ArticlePDF Available

Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice

Authors:

Abstract and Figures

Cancer development, progression, and metastasis are highly dependent on angiogenesis. The use of antiangiogenic drugs has been proposed as a novel strategy to interfere with tumor growth, but cancer cells respond by developing strategies to escape these treatments. In particular, animal models show that antiangiogenic drugs currently used in clinical settings reduce tumor tissue oxygenation and trigger molecular events that foster cancer resistance to therapy. Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in HPV16/E2 mice. By improving cancer tissue oxygenation and extending the normalization window, Sema3A counteracted sunitinib-induced activation of HIF-1α, Met tyrosine kinase receptor, epithelial-mesenchymal transition (EMT), and other hypoxia-dependent signaling pathways. Sema3A also reduced tumor hypoxia and halted cancer dissemination induced by DC101, a specific inhibitor of the VEGF pathway. As a result, reexpressing Sema3A in cancer cells converts metastatic PNETs and cervical carcinomas into benign lesions. We therefore suggest that this strategy could be developed to safely harnesses the therapeutic potential of the antiangiogenic treatment.
Content may be subject to copyright.
Research article
The Journal of Clinical Investigation http://www.jci.org
Semaphorin 3A overcomes cancer hypoxia
and metastatic dissemination induced
by antiangiogenic treatment in mice
Federica Maione,1,2,3 Stefania Capano,1,2,3 Donatella Regano,1,2,3 Lorena Zentilin,4 Mauro Giacca,4
Oriol Casanovas,5 Federico Bussolino,2,3,6,7 Guido Serini,2,3,7,8 and Enrico Giraudo1,2,3,7
1Laboratory of Transgenic Mouse Models, 2IRCC, and 3Department of Oncological Sciences, University of Torino School of Medicine, Candiolo, Italy.
4International Centre for Genetic Engineering and Biotechnology (ICGEB), Molecular Medicine Laboratory, Trieste, Italy. 5Tumor Angiogenesis Group,
Translational Research Laboratory, Catalan Institute of Oncology — IDIBELL, L’Hospitalet de Llobregat, Spain. 6Laboratory of Vascular Oncology,
University of Torino School of Medicine, Candiolo, Italy. 7Center for Complex Systems in Molecular Biology and Medicine (SysBioM), University of Torino,
Torino, Italy. 8Laboratory of Cell Adhesion Dynamics, University of Torino School of Medicine, Candiolo, Italy.
Cancer development, progression, and metastasis are highly dependent on angiogenesis. The use of antiangio-
genic drugs has been proposed as a novel strategy to interfere with tumor growth, but cancer cells respond by
developing strategies to escape these treatments. In particular, animal models show that antiangiogenic drugs
currently used in clinical settings reduce tumor tissue oxygenation and trigger molecular events that foster can-
cer resistance to therapy. Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive
and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor
sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in
HPV16/E2 mice. By improving cancer tissue oxygenation and extending the normalization window, Sema3A
counteracted sunitinib-induced activation of HIF-1α, Met tyrosine kinase receptor, epithelial-mesenchymal
transition (EMT), and other hypoxia-dependent signaling pathways. Sema3A also reduced tumor hypoxia
and halted cancer dissemination induced by DC101, a specific inhibitor of the VEGF pathway. As a result,
reexpressing Sema3A in cancer cells converts metastatic PNETs and cervical carcinomas into benign lesions.
We therefore suggest that this strategy could be developed to safely harnesses the therapeutic potential of the
antiangiogenic treatment.
Introduction
Angiogenesis is required for  invasive tumor growth and meta-
static dissemination, providing the rationale for the development 
of antiangiogenic therapies (1). Despite the generation of innova-
tive antiangiogenic strategies, such as inhibitors of the VEGF-A 
pathway, resistance  to  anti-VEGF  therapy has  been  recently 
observed in both preclinical and clinical trials (2, 3). For instance, 
preclinical studies provided evidence for anti-VEGF drug evasion 
by activation of alternate proangiogenic pathways, likely induced 
by a significant increase of tumor tissue hypoxia (4). Therefore, to 
extend the optimal therapeutic windows and design more effective 
antiangiogenic combinatory regimens that could prevent or block 
tumor invasion and metastasis formation, it is critical to identify 
new angiogenic modulators and uncover their molecular and cel-
lular mechanisms of action in vivo.
It is well documented that, as a result of architectural and bio-
logical abnormalities such as  tortuosity, leakiness, and lack of 
pericytes, tumor blood vessels are structurally and functionally 
aberrant (5), causing cancer tissue hypoxia (6). Notably, abnormal 
vascular permeability and chronic oxygen shortage promote tumor 
invasiveness, for example, by upregulating HIF-1α expression (3, 7), 
downregulating  E-cadherin expression (8), and hyperactivating 
hepatocyte growth factor/Met (HGF/Met) signaling (9). Further-
more, several independent preclinical studies (10, 11), which have 
not yet been paralleled by analogous clinical trials, revealed that 
although impairing cancer angiogenesis with different therapeu-
tic approaches initially causes remarkable shrinkage of the tumor 
mass, this approach eventually causes dramatic enhancement of 
tumor invasiveness and increased  distal metastasis formation. 
These data, together with the formal demonstration that improv-
ing oxygenation can suppress metastatization of cancer cells and 
promote their differentiation (12), further support the hypoth-
esis  that  vascularnormalization  could  represent  a  remarkably 
advantageous anticancer strategy, as it is also able to favor chemo-
therapy delivery and response to radiotherapy (6). We previously 
showed that endothelial semaphorin 3A (Sema3A) is an endog-
enous antiangiogenic agent that, when reexpressed in cancers that 
lost it, is able to normalize the vasculature and to block tumor 
growth, finally inducing a stable disease (13). In the present study, 
we investigated the molecular and cellular mechanisms by which 
Sema3A, alone or in combination with different antiangiogenic 
drugs, is able to impair tumor cell dissemination and overcome 
evasive resistance to angiogenesis inhibition (14).
Results
Sema3A halts tumor invasion and metastasis formation caused by antian-
giogenic treatment. We previously demonstrated that reexpressing 
Sema3A in tumors of a spontaneous mouse model of pancreatic 
neuroendocrine cancer (RIP-Tag2) by somatic gene transfer using 
adeno-associated virus–8 (AAV8) resulted in reduced vascular den-
sity, inhibition of tumor growth, significant survival extension, 
normalization of tumor vasculature, and decreased tumor hypoxia 
(13). Stemming from these data, we sought to investigate whether 
Conflict of interest: The authors have declared that no conflict of interest exists.
Citation for this article:J Clin Invest doi:10.1172/JCI58976.
research article
The Journal of Clinical Investigation http://www.jci.org
Sema3A also impairs tumor invasion and metastasis formation 
to overcome the evasive resistance observed in RIP-Tag2 mice and 
other mouse models in response to antiangiogenic therapies (10, 
11). We first compared the effect of AAV8-Sema3A (referred to here-
in as Sema3A) and sunitinib, a prototypical small-molecule tyro-
sine kinase (TK) inhibitor and antiangiogenic drug (15), on tumor 
dissemination in RIP-Tag2 mice by performing a 4-week regression 
trial between 12 and 16 weeks of age. Treatment of tumor-bearing 
RIP-Tag2 mice with sunitinib induced primary tumor shrinkage 
and strongly inhibited angiogenesis (see below), but at the same 
time promoted local invasiveness and distant metastasis formation 
compared with controls (Figure 1, Figure 2, A–G, and Supplemental 
Figure 1; supplemental material available online with this article; 
doi:10.1172/JCI58976DS1), consistent with previous findings (10). 
On the contrary, treatment with Sema3A alone not only reduced 
tumor burden and vascularization (ref. 13 and see below), but also 
dramatically decreased cancer invasion in the surrounding tissues 
as well as the incidence and volume of peripancreatic LN metas-
tases and the incidence, number, and volume of liver metastases 
compared with controls (Figure 1 and Figure 2, A–G). Hence, we 
showed Sema3A to be an angiogenesis inhibitor that, differently 
from sunitinib, also displayed a powerful antimetastatic activity. 
The recent observation that systemic delivery of Sema3A inhibits 
angiogenesis and metastatization in xenograft tumor models as 
well (16) further supports the role of Sema3A as an effective phar-
macological inhibitor of cancer progression. To start investigating 
whether Sema3A antagonizes the previously described proinvasive 
effect of sunitinib in RIP-Tag2 mice (10), we set up a combinatory 
therapeutic regimen, treating RIP-Tag2 mice simultaneously with 
Sema3A and sunitinib for 1 month, after which we assessed the 
frequency of invasive lesions and metastasis formation. Notably, 
the combination of Sema3A with sunitinib strongly reduced the 
incidence of fully invasive (IC2) tumors and the extent of both LN 
and liver metastases in sunitinib-treated animals (Figure 1, B and C, 
and Figure  2,  A–G). Together, these  data  demonstrated  that 
Sema3A not only impaired metastasis formation during spontane-
ous tumorigenesis, but also curbed the increased cancer aggressive-
ness stimulated by sunitinib treatment.
Sema3A and sunitinib synergize to enhance survival. Based on our 
observation that the combination of Sema3A with sunitinib effec-
tively hampered the evasive resistance elicited by sunitinib treat-
ment alone in RIP-Tag2 mice, we next investigated whether these 
2 drugs could synergistically impair tumor progression and hence 
extend RIP-Tag2 survival as well. We performed a longer survival 
trial in which RIP-Tag2 tumor-bearing mice were treated beginning 
at 12 weeks of age with AAV8-LacZ (referred to herein as LacZ) 
plus vehicle (control), Sema3A, sunitinib, or combined Sema3A 
and sunitinib. The median survival of control mice was 2.5 weeks. 
Similar to our previous observations (13), Sema3A significantly 
prolonged the survival of RIP-Tag2 mice by 9.0 weeks compared 
with control-treated animals (P < 0.001), 2.3 weeks longer than 
that observed with sunitinib treatment alone (P < 0.01). Treatment 
Figure 
Sema3A blocks tumor invasion caused by antiangiogenic treatment. (A) Total tumor volume in 4-week treatment regression trial (from 12
to 16 weeks of age) showed that sunitinib (SUN), AAV8-Sema3A in combination with sunitinib, and AAV8-Sema3A reduced tumor burden
64%, 63%, and 66%, respectively, compared with controls (AAV8-LacZ–injected RIP-Tag2 treated with vehicle; see Methods). (B) Percent
encapsulated (IT), microinvasive (IC1), and fully invasive (IC2) carcinomas. Combined AAV8-Sema3A and sunitinib decreased IC2 carcinoma
incidence 62% compared with sunitinib alone. Sema3A-treated tumors showed a statistically significant 64% decrease of IC2 carcinomas
compared with controls. (C) Analysis of tumor invasiveness by means of SV40 T-antigen immunostaining. Sunitinib-treated tumors displayed
an invasive front extensively intercalated into the surrounding tissue (arrows). T, tumor; Ac, acinar tissue. *P < 0.05, **P < 0.01, unpaired
Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
Figure 
Sema3A blocks distal metastasis formation caused by antiangiogenic treatment. (A and B) SV40 T-antigen immunostaining (arrows) of peri-
pancreatic LN and liver metastases (Met). Representative images were derived from serial section analysis in each animal (n = 30 per treat-
ment group). (C and D) Metastasis incidence per animal. Combined Sema3A and sunitinib decreased LN and liver metastases 57% and 54%,
respectively, compared with sunitinib alone. Sema3A reduced LN and liver metastases 62% and 50%, respectively, compared with controls.
(E) LN metastasis volume. Combined Sema3A and sunitinib decreased LN volume 89% compared with sunitinib alone; Sema3A decreased
LN volume 79% compared with controls. (F and G) Number (F) and volume (G) of liver metastases (n = 30 per treatment group). Combined
Sema3A and sunitinib diminished the number and volume of liver metastases 88% and 91%, respectively, compared with sunitinib alone;
Sema3A reduced liver metastasis number and volume 80% and 85%, respectively, compared with controls. (H) Survival trial in tumor-bearing
RIP-Tag2 mice treated continuously with 40 mg/kg/d sunitinib, Sema3A, combined Sema3A and sunitinib, or LacZ beginning at 12 weeks
(n = 20 per group). *P < 0.05, **P < 0.01, unpaired Mann-Whitney U test or log-rank test (for survival trials). Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
with sunitinib increased survival 6.7 weeks compared with con-
trols (P < 0.001) (Figure 2H), as previously shown (10). Interest-
ingly, this survival trial clearly demonstrated that the combination 
of Sema3A with sunitinib significantly enhanced the survival of 
RIP-Tag2 mice by 16.2, 7.2, and 9.5 weeks compared with control, 
Sema3A, and sunitinib, respectively (P< 0.001 for all comparisons; 
Figure 2H), suggestive of effective synergism of Sema3A and suni-
tinib regarding survival and tumor progression. Of note, 18 weeks 
after the initial treatment with combined Sema3A and sunitinib, 
6 of 20 mice  of  the survival trial were still alive,  and  2  of  these 
were tumor free. Interestingly, similarly to what we observed in the 
4-week regression trial, this combinatorial treatment resulted in very 
small and round tumors (Supplemental Figure 4A) and strongly 
halted tumor invasiveness in the RIP-Tag2 mice that survived until 
the end of the trial. Importantly, none of the 6 surviving mice had 
liver or peripancreatic LN metastases. Together, these data indicate 
that the combination of Sema3A with sunitinib results in a syner-
gistic effect by prolonging animal survival and inducing smaller, 
less invasive, and less frequent metastatic cancers.
