Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes. Nat Immunol. 2011;12(11):1096‐1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Cote B, Rao D, Alany RG, Kwon GS, Alani AWG. Lymphatic changes in cancer and drug delivery to the lymphatics in solid tumors. Adv Drug Deliv Rev. 2019;144:16‐34. [DOI] [PubMed] [Google Scholar]
  • 86. Dieterich LC, Detmar M. Tumor lymphangiogenesis and new drug development. Adv Drug Deliv Rev. 2016;99(Pt B):148‐160. [DOI] [PubMed] [Google Scholar]
  • 87. Farnsworth RH, Achen MG, Stacker SA. The evolving role of lymphatics in cancer metastasis. Curr Opin Immunol. 2018;53:64‐73. [DOI] [PubMed] [Google Scholar]
  • 88. Farnsworth RH, Karnezis T, Maciburko SJ, Mueller SN, Stacker SA. The interplay between lymphatic vessels and chemokines. Front Immunol. 2019;10:518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Karaman S, Detmar M. Mechanisms of lymphatic metastasis. J Clin Invest. 2014;124(3):922‐928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Stacker SA, Williams SP, Karnezis T, Shayan R, Fox SB, Achen MG. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat Rev Cancer. 2014;14(3):159‐172. [DOI] [PubMed] [Google Scholar]
  • 91. Heinz M, Niederleithner HL, Puujalka E, et al. Activin A is anti‐lymphangiogenic in a melanoma mouse model. J Invest Dermatol. 2015;135(1):212‐221. [DOI] [PubMed] [Google Scholar]
  • 92. Klein KR, Caron KM. Adrenomedullin in lymphangiogenesis: from development to disease. Cell Mol Life Sci. 2015;72(16):3115‐3126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Fagiani E, Lorentz P, Kopfstein L, Christofori G. Angiopoietin‐1 and ‐2 exert antagonistic functions in tumor angiogenesis, yet both induce lymphangiogenesis. Cancer Res. 2011;71(17):5717‐5727. [DOI] [PubMed] [Google Scholar]
  • 94. Kang YE, Kim KS, Park SJ, et al. High expression of angiopoietin‐1 is associated with lymph node metastasis and invasiveness of papillary thyroid carcinoma. World J Surg. 2017;41(12):3128‐3138. [DOI] [PubMed] [Google Scholar]
  • 95. Bracher A, Cardona AS, Tauber S, et al. Epidermal growth factor facilitates melanoma lymph node metastasis by influencing tumor lymphangiogenesis. J Invest Dermatol. 2013;133(1):230‐238. [DOI] [PubMed] [Google Scholar]
  • 96. Lee AS, Kim DH, Lee JE, et al. Erythropoietin induces lymph node lymphangiogenesis and lymph node tumor metastasis. Cancer Res. 2011;71(13):4506‐4517. [DOI] [PubMed] [Google Scholar]
  • 97. Platonova N, Miquel G, Regenfuss B, et al. Evidence for the interaction of fibroblast growth factor‐2 with the lymphatic endothelial cell marker LYVE‐1. Blood. 2013;121(7):1229‐1237. [DOI] [PubMed] [Google Scholar]
  • 98. Cao R, Ji H, Feng N, et al. Collaborative interplay between FGF‐2 and VEGF‐C promotes lymphangiogenesis and metastasis. Proc Natl Acad Sci USA. 2012;109(39):15894‐15899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Kajiya K, Hirakawa S, Ma B, Drinnenberg I, Detmar M. Hepatocyte growth factor promotes lymphatic vessel formation and function. EMBO J. 2005;24(16):2885‐2895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Jiang WG, Davies G, Martin TA, et al. The potential lymphangiogenic effects of hepatocyte growth factor/scatter factor in vitro and in vivo. Int J Mol Med. 2005;16(4):723‐728. [PubMed] [Google Scholar]
  • 101. Cao R, Björndahl MA, Gallego MI, et al. Hepatocyte growth factor is a lymphangiogenic factor with an indirect mechanism of action. Blood. 2006;107(9):3531‐3536. [DOI] [PubMed] [Google Scholar]
  • 102. Li ZJ, Ying XJ, Chen HL, et al. Insulin‐like growth factor‐1 induces lymphangiogenesis and facilitates lymphatic metastasis in colorectal cancer. World J Gastroenterol. 2013;19(43):7788‐7794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Björndahl M, Cao R, Nissen LJ, et al. Insulin‐like growth factors 1 and 2 induce lymphangiogenesis in vivo. Proc Natl Acad Sci USA. 2005;102(43):15593‐15598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Zhao G, Zhu G, Huang Y, et al. IL‐6 mediates the signal pathway of JAK‐STAT3‐VEGF‐C promoting growth, invasion and lymphangiogenesis in gastric cancer. Oncol Rep. 2016;35(3):1787‐1795. [DOI] [PubMed] [Google Scholar]
  • 105. Shinriki S, Jono H, Ueda M, et al. Interleukin‐6 signalling regulates vascular endothelial growth factor‐C synthesis and lymphangiogenesis in human oral squamous cell carcinoma. J Pathol. 2011;225(1):142‐150. [DOI] [PubMed] [Google Scholar]
  • 106. Olmeda D, Cerezo‐Wallis D, Riveiro‐Falkenbach E, et al. Whole‐body imaging of lymphovascular niches identifies pre‐metastatic roles of midkine. Nature. 2017;546(7660):676‐680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Liersch R, Shin JW, Bayer M, et al. Analysis of a novel highly metastatic melanoma cell line identifies osteopontin as a new lymphangiogenic factor. Int J Oncol. 2012;41(4):1455‐1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Cao R, Björndahl MA, Religa P, et al. PDGF‐BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis. Cancer Cell. 2004;6(4):333‐345. [DOI] [PubMed] [Google Scholar]
  • 109. Jitariu AA, Cimpean AM, Kundnani NR, Raica M. Platelet‐derived growth factors induced lymphangiogenesis: evidence, unanswered questions and upcoming challenges. Arch Med Sci. 2015;11(1):57‐66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Liu J, Liu C, Qiu L, Li J, Zhang P, Sun Y. Overexpression of both platelet‐derived growth factor‐BB and vascular endothelial growth factor‐C and its association with lymphangiogenesis in primary human non‐small cell lung cancer. Diagn Pathol. 2014;9:128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Cao Y. Direct role of PDGF‐BB in lymphangiogenesis and lymphatic metastasis. Cell Cycle. 2005;4(2):228‐230. [DOI] [PubMed] [Google Scholar]
  • 112. Cartier A, Hla T. Sphingosine 1‐phosphate: lipid signaling in pathology and therapy. Science. 2019;366:6463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Nagahashi M, Ramachandran S, Kim EY, et al. Sphingosine‐1‐phosphate produced by sphingosine kinase 1 promotes breast cancer progression by stimulating angiogenesis and lymphangiogenesis. Cancer Res. 2012;72(3):726‐735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Yu J, Zhang X, Kuzontkoski PM, et al. Slit2N and Robo4 regulate lymphangiogenesis through the VEGF‐C/VEGFR‐3 pathway. Cell Commun Signal. 2014;12:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Liu D, Li L, Zhang XX, et al. SIX1 promotes tumor lymphangiogenesis by coordinating TGFbeta signals that increase expression of VEGF‐C. Cancer Res. 2014;74(19):5597‐5607. [DOI] [PubMed] [Google Scholar]
