collaborative interplay between fgf-2 and vegf-c promotes ... · specific neutralizing antibody...

6
Collaborative interplay between FGF-2 and VEGF-C promotes lymphangiogenesis and metastasis Renhai Cao a,1 , Hong Ji a,1 , Ninghan Feng a , Yin Zhang a , Xiaojuan Yang a , Patrik Andersson a , Yuping Sun b , Katerina Tritsaris c , Anker Jon Hansen d,e , Steen Dissing c , and Yihai Cao a,f,2 a Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; b Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, Peoples Republic of China; c Department of Cellular and Molecular Medicine, Center for Healthy Aging, and d Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, Copenhagen 2200N, Denmark; e Novo Nordisk A/S, 2760 Måløv, Denmark; and f Department of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden Edited* by Tadamitsu Kishimoto, Immunology Frontier Research Center, Osaka University, Suita, Japan, and approved August 17, 2012 (received for review May 16, 2012) Interplay between various lymphangiogenic factors in promot- ing lymphangiogenesis and lymphatic metastasis remains poorly understood. Here we show that FGF-2 and VEGF-C, two lym- phangiogenic factors, collaboratively promote angiogenesis and lymphangiogenesis in the tumor microenvironment, leading to widespread pulmonary and lymph-node metastases. Coimplanta- tion of dual factors in the mouse cornea resulted in additive angiogenesis and lymphangiogenesis. At the molecular level, we showed that FGFR-1 expressed in lymphatic endothelial cells is a crucial receptor that mediates the FGF-2induced lymphangio- genesis. Intriguingly, the VEGFR-3mediated signaling was re- quired for the lymphatic tip cell formation in both FGF-2and VEGF-Cinduced lymphangiogenesis. Consequently, a VEGFR-3specic neutralizing antibody markedly inhibited FGF-2induced lymphangiogenesis. Thus, the VEGFR-3induced lymphatic endo- thelial cell tip cell formation is a prerequisite for FGF-2stimulated lymphangiogenesis. In the tumor microenvironment, the recipro- cal interplay between FGF-2 and VEGF-C collaboratively stimulated tumor growth, angiogenesis, intratumoral lymphangiogenesis, and metastasis. Thus, intervention and targeting of the FGF-2and VEGF-Cinduced angiogenic and lymphangiogenic synergism could be potentially important approaches for cancer therapy and prevention of metastasis. neovascularization | growth factors | signaling interplay | cancer spread | antiangiogenic therapy M etastasis is one of the hallmarks of malignant disease and is responsible for most cancer-related death (1, 2). Metas- tasis occurs in most cancer types and often affects multiple tis- sues and organs of the cancer-bearing host. Cancer metastasis is a complex disease that engages tight interactions between ma- lignant cells and the host microenvironment. The metastatic process consists of multiple steps of the tumorhost interaction, including: (i ) invasion of tumor cells into the surrounding tissue and dissemination from the primary site (3, 4); (ii ) intravasation of malignant cells in the circulation or lymphatic systems (59); (iii ) transport of tumor cells to the distal tissues and organs; (iv) extravasation of malignant cells from blood or lymphatic vessels (10); (v) formation of the initial metastatic niche and micro- metastatic foci in the distal tissues and organs (11); and (vi ) regrowth of metastatic foci into a clinical detectable mass. In some cases, tumor cells can escape from the primary metastatic site to further spread to other tissues and organs (5). At the primary site, intra- and peritumoral blood and lymphatic microvessels are the crucial structures that facilitate tumor cell dissemination; probably both vessel density and structures are key determinants for tumor cell dissemination (57). Once arriving at the distal site, tumor cells often form micrometastases caving around the existing blood vessels in which nutrients and O 2 can be freely diffused to tumor cells and angiogenesis can be initiated if further growth begins (12, 13). Thus, the entire metastatic process is tightly linked to the intimate interactions between tumor cells and hemvascular and lymphvascular systems. Tumors produce various angiogenic factors to stimulate angiogenesis and lym- phangiogenesis (1421). Both VEGF-C and FGF-2 are commonly expressed in various tumor tissues and their expression levels have been correlated with tumor growth, progression, and metastasis (22, 23). Although the individual roles of angiogenic factors in promoting tumor growth and metastasis are relatively well stud- ied, the cross-talk between various angiogenic factors in the tumor environment remains poorly understood. In this study, we provide a unique example of the intimate interaction between FGF-2 and VEGF-C in promoting lymphangiogenesis and metastasis. Our ndings may provide important information for therapeutic development of novel strategies for the treatment of cancer me- tastasis by intervention of the interaction between FGF-2 and VEGF-C. Results FGF-2 and VEGF-C Collaboratively Promote Corneal Angiogenesis and Lymphangiogenesis. To study interplay between various angio- genic factors in promoting lymphangiogenesis, we chose FGF-2 and VEGF-C, two commonly expressed angiogenic factors, in the tumor microenvironment for our study. First, we tested their angiogenic and lymphangiogenic effects in the mouse corneal micropocket assay (24). As the corneal tissue remains avascular, implantation of angiogenic factors in the cornea would allow us to study a specic factor-induced lymphangiogenesis and vascu- lar structures. FGF-2 and VEGF-C, together with a slow-release polymer composed of sucralfate and hydron, were implanted into the corneal micropockets of C57BL/6 mice. At day 6 post- implantation, gross-examination of corneas showed that FGF-2 displayed a robust angiogenic response (Fig. 1A). As expected, VEGF-C was also able to stimulate corneal angiogenesis, albeit the angiogenic activity was substantially weaker relative to FGF- 2 (Fig. 1A). Surprisingly, coimplantation of micropellets con- taining FGF-2 and VEGF-C resulted in angiogenic synergism (Fig. 1A). The slow-release polymer-implanted corneas without angiogenic factors were used as a negative control and lacked the ability of triggering an angiogenic response (Fig. 1A), sug- gesting that the implantation procedure per se does not induce angiogenesis. To further study synergisms of FGF-2and VEGF-Cinduced angiogenesis and lymphangiogenesis, at-mounted mouse corneas Author contributions: Y.C. designed research; R.C., H.J., N.F., Y.Z., X.Y., P.A., Y.S., K.T., and S.D. performed research; A.J.H. contributed new reagents/analytic tools; R.C., H.J., N.F., Y.Z., X.Y., P.A., K.T., S.D., and Y.C. analyzed data; and Y.C. wrote the paper. The authors declare no conict of interest. *This Direct Submission article had a prearranged editor. 1 R.C. and H.J. contributed equally to this work. 2 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1208324109/-/DCSupplemental. 1589415899 | PNAS | September 25, 2012 | vol. 109 | no. 39 www.pnas.org/cgi/doi/10.1073/pnas.1208324109 Downloaded by guest on October 22, 2020

Upload: others

Post on 04-Aug-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Collaborative interplay between FGF-2 and VEGF-C promotes ... · specific neutralizing antibody markedly inhibited FGF-2–induced lymphangiogenesis. Thus, the VEGFR-3–induced