Sema3A counteracts basal and sunitinib-elicited tumor hypoxia. Both 
primary tumors and metastases of mice treated with antiangiogen-
ic drugs are highly hypoxic (10), and preclinical studies suggest that 
evasion to antiangiogenic therapies could depend on the hypoxia-
driven induction of alternative proangiogenic pathways in tumor 
cells (4, 14). Based on the vascular normalizing effect of Sema3A we 
previously observed in RIP-Tag2 tumors (13), we hypothesized that 
this molecule could overcome the evasive resistance to angiogen-
esis inhibition by hampering tumor hypo-oxygenation. We there-
fore measured tissue hypoxia in RIP-Tag2 insulinomas treated with 
sunitinib, Sema3A, or both in combination. The strong reduction 
of vessel area induced by sunitinib was accompanied by an increase 
in intratumoral  hypoxia, as assessed  by pimonidazole staining 
(Figure 3, A and B). As previously shown, treating RIP-Tag2 mice 
with Sema3A for 1 month proportionally restrained the amount of 
blood vessels and normalized the remaining vasculature, abrogat-
ing the tumor hypoxia observed in control mice at both the begin-
ning and the end of the therapeutic trial (ref. 13 and Supplemental 
Figure 2). Remarkably, combinatory treatment with Sema3A com-
pletely reversed the sizeable hypoxia observed in sunitinib-treated 
RIP-Tag2 insulinomas (Figure 3, A and B).
To further characterize the extent of tumor hypoxia associated 
with the different therapeutic regimens, we assessed the expres-
sion of HIF-1α, a master regulator of cellular adaptation to oxygen 
deprivation that acts as a survival factor for hypoxic cancer cells, 
being expressed in many human cancers and associated with poor 
prognosis and treatment failure (3, 7). Remarkably, Western blot 
analysis revealed a strong increase of HIF-1α protein in sunitinib-
treated tumors that was dramatically reduced by simultaneous 
treatment with Sema3A  (Figure 3, C and D). Of note, adminis-
tering sunitinib, alone or in combination with Sema3A, resulted 
Figure 
Sema3A impairs basal and sunitinib-elicited tumor hypoxia and HIF-1α expression. (A) Tumor hypoxia was assessed by means of pimoni-
dazole adduct immunostaining (arrows) in serial sections of tumors of RIP-Tag2 mice treated for 4 weeks with sunitinib, Sema3A, and the
combination compared with control. Representative images were derived from serial section analysis in each animal (n = 15 per treatment
group). (B) Quantification of hypoxic tumors, expressed as percent of pimonidazole density area per tumor, per animal. Combined Sema3A
and sunitinib reduced hypoxic area 84% compared with sunitinib alone; Sema3A decreased hypoxic area 90% compared with controls.
(C) Western blot analysis of HIF-1α, CA9, and vinculin (as protein loading control) in tumors isolated from RIP-Tag2 mice treated as indicated.
(D) Relative levels of HIF-1α and CA9 protein expression, normalized to vinculin (n = 12 per treatment group). Combined Sema3A and sunitinib
diminished HIF-1α expression and CA9 amount 91% and 60%, respectively, compared with sunitinib alone; Sema3A decreased CA9 expres-
sion 55% compared with control. *P < 0.05, **P < 0.01, unpaired Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
Figure 
Sema3A alone or in combination with sunitinib enhances pericyte coverage, reduces blood vessel leakage, improves tissue perfusion, and
increases doxorubicin delivery to tumors. (A and B) As assessed by confocal analysis of Meca32 immunostaining, vessel density in RIP-Tag2
tumors after a 4-week regression trial was reduced 89% in sunitinib-treated tumors, 60% in Sema3A and sunitinib–treated tumors, and 47%
Sema3A-treated tumors compared with controls. (A and CF) Pericyte coverage, as evaluated by confocal colocalization analysis of Meca32
with NG2 (A, arrows, and C) or with α-SMA (D), PDGFR-β (E), or desmin (F). Combined Sema3A and sunitinib enhanced pericyte coverage
62% (NG2), 72% (α-SMA), 84% (PDGFR-β), and 69% (desmin) compared with sunitinib alone; Sema3A increased pericyte coverage 45%
(NG2), 47% (α-SMA), 40% (PDGFR-β), and 43% (desmin) compared with controls. (G) In tumors treated with Sema3A alone or in combination
with sunitinib, FITC-lectin perfused vessels (arrows) were increased compared with sunitinib-treated and control insulinomas. (H) FITC-dextran
extravasation (arrows) decreased in tumors treated with Sema3A alone or in combination with sunitinib compared with sunitinib-treated and con-
trol insulinomas. Results are from 5 fields per mouse (n = 12 per treatment group). (I) Amount of doxorubicin (DXR) present in tumors, expressed
as μg equivalent/g tumor. Combined Sema3A and sunitinib enhanced doxorubicin 87% compared with sunitinib alone; Sema3A enhanced
doxorubicin 83% compared with controls. *P < 0.05, **P < 0.01, unpaired Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
in similar modulation of the HIF-1α target gene carbonic anhy-
drase 9 (CA9) (Figure 3, C and D), which is also upregulated in 
several human cancers (3, 17). Moreover, in agreement with the 
normoxic tumor environment induced by Sema3A (ref. 13 and 
Supplemental Figure 2), we also observed a significant reduction 
of CA9 in animals treated with Sema3A alone compared with con-
trols (Figure 3, C and D). Thus, by virtue of its ability to normalize 
tumor blood vessels and to reestablish tissue normoxia, Sema3A 
efficiently overcame the invasive phenotype elicited by sunitinib 
in RIP-Tag2 mice.
The combination of Sema3A and sunitinib increases pericyte coverage,
reduces blood vessel leakage, enhances tumor tissue perfusion, and pro-
longs the vascular normalization window. Increased pericyte coverage 
and reduction in vascular density and branching are hallmarks 
of tumor  blood vessel normalization, a  process  that occurs in 
response to some antiangiogenic agents and allows for more effi-
cient delivery of oxygen and chemotherapeutic drugs (6, 18, 19). As 
expected (10, 15, 20), in sunitinib-treated tumors, in addition to a 
strong reduction of blood vessel area, we observed remarkable inhi-
bition of pericyte coverage, as revealed by confocal analysis of NG2 
staining (Figure 4, A–C). On the contrary, as previously described 
(13, 20), Sema3A treatment increased the number of perivascular 
NG2+ cells. Of note, simultaneous treatment with Sema3A and 
sunitinib significantly increased pericyte coverage (Figure 4, A 
and C). Similar observations with respect to pericyte coverage in 
the different treatment groups were obtained using other markers, 
such as α-SMA, PDGFR-β, and desmin (Figure 4, D–F). To better 
characterize Sema3A-elicited tumor blood vessel normalization, 
we further studied the perfusion and permeability of the tumor 
vasculature of RIP-Tag2 mice undergoing different therapeutic 
Figure 
Sema3A reduces both basal and sunitinib-induced EMT and inhibits NF-κB expression. (AD) RIP-Tag2 tumors were treated for 4 weeks
with sunitinib, AVV8-Sema3A, and the combination; shown are (A and C) representative confocal microscopy and (B and D) protein expres-
sion, quantified by MFI per animal. (A and B) Low E-cadherin expression levels in controls and sunitinib-treated insulinomas were significantly
upregulated by Sema3A treatment alone or in combination with sunitinib. (C and D) Increased vimentin expression in sunitinib-treated tumors
compared with controls was inhibited by treatment with Sema3A alone or in combination with sunitinib. (E) Western blot analysis of protein
lysates derived from tumors of the different treatment groups. Sema3A inhibited sunitinib-induced Snail1 and vimentin expression. E-cadherin
was highly expressed with Sema3A treatment, alone or in combination with sunitinib, compared with the control and sunitinib alone. Sema3A
decreased both basal and sunitinib-induced NF-κB expression. (F) Relative protein expression levels of EMT markers and NF-κB expression,
normalized to vinculin (n = 6 per treatment group). *P < 0.05, **P < 0.01, unpaired Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
regimens. At the end of each scheme of treat-
ment, blood vessel perfusion and permeabil-
ity were respectively assessed by tail-injecting 
animals with FITC-lectin and 70-kDa FITC-
dextran. Interestingly, Sema3A (both alone 
and in combination with sunitinib) strongly 
increased the amount of FITC-lectin in the 
vasculature of insulinomas (Figure 4G and 
Supplemental Figure  3,  A and C), demon-
strating a clear improvement of tumor blood 
vessel perfusion. Moreover, the therapy with 
Sema3A significantly reduced both basal and 
sunitinib-elicited leakiness of tumor blood 
vessels, as measured by confocal analysis of 
70-kDa FITC-dextran extravasation (Figure 
4H and Supplemental Figure 3, B and D).
To  ass ess  wh ether Sema3 A,  al one  o r  in 
combination with  sunitinib,  increases  the 
efficacy of tumor blood vessels in delivering a 
chemotherapeutic drug, we injected doxoru-
bicin in RIP-Tag2 mice that were previously 
treated for 4 weeks with  LacZ plus vehicle 
(control) or with Sema3A, sunitinib, or both 
in combination, then assessed the amount 
of drug present within tumor tissues. We 
detected decreased levels of doxorubicin in 
insulinomas of sunitinib-treated mice com-
pared with controls (Figure 4I). In contrast, 
Figure 
Sema3A inhibits basal and sunitinib-induced
expression and activation of Met TK receptor.
(A) Protein analysis showed greater total Met
and phospho-Met in RIP-Tag2 tumors treated
with sunitinib compared with controls; notably,
both were reduced by Sema3A, either alone or
in combination with sunitinib. Vinculin protein
was used as loading control. (B) Protein lev-
els of phospho-Met, normalized to total Met.
(CF) Presence and localization of Met and
phospho-Met in RIP-Tag2 tumors treated with
sunitinib, Sema3A, and the combination com-
pared with controls, as evaluated by colocaliza-
tion of anti-Met or anti–phospho-Met Abs with
Meca32. Shown are (C and E) representative
confocal microscopy, representative of 5 fields
per mouse, and (D and F) quantification by MFI
(n = 8 per treatment group). (C and D) Total
Met was present in a subset of tumor blood
vessels and in cancer cells in basal conditions
and was highly expressed in both vessels and
tumor cells after sunitinib treatment (arrows);
combined Sema3A and sunitinib reduced Met
both in vessels and tumor cells. (E and F) Phos-
pho-Met was present in some tumor vessels
(arrows) and in cancer cells in basal conditions,
but increased mainly in tumor cells in sunitinib-
treated animals. Notably, Sema3A alone or
combined with sunitinib inhibited both total and
phospho-Met. *P < 0.05, **P < 0.01, unpaired
Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
greater amounts of doxorubicin were present in Sema3A-treated 
tumors (alone or in combination with sunitinib) compared with 
control and sunitinib-treated insulinomas (Figure 4I). Based on 
these findings, we next investigated whether Sema3A is capable of 
increasing and sustaining the ability of the cancer vasculature to 
deliver a drug in tumors over a longer period of time, in the hopes 
of verifying the presence of an extended normalization window. 
We evaluated the degree of pericyte coverage, tissue hypoxia, and 
doxorubicin delivery efficiency to tumors of RIP-Tag2 mice treated 
with combined Sema3A and sunitinib and found to be alive at the 
end of the survival trial. Remarkably, similarly to what we observed 
in the 4-week regression trial (Figure 4, A–I), the insulinomas of 
the surviving mice treated with combined Sema3A and sunitinib 
were not hypoxic and displayed a functional, nonleaky vasculature, 
covered by pericytes (Supplemental Figure 4, B–E), that was able 
to efficiently deliver doxorubicin to tumors (24.6 ± 2.3 μg equiva-
lents/g tumor). Taken together, these data suggest that Sema3A, 
alone or in combination with sunitinib, substantially extends the 
normalization window of tumor blood vessel and improves the 
delivery efficiency of chemotherapeutic drugs to cancers.