  • 116. Gore J, Imasuen‐Williams IE, Conteh AM, Craven KE, Cheng M, Korc M. Combined targeting of TGF‐beta, EGFR and HER2 suppresses lymphangiogenesis and metastasis in a pancreatic cancer model. Cancer Lett. 2016;379(1):143‐153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Evans R, Flores‐Borja F, Nassiri S, et al. Integrin‐mediated macrophage adhesion promotes lymphovascular dissemination in breast cancer. Cell Rep. 2019;27(7):1967‐1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Song J, Chen W, Cui X, et al. CCBE1 promotes tumor lymphangiogenesis and is negatively regulated by TGFbeta signaling in colorectal cancer. Theranostics. 2020;10(5):2327‐2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Oka M, Iwata C, Suzuki HI, et al. Inhibition of endogenous TGF‐beta signaling enhances lymphangiogenesis. Blood. 2008;111(9):4571‐4579. [DOI] [PubMed] [Google Scholar]
  • 120. Ji H, Cao R, Yang Y, et al. TNFR1 mediates TNF‐alpha‐induced tumour lymphangiogenesis and metastasis by modulating VEGF‐C‐VEGFR3 signalling. Nat Commun. 2014;5:4944. [DOI] [PubMed] [Google Scholar]
  • 121. Hong H, Jiang L, Lin Y, et al. TNF‐alpha promotes lymphangiogenesis and lymphatic metastasis of gallbladder cancer through the ERK1/2/AP‐1/VEGF‐D pathway. BMC Cancer. 2016;16:240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Tacconi C, Correale C, Gandelli A, et al. Vascular endothelial growth factor C disrupts the endothelial lymphatic barrier to promote colorectal cancer invasion. Gastroenterology. 2015;148(7):1438‐1451. [DOI] [PubMed] [Google Scholar]
  • 123. Skobe M, Hawighorst T, Jackson DG, et al. Induction of tumor lymphangiogenesis by VEGF‐C promotes breast cancer metastasis. Nat Med. 2001;7(2):192‐198. [DOI] [PubMed] [Google Scholar]
  • 124. Zhang YQ, Chen WL, Zhang F, et al. Over‐expression of both VEGF‐C and Twist predicts poor prognosis in human breast cancer. Clin Transl Oncol. 2019;21(9):1250‐1259. [DOI] [PubMed] [Google Scholar]
  • 125. Li J, Xie Y, Wang X, et al. Overexpression of VEGF‐C and MMP‐9 predicts poor prognosis in Kazakh patients with esophageal squamous cell carcinoma. PeerJ. 2019;7:e8182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Stacker SA, Caesar C, Baldwin ME, et al. VEGF‐D promotes the metastatic spread of tumor cells via the lymphatics. Nat Med. 2001;7(2):186‐191. [DOI] [PubMed] [Google Scholar]
  • 127. Yonemura Y, Endo Y, Tabata K, et al. Role of VEGF‐C and VEGF‐D in lymphangiogenesis in gastric cancer. Int J Clin Oncol. 2005;10(5):318‐327. [DOI] [PubMed] [Google Scholar]
  • 128. Niederleithner H, Heinz M, Tauber S, et al. Wnt1 is anti‐lymphangiogenic in a melanoma mouse model. J Invest Dermatol. 2012;132(9):2235‐2244. [DOI] [PubMed] [Google Scholar]
  • 129. Wang SH, Chang JS, Hsiao JR, et al. Tumour cell‐derived WNT5B modulates in vitro lymphangiogenesis via induction of partial endothelial‐mesenchymal transition of lymphatic endothelial cells. Oncogene. 2017;36(11):1503‐1515. [DOI] [PubMed] [Google Scholar]
  • 130. Stacker SA, Stenvers K, Caesar C, et al. Biosynthesis of vascular endothelial growth factor‐D involves proteolytic processing which generates non‐covalent homodimers. J Biol Chem. 1999;274(45):32127‐32136. [DOI] [PubMed] [Google Scholar]
  • 131. Mäkinen T, Veikkola T, Mustjoki S, et al. Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF‐C/D receptor VEGFR‐3. EMBO J. 2001;20(17):4762‐4773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Kwon S, Velasquez FC, Sevick‐Muraca EM. Near‐infrared fluorescence lymphatic imaging in vascular endothelial growth factor‐C overexpressing murine melanoma. Biomed Opt Express. 2018;9(10):4631‐4637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Tammela T, Alitalo K. Lymphangiogenesis: molecular mechanisms and future promise. Cell. 2010;140(4):460‐476. [DOI] [PubMed] [Google Scholar]
  • 134. Ma Q, Dieterich LC, Ikenberg K, et al. Unexpected contribution of lymphatic vessels to promotion of distant metastatic tumor spread. Sci Adv. 2018;4(8):eaat4758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Wang J, Huang Y, Zhang J, et al. NRP‐2 in tumor lymphangiogenesis and lymphatic metastasis. Cancer Lett. 2018;418:176‐184. [DOI] [PubMed] [Google Scholar]
  • 136. Lee JY, Hong SH, Shin M, Heo HR, Jang IH. Blockade of FLT4 suppresses metastasis of melanoma cells by impaired lymphatic vessels. Biochem Biophys Res Commun. 2016;478(2):733‐738. [DOI] [PubMed] [Google Scholar]
  • 137. Shibata MA, Shibata E, Tanaka Y, Shiraoka C, Kondo Y. Soluble Vegfr3 gene therapy suppresses multi‐organ metastasis in a mouse mammary cancer model. Cancer Sci. 2020;111:2837‐2849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Morfoisse F, Renaud E, Hantelys F, Prats AC, Garmy‐Susini B. Role of hypoxia and vascular endothelial growth factors in lymphangiogenesis. Mol Cell Oncol. 2015;2(4):e1024821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Roy S, Kumaravel S, Sharma A, Duran CL, Bayless KJ, Chakraborty S. Hypoxic tumor microenvironment: implications for cancer therapy. Exp Biol Med. 2020;245(13):1073‐1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Wiig H, Swartz MA. Interstitial fluid and lymph formation and transport: physiological regulation and roles in inflammation and cancer. Physiol Rev. 2012;92(3):1005‐1060. [DOI] [PubMed] [Google Scholar]
  • 141. Boardman KC, Swartz MA. Interstitial flow as a guide for lymphangiogenesis. Circ Res. 2003;92(7):801‐808. [DOI] [PubMed] [Google Scholar]
  • 142. Jain RK, Tong RT, Munn LL. Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: insights from a mathematical model. Cancer Res. 2007;67(6):2729‐2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Rofstad EK, Galappathi K, Mathiesen BS. Tumor interstitial fluid pressure—a link between tumor hypoxia, microvascular density, and lymph node metastasis. Neoplasia. 2014;16(7):586‐594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Nathan SS, Huvos AG, Casas‐Ganem JE, et al. Tumor interstitial fluid pressure may regulate angiogenic factors in osteosarcoma. J Orthop Res. 2008;26(11):1520‐1525. [DOI] [PubMed] [Google Scholar]
  • 145. Hunter S, Nault B, Ugwuagbo KC, Maiti S, Majumder M. Mir526b and Mir655 promote tumour associated angiogenesis and lymphangiogenesis in breast cancer. Cancers. 2019;11(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Martini T, Heinkele J, Mayr R, et al. Predictive value of lymphangiogenesis and proliferation markers on mRNA level in urothelial carcinoma of the bladder after radical cystectomy. Urol Oncol. 2018;36(12):530.e19‐530.e27. [DOI] [PubMed] [Google Scholar]
  • 147. Qin T, Xia J, Liu S, et al. Clinical importance of VEGFC and PD‐L1 co‐expression in lung adenocarcinoma patients. Thorac Cancer. 2020;11(5):1139‐1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Mlecnik B, Bindea G, Kirilovsky A, et al. The tumor microenvironment and Immunoscore are critical determinants of dissemination to distant metastasis. Sci Transl Med. 2016;8(327):327ra326. [DOI] [PubMed] [Google Scholar]
  • 149. Fankhauser M, Broggi MAS, Potin L, et al. Tumor lymphangiogenesis promotes T cell infiltration and potentiates immunotherapy in melanoma. Sci Transl Med. 2017;9(407). [DOI] [PubMed] [Google Scholar]
  • 150. Hong YK, Lange‐Asschenfeldt B, Velasco P, et al. VEGF‐A promotes tissue repair‐associated lymphatic vessel formation via VEGFR‐2 and the alpha1beta1 and alpha2beta1 integrins. FASEB J. 2004;18(10):1111‐1113. [DOI] [PubMed] [Google Scholar]