Collaborative interplay between FGF-2 and VEGF-Cpromotes lymphangiogenesis and metastasisRenhai Caoa,1, Hong Jia,1, Ninghan Fenga, Yin Zhanga, Xiaojuan Yanga, Patrik Anderssona, Yuping Sunb,Katerina Tritsarisc, Anker Jon Hansend,e, Steen Dissingc, and Yihai Caoa,f,2

aDepartment of Microbiology, Tumor, and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; bDepartment of Oncology, Jinan Central Hospital,Shandong University, Jinan, Shandong 250013, People’s Republic of China; cDepartment of Cellular and Molecular Medicine, Center for Healthy Aging, anddDepartment of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, Copenhagen 2200N, Denmark; eNovo Nordisk A/S, 2760Måløv, Denmark; and fDepartment of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden

Edited* by Tadamitsu Kishimoto, Immunology Frontier Research Center, Osaka University, Suita, Japan, and approved August 17, 2012 (received for reviewMay 16, 2012)

Interplay between various lymphangiogenic factors in promot-ing lymphangiogenesis and lymphatic metastasis remains poorlyunderstood. Here we show that FGF-2 and VEGF-C, two lym-phangiogenic factors, collaboratively promote angiogenesis andlymphangiogenesis in the tumor microenvironment, leading towidespread pulmonary and lymph-node metastases. Coimplanta-tion of dual factors in the mouse cornea resulted in additiveangiogenesis and lymphangiogenesis. At the molecular level, weshowed that FGFR-1 expressed in lymphatic endothelial cells isa crucial receptor that mediates the FGF-2–induced lymphangio-genesis. Intriguingly, the VEGFR-3–mediated signaling was re-quired for the lymphatic tip cell formation in both FGF-2– andVEGF-C–induced lymphangiogenesis. Consequently, a VEGFR-3–specific neutralizing antibody markedly inhibited FGF-2–inducedlymphangiogenesis. Thus, the VEGFR-3–induced lymphatic endo-thelial cell tip cell formation is a prerequisite for FGF-2–stimulatedlymphangiogenesis. In the tumor microenvironment, the recipro-cal interplay between FGF-2 and VEGF-C collaboratively stimulatedtumor growth, angiogenesis, intratumoral lymphangiogenesis,and metastasis. Thus, intervention and targeting of the FGF-2–and VEGF-C–induced angiogenic and lymphangiogenic synergismcould be potentially important approaches for cancer therapy andprevention of metastasis.

neovascularization | growth factors | signaling interplay | cancer spread |antiangiogenic therapy

Metastasis is one of the hallmarks of malignant disease and isresponsible for most cancer-related death (1, 2). Metas-

tasis occurs in most cancer types and often affects multiple tis-sues and organs of the cancer-bearing host. Cancer metastasis isa complex disease that engages tight interactions between ma-lignant cells and the host microenvironment. The metastaticprocess consists of multiple steps of the tumor–host interaction,including: (i) invasion of tumor cells into the surrounding tissueand dissemination from the primary site (3, 4); (ii) intravasationof malignant cells in the circulation or lymphatic systems (5–9);(iii) transport of tumor cells to the distal tissues and organs; (iv)extravasation of malignant cells from blood or lymphatic vessels(10); (v) formation of the initial metastatic niche and micro-metastatic foci in the distal tissues and organs (11); and (vi)regrowth of metastatic foci into a clinical detectable mass. Insome cases, tumor cells can escape from the primary metastaticsite to further spread to other tissues and organs (5).At the primary site, intra- and peritumoral blood and lymphatic

microvessels are the crucial structures that facilitate tumor celldissemination; probably both vessel density and structures are keydeterminants for tumor cell dissemination (5–7). Once arriving atthe distal site, tumor cells often form micrometastases cavingaround the existing blood vessels in which nutrients and O2 can befreely diffused to tumor cells and angiogenesis can be initiated iffurther growth begins (12, 13). Thus, the entire metastatic processis tightly linked to the intimate interactions between tumor cells

and hemvascular and lymphvascular systems. Tumors producevarious angiogenic factors to stimulate angiogenesis and lym-phangiogenesis (14–21). Both VEGF-C and FGF-2 are commonlyexpressed in various tumor tissues and their expression levels havebeen correlated with tumor growth, progression, and metastasis(22, 23). Although the individual roles of angiogenic factors inpromoting tumor growth and metastasis are relatively well stud-ied, the cross-talk between various angiogenic factors in the tumorenvironment remains poorly understood. In this study, we providea unique example of the intimate interaction between FGF-2and VEGF-C in promoting lymphangiogenesis and metastasis.Our findings may provide important information for therapeuticdevelopment of novel strategies for the treatment of cancer me-tastasis by intervention of the interaction between FGF-2 andVEGF-C.

ResultsFGF-2 and VEGF-C Collaboratively Promote Corneal Angiogenesis andLymphangiogenesis. To study interplay between various angio-genic factors in promoting lymphangiogenesis, we chose FGF-2and VEGF-C, two commonly expressed angiogenic factors, inthe tumor microenvironment for our study. First, we tested theirangiogenic and lymphangiogenic effects in the mouse cornealmicropocket assay (24). As the corneal tissue remains avascular,implantation of angiogenic factors in the cornea would allow usto study a specific factor-induced lymphangiogenesis and vascu-lar structures. FGF-2 and VEGF-C, together with a slow-releasepolymer composed of sucralfate and hydron, were implantedinto the corneal micropockets of C57BL/6 mice. At day 6 post-implantation, gross-examination of corneas showed that FGF-2displayed a robust angiogenic response (Fig. 1A). As expected,VEGF-C was also able to stimulate corneal angiogenesis, albeitthe angiogenic activity was substantially weaker relative to FGF-2 (Fig. 1A). Surprisingly, coimplantation of micropellets con-taining FGF-2 and VEGF-C resulted in angiogenic synergism(Fig. 1A). The slow-release polymer-implanted corneas withoutangiogenic factors were used as a negative control and lackedthe ability of triggering an angiogenic response (Fig. 1A), sug-gesting that the implantation procedure per se does not induceangiogenesis.To further study synergisms of FGF-2– and VEGF-C–induced

angiogenesis and lymphangiogenesis, flat-mounted mouse corneas

Author contributions: Y.C. designed research; R.C., H.J., N.F., Y.Z., X.Y., P.A., Y.S., K.T., andS.D. performed research; A.J.H. contributed new reagents/analytic tools; R.C., H.J., N.F.,Y.Z., X.Y., P.A., K.T., S.D., and Y.C. analyzed data; and Y.C. wrote the paper.

The authors declare no conflict of interest.

*This Direct Submission article had a prearranged editor.1R.C. and H.J. contributed equally to this work.2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1208324109/-/DCSupplemental.