Sema3A impairs sunitinib-induced epithelial-mesenchymal transition
and NF-κB expression. To better investigate the molecular mecha-
nisms by which Sema3A effectively counteracts the evasive resis-
tance induced by angiogenesis inhibition in RIP-Tag2 mice, we 
first evaluated the expression of proteins that are involved in epi-
thelial-mesenchymal transition (EMT) and support cancer cell 
Figure 
Sema3A halts sunitinib-elicited tumor invasion and metastasis formation in HPV16/E2 mice. (A) Total tumor volume of HPV16/E2 mice in a
4-week regression trial (from 5 to 6 months of age) revealed that sunitinib, Sema3A combined with sunitinib, and Sema3A alone reduced tumor
burden 58%, 76%, and 53%, respectively, compared with LacZ-injected, vehicle-treated HPV16/E2 controls. (B and C) Liver and lung metas-
tases (arrows), as assessed by oncogene E7 immunostaining. (D and E) Combined Sema3A and sunitinib decreased lung (D) and liver (E)
metastasis incidence 67% and 70%, respectively, compared with sunitinib; Sema3A reduced lung and liver metastasis incidence 84% and 74%,
respectively, compared with controls. (F and G) Combined Sema3A and sunitinib diminished the number (F) and volume (G) of liver metastases
82% and 94%, respectively, compared with sunitinib; Sema3A reduced liver metastasis number and volume 60% and 92%, respectively, com-
pared with controls. Representative images were derived from serial section analysis of each animal ( n = 12 per treatment group). *P < 0.05,
**P < 0.01, unpaired Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org
research article
0 The Journal of Clinical Investigation http://www.jci.org
metastatic dissemination (21). In EMT, tumor cells typically lose 
the epithelial marker E-cadherin and gain mesenchymal markers, 
such as vimentin and N-cadherin (21, 22). In addition, E-cadherin 
transcriptional repressors, such as Snail1, are also upregulated 
during EMT (23). Sunitinib-treated tumors demonstrated high 
expression of Snail1 and the mesenchymal markers vimentin and, 
to a lesser extent, N-cadherin; in contrast, the Snail1 target E-cad-
herin was strongly inhibited (Figure 5). Hence, sunitinib treatment 
promoted invasiveness by activating an EMT program. Remark-
ably, addition of Sema3A completely reverted the effects of suni-
tinib, dramatically inhibiting Snail1 and vimentin and enhancing 
E-cadherin expression (Figure 5). In addition, treating animals with 
Sema3A alone similarly inhibited the synthesis of mesenchymal 
markers and promoted E-cadherin expression as well.
NF-κB is involved in both physiological and pathological pro-
cesses and plays pivotal roles in promoting the EMT-dependent 
invasive phenotype  of  several cancers (24, 25). NF-κB induces 
HIF-1α(26), is activated by hypoxia (27), and is a critical component 
of the molecular machinery that senses low oxygen levels (28). In 
agreement with the above data, we observed that NF-κB protein lev-
els were high in tumors treated with sunitinib and that cotreatment 
with Sema3A returned NF-κB expression levels to those observed 
with control or Sema3A treatment alone (Figure 5, E and F).
Sema3A inhibits both basal and sunitinib-induced expression and activa-
tion of the Met TK receptor. Based on the known inductive effects of 
hypoxia on the expression and activation of the proinvasive TK 
receptor Met (9), we assessed total protein and tyrosine phosphor-
ylation levels of Met in treated RIP-Tag2 mice. Western blot analy-
sis revealed that sunitinib treatment caused a significant increase 
of both total Met and phospho-Met in tumors (Figure 6, A and B). 
However, whereas total Met immunoreactivity was observed in 
both blood  vessels  and tumor cells, phospho-Met was mainly 
detected in cancer cells (Figure 6, C–F). Interestingly, concomitant 
Sema3A administration fully inhibited the induction of both total 
Met and phospho-Met observed with sunitinib treatment alone. 
Tumors receiving Sema3A alone displayed a similar reduction of 
Met activation (Figure 6, A–C and E). The clear inhibition of Met 
TK receptor phosphorylation we observed identified a potential 
mechanism through which Sema3A might inhibit metastatization, 
namely the inhibition of Met receptor signaling in tumor cells as 
result of the reduced tumor hypoxia induced by Sema3A itself.
Sema3A overcomes metastasis formation caused by sunitinib treatment in
a mouse model of spontaneous cervical cancer. To evaluate whether the 
effects of Sema3A on tumor progression during angiogenesis inhi-
bition in RIP-Tag2 mice are recapitulated in another tumor model 
and histotype, we used the 17β-estradiol–treated K14-HPV16 trans-
genic mouse model of spontaneous cervical carcinogenesis (referred 
to herein as HPV16/E2 mice). In our previous work (13), we iden-
tified a similar expression pattern of Sema3A in both HPV16/E2
and RIP-Tag2 transgenic mouse models; in fact, Sema3A was sig-
nificantly expressed in normal and angiogenic premalignant stages, 
but was lost in invasive cervical carcinomas. This cervical cancer 
model, which recapitulates the progressive development of human 
cancers, has high and remarkable similarity in terms of both his-
tological and molecular cross-correlation with its corresponding 
human counterpart (29) and has been widely used to test the effect 
of antiangiogenic drugs on tumor progression (30).
First, we established an AAV8-based strategy that we believe to 
be novel to deliver Sema3A in vivo to cervical tumors of HPV16/E2
mice. To achieve specific gene delivery to the cervix, we established 
a route of administration by injecting the recombinant AAV8 virus 
expressing either LacZ (control) or Sema3A-myc in the distal por-
tion of the abdominal aorta, just before its bifurcation into the 
2 common iliac arteries. This procedure allowed us to efficiently 
target and express within the cervix these 2 exogenous gene con-
structs. We observed that the transformation zone — the area at 
the border between endo- and ectocervix that undergoes physi-
ological metaplasia, where cervical carcinomas most frequently 
develop (30, 31) — was efficiently transduced by AAV8 (see Meth-
ods and Supplemental Figure 5). This Sema3A delivery strategy 
allowed us to perform a regression trial with HPV16/E2 mice, as 
previously described (30). Tumor-bearing animals were treated 
with LacZ control or with Sema3A, sunitinib, or the combination 
for 4 weeks, from the fifth to the sixth month of age. Compared 
with controls, treatment with either sunitinib or Sema3A alone 
strongly reduced tumor volume. Interestingly, the combination 
resulted in much more efficient tumor growth inhibition com-
pared with single drug treatments (Figure 7A).
To assess the effect of  the different therapeutic regimens on 
the formation of distant metastases in HPV16/E2 mice, we evalu-
ated incidence, volume, and number of micrometastases in vital 
organs. We evaluated tumor dissemination by means of immuno-
fluorescence and immunohistochemistry using an anti-E7 antibody 
that specifically detects the HPV16 viral oncogene E7-positive can-
cer cells in HPV16/E2 mice organs (Figure 7, B and C). Metastasis 
incidence in 6-month-old LacZ-treated (control) HPV16/E2 mouse 
liver and lungs was 58% and 50%, respectively (Figure 7, D and E). 
Interestingly, as observed in RIP-Tag2 mice, 1 month of treatment 
with sunitinib increased liver and lung metastasis incidence to 83% 
and 75%, respectively. Remarkably, Sema3A, alone and in combina-
tion with sunitinib, strongly reduced the percentage of metastases 
compared with sunitinib- or control-treated mice.
Analysis of metastasis number and volume demonstrated that 
sunitinib induced a greater number  of liver metastases, with 
Figure 
Sema3A decreases basal and sunitinib-induced hypoxia in HPV16/
E2 mice by normalizing the tumor vasculature. (A) Tumor hypoxia,
assessed by pimonidazole adduct immunostaining in serial sections
of tumors from HPV16/E2 mice treated as indicated for 4 weeks. (B)
Quantification of hypoxic tumors, expressed as percent pimonidazole
density area per tumor, per animal. Sema3A and sunitinib combined
decreased hypoxia 92% compared with sunitinib alone; Sema3A
decreased hypoxia 88% compared with controls. (C) Vessel density,
as assessed by confocal analysis of Meca32 immunostaining, was
reduced 67%, 62%, and 52% by treatment with sunitinib, sunitinib plus
Sema3A, and Sema3A, respectively, compared with controls. (DH)
Pericyte coverage, as evaluated by confocal colocalization (arrows) of
Meca32 with α-SMA (D and F) or with NG2 (E), PDGFR-β (G), or des-
min (H) and expressed as percent colocalization of pericyte markers on
tumor ECs. Combined Sema3A and sunitinib enhanced pericyte cover-
age 71% (NG2), 68% (α-SMA), 82% (PDGFR-β), and 69% (desmin)
compared with sunitinib alone. Sema3A increased pericyte coverage
40% (NG2), 48% (α-SMA), 39% (PDGFR-β), and 32% (desmin) com-
pared with controls. (I) Increased incidence of FITC-lectin–perfused
vessels (arrows) in Sema3A-treated (alone or in combination with
sunitinib) tumors compared with sunitinib-treated and control insulino-
mas. (J) Decreased FITC-dextran extravasation (arrows) in Sema3A-
treated (alone or in combination with sunitinib) tumors compared with
sunitinib-treated and control insulinomas. Results are from 5 fields per
mouse (n = 8 per treatment group). *P < 0.05, **P < 0.01, unpaired
Mann-Whitney U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org 
larger volumes,  compared with controls  (Figure  7, F and G). 
These data further corroborated the sunitinib-induced evasive 
resistance previously observed in RIP-Tag2 and other models 
(10) in a different spontaneous mouse model of carcinogenesis. 
Notably, Sema3A alone or combined with sunitinib dramatically
decreased the degree of liver and lung metastasis formation (Fig-
ure 7, B–G). These findings bolster our results obtained with RIP-
Tag2 insulinomas, demonstrating that in a very different tumor 
Figure 
Sema3A blocks DC101-induced cancer invasion and metastatic dissemination. (A) Total tumor volume of RIP-Tag2 mice in a 4-week regres-
sion trial (from 12 to 16 weeks of age). Treatment with DC101, DC101 plus Sema3A, and Sema3A reduced tumor burden 60%, 79%, and 68%,
respectively, compared with LacZ-injected, purified rat IgG–treated RIP-Tag2 controls. (B) Percent encapsulated IT, IC1, and IC2 carcinomas.
Combined treatment with Sema3A and DC101 decreased IC2 carcinoma incidence 67% compared with DC101 alone. (C) DC101-treated tumors
displayed highly invasive tumors (arrows), as shown by SV40 T-antigen immunostaining. (DG) Analysis of peripancreatic LN (D and E) and liver
(F and G) metastasis formation. Shown is (D and F) representative SV40 T-antigen immunostaining, derived from serial sections analysis of each
animal, and (E and G) quantification of metastasis incidence per animal (n = 15 per group). Combined Sema3A and DC101 decreased LN and
liver metastasis incidence 64% and 75%, respectively, compared with DC101. Sema3A treatment reduced LN and liver metastasis incidence
62% and 50%, respectively, compared with controls. *P < 0.05, **P < 0.01, unpaired Mann-Whitney U test. Scale bars: 50 μm.
research article
 The Journal of Clinical Investigation http://www.jci.org
type (i.e., cervical cancer), treatment with Sema3A alone ham-
pered tumor invasiveness and dissemination. Moreover, these 
data strengthen the notion that Sema3A can be conceived as a 
drug able to overcome the proinvasive and prometastatic effect 
of sunitinib in different cancer types.
Sema3A reduces basal and sunitinib-induced hypoxia in cervical cancer
by normalizing the vasculature. We next sought to determine whether 
Sema3A counteracts the prometastatic effect of sunitinib in the 
HPV16/E2 model by reducing tumor  hypoxia, as  was observed 
in RIP-Tag2 mice. We analyzed tumor hypoxia by pimonidazole 
immunostaining. Control HPV16/E2 mice displayed a substantial 
amount of hypoxia both in CIN3 premalignant lesions and in cer-
vical tumors (Figure 8, A and B). As observed in RIP-Tag2 mice, 
treating HPV16/E2mice with  sunitinib significantly enhanced 
hypoxic levels in tumors, in CIN3 lesions, and in the transforma-
tion zone (Figure 8A). Sema3A, alone and in combination with 
sunitinib, strongly reduced  both  basal  and  sunitinib-induced 
tumor hypoxia (Figure 8, A and B). Interestingly, the increase in 
tissue hypoxia is a critical factor that promotes cervical carcino-
genesis and has been associated with shorter progression-free and 
overall survival and with treatment failure in clinic (32). Therefore, 
the reduced hypoxia may represent a major mechanism by which 
Sema3A reduces cervical cancer progression and the metastatic 
spreading induced by sunitinib.