  • 151. Nagy JA, Vasile E, Feng D, et al. Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J Exp Med. 2002;196(11):1497‐1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Hirakawa S, Kodama S, Kunstfeld R, Kajiya K, Brown LF, Detmar M. VEGF‐A induces tumor and sentinel lymph node lymphangiogenesis and promotes lymphatic metastasis. J Exp Med. 2005;201(7):1089‐1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Hwang‐Bo J, Bae MG, Park JH, Chung IS. 3‐O‐Acetyloleanolic acid inhibits VEGF‐A‐induced lymphangiogenesis and lymph node metastasis in an oral cancer sentinel lymph node animal model. BMC Cancer. 2018;18(1):714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Li XP, Jing W, Sun JJ, et al. A potential small‐molecule synthetic antilymphangiogenic agent norcantharidin inhibits tumor growth and lymphangiogenesis of human colonic adenocarcinomas through blocking VEGF‐A,‐C,‐D/VEGFR‐2,‐3 "multi‐points priming" mechanisms in vitro and in vivo. BMC Cancer. 2015;15:527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Miyazaki H, Yoshimatsu Y, Akatsu Y, et al. Expression of platelet‐derived growth factor receptor beta is maintained by Prox1 in lymphatic endothelial cells and is required for tumor lymphangiogenesis. Cancer Sci. 2014;105(9):1116‐1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Zhang QH, Qian K, Li XJ, Pu J, Wu XT. [Experimental study of the hepatocyte growth factor contributing to lymphangiogenesis and lymphatic metastasis in gastric cancer]. Zhonghua Wei Chang Wai Ke Za Zhi. 2007;10(3):212‐216. [PubMed] [Google Scholar]
  • 157. Duong T, Proulx ST, Luciani P, et al. Genetic ablation of SOX18 function suppresses tumor lymphangiogenesis and metastasis of melanoma in mice. Cancer Res. 2012;72(12):3105‐3114. [DOI] [PubMed] [Google Scholar]
  • 158. Huaqi Y, Caipeng Q, Qiang W, et al. Transcription factor SOX18 promotes clear cell renal cell carcinoma progression and alleviates cabozantinib‐mediated inhibitory effects. Mol Cancer Ther. 2019;18(12):2433‐2445. [DOI] [PubMed] [Google Scholar]
  • 159. Zhang N, Xie F, Gao W, et al. Expression of hepatocyte growth factor and c‐Met in non‐small‐cell lung cancer and association with lymphangiogenesis. Mol Med Rep. 2015;11(4):2797‐2804. [DOI] [PubMed] [Google Scholar]
  • 160. Giacomini A, Grillo E, Rezzola S, et al. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev. 2020;101:569‐610. [DOI] [PubMed] [Google Scholar]
  • 161. Shin JW, Min M, Larrieu‐Lahargue F, et al. Prox1 promotes lineage‐specific expression of fibroblast growth factor (FGF) receptor‐3 in lymphatic endothelium: a role for FGF signaling in lymphangiogenesis. Mol Biol Cell. 2006;17(2):576‐584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Cai Y, Yao S, Zhong J, et al. Inhibition activity of a disulfide‐stabilized diabody against basic fibroblast growth factor in lung cancer. Oncotarget. 2017;8(12):20187‐20197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Saharinen P, Leppanen VM, Alitalo K. SnapShot: angiopoietins and their functions. Cell. 2017;171(3):724. [DOI] [PubMed] [Google Scholar]
  • 164. Holopainen T, Saharinen P, D'Amico G, et al. Effects of angiopoietin‐2‐blocking antibody on endothelial cell‐cell junctions and lung metastasis. J Natl Cancer Inst. 2012;104(6):461‐475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Cai E, Yang D, Zhang Y, et al. Angiopoietin‐1 is associated with a decreased risk of lymph node metastasis in early stage cervical cancer. Histol Histopathol. 2020;35:18234‐1034. [DOI] [PubMed] [Google Scholar]
  • 166. Jeltsch M, Jha SK, Tvorogov D, et al. CCBE1 enhances lymphangiogenesis via A disintegrin and metalloprotease with thrombospondin motifs‐3‐mediated vascular endothelial growth factor‐C activation. Circulation. 2014;129(19):1962‐1971. [DOI] [PubMed] [Google Scholar]
  • 167. Nagahashi M, Abe M, Sakimura K, Takabe K, Wakai T. The role of sphingosine‐1‐phosphate in inflammation and cancer progression. Cancer Sci. 2018;109(12):3671‐3678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Schulz MM, Reisen F, Zgraggen S, et al. Phenotype‐based high‐content chemical library screening identifies statins as inhibitors of in vivo lymphangiogenesis. Proc Natl Acad Sci USA. 2012;109(40):E2665‐E2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Guerrero PA, McCarty JH. Integrins in vascular development and pathology. Adv Pharmacol. 2018;81:129‐153. [DOI] [PubMed] [Google Scholar]
  • 170. Avraamides CJ, Garmy‐Susini B, Varner JA. Integrins in angiogenesis and lymphangiogenesis. Nat Rev Cancer. 2008;8(8):604‐617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Black SA, Nelson AC, Gurule NJ, Futscher BW, Lyons TR. Semaphorin 7a exerts pleiotropic effects to promote breast tumor progression. Oncogene. 2016;35(39):5170‐5178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Garmy‐Susini B, Avraamides CJ, Schmid MC, et al. Integrin alpha4beta1 signaling is required for lymphangiogenesis and tumor metastasis. Cancer Res. 2010;70(8):3042‐3051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Bieniasz‐Krzywiec P, Martín‐Pérez R, Ehling M, et al. Podoplanin‐expressing macrophages promote lymphangiogenesis and lymphoinvasion in breast cancer. Cell Metab. 2019;30(5):917‐936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Crnic I, Strittmatter K, Cavallaro U, et al. Loss of neural cell adhesion molecule induces tumor metastasis by up‐regulating lymphangiogenesis. Cancer Res. 2004;64(23):8630‐8638. [DOI] [PubMed] [Google Scholar]
  • 175. Yan H, Zhang C, Wang Z, et al. CD146 is required for VEGF‐C‐induced lymphatic sprouting during lymphangiogenesis. Sci Rep. 2017;7(1):7442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Wang Z, Xu Q, Zhang N, Du X, Xu G, Yan X. CD146, from a melanoma cell adhesion molecule to a signaling receptor. Signal Transduct Target Ther. 2020;5(1):148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Md Yusof K, Rosli R, Abdullah M, AA‐K K. The roles of non‐coding RNAs in tumor‐associated lymphangiogenesis. Cancers. 2020;12(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Yan S, Wang H, Chen X, et al. MiR‐182‐5p inhibits colon cancer tumorigenesis, angiogenesis, and lymphangiogenesis by directly downregulating VEGF‐C. Cancer Lett. 2020;488:18‐26. [DOI] [PubMed] [Google Scholar]
  • 179. Su CM, Tang CH, Chi MJ, et al. Resistin facilitates VEGF‐C‐associated lymphangiogenesis by inhibiting miR‐186 in human chondrosarcoma cells. Biochem Pharmacol. 2018;154:234‐242. [DOI] [PubMed] [Google Scholar]