15894–15899 | PNAS | September 25, 2012 | vol. 109 | no. 39 www.pnas.org/cgi/doi/10.1073/pnas.1208324109

Dow

nloa

ded

by g

uest

on

Oct

ober

22,

202

0

Page 2: Collaborative interplay between FGF-2 and VEGF-C promotes ... · specific neutralizing antibody markedly inhibited FGF-2–induced lymphangiogenesis. Thus, the VEGFR-3–induced

were immunohistologically stained with CD31 and LYVE-1(lymphatic vessel endothelial hyaluronan receptor-1), two spe-cific pan-markers for blood vessel endothelial cells and lymphaticendothelial cells (LECs), respectively. To comparatively andoptimally quantify the results, the circumferential area of eachflat-mounted cornea was divided into clock-hours (24), where theimplanted pellet was positioned at the 6 h of the anterior front(Fig. 1B). Angiogenic and lymphangiogenic responses in eachcornea of various groups were compared at 4.5–7.5 h, 7.5–10.5 h,10.5–1.5 h, and 1.5–4.5 h positions. Consistent with gross-examinations, CD31 staining demonstrated that FGF-2 andVEGF-C collaboratively induced corneal neovascularization(Fig. 1 C and E). LYVE-1 staining showed that both FGF-2 andVEGF-C induced relative robust lymphangiogenic responses(Fig. 1 C and F). Interestingly, an additive lympangiogenic effectbetween FGF-2 and VEGF-C was detected in all clock-hourpositions of the cornea (Fig. 1 C and F). The additive angiogenicand lymphangiogenic effects of FGF-2 and VEGF-C were fur-ther validated by quantifying the entire circumferential area ofeach cornea (Fig. 1 G and H). Double immunohistochemicalstaining showed that CD31+ and LYVE-1+ vessels lacked over-lapping signals, demonstrating high specificities of these anti-bodies. Staining with a VEGFR-3 specific antibody furthervalidated the fact that lymphatic vessels and blood vessels aretwo independent vascular structures (Fig. 1D). Taken together,FGF-2 and VEGF-C collaboratively stimulated corneal angio-genesis and lymphangiogenesis.

FGF-2 and VEGF-C Independently Stimulate LEC Proliferation andMigration. To study the cellular basis of lymphangiogenic syner-gism between FGF-2 and VEGF-C, primary LECs were used forin vitro proliferation and migration assays. As expected, VEGF-C significantly stimulated human LEC (hLEC) proliferation(Fig. 2A). Interestingly, FGF-2 also potently induced hLECproliferation, suggesting that FGF-2 directly acts on hLEC toinduce lymphangiogenic activity. Costimulation of hLECs withFGF-2 and VEGF-C resulted in significantly increased cellproliferation relative to FGF-2 or VEGF-C alone (Fig. 2A).Consistent with hLEC proliferation, FGF-2 was able to directlyinduce phosphorylation of several intracellular signaling com-ponents in LECs, including Akt, Erk, and ribosomal protein S6,further supporting its direct effect on hLECs (Fig. 2B). Similarly,stimulation of hLECs with VEGF-C also led to activation ofthese intracellular signaling components (Fig. 2C).RT-PCR analysis demonstrated that Fgfr-1 was expressed in

hLECs (Fig. 2D), whereas other forms of Fgfrs including Fgfr-2,Fgfr-3, and Fgfr-4 were weakly expressed or remained undetect-able. To study the molecular mechanism that underlies thelymphangiogenic synergism, reciprocal regulation of Fgfr-1 andVegfr-3 in hLECs by both VEGF-C and FGF-2 were analyzed.Interestingly, Fgfr-1 expression was up-regulated by both VEGF-C and FGF-2 (Fig. 2E). Similarly, significant up-regulation ofVegfr-3 was detected in FGF-2– and VEGF-C–stimulated LECs(Fig. 2F). These findings suggested that FGF-2 and VEGF-Creciprocally amplify their receptor-mediated signaling systems inLECs, leading to additive lymphangiogenic activity when bothfactors are coexposed to LECs.We next used siRNA specific for Fgfr-1 to inhibit FGF-2–in-

duced hLEC proliferation. As expected, siRNA specific for Fgfr-1, but not scrambled siRNA, significantly suppressed FGF-2–induced hLEC proliferation (Fig. 2G). To further study potentialinvolvement of VEGFR-3 and FGFR-1 in mediating FGF-2–induced LEC proliferation and migration, anti–VEGFR-3 andanti-FGFR-1 neutralizing antibodies (hereafter called VEGFR-3blockade and FGFR-1 blockade) (25, 26) were used to func-tionally block the VEGFR-3 signaling system or the FGFR-1signaling system. FGF-2 displayed potent proliferative and mi-gratory effects on primary mouse LECs (mLECs), which were

PBS FGF-2 VEGF-C FGF-2+VEGF-C

LYV

E-1

/CD

31LY

VE

-1

A B

C PBS FGF-2 VEGF-C FGF-2+VEGF-C

CD

31

CD

31V

EG

FR3

CD

31/

VE

GFR

-3

D

P PPP

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

P - - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -

P- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - -- - - - - - -

P- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - -- -

P

E

F

P

0

6

2

4

8

0

6

2

4

8

G H

2B

lood

ves

sel a

rea

per c

orne

a (m

m )

Lym

phat

ic v

esse

l are

ape

r cor

nea

(mm

)2

0

3

FGF-

2VEGF-

C

1

PBS

- - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - -

CD314.5-7.5 h 7.5-10.5 h 10.5-1.5 h 1.5-4.5 h

2P < 0.05

FGF-

2+VEGF-

C

2B

lood

ves

sel a

rea

per c

orne

a (m

m )

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

- - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - -

LYVE-1

0

3

1

2

Lym

phat

ic v

esse

l are

ape

r cor

nea

(mm

)2

4.5-7.5h

10.5-1.5h

1.5-4.5h

7.5-

10.5

h

P < 0.01P < 0.001

P < 0.001P < 0.001

P < 0.001

P < 0.01P < 0.001P < 0.001

P < 0.01P < 0.001

P < 0.01P < 0.01

P < 0.001P < 0.001

P < 0.001P < 0.001P < 0.001

P < 0.01P < 0.001

P < 0.05P < 0.01

P < 0.001P < 0.01

P < 0.001

P < 0.001P < 0.001

P < 0.001P < 0.001

P < 0.001

P < 0.01P < 0.001P < 0.001

P < 0.01P < 0.001

P < 0.001P < 0.001P < 0.001

P < 0.001P < 0.001

P < 0.05P < 0.001P < 0.001

P < 0.001P < 0.001

P < 0.001

P < 0.05P < 0.001P < 0.001

P < 0.001P < 0.001

P < 0.001

400 μm 400 μm 400 μm 400 μm

100 μm 100 μm 100 μm 100 μm

100 μm 100 μm 100 μm 100 μm

100 μm 100 μm 100 μm 100 μm

100 μm

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

FGF-

2VEGF-

C

PBS

FGF-

2+VEGF-

C

4.5-7.5 h 7.5-10.5 h 10.5-1.5 h 1.5-4.5 h

Fig. 1. FGF-2 and VEGF-C collaboratively stimulate hem- and lym-phangiogensis. (A) Micropellets containing FGF-2, VEGF-C, or FGF-2 plusVEGF-C together with the slow-release polymer were implanted intomicropockets of mouse corneas (n = 5–6 per group). At day 6 after im-plantation, corneal neovascularization was examined and photographed.Slow-release polymer containing PBS was used as a negative control. Pindicates the position of the implanted pellet. (B) Schematic diagramdemonstrates the flat-mounted cornea and clock-hours of the circumfer-ential cornea. (C ) A representative cornea from each group that weredouble immunostained with CD31 (red) and LYVE-1 (green), whichshowed no overlapping signals. (D) Double immunostaining of cornealblood and lymphatic vessels using VEGFR-3 (green) and CD31 (red) specificantibodies. (E ) Quantification of corneal CD31+ blood vessels at differentclock hours (n = 5–6 /group). (F ) Quantification of corneal LYVE-1 positivelympghatic vessels at different clockhours (n = 5–6 per group). (G)Quantification of the total CD31+ vessels in the entire of circumferentialarea of each cornea (n = 5–6 per group). (H) Quantification of the totalLYVE-1+ lymphatic vessels in the entire of circumferential area of eachcornea (n = 5–6 per group).