To confirm that Sema3A decreases tumor hypoxia by promoting 
blood vessel normalization in the HPV16/E2 model, we character-
ized pericyte coverage of tumor blood vessels by confocal micros-
copy analysis of the pericyte markers NG2, α-SMA, PDGFR-β, and 
desmin. Similar to our findings in RIP-Tag2 mice (Figure 4, A–F), 
we observed that 1 month of sunitinib treatment in tumor-bear-
ing HPV16/E2 mice significantly decreased blood vessel area and 
greatly reduced pericyte coverage compared with controls (Figure 
8, C–H). As expected, Sema3A lessened blood vessel area, but at 
the same time  induced blood vessel normalization by reducing 
vascular branching and increasing the extent of pericyte coverage 
of the vasculature compared with sunitinib or control treatments. 
Notably, combined Sema3A and sunitinib treatment restored 
pericyte coverage compared with sunitinib alone and induced a 
vascular phenotype similar to that observed in tumors treated with 
Sema3A alone. Next, we assessed blood vessel perfusion and per-
meability in order to determine the functionality of the tumor vas-
culature. In line with the pericyte coverage analysis, we found very 
poorly perfused and highly permeable blood vessels in sunitinib-
treated carcinomas, whereas treatment with Sema3A (alone or in 
combination with sunitinib) promoted the formation of a highly 
perfused and less leaky tumor vasculature (Figure 8, I and J, and 
Supplemental Figure 6, A–D). Together, these data in a transgenic 
mouse model of cancer other than RIP-Tag2 compellingly suggest 
that, by normalizing tumor vasculature and consequently reduc-
ing hypoxia, Sema3A is capable of halting cancer invasiveness and 
metastatic spreading while inhibiting tumor angiogenesis.
Sema3A overcomes the evasive resistance induced by an inhibitor of
the VEGF pathway. In order to evaluate whether Sema3A is able to 
overcome the resistance to antiangiogenic therapies that specifi-
cally and selectively interfere with the VEGF signaling pathway, we 
used DC101, a function-blocking rat monoclonal antibody raised 
against VEGFR-2  and previously used in the  RIP-Tag2 mouse 
model to assess the evasive resistance to angiogenesis inhibition 
(4, 10). Similar to the trials performed with sunitinib, we treated 
RIP-Tag2 mice for 4 weeks with DC101 alone or in combination 
with Sema3A and compared them with mice treated with Sema3A 
alone or LacZ plus purified IgG control. Treatment with DC101 
exerted effects similar to those we obtained with sunitinib in RIP-
Tag2 mice as well as previously described findings (10). Indeed, 
DC101 inhibited tumor angiogenesis and growth, but at the same 
time it increased cancer invasiveness as well as the incidence, vol-
ume, and number of LN and liver metastases (Figure 9 and Supple-
mental Figure 7, A–C). Interestingly, when Sema3A treatment was 
combined with DC101, we observed a strong reduction of tumor 
invasiveness and metastasis formation compared with DC101-
treated mice and controls (Figure 9, B–G).
In addition, whereas insulinomas treated with DC101 were highly 
hypoxic and displayed a less pericyte-covered, leakier, and poorly 
perfused vasculature, the combination of DC101 with Sema3A 
strongly reduced tumor hypoxia, increased blood vessel coverage, 
and restored the functionality of the tumor vasculature (Figure 
10). Additionally, DC101  exerted a milder effect on the tumor 
vasculature than did sunitinib. In fact, although it strongly inhib-
ited the blood vessel area, DC101 reduced the pericyte coverage of 
blood vessels less severely than sunitinib did (compare Figure 10, 
C–H, and Figure 4, A–F). Of note, in tumors treated with DC101, 
we observed a decreased number of NG2+, PDGFR-β+, and desmin+
pericytes, but a significant increase of α-SMA+ perivascular cells, 
compared  with controls  (Figure 10,  D–H).  These  observations 
corroborate recent data showing that treatment of RIP-Tag2 mice 
with DC101 specifically increased the content of α-SMA+pericytes, 
but not perivascular cells identified by other markers (33).
Compared with the control, DC101 significantly impaired the 
perfusion and increased the permeability of tumor blood vessels, 
(Figure 10, I and J, and Supplemental Figure 8, A–D), which sug-
gests that the DC101-induced rise of α-SMA+ pericytes was not 
sufficient to maintain the function of blood vessels and indicates 
that the other pericyte subpopulations would indeed  be neces-
sary to fully normalize and improve the function of the tumor 
vasculature. Accordingly, simultaneous treatment with DC101 
and Sema3A strongly increased all the subpopulation of pericytes 
(NG2+, PDGFR-β+, desmin+, andα-SMA+)  and simultaneously 
improved the perfusion and reduced the vascular leakage, simi-
lar to Sema3A treatment alone (Figure 10, D–J, and Supplemental 
Figure 8, A–D). Together, these findings indicate that Sema3A is 
able to counteract the evasive resistance induced by the specific 
inhibition of VEGF signal pathways.
Discussion
Using 2 transgenic mouse models of spontaneous tumorigenesis, 
RIP-Tag2 and HPV16/E2, we here demonstrate what we believe to 
be a novel role for Sema3A in overcoming the evasive resistance 
previously observed in preclinical mouse models upon angiogen-
esis inhibition (10, 11). When used as single therapeutic agent, 
Sema3A strongly inhibited tumor growth, similar to the effects of 
sunitinib and DC101; however, different from these latter drugs, 
Sema3A also impaired tumor invasion and dissemination to dis-
tal organs. Moreover, thanks to its vascular normalizing activity, 
Sema3A ameliorated blood vessel function, improved cancer tis-
sue oxygenation, and lessened several hypoxia-regulated signaling 
pathways that support tumor progression and invasion. Conse-
quently, Sema3A efficiently drove sunitinib- or  DC101-treated 
tumors back from a prometastatic to a benign phenotype.
Recently, several reports on acquired resistance to antiangiogenic 
therapies highlighted the need to revisit the current therapies and 
research article
The Journal of Clinical Investigation http://www.jci.org 
research article
 The Journal of Clinical Investigation http://www.jci.org
investigate the possibility of combining tumor shrinkage (induced 
by cancer starvation from oxygen and nutrients) with blood vessel 
normalization to effectively counteract the metastatic dissemina-
tion of cancer cells, favored, for example, by a hypoxic microenvi-
ronment (34). Here, we showed that the combination of Sema3A 
with sunitinib synergistically enhanced RIP-Tag2 mouse survival 
and reduced HPV16/E2mouse tumor burden, finally inducing less 
invasive and less frequent metastatic cancers in both transgenic 
mouse models. Thus, administration of Sema3A in combination 
with sunitinib may represent  an innovative and more efficient 
therapeutic strategy, thanks to the coupling of sunitinib’s robust 
antitumorigenic and antiangiogenic activities (observed in several 
human tumors; refs. 35, 36) with Sema3A’s pronormalizing, anti-
invasive, and antimetastatic activities. The main mechanism by 
which Sema3A overcame the evasive resistance both to sunitinib 
and to DC101 was the ability of this repulsive guidance cue to 
restore tumor tissue oxygenation as a result of its powerful blood 
vessel normalizing activity. It is known that the pharmacological 
targeting of pericytes may disrupt the integrity of the tumor vas-
culature, thus enabling cancer cells to transit into the circulation 
system and metastasize (37). Therefore, our data suggest that the 
vascular normalizing effect of Sema3A could reduce the proinva-
sive effects of sunitinib by simultaneously inducing tumor tissue 
normoxia and blocking cancer cell extravasation.
It has been proposed that the duration of the vascular normal-
ization window is critical to the achievement of long-lasting and 
successful therapeutic synergy between antiangiogenic and che-
motherapeutic drugs (6, 18, 19,  38). Notably, all our different 
trials demonstrated that Sema3A, alone or in combination with 
sunitinib, significantly extended the normalization window of 
tumor blood vessels and improved the delivering efficiency of che-
motherapeutic drugs to cancer tissues, by proportionally restrain-
ing the number of blood vessels and simultaneously favoring their 
coverage, maturation, and function. It is therefore conceivable to 
hypothesize new treatment strategies in which Sema3A could be 
combined with other clinically approved chemotherapeutic and/
or antiangiogenic compounds.
The strong inhibition of HIF-1α protein expression we observed, 
due to the restoration of tumor tissue oxygenation upon combined 
treatment with Sema3A and sunitinib, highlights the crucial role 
played by Sema3A in overcoming the resistance to antiangiogenic 
therapies. Several HIF-1α inhibitors identified thus far strongly 
impair tumor progression in xenograft tumor models and are either 
in the early stages of clinical trials or FDA approved for anticancer 
therapy (3, 39). Notably, it has previously been shown that the com-
bination of bevacizumab and irinotecan, a topoisomerase I inhibitor 
that also inhibits HIF-1α, induced clinical benefit in glioblastoma 
patients (40). By correlating the anti-invasive and antimetastatic 
effects of Sema3A on hypoxia-stressed cancers with the inhibition 
of expression of HIF-1α and its target genes, such as the TK receptor 
Met, our data further corroborate the main concept that combining 
HIF-1αinhibitors with antiangiogenic drugs can increase the thera-
peutic efficacy and avoid the described side effects (3).
In recent years, several mechanisms of intrinsic and acquired 
resistance to antiangiogenic agents have been described (14, 41). 
For instance, preclinical studies provided evidence of anti-VEGF 
drug evasion by activation of alternative pathways of angiogen-
esis and tumor progression (2). RIP-Tag2 mice have shown rapid 
adaptation to anti-VEGF agents, followed by tumor regrowth as 
a result of FGF signaling activation (4). An additional possibility 
could therefore be that activation of proangiogenic pathways, such 
as those triggered by FGFs, can be involved in developing suni-
tinib resistance in RIP-Tag2 mice and that the addition of Sema3A 
can inhibit  the  activation  of these compensatory signal path-
ways. Interestingly, the data we obtained by combining Sema3A 
with DC101 further corroborated and strengthened the results 
obtained with sunitinib and demonstrated that by normalizing 
the vasculature and reducing tumor hypoxia, Sema3A overcame 
the evasive resistance  induced  by  inhibition of both  VEGF (via 
DC101) and multiple TK receptor–dependent signaling pathways 
(via sunitinib). Notably, our observation of an enhanced number 
of α-SMA+ mural cells, paralleled by a simultaneous reduction 
of the other pericyte subpopulations (i.e., NG2+, PDGFR-β+, and 
desmin+ cells), corroborates recent data showing that treatment 
of RIP-Tag2 mice with DC101 specifically increases the content 
of α-SMA+pericytes (33). As those authors suggested, DC101 is 
likely to induce a subpopulation of tumor blood vessels covered 
by α-SMA+pericytes deriving from co-opted blood vessels. One 
could hence speculate that the increased amount of tumor blood 
vessels surrounded by α-SMA+ pericytes may be attributable to the 
milder effect that DC101 exerts on blood vessel perfusion and per-
meability compared with sunitinib. However, since we observed 
that DC101 significantly impaired perfusion and increased the 
permeability of tumor blood vessels compared with controls, such 
a DC101-induced rise in α-SMA+ pericytes does not appear suf-
ficient to support the reconstitution of physiologically function-
ing blood vessels. Therefore, the other pericyte subpopulations 
seem to be necessary to warrant the appearance of an efficiently 
normalized tumor vasculature. Accordingly, simultaneous treat-
ment with DC101 and Sema3A, similar to what we observed with 
Sema3A alone, strongly increased all the pericyte subpopulations 
and simultaneously improved the  perfusion and reduced the 
vascular leakage. These observations indicate that sunitinib and 
DC101 exert different effects on the tumor vasculature, suggesting 
Figure 0
Sema3A impairs DC101-induced hypoxia and improves tumor blood
vessel function. (A) Tumor hypoxia, assessed by pimonidazole adduct
immunostaining (arrows) in tumors from mice treated as indicated
for 4 weeks. (B) Quantification of hypoxic tumors, expressed as per-
cent pimonidazole density area per tumor, per animal. Sema3A and
DC101 combined decreased hypoxia 88% compared with DC101
alone; Sema3A decreased hypoxia 90% compared with controls.