  • 180. Lin CY, Wang SW, Chen YL, et al. Brain‐derived neurotrophic factor promotes VEGF‐C‐dependent lymphangiogenesis by suppressing miR‐624‐3p in human chondrosarcoma cells. Cell Death Dis. 2017;8(8):e2964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Tzeng HE, Chang AC, Tsai CH, Wang SW, Tang CH. Basic fibroblast growth factor promotes VEGF‐C‐dependent lymphangiogenesis via inhibition of miR‐381 in human chondrosarcoma cells. Oncotarget. 2016;7(25):38566‐38578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Yang WH, Chang AC, Wang SW, et al. Leptin promotes VEGF‐C production and induces lymphangiogenesis by suppressing miR‐27b in human chondrosarcoma cells. Sci Rep. 2016;6:28647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Huang CY, Chang AC, Chen HT, Wang SW, Lo YS, Tang CH. Adiponectin promotes VEGF‐C‐dependent lymphangiogenesis by inhibiting miR‐27b through a CaMKII/AMPK/p38 signaling pathway in human chondrosarcoma cells. Clin Sci. 2016;130(17):1523‐1533. [DOI] [PubMed] [Google Scholar]
  • 184. Wang LH, Lin CY, Liu SC, et al. CCL5 promotes VEGF‐C production and induces lymphangiogenesis by suppressing miR‐507 in human chondrosarcoma cells. Oncotarget. 2016;7(24):36896‐36908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Lien MY, Tsai HC, Chang AC, et al. Chemokine CCL4 induces vascular endothelial growth factor C expression and lymphangiogenesis by miR‐195‐3p in oral squamous cell carcinoma. Front Immunol. 2018;9:412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Lin CC, Chen PC, Lein MY, et al. WISP‐1 promotes VEGF‐C‐dependent lymphangiogenesis by inhibiting miR‐300 in human oral squamous cell carcinoma cells. Oncotarget. 2016;7(9):9993‐10005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Hu J, Cheng Y, Li Y, et al. microRNA‐128 plays a critical role in human non‐small cell lung cancer tumourigenesis, angiogenesis and lymphangiogenesis by directly targeting vascular endothelial growth factor‐C. Eur J Cancer. 2014;50(13):2336‐2350. [DOI] [PubMed] [Google Scholar]
  • 188. Keklikoglou I, Hosaka K, Bender C, et al. MicroRNA‐206 functions as a pleiotropic modulator of cell proliferation, invasion and lymphangiogenesis in pancreatic adenocarcinoma by targeting ANXA2 and KRAS genes. Oncogene. 2015;34(37):4867‐4878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Wei L, Sun C, Zhang Y, Han N, Sun S. miR‐503‐5p inhibits colon cancer tumorigenesis, angiogenesis, and lymphangiogenesis by directly downregulating VEGF‐A. Gene Ther. 2020. [DOI] [PubMed] [Google Scholar]
  • 190. Sasahira T, Kurihara M, Bhawal UK, et al. Downregulation of miR‐126 induces angiogenesis and lymphangiogenesis by activation of VEGF‐A in oral cancer. Br J Cancer. 2012;107(4):700‐706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Zhao Z, Li L, Du P, et al. Transcriptional downregulation of miR‐4306 serves as a new therapeutic target for triple negative breast cancer. Theranostics. 2019;9(5):1401‐1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Liu C, Li M, Hu Y, et al. miR‐486‐5p attenuates tumor growth and lymphangiogenesis by targeting neuropilin‐2 in colorectal carcinoma. OncoTargets Ther. 2016;9:2865‐2871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Qian Q, Sun W, Zhu W, et al. The role of microRNA‐93 regulating angiopoietin2 in the formation of malignant pleural effusion. Cancer Med. 2017;6(5):1036‐1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Yu D, Han GH, Zhao X, et al. MicroRNA‐129‐5p suppresses nasopharyngeal carcinoma lymphangiogenesis and lymph node metastasis by targeting ZIC2. Cell Oncol. 2020;43(2):249‐261. [DOI] [PubMed] [Google Scholar]
  • 195. Ye T, Yang M, Huang D, et al. MicroRNA‐7 as a potential therapeutic target for aberrant NF‐kappaB‐driven distant metastasis of gastric cancer. J Exp Clin Cancer Res. 2019;38(1):55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Yin Q, Wang PP, Peng R, Zhou H. MiR‐19a enhances cell proliferation, migration, and invasiveness through enhancing lymphangiogenesis by targeting thrombospondin‐1 in colorectal cancer. Biochem Cell Biol. 2019;97(6):731‐739. [DOI] [PubMed] [Google Scholar]
  • 197. Chang Y, Cui M, Fu X, et al. MiRNA‐155 regulates lymphangiogenesis in natural killer/T‐cell lymphoma by targeting BRG1. Cancer Biol Ther. 2019;20(1):31‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Zhou CF, Ma J, Huang L, et al. Cervical squamous cell carcinoma‐secreted exosomal miR‐221‐3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene. 2019;38(8):1256‐1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Zhang W, Hong R, Li L, et al. The chromosome 11q13.3 amplification associated lymph node metastasis is driven by miR‐548k through modulating tumor microenvironment. Mol Cancer. 2018;17(1):125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Sun Z, Ou C, Ren W, Xie X, Li X, Li G. Downregulation of long non‐coding RNA ANRIL suppresses lymphangiogenesis and lymphatic metastasis in colorectal cancer. Oncotarget. 2016;7(30):47536‐47555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Li X, Lv X, Li Z, et al. Long noncoding RNA ASLNC07322 functions in VEGF‐C expression regulated by Smad4 during colon cancer metastasis. Mol Ther Nucleic Acids. 2019;18:851‐862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. He W, Zhong G, Jiang N, et al. Long noncoding RNA BLACAT2 promotes bladder cancer‐associated lymphangiogenesis and lymphatic metastasis. J Clin Invest. 2018;128(2):861‐875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Liu HT, Ma RR, Lv BB, et al. LncRNA‐HNF1A‐AS1 functions as a competing endogenous RNA to activate PI3K/AKT signalling pathway by sponging miR‐30b‐3p in gastric cancer. Br J Cancer. 2020;122(12):1825‐1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Zheng S, Yang L, Zou Y, et al. Long non‐coding RNA HUMT hypomethylation promotes lymphangiogenesis and metastasis via activating FOXK1 transcription in triple‐negative breast cancer. J Hematol Oncol. 2020;13(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Chen C, He W, Huang J, et al. LNMAT1 promotes lymphatic metastasis of bladder cancer via CCL2 dependent macrophage recruitment. Nat Commun. 2018;9(1):3826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Chen C, Luo Y, He W, et al. Exosomal long noncoding RNA LNMAT2 promotes lymphatic metastasis in bladder cancer. J Clin Invest. 2020;130(1):404‐421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Karnezis T, Shayan R, Caesar C, et al. VEGF‐D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium. Cancer Cell. 2012;21(2):181‐195. [DOI] [PubMed] [Google Scholar]
  • 208. Proulx ST, Luciani P, Derzsi S, et al. Quantitative imaging of lymphatic function with liposomal indocyanine green. Cancer Res. 2010;70(18):7053‐7062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Clasper S, Royston D, Baban D, et al. A novel gene expression profile in lymphatics associated with tumor growth and nodal metastasis. Cancer Res. 2008;68(18):7293‐7303. [DOI] [PubMed] [Google Scholar]
  • 210. Dieterich LC, Kapaklikaya K, Cetintas T, et al. Transcriptional profiling of breast cancer‐associated lymphatic vessels reveals VCAM‐1 as regulator of lymphatic invasion and permeability. Int J Cancer. 2019;145(10):2804‐2815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Tutunea‐Fatan E, Majumder M, Xin X, Lala PK. The role of CCL21/CCR7 chemokine axis in breast cancer‐induced lymphangiogenesis. Mol Cancer. 2015;14:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Issa A, Le TX, Shoushtari AN, Shields JD, Swartz MA. Vascular endothelial growth factor‐C and C‐C chemokine receptor 7 in tumor cell‐lymphatic cross‐talk promote invasive phenotype. Cancer Res. 2009;69(1):349‐357. [DOI] [PubMed] [Google Scholar]