Cao et al. PNAS | September 25, 2012 | vol. 109 | no. 39 | 15895

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Oct

ober

22,

202

0

Page 3: Collaborative interplay between FGF-2 and VEGF-C promotes ... · specific neutralizing antibody markedly inhibited FGF-2–induced lymphangiogenesis. Thus, the VEGFR-3–induced

completely neutralized by FGFR-1 blockade, but not byVEGFR-3 blockade (Fig. 2 H and I). Similarly, VEGF-C–inducedproliferative and migratory activities were only inhibited byVEGFR-3 blockade, but not by FGFR-1 blockade (Fig. 2 Hand I). These findings further support the fact that FGF-2 andVEGFR-3 independently mediate their specific ligand-trig-gered lymphangiogenic activity.

VEGFR-3–Mediated Lymphatic Tips Cell Formation Are Prerequisitefor FGF-2–Induced Lymphangiogenesis. To study the role of theFGFR-1–mediated signaling system in stimulation of hem- andlymphangiogenesis, a FGFR-1 neutralizing antibody was used fortreatment. Interestingly, FGFR-1 blockade significantly inhibitedFGFR-2 plus VEGF-C–induced angiogenesis and lymphangio-genesis (Fig. 2 J–M). Quantification of CD31+ vascular structuresshowed that ∼50% reduction of corneal neovascularization byFGFR-1 blockade (Fig. 2L). A similar potent antilymphan-giogenic activity was also observed in FGFR-1 blockade-treatedcorneas (Fig. 2M).Expectedly, VEGFR-3 blockade completely blocked VEGF-

C–induced lymphangiogenesis and hemangiogenesis (Fig. 3 A, B,D, and F), suggesting the VEGFR-3–transduced signals are es-sential for VEGF-C–stimulated angiogenesis and lymphangio-genesis. Surprisingly, VEGFR-3 blockade almost entirelyinhibited FGF-2–induced lymphangiogenesis in this in vivomodel (Fig. 3 B and D). These findings are unexpected becausethe same VEGFR-3 blockade did not significantly affect FGF-2–induced LEC proliferation and migration in vitro.To gain a mechanistic insight by which the VEGFR-3 signal-

ing system is a prerequisite for FGF-2–induced lymphangio-genesis, we analyzed the tip cell formation at the leading edge ofgrowing lymphatics. Recent studies have shown that the for-mation of tip endothelial cells in growing angiogenic and lym-phangiogenic vessels is an essential process for development ofvascular networks (27–29). Indeed, both FGF-2 and VEGF-Cwere able to induce endothelial tips at the leading front of thegrowing lymphatics (Fig. 3C). Quantification analysis showedthat coimplantation of FGF-2 and VEGF-C resulted in an ad-ditive effect on the formation of LEC tips (Fig. 3 C and E).Surprisingly, VEGFR-3 blockade not only completely inhibitedthe lymphatic tip cell formation in the VEGF-C–induced lym-phatic vessels, but also in FGF-2–stimulated lymphatics (Fig. 3 Cand E). Similarly, FGF-2 plus VEGF-C–induced tip cell for-mation at the leading edge of the lymphatics was also completelysuppressed by VEGFR-3 blockade. Unlike antilymphangiogenicactivity, VEGFR-3 blockade did not inhibit FGF-2–inducedblood vessel growth, suggesting that FGF-2 stimulates angio-genesis via a VEGFR-3 independent pathway. Interestingly,VEGFR-3 blockade did not significantly affect the VEGF-C–induced hemangiogenesis, supporting the fact that the in-teraction of VEGF-C with VEGFR-2 but not with VEGFR-3 isthe key pathway for triggering angiogenesis (13). These findingsdemonstrate that VEGFR-3–induced tip formation is a pre-requisite for FGF-2–stimulated lymphangiogenesis but not forhemangiogenesis.

FGF-2 and VEGF-C Collaboratively Promote Tumor Hemangiogenesisand Lymphangiogenesis. The lymphangiogenic synergism of FGF-2 and VEGF-C in mouse corneas promoted us to study its rel-evance to tumor lymphatic vessel development and lymphaticmetastasis. We transfected a murine fibrosarcoma cell line toexpress secreted forms of FGF-2 and VEGF-C, as previouslydescribed (18, 30). As expected, both FGF-2 and VEGF-C sig-nificantly promoted tumor growth when subcutaneously implantedin immunodeficient SCID mice (Fig. 4A). Interestingly, im-plantation of the same number of tumor cells consisting of 50%FGF-2 and 50% VEGF-C cells resulting in a markedly acceler-ated tumor growth rate (Fig. 4A), demonstrating that these two

A B C

D

E

F

0

0.2

0.4

Scrambled

FGF-2 alone

FGF-2+

siFGFR-1

0.6

-FGF-2

- - -

- - -

- - -

- - -

- - -

- - -

- - -

- -

Pro

lifer

atio

nO

D (4

50-6

30 n

m) P < 0.001

320 bp

242 bp

FGFR-1DNA ladder NTC

FGFR-1

P

P

- - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - --- - -

-- - -

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - P

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - -- - - - ----

- ---

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

P

- - - - - - - - - - - - - - -

- - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - -

CD31 LYVE-1/CD31LYVE-1

- - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - -Ant

i-FG

FR-1

NT

0

4

8

2

6

2B

lood

ves

sel a

rea

per c

orne

a (m

m )

P < 0.001

0

4

8

2

6

Lym

phat

ic v

esse

l are

ape

r cor

nea

(mm

)2

P < 0.001

G

H I

J

400 μm 100 μm 100 μm 100 μm

400 μm 100 μm 100 μm 100 μm

0

0.20.4

Control

FGF-2

FGF-2+

VEGF-C

Pro

lifer

atio

nO

D (4

50-6

30 n

m)

0.6

P < 0.001

P < 0.001P < 0.001P < 0.001

VEGF-C

- - - - - -

- - - - - -

- - - p-ERK1/2p-Akt

GAPDHp-rpS6

-FGF-2 5 10 20 30 45 60(min)

FGF-2

p-ERK1/2p-Akt

GAPDH

-VEGF-C

p-rpS6

VEGF-C

FGFR

-1R

elat

ive

quan

tific

atio

n

0

1

2

3

- - - -

- - - -

- - - -

- - -

P < 0.001

P < 0.01

P < 0.001

- - - -

- - - -

- - - -

- - -

P < 0.001

24 24 48 24 48 (h)FGF-2 VEGF-CPBS (10 ng/ml)

VE

GFR

-3R

elat

ive

quan

tific

atio

n

0

0.5

1.5

2

- - - -

- - - -

- - - -

- - -

P < 0.01

P < 0.01

P < 0.01

- - - -

- - - -

- - - -

- - -

P < 0.01

24 24 48 24 48 (h)FGF-2 VEGF-CPBS (10 ng/ml)

