ArticlePDF AvailableLiterature Review

Angiogenesis in Lymph Nodes Is a Critical Regulator of Immune Response and Lymphoma Growth

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the formation of blood vessels, which goes beyond the regulation of metabolism, and shaping a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-angiogenesis, hematopoietic malignancies usually grow within pre-vascularized autochthonous niches in secondary lymphatic organs or the bone marrow. The mechanisms of vascular remodeling in expanding LNs during infection-induced responses have been studied in more detail; in contrast, insights into the conditions of lymphoma growth and lodging remain enigmatic. Based on previous murine studies and clinical trials in human, we conclude that there is not a universal LN-specific angiogenic program applicable. Instead, signaling pathways that are tightly connected to autochthonous and infiltrating cell types contribute variably to LN vascular expansion. Inflammation related angiogenesis within LNs relies on dendritic cell derived pro-inflammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A) expression in fibroblastic reticular cells, which in turn triggers vessel growth. In high-grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical inflammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The dissemination routes of neoplastic lymphocytes are usually disease stage dependent. Early seeding via the blood stream requires the expression of the homeostatic chemokine receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV route is not only relevant for lymphoma cell homing, but also for a continuous immunosurveillance. We envision that HEV functional and structural alterations during lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell accessibility when targeted by T cell–mediated immunotherapies.
Lymph node vascularization in development and under homeostatic conditions. The LN compartments during LN development (left) and homeostatic conditions (right). Left: Lymphoid organogenesis is driven by recruitment of Lymphoid tissue-inducer (LTi) cells that stimulate lymphoid-organizer (LTo) cells via lymphotoxin (LT) α1β2 - LT β receptor signaling, which secrete LTi-recruiting CXCL13 in turn. LTi recruitment from the blood circulation and the afferent lymphatics accumulates LTi cells within the LN anlagen resulting in a self-amplifying process of LN development. α4β7 integrin-expressing LTi recruitment and extravasation utilizes the mucosal vascular addressin cell adhesion molecule-1 (MadCAM-1) on the luminal surface of blood vessels. MadCAM-1 switches to peripheral node addressin (PNAd) expression during differentiation of mature high endothelial venules (HEVs) within peripheral LNs. The formation of the blood vessel network comprises sprouting and branching of expanding blood vessels driven by retinoic acid (RA) stimulation of the RA receptor (RAR) on blood endothelial cells (BECs). Right: The blood circulation enters the LN during homeostatic conditions via the feeding arteriole at the LN hilum, proceeds along the medullary cord and branches into metarterioles that feed the capillary networks around the medulla and at the subcapsular sinus. HEVs are post-capillary venules with a characteristically enlarged vessel diameter. The venous backflow leaves the LN in a bundle of venules at the hilum. Bottom: Representative histochemistry sections (vessels: Cadherin5 fluoresent_reporter, red) of murine LNs during homeostasis and during progression of a murine high-grade B cell lymphoma.
… 
Lymphoma induced angiogenesis in LNs and participating immune cells. Top: The LN compartments represented under homeostatic conditions (left) and lymphoma-activated angiogenesis (right). Lymphoma growth is characterized by a strong LN volume expansion and blood vasculature growth. Remodeling of the stromal infrastructure involves an increase of the microvessel density (MVD), as effectuated by direct angiogenic stimulation through lymphoma B cells cells, but concomitantly also through reciprocal crosstalk of cells in the TME and recruited immune cells. Notably, the initiation of the angiogenic switch in lymphoma is independent from hypoxia-induced HIF1α pathway activation. Tumor polarized DCs (CEBP/βhigh) control the HEV differentiation status via LTα1β2 and LIGHT presentation; they release IL-1β and hereby take part in the blood vessel growth by inducing VEGF-A expression in FRCs. They also secrete the angiogenic factors VEGF-A and FGF2. B cells express LTα1β2, which exerts minor effects on HEVs, but a predominating stimulatory effect on FRCs. Expression of the chemokines CCL2, CXCL12, and MIF recruits additional immune cells into the LN. Regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), M2-polarized macrophages, neutrophils and mast cells are capable of producing the pro-angiogenic factors VEGF-A, VEGF-B, VEGF-C, MMP9, IL-8, IL-10, TGFβ, and FGF1/2. Bottom left: HEVs express PNAd, CCL21, and ICAM1 and thereby constitute the transmigration routes for lymphocytes under homeostatic conditions. Interaction of CD62L, CCR7, and LFA-1 on naïve lymphocytes with these HEV-associated surface receptors and chemokines initiates lymphocyte rolling, HEV wall adhesion and eventually, transmigration into the LN parenchyma. Bottom middle: Inflammatory vessels in reactive LNs recruit activated lymphocytes by CXCL9 secretion and replace the homeostatic receptors on endothelial cells with CD62P, CD63E, and VCAM1 that are interaction partners of leukocyte-expressed CD44, PSGL1, and VLA4. Bottom right: The lymphoma induced expansion of the blood vessel network favors the assembly of smaller anergic endothelium that is insufficiently equipped for lymphocyte extravasation.
… 
This content is subject to copyright.
Angiogenesis in Lymph Nodes Is a
Critical Regulator of Immune
Response and Lymphoma Growth
Lutz Menzel
1
*, Uta E. Höpken
2
and Armin Rehm
1
*
1
Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany,
2
Microenvironmental
Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the
formation of blood vessels, which goes beyond the regulation of metabolism, and shaping
a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-
angiogenesis, hematopoietic malignancies usually grow within pre-vascularized
autochthonous niches in secondary lymphatic organs or the bone marrow. The
mechanisms of vascular remodeling in expanding LNs during infection-induced
responses have been studied in more detail; in contrast, insights into the conditions of
lymphoma growth and lodging remain enigmatic. Based on previous murine studies and
clinical trials in human, we conclude that there is not a universal LN-specic angiogenic
program applicable. Instead, signaling pathways that are tightly connected to
autochthonous and inltrating cell types contribute variably to LN vascular expansion.
Inammation related angiogenesis within LNs relies on dendritic cell derived pro-
inammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A)
expression in broblastic reticular cells, which in turn triggers vessel growth. In high-
grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells
immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In
contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical
inammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are
unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The
dissemination routes of neoplastic lymphocytes are usually disease stage dependent.
Early seeding via the blood stream requires the expression of the homeostatic chemokine
receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and
also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV
route is not only relevant for lymphoma cell homing, but also for a continuous
immunosurveillance. We envision that HEV functional and structural alterations during
lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell
accessibility when targeted by T cellmediated immunotherapies.
Keywords: lymphoma, B cell malignancy, angiogenesis, lymph node, tumor microenvironment, reactive
endothelium, lymphocyte trafcking, high endothelial venule
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917411
Edited by:
Martina Seiffert,
German Cancer Research Center
(DKFZ), Germany
Reviewed by:
Tanja Nicole Hartmann,
University of Freiburg Medical Center,
Germany
Patricia Perez Galan,
Institut de Recerca Biomèdica August
Pi i Sunyer (IDIBAPS), Spain
*Correspondence:
Lutz Menzel
lutz.menzel@mdc-berlin.de
Armin Rehm
arehm@mdc-berlin.de
Specialty section:
This article was submitted to
Cancer Immunity
and Immunotherapy,
a section of the journal
Frontiers in Immunology
Received: 05 August 2020
Accepted: 19 October 2020
Published: 03 December 2020
Citation:
Menzel L, Höpken UE and Rehm A
(2020) Angiogenesis in Lymph Nodes
Is a Critical Regulator of Immune
Response and Lymphoma Growth.
Front. Immunol. 11:591741.
doi: 10.3389/fimmu.2020.591741
REVIEW
published: 03 December 2020
doi: 10.3389/fimmu.2020.591741
INTRODUCTION
Lymph nodes (LNs) are strategically positioned hubs of the immune
system, connecting the lymphatic system with the blood circulation,
ltering antigens and organizing the encounter of lymphocytes with
antigen presenting cells (APCs). The LN parenchyma is tightly
packed with numerous types of immune cells and susceptible for
their immigration and release during conditions of homeostasis,
inammation and tumor transformation. The complex reciprocal
interactions of stromal cells and immune cells in LNs shape an
adapted microenvironment that supports angiogenesis and
increased LN vascularization (1). Although numerous studies
reported vascular remodeling and expansion in LNs upon
pathogen or tumor cell encounter, the detailed mechanisms and
the participating cells of these angiogenic processes are not yet
identied. In this review, we delineate the current state of knowledge
and propose probable cellular interactions that participate in
vascular growth in LNs. In particular, we will focus on the
intricate relationship between immune cells and vascular cells as a
majorpillarofthetumormicroenvironment(TME).
B cell non-Hodgkin lymphoma (B-NHL) is a heterogenous group
of hematological malignancies that arise from B lymphocytes at
various stages of differentiation. Lymphomas grow in the bone
marrow and in the secondary lymphatic organs (SLOs), with a
predominance of LNs and spleen, but they can also manifest in
non-lymphoid tissues (2). The genetic and epigenetic alterations and
the intracellular pathway dysregulations responsible for the
pathogenesis and progression of lymphomas have been extensively
studied and led to tremendous advancements in therapeutic
intervention strategies (3). The idea of tumor dependency on cells
in the surrounding of a a priori benign environment and on adapted
organ properties goes back to Rudolph Virchow in the 19
th
century
(4). The crucial inuence of the cellular context in which lymphoma
cells arise and lodge attracts growing interest, and the investigation of
the TME became an increasingly appreciated eld in cancer research
(5,6). The TME constitutes about half of the tumor mass in indolent
follicular lymphoma (FL) and marginal zone lymphoma (MCL),
whereas the proportion in aggressive diffuse large B cell lymphoma
(DLBCL) is generally lower and scarce in Burkittslymphoma(BL)
(7). On the extreme, in classical Hodgkin lymphoma (cHL) only
about 2%3% of the cells comprise the malignant Hodgkin-Reed-
Sternberg cells (8).Hence, the composition and the dependency of the
different B-NHL and cHL on the TME differ substantially between
the entities (7). What distinguishes solid tumors and their metastasis
most from lymphoma is that within SLOs, transformed B cells
encounter a TME infrastructure that genuinely supports survival of
benign B cells. These tissues undergo renement to the needs of the
tumor cells induced by a continuous reciprocal crosstalk of tumor,
immune and mesenchymal stromal cells (9).
The complex interactions of transformed B cells and the TME lead
to extensive changes of the vasculature within the affected organs,
which is considered to have a substantial prognostic impact on the
patientsdisease outcome (7,10). The stromal compartment, mainly
comprised of blood vessels, lymphatic sinuses and the broblastic
reticular network is tightly interconnected and regulated. In some
respect, it can be considered to represent a joint structural
compartment in which its distinct subcompartments grow and
remodel in a synchronized manner (11,12). While the crucial role
of lymphatic vessels during lymphoma growth and dissemination is
undisputed (13,14), here we will highlight the inuence of blood
endothelial cells (BECs) and the blood vasculature, which comprise
the main provider of nutrition for proliferating and differentiating
immune and tumor cells. In addition, the blood vasculature shapes a
major dissemination route for benign immune and transformed cells
(15,16).
EXPANSION OF BLOOD VASCULATURE
IN LNs DURING DEVELOPMENT,
INFLAMMATION, AND CANCER
Tumors often recapitulate developmental traits of tissues in which
they arise. The stem cell-like phenotype of many tumors is
characterized by gene expression signatures that are associated with
embryonic stem cell identity and underlines the close transcriptional
relationship between neoplastic and developmental tissue (17,18).
Similar to rapidly developing and growing organs, tumors require
blood vessels to access oxygen and nutrients. The initiation of blood
vessel expansion, referred to as angiogenic switch, occurs at different
stages during tumorigenesis, depending on the tumor type and the
respective TME. The onset of neo-vascularization and vascular
remodeling is a multifactorial processes orchestrated by activating
and inhibiting factors whose balance determines whether BECs stay
quiescent or get activated (19).
Therefore, it is useful to recapitulate the essential steps during
development to understand the basal mechanisms of the
microenvironmental remodeling in LNs. Blood vessels in LNs
reside within the stromal scaffold and are crucial for the delivery of
oxygen, nutrients, and cells. The critical delivery function was
demonstrated by the rapid occurrence of necrosis in LNs upon
ablation of the arterial feeding vessel in rats (20). During
development (Figure 1), LNs evolve from budding lymphatic veins
that form a primordial lymph sac, also known as LN anlagen. Studies
with transgenic mice lacking lymphatic vessels due to the deciency
for the transcription factor (TF) Prox1 or appropriate
lymphangiogenesis factors, e.g., vascular endothelial growth factor-
c(Vegfc
+/
), revealed a compromised LN development (21,22). LN
anlagen recruit hematopoietic lymphoid tissue-inducer (LTi) cells,
which in turn stimulate local mesenchymal cell differentiation into
lymphoid tissue-organizer (LTo) cells. The accumulation and
interaction of lymphotoxin (LT) a
1
b
2
on LTi cells and LTb
receptor (LTbR) expressing LTo cells results in a self-amplifying
loop of LTi recruitment and LTo differentiation that drives the LN
development (23). The lymphoid organogenesis is accompanied by
the maturation of blood vasculature driven by locally generated
retinoic acid (RA) (24). RA is presumably provided by neurons
localized adjacent to the developing LN. It directly regulates the
proliferation of endothelial cells, but also induces CXCL13 expression
in LTo cells via binding to the RA receptor-related orphan receptor
(RORgt). CXCL13 in cooperation with its receptor CXCR5 is the
exclusive inducer of the initial clustering of LTi cells in LN anlagen
independently of LT-LTbR signaling (25). A ubiquitous expression of
the mucosal addressin cell adhesion molecule-1 (MadCAM-1) on
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917412
developing venous blood vessels in the LN mediates the directed
immigration of the a4b7 integrin expressing LTi cells (2628).
Notably, the expression of MadCAM-1 in peripheral LNs of
newborns switches during the formation and maturation of high
endothelial venules (HEVs) into the expression of the peripheral
node addressin (PNAd). PNAd expression marks the completion of
the maturation of the postcappillary vessels to highly differentiated
HEVs that provide all prerequisites for the functional transmigration
of blood-borne lymphocytes into the developing and homeostatic LN
(Figure 1)(26).
The vascular system of LNs in adult mammals is composed of
arteries, capillaries, post-capillary venules and veins (29).
Arteries are characteristically located at the periphery of the
LN. The feeding arteriole enters the LN at the hilum and exhibits
a gradual decrease in diameter at its few branching points
alongside the medullary cords until it reaches the subcapsular
capillary network. Capillaries form a dense network under the
subcapsular sinus and around the medullary cords, whereas they
are markedly less dense in cortex regions and sparse within the
paracortex under homeostatic conditions. The vessel diameter
abruptly increases at the capillary to post-capillary transitions.
These post-capillary venules, referred to as HEVs, are primarily
located within the cortex in the interfollicular space. HEVs form
loop-like structures following a centripetal course that ends in
FIGURE 1 | Lymph node vascularization in development and under homeostatic conditions. The LN compartments during LN development (left) and homeostatic
conditions (right). Left: Lymphoid organogenesis is driven by recruitment of Lymphoid tissue-inducer (LTi) cells that stimulate lymphoid-organizer (LTo) cells via
lymphotoxin (LT) a
1
b
2
-LTbreceptor signaling, which secrete LTi-recruiting CXCL13 in turn. LTi recruitment from the blood circulation and the afferent lymphatics
accumulates LTi cells within the LN anlagen resulting in a self-amplifying process of LN development. a
4
b
7
integrin-expressing LTi recruitment and extravasation
utilizes the mucosal vascular addressin cell adhesion molecule-1 (MadCAM-1) on the luminal surface of blood vessels. MadCAM-1 switches to peripheral node
addressin (PNAd) expression during differentiation of mature high endothelial venules (HEVs) within peripheral LNs. The formation of the blood vessel network
comprises sprouting and branching of expanding blood vessels driven by retinoic acid (RA) stimulation of the RA receptor (RAR) on blood endothelial cells (BECs).
Right: The blood circulation enters the LN during homeostatic conditions via the feeding arteriole at the LN hilum, proceeds along the medullary cord and branches
into metarterioles that feed the capillary networks around the medulla and at the subcapsular sinus. HEVs are post-capillary venules with a characteristically enlarged
vessel diameter. The venous backow leaves the LN in a bundle of venules at the hilum. Bottom: Representative histochemistry sections (vessels:
Cadherin5
uoresent_reporter
, red) of murine LNs during homeostasis and during progression of a murine high-grade B cell lymphoma.
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917413
transitions to veins at the corticomedullary junctions. Finally, a
bundle of larger main veins leave the LN though the hilum
(Figure 1)(29,30).
Tumor growth is often accompanied by the ingrowth of blood
vessels and the formation of a vascular network, consistent with the
need for malignant cells to have access to the circulation system.