  • 213. Das S, Sarrou E, Podgrabinska S, et al. Tumor cell entry into the lymph node is controlled by CCL1 chemokine expressed by lymph node lymphatic sinuses. J Exp Med. 2013;210(8):1509‐1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Marcuzzi E, Angioni R, Molon B, Cali B. Chemokines and chemokine receptors: orchestrating tumor metastasization. Int J Mol Sci. 2018;20(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Padera TP, Kadambi A, di Tomaso E, et al. Lymphatic metastasis in the absence of functional intratumor lymphatics. Science. 2002;296(5574):1883‐1886. [DOI] [PubMed] [Google Scholar]
  • 216. Stanczyk M, Olszewski WL, Gewartowska M, Domaszewska‐Szostek A. Lack of functioning lymphatics and accumulation of tissue fluid/lymph in interstitial "lakes" in colon cancer tissue. Lymphology. 2010;43(4):158‐167. [PubMed] [Google Scholar]
  • 217. Olszewski WL, Stanczyk M, Gewartowska M, Domaszewska‐Szostek A, Durlik M. Lack of functioning intratumoral lymphatics in colon and pancreas cancer tissue. Lymphat Res Biol. 2012;10(3):112‐117. [DOI] [PubMed] [Google Scholar]
  • 218. Wong SY, Hynes RO. Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle. 2006;5(8):812‐817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Dihge L, Vallon‐Christersson J, Hegardt C, et al. Prediction of lymph node metastasis in breast cancer by gene expression and clinicopathological models: development and validation within a population‐based cohort. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25(21):6368‐6381. [DOI] [PubMed] [Google Scholar]
  • 220. Cochran AJ, Wen DR, Huang RR, Wang HJ, Elashoff R, Morton DL. Prediction of metastatic melanoma in nonsentinel nodes and clinical outcome based on the primary melanoma and the sentinel node. Mod Pathol Off J US Can Acad Pathol Inc. 2004;17(7):747‐755. [DOI] [PubMed] [Google Scholar]
  • 221. Karnezis T, Shayan R, Fox S, Achen MG, Stacker SA. The connection between lymphangiogenic signalling and prostaglandin biology: a missing link in the metastatic pathway. Oncotarget. 2012;3(8):893‐906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Le CP, Nowell CJ, Kim‐Fuchs C, et al. Chronic stress in mice remodels lymph vasculature to promote tumour cell dissemination. Nat Commun. 2016;7:10634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Kerjaschki D, Bago‐Horvath Z, Rudas M, et al. Lipoxygenase mediates invasion of intrametastatic lymphatic vessels and propagates lymph node metastasis of human mammary carcinoma xenografts in mouse. J Clin Invest. 2011;121(5):2000‐2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Gogineni A, Caunt M, Crow A, et al. Inhibition of VEGF‐C modulates distal lymphatic remodeling and secondary metastasis. PLOS One. 2013;8(7):e68755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Follain G, Herrmann D, Harlepp S, et al. Fluids and their mechanics in tumour transit: shaping metastasis. Nat Rev Cancer. 2020;20(2):107‐124. [DOI] [PubMed] [Google Scholar]
  • 226. Stylianopoulos T, Martin JD, Chauhan VP, et al. Causes, consequences, and remedies for growth‐induced solid stress in murine and human tumors. Proc Natl Acad Sci USA. 2012;109(38):15101‐15108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Jain RK, Martin JD, Stylianopoulos T. The role of mechanical forces in tumor growth and therapy. Annu Rev Biomed Eng. 2014;16:321‐346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Cornelison RC, Brennan CE, Kingsmore KM, Munson JM. Convective forces increase CXCR4‐dependent glioblastoma cell invasion in GL261 murine model. Sci Rep. 2018;8(1):17057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Huang YL, Tung CK, Zheng A, Kim BJ, Wu M. Interstitial flows promote amoeboid over mesenchymal motility of breast cancer cells revealed by a three dimensional microfluidic model. Integr Biol. 2015;7(11):1402‐1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Shields JD, Fleury ME, Yong C, Tomei AA, Randolph GJ, Swartz MA. Autologous chemotaxis as a mechanism of tumor cell homing to lymphatics via interstitial flow and autocrine CCR7 signaling. Cancer Cell. 2007;11(6):526‐538. [DOI] [PubMed] [Google Scholar]
  • 231. Li R, Serrano JC, Xing H, et al. Interstitial flow promotes macrophage polarization toward an M2 phenotype. Mol Biol Cell. 2018;29(16):1927‐1940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Harney AS, Arwert EN, Entenberg D, et al. Real‐time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage‐derived VEGFA. Cancer Discov. 2015;5(9):932‐943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Lee HJ, Diaz MF, Price KM, et al. Fluid shear stress activates YAP1 to promote cancer cell motility. Nat Commun. 2017;8:14122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Lee HJ, Ewere A, Diaz MF, Wenzel PL. TAZ responds to fluid shear stress to regulate the cell cycle. Cell Cycle. 2018;17(2):147‐153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Fares J, Fares MY, Khachfe HH, Salhab HA, Fares Y. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct Target Ther. 2020;5(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Moose DL, Krog BL, Kim TH, et al. Cancer cells resist mechanical destruction in circulation via RhoA/actomyosin‐dependent mechano‐adaptation. Cell Rep. 2020;30(11):3864‐3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Moore JE Jr, Bertram CD. Lymphatic system flows. Annu Rev Fluid Mech. 2018;50:459‐482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Ubellacker JM, Tasdogan A, Ramesh V, et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature. 2020;585(7823):113‐118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Dadiani M, Kalchenko V, Yosepovich A, et al. Real‐time imaging of lymphogenic metastasis in orthotopic human breast cancer. Cancer Res. 2006;66(16):8037‐8041. [DOI] [PubMed] [Google Scholar]
  • 240. Giampieri S, Manning C, Hooper S, Jones L, Hill CS, Sahai E. Localized and reversible TGFbeta signalling switches breast cancer cells from cohesive to single cell motility. Nature Cell Biol. 2009;11(11):1287‐1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Luzzi KJ, MacDonald IC, Schmidt EE, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153(3):865‐873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Alitalo A, Detmar M. Interaction of tumor cells and lymphatic vessels in cancer progression. Oncogene. 2012;31(42):4499‐4508. [DOI] [PubMed] [Google Scholar]
  • 243. Lee CK, Jeong SH, Jang C, et al. Tumor metastasis to lymph nodes requires YAP‐dependent metabolic adaptation. Science. 2019;363(6427):644‐649. [DOI] [PubMed] [Google Scholar]