Num

ber o

f mig

rate

d ce

lls

0

20

40

60

80

- - - -

- - - -

- - - -

- - -

- - - -

- - - -

- - - -

- - -

P < 0.01P < 0.001

P < 0.001

P < 0.001NS

NS

1

K

L M

Control

Pro

lifer

atio

n (O

D 4

90 n

m)

0

FGF-2

FGF-2+Anti-VEGFR-3

0.2

0.4

0.6

VEGF-C

VEGF-C+Anti-F

GFR-1

- - - -

- - - -

- - - -

- - -

P < 0.01P < 0.001

P < 0.001

VEGF-C+Anti-V

EGFR-3

- - - -

- - - -

- - - -

- - -

FGF-2+Anti-FGFR-1

P < 0.001NS

NS

P < 0.01 5 10 20 30 45 60(min)

Control

FGF-2

FGF-2+Anti-VEGFR-3

VEGF-C

VEGF-C+Anti-F

GFR-1

VEGF-C+Anti-V

EGFR-3

FGF-2+Anti-FGFR-1

Anti-FGFR-1NT Anti-FGFR-1NT

Fig. 2. In vitro LEC activity and signaling and suppression of hem- andlymphangiogenesis by FGFR-1 blockade. (A) Stimulation of hLEC pro-liferation by FGF-2, VEGF-C, FGF-2 plus VEGF-C or buffer alone (16 samplesper group). (B) Immunoblot analysis of activation of signaling components inhLECs by FGF-2. GDPDH showed the standard level of sample loading. (C)Immunoblot analysis of activation of signaling components in hLECs byVEGF-C. GDPDH showed the standard level of sample loading. (D) Amplifi-cation of Fgfr-1 by reverse-transcription PCR using cDNA extracted fromLECs. (E) qPCR analysis of Fgfr-1 mRNA expression levels in hLECs stimulatedby FGF-2 or VEGF-C. PBS-stimulated cells were used as a control. (F) Quan-titative PCR analysis of Vegfr-3 mRNA expression levels in hLECs stimulatedby FGF-2 or VEGF-C. PBS-stimulated cells were used as a control. (G) Fgfr-1specific siRNA substantially inhibited FGF-2–induced LEC proliferation. Ascrambled nucleotide sequence was used as a control. (H) Inhibition of FGF-2– or VEGF-C–induced mLEC proliferation by FGFR-1 or VEGFR-3 blockade(six samples per group). (I) Inhibition of FGF-2– or VEGF-C–induced mLECmigration by FGFR-1 or VEGFR-3 blockade (six samples per group). (J) Sup-pression of FGF-2 plus VEGF-C–induced corneal neovascularization by FGFR-1blockade. (K) Suppression of FGF-2 plus VEGF-C–induced corneal heman-giogenesis (CD31+ blood vessels) and lymphangiogenesis (LYVE-1+ lymphaticvessels) by FGFR-1 blockade. P, pellet. (L) Quantification of antiangiogenicactivity by FGFR-1 blockade (n = 5–6 per group). (M) Quantification of anti-lymphangiogenic activity by FGFR-1 blockade (n = 5–6 per group).

15896 | www.pnas.org/cgi/doi/10.1073/pnas.1208324109 Cao et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

22,

202

0

Page 4: Collaborative interplay between FGF-2 and VEGF-C promotes ... · specific neutralizing antibody markedly inhibited FGF-2–induced lymphangiogenesis. Thus, the VEGFR-3–induced

factors collaboratively stimulate tumor growth. Consistent withthe markedly accelerated tumor growth rate, FGF-2– and VEGF-C–coexpressing tumors contained a higher density of disorga-nized vascular networks compared with tumors expressing FGF-2,VEGF-C, or vector alone (Fig. 4 B and C). These findings dem-onstrate that additive stimulation of tumor angiogenesis by FGF-2and VEGF-C significantly contributes to the accelerated tumorgrowth rate.We next analyzed tumor lymphatic vessels using LYVE-1 as

a specific marker. In the vector-transfected control tumors,LYVE-1+ lymphatic vessels were primarily distributed in theperitumoral area and intratumoral lymphatics were rarely de-tectable (Fig. 5A). In contrast, both FGF-2 and VEGF-C tumorssignificantly stimulated intratumoral lymphatic vessel growth,albeit VEGF-C exhibited more potent lymphangiogenic activitythan FGF-2 (Fig. 5A). Surprisingly, tumors expressing both fac-tors contained a markedly higher density of intratumoral lym-phatic vessels than tumors expressing FGF-2 or VEGF-C alone(Fig. 5A). Quantification analysis showed that FGF-2 plus

VEGF-C collaboratively promoted tumor lymphangiogenesis(Fig. 5B). FGF-2 plus VEGF-C–induced lymphatic vesselsappeared as disorganized vascular networks compared withtumors expressing FGF-2, VEGF-C or vector alone (Fig. 5A).Additionally, peritumoral lymphatics became highly dilated andoften contained EGFP+ tumor cells (Fig. S1). These findingsfurther validate the mouse corneal data that FGF-2 and VEGF-C collaboratively stimulated lymphangiogenesis, which occurredunder pathological conditions such as in the tumor environment.

FGF-2 and VEGF-C Collaboratively Stimulate Hematogenous andLymphatic Metastasis. Knowing that FGF-2 and VEGF-C collab-oratively stimulated tumor hem- and lymphangiogenesis, andinduced disorganization, we next studied their potentials inpromoting hematogenous and lymphatic metastasis. Necropathyof tumor-bearing mice showed that ∼70% of FGF-2 plus VEGF-C tumor-bearing mice had pulmonary metastases on the surfaceof their lungs (Fig. 4 D and E). In contrast, no pulmonary me-tastasis was detected in FGF-2 tumor-bearing mice (Fig. 4 D andE) and only less than 40% of VEGF-C tumor-bearing mice hadlung surface metastases (Fig. 4 D and E). Histological exami-nation further confirmed the presence of large metastatic nod-ules, whereas only small microscopic micrometastases werefound in the VEGF-C tumor-bearing mice (Fig. 4F). Both grossand histological examinations showed that mice with vectortumors completely lacked pulmonary metastases (Fig. 4 E andF). These findings provide compelling evidence demonstratingthat FGF-2 and VEGF-C collaboratively promote hematogenousmetastasis, most likely mediated by the high density of disorga-nized vasculatures in dual factor-expressing tumors.