Tumor vascularization occurs either through co-option of the pre-
existing vasculature, or by induction of neovascularization. Vessel
co-option is a non-angiogenic process in which tumor cells utilize
pre-existing blood vessels of surrounding tissue to support tumor
growth, survival and metastasis (31). In contrast,
neovascularization involves a series of complex and sequential
events: (I) activation of microvascular endothelial cells, (II)
enzymatic degradation of the vascular basal membrane, (III)
gradual degradation of other extracellular matrix (ECM)
components, (IV) endothelial cell migration and proliferation,
(V) lumen formation within neo-sprouts, (VI) branching of the
neo-vessel, and (VII) formation of a functional vessel network by
fusion with neighboring vessels to initiate blood ow (32,33).
Located at the leading edge of the vascular sprout, tip cells form
cellular protrusions or lopodia to guide migration toward a source
of angiogenic growth factors. Simultaneously, they signal to
adjacent endothelial cells via Delta-like ligand (DLL)-Notch
interactions not to adapt the tip cell phenotype, but to maintain
the proliferative stalk cell phenotype and toform a vascular lumen
(34,35). The vascular endothelial growth factors (VEGFs) are the
major contributors to angiogenesis. The local secretion of VEGF-A
andits gradientformingdepositionon theECMtriggersendothelial
tip cell formation via binding to VEGFR2, resulting in endothelial
cell proliferation and migration and eventually, formation of tube
structures resembling new capillaries (3538). VEGF-B, VEGF-C,
and VEGF-D are other members of the VEGF family of which
VEGF-C plays a critical role upon LN remodeling because it is the
most potent inducer of lymphangiogenesis as a ligand of VEGFR3.
VEGFR3 is known for its involvement in physiological and tumor-
associated lymphangiogenesis and lymphatic metastasis (39,40).
Apart from lymphatics, VEGFR3 is highly expressed at the leading-
edge of BECs that undergo sprouting (41) and was recently shown
to coregulate the expansion of the blood vessel network in LNs in a
Myc-driven high-grade B cell lymphoma mouse model (42).
Fibroblast growth factors (FGFs) stimulate endothelial cell
migration and proliferation in a very potent manner, which in in
vitro experiments evenexceeds the stimulation capacity of VEGF-A
(43,44). FGF-1 stimulates proliferation anddifferentiation of all cell
types necessary for the formation of arterial vessels, including
endothelial and smooth muscle cells. The angiogenic potency of
FGFs extends to prompt broblastic cells (e.g., pericytes, smooth
muscle cells, and mural cells) and recruits them for vesselformation
and maturation during tumorigenesis(45). FGF-2, the second most
abundant growth factor of the FGF family, promotes endothelial
cell proliferation and the physical organization of the endothelial
cell tube-like formation during developmental vessel assembly
(46,47).
The integral investigation of the highly complex vascular
network and the unique features of its parts in context of the
compartmentalized architecture of theLN has long been a challenge
for microscopic image analysis. Because higher order anatomical
data sets were obtained from such advanced optical imaging
approaches, algorithms for data handling were also demanding to
generate. Over the last couple of years, novel tissue preparation
methods (48,49), imaging systems and computational rendering
strategies evolved, which enable contextual and organ-wide
topological analyses in three-dimensional spaces and over time.
In particular, optical projection tomography (OPT) and light sheet
microscopy have been established to study anatomical and
functional features of LN, e.g., to quantify capillary and HEV
structures and their contextual relationship to B cell follicles and
dendriticcells (DCs) throughout the organ (5052). A combination
of microscopic imaging and computational modulation of the
hydrodynamic properties of vessels in LNs revealed a tight
connection of the hydraulic conductivity between lymphatic and
blood vessels and the respective hydrodynamic conditions within
the LN. These biophysical conditions are vital for inter- and intra-
LN transport mechanisms and immunological functions, and most
likely for lymphoma B cell dissemination and immunosurveillance
as well (53,54). Up to date, these dynamic conditions are not easy to
mimic in organoid models. However, in an early 3D organoid
model mimicking a LN exposed to tissue injury or inammation,
the interstitial ow affected the broblastic reticular cells (FRCs)
that enwrap conduits transporting uid from the subcapsular sinus
to HEVs. Blocking this ow led to CCL21 downregulation,
indicating that increased lymph ow as a hydrodynamic factor
acts on the paracortex and thus, affects the remodeling and
functionality of conduits and FRCs (55). In line, mechanosensing
of conduit ow deprivation by FRCs in Peyers patches resulted in
dysfunctional HEVs and disturbed mucosal immune responses
(56). Similar processes are also conceivable during lymphoma
growth within LNs, where a gradual loss of HEVs in numerous B-
NHL was described many years ago (57). A comprehensive and
continuous blood vessel network of LNs under homeostatic
conditions has been revealed (54,58) and brought up an analysis
pipeline for detailed and whole-organ investigations of the LN
vasculature upon perturbations through inammation, lymphoma
homing and LN solid tumor metastasis. Recently, utilization of
single cell transcriptome analysis methods revealed a broad
overview of the heterogeneity of ECs throughout several different
murine organs, including the spleen and LN as representatives for
SLOs (59,60).
THE BLOOD VASCULATURE
IS PART OF THE REACTIVE
STROMAL INFRASTRUCTURE
DURING INFLAMMATION AND
CANCER DEVELOPMENT
Inammation, vessel reorganization and angiogenesis are
intimately connected processes. In adults, angiogenesis usually
occurs during pathological settings such as infection, wound
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917414
healing and cancer. Notably, hematopoietic cells and endothelial
precursors share common CD34
+
stem and progenitor cells (61).
Growthof solidtumorsis typicallyassociatedwithinammation
that triggers tissue-protective and pro-tumorigenic mechanisms.
Inammatory responses in normal tissue and cancer are initiated
and maintained by local tissue or cancer associated macrophages
(TAMs) and DCs. Sustained inammation further leads to
recruitment of bone marrowderived monocytes, neutrophilic
granulocytes, myeloid-derived suppressor cells (MDSC), and
tissue or tumor inltration of lymphocytes from the SLOs.
Especially cytokines and chemokines, transcriptionally regulated
downstream of NF-kB signaling pathways in immune cells,
promote cell survival and proliferation, recruit more immune
cells and re-shape the TME. Pro-inammatory cytokines like IL-
6, TNFaand IL-17, increase the proliferation rate of other
inammatory immune cells and prime the tumor to overcome
suboptimal microenvironmental conditions including lack of
nutrients, growth factors and hypoxia (62). Inamed tissue and
solid tumors are often characterized by insufcient oxygen supply
that triggers angiogenesis. Hypoxia, which is the major driver of
vascular alterations in solid tumors, stabilizes the TF HIF-1a,the
master regulator of pro-angiogenic factor expression such as
VEGFs, CXCL12, and COX-2 (6365). The presence of a
constant pro-angiogenic milieu in solid tumors often causes a
disturbed maturation and pruning of blood vessels. The division
inarterioles,capillariesandvenulescanbe decientandresultsin an
aberrant distribution of vessel caliber, inuencing the blood ow.
Morphologically, a poorly organized, malformed vessel network
develops under these conditions (66,67). The endothelial junctions
in such malformed networks are often defective and lead to
enhanced permeability and elevated interstitial uid pressure
(68). Pericytes can be partially detached and newly build blood
vessels often fail to recruit sufcient pericyte coverage, causing an
unevenly distributed basement membrane, vessel fragility, and risk
of hemorrhage (69,70). Besides the structural and functional
defects, the specic transcriptional response of tumor vasculature
is not only related to angiogenesis and vessel integrity, but affects
endothelial activation and recruitment of leukocytes as well. Pro-
angiogenic signaling leads to endothelial anergy, reduced response
to pro-inammatory signaling and decreased expression of
adhesion molecules and chemokines necessary for capture and
trans-endothelial migration of leukocytes (71,72).
In LNs, which are the authochthonous environment for most
B-NHL, the pre-existing vasculature takes part in the massive
remodeling process during immune responses, best studied for
strong inammatory stimuli in mice (50,73,74). LNs are plastic
organs able to expand to a multiple of their normal size within
days including an extensive remodeling of the vascular-stromal
compartment. The rapid expansion of the LN size and cellularity
includes early events of remodeling of the feeding artery, causing
an increased blood ow and LN hypertension accompanied by
an increase of the vascular permeability (75,76) and increased
interstitial pressure. The capillary network within the cortex and
medulla expands toward the paracortex, and post-capillary
venules are reorganized (30). Skin allograft-draining LNs in
rats exhibited a progressive elongation and branching of HEVs
resulting from focal proliferation of endothelial cells in the
transition zone from high to low endothelium (77). Several
years later, Bajenoff and colleagues revisited these observations
and investigated the BEC proliferation applying a multicolor
uorescence fate-mapping mouse model. They found similar
proliferation foci in post-capillary venules as proposed by
Anderson and Anderson. In addition, an extensive expansion
of the LN vasculature relying on the sequential assembly of
endothelial cell proliferative units upon inammation was
observed. Clonally proliferating HEV cells (73) and capillary
resident precursors (60) comprised local progenitors for HEV
elongation and capillary neo-vessels during BEC turnover and
vessel sprouting. Interestingly, recruitment of bone marrow
derived endothelial cell progenitors did not contribute to the
local LN vascular alterations in this model. LN expansion
stimulated by several immunization strategies in mouse
experiments, e.g., bone marrowderived DCs (78), ovalbumin/
complete Freunds adjuvant (OVA/CFA) (79), OVA/alum (80),
oxazalone (11), and lymphocytic choriomeningitis virus
(LCMV) infection (50) indicated similar courses of vessel
expansion, starting with early proliferation events that last for
up to 58 days. The remodeling eventually ends with a gradual
re-establishment of the vascular endothelial cell quiescence, a
normalization of the vascular bed and restoration of the normal
LN size (30,73,78).
IMMUNE CELLS ARE MEDIATORS
OF ANGIOGENESIS
Both innate and adaptive immune cells have an intricate
relationship with angiogenesis. They are involved in regulation of
BECproliferation,migrationand activationand theyprovidea large
spectrum of pro-angiogenic mediators apart from their genuine
immunological function. Hence, immune cells induce, support or
antagonize angiogenic processes during inammation and tumor
growth (Figure 2 and Table 1)(124,125). Angiogenesis is also
importantfor the progression of B cell lymphoma, howeverthe role
of angiogenic factors and the composition of pro-angiogenic
immune cells within LNs varies between different entities.
A leading immune cell source for growth factors and chemokines
to promote angiogenesis under inammatory and tumorous
conditions are myeloid cells (126). Macrophages are phagocytic
immune cells and important regulators of tissue homeostasis,
morphogenesis and repair. In LNs, macrophages are an abundant
immune cell population that is divided into subcapsular sinus
macrophages (SSM), medullary sinus macrophages (MSMs), and
medullary cord macrophages (MCMs) (127). Monocytes from the
blood stream and macrophages from LN remote tissues (e.g., bone
marrow)inltrate the LN attracted by a variety of chemotactic factors,
among others CCL2, CXCL12, and the macrophage migration
inhibitory factor (MIF) (128130). Tumor-associated macrophages
(TAMs) play a prominent role during progression of chronic
lymphocytic leukemia (CLL) by supporting tumor cell survival
(131) and regulation of the TME (132). The presence and
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917415
polarization of macrophages during CLL is critical for the tumor
progression, as indicated by a CLL-associated skewing of T cells
toward antigen-experienced phenotypes and T cell exhaustion, which
could be reversed by monocyte and macrophage depletion. Thus,
interference with macrophage polarization in CLL turned out to be a
promising target for immunotherapy (133). Similar to TAMs in
leukemia, macrophages likely support angiogenesis in lymphoma as
well, both in cHL and B-NHL. M2-polarized macrophages induce an
immunosuppressive milieu in cHL, comprising the majority of the
PD-L1 expressing cells, located in close proximity to the Hodgkin
Reed Sternberg (HRS) cells (134). In this disease-dening tumor cell
population, high frequencies of alterations on chromosome 9p24.1,
involving copy number gain and amplications, have been shown to
increase the abundance of the PD-1 ligands, PD-L1 and PD-L2 (135).
Furthermore, Epstein-Barr virus (EBV) infection can increase
expression of PD-1 ligands in cHL as well (136). The TAM derived
PD-L1 in conjunction with the HRS-cell derived PD-1 ligands PD-L1
and PD-L2 may neutralize the anticancer activity of PD-1+ T cells
and natural killer cells, a process that can be reversed by utilizing PD-1
blocking antibodies (137). TAMs were also frequently found in FL
and DLBCL, among them often polarized and pro-angiogenic M2-
like macrophages, which secrete angiogenic factors and re-arrange
the ECM by matrix metalloproteinase (MMP) release for vascular
expansion (138,139).
FIGURE 2 | Lymphoma induced angiogenesis in LNs and participating immune cells. Top: The LN compartments represented under homeostatic conditions (left)
and lymphoma-activated angiogenesis (right). Lymphoma growth is characterized by a strong LN volume expansion and blood vasculature growth. Remodeling of
the stromal infrastructure involves an increase of the microvessel density (MVD), as effectuated by direct angiogenic stimulation through lymphoma B cells cells, but
concomitantly also through reciprocal crosstalk of cells in the TME and recruited immune cells. Notably, the initiation of the angiogenic switch in lymphoma is
independent from hypoxia-induced HIF1apathway activation. Tumor polarized DCs (CEBP/b
high
) control the HEV differentiation status via LTa
1
b
2
and LIGHT
presentation; they release IL-1band hereby take part in the blood vessel growth by inducing VEGF-A expression in FRCs. They also secrete the angiogenic factors
VEGF-A and FGF2. B cells express LTa
1
b
2
, which exerts minor effects on HEVs, but a predominating stimulatory effect on FRCs. Expression of the chemokines
CCL2, CXCL12, and MIF recruits additional immune cells into the LN. Regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), M2-polarized
macrophages, neutrophils and mast cells are capable of producing the pro-angiogenic factors VEGF-A, VEGF-B, VEGF-C, MMP9, IL-8, IL-10, TGFb, and FGF1/2.
Bottom left: HEVs express PNAd, CCL21, and ICAM1 and thereby constitute the transmigration routes for lymphocytes under homeostatic conditions. Interaction of
CD62L, CCR7, and LFA-1 on naïve lymphocytes with these HEV-associated surface receptors and chemokines initiates lymphocyte rolling, HEV wall adhesion and
eventually, transmigration into the LN parenchyma. Bottom middle: Inammatory vessels in reactive LNs recruit activated lymphocytes by CXCL9 secretion and
replace the homeostatic receptors on endothelial cells with CD62P, CD63E, and VCAM1 that are interaction partners of leukocyte-expressed CD44, PSGL1, and
VLA4. Bottom right: The lymphoma induced expansion of the blood vessel network favors the assembly of smaller anergic endothelium that is insufciently equipped
for lymphocyte extravasation.
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917416
Regarding angiogenesis, macrophages play a crucial role at each
step of the angiogenic cascade, starting from blood vessel sprouting
to vessel maturation and remodeling of the vascular network. Pro-
inammatory conditions polarize classical activated macrophages
(M1), whereas anti-inammatory conditions give rise to
alternatively activated macrophages (M2) including angiogenesis
associated macrophages (140142). For example, in cHL HRS-cell
derived TGF-b, IL-13 and M-CSF educate monocytes or TAMs to
become immunosuppressive M2-polarized TAMs (Aldinucci D,
Casagrande N, 2016, Cancer Letters; Skinnidder and Tak Mak,
2002, Blood).
Macrophage-derived MMP2 and MMP9 proteases cleave the
ECM to break open matrix remodeling to pave the way for
endothelial sprout migration (85). Pro-angiogenic growth factors,
such as VEGF-A, MMPs, IL-1b, FGF2, and transforming growth
factor beta 1 (TGFb1), are part of the repertoire secreted by
macrophages in a pro-angiogenic milieu (8589). Upon hypoxic
conditions that lead to HIF1aactivation, macrophages are able to
establish capillary-like networks in which they line a vessel micro-
tunnel and express lineage aberrant endothelial markers such as
CD31, von Willebrand factor and Cadherin-5, leading to the
assumption that macrophages may transdifferentiate into ECs
under specic conditions (143,144). Macrophages also function
as cellular chaperones during anastomosis of vascular sprouts by
guiding endothelial tip cells to undergo sprout fusion (82). Such
macrophages expressed the markers tyrosine kinase with
immunoglobulin-like and EGF-like domains (TIE2) and
neuropilin-1 (Nrp1), indicating that they are M2 polarized cells
TABLE 1 | Immune cells derived pro-angiogenic factors.