  • 244. Li S, Li Q. Cancer stem cells, lymphangiogenesis, and lymphatic metastasis. Cancer Lett. 2015;357(2):438‐447. [DOI] [PubMed] [Google Scholar]
  • 245. Bao S, Wu Q, Sathornsumetee S, et al. Stem cell‐like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006;66(16):7843‐7848. [DOI] [PubMed] [Google Scholar]
  • 246. Yao XH, Ping YF, Chen JH, et al. Glioblastoma stem cells produce vascular endothelial growth factor by activation of a G‐protein coupled formylpeptide receptor FPR. J Pathol. 2008;215(4):369‐376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Krishnapriya S, Sidhanth C, Manasa P, et al. Cancer stem cells contribute to angiogenesis and lymphangiogenesis in serous adenocarcinoma of the ovary. Angiogenesis. 2019;22(3):441‐455. [DOI] [PubMed] [Google Scholar]
  • 248. Kim M, Koh YJ, Kim KE, et al. CXCR4 signaling regulates metastasis of chemoresistant melanoma cells by a lymphatic metastatic niche. Cancer Res. 2010;70(24):10411‐10421. [DOI] [PubMed] [Google Scholar]
  • 249. Kimura T, Sugaya M, Oka T, Blauvelt A, Okochi H, Sato S. Lymphatic dysfunction attenuates tumor immunity through impaired antigen presentation. Oncotarget. 2015;6(20):18081‐18093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Kataru RP, Ly CL, Shin J, et al. Tumor lymphatic function regulates tumor inflammatory and immunosuppressive microenvironments. Cancer Immunol Res. 2019;7(8):1345‐1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Lund AW, Wagner M, Fankhauser M, et al. Lymphatic vessels regulate immune microenvironments in human and murine melanoma. J Clin Invest. 2016;126(9):3389‐3402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Song E, Mao T, Dong H, et al. VEGF‐C‐driven lymphatic drainage enables immunosurveillance of brain tumours. Nature. 2020;577(7792):689‐694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Shields JD, Emmett MS, Dunn DB, et al. Chemokine‐mediated migration of melanoma cells towards lymphatics—a mechanism contributing to metastasis. Oncogene. 2007;26(21):2997‐3005. [DOI] [PubMed] [Google Scholar]
  • 254. Schietinger A, Philip M, Krisnawan VE, et al. Tumor‐specific T cell dysfunction is a dynamic antigen‐driven differentiation program initiated early during tumorigenesis. Immunity. 2016;45(2):389‐401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Tormoen GW, Crittenden MR, Gough MJ. Role of the immunosuppressive microenvironment in immunotherapy. Adv Radiat Oncol. 2018;3(4):520‐526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Lane RS, Femel J, Breazeale AP, et al. IFNgamma‐activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J Exp Med. 2018;215(12):3057‐3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Dieterich LC, Ikenberg K, Cetintas T, Kapaklikaya K, Hutmacher C, Detmar M. Tumor‐associated lymphatic vessels upregulate PDL1 to inhibit T‐cell activation. Front Immunol. 2017;8:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Hornyák L, Dobos N, Koncz G, et al. The role of indoleamine‐2,3‐dioxygenase in cancer development, diagnostics, and therapy. Front Immunol. 2018;9:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Broggi MAS, Maillat L, Clement CC, et al. Tumor‐associated factors are enriched in lymphatic exudate compared to plasma in metastatic melanoma patients. J Exp Med. 2019;216(5):1091‐1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Commerford CD, Dieterich LC, He Y, et al. Mechanisms of tumor‐induced lymphovascular niche formation in draining lymph nodes. Cell Rep. 2018;25(13):3554‐3563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. García‐Silva S, Benito‐Martín A, Sánchez‐Redondo S, et al. Use of extracellular vesicles from lymphatic drainage as surrogate markers of melanoma progression and BRAF (V600E) mutation. J Exp Med. 2019;216(5):1061‐1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Nogues L, Benito‐Martin A, Hergueta‐Redondo M, Peinado H. The influence of tumour‐derived extracellular vesicles on local and distal metastatic dissemination. Mol Aspects Med. 2018;60:15‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Ma Q, Dieterich LC, Detmar M. Multiple roles of lymphatic vessels in tumor progression. Curr Opin Immunol. 2018;53:7‐12. [DOI] [PubMed] [Google Scholar]
  • 264. Hirakawa S, Brown LF, Kodama S, Paavonen K, Alitalo K, Detmar M. VEGF‐C‐induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood. 2007;109(3):1010‐1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Garmy‐Susini B, Avraamides CJ, Desgrosellier JS, et al. PI3Kalpha activates integrin alpha4beta1 to establish a metastatic niche in lymph nodes. Proc Natl Acad Sci USA. 2013;110(22):9042‐9047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. Harrell MI, Iritani BM, Ruddell A. Tumor‐induced sentinel lymph node lymphangiogenesis and increased lymph flow precede melanoma metastasis. Am J Pathol. 2007;170(2):774‐786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. García‐Caballero M, Van de Velde M, Blacher S, et al. Modeling pre‐metastatic lymphvascular niche in the mouse ear sponge assay. Sci Rep. 2017;7:41494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Qian CN, Berghuis B, Tsarfaty G, et al. Preparing the "soil": the primary tumor induces vasculature reorganization in the sentinel lymph node before the arrival of metastatic cancer cells. Cancer Res. 2006;66(21):10365‐10376. [DOI] [PubMed] [Google Scholar]
  • 269. Ogawa F, Amano H, Eshima K, et al. Prostanoid induces premetastatic niche in regional lymph nodes. J Clin Invest. 2014;124(11):4882‐4894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Filippou PS, Karagiannis GS, Constantinidou A. Midkine (MDK) growth factor: a key player in cancer progression and a promising therapeutic target. Oncogene. 2020;39(10):2040‐2054. [DOI] [PubMed] [Google Scholar]
  • 271. Patel V, Marsh CA, Dorsam RT, et al. Decreased lymphangiogenesis and lymph node metastasis by mTOR inhibition in head and neck cancer. Cancer Res. 2011;71(22):7103‐7112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res Off J Am Assoc Cancer Res. 2015;21(4):687‐692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. Spranger S, Gajewski TF. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer. 2018;18(3):139‐147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Gardner A, Ruffell B. Dendritic cells and cancer immunity. Trends Immunol. 2016;37(12):855‐865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275. Saxena V, Li L, Paluskievicz C, et al. Role of lymph node stroma and microenvironment in T cell tolerance. Immunol Rev. 2019;292(1):9‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276. Niedbala W, Cai B, Liu H, Pitman N, Chang L, Liew FY. Nitric oxide induces CD4+CD25+ Foxp3 regulatory T cells from CD4+CD25 T cells via p53, IL‐2, and OX40. Proc Natl Acad Sci USA. 2007;104(39):15478‐15483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277. Knoblich K, Cruz Migoni S, Siew SM, et al. The human lymph node microenvironment unilaterally regulates T‐cell activation and differentiation. PLOS Biol. 2018;16(9):e2005046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Roberts EW, Broz ML, Binnewies M, et al. Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell. 2016;30(2):324‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Ruhland MK, Roberts EW, Cai E, et al. Visualizing synaptic transfer of tumor antigens among dendritic cells. Cancer Cell. 2020;37(6):786‐799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Mohammed SI, Torres‐Luquis O, Walls E, Lloyd F. Lymph‐circulating tumor cells show distinct properties to blood‐circulating tumor cells and are efficient metastatic precursors. Mol Oncol. 2019;13(6):1400‐1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Brown M, Assen FP, Leithner A, et al. Lymph node blood vessels provide exit routes for metastatic tumor cell dissemination in mice. Science. 2018;359(6382):1408‐1411. [DOI] [PubMed] [Google Scholar]