- - - - - - - -

- - -

- - - - - - - -

- - -

FGF-2 VEGF-C VEGF-C+FGF-2 FGF-2 VEGF-C VEGF-C+FGF-2Non-treated VEGFR-3 blockade

P P PP PP

LYV

E-1

/CD

31LY

VE

-1C

D31

LYV

E-1

- - - - - - - -

- - - - - - - -

- - - - - - - -- - - - - - - -

A

B

C

- - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - -- - - -

-- --

- - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - -

- - - - - - - - - - - - - - - - - - - - - - - -P P

P P P P- - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - --- - -

-- -- - - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - --- - -

-- --- - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - --- - -

-- --- - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - -

- -- -

---- - - - - - - - -

- - - - - - - -

- - - -- - - -

- - - -- - - --- - -

-- --

- - - - - - -

- - - - - - -

- - - - - -

- - - - - - - - - - - - - -

- - - - -

- - - - -

- - - - - -

- - - - - - - - - - -

- - - - -

- - - - -

- - - - -- - - - - - -

- - - - - - - - - - - - - - -

- - - - -

- - - - -- - - - - - - - - -

- - - - - - - - - - - - - - -

- - -

- - - - -

- - -

- - - - - - - - - - - - - - - - - - - - -

NT Anti-VEGFR-3

0

6

2

4

Num

ber o

f Filo

podi

ape

r fie

ld

048

1216 - - - - - - - - - - - - - - - - - - - - -

NT Anti-VEGFR-3

FGF-

2VEGF-

C

FGF-

2+VE

GF-C

- - - - - - - - - - - - - - - - - - - - -

FGF-

2VEGF-

C

FGF-

2+VE

GF-C

0

4

Lym

phat

ic v

esse

l are

ape

r cor

nea

(mm

)2

2

6

D E F

Blo

od v

esse

l are

ape

r cor

nea

(mm

)2P < 0.05P < 0.01

P < 0.001

P < 0.001P < 0.001

P < 0.001

NSNS

NSNT Anti-VEGFR-3

P < 0.001P < 0.001

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

100 μm

400 μm 400 μm 400 μm 400 μm 400 μm 400 μmFG

F-2

VEGF-C

FGF-

2+VE

GF-C

FGF-

2VEGF-

C

FGF-

2+VE

GF-C

FGF-

2VEGF-

C

FGF-

2+VE

GF-C

FGF-

2VEGF-

C

FGF-

2+VE

GF-C

20 μm20 μm20 μm20 μm20 μm20 μm

Fig. 3. Anti-VEGFR-3 blocks FGF-2–induced lymphangiogenesis and lym-phatic tip formation. (A) FGF-2–, VEGF-C–, or FGF-2 plus VEGF-C–inducedcorneal neovascularization was treated with or without VEGFR-3 blockade.(B) FGF-2–, VEGF-C–, or FGF-2 plus VEGF-C–induced corneas in response toanti–VEGFR-3 treatment were double immunostained with CD31 (red) andLYVE-1 (green). VEGFR-3 blockade almost completely suppressed FGF-2–,VEGF-C–, or FGF-2 plus VEGF-C–induced corneal lymphangiogenesis but nothemangiogenesis. (C) Suppression of FGF-2–, VEGF-C–, or FGF-2 plus VEGF-C–induced tip formation of lymphatic vessels by VEGFR-3 blockade. (D)Quantification of LYVE-1+ lymphatic vessels in various groups treated withor without VEGFR-3 blockade (n = 4–6 per group). (E) Quantification oflymphatic tips in various groups treated with or without VEGFR-3 blockade(n = 4–6 per group). (F) Quantification of CD31+ blood vessels in variousgroups treated with or without VEGFR-3 blockade (n = 4–6 per group).

80

0204060

Vecto

rFG

F-2

VEGF-C

FGF-

2+VEGF-

C

% o

f mic

e w

ith v

isib

le p

ulm

onar

y m

ets

Tumor

free

Vector FGF-2 VEGF-CFGF-2+VEGF-CTumor free

1 cm

A

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - - - -

- - - - - -

CD

31

Vector FGF-2 VEGF-C FGF-2+VEGF-C

CD

31

- - - - - -

- - - - - -- - - -

- -

- - - -

- -

- -- -- - - - - - - -- - - - - - - -- - - - - - -- - - - - - -- -

- - -

- - - -

-----

- ---

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - -- - - - ----

- ---

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - -- - - - ----

- ---

- - - -- - - -

- - - -- - - -

- - - -- - - -

- - - -- - - - ----

- ---

50 μm 50 μm50 μm 50 μm

B

0

4

8

Vecto

rFG

F-2

VEGF-C

Are

a of

CD

31 p

ositi

ve

stai

ning

per

fiel

d (x

10

μm )

42

2

6

P < 0.001

P < 0.001P < 0.01

P < 0.001P < 0.01

C D

E FVector FGF-2 VEGF-C

FGF-2+VEGF-CTumor free

H&

EE

GFP

TT

TT

100 μm100 μm100 μm100 μm100 μm

100 μm100 μm100 μm100 μm100 μm

0

0.4

0.8

1.2

1.6

7 9 11Tum

or v

olum

e (c

m )3

Time (days)

FGF-2VEGF-CVEGF-C+FGF-2

Vector

P <

0.0

01P

< 0

.05

0

FGF-

2+VEGF-

C

Fig. 4. FGF-2 and VEGF-C collaboratively promote tumor angiogenesis andhematogenous metastasis. (A) Growth rates of vector, FGF-2–, VEGF-C–, andFGF-2 plus VEGF-C–expressing mouse fibrosarcomas. (B) Detection of tumormicrovessels in various tumor groups by CD31 immunostaining. (C) Quanti-fication of CD31+ vessel density in various tumor groups (n = 6 per group).(D) Lung morphology of a representative mouse from each tumor-bearinggroup (n = 6 per group). Arrowheads point to lung surface metastaticnodules. (E) Quantification of percentage of tumor-bearing mice that hadlung metastases (n = 6 per group). (F) Histological examination of pulmo-nary metastases in mice bearing various tumors. Dashed lines encircle met-astatic tumors that express EGFP (green). T, tumor.

Cao et al. PNAS | September 25, 2012 | vol. 109 | no. 39 | 15897

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Oct

ober

22,

202

0

Page 5: Collaborative interplay between FGF-2 and VEGF-C promotes ... · specific neutralizing antibody markedly inhibited FGF-2–induced lymphangiogenesis. Thus, the VEGFR-3–induced

Similar to the synergism of promoting hematogenous metas-tasis, examination of sentinel lymph nodes of tumor-bearingmice showed that 100% of VEGF-C and FGF-2 plus VEGF-Ctumor-bearing mice carried enlarged sentinel lymph nodes (Fig.5C). Notably, both weights and volumes of lymph nodes in FGF-2 plus VEGF-C tumor-bearing mice were significantly greaterthan those lymph nodes in VEGF-C tumor-bearing mice (Fig. 5C–F). In contrast to VEGF-C and FGF-2 plus VEGF-C tumor-bearing mice, only a small number (less than 40%) of FGF-2tumor-bearing mice suffered from enlargement of lymph nodes,which were marginally greater than those found in tumor-freemice (Fig. 5 C–F). Histology confirmed the presence of sentinellymph node metastases in FGF-2, VEGF-C and FGF-2 plusVEGF-C tumor-bearing mice (Fig. 5G). Collectively, these datademonstrate that FGF-2 and VEGF-C collaboratively stimulatelymph node metastasis.