Cells Condition Angiogenic Factors Reference
MFInammation (mouse, LPS, LTA/MDP) VEGF-A/C/D (81)
Development (zebra sh) VEGF-A (82)
Inammation (mouse, OVA/CFA) IL-1bvia FRCs (83)
Hypoxia (in vitro) VEGF, bFGF, CXCL8, COX2, HGF, MMP12 (84)
Mouse/chicken angiogenesis model MMP2, MMP9 (85)
Human Monocytes (in vitro) VEGF-A (86)
Mouse Matrigel Assay (in vivo) IL-1b(87)
Mouse Matrigel Assay (in vivo) FGF2, PlGF (88)
Human atherosclerotic plaques VEGF-A (89)
Mouse solid tumors PDGFb(90)
Squamous carcinoma VEGF-C (91)
human cell lines in vitro TP (92)
Ovarian carcinoma MMP9 (93)
Breast carcinoma VEGF-A (94)
DC OVA/CFA inammation (mouse) VEGF-A (95,96)
Inammation (mouse, LPS) PGE
2
(97)
LPS, PGE
2
in vitro (mouse) FGF2 (95)
development and homeostasis (mouse) LTa
1
b
2
(98,99,100)
Inammation (mouse, OVA/CFA) IL-1bvia FRCs (83)
co-culture with NK cells (in vitro) VEGF-C (101)
Il-10 stimulation (in vitro) Osteopontin (102)
NFMouse Matrigel Assay (in vivo) VEGF-A, MMP9 (103)
Human cells, angiogenesis assay (in vitro) VEGF-A, IL-8 (104)
Mouse wound healing assay VEGF-A (105)
MC human skin VEGF-A, IL-8, MCP-1 (106)
(107)
Human lung mast cells (in vitro) MMP9, VEGF-A/B/C/D (108)
Thyroid cancer IL-8 (109)
MDSC Mouse tumor models VEGF-A, G-CSF, MMP9 (110)
Mouse melanoma model VEGF-A (111)
Mouse ovarian cancer model VEGF-A (112)
Multiple myeloma mouse model MMP9 (113)
Colorectal cancer mouse model MMP9 (114)
T cells Inammation (mouse, OVA/Montanide) LTa
1
b
2
via FRCs (115)
HUVECs (in vitro) GM-CSF, IL-8 (116)
Ischemia mouse model and in vitro IL-10, amphiregulin (117,118)
Type 2 Diabetes Amphiregulin, IL-10 (119)
Systemic sclerosis IL-8, MMP9, VEGF-A
induce EPCs diff.
(120,121)
Hypoxia (in vitro, ovarian cancer) VEGF-A (118)
B cells Inammation (mouse, LPS) VEGF-A (122)
Inammation (mouse, OVA/Montanide) LTa
1
b
2
via FRCs (115)
Virus infection (mouse, LCMV) LTa
1
b
2
(50)
In vitro tube formation VEGF-A, FGF2, PDGFA (123)
MF(macrophages), DC (dendritic cells), NF(neutrophils), MC (mast cells), MDSC (myeloid-derived suppressor cells).
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917417
with properties similar to TAMs (82,145). New blood vessels need
to undergo maturation to become functionally stable. A crucial step
in this process is the integration of new blood vessels into the
established stromal environment and the recruitment of pericytes to
strengthen vascular junctions. Macrophages are highly abundant
around new blood vessels and help to recruit pericytes by secretion
of the platelet-derived growth factor b(PDGFb)(90,146).
Dendritic cells (DCs) are sentinel cells that connect the
innate and adaptive branches of the immune system wherein
they have important roles in host defense against pathogens and
in generating anti-tumor immune responses. The classical DC
compartment of the spleen is comprised of lymphoid tissue-
resident DCs, whereas LNs also include non-lymphoid tissue-
migratory DCs (147). Especially CD11c
medium
MHCII
high
DCs are
associated with the initiation of vascular expansion after bone marrow
derived DC (BMDC) transfer, whereas CD11c
high
MHCII
medium
DCs
accumulate later in the process and promote the re-establishment of
vascular quiescence (78). Apart from their predominant immunologic role
as professional APC population, DCs carry a wide range of angiogenic
mediatorstomodulatevascularization.Theydosobyengagingcognate
signaling receptors, such as VEGFR2 on endothelial cells or by recruiting
and stimulating adjacent cells or cells of the TME (96). LN-resident DCs
are closely associated with FRCs and sample conduit-conveyed antigens
within the paracortical and interfollicular zone, where they are located in
the proximity of HEVs (148). This spatial proximity suggests that DCs are
likely to be a link between immune cells, vasculature and mesenchymal
stromal cells. The development, maturation and lineage commitment of
DC subsets is differentially regulated by acomplex TF network, depending
on homeostatic, inammatory, and tumorous conditions. We recently
demonstrated that the TF C/EBPbplays a crucial role in murine DC
maturation and immunogenic functionality under homeostatic and
lymphoma-transformed conditions (149). In the presence of lymphoma
cells, enhanced expression of C/EBPbin DCs was observed which
transformed them into an immature, tolerogenic and pro-tumorigenic
subtype (150). Such aberrant maturation stages may potentially affect the
angiogenic capacities of the DCs as well. The crucial role of DCs for the LN
vasculature was elaborated in several studies (12,78,79,96)andrevealed
the DC-coordinated remodeling mechanisms of blood- and lymph-
vasculature, and the FRC network as well. The DC associated increase
of VEGF-A in reactive LNs further includes stimulation of a pro-
angiogenic program in FRCs and the recruitment of blood-borne cells
that participate in the angiogenic process. IL-1bexpression by recruited
CCR7
+
CD11c
+
DCs is associated with the enrichment of VEGF-A
expressing FRCs at the border of the LN paracortex (83). The angiogenic
role of DCs in lymphoma LNs has not been investigated yet; however, in
reactive LNs, resident classic DCs produce biologically active VEGF-A
downstream of the inammation-associated TFs HIF-1a, STAT3, and
CREB. HIF-1aand STAT3 are generally related to hypoxic conditions,
whereas CREB phosphorylation is the consequence of autocrine and
paracrine prostaglandin E2 (PGE
2
) signaling (151). The PGE
2
production
is directly connected to pathogen induced toll like receptor-4 (TLR4)
signaling and therefore, delineates the connection of infection induced LN
reactivity with angiogenic vessel formation (152). DCs also release other
classical angiogenic growth factors like FGF2, endothelin-1 (ET-1),
CXCL12, and COX-2. FGF2 activates endothelial cells and induces
VEGF-A expression in mesenchymal cells, but also recruits and activates
macrophages and mast cells that in turn exhibit angiogenic
properties (152,153). DCs further have the capacity to modulate
angiogenesis in an indirect manner through secretion of the
monocyte and granulocyte attracting chemokines CXCL8,
CXCL1, CXCL2, CXCL3, and CXCL5 (154). The recruited
myeloid cells can be triggered to secrete the pro-angiogenic IL-1b
by a signaling pathway that includes classical DC-derived
osteopontin (155). DCs are not only associated with vascular
expansion, but also with the re-establishment of vascular
quiescence and stability in the process of reinstallation of the LN
homeostasis (78).
Neutrophilic granulocytes are the most abundant type of
leukocytes throughout the body, representing the pioneering cells
that are recruited to injuries and thus, they are frontline defenders
against pathogens.Neutrophilsinltrate LNs guided by
inammatory cytokines like IL-1band TNFa,thecomplement
factors C3a and C5a, along the CXCR4-CXCL12 axis, and
eventually they are also attracted by a plethora of inammatory
chemokines (156,157) (Capucetti, Albano, Bonecchi, Frontiers in
Immunology, 2020). Neutrophils are a source ofsoluble mediators
that exert important angiogenic functions. VEGF-A, IL-8,
hepatocyte growth factor (HGF), granulocyte colony-stimulating
factor (G-CSF), and MMP9 are the most important activators of
angiogenesis produced by these cells (103,158). Interestingly,
neutrophils are able to release VEGF-A-enriched granules upon
TNFastimulation and thus, promote vessel growth during
inammation (105). This can become an self-amplifying process
since neutrophil-derived VEGFstimulates neutrophil migration via
an autocrine amplication mechanism, a process that likely
contributes to pathological angiogenesis during inammation and
cancer (159). Human polymorphonuclear granulocytes have been
demonstrated to directly induce the sprouting of capillary-like
structures in an in vitro angiogenesis assay, mediated by secretion
of both pre-formed VEGF from cell stores and de novo synthesized
IL-8 (104). In the murineEm-Tcl1 model, mimicking CLL, a tumor-
associated neutrophil (TAN) population with a B cell helper-like
polarization was identied. Selective depletion of these TANs
retarded leukemia progression in SLOs substantially (160).
Mast cells (MC) are hematopoietic tissue resident immune
cells that are classically recognized as the main effector cell type
of IgE-mediated immediate allergic reactions, however they are
also frequently associated with tumorigenesis (161163).
According to their protease expression, mast cells are divided
in two phenotypical populations: the trypase
+
chymase
(MC
T
)
and the trypase
+
and chymase
+
(MC
TC
) cells (164,165). MC
produce several proangiogenic factors, among them VEGF-A,
VEGF-B, MMP9, and FGF-2. In addition, mast cells
chemotactically respond to VEGF-A and FGF2, indicating that
a connection between mast cell accumulation at tumor sites,
angiogenesis and tumor growth exists (166,167).
Myeloid derived suppressor cells (MDSCs) contribute to the
induction of an immune suppressive and tumor permissive
microenvironment. They are frequently found in SLOs like
spleen, but they are rare in LNs (168). However, they are able
to modulate the L-selectin expression of naïve T and B cells,
preventing efcient HEV adhesion, transmigration, and
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917418
subsequent antigen encounter within LN parenchyma (169,170).
MDSCs promote the formation of T regulatory cells (Tregs), the
secretion of immunosuppressive IL-10 and TGF-b, and inhibit
the activity of cytotoxic CD8 T cells via expression of arginase-1
(Arg1) and inducible nitric oxidase (iNOS) within the TME of
several tumor entities including B cell lymphoma (171,172).
Moreover, MDSCs directly inuence the tumor stroma by
inducing differentiation of cancer-associated broblasts (CAFs)
(173,174). Pro-angiogenic properties of MDSCs during tumor
progression have been reviewed recently (175). MDSCs and their
progenitors, immature myeloid cells are usually not present in
LNs during steady state conditions. However, inammatory
conditions and tumor-derived factors (e.g., CXCL12, GM-CSF,
and CCL2) induce the activation and accumulation of MDSC in
SLOs (176178). MDSCs exhibit numerous immunomodulatory
properties that have considerable potential to inuence
angiogenic processes in LNs, either through direct triggering of
ECs, or by stimulating leukocyte and stromal cells to establish an
angiogenic milieu (175). MDSCs are able to promote tumor
angiogenesis through releasing VEGF-A and MMP9. Mouse
models suggest that MDSCs integrate into the line of vessel-
decorating endothelial cells (179). In mouse melanoma, MDSC
contribute to A2B adenosine receptor-induced VEGF-A
production (111,180). VEGF-A in turn stimulates MDSC
recruitment from the bone marrow, creating a self-enhancing
feedback loop that promotes immunosuppression and vessel
growth (112). One of the reasons why several angiogenic
tumors occur to be insensitive to VEGF-A-targeted therapy is
the presence and recruitment of MDSCs. These cells secrete high
amounts of VEGF-A which might lead to neutralization of the
VEGF-inhibition and additionally, they establish pro-angiogenic
signaling pathways involving several other cells of the TME (110,
181). Moreover, MDSCs limit T cell adhesion and extravasation
by VEGF-A stimulated suppression of endothelial ICAM-1 and
VCAM-1 expression during tumor angiogenesis (71,182).
Lymphocytes, the major regulatory and executive cell subset
of the adaptive immune response are also able to inuence
angiogenesis during inammation and cancer, although their
specic implications are still enigmatic.
T cells comprise different subsets involved in lymphomagenesis,
including naive T cells, memory T cells, and Treg cells (183). Several
negative regulators of T cell activation act as checkpoints to ne-tune
the immune response and regulate hyperactivation. Cytotoxic T
lymphocyte antigen 4 (CTLA-4) andprogrammedcelldeath1(PD-
1) are the most potent examples of T cell immune checkpoint
molecules (ICB) (184). Cancer patients often display dysfunctional
antitumor T cell responses because of the signaling pathways
downstream of these receptors. PD-1 and CTLA-4 inhibition are
subject of extended clinical studies and led already to impressive
response rates in some tumor entities, among them melanoma, non
small cell lung cancer and for hematopoietic tumors, (184186), in
cHL as well (187,188). By targeting abnormal formation of tumor
vessels, anti-angiogenic agents potentially result in an enhanced
inltration of anti-tumor effector cells, making the combination of
immune checkpoint inhibitors and anti-angiogenic agents a
promising and complementary approach in cancer adoptive T cell
therapy (189). On the other hand, as a result of IFNgand IL-12
stimulation, microvascular endothelial cells express checkpoint
molecules like PD-L1 (190,191). In line, arterial vessels express
PD-L1 and PD-L2 after toll like receptor (TLR)-3 activation upon
bacterial infection (192). The regulatory and angiogenic effects of
CD4
+
T helper cells (Th cells) are strictly associated with their
differentiation. Cytotoxic CD8
+
TcellsandCD4
+
Th1 cells produce
IFNgthat restrains endothelial cell proliferation and induces
expression of the angiostatic chemokines CXCL9/10/11 in TAMs
(126,193). In vitro studies revealed that Th2- and Th17 cell-
conditioned medium triggered endothelial sprouting, whereas
medium of Th1 cultures induced vascular regression. Conditioned
medium from Tregs had a minor or no effect (116). In vivo, CD4
+
T
cells display opposing effects on vascularization depending on their
subset differentiation. Th1 cell-derived IFNgimpairs angiogenesis in
ischemic tissue, an effect that is counteracted by regulatory CD4
+
T
cells (Tregs) that antagonize the immunologic Th1 cell response by
secreting anti-inammatory IL-10 and TGFb. Thus, Tregs display
rather indirect pro-angiogenic properties, most likely by paracrine
effects on other potentially pro-angiogenic immune cells (e.g.,
macrophages, DCs, mast cells) (119,194). T cell recruitment,
survival and functionality are highly dependent on tumor-
polarized myeloid cells and tumor-derived factors. The typical
immuno-suppressive milieu of the TME is characterized by
polarizing factors, shifting CD4+ T cell differentiation toward
CD4
+
CD25
+
FOXP3
+
Tregs. In the aggressive Myc-driven murine
lymphoma model, this polarization process is promoted by DCs
expressing increased amounts of the TF C/EBPb(144).
In ovarian cancer, Tregs were selectively recruited into the
tumor tissue via CCL22 and CCL28 production by the tumor
cells and subsequently, Treg-induced secretion of high amounts
of VEGF-A to promote endothelial cell proliferation (118,195).
A striking example for Treg recruitment represents cHL; here,
Tregs are attracted via the Hodgkin-Reed-Sternberg cell-secreted
chemokines CCL17 and CCL22, which engage the Treg-
expressed chemokine receptor CCR4 (196), or by the
chemokine CCL20 that binds to CCR6 (Baumforth,
Birgersdotter, Machado, Am J Pathol, 2008). Th cells and
cytotoxic T cells are required to mediate the anti-angiogenic
effect of IL-12. IL-12activated lymphocytes effectuate inhibition
of tumor growth and function as anti-vascular agents that release
higher amounts of IFNgwhile they down-regulate VEGF in
neighboring cells (197,198). Noteworthy, the presence of IFNg
comes at the expense of an induction of PD-L1 on numerous
stromal cell types, among them endothelial cells (199,200); this
process is likely to counteract the benecial effects of IFNg-
secreting effector T cells which may be rendered dysfunctional
(201). The inltration of tumor sites by cytotoxic CD8
+
T cells is
usually correlated with a favorable clinical prognosis, however
immunosuppressive conditions can polarize these cells to
CD8
+
FOXP3
+
regulatory cells with similar immunomodulatory
and angiogenic properties as CD4
+
Tregs (202205). Studies of
coronary artery disease and systemic sclerosis found T cells with
angiogenic potential in blood samples of patients and
demonstrated that these CD3
+
CD31
+
CXCR4
+
cells (referred to
as angiogenic T cells) play a vital role for the colony formation
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 5917419
and differentiation of endothelial progenitor cells (EPCs) in the
bone marrow (110,206). Such EPCs have been detected in the
circulation and in LN samples from patients with B-NHL as well,
although their inuence on lymphoma-induced vessel growth is
still elusive (207,208). However, inammation models argue
against a signicant functional role of BM-recruited EPCs in LN
vascularization (73).
B cells are frequently found to be part of the TME (209);
however, their role in tumor progression and vascularization is
still unclear. They can directly promote angiogenesis by secreting
pro-angiogenic factors such as VEGF-A, FGF2, and MMP9
(210), or indirectly by polarizing macrophages to the M2 pro-
angiogenic phenotype (211). Transgenic mice (CD19
Cre
/hVEGF-
A
) overexpressing human VEGF-A in murine B cells exhibited a
VEGF-A induced lymphangiogenesis and an expansion of HEVs
in LNs. The authors of the study speculated that the
unphysiologically high levels of human VEGF-A might not
directly inuence the LN lymph- and blood vasculature, but
may rather cause an accumulation of pro-angiogenic
macrophages (122). In a mouse model of LCMV infection, B
cells were shown to be required for LN tissue remodeling and
vessel expansion. Surprisingly, the latter was independent of
VEGF-A signaling pathways, but required LTa
1
b
2
-expressing B
cells (50). A recent study emphasized the angiogenic capacity of a
B cell subset during eosinophilic esophagitis and in patients with
melanoma. These pro-angiogenic B cells were identied by the
surface markers IgG4
+
CD49b
+
CD73
+
and shown to promote
vascular tube formation in vitro through VEGF-A, FGF2, and
PDGFA expression (123). Taken together, although B cells
express VEGF-A and LTa
1
b
2
during certain conditions, their
role in LN angiogenesis is not well understood. Potentially, B
cells may exert pro-angiogenic effects themselves, but also
through stimulation of other stromal cell types, such as FRCs
and macrophages (Figure 2).