  • 282. Pereira ER, Kedrin D, Seano G, et al. Lymph node metastases can invade local blood vessels, exit the node, and colonize distant organs in mice. Science. 2018;359(6382):1403‐1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283. Naxerova K, Reiter JG, Brachtel E, et al. Origins of lymphatic and distant metastases in human colorectal cancer. Science. 2017;357(6346):55‐60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Gundem G, Van Loo P, Kremeyer B, et al. The evolutionary history of lethal metastatic prostate cancer. Nature. 2015;520(7547):353‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Gervasoni JE, Jr , Sbayi S, Cady B. Role of lymphadenectomy in surgical treatment of solid tumors: an update on the clinical data. Ann Surg Oncol. 2007;14(9):2443‐2462 [DOI] [PubMed] [Google Scholar]
  • 286. Faries MB, Thompson JF, Cochran AJ, et al. Completion dissection or observation for sentinel‐node metastasis in melanoma. N Engl J Med. 2017;376(23):2211‐2222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Dadras SS, Paul T, Bertoncini J, et al. Tumor lymphangiogenesis: a novel prognostic indicator for cutaneous melanoma metastasis and survival. Am J Pathol. 2003;162(6):1951‐1960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Schietroma C, Cianfarani F, Lacal PM, et al. Vascular endothelial growth factor‐C expression correlates with lymph node localization of human melanoma metastases. Cancer. 2003;98(4):789‐797. [DOI] [PubMed] [Google Scholar]
  • 289. Monteiro AC, Muenzner JK, Andrade F, et al. Gene expression and promoter methylation of angiogenic and lymphangiogenic factors as prognostic markers in melanoma. Mol Oncol. 2019;13(6):1433‐1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Dadras SS, Lange‐Asschenfeldt B, Velasco P, et al. Tumor lymphangiogenesis predicts melanoma metastasis to sentinel lymph nodes. Mod Pathol Off J US Can Acad Pathol Inc. 2005;18(9):1232‐1242. [DOI] [PubMed] [Google Scholar]
  • 291. Van den Eynden GG, Vandenberghe MK, van Dam PJ, et al. Increased sentinel lymph node lymphangiogenesis is associated with nonsentinel axillary lymph node involvement in breast cancer patients with a positive sentinel node. Clin Cancer Res Off J Am Assoc Cancer Res. 2007;13(18 Pt 1):5391‐5397. [DOI] [PubMed] [Google Scholar]
  • 292. Fisher B, Bauer M, Wickerham DL, et al. Relation of number of positive axillary nodes to the prognosis of patients with primary breast cancer. An NSABP update. Cancer. 1983;52(9):1551‐1557. [DOI] [PubMed] [Google Scholar]
  • 293. Foster RS Jr. The biologic and clinical significance of lymphatic metastases in breast cancer. Surg Oncol Clin N Am. 1996;5(1):79‐104. [PubMed] [Google Scholar]
  • 294. Jones D, Pereira ER, Padera TP. Growth and immune evasion of lymph node metastasis. Front Oncol. 2018;8:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Morton DL, Thompson JF, Cochran AJ, et al. Final trial report of sentinel‐node biopsy versus nodal observation in melanoma. N Engl J Med. 2014;370(7):599‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Harter P, Sehouli J, Lorusso D, et al. A randomized trial of lymphadenectomy in patients with advanced ovarian neoplasms. N Engl J Med. 2019;380(9):822‐832. [DOI] [PubMed] [Google Scholar]
  • 297. Erdem S, Bolli M, Muller SA, von Flue M, White R, Worni M. Role of lymphadenectomy in resectable pancreatic cancer. Langenbecks Arch Surg. 2020;405(7):889‐902. [DOI] [PubMed] [Google Scholar]
  • 298. Watanabe S. Lymph node dissection for lung cancer: past, present, and future. Gen Thorac Cardiovasc Surg. 2014;62(7):407‐414. [DOI] [PubMed] [Google Scholar]
  • 299. Ronca R, Benkheil M, Mitola S, Struyf S, Liekens S. Tumor angiogenesis revisited: regulators and clinical implications. Med Res Rev. 2017;37(6):1231‐1274. [DOI] [PubMed] [Google Scholar]
  • 300. Haibe Y, Kreidieh M, El Hajj H, et al. Resistance mechanisms to anti‐angiogenic therapies in cancer. Front Oncol. 2020;10:221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Hsu MC, Pan MR, Hung WC. Two birds, one stone: double hits on tumor growth and lymphangiogenesis by targeting vascular endothelial growth factor receptor 3. Cells. 2019;8(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Saif MW, Knost JA, Chiorean EG, et al. Phase 1 study of the anti‐vascular endothelial growth factor receptor 3 monoclonal antibody LY3022856/IMC‐3C5 in patients with advanced and refractory solid tumors and advanced colorectal cancer. Cancer Chemother Pharmacol. 2016;78(4):815‐824. [DOI] [PubMed] [Google Scholar]
  • 303. Falchook GS, Goldman JW, Desai J, et al. A first‐in‐human phase I study of VGX‐100, a selective anti‐VEGF‐C antibody, alone and in combination with bevacizumab in patients with advanced solid tumors. J Clin Oncol. 2014;32:2524. [Google Scholar]
  • 304. Prenen H, Vecchione L, Van Cutsem E. Role of targeted agents in metastatic colorectal cancer. Target Oncol. 2013;8(2):83‐96. [DOI] [PubMed] [Google Scholar]
  • 305. Yamakawa M, Doh SJ, Santosa SM, et al. Potential lymphangiogenesis therapies: Learning from current antiangiogenesis therapies—a review. Med Res Rev. 2018;38(6):1769‐1798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. Jones D. Parallels of resistance between angiogenesis and lymphangiogenesis inhibition in cancer therapy. Cells. 2020;9(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307. Sung HK, Morisada T, Cho CH, et al. Intestinal and peri‐tumoral lymphatic endothelial cells are resistant to radiation‐induced apoptosis. Biochem Biophys Res Commun. 2006;345(2):545‐551. [DOI] [PubMed] [Google Scholar]
  • 308. Allam O, Park KE, Chandler L, et al. The impact of radiation on lymphedema: a review of the literature. Gland Surg. 2020;9(2):596‐602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. Hu T, Wang Z, Shen W, Liang R, Yan D, Wei M. Recent advances in innovative strategies for enhanced cancer photodynamic therapy. Theranostics. 2021;11(7):3278‐3300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310. Agostinis P, Berg K, Cengel KA, et al. Photodynamic therapy of cancer: an update. CA Cancer J Clin. 2011;61(4):250‐281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311. Flegar L, Buerk B, Proschmann R, et al. Vascular‐targeted photodynamic therapy in unilateral low‐risk prostate cancer in Germany: 2‐yr single‐centre experience in a real‐world setting compared with radical prostatectomy. Eur Urol Focus. 2021. [DOI] [PubMed] [Google Scholar]