(C) Vessel density, as assessed by confocal microscopy of Meca32
immunostaining, was reduced 75%, 73%, and 50% by treatment with
DC101, DC101 plus Sema3A, and Sema3A, respectively, compared
with controls. (DH) Pericyte coverage, as evaluated by confocal colo-
calization (arrows) of Meca32 with NG2 (D and E) or with α-SMA (F),
PDGFR-β (G), or desmin (H) and expressed as percent colocalization
of pericyte markers on tumor ECs. Combined Sema3A and DC101
enhanced pericyte coverage 56% (NG2), 59% (PDGFR-β), and 48%
(desmin) compared with DC101. Sema3A increased pericyte coverage
45% (NG2), 47% (PDGFR-β), and 40% (desmin) compared with con-
trols. DC101 increased α-SMA+ pericyte coverage 46% versus con-
trols. (I) Increased incidence of FITC-lectin–perfused vessels (arrows)
in Sema3A-treated (alone or in combination with DC101) tumors com-
pared with DC101-treated and control insulinomas. (J) Decreased
FITC-dextran extravasation (arrows) in Sema3A-treated (alone or
in combination with DC101) tumors compared with DC101-treated
and control insulinomas. Results are from 5 fields per mouse (n = 8
per treatment group). *P < 0.05, **P < 0.01, unpaired Mann-Whitney
U test. Scale bars: 50 μm.
research article
The Journal of Clinical Investigation http://www.jci.org 
how these 2 drugs may induce evasive resistance to angiogenesis 
inhibition through different molecular mechanisms. Indeed, simi-
lar to sunitinib, DC101 triggered tumor hypoxia, but, differently 
from sunitinib, also co-opted blood vessels, a phenomenon that 
has previously been correlated with the development of acquired 
resistance to antiangiogenic therapies in RIP-Tag2 mice (4).
In this study,  we  showed that treating RIP-Tag2 tumors with 
sunitinib highly increased NF-κB expression. Since NF-κB acti-
vates HIF-1α and promotes EMT, cancer invasion, and  tumor 
angiogenesis in several tumor types (25, 26, 42), our data suggest 
that NF-κB plays an important role in the development of eva-
sive resistance in response to standard antiangiogenic therapies 
and that inhibition of NF-κB expression may represent a further 
mechanism by which Sema3A can overcome the side effects caused 
by angiogenesis inhibition. It has also been observed that during 
progression, tumors recruit proangiogenic myeloid cells that can 
contribute to the intrinsic resistance to antiangiogenic therapies 
(43). Of note, Gr1+MMP9+ cells, which increase the bioavailability 
of VEGF for its receptors (44), and tumor-associated macrophages 
(TAMs) expressing cathepsins B and S are critical promoters of 
tumor growth, angiogenesis, and invasion in RIP-Tag2 mice (45). 
Because NF-κB  orchestrates the tissue inflammatory response 
induced by hypoxia, including leukocyte infiltration (25, 46, 47), 
it is conceivable that, by upregulating NF-κB expression, sunitinib 
could induce the recruitment and activation of neutrophils, TAMs, 
and other protumoral myeloid cells. Simultaneous administration 
of Sema3A together with sunitinib restored tumor tissue normoxia 
and reduced NF-κB, and could therefore inhibit the appearance of 
these inflammatory cell populations. Further studies are required 
to clarify the effect of Sema3A on bone marrow–derived proangio-
genic cells during angiogenesis inhibition.
Inhibition of tumor angiogenesis by sunitinib strongly increased 
the expression and tyrosine phosphorylation of Met receptor in 
RIP-Tag2 tumors. Met and phospho-Met were present in cancer 
cells and, to a lesser extent, in vessels of untreated mice, highlight-
ing the key role played by the Met receptor in tumor angiogenesis 
and progression (48–50). Notably, 1 month of sunitinib treatment 
strongly increased Met phosphorylation in tumor cells and not in 
ECs, suggestive of specific activation of the proinvasive HGF/Met 
pathway in cancer cells, but not in the tumor vasculature. The dra-
matic inhibition of Met expression and phosphorylation induced 
by Sema3A, alone or in combination with sunitinib, along with 
the substantial reduction of tumor spreading and metastatization 
indicated that HGF/Met signaling inhibition is an additional key 
mechanism by which Sema3A can overcome the evasive resistance 
to antiangiogenic therapies.
It is worth noting that treatment of tumor-bearing RIP-Tag2 
mice with AVV8-Sema3A as a single agent reduced invasiveness 
and metastasis  formation by increasing E-cadherin expression 
and inhibiting Met TK receptor activation in cancer cells com-
pared with control insulinomas. Even though untreated tumors 
displayed a milder hypoxia than did sunitinib-treated insulino-
mas, 2 hypoxia-induced genes, CA9 and NF-κB, were significantly 
reduced in Sema3A-treated mice compared with controls. We spec-
ulate that Sema3A, by completely  restoring tumor oxygenation 
and by inhibiting hypoxia-induced signal pathways in end-stage 
RIP-Tag2 tumors, may be responsible for the observed increase of 
E-cadherin levels, inhibition of Met activation, and consequent 
reduction of tumor invasion and metastatization. However, given 
the complexity of the tumor microenvironment (14, 41), comple-
mentary mechanisms may also mediate the effects of Sema3A on 
tumor angiogenesis and cancer progression. Further investigation 
is hence needed to clarify these aspects.
In conclusion, our studies indicate that Sema3A administration 
may represent a new therapeutic approach to inhibit angiogenesis, 
while promoting the maturation of the surviving vasculature and 
hence avoiding the commonly observed long-lasting tumor hypox-
ia that, if not hindered, can support the lethal dissemination of 
cancer cells throughout the body. This pharmacological strategy 
may help to better and safely harness the therapeutic potential of 
antiangiogenic drugs for the final benefit of oncologic patients.
Methods
Further information can be found in Supplemental Methods.
Transgenic tumor model. The RIP-Tag2 transgenic mouse model has been 
previously described (10, 13, 51). RIP-Tag2 mice were generated and main-
tained in the C57BL/6 background (Jackson Laboratory). From 12 weeks 
of age, all RIP-Tag2 mice received 50% sugar food (Harlan Teklad) and 5% 
sugar water to relieve hypoglycemia induced by the insulin-secreting tumors. 
Generation of K14-HPV16 transgenic mice (52) and E2 treatment for cervi-
cal carcinogenesis has been previously reported (30, 31). Briefly, 1-month-
old virgin female transgenic (heterozygous K14-HPV16) were anesthetized, 
and continuous-release pellets that deliver E2 at 0.05-mg doses over 60 days 
(Innovative Research of America Inc.) were implanted s.c. in the dorsal back 
skin. Subsequent pellets were implanted at 3 and 5 months of age. The result-
ing HPV16/E2mice were maintained in the FVB/n background (The Jackson 
Laboratory). Mice were monitored throughout the experiments for compli-
cations caused by the dysplastic nature of their skin or by E2treatment. 
Therapeutic treatments. Tumor- bearing RIP-Tag2 or HPV16/E2 mice were 
treated for 4 weeks, from 12 until 16 weeks or from 5 until 6 months of 
age, respectively (regression trial). Different regression trials were designed: 
(a) 40 mg/kg/d sunitinib l-malate (Axon Medchem BV) was administered 
daily by oral gavage (n = 30); (b) 1  mg/mouse rat monoclonal function-
blocking antibodies against VEGFR-2 (DC101), obtained in bulk by affin-
ity purification from the supernatant of a hybridoma culture (DC101) 
(Translational Research Laboratory, Catalan Institute of Oncology), was 
administered twice weekly i.p. (n = 15), as previously reported (4); (c) 100 μl 
Sema3A was injected slowly through  the abdominal  aorta of RIP-Tag2 
mice using a 30-gauge needle (Roboz Surgical Instrument) (n =  30), as 
previously described (13), or through the distal portion of the abdomi-
nal aorta just before its bifurcation into the 2 common iliac arteries of 
HPV16/E2 mice (n = 12 per group) (see In vivo AAV8 administration); and (d) 
Sema3A-injected mice were treated daily by oral gavage with 40 mg/kg/d 
sunitinib l-malate or twice weekly with 1 mg/mouse DC101 (n = 15). Con-
trol mice were injected with LacZ (13) and treated with methylcellulose 
vehicle daily by oral gavage (n = 30) or with 1 mg/mouse purified rat IgG 
(Jackson Immunoresearch) i.p. (n = 15). For the survival trial, 12-week-old 
Rip-Tag2 mice were treated with 40 mg/kg/d sunitinib, Sema3A, combined 
Sema3A and sunitinib, or LacZ plus vehicle (n = 20 per group), and their 
survival was monitored over time.
In vivo AAV8 administration. AAV8-Sema3A was administered in RIP-Tag2 
mice as previously described (13). For AAV8-LacZ or AAV8-Sema3A delivery 
in HPV16/E2 mice, animals were anesthetized by 1.5% isoflurane anesthesia. 
The distal portion of the abdominal aorta just before its bifurcation into 
the 2 common iliac arteries was exposed following a displacement of intes-
tine and urinary bladder and isolated from the surrounding fat tissue. 50 μl 
recombinant AAV8-Sema3A or AAV8-LacZ virus was injected slowly through 
the abdominal aorta, by means of a 31-gauge needle of an insulin syringe. 
After injection, homeostasis was performed. The abdomen was then closed 
layer-to-layer with 5-0 chromic gut sutures. Animals were subsequently mon-
research article
 The Journal of Clinical Investigation http://www.jci.org
itored and allowed to recover 1–2 hours after surgery. Postsurgical analgesia 
was achieved by buprenorphine (0.1 mg/kg s.c. every 12 hours for 1 day) and 
antibiotic prophylaxis with ampicillin (150 mg/kg s.c.).
Invasion index and metastasis analysis. Paraffin sections of pancreas, liver, 
and LN from each group of treatment were serially cut (10 μm) and rehy-
drated  through 100%  xylene  and  100%,  95%,  and  70%  ethanolbefore 
immersion in  1× PBS.  Sections were stained with  H&E  as  previously 
described (10). Tissues were visualized with a BX-60 microscope (Olym-
pus) equipped with a color Qicam Fast 1394-digital CCD camera (12 bits; 
QImaging Corp.).  The  invasion index  of  tumors was determined using 
5 H&E-stained sections  per animal. Based on gross morphological and 
detailed histopathological characteristics, tumor islets were subdivided 
into insulinomas with well-defined margins and frequent fibrous capsules 
(IT), carcinomas with focal regions of invasion (IC1), or fully invasive car-
cinomas (IC2) (10). The presence or absence of tumor cell dissemination 
was first evaluated by H&E staining and confirmed by rabbit anti-SV40 
T-antigen (sc-20800, 1:50; Santa Cruz) or HPV16-E7 (250629, 1:100; Abbio-
tec) Abs and by immunof luorescence, according to previously described 
protocols. LN and liver metastasis incidence in RIP-Tag2 mice and lung 
and liver metastasis incidence in HPV16/E2 transgenic mice was evaluated 
by scoring for presence or absence in each animal using 10 sections per 
animal. The number of liver metastases was measured as SV40 T-antigen 
or HPV16-E7 protein-positive regions in 10 images per mouse per treat-
ment group. To quantify the metastatic volume, we used ImageJ software 
to compare the metastatic mass with a spheroid. In each image, we drew 
a line corresponding to the width (w) of the metastatic region and a line 
corresponding to the length (l), then calculated the LN metastatic volume 
as 0.52 ×w2×l, as for a spheroid.
Tumor hypoxia analysis. The amount of tumor hypoxia was determined 
2 hours after injection of 60  mg/kg pimonidazole hydrochloride (HP2-
100 Hypoxyprobe Kit-Plus;  Natural Pharmacia International Inc.) into 
RIP-Tag2 mice (13). The formation of pimonidazole adducts was detected 
by immunostaining with Hypoxyprobe-1-Mab1 FITC Ab according to the 
manufacturer’s instructions. Immunofluorescence images were captured 
and analyzed using a Leica TCS SP2 AOBS confocal laser-scanning micro-
scope (Leica Microsystems) and then evaluated by Image-ProPlus 6.2 soft-
ware (Media Cybernetics). Quantification was done by analyzing at least 
5 sections and 5 fields per tumor.
Tumor vessel perfusion and vascular permeability. To detect  tumor vessel 
perfusion and vascular permeability, mice were injected i.v. with 0.05 mg 
FITC-labeled tomato lectin (Lycopersicon esculentum; Vector Laborato-
ries) or 0.05 mg 70-kDa FITC-conjugated dextran (Molecular Probes). After 
2 minutes, the animals were euthanized, and the heart was perfused with 
saline solution followed by 2% PFA. Lectin and dextran distribution was 
visualized by fluorescent confocal microscopy z-sectioning that allowed 
for 3D reconstruction of the vascular network.
Confocal microscopy quantifications. To quantify pericyte coverage (NG2, 
α-SMA, PDGFR-β, or desmin, shown by red channels) in each image, we 
drew a region of interest (ROI) close to each blood vessel (Meca32, shown 
by green channels) and then quantified the MFI of red and green channels 
using the Leica Confocal Software Histogram Quantification Tool. We 
then calculated the ratio between red and green channel MFI; values are 
expressed as percentage of red-green costaining. A similar procedure was 
followed to quantify vascular perfusion by FITC-labeled lectin. Blood vessel 
permeability was analyzed by measuring the area of dextran extravasation. 
To determine the expression levels of E-cadherin (green channel), vimentin 
(red channel), total Met, and phospho-Met (red channel) in each analyzed 
image, we considered 5 random ROIs of the same size. Then we measured 
the ratio between the red or green channel and the blue (DAPI) channel MFI; 
data are presented as percent positive cells relative to total cell number.