B CELL LYMPHOMA-INDUCED
VASCULAR CHANGES ARE DEPENDENT
ON THE ENTITY AND STATE OF
LYMPHOMA PROGRESSION
The clinical importance of angiogenic processes and mechanisms
for the growth of solid tumors is well recognized (212,213).
Therapeutic concepts from solid tumors targeting the VEGF-A/
VEGFR1/2 axis have been adopted for combinatorial therapies of
B-NHL, resulting in rather disappointing clinical outcomes (214,
215). We recently showed that angiogenic processes in LNs in a
mouse model of high-grade B cell lymphoma are induced by
signaling pathways distinct from solid tumors. In sharp contrast
to most solid tumors, lymphoma growth in LNs was not
associated with hypoxic conditions or inammation. Instead,
lymphoma affected vessel expansion via the VEGF-C/VEGFR3
and LTa
1
b
2
/LTbR signaling axes (42). In patients, the growth of
tumor cells in low-grade B-NHLs is usually exponential for a few
months and remains in a steady state as indolently growing
tumor mass for years. This indolent lymphoma is considered to
be avascular with dormant endothelial cells within the TME. In
contrast, high-grade B-NHL progression is often accompanied
by a so called vascular phase, which represents extensive
vascularization of LNs (216,217). Such intermediate- and
high-grade B-NHLs grow exponentially without intermission
phase until they reach a mass critical for a patients survival.
As a clinical indicator of the vascularization, B-NHLs are usually
quantied by terms of the microvessel density (MVD).
Immunohistology using anti-CD31 antibody staining is still
considered the gold standardof blood vessel detection, even
though there is substantial variation between different studies
due to the heterogeneity of the lymphoma stroma and different
scoring methodologies. In some cases, the marker CD34 is used
to detect the blood vasculature. Notably, lymphatic vasculature
also expresses CD31, but at much lower levels (42,218,219).
Non-invasive assessment of tumor vascularization in vivo is
possible by using Doppler sonography, contrast-enhanced
dynamic magnetic resonance imaging (dMRI) and positron
emission tomography-computer tomography (PET-CT). These
techniques do not allow a direct quantication of the blood
vessel density but provide information on the functional status
of the blood vessels, e.g., vessel integrity, permeability, perfusion
and metabolism (220). Another diagnostic approach to detect
ongoing angiogenesis in vivo is the serological quantication of
growth factors. VEGF-A levels in the serum of patients with
progressive NHL were signicantly elevated in comparison to
patients in complete remission (221,222). Elevated VEGF-A levels
have been found in aggressive B cell lymphoma subtypes including
MCL, DLBCL, but also in indolent lymphoma, such as CLL and
small lymphocytic lymphoma (SLL), respectively (223225). A
variety of commonly used B-NHL cell lines secrete measurable
VEGF amounts under serum starvation conditions, whereas other
angiogenic factors like the placental growth factor (PlGF) and
FGF-2 are not expressed (226). However, the detection of
angiogenic factors in clinical serum samples gives no
information on the cellular source of these molecules and is not
a reliable indicator of angiogenesis in the compartment of interest.
Previously, a group of angiogenesis experts published consensus
guidelines for the use and interpretation of angiogenesis assays,
which involve in vivo, ex vivo explantation, and in vitro bioassays.
They explicitly highlighted critical aspects that are relevant for the
execution of angiogenesis detection and proper interpretation (227).
Mantle cell lymphoma (MCL) is an aggressive B cell
neoplasm that comprises 6% of all NHL cases (228,229). It is
susceptible to paracrine signaling from the microenvironment
and in turn shapes the microenvironment by secreting soluble
factors (230). MCL is genetically characterized by overexpression
of the CCND1gene, encoding for cyclin D1 (231). Recent studies
identied a subgroup of MCL that has a more indolent behavior
with a clinical presentation as leukemic disease, exhibiting
minimal LN distribution and a frequent splenomegaly. These
tumors also overexpress cyclin D1 but lack expression of the sex
determining region-Y-box11 (SOX11), a TF specically
expressedinconventionalMCLandassociatedwithan
aggressive and angiogenic phenotype (232). These results have
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174110
been conrmed in MCL patient samples by using
immunohistochemistry, demonstrating a correlation between
an increased MVD and high levels of SOX11 expression (233).
Experiments with MCL tumor xenotransplants in mice, in cell
lines, and in primary MCL samples revealed that SOX11 actively
modulates angiogenesis by up-regulation of the platelet-derived
growth factor a(PDGFa), which is a competent inducer of an
FRC-associated pro-angiogenic program (234,235). Moreover,
SOX11 overexpression promotes B cell receptor signaling
represses Bcl6 transcription and upregulates PAX5 to avoid B
cell differentiation into memory B cells or plasma cells. PAX5
supports tumor cell homing and invasion via up-regulation of
CXCR4 and the focal adhesion kinase (FAK) (236238). The
absolute monocyte count in MCL correlated with the prognosis
and supports the hypothesis that the TME is relevant for MCL
tumor progression (239). CD68
+
and CD163
+
macrophages were
found in MCL LNs without exception. Substantial numbers of
VEGF-C expressing macrophages were found in a mouse
xenotransplantation model as well (240). Treatment with the
immunomodulator lenalidomide depleted monocytes and
VEGF-C expressing macrophages, resulted in impaired
functional lymphangiogenesis. However, a relevant impact on
lymphoma-associated blood vessel growth in MCL was not
investigated in this study. Of note, in human MCL anti-
inammatory and pro-angiogenic CD163
+
cells (M2-like)
outnumbered the more inammatory CD68
+
CD163
-
macrophages (233), indicating a propensity to stimulate
angiogenesis. This M2-like polarization of macrophages is
actively driven by MCL derived CSF-1 and IL-10 (241). MCL
cells exhibit increased expression of the T cell, B cell, and
monocyte recruiting chemokines CCL4 and CCL5 compared to
normal B cells (242). T cell inltration has been considered as a
prognostic marker in MCL in which CD8
+
, and particularly
CD4
+
T cell frequencies are higher in indolent MCL and decrease
with more aggressive histological and clinical presentation (243).
In contrast, a recent study reported an expanded vascularization
of MCL associated with a high inltration of CD4
+
and CD8
+
T
lymphocytes (233). The differences might be explained by a weak
comparability of data that were either correlated with the clinical
outcome, or with the SOX11 expression level in MCL, two
hallmarks that are not always correlated. A more detailed T
cell characterization of CD4/CD8 T cell subsets is required for a
more reliable assessment of the T cellrelated inuence on
angiogenesis and the clinical outcome in MCL. Interestingly,
MCL cells itself express the VEGFR-1, providing a strong
rationale to target VEGF in order to interfere with angiogenic
processes and concomitantly, with autocrine survival signals
(230,244).
Angiogenesis is likely a part of MCL progression, driven by
MCL derived PDGFa. Therapeutical interference with PDGFR-b
signaling, the receptor for PDGFa, can be achieved with receptor
tyrosine kinase (RTK) inhibitors. Some PDGFR-btargeted drugs
have been tested in clinical trials for B-NHL but failed to bring
signicant benet(217). In contrast, immunomodulating drugs
(IMiD) like thalidomide and lenalidomide have anti-angiogenic
properties and showed great potential in combination with
rituximab for the treatment of untreated or relapsed MCL
patients (245,246).
Follicular lymphoma (FL) is the second most common B-
NHL, accounting for 20% of all B-NHL cases (247). The disease
affects LNs, spleen and frequently also the bone marrow.
Neoplastic follicles in FL have a lower proliferative index in
comparison to reactive germinal centers. However, the
proliferative capacity of FL cells increases gradually with the
FL grade. FL progression requires the supporting infrastructure
of the follicular TME to maintain survival, a requirement that
gets progressively lost in the process of transformation to
aggressive DLBCL (248,249). Follicular dendritic cells (FDCs)
are one branch of this supporting infrastructure. They are of
mesenchymal origin and represent a crucial stromal cell
population supporting the germinal center reaction and
maintenance of the B cell follicle in LN and spleen (250,251).
FDC secreted B cell survival factors such as Indian hedgehog
(HH), the B cell activating factor (BAFF), and IL-15 are
potentially pro-tumorigenic (252,253). CXCR5-controlled
access to FDCs conferred survival and proliferation stimuli to
CLL B cells in the murine Em-Tcl1 model, which mimics some
aspects of indolent tumor growth (253). Similar to reactive LN
follicles, neoplastic follicles in FL preserve the organized FDC
network structure at least in early stages of the disease
progression (254). FL-FDC cross-talk induces a pro-angiogenic
expression pattern in FL cells, including secretion of VEGF-A
and VEGF-C (255). This cross-talk is crucially dependent on the
phosphoinositide-3-kinase d(PI3Kd), providing therapeutic
intervention options with PI3K specic inhibitors like
idelalisib, which is approved for the treatment of FL, CLL, and
SLL (256). The second branch of the supportive infrastructure in
follicles are the CD4
+
CXCR5
+
PD1
+
T follicular helper (Tfh)
cells, which provide vital survival signals for FL cells by secreting
IL2, IL4, IFNg, and by CD40L presentation (9,257). FL cells are
further dependent on proliferation and survival signals of the B
cell receptor (BCR) in interaction with FDCs and TAMs (258).
Elevated numbers of M2-like TAMs are found in the immediate
microenvironment of FL cells and neo-vascular sprouts within
the follicle (138). However, the prognostic value of CD163
+
TAMs remains controversial and is highly dependent on the
prior course of treatment (259). In sum, FL appeared to be less
prone to induce relevant vascular changes, whereas LNs of high-
grade B-NHLs exhibited a dense and aberrantly distributed
vasculature within the paracortical zone. In contrast to most
other B-NHL malignancies in which high levels of pro-
angiogenic factors and an increased MVD is associated with
an adverse prognosis, high level FL vascularization correlates
with a benecial disease course (260262). The improved
clinical outcome apparently correlated with the increased
vascularization, but was surprisingly independent of follicular
VEGF-A expression (223,263). Some studies stated a minor
vascular remodeling in FL compared to reactive LN or follicular
hyperplasia, or even vascular regression constraining the growth
of reactive and neoplastic follicles (260,264). Therefore, the
clinical signicance of angiogenesis in FL remains uncertain. In
one clinical trial, addition of the anti-VEGF bevacizumab during
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174111
rituximab treatment of relapsed FL signicantly improved the
progression-free survival (265). The potential of angiogenesis
inhibition upon treatment of FL requires further evaluation in
larger clinical trials.
Collectively, according to the data currently available it seems
that angiogenesis is important for high-grade lymphoma, but has
less impact on indolent FL growth.
Diffuse large B-cell lymphoma (DLBCL) is the most
common type of lymphoid tumors worldwide accounting for
30% of all diagnosed NHL and characterized by the large size of
neoplastic B cells and usually a very aggressive clinical
presentation (266). Lenz et al. identied gene expression
proles in LN from patients pre-treated with the combination
therapy anti-CD20 antibody, cyclophosphamide, doxorubicin,
vincristin, prednisolone (R-CHOP), dividing DLBCL in two
distinct subgroups that are predictive of the clinical outcome
(267). The stromal-1signature includes expression of
extracellular matrix (ECM) elements, ECM remodeling factors
(MMP2,MMP9,M1-MMP,PLAU,TIMP2) and is associated
with a favorable prognosis. The stromal-2signaturewas
found in tumors with an increased MVD and is characterized
by markers of endothelial cells (Pecam1,Vwf,Kdr,Tek). The
latter signature is associated with a poor clinical outcome,
emphasizing the critical impact of angiogenic processes on
aggressive B-NHL progression. Several studies investigated the
clinical consequences of the stromal-2signature and conrmed
the correlation of a high MVD with an adverse outcome and a
shorter overall survival rate (268271). The relationship between
MVD and DLBCL behavior was the object of many studies and
was found to be associated with poor prognostic parameters such
as splenic involvement, high mitotic rate, and capsular invasion
(268272). Gomez-Gelvez et al. reported contradictory results,
showing that high MVD is associated with rather better
progression-free survival (PFS) and event-free survival (EFS)
(273). Several other studies also failed to draw a connection
between the MVD, tumor grade and prognostic outcome (274
277). A DLBCL mouse xenotransplantation model demonstrated
that the inhibition of the paracrine VEGFR-2 pathway reduced
growth of an established lymphoma and correlated with
decreased tumor angiogenesis (226). DLBCL cells often
overexpress the phosphodiesterase 4B (PDE4B), which
intracellularly catalyzes the hydrolysis of cyclic-AMP (cAMP).
The cAMP-PDE4B axis modulates signaling of PI3K and AKT
and therefore acts upstream of VEGF-A expression. Experiments
with genetically or pharmacologically inhibited PDE4B resulted
in decreased VEGF-A expression in lymphoma cells and reduced
angiogenesis in the Eµ-Myc high-grade lymphoma mouse
model (278).
In a gene expression study on relapsed or refractory DLBCL,
patients with the ABC-like DLBCL subtype that had low VEGF
121
isoform expression, exhibited a signicantly better overal survival
than those with high VEGF
121
gene expression levels (279).
Interestingly, VEGF
121
low transcript levels were associated to a
gene signature reecting immune response and T cell activation.
DCs are likely a major source of VEGF-A in LNs with DLBCL.
Functionally, DCs could be involved in lymphoma TME
remodeling, but their number in DLBCL LNs is signicantly
lower than in reactive LNs. Lower expression levels of the LN
homing receptors CD62L and CCR7 in DCs in LNs of DLBCL
patients were thought to result in reduced DC immigration.
However, it remains elusive if the DCs lose the receptor
expression upon arrival in the LN, or whether these cells are
recruited via alternative routes (280). In an aggressive Myc-driven
lymphoma model in mice a tumor-specic DC differentiation
occurs that promotes tumor cell survival and favors the
maturation of monocytic-derived DCs (MHCII
medium
)(149,150).
Alongside tumor repressing M1 macrophages, alternatively
activated M2 macrophages exhibit angiogenic capacities, they are
frequently found in DLBCL and often correlate with a poor
prognosis (281283). Although numerous studies reported an
association between TAMs and MVD in DLBCL, others could
not nd a correlation between CD68
+
macrophages and an
increased MVD (270). Such controversies can probably be best
explained by variabilities in the methodological approaches. The
macrophage marker CD68 represents M1 and M2 macrophages
and therefore, produces inaccuracies in the interpretation of studies
concerning the macrophage-MVD correlation. The addition of the
marker CD163, which rather recognizes M2 activated
macrophages, including angiogenic macrophages, provides a
more reliable view on the role of macrophages in DLBCL (284).
Elevated numbers of macrophages have been correlated with poor
prognosis in DLBCL (282). However, in therapeutic setting
macrophages are required to confer treatment effects when
patients were treated with anti-CD20 antibody (e.g., Rituximab).
Here, macrophages mediate tumor cell depletion via the
macrophage Fc-gamma receptor (FcgR) expression (215).
Another abundant immune cell population in LNs of DLBCL
patients is mast cells with a predominance of MC
T
-type (tryptase-
positive) cells. M C
TC
-type (tryptase-positive andchymasepositive)
and CD4
+
Th
2
were shown to express IL-4 in DLBCL and therefore,
they may actively promote survival of the tumor cells (285).
Hedström and coworkers examined 154 DLBCL cases and
suggested that the inltration of mast cells reects the
inammatory immune response of the endogenous anti-tumor
defense and is therefore related to a favorable outcome (286). The
gradual increase of the MVD was correlated with an increasing
number of mast cells. Although mast cells are considered to be
bystandersin tumor immunology,additionalpro-angiogeniceffects
of these cells are likely as they secrete relevant amounts of different
VEGFs, FGF-2, trypase, and granzyme B. The latter has a pro-
angiogenic effect via the enzymatic mobilization of ECM-bound
FGF-1 (287,288). The wide range of physiological conditions and
tumor entities that include mast cell-supported angiogenesis and
the respective recruitment and signaling pathways were excellently
reviewed by Ribatti et al. (289).
Apart from the direct effect on immune and tumor cells,
surprisingly, the application of the VEGF-A inhibiting antibody
bevacizumab to R-CHOP therapy increased adverse cardiac events,
yet without increasing the therapeutic efcacy in DLBCL patients
(214,215). From these studies it can be inferred that the increased
MVD in DLBCL patients may be simply a correlation with minor
importance for the disease course, or that other non-VEGF-A
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174112
angiogenic pathwaysprevail and cause enhanced vascular assembly
instead. Ina study of Pazgal et al., VEGF-C, VEGF-D, and VEGFR3
were expressed in both lymphoma cells and endothelial cells of the
blood and lymphatic vasculature. They reported a signicant
correlation of the VEGF-C expression and the presence of blood
vessels. VEGF-D expression correlated with the patient
International Prognostic Index (IPI) Score and the patients
overall survival (14). These results may indicate that apart from
its role as primary signaling pathway for lymphatic vessels, the
VEGF-CVEGFR3 axis also has implications on angiogenic
processes of LN blood vessels. A study in breast cancer
demonstrated that VEGFR3 is signicantly upregulated in the
endothelium of new blood vessels. The results also suggested that
VEGF-C secreted by the intraductal carcinoma cells acts
predominantly as an angiogenic growth factor for blood vessels,
although other immune or stromal cells might be involved in this
paracrine signaling network as well (290). An experimental study
using a Myc-driven aggressive lymphoma mouse model, which
resembles important aspects of aggressive B-NHL, supported this
hypothesis, showing that the MVD expansion was triggered by
lymphoma-provided VEGF-C, in a synergistic activity with
LTa
1
b
2
(42).