DiscussionIn this study we provide several unique mechanistic insights onlymphangiogenesis regulated by FGF-2 and VEGF-C that stim-ulate discrete steps of lymphatic vessel growth (Fig. 5H). First,we show that FGF-2 directly acts on LECs to promote pro-liferation and migration via activation of the FGFR-1–mediatedsignaling pathway. These findings demonstrate that FGF-2 isa direct lymphangiogenic factor and have validated other pub-lished data (31). Second, we provide evidence that FGF-2 is ableto induce tip cell formation at the leading front of growinglymphatic vessels. Third, FGF-2–induced LEC tips are de-pendent on the VEGFR-3 signaling system. This finding is un-expected because FGF-2 can directly promote VEGFR-3–independent LEC proliferation and migration in vitro. The factthat VEGFR-3 blockade completely inhibited FGF-2–inducedlymphangiogenesis in vivo demonstrates the formation of LECtips is an essential process for lymphatic vessel growth. Thisprocess is tightly controlled by the VEGFR-3–mediated signalingsystem and may not be replaced by other lymphangiogenic sig-nals. If this is the case, the pivotal role of the VEGFR-3 systemmay reasonably be generalized to other lymphangiogenic factors.For example, hepatocyte growth factor (HGF)-induced lym-phangiogenesis is dependent on the VEGFR-3 signaling system(16), although the involvement of the LEC tips in HGF-stimu-lated lymphatic vessels has not been studied. The essential roleof VEGFR-3–induced LEC tips in FGF-2–induced lym-phangiogenesis has raised several important issues, including:How does VEGFR-3 become activated? Does FGF-2 induceVEGF-C/-D expression or transactivate the VEGFR-3 signalingin the absence of these ligands? It is also plausible that FGF-2induces VEGFR-3 expression in LECs, leading to more re-sponsive to VEGF-C/-D stimulation. These interesting questionswarrant further investigation. Finally, FGF-2 and VEGF-C col-laboratively stimulate lymphangiogenesis in vivo, probably byenhancing the VEGF-C–VEGFR-3–induced LEC tips and FGF-2–triggered proliferative signals.Unlike lymphangiogenesis, FGF-2–induced hemangiogenesis

is completely insensitive to VEGFR-3 blockade, indicating thatFGF-2 on its own is sufficiently potent to trigger angiogenicsignals. Although the formation of endothelial cell tips at theleading edge of angiogenic vessels is probably also essential forhemangiogenesis, this process does not seem to require activa-tion of VEGFR-3, which is not prominently expressed in bloodvessels. However, FGF-2 and VEGF-C also display marked

0

10

20

30

Vecto

rFG

F-2

VEGF-C

FGF-

2+VEGF-

C

Are

a of

LY

VE

-1 p

ositi

vest

aini

ng p

er fi

eld

(x 1

0 μm

)4

2

P < 0.001

P < 0.001

P < 0.001

P < 0.001

P < 0.001

0

50

100

% o

f mic

e w

ithvi

sibl

e LN

met

s

Vecto

rFG

F-2

VEGF-C

FGF-

2+VEGF-

C

Tumor

free

3

0.8

00.20.40.6

Vecto

rFG

F-2

VEGF-C

FGF-

2+VEGF-

C

Tumor

free L

N v

olum

e (c

m ) P < 0.001

P < 0.01

P < 0.001

P < 0.001

P < 0.001P < 0.01

0.9

00.30.6

Vecto

rFG

F-2

VEGF-C

FGF-

2+VEGF-

C

Tumor

free L

N w

eigh

t (g)

P < 0.001

P < 0.05

P < 0.001

P < 0.001

P < 0.001P < 0.001

Tumor freeVectorFGF-2

VEGF-C

FGF-2+VEGF-C 1 cm

A

B C

D E F

LYV

E-1

/EG

FPVector FGF-2 VEGF-C FGF-2+VEGF-C

LYV

E-1

- - -

PTL

ITL

PTL

ITL

PTL

ITLPTL

PTL

ITL

PTL

ITLPTL

PTL

ITL50 μm50 μm50 μm50 μm

G Vector FGF-2 VEGF-C FGF-2+VEGF-CTumor free

H&

EE

GFP

TT

TTT

100 μm100 μm100 μm100 μm100 μm

100 μm100 μm100 μm100 μm100 μm

H

Fig. 5. FGF-2 and VEGF-C collaboratively promotes tumor lymphangio-genesis and lymphatic metastasis. (A) Detection of peritumoral and intra-tumoral lymphatic vessels (PTL and ITL) in various tumor groups by LYVE-1immunostaining. Dashed line defines the rim between the tumor and peri-tumoral regions. Arrowheads indicate the tumoral lymphatics. (B) Quanti-fication of intratumoral LYVE-1+ lymphatic vessel density in various tumorgroups (n = 6 per group). (C) Sentinel lymph node morphology of varioustumor-bearing groups (n = 6 per group). (D) Quantification of percentage oftumor-bearing mice that had sentinel lymph-node metastases (n = 6 pergroup). (E) Average weight of sentinel lymph nodes in various tumor-bear-ing mice (n = 6 /group). (F) Average volume of sentinel lymph nodes invarious tumor-bearing mice (n = 6 per group). (G) Sentinel lymph nodehistology showed the presence of lymphatic metastases in FGF-2, VEGF-C,and FGF-2 plus VEGF-C tumor-mice. Dashed lines encircle metastatic tumorsthat express EGFP (green). T, tumor. (H) Schematic diagram of molecularmechanisms by which FGF-2 and VEGF-C collaboratively induce hem-/lym-phangiogenesis and metastasis. VEGF-C activates VEGFR-3 receptor on LECs,leading to LEC tip formation, proliferation, and migration. FGF-2 via acti-vation of FGFR-1 stimulates LEC proliferation and migration. However,VEGFR-3–triggered tip cell formation is a prerequisite for FGF-2–inducedlymphangiogenesis. Blockade of the VEGFR-3 signaling system completelyinhibits FGF-2–induced lymphangiogenesis. Lymhangiogenic interplay be-tween FGF-2 and VEGF-C in the tumor environment promoted lymphatic

metastasis. On blood vessel endothelial cells (BVECs), VEGF-C binds toVEGFR-2 and induced hem-angiogenic signals. FGF-2 directly induceshemangiogenesis by activation of FGFR-1 to -4 expressed on BVECs. The FGF-2 plus VEGF-C–induced high numbers of disorganized tumor blood vesselsfacilitate hematogenous metastasis.

15898 | www.pnas.org/cgi/doi/10.1073/pnas.1208324109 Cao et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

22,

202

0

Page 6: Collaborative interplay between FGF-2 and VEGF-C promotes ... · specific neutralizing antibody markedly inhibited FGF-2–induced lymphangiogenesis. Thus, the VEGFR-3–induced

additive activity in stimulation of hemangiogenesis. Because ofits binding to VEGFR-2 receptor (13), the angiogenic synergismof FGF-2 and VEGF-C is likely achieved by the intimate sig-naling interaction between FGFRs and VEGFR-2. Consistentwith this notion, previous studies demonstrated that FGF-2 andVEGF-A (another VEGFR-2 binding ligand) collaborativelypromote angiogenesis in vivo (32).Hem- and lymphangiogenic synergisms between FGF-2 and

VEGF-C in the tumor microenvironment lead to increasedbloodstream and lymphatic metastases. The dual factor-inducedangiogenic and lymphangiogenic vessels within tumors appearedto be highly disorganized and premature, a susceptible feature forfacilitating tumor-cell dissemination and intravasation. Further-more, peritumoral lymphatic vessels are highly dilated and maysubstantially contribute to lymphatic metastasis. Once metasta-sized to sentinel lymph nodes, the growth of metastases is furtherdependent on hemangiogenesis. In our tumor models, we showthat FGF-2 andVEGF-C not only facilitate themetastatic process,but also further stimulate metastatic growth to become visiblemetastatic masses. Thus, FGF-2 and VEGF-C accelerate metas-tasis by acting on several metastatic steps; that is, facilitating tumor

cell dissemination via intratumoral disorganized blood or lym-phatic vessels, propagating or co-opting surrounding blood orlymphatic vessels, and supporting further growth of metastaticmasses in distal organs, where tumor cells can be further spread toother tissues and organs. It is therefore critically important todevelop therapeutic agents that interfere with the interplay be-tween various tumor angiogenic factors.