  • 312. Nogueira L, Tracey AT, Alvim R, et al. Developments in vascular‐targeted photodynamic therapy for urologic malignancies. Molecules. 2020;25(22). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Kilarski WW, Muchowicz A, Wachowska M, et al. Optimization and regeneration kinetics of lymphatic‐specific photodynamic therapy in the mouse dermis. Angiogenesis. 2014;17(2):347‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Tammela T, Saaristo A, Holopainen T, et al. Photodynamic ablation of lymphatic vessels and intralymphatic cancer cells prevents metastasis. Sci Transl Med. 2011;3(69):69ra11. [DOI] [PubMed] [Google Scholar]
  • 315. Bucher F, Bi Y, Gehlsen U, Hos D, Cursiefen C, Bock F. Regression of mature lymphatic vessels in the cornea by photodynamic therapy. Br J Ophthalmol. 2014;98(3):391‐395. [DOI] [PubMed] [Google Scholar]
  • 316. Muchowicz A, Wachowska M, Stachura J, et al. Inhibition of lymphangiogenesis impairs antitumour effects of photodynamic therapy and checkpoint inhibitors in mice. Eur J Cancer. 2017;83:19‐27. [DOI] [PubMed] [Google Scholar]
  • 317. Zhang S, Zhang D, Yi S, et al. The relationship of lymphatic vessel density, lymphovascular invasion, and lymph node metastasis in breast cancer: a systematic review and meta‐analysis. Oncotarget. 2017;8(2):2863‐2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318. Trevaskis NL, Kaminskas LM, Porter CJ. From sewer to saviour – targeting the lymphatic system to promote drug exposure and activity. Nat Rev Drug Discov. 2015;14(11):781‐803. [DOI] [PubMed] [Google Scholar]
  • 319. Chen J, Wang L, Yao Q, Ling R, Li K, Wang H. Drug concentrations in axillary lymph nodes after lymphatic chemotherapy on patients with breast cancer. Breast Cancer Res. 2004;6(4):R474‐R477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Wilson KD, Raney SG, Sekirov L, et al. Effects of intravenous and subcutaneous administration on the pharmacokinetics, biodistribution, cellular uptake and immunostimulatory activity of CpG ODN encapsulated in liposomal nanoparticles. Int Immunopharmacol. 2007;7(8):1064‐1075. [DOI] [PubMed] [Google Scholar]
  • 321. Supersaxo A, Hein WR, Steffen H. Effect of molecular weight on the lymphatic absorption of water‐soluble compounds following subcutaneous administration. Pharm Res. 1990;7(2):167‐169. [DOI] [PubMed] [Google Scholar]
  • 322. Rohner NA, Thomas SN. Flexible macromolecule versus rigid particle retention in the injected skin and accumulation in draining lymph nodes are differentially influenced by hydrodynamic size. ACS Biomater Sci Eng. 2017;3(2):153‐159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323. Khullar OV, Griset AP, Gibbs‐Strauss SL, et al. Nanoparticle migration and delivery of Paclitaxel to regional lymph nodes in a large animal model. J Am Coll Surg. 2012;214(3):328‐337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324. Sarin H. Physiologic upper limits of pore size of different blood capillary types and another perspective on the dual pore theory of microvascular permeability. J Angiogenes Res. 2010;2:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325. Oussoren C, Zuidema J, Crommelin DJ, Storm G. Lymphatic uptake and biodistribution of liposomes after subcutaneous injection. II. Influence of liposomal size, lipid compostion and lipid dose. Biochim Biophys Acta. 1997;1328(2):261‐272. [DOI] [PubMed] [Google Scholar]
  • 326. Schudel A, Francis DM, Thomas SN. Material design for lymph node drug delivery. Nat Rev Mater. 2019;4(6):415‐428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327. Manspeaker MP, Thomas SN. Lymphatic immunomodulation using engineered drug delivery systems for cancer immunotherapy. Adv Drug Deliv Rev. 2020;160:19‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328. Akamo Y, Mizuno I, Yotsuyanagi T, et al. Chemotherapy targeting regional lymph nodes by gastric submucosal injection of liposomal adriamycin in patients with gastric carcinoma. Jpn J Cancer Res Gann. 1994;85(6):652‐658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329. Kwon S, Velasquez FC, Rasmussen JC, et al. Nanotopography‐based lymphatic delivery for improved anti‐tumor responses to checkpoint blockade immunotherapy. Theranostics. 2019;9(26):8332‐8343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330. Francis DM, Manspeaker MP, Schudel A, et al. Blockade of immune checkpoints in lymph nodes through locoregional delivery augments cancer immunotherapy. Sci Transl Med. 2020;12(563). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331. Kim CK, Han JH. Lymphatic delivery and pharmacokinetics of methotrexate after intramuscular injection of differently charged liposome‐entrapped methotrexate to rats. J Microencapsul. 1995;12(4):437‐446. [DOI] [PubMed] [Google Scholar]
  • 332. Takakura Y, Matsumoto S, Hashida M, Sezaki H. Enhanced lymphatic delivery of mitomycin C conjugated with dextran. Cancer Res. 1984;44(6):2505‐2510. [PubMed] [Google Scholar]
  • 333. Kato S, Shirai Y, Sakamoto M, Mori S, Kodama T. Use of a lymphatic drug delivery system and sonoporation to target malignant metastatic breast cancer cells proliferating in the marginal sinuses. Sci Rep. 2019;9(1):13242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334. Laakkonen P, Porkka K, Hoffman JA, Ruoslahti E. A tumor‐homing peptide with a targeting specificity related to lymphatic vessels. Nat Med. 2002;8(7):751‐755. [DOI] [PubMed] [Google Scholar]
  • 335. Song N, Zhao L, Zhu M, Zhao J. Recent progress in LyP‐1‐based strategies for targeted imaging and therapy. Drug Deliv. 2019;26(1):363‐375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336. Wang Z, Yu Y, Ma J, et al. LyP‐1 modification to enhance delivery of artemisinin or fluorescent probe loaded polymeric micelles to highly metastatic tumor and its lymphatics. Mol Pharm. 2012;9(9):2646‐2657. [DOI] [PubMed] [Google Scholar]
  • 337. Yan Z, Wang F, Wen Z, et al. LyP‐1‐conjugated PEGylated liposomes: a carrier system for targeted therapy of lymphatic metastatic tumor. J Control Release. 2012;157(1):118‐125. [DOI] [PubMed] [Google Scholar]
  • 338. Luo G, Yu X, Jin C, et al. LyP‐1‐conjugated nanoparticles for targeting drug delivery to lymphatic metastatic tumors. Int J Pharm. 2010;385(1–2):150‐156. [DOI] [PubMed] [Google Scholar]
  • 339. Yan Z, Zhan C, Wen Z, et al. LyP‐1‐conjugated doxorubicin‐loaded liposomes suppress lymphatic metastasis by inhibiting lymph node metastases and destroying tumor lymphatics. Nanotechnology. 2011;22(41):415103. [DOI] [PubMed] [Google Scholar]

Articles from Medicinal Research Reviews are provided here courtesy of Wiley

RESOURCES