Representing a high-grade and angiogenesis-associated
lymphoma type, multiple clinical trials with anti-angiogenic
agents for the treatment of DLBCL have been conducted. Most of
the treatment approaches using single agent angiogenesis inhibitors
failed to prove a benecial effect. However, combinatorial treatment
strategies such as R2-CHOP (lenalidomide, R-CHOP) (291,292),
brought encouraging results. Such oberservations emphasize that
anti-angiogenesis therapies might not be effective when applied
alone, even in highly vascularized lymphoma, but are valuable
components in combination with other drugs.
Burkittslymphoma(BL) represents around half of all malignant
non-Hodgkin lymphoma in children and around 2% in adults (293).
The BL pathogenicity is usually associated with the infection of B cells
with the Epstein-Barr virus (EBV). EBV gene products induce BL
cell-derived soluble factors that result in inhibition of neo-
vascularization and eventually tumor necrosis and regression (294).
However, in in vivo experiments, EBV-positive cells induced massive
recruitment of leukocytes at the tumor border and the development
of granulation tissue with large numbers of blood and lymphatic
vessels (295). Surprisingly, aggressive BL displayed the highest MVD
in comparison to intermediate DLBCL and indolent B-NHL (42,262,
287). In support of this observation, BL showed increased
vascularization relative to benign lymphadenopathies and can
produce several angiogenic factors, although it is not yet known
whether this is due to Myc gene overexpression or the EBV
transformation (296298). BL were found to be closely associated
with VEGF-producing CD68
+
VEGFR1
+
myeloid cells located
around the neo-vasculature. The newly formed blood vessels were
identied by the absence of pericyte coverage as result of the rapid
vessel growth (299,300). Genetic depletion of this subpopulation of
CD68+VEGFR1+ myeloid cells was sufcient to inhibit angiogenesis
in experimental lymphoma (301). To our knowledge, to date there are
no clinical data or published treatment strategies of BL that target
angiogenesis specically.
Classical Hodgkin Lymphoma (cHL) is characterized by
mono-nucleated Hodgkin and multi-nucleated Reed-Sternberg
(HRS) cells, which comprises tumors with mixed cellularity,
nodular sclerosis and lymphocyte-rich or lymphocyte-depleted
subtypes. Different from other lymphoma, HRS cells are the
minority of cells within the affected LN. Most of the cells in cHL
tumors are cells of the TME, indicating a prominent role ofbenign
immune cells and the LN stroma (302). A crucial role of
angiogenesis and increased MVD have been reported for cHL
and correlate with a poor prognosis (303). Similar to observations
in highly vascularized LNs in an aggressive B-NHL mouse model
(42) and in immunohistochemically characterized B-NHL patient
specimen (277), in cHL HIF-1awas only moderately expressed
(304), suggesting that angiogenesis in cHL is not hypoxia-driven
and may utilize other angiogenic pathways instead. In childhood
cHL, HRS cells express VEGF, MMP-2 and MMP-9. However, the
expression of these factors did not correlate with the MVD and
neovascularization level (305,306). On the other hand, VEGF-D, a
ligand for VEGFR3 and usually associated with lymphangiogenesis,
is expressed in HRS cells at high abundance and correlated with
high numbers of microvessels (307). Moreover, in vitro HRS cell-
derived TGF-b,FGF-2,andVEGFsupportedHUVEC
tubulogenesis (308,309). Secretion of Ltaby HRS cells activated
endothelial cells, which enhances adhesion molecule expression
and consequently, recruitment of T cells. This mechanism amplies
the inammatory milieu in the cHL TME through conditioning of
the blood vasculature (310).
Commonly attributed to the occurrence of angiogenic M2
macrophages, TAMs are linked to poor outcome in HL.
Interestingly, lack of macrophages, but also high numbers of
macrophages is associated with a poorer disease-free survival and
overall survival, whereas intermediate numbers are associated
with a better outcome. This macrophage paradox suggests that a
lack of TAMs is benecial for HL growth, while TAMs have an
inhibitory effect with increasing numbers (311). The inhibitory
effect seems to be displaced by an adverse effect of TAM-induced
angiogenesis, supposedly predominated by CD163
+
M2-like
TAMs (312). High numbers of CD163
+
TAMs correlate with
elevated VEGF-A levels and an increased MVD, indicating that
CD163 is an independent prognostic marker in cHL (313).
Interestingly, although the particular signaling pathways within
TAMs remain elusive, pre-clinical experiments with PI3K-Akt
pathway inhibition suggested a connection to macrophage M2-
polarization (314,315), which could be a promising anti-
angiogenic intervention clue by prevention of pro-angiogenic
activity of M2-like TAMs.
ANTI-ANGIOGENIC THERAPIES IN
COMBINATION WITH CHEMOTHERAPIES
Cancer therapy earlier than the 1970s was solely focused on
targeting the actual cancer cells. Judah Folkmans discovery that
tumor growth is angiogenesis-dependent led to a profound
paradigm shift in cancer therapy (316,317). Sprouting
angiogenesis plays an essential role in tumor growth, invasion,
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174113
progression, and metastasis, targeting this process is a promising
strategy to inhibit growth and spread of solid tumors. Clinical
trials and treatment strategies of anti-angiogenesis therapy in B-
NHL were recently reviewed (217). Angiogenesis inhibitors are
classied into direct and indirect agents. Direct inhibitors target
vascular ECs and include endostatin, arrestin, and tumstatin.
Indirect angiogenesis inhibitors target tumor cells or cells of the
TME to prevent the expression of pro-angiogenic factors or
block their activity (318). The anti-VEGF monoclonal antibody
Bevacizumab was the rst anti-angiogenesis drug approved by
the FDA for the treatment of metastatic colon, ovarian, renal,
non-squamous cell lung cancer, and glioblastoma multiforme.
Unfortunately, clinical signicance was only reached in
glioblastoma multiforme treatment (319,320), a result that
could not be conrmed in other studies (321). In contrast to
Bevacizumab, treatment with tyrosine kinase inhibitors (e.g.,
Sorafenib) that interfere with the signal transmission of
VEGFRs resulted in remarkable effects throughout several
cancer entities. Combination of tyrosine kinase inhibitors and
conventional chemotherapy have not been benecial (322).
Conventional chemotherapy can cause direct cytotoxicity of
endothelial cells, but this effect is non-selective and only
observed upon the maximal tolerated dose (MTD). Insufcient
tumor and vascular bed destruction can effectuate a strong
hypoxic condition, which results in release of chemoattractant
CXCL12. Accordingly, MTD chemotherapy potentially increases
systemic CXCL12, which recruits bone marrowderived EPCs.
These cells can cause recurring angiogenesis in mouse models of
solid tumors (323,324). Therefore, an anti-angiogenesis therapy
that is complementary to chemo- or immunotherapy is aimed at
restricting pro-angiogenic bystander effects of the tumor
treatment. In addition, instead of aiming for a complete
vascular eradication, the paradigm in anti-angiogenic therapies
shifted to vascular normalization (325,326).
Rituximab has become an essential part ofrst-line treatment of
several B cell lymphoma entities, foremost of DLBCL. However,
ongoing research aims to improvethe therapeutic efciency and the
reduction of the relapse rate of drug-resistant lymphoma cells.
Tumor anti-angiogenesis therapy approaches are one branch of
such research, in whichBevacizumab and Endostatin werethe most
promising representatives for lymphoma treatment (327,328).
VEGF-A has a crucial role in promoting vessel growth, but is also
considered to be an immunosuppressive factor that modulates the
migration and function of several immune cells, e.g., DCs and mast
cells. The potential pharmaceutical targeting of the VEGF/VEGFR
axis to modulate anti-tumor immunity has been reviewed
recently (329).
An important challenge of anti-angiogenic therapy in solid
tumors as well as in lymphoma is the identication of the
particular angioactive receptors throughout different tumor entities
and individual patients. The inhibition of intracellular signaling hubs
is a strategy to overcome the targeting of distinct angiogenic tyrosine-
kinase receptors. Class I PI3Ks are involved in the signal transduction
of many pro-angiogenic signals and control cell growth, survival,
motility, and metabolism (330). PI3Kdinhibition in lymphoma
potentially also interferes with tonic signaling in tumor cells, e.g.,
via the BCR signaling pathway (331), or breaks the Treg-mediated
immune tolerance (332). Interestingly, PI3K activity is essential for
macrophage M2 polarization (333) and therefore, a potential target to
hamper M2-like angiogenic macrophages. Inhibition of PI3K
signaling represents a valuable therapeutic strategy to target
different indolent B cell lymphoma entities, among them FL, CLL,
SLL, and more recently, they showed promise in T cell lymphomas as
well (334,335). The combinatorial treatment of the rst generation
PI3K inhibitor idelalisib with rituximab or bendamustine revealed
favorable response rates in FL patients (334), but serious adverse
effects due to bacterial and viral infections were observed.
Additionally, immune-mediated and hematologic adverse events
occurred. Beyond that rst generation PI3K inhibitor, newer PI3K
inhibitors such as copanlisib and duvelisib were introduced for
patients with relapsed and progressive FL, CLL, SLL, respectively.
These inhibitors differ in their preference for PI3K isoforms which are
expressed differentially in various tissues (336). Despite relevant side
effects of PI3K inhibitors, they have been judged clinically
manageable and thus, prompted an FDA approval for relapsed and
refractory indolent B-NHL (335,337). Published reports on anti-
angiogenic therapies in B-NHL allow the conclusion that the complex
mechanisms of angiogenesis in lymphoma are incompletely
understood and require further pre-clinical and translational
research to develop reliable and effective anti-angiogenic treatment
strategies. Moreover, new anti-angiogenic treatment regimens need
to be validated regarding an actual reduction of tumor growth, since
sole targeting of angiogenic factors often fail to cause substantial
tumor regression (Figure 3)(340).
OUTLOOK
Vascular remodeling and angiogenesis have been increasingly
recognized as crucial factors in the pathophysiology of B-NHLs.
We here present an integrated concept that includes angiogenic
processes of the LN TME beyond the proliferation and survival
of endothelial cells stimulated by the VEGF-VEGFR axis. In
human, the angiogenic properties of LN resident and recruited
immune cells are still insufciently understood. Studies to
address such functional states are limited because tissues from
appropriate human patients are rarely available. Notably, most of
the human data available so far are observations on clinically
progressed and even terminal stage lymphoma LNs. Flow
cytometry analysis of blood samples is usually limited to a few
entities (e.g., FL, CLL, and MCL), common markers and cannot
readily be correlated with pathohistological observations due to
the lack of tissue specimen. Availability of LN tissue from
progressed disease stages is often limited to scarce material
from ne-needle biopsies. As a useful surrogate, mouse models
of reactive LNs and LNs with lymphoma growth demonstrated
that the angiogenic processes are regulated by a timely complex
interplay of immune, tumor, and stromal cells (42).
In the future, modern methods like single-cell RNA sequencing
alone or with spatial resolution, or single-cell analysis in
combination with proteomics will help to resolve the complexity
of participating cells and their heterogenous differentiation status.
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174114
This technique requires much less input material for a high
resolution analysis at the genome, protein, or epigenome level
(341343). Even patient-derived specimen from ne needle
biopsies seem amenable to such analysis, allowing then a kinetic
description of LN remodeling in the course of diagnosis, treatment
response, and eventually relapse. Single-cell RNA sequencing will
further enable the discrimination of different endothelial cell
subtypes and their differentiation traits (344). The compartment
of BECs is comprised of several functionally distinguishable
subpopulations that further differentiate during angiogenesis. To
date, we know little about the role, differentiation conditions and
distribution of these subpopulations in LNs. Such transcriptional
observations need to be correlated with the topology of the single
cells and cell networks within the complex LN (345). Modern
imaging methods, e.g., light sheet microscopy and intravital 2-
photon microscopy enable the complex spatial integration and the
investigation of dynamic processes in situ, which have long been
restricted to snapshot observations. Very recently, a new generation
of ow cytometry devices became available that, based on a
spectrum wide detection of uorophores, allow a simultaneous
detection of a multi-fold higher number of cell markers. The
possibility to determine extended marker panels with small
sample sizes will not only improve basic knowledge in the pre-
clinical context, but will also provide innovative approaches for
clinical diagnostics (346,347).
Improving insight into angiogenesis is also of considerable
relevance for the emerging immunotherapies using chimeric
antigen receptor (CAR)- and T cell receptor (TCR)-transgenic
T cells and NK cells. It is reasonable to suggest that tumor blood
vessels have a leading role in granting effector T cell access to the LN
and the tumor niche formed therein. For example, solid tumors
condition an endothelial activation status that can be considered
immunologically silent(348). However, reactivation of such
vessel-lining endothelial layers is a prerequisite for the adhesion
and transmigration cascade of naive and therapeutic T cell
populations. We envision that this endothelial tuning is not only
applicable to solid tumors, but also to LN-localized lymphatic
neoplasm. Except for cHL, immune checkpoint blockade (ICB)
targeting PD-1 or CTLA-4 has not shown relevant benetinotherB
cell neoplasm. Because ICB efcacy depends on the presence of a
repertoire of antigen-specic T cells, a rational sequence of
immunotherapeutic interference in B-NHL would start with a
vessel induction toward a more activated or even inammatory
state. It seems not even necessary to overactivate local endothelial
cells, as shown by the application of a modied TNFacytokine that
upregulatesadhesionmolecules,buttheneveneradicatessolid
tumors through rapid destruction of the tumor neovasculature
(349). Enhanced adhesion, e.g., involving ICAM-1 and VCAM-1
up-regulation, may be sufcient to allow T cells to get access to the
primary lymphoma site in the deep parenchyma. Finally, in a time
window to be dened, application of ICB might then unleash the
activity of effector T cells that already invaded the tumor site.
Collectively, efforts to target tumor cells only or single
lymphoma-promoting cellular stromal elements in the TME are
unlikely to confer long lasting remissions. For example, although
anti-CD19 CAR T-cell therapies have proven remarkable efcacy in
B cell malignancies, they become ineffective due to CD19 antigen
loss or downregulation (350,351). Other contributing factors to
A
B
C
FIGURE 3 | Therapeutic strategies to induce vessel normalization and revert endothelial anergy in B-NHL. (A) Anti-angiogenesis therapy tageted at VEGFR-2 or
VEGFR-3 can restore a normalized vessel network. (B) Targeting of the LTbR with LTa
1
b
2
and LIGHT expressing DCs, agonistic antibodies or recombinant factors
potentially circumvents impaired lymphocyte homing by establishing or stabilizing HEV integrity within the lymphoma TME (338). (C) Vessel anergy can be changed
by a targeted conversion of the endothelium toward a reactive endothelium using inammatory cytokines, which might be site directed to avoid unintended systemic
effects. Normalization of aberrant vessels and activation of the endothelium can also be achieved by locally applied low-dose gamma irradiation (339). Reactive
endothelium within LNs is a prerequisite for an effective inltration of effector T cells during cellular immunotherapy.
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174115
substantial rates of treatment failure might be the nodal
immunosuppressive microenvironments in B-NHL (9,352).What
is needed is an integrative concept that blocks vicious feedback
cycles in lymphoma. We suggest that combinatorial targeting of
aberrantly polarized myeloid cell populations, blood endothelial
activation, angiogenesis, and effector T cell dysfunction is a rational
stepwise strategy (Figure 3). Advanced CAR T cell technologies try
to integrate a few of these demands, for example by deleting the
functionality of PD-1 (353,354) and secretion of immune-
stimulatory cytokines such as IL-12, IL-21, or IL-18 (355357).
We envision that the vasculature is important for control of
lymphoma relapse. In this process, mutual stimulation of residual
tumor cells, mesenchymal and hematopoietic stromal cells, and
endothelial cells might favor neo-angiogenesis and eventually, re-
shaping a growth supporting niche for lymphoma B cells.
AUTHOR CONTRIBUTIONS
LM: conceived the general idea, wrote the manuscript, and
created the gures and tables. AR: conceived the general idea,
co-wrote the manuscript, and edited gures and tables. UH:
provided expert opinion/knowledge input and edited
manuscript, gures, and tables. All authors contributed to the
article and approved the submitted version.
FUNDING
This work was funded by the Wilhelm Sander-Stiftung (grant
number 213.100.02), and by the Deutsche Krebshilfe (grant
number 107749) awarded to AR and UH.