Measurement of in vivo tumor bioavailability of doxorubicin. A group of RIP-
Tag2 mice, previously treated for 4 weeks with LacZ  plus vehicle  (con-
trol) or with Sema3A, sunitinib, or Sema3A and sunitinib combined, was 
injected with 10 mg/kg doxorubicin hydrochloride (Sigma-Aldrich) via the 
lateral tail vein 4 hours before sacrifice. Pancreatic tumors and kidneys 
as controls were collected from each mouse and weighed. Samples were 
resuspended in a lysis buffer (0.25 M sucrose, 5 mM TrisHCl pH 7.6, 1 mM 
MgSO4, 1 mM CaCl2) and homogenized in an ice-cold Potter homogenizer. 
200 μl of each homogenate was placed into a new microcentrifuge tube, 
and 100μl 10% Triton X-100, 200 μl water, and 1.5 ml acidified isopro-
panol was added. The mixture was vortexed and kept at –20°C overnight. 
The next day, samples were warmed to room temperature ad centrifuged at 
15,000 g for 20 minutes. Doxorubicin was quantified by spectrophotomet-
ric analysis (λ ex, 470 nm; λ em, 590 nm) using the Synergy HT (BioTek) 
plate reader. These values were compared with a standard curve of known 
amounts of doxorubicin and normalized based on the weight of the each 
organ and on the fluorescence emission of the control tissue, calculated 
as the fluorescence/weight ratio of the tumor divided by the fluorescence/
weight ratio of the kidney. Data are mean ± SD of triplicate aliquots from 
tumor homogenates expressed as μ equivalents/g tissue of doxorubicin.
Statistics. All values are expressed as mean ± SD. For all statistical analyses, 
a 2-tailed, unpaired Mann-Whitney U test was used. Statistical analysis for 
the survival trial was performed using the log-rank test. A P value less than 
0.05 was considered significant.
Study approval. All animal procedures were approved by the Ethical Com-
mission of the University of Torino and by the Italian Ministry of Health 
in compliance with the international laws and policies.
Acknowledgments
We thank Marta Paez-Ribes for helping to evaluate tumor invasive-
ness and metastasis formation and Doug Hanahan for discussion 
and insightful suggestions. This work was supported by Associazione 
Italiana per la Ricerca sul Cancro (AIRC) investigator grants (5837 to 
E. Giraudo, 9211 to G. Serini, and 10133 to F. Bussolino); by AIRC 
2010 Special Program in Molecular Clinical Oncology 5% Project no. 
9970 (to E. Giraudo and F. Bussolino); by Regione Piemonte Ricerca 
Sanitaria Finalizzata 2007, 2008, and 2008 bis (to E. Giraudo, G. 
Serini, and F. Bussolino); by Ricerca industriale e sviluppo precom-
petitivo 2006 grants PRESTO and SPLASERBA (to F. Bussolino); by 
Associazione Augusto per la Vita (to G. Serini); by Compagnia di San 
Paolo — Neuroscience Program Multicentre Projects (to G. Serini); by 
Ministero della Salute Programma di Ricerca Finalizzata 2006 and 
Programma Straordinario di Ricerca Oncologica 2006 (to E. Giraudo 
and F. Bussolino); by Converging Technologies grant PHOENICS (to 
E. Giraudo and F. Bussolino); by Fondazione Guido Berlucchi (to E. 
Giraudo and G. Serini); by Fondazione Cassa di Risparmio Torino 
(CRT) (to E. Giraudo and F. Bussolino); by Fondazione Piemontese 
per la Ricerca sul Cancro-ONLUS (Intramural Grant 5% 2008) (to 
E. Giraudo and G. Serini); and by Telethon Italy (to G. Serini). F. 
Maione was supported by fellowship “26 fellowship — FIRC” granted 
by Fondazione Italiana per la Ricerca sul Cancro (FIRC).
Received for publication May 12, 2011, and accepted in revised 
form February 22, 2012.
Address correspondence to: Enrico Giraudo, Laboratory of Trans-
genic Mouse Models, IRCC, and Department of Oncological Scienc-
es, University of Torino School of Medicine, Strada Provinciale 142, 
Km 3.95, I-10060 Candiolo, Turin, Italy. Phone: 39.011.9933505; 
Fax: 39.011.9933524; E-mail: enrico.giraudo@ircc.it.
research article
The Journal of Clinical Investigation http://www.jci.org 
1. Folkman J. Angiogenesis.  Annu Rev Med.  200 6; 
57:1–18.
2. Duda  DG,  Batchelor  TT, Willett CG, Jain  RK. 
VEGF-targeted cancer therapy strategies: current 
progress, hurdles and future prospects. Trends Mol
Med. 2007;13(6):223–230.
3. Rapisarda A, Melillo G. Role of the hypoxic tumor 
microenvironment in the resistance to anti-angio-
genic therapies. Drug Resist Updat. 2009;12(3):74–80.
4. Casanovas O, Hicklin DJ, Bergers G, Hanahan D. 
Drug resistance by evasion of  antiangiogenic tar-
geting of VEGF signaling in late-stage pancreatic 
islet tumors. Cancer Cell. 2005;8(4):299–309.
5. Baluk P, Hashizume H, McDonald  DM. Cellular 
abnormalities of blood vessels as targets in cancer. 
Curr Opin Genet Dev. 2005;15(1):102–111.
6. Jain RK. Normalization of tumor vasculature: an 
emerging concept in antiangiogenic therapy. Sci-
ence. 2005;307(5706):58–62.
7. Semenza GL. Hypoxia and cancer. Cancer Metastasis
Rev. 2007;26(2):223–224.
8. Sabbah M, et al. Molecular signature and therapeu-
tic perspective of the  epithelial–to–mesenchymal 
transitions in epithelial cancers. Drug Resist Updat. 
2008;11(4–5):123–151.
9. Pennacchietti S, Michieli P, Galluzzo M, Mazzone 
M, Giordano S, Comoglio PM. Hypoxia promotes 
invasive growth by transcriptional activation of the 
met protooncogene. Cancer Cell. 2003;3(4):347–361.
10. Paez-Ribes M, et al. Antiangiogenic therapy elic-
its malignant progression of tumors to increased 
local invasion and distant metastasis.  Cancer Cell. 
2009;15(3):220–231.
11. Ebos JM, Lee  CR, Cruz-Munoz W, Bjarnason  GA, 
Christensen JG, Kerbel  RS. Accelerated  metastasis 
after short-term treatment with a potent inhibitor of 
tumor angiogenesis. Cancer Cell. 2009;15(3):232–239.
12. Galluzzo M, Pennacchietti S, Rosano S, Comoglio 
PM, Michieli P. Prevention of hypoxia by myoglo-
bin expression in human tumor cells promotes dif-
ferentiation and inhibits metastasis. J Clin Invest. 
2009;119(4):865–875.
13. Maione F, et al. Semaphorin 3A  is an endogenous 
angiogenesis inhibitor that blocks tumor growth and 
normalizes tumor vasculature in transgenic mouse 
models. J Clin Invest. 2009;119(11):3356–3372.
 14. Bergers G,  Hanahan D.  Modes  of resistance to 
anti-angiogenic therapy.  Nat Rev Cancer.  2008; 
8(8):592–603.
15. Christensen JG. A preclinical review of  sunitinib, 
a multitargeted receptor tyrosine kinase inhibitor 
with anti-angiogenic  and antitumour activities. 
Ann Oncol. 2007;18 suppl 10:x3–x10.
16. Casazza A, et al. Systemic and targeted delivery of 
semaphorin 3A inhibits  tumor angiogenesis and 
progression in mouse tumor models. Arterioscler
Thromb Vasc Biol. 2011;31(4):741–749.
17. Sathornsumetee S, et  al. Tumor angiogenic  and 
hypoxic profiles predict radiographic response and 
survival in malignant astrocytoma patients treated 
with bevacizumab  and  irinotecan.  J Clin Oncol. 
2008;26(2):271–278.
18. Winkler F, et al.  Kinetics of  vascular normaliza-
tion by  VEGFR2 blockade  governs  brain tumor 
response to radiation: role of oxygenation, angio-
poietin-1, and matrix metalloproteinases. Cancer
Cell. 2004;6(6):553–563.
19. Goel S, et al. Normalization of the vasculature for 
treatment of cancer and other diseases. Physiol Rev. 
2011;91(3):1071–1121.
20. Bergers G, Song  S, Meyer-Morse N,  Bergsland E, 
Hanahan D. Benefits of targeting both pericytes and 
endothelial cells in the tumor vasculature with kinase 
inhibitors. J Clin Invest. 2003;111(9):1287–1295.
21. Yilmaz M,  Christofor i G. EM T, the cy toskeleton, 
and cancer cell  invasion.  Cancer Metastasis Rev. 
2009;28(1–2):15–33.
22. Perl AK, Wilgenbus P, Dahl U,  Semb H, Christo-
fori G. A causal  role for E-cadherin in the transi-
tion from  adenoma to  carcinoma. Nature.  1998; 
392(6672):190–193.
23. McConkey DJ, et al. Role of epithelial–to–mesen-
chymal  transition  (EMT)  in  drug  sensitivity  and 
metastasis in bladder cancer. Cancer Metastasis Rev. 
2009;28(3–4):335–344.
24. Min C, Eddy SF, Sherr DH, Sonenshein GE.  NF-
kappaB and epithelial to mesenchymal transition 
of cancer. J Cell Biochem. 2008;104(3):733–744.
 25. Sark ar  FH,  Li Y,  Wang Z,  Kong  D. NF-kappaB 
signaling pathway and  its therapeutic  implica-
tions in human diseases. Int Rev Immunol. 2008; 
27(5):293–319.
26. van Uden P,  Kenneth NS,  Rocha S. R egulation of 
hypoxia-inducible factor-1alpha by  NF-kappaB. 
Biochem J. 2008;412(3):477–484.
27. Cummins EP, et al. Prolyl hydroxylase-1 negatively 
regulates IkappaB kinase-beta, giving insight into 
hypoxia-induced NFkappaB activity. Proc Natl Acad
Sci U S A. 2006;103(48):18154–18159.
28. Culver C, Sundqvist A, Mudie S, Melvin A, Xirodi-
mas D, Rocha S.  Mechanism of hypoxia-induced 
NF-kappaB. Mol Cell Biol. 2010;30(20):4901–4921.
29. Smith-McCune K, Zhu YH, Hanahan D, Arbeit  J. 
Cross-species comparison of angiogenesis during 
the premalignant stages of squamous carcinogene-
sis in the human cervix and K14-HPV16 transgenic 
mice. Cancer Res. 1997;57(7):1294–1300.
30. Giraudo E,  Inoue  M,  Hanahan  D.  An  amino-
bisphosphonate targets MMP-9-expressing macro-
phages and angiogenesis to impair cervical carcino-
genesis. J Clin Invest. 2004;114(5):623–633.
31. Arbeit JM,  Howley  PM,  Hanahan  D.  Chronic 
estrogen-induced cervical and vaginal squamous 
carcinogenesis in human papillomavirus  type 16 
transgenic mice.  Proc Natl Acad Sci U S A.  1996; 
93(7):2930–2935.
32. Seeber LM, et al. The role of hypoxia inducible fac-
tor-1alpha in gynecological cancer. Crit Rev Oncol
Hematol. 2011;78(3):173–184.
33. Franco M,  Paez-Ribes  M, Cor tez E,  Casanovas  O, 
Pietras K. Use of a mouse model of pancreatic neu-
roendocrine tumors to find pericyte biomarkers of 
resistance to anti-angiogenic therapy. Horm Metab
Res. 2011;43(12):884–889.
34. Carmeliet P, Jain RK. Principles and mechanisms of 
vessel normalization for cancer and other angiogen-
ic diseases. Nat Rev Drug Discov. 2011;10(6):417–427.
 35. Goodman VL,  et  al.  Approval  summar y:  suni-
tinib for the  treatment of  imatinib refractory  or 
intolerant gastrointestinal  stromal  tumors and 
advanced renal  cell  carcinoma.  Clin Cancer Res. 
2007;13(5):1367–1373.
36. Raymond E,  et al . Sunitin ib malat e for t he treat -
ment of pancreatic neuroendocrine tumors. N Engl
J Med. 2011;364(6):501–513.
37. Xian X, et al. Pericytes limit tumor cell metastasis. 
J Clin Invest. 2006;116(3):642–651.
 38. Sorensen AG,  et  al.  A  “vascular  normalization 
index”  as  potential  mechanistic  biomarker  to 
predict survival after a single dose of cediranib in 
recurrent glioblastoma patients. Cancer Res. 2009; 
69(13):5296–5300.