Experimental ProceduresAll animal studies were approved by the Northern Stockholm ExperimentalAnimal Ethical Committee. Statistical analyses were performed using a stan-dard two-tail Student’s t test. Mean determinants were presented as ± SEM.Details are provided in SI Experimental Procedures.

ACKNOWLEDGMENTS. We thank Sharon Lim for the artistic work presentedin Fig. 5, and ImClone/EliLilly for providing FGFR-1– and VEGFR-3–specific neu-tralizing antibodies. Y.C.’s laboratory is supported by research grants from theSwedish Research Council, the Swedish Cancer Foundation, the Karolinska In-stitute Foundation, the Karolinska Institute distinguished professor award, theTorsten Soderbergs Foundation, ImClone Systems/EliLilly, European Union In-tegrated Project of Metoxia Project 222741, the Nordea Foundation, and theEuropean Research Council advanced Grant ANGIOFAT, Project 250021.

1. Fidler IJ, Ellis LM (1994) The implications of angiogenesis for the biology and therapyof cancer metastasis. Cell 79:185–188.

2. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: The next generation. Cell 144:646–674.

3. Rouhi P, et al. (2010) Hypoxia-induced metastasis model in embryonic zebrafish. NatProtoc 5:1911–1918.

4. Lee SL, et al. (2009) Hypoxia-induced pathological angiogenesis mediates tumor celldissemination, invasion, and metastasis in a zebrafish tumor model. Proc Natl Acad SciUSA 106:19485–19490.

5. Cao Y (2005) Opinion: Emerging mechanisms of tumour lymphangiogenesis andlymphatic metastasis. Nat Rev Cancer 5:735–743.

6. Alitalo K, Carmeliet P (2002) Molecular mechanisms of lymphangiogenesis in healthand disease. Cancer Cell 1:219–227.

7. Stacker SA, Achen MG, Jussila L, Baldwin ME, Alitalo K (2002) Lymphangiogenesis andcancer metastasis. Nat Rev Cancer 2:573–583.

8. Kedrin D, et al. (2008) Intravital imaging of metastatic behavior through a mammaryimaging window. Nat Methods 5:1019–1021.

9. Zijlstra A, Lewis J, Degryse B, Stuhlmann H, Quigley JP (2008) The inhibition of tumorcell intravasation and subsequent metastasis via regulation of in vivo tumor cellmotility by the tetraspanin CD151. Cancer Cell 13:221–234.

10. Kienast Y, et al. (2010) Real-time imaging reveals the single steps of brain metastasisformation. Nat Med 16:116–122.

11. Kaplan RN, et al. (2005) VEGFR1-positive haematopoietic bone marrow progenitorsinitiate the pre-metastatic niche. Nature 438:820–827.

12. Cao Y, et al. (1998) Expression of angiostatin cDNA in a murine fibrosarcoma sup-presses primary tumor growth and produces long-term dormancy of metastases. J ClinInvest 101:1055–1063.

13. Cao Y, et al. (1998) Vascular endothelial growth factor C induces angiogenesis in vivo.Proc Natl Acad Sci USA 95:14389–14394.

14. Cao Y (2008) Why and how do tumors stimulate lymphangiogenesis? Lymphat ResBiol 6:145–148.

15. Cao Y, Zhong W (2007) Tumor-derived lymphangiogenic factors and lymphatic me-tastasis. Biomed Pharmacother 61:534–539.

16. Cao R, et al. (2006) Hepatocyte growth factor is a lymphangiogenic factor with anindirect mechanism of action. Blood 107:3531–3536.

17. Björndahl M, et al. (2005) Insulin-like growth factors 1 and 2 induce lymphangio-genesis in vivo. Proc Natl Acad Sci USA 102:15593–15598.

18. Cao Y (2005) Direct role of PDGF-BB in lymphangiogenesis and lymphatic metastasis.Cell Cycle 4:228–230.

19. Morisada T, et al. (2005) Angiopoietin-1 promotes LYVE-1-positive lymphatic vesselformation. Blood 105:4649–4656.

20. Veikkola T, Alitalo K (2002) Dual role of Ang2 in postnatal angiogenesis and lym-phangiogenesis. Dev Cell 3:302–304.

21. Xue Y, et al. (2011) PDGF-BB modulates hematopoiesis and tumor angiogenesis byinducing erythropoietin production in stromal cells. Nat Med 18:100–110.

22. Nguyen M, Watanabe H, Budson AE, Richie JP, Folkman J (1993) Elevated levels of theangiogenic peptide basic fibroblast growth factor in urine of bladder cancer patients.J Natl Cancer Inst 85:241–242.

23. Van der Auwera I, et al. (2006) First international consensus on the methodology oflymphangiogenesis quantification in solid human tumours. Br J Cancer 95:1611–1625.

24. Cao R, et al. (2011) Mouse corneal lymphangiogenesis model. Nat Protoc 6:817–826.25. Kubo H, et al. (2002) Blockade of vascular endothelial growth factor receptor-3 sig-

naling inhibits fibroblast growth factor-2-induced lymphangiogenesis in mouse cor-nea. Proc Natl Acad Sci USA 99:8868–8873.

26. Pytowski B, et al. (2005) Complete and specific inhibition of adult lymphatic re-generation by a novel VEGFR-3 neutralizing antibody. J Natl Cancer Inst 97:14–21.

27. Tammela T, et al. (2011) VEGFR-3 controls tip to stalk conversion at vessel fusion sitesby reinforcing Notch signalling. Nat Cell Biol 13:1202–1213.

28. Tammela T, et al. (2008) Blocking VEGFR-3 suppresses angiogenic sprouting andvascular network formation. Nature 454:656–660.

29. Hellström M, et al. (2001) Lack of pericytes leads to endothelial hyperplasia and ab-normal vascular morphogenesis. J Cell Biol 153:543–553.

30. Nissen LJ, et al. (2007) Angiogenic factors FGF2 and PDGF-BB synergistically promotemurine tumor neovascularization and metastasis. J Clin Invest 117:2766–2777.

31. Shin JW, et al. (2006) Prox1 promotes lineage-specific expression of fibroblastgrowth factor (FGF) receptor-3 in lymphatic endothelium: A role for FGF signaling inlymphangiogenesis. Mol Biol Cell 17(2):576–584.

32. Nico B, de Falco G, Vacca A, Roncali L, Ribatti D (2001) In vivo absence of synergismbetween fibroblast growth factor-2 and vascular endothelial growth factor. J Hem-atother Stem Cell Res 10:905–912.

Cao et al. PNAS | September 25, 2012 | vol. 109 | no. 39 | 15899

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Oct

ober

22,

202

0