REFERENCES
1. Krishnamurty AT, Turley SJ. Lymph node stromal cells: cartographers of the
immune system. Nat Immunol (2020). doi: 10.1038/s41590-020-0635-3
2. Till KJ, Coupland SE, Pettitt AR. Motility and trafcking in B-cell non-
Hodgkinslymphoma(Review).Int J Oncol (2014). doi: 10.3892/
ijo.2014.2395
3. Wang L, Qin W, Huo YJ, Li X, Shi Q, Rasko JEJ, et al. Advances in targeted
therapy for malignant lymphoma. Signal Transduct Target Ther (2020). doi:
10.1038/s41392-020-0113-2
4. Maman S, Witz IP. A history of exploring cancer in context. Nat Rev Cancer
(2018). doi: 10.1038/s41568-018-0006-7
5. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell
(2011). doi: 10.1016/j.cell.2011.02.013
6.BurgerJA,GhiaP,RosenwaldA,Caligaris-CappioF.The
microenvironment in mature B-cell malignancies: A target for new
treatment strategies. Blood (2009). doi: 10.1182/blood-2009-06-225326
7. Menter T, Tzankov A. Lymphomas and their microenvironment: A
multifaceted relationship. Pathobiology (2019). doi: 10.1159/000502912
8. Wolf J, Diehl V. Hodgkin diseasepathogenesis and therapy. Ther Umsch
(1996).
9. Höpken UE, Rehm A. Targeting the Tumor Microenvironment of Leukemia
and Lymphoma. Trends Cancer (2019). doi: 10.1016/j.trecan.2019.05.001
10. Cao Z, Ding BS, Guo P, Lee SB, Butler JM, Casey SC, et al. Angiocrine factors
deployed by tumor vascular niche induce B cell lymphoma invasiveness and
chemoresistance. Cancer Cell (2014). doi: 10.1016/j.ccr.2014.02.005
11. Liao S, Ruddle NH. Synchrony of High Endothelial Venules and Lymphatic
Vessels Revealed by Immunization. J Immunol (2006). doi: 10.4049/
jimmunol.177.5.3369
12. Dasoveanu DC, Shipman WD, Chia JJ, Chyou S, Lu TT. Regulation of
Lymph Node VascularStromal Compartment by Dendritic Cells. Trends
Immunol (2016). doi: 10.1016/j.it.2016.08.013
13. Habenicht LM, Kirschbaum SB, Furuya M, Harrell MI, Ruddell A. Tumor
regulation of lymph node lymphatic sinus growth and lymph ow in mice
and in humans. Yale J Biol Med (2017).
14. Pazgal I, Boycov O, Shpilberg O, Okon E, Bairey O. Expression of VEGF-C,
VEGF-D and their receptor VEGFR-3 in diffuse large B-cell lymphomas.
Leuk Lymphoma (2007). doi: 10.1080/10428190701632822
15. Girard JP, Moussion C, Förster R. HEVs, lymphatics and homeostatic
immune cell trafcking in lymph nodes. Nat Rev Immunol (2012). doi:
10.1038/nri3298
16. Ito K, Smith BR, Parashurama N, Yoon JK, Song SY, Miething C, et al.
Unexpected dissemination patterns in lymphoma progression revealed by
serial imaging within a murine lymph node. Cancer Res (2012). doi: 10.1158/
0008-5472.CAN-12-2579
17. Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, et al. An
embryonic stem cell-like gene expression signature in poorly differentiated
aggressive human tumors. Nat Genet (2008). doi: 10.1038/ng.127
18. Naxerova K, Bult CJ, Peaston A, Fancher K, Knowles BB, Kasif S, et al.
Analysis of gene expression in a developmental context emphasizes distinct
biological leitmotifs in human cancers. Genome Biol (2008). doi: 10.1186/gb-
2008-9-7-r108
19. Mazzone M, Bergers G. Regulation of Blood and Lymphatic Vessels by
Immune Cells in Tumors and Metastasis. Annu Rev Physiol (2019). doi:
10.1146/annurev-physiol-020518-114721
20. Osogoe B, Courtice FC. The effects of occlusion of the blood supply to the
popliteal lymph node of the rabbit on the cell and protein content of the
lymph and on the histology of the node. Aust J Exp Biol Med Sci (1968). doi:
10.1038/icb.1968.159
21. Lee Y-e, Koh GY. Coordinated lymphangiogenesis is critical in lymph node
development and maturation. Dev Dyn (2016). doi: 10.1002/dvdy.24456
22. Vondenhoff MF, van de Pavert SA, Dillard ME, Greuter M, Goverse G,
Oliver G, et al. Lymph sacs are not required for the initiation of lymph node
formation. Development (2009). doi: 10.1242/dev.028456
23. Bovay E, Sabine A, Prat-Luri B, Kim S, Son K, Willrodt AH, et al. Multiple
roles of lymphatic vessels in peripheral lymph node development. J Exp Med
(2018). doi: 10.1084/jem.20180217
24. Lai L, Bohnsack BL, Niederreither K, Hirschi KK. Retinoic acid regulates
endothelial cell proliferation during vasculogenesis. Development (2003).
doi: 10.1242/dev.00887
25. Van De Pavert SA, Olivier BJ, Goverse G, Vondenhoff MF, Greuter M, Beke
P, et al. Chemokine cxcl13 is essential for lymph node initiation and is
induced by retinoic acid and neuronal stimulation. Nat Immunol (2009). doi:
10.1038/ni.1789
26. Mebius RE, Streeter PR, Michie S, Butcher EC, Weissman IL. A
developmental switch in lymphocyte homing receptor and endothelial
vascular addressin expression regulates lymphocyte homing and permits
CD4+CD3- cells to colonize lymph nodes. Proc Natl Acad Sci U S A (1996).
doi: 10.1073/pnas.93.20.11019
27. Mebius RE, Rennert P, Weissman IL. Developing lymph nodes collect CD4
+CD3- LTb+ cells that can differentiate to APC, NK cells, and follicular cells
but not T or B cells. Immunity (1997). doi: 10.1016/S1074-7613(00)80371-4
28. Mebius RE, Dowbenko D, Williams A, Fennie C, Lasky LA, Watson SR.
Expression of GlyCAM-1, an endothelial ligand for L-selectin, is affected by
afferent lymphatic ow. J Immunol (1993).
29. Anderson AO, Anderson ND. Studies on the structure and permeability of
the microvasculature in normal rat lymph nodes. Am J Pathol (1975).
30. Herman PG, Yamamoto I, Mellins HZ. Blood microcirculation in the lymph
node during the primary immune response. J Exp Med (1972). doi: 10.1097/
00004424-197209000-00038
31. Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR. Vessel
co-option in cancer. Nat Rev Clin Oncol (2019). doi: 10.1038/s41571-019-
0181-9
32. Krishnan L, Chang CC, Nunes SS, Williams SK, Weiss JA, Hoying JB.
Manipulating the microvasculature and its microenvironment. Crit Rev
BioMed Eng (2013). doi: 10.1615/CritRevBiomedEng.2013008077
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174116
33. Lamalice L, Le Boeuf F, Huot J. Endothelial cell migration during
angiogenesis. Circ Res (2007). doi: 10.1161/01.RES.0000259593.07661.1e
34. Hellström M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, Lindblom P,
et al. Dll4 signalling through Notch1 regulates formation of tip cells during
angiogenesis. Nature (2007). doi: 10.1038/nature05571
35. Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, Abramsson
A, et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell
lopodia. J Cell Biol (2003). doi: 10.1083/jcb.200302047
36. Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of
angiogenesis. Cell (2011). doi: 10.1016/j.cell.2011.08.039
37. Akeson A, Herman A, Wiginton D, Greenberg J. Endothelial cell activation
in a VEGF-A gradient: Relevance to cell fate decisions. Microvasc Res (2010).
doi: 10.1016/j.mvr.2010.02.001
38. Bautch VL. VEGF-directed blood vessel patterning: From cells to organism.
Cold Spring Harb Perspect Med (2012). doi: 10.1101/cshperspect.a006452
39. Zhang L, Zhou F, Han W, Shen B, Luo J, Shibuya M, et al. VEGFR-3 ligand-
binding and kinase activity are required for lymphangiogenesis but not for
angiogenesis. Cell Res (2010). doi: 10.1038/cr.2010.116
40. Matsumoto M, Roufail S, Inder R, Caesar C, Karnezis T, Shayan R, et al.
Signaling for lymphangiogenesis via VEGFR-3 is required for the early
events of metastasis. Clin Exp Metastasis (2013). doi: 10.1007/s10585-013-
9581-x
41. Tammela T, Zarkada G, Wallgard E, Murtomäki A, Suchting S, Wirzenius
M, et al. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular
network formation. Nature (2008). doi: 10.1038/nature07083
42. Gloger M, Menzel L, Grau M, Vion A-C, Anagnostopoulos I, Zapukhlyak M,
et al. Lymphoma Angiogenesis Is Orchestrated by Noncanonical Signaling
Pathways. Cancer Res (2020). doi: 10.1158/0008-5472.CAN-19-1493
43. Fahmy RG, Dass CR, Sun LQ, Chesterman CN, Khachigian LM.
Transcription factor Egr-1 supports FGF-dependent angiogenesis during
neovascularization and tumor growth. Nat Med (2003). doi: 10.1038/nm905
44. Yu P, Wilhelm K, Dubrac A, Tung JK, Alves TC, Fang JS, et al. FGF-
dependent metabolic control of vascular development. Nature (2017). doi:
10.1016/j.jvs.2017.07.055
45. Turner N, Grose R. Fibroblast growth factor signalling: From development
to cancer. Nat Rev Cancer (2010). doi: 10.1038/nrc2780
46. Kroon ME, Koolwijk P, Van Der Vecht B, Van Hinsbergh VWM. Hypoxia in
combination with FGF-2 induces tube formation by human microvascular
endothelial cells in a brin matrix: Involvement of at least two signal
transduction pathways. J Cell Sci (2001).
47. Tsuda S, Ohtsuru A, Yamashita S, Kanetake H, Kanda S. Role of c-Fyn in
FGF-2-mediated tube-like structure formation by murine brain capillary
endothelial cells. Biochem Biophys Res Commun (2002). doi: 10.1006/
bbrc.2002.6345
48. Susaki EA, Tainaka K, Perrin D, Kishino F, Tawara T, Watanabe TM, et al.
Whole-brain imaging with single-cell resolution using chemical cocktails
and computational analysis. Cell (2014). doi: 10.1016/j.cell.2014.03.042
49. Susaki EA, Tainaka K, Perrin D, Yukinaga H, Kuno A, Ueda HR. Advanced
CUBIC protocols for whole-brain and whole-body clearing and imaging.
Nat Protoc (2015). doi: 10.1038/nprot.2015.085
50. Kumar V, Scandella E, Danuser R, Onder L, NitschkeM, Fukui Y, et al.
Global lymphoid tissue remodeling during a viral infection is orchestrated by
a B cell-lymphotoxin-dependent pathway. Blood (2010). doi: 10.1182/blood-
2009-10-250118
51. Mayer J, Swoger J, Ozga AJ, Stein JV, Sharpe J. Quantitative measurements
in 3-dimensional datasets of mouse lymph nodes resolve organ-wide
functional dependencies. Comput Math Methods Med (2012). doi:
10.1155/2012/128431
52. Abe J, Ozga AJ, Swoger J, Sharpe J, Ripoll J, Stein JV. Light sheet uorescence
microscopy for in situ cell interaction analysis in mouse lymph nodes. J
Immunol Methods (2016). doi: 10.1016/j.jim.2016.01.015
53. Jafarnejad M, Woodruff MC, Zawieja DC, Carroll MC, Moore JE. Modeling
lymph ow and uid exchange with blood vessels in lymph nodes. Lymphat
Res Biol (2015). doi: 10.1089/lrb.2015.0028
54. Jafarnejad M, Ismail AZ, Duarte D, Vyas C, Ghahramani A, Zawieja DC,
et al. Quantication of the Whole Lymph Node Vasculature Based on
Tomography of the Vessel Corrosion Casts. Sci Rep (2019). doi: 10.1038/
s41598-019-49055-7
55. Tomei AA, Siegert S, Britschgi MR, Luther SA, Swartz MA. Fluid Flow
Regulates Stromal Cell Organization and CCL21 Expression in a Tissue-
Engineered Lymph Node Microenvironment. J Immunol (2009). doi:
10.4049/jimmunol.0900835
56. Chang JE, Buechler MB, Gressier E, Turley SJ, Carroll MC.
Mechanosensing by Peyers patch stroma regulates lymphocyte
migration and mucosal antibody responses. Nat Immunol (2019). doi:
10.1038/s41590-019-0505-z
57. Pajor K, Pap T, Csanaky G, Kalasz V. High endothelial venules in B-cell non-
Hodgkin malignant lymphomas. JPathol(1990). doi: 10.1002/
path.1711610213
58. Kelch ID, Bogle G, Sands GB, Phillips ARJ, Legrice IJ, Rod Dunbar P. Organ-
wide 3D-imaging and topological analysis of the continuous microvascular
network in a murine lymph node. Sci Rep (2015). doi: 10.1038/srep16534
59. Kalucka J, de Rooij LPMH, Goveia J, Rohlenova K, Dumas SJ, Meta E, et al.
Single-Cell Transcriptome Atlas of Murine Endothelial Cells. Cell (2020).
doi: 10.1016/j.cell.2020.01.015
60. Brulois K, Rajaraman A, Szade A, Nordling S, Bogoslowski A, Dermadi D,
et al. A molecular map of murine lymph node blood vascular endothelium at
single cell resolution. Nat Commun (2020) 11(1):3798. doi: 10.1038/s41467-
020-17291-5
61. Wood HB, May G, Healy L, Enver T, Morriss-Kay GM. Cd34 expression
patterns during early mouse development are related to modes of blood
vessel formation and reveal additional sites of hematopoiesis. Blood (1997).
doi: 10.1182/blood.V90.6.2300.2300_2300_2311
62. Greten FR, Grivennikov SI. Inammation and Cancer: Triggers,
Mechanisms, and Consequences. Immunity (2019). doi: 10.1016/
j.immuni.2019.06.025
63. Oladipupo S, Hu S, Kovalski J, Yao J, Santeford A, Sohn RE, et al. VEGF is
essential for hypoxia-inducible factor-mediated neovascularization but
dispensable for endothelial sprouting. Proc Natl Acad Sci U S A (2011).
doi: 10.1073/pnas.1101321108
64. Hitchon C, Wong K, Ma G, Reed J, Lyttle D, El-Gabalawy H. Hypoxia-
induced production of stromal cell-derived factor 1 (CXCL12) and vascular
endothelial growth factor by synovial broblasts. Arthritis Rheumatol
(2002). doi: 10.1002/art.10520
65. Semenza GL. Cancer-stromal cell interactions mediated by hypoxia-
inducible factors promote angiogenesis, lymphangiogenesis, and
metastasis. Oncogene (2013). doi: 10.1038/onc.2012.578
66. McDonald DM, Baluk P. Imaging of angiogenesis in inamed airways and
tumors: Newly formed blood vessels are not alike and may be wildly
abnormal. Parker B. Francis lecture. Chest (2005). doi: 10.1378/
chest.128.6_suppl.602S-a
67. Siemann DW. The unique characteristics of tumor vasculature and
preclinical evidence for its selective disruption by Tumor-Vascular
Disrupting Agents. Cancer Treat Rev (2011). doi: 10.1016/j.ctrv.2010.
05.001
68. Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G, Roberge S,
et al. Openings between defective endothelial cells explain tumor vessel
leakiness. Am J Pathol (2000). doi: 10.1016/S0002-9440(10)65006-7
69. Morikawa S, Baluk P, Kaidoh T, Haskell A, Jain RK, McDonald DM.
Abnormalities in pericytes on blood vessels and endothelial sprouts in
tumors. Am J Pathol (2002). doi: 10.1016/S0002-9440(10)64920-6
70. Baluk P, Morikawa S, Haskell A, Mancuso M, McDonald DM.
Abnormalities of Basement Membrane on Blood Vessels and Endothelial
Sprouts in Tumors. Am J Pathol (2003). doi: 10.1016/S0002-9440(10)
63540-7
71. Dirkx AEM, Oude Egbrink MGA, Kuijpers MJE, Van der Niet ST, Heijnen
VVT, Bouma-ter Steege JCA, et al. Tumor angiogenesis modulates
leukocyte-vessel wall interactions in Vivo by reducing endothelial
adhesion molecule expression. Cancer Res (2003).
72. Huang H, Langenkamp E, Georganaki M, Loskog A, Fuchs PF, Dieterich LC,
et al. VEGF suppresses T-lymphocyte inltration in the tumor
microenvironment through inhibition of NF-kB-induced endothelial
activation. FASEB J (2015). doi: 10.1096/fj.14-250985
73. Mondor I, Jorquera A, Sene C, Adriouch S, Adams RH, Zhou B, et al. Clonal
Proliferation and Stochastic Pruning Orchestrate Lymph Node Vasculature
Remodeling. Immunity (2016). doi: 10.1016/j.immuni.2016.09.017
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174117
74. Martinez VG, Pankova V, Krasny L, Singh T, Makris S, White IJ, et al.
Fibroblastic Reticular Cells Control Conduit Matrix Deposition during
Lymph Node Expansion. Cell Rep (2019). doi: 10.1016/j.celrep.