39. Rapisarda A,  et  al.  Schedule-dependent  inhibi-
tion of  hypoxia-inducible factor-1alpha protein 
accumulation, angiogenesis, and tumor growth by 
topotecan in U251-HRE glioblastoma xenografts. 
Cancer Res. 2004;64(19):6845–6848.
40. Vredenburgh JJ, et al. Bevacizumab plus irinotecan 
in recurrent glioblastoma multiforme. J Clin Oncol. 
2007;25(30):4722–4729.
41. Carmeliet  P,  Jain  RK.  Molecular  mechanisms 
and clinical applications of  angiogenesis. Nature. 
2011;473(7347):298–307.
42. Nam SY, et al. A hypoxia-dependent upregulation 
of hypoxia-inducible factor-1 by nuclear factor-
kappaB  promotes  gastric  tumour  growth and 
angiogenesis. Br J Cancer. 2011;104(1):166–174.
 43. Shojaei  F,  et  al.  Bv8  regulates  myeloid-cell-
dependent tumour  angiogenesis.  Nature.  2007; 
450(7171):825–831.
44. Nozawa H, Chiu C, Hanahan D. Infiltrating  neu-
trophils mediate the  initial angiogenic switch in 
a mouse model of multistage carcinogenesis. Proc
Natl Acad Sci U S A. 2006;103(33):12493–12498.
45. Gocheva V, et al. IL-4 induces cathepsin  protease 
activity in tumor-associated macrophages to pro-
mote cancer growth and invasion. Genes Dev. 2010; 
24(3):241–255.
46. Fitzpatrick SF, et al. An intact canonical NF-kap-
paB pathway is required  for inf lammatory gene 
expression in response to hypoxia. J Immunol. 2011; 
186(2):1091–1096.
47. Porta C,  et al. Cellular  and molecular  pathways 
linking inflammation and cancer. Immunobiology. 
2009;214(9–10):761–777.
48. Christensen J, Anderes K.  Beyond VEGF:  target-
ing tumor growth and angiogenesis via alternative 
mechanisms. Adv Exp Med Biol. 2008;610:43–53.
49. Abounader R, Laterra J. Scatter factor/hepatocyte 
growth factor  in brain tumor growth and angio-
genesis. Neuro Oncol. 2005;7(4):436–451.
50. Bussolino F, et al. Hepatocyte growth factor is a potent 
angiogenic factor which stimulates endothelial cell 
motility and growth. J Cell Biol. 1992;119(3):629–641.
51. Hanahan D.  Heritable  formation  of  pancreatic 
beta-cell tumours  in transgenic  mice expressing 
recombinant insulin/simian virus 40 oncogenes. 
Nature. 1985;315(6015):115–122.
52. Coussens LM, Hanahan D, Arbeit JM. Genetic pre-
disposition and parameters of malignant progres-
sion in K14-HPV16  transgenic mice. Am J Pathol. 
1996;149(6):1899–1917.
... However, mice treated with sunitinib demonstrated an increase in local invasion, distant metastasis and hypoxia compared to the control mice. These unhelpful effects of sunitinib were overcome with combined SEMA3A treatment, with the combination demonstrating not only a reduced tumour burden and reduced angiogenesis but also reduced cancer invasion into surrounding tissue, including lymph node and liver metastases [55]. Combining SEMA3A treatment with sunitinib also reversed hypoxia and promoted blood vessel normalisation by reducing vascular branching [55]. ...
... These unhelpful effects of sunitinib were overcome with combined SEMA3A treatment, with the combination demonstrating not only a reduced tumour burden and reduced angiogenesis but also reduced cancer invasion into surrounding tissue, including lymph node and liver metastases [55]. Combining SEMA3A treatment with sunitinib also reversed hypoxia and promoted blood vessel normalisation by reducing vascular branching [55]. In breast cancer, a phase II clinical trial evaluated the efficacy of sunitinib in metastatic breast cancer patients with favourable outcomes in TNBC and HER-2-positive tumours (NCT00471276). ...
Article
Full-text available
Breast cancer is the most common cancer worldwide and a leading cause of cancer-related deaths in women. The clinical management of breast cancer is further complicated by the heterogeneous nature of the disease, which results in varying prognoses and treatment responses in patients. The semaphorins are a family of proteins with varied roles in development and homoeostasis. They are also expressed in a wide range of human cancers and are implicated as regulators of tumour growth, angiogenesis, metastasis and immune evasion. More recently, semaphorins have been implicated in drug resistance across a range of malignancies. In breast cancer, semaphorins are associated with resistance to endocrine therapy as well as breast cancer chemotherapeutic agents such as taxanes and anthracyclines. This review will focus on the semaphorins involved in breast cancer progression and their association with drug resistance.
... While the mechanisms by which blocking VEGFA improves tumor response to chemotherapy are incompletely understood 17,18 , VEGFA signaling stimulates vascular ECs and TAMs to adopt immunosuppressive phenotypes that limit the efficacy of chemotherapy and other anticancer agents 44,150,187 . Beyond VEGF inhibition, additional strategies can demonstrably normalize TBVs, including enforced expression of semaphorin 3A (SEMA3A) 188 , delivery of agonists of the lymphotoxin-β receptor (LTβR) 189 , blocking ANGPT2 (refs. 79,80,85) or activating TIE2 (ref. ...
Article
Research into the mechanisms and manifestations of solid tumor vascularization was launched more than 50 years ago with the proposition and experimental demonstrations that angiogenesis is instrumental for tumor growth and was, therefore, a promising therapeutic target. The biological knowledge and therapeutic insights forthcoming have been remarkable, punctuated by new concepts, many of which were not foreseen in the early decades. This article presents a perspective on tumor vascularization and its therapeutic targeting but does not portray a historical timeline. Rather, we highlight eight conceptual milestones, integrating initial discoveries and recent progress and posing open questions for the future.
Article
Full-text available
Semaphorin-3A (SEMA3A) functions as a chemorepulsive signal during development and can affect T cells by altering their filamentous actin (F-actin) cytoskeleton. The exact extent of these effects on tumour-specific T cells are not completely understood. Here we demonstrate that Neuropilin-1 (NRP1) and Plexin-A1 and Plexin-A4 are upregulated on stimulated CD8⁺ T cells, allowing tumour-derived SEMA3A to inhibit T cell migration and assembly of the immunological synapse. Deletion of NRP1 in both CD4⁺ and CD8⁺ T cells enhance CD8⁺ T-cell infiltration into tumours and restricted tumour growth in animal models. Conversely, over-expression of SEMA3A inhibit CD8⁺ T-cell infiltration. We further show that SEMA3A affects CD8⁺ T cell F-actin, leading to inhibition of immune synapse formation and motility. Examining a clear cell renal cell carcinoma patient cohort, we find that SEMA3A expression is associated with reduced survival, and that T-cells appear trapped in SEMA3A rich regions. Our study establishes SEMA3A as an inhibitor of effector CD8⁺ T cell tumour infiltration, suggesting that blocking NRP1 could improve T cell function in tumours.
Article
Full-text available
Vessel co-option (VCO) is a non-angiogenic mechanism of vascularization that has been associated to anti-angiogenic therapy. In VCO, cancer cells hijack the pre-existing blood vessels and use them to obtain oxygen and nutrients and invade adjacent tissue. Multiple primary tumors and metastases undergo VCO in highly vascularized tissues such as the lungs, liver or brain. VCO has been associated with a worse prognosis. The cellular and molecular mechanisms that undergo VCO are poorly understood. Recent studies have demonstrated that co-opted vessels show a quiescent phenotype in contrast to angiogenic tumor blood vessels. On the other hand, it is believed that during VCO, cancer cells are adhered to basement membrane from pre-existing blood vessels by using integrins, show enhanced motility and a mesenchymal phenotype. Other components of the tumor microenvironment (TME) such as extracellular matrix, immune cells or extracellular vesicles play important roles in vessel co-option maintenance. There are no strategies to inhibit VCO, and thus, to eliminate resistance to anti-angiogenic therapy. This review summarizes all the molecular mechanisms involved in vessel co-option analyzing the possible therapeutic strategies to inhibit this process.
Preprint
Full-text available
Background More than 60% of patients with head and neck squamous carcinoma (HNSCC) are diagnosed at advanced stages and miss radical treatment. This has prompted the need to find new biomarkers to achieve early diagnosis and predict early recurrence and metastasis of tumors. Methods Single-cell RNA sequencing (scRNA-seq) data from HNSCC tissues and peripheral blood samples were obtained through the Gene Expression Omnibus (GEO) database (GSE164690) to characterize the B-cell subgroups, differentiation trajectories, and intercellular communication networks in HNSCC and to construct a prognostic model of the associated risks. In addition, this study analyzed the differences in clinical features, immune cell infiltration, functional enrichment, tumor mutational burden (TMB), and drug sensitivity between the high- and low-risk groups. Results Using scRNA-seq of HNSCC, we classified B and plasma cells into a total of four subgroups for the first time: naive B cells (NBs), germinal center B cells (GCBs), memory B cells (MBs), and plasma cells (PCs) and analyzed each subgroup separately. Pseudotemporal trajectory analysis revealed that NBs and GCBs were at the early stage of B cell differentiation, while MBs and PCs were at the end stage. Cellular communication revealed that GCBs acted on tumor cells through the CD99 and SEMA4 signaling pathways. By employing univariate Cox regression, Lasso, and multivariate Cox regression analysis, we developed several risk scores based on marker genes of GCBs subgroups, especially the independent prognostic value of MEF2B⁺ GCB score was validated. The results of immune cell infiltration, TMB, and drug sensitivity assays were significantly different in HNSCC samples from high and low- MEF2B⁺ GCB score groups. Conclusions This study provides ideas to unravel the fundamental biological functions of B cells and their complex mechanistic roles. For the first time, we identified GCBs as B cell-specific prognostic biomarkers. The MEF2B⁺ GCB score fills the research gap in the genetic prognostic prediction model of HNSCC and is expected to provide a theoretical basis for finding new therapeutic targets for HNSCC.
Article
Tumor hypoxia resulting from abnormal and dysfunctional tumor vascular network poses a substantial obstacle to immunotherapy. In fact, hypoxia creates an immunosuppressive tumor microenvironment (TME) through promoting angiogenesis, metabolic reprogramming, extracellular matrix remodeling, epithelial-mesenchymal transition (EMT), p53 inactivation, and immune evasion. Vascular normalization, a strategy aimed at restoring the structure and function of tumor blood vessels, has been shown to improve oxygen delivery and reverse hypoxia-induced signaling pathways, thus alleviates hypoxia and potentiates cancer immunotherapy. In this review, we discuss the mechanisms of tumor tissue hypoxia and its impacts on immune cells and cancer immunotherapy, as well as the approaches to induce tumor vascular normalization. We also summarize the evidence supporting the use of vascular normalization in combination with cancer immunotherapy, and highlight the challenges and future directions of this overlooked important field. By targeting the fundamental problem of tumor hypoxia, vascular normalization proposes a promising strategy to enhance the efficacy of cancer immunotherapy and improve clinical outcomes for cancer patients.
Article
Perineural invasion and neurogenesis are frequently observed in pancreatic ductal adenocarcinoma (PDAC), and they are associated with a poor prognosis. Axon guidance factor semaphorin 3A (SEMA3A) is upregulated in PDAC. However, it remains unclear whether cancer-derived SEMA3A influences nerve innervation and pancreatic tumorigenesis. In silico analyses were performed using PROGgene and NetworkAnalyst to clarify the importance of SEMA3A and its receptors, plexin A1 (PLXNA1) and neuropilin 2 (NRP2), in pancreatic cancer. In vitro assays, including migration, neurite outgrowth, and 3D recruitment, were performed to study the effects of SEMA3A on neuronal behaviors. Additionally, an orthotopic animal study using C57BL/6 mice was performed to validate the in vitro findings. Expression of SEMA3A and its receptors predicted worse prognosis for PDAC. Cancer-derived SEMA3A promoted neural migration, neurite outgrowth, and neural recruitment. Furthermore, SEMA3A-induced effects depended on PLXNA1, NRP2, and MAPK activation. Trametinib, an approved MAPK kinase (MEK) inhibitor, counteracted SEMA3A-enhanced neuronal activity in vitro. Inhibition of SEMA3A by shRNA in pancreatic cancer cells resulted in decreased neural recruitment, tumor growth, and dissemination in vivo. Our results suggested that cancer-secreted SEMA3A plays an important role in promoting neo-neurogenesis and progression of PDAC.
Article
Semaphorins were originally identified as guidance molecules in neural development. However, accumulating evidence indicates that 'immune semaphorins' are critically involved in regulating immune cell activation, differentiation, mobility and migration. Semaphorins are also intimately associated with the pathogenesis of allergic diseases including asthma, allergic rhinitis, atopic dermatitis, allergic conjunctivitis, and eosinophilic chronic rhinosinusitis. Interestingly, reflecting their function in positive or negative regulation of immune cells, levels of some semaphorins are increased while others are decreased in patients with allergic diseases. This review presents the pathogenic functions of immune semaphorins in allergic inflammation and discusses the potential use of these molecules as therapeutic targets for allergic diseases.