2019.10.103
75. Anderson ND, Anderson AO, Wyllie RG. Microvascular changes in lymph
nodes draining skin allografts. Am J Pathol (1975).
76. Soderberg KA, Payne GW, Sato A, Medzhitov R, Segal SS, Iwasaki A. Innate
control of adaptive immunity via remodeling of lymnh node feed arteriole.
Proc Natl Acad Sci U S A (2005). doi: 10.1073/pnas.0506190102
77. Anderson AO, Anderson ND. Lymphocyte emigration from high
endothelial venules in rat lymph nodes. Immunol Commun (1976).
78. Tzeng T-C, Chyou S, Tian S, Webster B, Carpenter AC, Guaiquil VH, et al.
CD11c hi Dendritic Cells Regulate the Re-establishment of Vascular
Quiescence and Stabilization after Immune Stimulation of Lymph Nodes.
J Immunol (2010). doi: 10.4049/jimmunol.0902914
79. Chyou S, Benahmed F, Chen J, Kumar V, Tian S, Lipp M, et al. Coordinated
Regulation of Lymph Node VascularStromal Growth First by CD11c +
Cells and Then by T and B Cells. J Immunol (2011). doi: 10.4049/
jimmunol.1101724
80. Kumar V, Dasoveanu DC, Chyou S, Tzeng TC, Rozo C, Liang Y, et al. A
Dendritic-Cell-Stromal Axis Maintains Immune Responses in Lymph
Nodes. Immunity (2015). doi: 10.1016/j.immuni.2015.03.015
81. Kataru RP, Jung K, Jang C, Yang H, Schwendener RA, Jung EB, et al. Critical
role of CD11b+ macrophages and VEGF in inammatory lymphangiogenesis,
antigen clearance, and inammation resolution. Blood (2009). doi: 10.1182/
blood-2008-09-176776
82. Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, et al. Tissue
macrophages act as cellular chaperones for vascular anastomosis
downstream of VEGF-mediated endothelial tip cell induction. Blood
(2010). doi: 10.1182/blood-2009-12-257832
83. Benahmed F, Chyou S, Dasoveanu D, Chen J, Kumar V, Iwakura Y, et al.
Multiple CD11c + Cells Collaboratively Express IL-1bTo Modulate Stromal
Vascular Endothelial Growth Factor and Lymph Node VascularStromal
Growth. J Immunol (2014). doi: 10.4049/jimmunol.1301765
84. White JR, Harris RA, Lee SR, Craigon MH, Binley K, Price T, et al. Genetic
amplication of the transcriptional response to hypoxia as a novel means of
identifying regulators of angiogenesis. Genomics (2004). doi: 10.1016/S0888-
7543(03)00215-5
85. Zajac E, Schweighofer B, Kupriyanova TA, Juncker-Jensen A, Minder P,
Quigley JP, et al. Angiogenic capacity of M1- and M2-polarized
macrophages is determined by the levels of TIMP-1 complexed with their
secreted proMMP-9. Blood (2013). doi: 10.1182/blood-2013-05-501494
86. Eubank TD, Galloway M, Montague CM, Waldman WJ, Marsh CB. M-CSF
Induces Vascular Endothelial Growth Factor Production and Angiogenic
Activity From Human Monocytes. JImmunol(2003). doi: 10.4049/
jimmunol.171.5.2637
87. Carmi Y, Voronov E, Dotan S, Lahat N, Rahat MA, Fogel M, et al. The Role
of Macrophage-Derived IL-1 in Induction and Maintenance of
Angiogenesis. J Immunol (2009). doi: 10.4049/jimmunol.0901511
88. Jetten N, Verbruggen S, Gijbels MJ, Post MJ, De Winther MPJ, Donners
MMPC. Anti-inammatory M2, but not pro-inammatory M1
macrophages promote angiogenesis in vivo.Angiogenesis (2014). doi:
10.1007/s10456-013-9381-6
89. Guo L, Akahori H, Harari E, Smith SL, Polavarapu R, Karmali V, et al.
CD163+ macrophages promote angiogenesis and vascular permeability
accompanied by inammation in atherosclerosis. J Clin Invest (2018). doi:
10.1172/JCI93025doi:10.1172/JCI93025
90. Abramsson A, Lindblom P, Betsholtz C. Endothelial and nonendothelial
sources of PDGF-B regulate pericyte recruitment and inuence vascular
patternformationintumors.JClinInvest(2003). doi: 10.1172/
JCI200318549
91.BirnerP,StöcklJ,KaltR,UllrichR,CaucigC.Tumor-associated
macrophages express lymphatic endothelial growth factors and are related
to peritumoral lymphangiogenesis. Am J Pathol (2002). doi: 10.1016/S0002-
9440(10)64255-1
92. Huang S. Contributions of Stromal Metalloproteinase-9 to Angiogenesis and
Growth of Human Ovarian Carcinoma in Mice. CancerSpectrum Knowl
Environ (2002). doi: 10.1093/jnci/94.15.1134
93. Lewis JS, Landers RJ, Underwood JCE, Harris AL, Lewis CE. Expression of
vascular endothelial growth factor by macrophages is up-regulated in poorly
vascularized areas of breast carcinomas. J Pathol (2000). doi: 10.1002/1096-
9896(2000)9999:9999::AID-PATH6873.0.CO;2-G
94. Tsutsui S, Yasuda K, Suzuki K, Tahara K, Higashi H, Era S. Macrophage
inltration and its prognostic implications in breast cancer: The relationship
with VEGF expression and microvessel density. Oncol Rep (2005). doi:
10.3892/or.14.2.425
95. Riboldi E, Musso T, Moroni E, Urbinati C, Bernasconi S, Rusnati M, et al.
Proangiogenic Properties of Alternatively Activated Dendritic Cells.
J Immunol (2005). doi: 10.4049/jimmunol.175.5.2788
96. Webster B, Ekland EH, Agle LM, Chyou S, Ruggieri R, Lu TT. Regulation of
lymph node vascular growth by dendritic cells. J Exp Med (2006). doi:
10.1084/jem.20052272
97. Salvi V, Vaira X, Gianello V, Vermi W, Bugatti M, Sozzani S, et al. TLR
Signalling Pathways Diverge in Their Ability to Induce PGE2. Mediators
Inammation (2016). doi: 10.1155/2016/5678046
98. Browning JL, Allaire N, Ngam-Ek A, Notidis E, Hunt J, Perrin S, et al.
Lymphotoxin-breceptor signaling is required for the homeostatic control of
HEV differentiation and function. Immunity (2005). doi: 10.1016/
j.immuni.2005.10.002
99. Moussion C, Girard JP. Dendritic cells control lymphocyte entry to lymph
nodes through high endothelial venules. Nature (2011). doi: 10.1038/
nature10540
100. Onder L, Danuser R, Scandella E, Firner S, Chai Q, Hehlgans T, et al.
Endothelial cellspecic lymphotoxin-breceptor signaling is critical for
lymph node and high endothelial venule formation. J Exp Med (2013). doi:
10.1084/jem.20121462
101. Gagliostro V, Seeger P, Garrafa E, Salvi V, Bresciani R, Bosisio D, et al. Pro-
lymphangiogenic properties of IFN-g-activated human dendritic cells.
Immunol Lett (2016). doi: 10.1016/j.imlet.2016.03.008
102. Konno S, Eckman JA, Plunkett B, Li X, Berman JS, Schroeder J, et al.
Interleukin-10 and Th2 cytokines differentially regulate osteopontin
expression in human monocytes and dendritic cells. J Interf Cytokine Res
(2006). doi: 10.1089/jir.2006.26.562
103. Ohki Y, Heissig B, Sato Y, Akiyama H, Zhu Z, Hicklin DJ, et al. Granulocyte
colony-stimulating factor promotes neovascularization by releasing vascular
endothelial growth factor from neutrophils. FASEB J (2005). doi: 10.1096/
fj.04-3496fje
104. Schruefer R, Lutze N, Schymeinsky J, Walzog B. Human neutrophils
promote angiogenesis by a paracrine feedforward mechanism involving
endothelial interleukin-8. Am J Physiol Heart Circ Physiol (2005). doi:
10.1152/ajpheart.00237.2004
105. Gaudry M, Bregerie O, Andrieu V, El Benna J, Pocidalo MA, Hakim J.
Intracellular pool of vascular endothelial growth factor in human
neutrophils. Blood (1997). doi: 10.1182/blood.V90.10.4153
106. McHale C, Mohammed Z, Gomez G. Human skin-derived mast cells
spontaneously secrete several angiogenesis-related factors. Front Immunol
(2019). doi: 10.3389/mmu.2019.01445
107. Tanaka A, Arai K, Kitamura Y, Matsuda H. Matrix metalloproteinase-9
production, a newly identied function of mast cell progenitors, is
downregulated by c-kit receptor activation. Blood (1999). doi: 10.1182/
blood.v94.7.2390.419k16_2390_2395
108. Detoraki A, Staiano RI, Granata F, Giannattasio G, Prevete N, de Paulis A,
et al. Vascular endothelial growth factors synthesized by human lung mast
cells exert angiogenic effects. J Allergy Clin Immunol (2009). doi: 10.1016/
j.jaci.2009.01.044
109. Melillo RM, Guarino V, Avilla E, Galdiero MR, Liotti F, Prevete N, et al. Mast
cells have a protumorigenic role in human thyroid cancer. Oncogene (2010).
doi: 10.1038/onc.2010.348
110. Shojaei F, Wu X, Qu X, Kowanetz M, Yu L, Tan M, et al. G-CSF-initiated
myeloid cell mobilization and angiogenesis mediate tumor refractoriness to
anti-VEGF therapy in mouse models. Proc Natl Acad Sci U S A (2009). doi:
10.1073/pnas.0902280106
111. Iannone R, Miele L, Maiolino P, Pinto A, Morello S. Blockade of A2b
adenosine receptor reduces tumor growth and immune suppression
mediated by myeloid-derived suppressor cells in a mouse model of
melanoma. Neoplasia (United States) (2013). doi: 10.1593/neo.131748
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174118
112. Horikawa N, Abiko K, Matsumura N, Hamanishi J, Baba T, Yamaguchi K,
et al. Expression of vascular endothelial growth factor in ovarian cancer
inhibits tumor immunity through the accumulation of myeloid-derived
suppressor cells. Clin Cancer Res (2017). doi: 10.1158/1078-0432.CCR-16-
0387
113. Binsfeld M, Muller J, Lamour V, De Veirman K, De Raeve H, Bellahcène A,
et al. Granulocytic myeloid-derived suppressor cells promote angiogenesis in
the context of multiple myeloma. Oncotarget (2016). doi: 10.18632/
oncotarget.9270
114. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, et al.
Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-
bearing host directly promotes tumor angiogenesis. Cancer Cell (2004).
doi: 10.1016/j.ccr.2004.08.031
115. Yang CY, Vogt TK, Favre S, Scarpellino L, Huang HY, Tacchini-Cottier F,
et al. Trapping of naive lymphocytes triggers rapid growth and remodeling of
the broblast network in reactive murine lymph nodes. Proc Natl Acad Sci
USA(2014). doi: 10.1073/pnas.1312585111
116. Kwee BJ, Budina E, Najibi AJ, Mooney DJ. CD4 T-cells regulate angiogenesis
and myogenesis. Biomaterials (2018). doi: 10.1016/j.biomaterials.
2018.06.003
117. Freeman MR, Schneck FX, Niknejad K, Gagnon ML, Soker S, Klagsbrun M,
et al. Peripheral Blood T Lymphocytes and Lymphocytes Inltrating Human
Cancers Express Vascular Endothelial Growth Factor: A Potential Role for T
Cells in Angiogenesis. Cancer Res (1995).
118. Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, et al.
Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T reg
cells. Nature (2011). doi: 10.1158/1538-7445.AM2012-308
119. Leung OM, Li J, Li X, Chan VW, Yang KY, Ku M, et al. Regulatory T Cells
Promote Apelin-Mediated Sprouting Angiogenesis in Type 2 Diabetes. Cell
Rep (2018). doi: 10.1016/j.celrep.2018.07.019
120. Hur J, Yang HM, Yoon CH, Lee CS, Park KW, Kim JH, et al. Identication of
a novel role of T cells in postnatal vasculogenesis: Characterization of
endothelial progenitor cell colonies. Circulation (2007). doi: 10.1161/
CIRCULATIONAHA.107.694778
121. Manetti M, Pratesi S, Romano E, Bellando-Randone S, Rosa I, Guiducci S,
et al. Angiogenic T cell expansion correlates with severity of peripheral
vascular damage in systemic sclerosis. PLoS One (2017). doi: 10.1371/
journal.pone.0183102
122. Shrestha B, Hashiguchi T, Ito T, Miura N, Takenouchi K, Oyama Y, et al. B
Cell-Derived Vascular Endothelial Growth Factor A Promotes
Lymphangiogenesis and High Endothelial Venule Expansion in Lymph
Nodes. J Immunol (2010). doi: 10.4049/jimmunol.0903063
123. van de Veen W, Globinska A, Jansen K, Straumann A, Kubo T, Verschoor D,
et al. A novel proangiogenic B cell subset is increased in cancer and chronic
inammation. Sci Adv (2020). doi: 10.1126/sciadv.aaz3559
124. Missiaen R, Mazzone M, Bergers G. The reciprocal function and regulation
of tumor vessels and immune cells offers new therapeutic opportunities in
cancer. Semin Cancer Biol (2018). doi: 10.1016/j.semcancer.2018.06.002
125. Hua Y, Bergers G. Tumors vs. Chronic Wounds: An Immune Cells
Perspective. Front Immunol (2019). doi: 10.3389/mmu.2019.02178
126. Lewis CE, Harney AS, Pollard JW. The Multifaceted Role of Perivascular
Macrophages in Tumors. Cancer Cell (2016). doi: 10.1016/j.ccell.2016.07.009
127. Gray EE, Cyster JG. Lymph node macrophages. J Innate Immun (2012). doi:
10.1159/000337007
128. Gregory JL, Morand EF, McKeown SJ, Ralph JA, Hall P, Yang YH, et al.
Macrophage Migration Inhibitory Factor Induces Macrophage Recruitment
via CC Chemokine Ligand 2. J Immunol (2006). doi: 10.4049/
jimmunol.177.11.8072
129. Argyle D, Kitamura T. Targeting macrophage-recruiting chemokines as a
novel therapeutic strategy to prevent the progression of solid tumors. Front
Immunol (2018). doi: 10.3389/mmu.2018.02629
130. Reinart N, Nguyen PH, Boucas J, Rosen N, Kvasnicka HM, Heukamp L, et al.
Delayed development of chronic lymphocytic leukemia in the absence of
macrophage migration inhibitory factor. Blood (2013). doi: 10.1182/blood-
2012-05-431452
131. Hanna BS, Öztürk S, Seiffert M. Beyond bystanders: Myeloid cells in chronic
lymphocytic leukemia. Mol Immunol (2019). doi: 10.1016/j.molimm.
2017.11.014
132. Haderk F, Schulz R, Iskar M, Cid LL, Worst T, Willmund KV, et al. Tumor-
derived exosomes modulate PD-L1 expression in monocytes. Sci Immunol
(2017). doi: 10.1126/sciimmunol.aah5509
133. Hanna BS, Mcclanahan F, Yazdanparast H, Zaborsky N, Kalter V, Rößner
PM, et al. Depletion of CLL-associated patrolling monocytes and
macrophagescontrolsdiseasedevelopmentandrepairsimmune
dysfunction in vivo.Leukemia (2016). doi: 10.1038/leu.2015.305
134. Carey CD, Gusenleitner D, Lipschitz M, Roemer MGM, Stack EC, Gjini E,
et al. Topological analysis reveals a PD-L1-associated microenvironmental
niche for Reed-Sternberg cells in Hodgkin lymphoma. Blood (2017). doi:
10.1182/blood-2017-03-770719
135. Roemer MGM, Advani RH, Ligon AH, Natkunam Y, Redd RA, Homer H,
et al. PD-L1 and PD-L2 genetic alterations dene classical hodgkin
lymphoma and predict outcome. JClinOncol(2016). doi: 10.1200/
JCO.2016.66.4482
136. Green MR, Rodig S, Juszczynski P, Ouyang J, Sinha P, ODonnell E, et al.
Constitutive AP-1 activity and EBV infection induce PD-l1 in Hodgkin
lymphomas and posttransplant lymphoproliferative disorders: Implications
for targeted therapy. Clin Cancer Res (2012). doi: 10.1158/1078-0432.CCR-
11-1942
137. Vari F, Arpon D, Keane C, Hertzberg MS, Talaulikar D, Jain S, et al. Immune
evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more
prominent in Hodgkin lymphoma than DLBCL. Blood (2018). doi: 10.1182/
blood-2017-07-796342
138. Clear AJ, Lee AM, Calaminici M, Ramsay AG, Morris KJ, Hallam S, et al.
Increased angiogenic sprouting in poor prognosis FL is associated with
elevated numbers of CD163+ macrophages within the immediate
sprouting microenvironment. Blood (2010). doi: 10.1182/blood-2009-11-
253260
139. Shen L, Li H, Shi Y, Wang D, Gong J, Xun J, et al. M2 tumour-associated
macrophages contribute to tumour progression via legumain remodelling
the extracellular matrix in diffuse large B cell lymphoma. Sci Rep (2016). doi:
10.1038/srep30347
140. Murray PJ. Macrophage Polarization. Annu Rev Physiol (2017). doi: 10.1146/
annurev-physiol-022516-034339
141. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al.