Article
Prostate cancer (PCa) is among the most commonly diagnosed solid cancers in male adults. However, most anti-angiogenic therapies and immunotherapies fail to achieve durable remission in advanced PCa. Integrative analysis indicated that Sema3A was negatively correlated with the pathological malignancy and was involved in angiogenesis, cell adhesion, and immune infiltrates in PCa. Sema3A significantly inhibited vascular endothelial growth factor (VEGFA)-induced colony formation, cell proliferation, and PD-L1 expression in PCa cells. Network pharmacological analysis demonstrated that evodiamine, a natural alkaloid compound derived from Evodiae fructus fruits, might regulate Sema3A, lipid metabolism, and monocarboxylic acid transport signaling of PCa. Evodiamine evidently inhibited PCa cell viability in a time-dose-dependent manner. Furthermore, evodiamine impaired angiogenesis by increasing Sema3A expression, and induced ferroptosis by reducing glutathione peroxidase 4 (GPX4) expression, which could be reversed by the ferroptosis blocker ferrostatin-1. Lactate treatment increased hypoxia-inducible factor (HIF)-1α and PD-L1 expressions while restricting Sema3A expression in PCa cells, which could be reversed by silencing monocarboxylate transporter 4 (MCT4) expression. Moreover, evodiamine markedly blocked lactate-induced angiogenesis by restricting histone lactylation and expression of HIF1A in PCa cells, further enhancing Sema3A transcription while inhibiting that of PD-L1. In vivo, evodiamine remarkably inhibited PCa xenograft growth in nude mice, repressing expressions of HIF1α, H3K18la, GPX4, PD-L1, and proliferation, while hindering angiogenesis by increasing Sema3A expression. Therefore, Sema3A represents an essential antineoplastic biomarker, while evodiamine may act as a metabolic-epigenetic modulator, as well as a promising agent in either PCa anti-angiogenic therapy or immunotherapy.
Article
Full-text available
The successful introduction of rationally targeted agents into standard cancer care is a testimony of the vast knowledge base in tumor biology. However, in order to provide individually tailored therapy to patients and to identify small subsets of patients with a high likelihood to benefit from treatment, the identification of biomarkers for response or resistance to a particular therapeutic regimen is imperative. Herein, by the use of a genetically engineered mouse model of pancreatic neuroendocrine tumors, we have assessed the utility of pericyte characteristics in terms of differential marker expression to serve as surrogate markers for response or evasive resistance to anti-angiogenic therapy. We found that tumors refractory to therapy following long-term treatment with a vascular endothelial growth factor receptor-2 blocking antibody contained blood vessels with a prolific investment of pericytes expressing α-smooth muscle actin. Further analysis by simultaneous immunostaining for different pericyte markers led to the conclusion that the increased abundance of this particular subtype of blood vessels most likely occurred by co-option of vessels from the surrounding exocrine pancreas. Our findings may form the basis for retrospective analysis of pancreatic neuroendocrine tumors from patients having undergone treatment with anti-angiogenic agents in order to validate the occurrence of pericytes expressing α-smooth muscle actin as a biomarker for tumors refractory to therapy.
Article
Full-text available
Previously we observed that neural cell adhesion molecule (NCAM) deficiency in beta tumor cells facilitates metastasis into distant organs and local lymph nodes. Here, we show that NCAM-deficient beta cell tumors grew leaky blood vessels with perturbed pericyte-endothelial cell-cell interactions and deficient perivascular deposition of ECM components. Conversely, tumor cell expression of NCAM in a fibrosarcoma model (T241) improved pericyte recruitment and increased perivascular deposition of ECM molecules. Together, these findings suggest that NCAM may limit tumor cell metastasis by stabilizing the microvessel wall. To directly address whether pericyte dysfunction increases the metastatic potential of solid tumors, we studied beta cell tumorigenesis in primary pericyte-deficient Pdgfb(ret/ret) mice. This resulted in beta tumor cell metastases in distant organs and local lymph nodes, demonstrating a role for pericytes in limiting tumor cell metastasis. These data support a new model for how tumor cells trigger metastasis by perturbing pericyte-endothelial cell-cell interactions.
Article
Full-text available
Development of malignant tumours is in part characterized by the ability of a tumour cell to overcome cell-cell adhesion and to invade surrounding tissue. E-cadherin is the main adhesion molecule of epithelia, and it has been implicated in carcinogenesis because it is frequently lost in human epithelial cancers. Re-establishing the functional cadherin complex in tumour cell lines results in a reversion from an invasive to a benign epithelial phenotype. However, it remained unresolved whether the loss of E-cadherin-mediated cell adhesion was a cause or a consequence of tumour progression in vivo. Here we report that the loss of E-cadherin expression coincides with the transition from well differentiated adenoma to invasive carcinoma in a transgenic mouse model of pancreatic beta-cell carcinogenesis (Rip1Tag2). Intercrossing Rip1Tag2 mice with transgenic mice that maintain E-cadherin expression in beta-tumour cells results in arrest of tumour development at the adenoma stage, whereas expression of a dominant-negative form of E-cadherin induces early invasion and metastasis. The results demonstrate that loss of E-cadherin-mediated cell adhesion is one rate-limiting step in the progression from adenoma to carcinoma.
Article
Full-text available
The multitargeted tyrosine kinase inhibitor sunitinib has shown activity against pancreatic neuroendocrine tumors in preclinical models and phase 1 and 2 trials. We conducted a multinational, randomized, double-blind, placebo-controlled phase 3 trial of sunitinib in patients with advanced, well-differentiated pancreatic neuroendocrine tumors. All patients had Response Evaluation Criteria in Solid Tumors-defined disease progression documented within 12 months before baseline. A total of 171 patients were randomly assigned (in a 1:1 ratio) to receive best supportive care with either sunitinib at a dose of 37.5 mg per day or placebo. The primary end point was progression-free survival; secondary end points included the objective response rate, overall survival, and safety. The study was discontinued early, after the independent data and safety monitoring committee observed more serious adverse events and deaths in the placebo group as well as a difference in progression-free survival favoring sunitinib. Median progression-free survival was 11.4 months in the sunitinib group as compared with 5.5 months in the placebo group (hazard ratio for progression or death, 0.42; 95% confidence interval [CI], 0.26 to 0.66; P<0.001). A Cox proportional-hazards analysis of progression-free survival according to baseline characteristics favored sunitinib in all subgroups studied. The objective response rate was 9.3% in the sunitinib group versus 0% in the placebo group. At the data cutoff point, 9 deaths were reported in the sunitinib group (10%) versus 21 deaths in the placebo group (25%) (hazard ratio for death, 0.41; 95% CI, 0.19 to 0.89; P=0.02). The most frequent adverse events in the sunitinib group were diarrhea, nausea, vomiting, asthenia, and fatigue. Continuous daily administration of sunitinib at a dose of 37.5 mg improved progression-free survival, overall survival, and the objective response rate as compared with placebo among patients with advanced pancreatic neuroendocrine tumors. (Funded by Pfizer; ClinicalTrials.gov number, NCT00428597.).
Article
A mouse model involving the human papillomavirus type-16 oncogenes develops cervical cancers by lesional stages analogous to those in humans. In this study the angiogenic-phenotype was characterized, revealing intense angiogenesis in high-grade cervical intraepithelial neoplasias (CIN-3) and carcinomas. MMP-9, a proangiogenic protease implicated in mobilization of VEGF, appeared in the stroma concomitant with the angiogenic switch, expressed by infiltrating macrophages, similar to what has been observed in humans. Preclinical trials sought to target MMP-9 and angiogenesis with a prototypical MMP inhibitor and with a bisphosphonate, zoledronic acid (ZA), revealing both to be antiangiogenic, producing effects comparable to a Mmp9 gene KO in impairing angiogenic switching, progression of premalignant lesions, and tumor growth. ZA therapy increased neoplastic epithelial and endothelial cell apoptosis without affecting hyperproliferation, indicating that ZA was not antimitotic. The analyses implicated cellular and molecular targets of ZA's actions: ZA suppressed MMP-9 expression by infiltrating macrophages and inhibited metalloprotease activity, reducing association of VEGF with its receptor on angiogenic endothelial cells. Given its track record in clinical use with limited toxicity, ZA holds promise as an "unconventional" MMP-9 inhibitor for antiangiogenic therapy of cervical cancer and potentially for additional cancers and other diseases where MMP-9 expression by infiltrating macrophages is evident.
Article
New vessel formation (angiogenesis) is an essential physiological process for embryologic development, normal growth, and tissue repair. Angiogenesis is tightly regulated at the molecular level. Dysregulation of angiogenesis occurs in various pathologies and is one of the hallmarks of cancer. The imbalance of pro- and anti-angiogenic signaling within tumors creates an abnormal vascular network that is characterized by dilated, tortuous, and hyperpermeable vessels. The physiological consequences of these vascular abnormalities include temporal and spatial heterogeneity in tumor blood flow and oxygenation and increased tumor interstitial fluid pressure. These abnormalities and the resultant microenvironment fuel tumor progression, and also lead to a reduction in the efficacy of chemotherapy, radiotherapy, and immunotherapy. With the discovery of vascular endothelial growth factor (VEGF) as a major driver of tumor angiogenesis, efforts have focused on novel therapeutics aimed at inhibiting VEGF activity, with the goal of regressing tumors by starvation. Unfortunately, clinical trials of anti-VEGF monotherapy in patients with solid tumors have been largely negative. Intriguingly, the combination of anti-VEGF therapy with conventional chemotherapy has improved survival in cancer patients compared with chemotherapy alone. These seemingly paradoxical results could be explained by a "normalization" of the tumor vasculature by anti-VEGF therapy. Preclinical studies have shown that anti-VEGF therapy changes tumor vasculature towards a more "mature" or "normal" phenotype. This "vascular normalization" is characterized by attenuation of hyperpermeability, increased vascular pericyte coverage, a more normal basement membrane, and a resultant reduction in tumor hypoxia and interstitial fluid pressure. These in turn can lead to an improvement in the metabolic profile of the tumor microenvironment, the delivery and efficacy of exogenously administered therapeutics, the efficacy of radiotherapy and of effector immune cells, and a reduction in number of metastatic cells shed by tumors into circulation in mice. These findings are consistent with data from clinical trials of anti-VEGF agents in patients with various solid tumors. More recently, genetic and pharmacological approaches have begun to unravel some other key regulators of vascular normalization such as proteins that regulate tissue oxygen sensing (PHD2) and vessel maturation (PDGFRβ, RGS5, Ang1/2, TGF-β). Here, we review the pathophysiology of tumor angiogenesis, the molecular underpinnings and functional consequences of vascular normalization, and the implications for treatment of cancer and nonmalignant diseases.
Article
Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signalling pathways are affording demonstrable therapeutic efficacy in mouse models of cancer and in an increasing number of human cancers. However, in both preclinical and clinical settings, the benefits are at best transitory and are followed by a restoration of tumour growth and progression. Emerging data support a proposition that two modes of unconventional resistance underlie such results: evasive resistance, an adaptation to circumvent the specific angiogenic blockade; and intrinsic or pre-existing indifference. Multiple mechanisms can be invoked in different tumour contexts to manifest both evasive and intrinsic resistance, motivating assessment of their prevalence and importance and in turn the design of pharmacological strategies that confer enduring anti-angiogenic therapies.
Article
Despite having an abundant number of vessels, tumours are usually hypoxic and nutrient-deprived because their vessels malfunction. Such abnormal milieu can fuel disease progression and resistance to treatment. Traditional anti-angiogenesis strategies attempt to reduce the tumour vascular supply, but their success is restricted by insufficient efficacy or development of resistance. Preclinical and initial clinical evidence reveal that normalization of the vascular abnormalities is emerging as a complementary therapeutic paradigm for cancer and other vascular disorders, which affect more than half a billion people worldwide. Here, we discuss the mechanisms, benefits, limitations and possible clinical translation of vessel normalization for cancer and other angiogenic disorders.
Article
Blood vessels deliver oxygen and nutrients to every part of the body, but also nourish diseases such as cancer. Over the past decade, our understanding of the molecular mechanisms of angiogenesis (blood vessel growth) has increased at an explosive rate and has led to the approval of anti-angiogenic drugs for cancer and eye diseases. So far, hundreds of thousands of patients have benefited from blockers of the angiogenic protein vascular endothelial growth factor, but limited efficacy and resistance remain outstanding problems. Recent preclinical and clinical studies have shown new molecular targets and principles, which may provide avenues for improving the therapeutic benefit from anti-angiogenic strategies.