Macrophage Activation and Polarization: Nomenclature and Experimental
Guidelines. Immunity (2014). doi: 10.1016/j.immuni.2014.07.009
142. De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of
tumour angiogenesis. Nat Rev Cancer (2017). doi: 10.1038/nrc.2017.51
143. Barnett FH, Rosenfeld M, Wood M, Kiosses WB, Usui Y, Marchetti V, et al.
Macrophages form functional vascular mimicry channels in vivo.Sci Rep
(2016). doi: 10.1038/srep36659
144. Schmeisser A, Garlichs CD, Zhang H, EskaS, Graffy C, Ludwig J, et al.
Monocytes coexpress endothelial and macrophagocytic lineage markers and
form cord-like structures in Matrigel®under angiogenic conditions.
Cardiovasc Res (2001). doi: 10.1016/S0008-6363(00)00270-4
145. Lewis CE, De Palma M, Naldini L. Tie2-expressing monocytes and tumor
angiogenesis: Regulation by hypoxia and angiopoietin-2. Cancer Res (2007).
doi: 10.1158/0008-5472.CAN-07-1684
146. He H, Mack JJ, GücE, Warren CM, Squadrito ML, Kilarski WW, et al.
Perivascular Macrophages Limit Permeability. Arterioscler Thromb Vasc Biol
(2016). doi: 10.1161/ATVBAHA.116.307592
147. Acton SE, Reis e Sousa C. Dendritic cells in remodeling of lymph
nodes during immune responses. Immunol Rev (2016). doi: 10.1111/
imr.12414
148. Bajenoff M, Granjeaud S, Guerder S. The strategy of T cell antigen-presenting
cell encounter in antigen-draining lymph nodes revealed by imaging of initial
T cell activation. J Exp Med (2003). doi: 10.1084/jem.20030167
149. Scholz F, Grau M, Menzel L, Graband A, Zapukhlyak M, Leutz A, et al. The
transcription factor C/EBPborchestrates dendritic cell maturation and
functionality under homeostatic and malignant conditions. Proc Natl Acad
Sci (2020). http://www.pnas.org/content/early/2020/09/30/2008883117.
abstract.
150. Rehm A, Gätjen M, Gerlach K, Scholz F, Mensen A, Gloger M, et al.
Dendritic cell-mediated survival signals in EI 1/4-Myc B-cell lymphoma
depend on the transcription factor C/EBPI 2. Nat Commun (2014). doi:
10.1038/ncomms6057
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174119
151. Kocieda VP, Adhikary S, Emig F, Yen JH, Toscano MG, Ganea D.
Prostaglandin E2-induced IL-23p19 subunit is regulated by cAMP-
responsive element-binding protein and C/AATT enhancer-binding
protein bin bone marrow-derived dendritic cells. J Biol Chem (2012). doi:
10.1074/jbc.M112.402958
152. Salvi V, Vermi W, Gianello V, Lonardi S, Gagliostro V, Naldini A, et al.
Dendritic cell-derived VEGF-A plays a role in inammatory angiogenesis of
human secondary lymphoid organs and is driven by the coordinated
activation of multiple transcription factors. Oncotarget (2016). doi:
10.18632/oncotarget.9684
153. Fainaru O, Adini A, Benny O, Adini I, Short S, Bazinet L, et al. Dendritic cells
support angiogenesis and promote lesion growth in a murine model of
endometriosis. FASEB J (2008). doi: 10.1096/fj.07-9034com
154. Sozzani S, Rusnati M, Riboldi E, Mitola S, Presta M. Dendritic cell-
endothelial cell cross-talk in angiogenesis. Trends Immunol (2007). doi:
10.1016/j.it.2007.07.006
155. Scimone ML, Lutzky VP, Zittermann SI, Mafa P, Jancic C, Buzzola F, et al.
Migration of polymorphonuclear leucocytes is inuenced by dendritic cells.
Immunology (2005). doi: 10.1111/j.1365-2567.2005.02104.x
156. Hampton HR, Chtanova T. The lymph node neutrophil. Semin Immunol
(2016). doi: 10.1016/j.smim.2016.03.008
157. Wang HW, Tedla N, Lloyd AR, Wakeeld D, McNeil HP. Mast cell
activation and migration to lymph nodes during induction of an immune
response in mice. J Clin Invest (1998). doi: 10.1172/JCI3704
158. Schruefer R, Sulyok S, Schymeinsky J, Peters T, Scharffetter-Kochanek K, Walzog
B. The proangiogenic capacity of polymorphonuclear neutrophils delineated
by microarray technique and by measurement of neovascularization
in wounded skin of CD18-decient mice. JVascRes(2005). doi: 10.1159/
000088975
159. Ancelin M, Chollet-Martin S, HerveMA, Legrand C, El Benna J, Perrot-
Applanat M. Vascular endothelial growth factor VEGF189 induces human
neutrophil chemotaxis in extravascular tissue via an autocrine amplication
mechanism. Lab Investig (2004). doi: 10.1038/labinvest.3700053
160. Gätjen M, Brand F, Grau M, Gerlach K, Kettritz R, Westermann J, et al.
Splenic marginal zone granulocytes acquire an accentuated neutrophil B-cell
helper phenotype in chronic lymphocytic leukemia. Cancer Res (2016). doi:
10.1158/0008-5472.CAN-15-3486
161. Rabenhorst A, Schlaak M, Heukamp LC, Förster A, Theurich S, Von
Bergwelt-Baildon M, et al. Mast cells play a protumorigenic role in
primary cutaneous lymphoma. Blood (2012). doi: 10.1182/blood-2012-03-
415638
162. Vyzoukaki R, Tsirakis G, Pappa CA, Devetzoglou M, Tzardi M, Alexandrakis
MG. The Impact of Mast Cell Density on the Progression of Bone Disease in
Multiple Myeloma Patients. Int Arch Allergy Immunol (2016). doi: 10.1159/
000443275
163. Franco G, Guarnotta C, Frossi B, Piccaluga PP, Boveri E, Gulino A, et al.
Bone marrow stroma CD40 expression correlates with inammatory mast
cell inltration and disease progression in splenic marginal zone lymphoma.
Blood (2014). doi: 10.1182/blood-2013-04-497271
164. Bradding P, Okayama Y, Howarth PH, Church MK, Holgate ST.
Heterogeneity of human mast cells based on cytokine content. J Immunol
(1995).
165. Irani AA, Schechter NM, Craig SS, DeBlois G, Schwartz LB. Two types of
human mast cells that have distinct neutral protease compositions. Proc Natl
Acad Sci U S A (1986). doi: 10.1073/pnas.83.12.4464
166. Ronca R, Tamma R, Coltrini D, Ruggieri S, Presta M, Ribatti D. Fibroblast
growth factor modulates mast cell recruitment in a murine model of prostate
cancer. Oncotarget (2017). doi: 10.18632/oncotarget.19773
167. Varricchi G, Granata F, Loffredo S, Genovese A, Marone G. Angiogenesis
and lymphangiogenesis in inammatory skin disorders. J Am Acad Dermatol
(2015). doi: 10.1016/j.jaad.2015.03.041
168. Ostrand-Rosenberg S, Sinha P. Myeloid-Derived Suppressor Cells: Linking
Inammation and Cancer. J Immunol (2009). doi: 10.4049/
jimmunol.0802740
169. Hanson EM, Clements VK, Sinha P, Ilkovitch D, Ostrand-Rosenberg S.
Myeloid-Derived Suppressor Cells Down-Regulate L-Selectin Expression on
CD4 + and CD8 + T Cells. J Immunol (2009). doi: 10.4049/
jimmunol.0804253
170. Parker KH, Sinha P, Horn LA, Clements VK, Yang H, Li J, et al. HMGB1
enhances immune suppression by facilitating the differentiation and
suppressive activity of myeloid-derived suppressor cells. Cancer Res (2014).
doi: 10.1158/0008-5472.CAN-13-2347
171. MacDonald KPA, Rowe V, Clouston AD, Welply JK, Kuns RD, Ferrara JLM,
et al. Cytokine Expanded Myeloid Precursors Function as Regulatory
Antigen-Presenting Cells and Promote Tolerance through IL-10-Producing
Regulatory T Cells. J Immunol (2005). doi: 10.4049/jimmunol.174.4.1841
172. Serani P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived suppressor
cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T
cells. Cancer Res (2008). doi: 10.1158/0008-5472.CAN-07-6621
173. Alkasalias T, Moyano-Galceran L, Arsenian-Henriksson M, Lehti K.
Fibroblasts in the tumor microenvironment: Shield or spear? Int J Mol Sci
(2018). doi: 10.3390/ijms19051532
174. Gunes EG, Rosen ST, Querfeld C. The role of myeloid-derived suppressor
cells in hematologic malignancies. Curr Opin Oncol (2020). doi: 10.1097/
CCO.0000000000000662
175. Bruno A, Mortara L, Baci D, Noonan DM, Albini A. Myeloid Derived
Suppressor Cells Interactions With Natural Killer Cells and Pro-angiogenic
Activities: Roles in Tumor Progression. Front Immunol (2019). doi: 10.3389/
mmu.2019.00771
176. Serani P, Carbley R, Noonan KA, Tan G, Bronte V, Borrello I. High-dose
granulocyte-macrophage colony-stimulating factor-producing vaccines
impair the immune response through the recruitment of myeloid
suppressor cells. Cancer Res (2004). doi: 10.1158/0008-5472.CAN-04-0757
177. Huang B, Lei Z, Zhao J, Gong W, Liu J, Chen Z, et al. CCL2/CCR2 pathway
mediates recruitment of myeloid suppressor cells to cancers. Cancer Lett
(2007). doi: 10.1016/j.canlet.2006.12.012
178. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of
the immune system. Nat Rev Immunol (2009). doi: 10.1038/nri2506
179. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in
the promotion of tumour angiogenesis. Nat Rev Cancer (2008). doi: 10.1038/
nrc2444
180. Sorrentino C, Miele L, Porta A, Pinto A, Morello S. Myeloid-derived
suppressor cells contribute to A2B adenosine receptor-induced VEGF
production and angiogenesis in a mouse melanoma model. Oncotarget
(2015). doi: 10.18632/oncotarget.4393
181. Ferrara N. Role of myeloid cells in vascular endothelial growth factor-
independent tumor angiogenesis. Curr Opin Hematol (2010). doi: 10.1097/
MOH.0b013e3283386660
182. Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-
angiogenic treatments and vice versa. Nat Rev Clin Oncol (2018). doi:
10.1038/nrclinonc.2018.9
183. Kumar BV, Connors TJ, FarberDL.HumanTCellDevelopment,
Localization, and Function throughout Life. Immunity (2018). doi:
10.1016/j.immuni.2018.01.007
184. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy:
from T cell basic science to clinical practice. Nat Rev Immunol (2020). doi:
10.1038/s41577-020-0306-5
185. McGranahan N, Furness AJS, Rosenthal R, Ramskov S, Lyngaa R, Saini SK,
et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to
immune checkpoint blockade. Science (2016). doi: 10.1126/science.aaf1490
186. Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, et al. Clonal
replacement of tumor-specic T cells following PD-1 blockade. Nat Med
(2019). doi: 10.1101/648899
187. Sasse S, Reddemann K, Diepstra A, Oschlies I, Schnitter A, Borchmann S,
et al. Programmed cell death protein-1 (PD-1)-expression in the
microenvironment of classical hodgkin lymphoma at relapse during
anti-PD-1-treatment. Haematologica (2019). doi: 10.3324/haematol.2018.
196279
188. Chen Y-B, Armand P, Redd RA, Bsat J, Merryman RW, Coleman K, et al.
PD-1 Blockade for Diffuse Large B-Cell Lymphoma after Autologous Stem
Cell Transplantation. Blood (2018). doi: 10.1182/blood-2018-99-114914
189. Ciciola P, Cascetta P, Bianco C, Formisano L, Bianco R. Combining Immune
Checkpoint Inhibitors with Anti-Angiogenic Agents. J Clin Med (2020). doi:
10.3390/jcm9030675
190. Eppihimer MJ, Gunn J, Freeman GJ, Greeneld EA, Chernova T, Erickson J,
et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174120
micro vascular endothelial cells. Microcirculation (2002). doi: 10.1080/
713774061
191. Mazanet MM, Hughes CCW. B7-H1 Is Expressed by Human Endothelial
Cells and Suppresses T Cell Cytokine Synthesis. J Immunol (2002). doi:
10.4049/jimmunol.169.7.3581
192. Cole JE, Navin TJ, Cross AJ, Goddard ME, Alexopoulou L, Mitra AT, et al.
Unexpected protective role for Toll-like receptor 3 in the arterial wall. Proc
Natl Acad Sci U S A (2011). doi: 10.1073/pnas.1018515108
193. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawk D, Kolhatkar N, et al.
CD4+ T Cells Regulate Pulmonary Metastasis of Mammary Carcinomas by
Enhancing Protumor Properties of Macrophages. Cancer Cell (2009). doi:
10.1016/j.ccr.2009.06.018
194. Hata T, Takahashi M, Hida S, Kawaguchi M, Kashima Y, Usui F, et al.
Critical role of Th17 cells in inammation and neovascularization after
ischaemia. Cardiovasc Res (2011). doi: 10.1093/cvr/cvq397
195. Hoechst B, Gamrekelashvili J, Manns MP, Greten TF, Korangy F. Plasticity
of human Th17 cells and iTregs is orchestrated by different subsets of
myeloid cells. Blood (2011). doi: 10.1182/blood-2010-11-317321
196. Ishida T, Ishii T, Inagaki A, Yano H, Komatsu H, Iida S, et al. Specic
recruitment of CC chemokine receptor 4-positive regulatory T cells in
Hodgkin lymphoma fosters immune privilege. Cancer Res (2006). doi:
10.1158/0008-5472.CAN-06-0261
197. Cavallo F, Quaglino E, Cifaldi L, Di Carlo E, AndreA, Bernabei P, et al.
Interleukin 12-activated lymphocytes inuence tumor genetic programs.
Cancer Res (2001).
198. Strasly M, Cavallo F, Geuna M, Mitola S, Colombo MP, Forni G, et al. IL-12
Inhibition of Endothelial Cell Functions and Angiogenesis Depends on
Lymphocyte-Endothelial Cell Cross-Talk. J Immunol (2001). doi: 10.4049/
jimmunol.166.6.3890
199. Lucas ED, Finlon JM, Burchill MA, McCarthy MK, Morrison TE, Colpitts
TM, et al. Type 1 IFN and PD-L1 Coordinate Lymphatic Endothelial Cell
Expansion and Contraction during an Inammatory Immune Response.
J Immunol (2018). doi: 10.4049/jimmunol.1800271
200. Lucas ED, Tamburini BAJ. Lymph node lymphatic endothelial cell expansion
and contraction and the programming of the immune response. Front
Immunol (2019). doi: 10.3389/mmu.2019.00036
201. Kammertoens T, Friese C, Arina A, Idel C, Briesemeister D, Rothe M, et al.
Tumour ischaemia by interferon-gresembles physiological blood vessel
regression. Nature (2017). doi: 10.1038/nature22311
202. Shafer-Weaver KA, Anderson MJ, Stagliano K, Malyguine A, Greenberg
NM, Hurwitz AA. Cutting Edge: Tumor-Specic CD8 + T Cells Inltrating
Prostatic Tumors Are Induced to Become Suppressor Cells. J Immunol
(2009). doi: 10.4049/jimmunol.0900848
203. Yu Y, Ma X, Gong R, Zhu J, Wei L, Yao J. Recent advances in CD8+
regulatory t cell research (Review). Oncol Lett (2018). doi: 10.3892/
ol.2018.8378
204. Kasakovski D, Zeng X, Xu L, Li Y. Increasing CD28-FOXP3+CD8+ Treg and
Senescent CD8+NK2GA+Eomes+ NK-like T Cells in Peripheral Blood of
Patients with Multiple Myeloma. Blood (2018). doi: 10.1182/blood-2018-99-
118088
205. Machicote A, Belen S, Baz P, Billordo LA, Fainboim L. Human CD8+HLA-
DR+Regulatory T Cells, similarly to classical CD4+Foxp3+cells, suppress
immune responses via PD-1/PD-L1 axis. Front Immunol (2018). doi:
10.3389/mmu.2018.02788
206. Manetti M, Pratesi S, Romano E, Bellando-Randone S, Rosa I, Guiducci S,
et al. Angiogenic T cell expansion correlates with severity of peripheral
vascular damage in systemic sclerosis. PloS One (2017). doi: 10.1136/
annrheumdis-2017-eular.3017
207. Igreja C, Courinha M, Cachaco AS, Pereira T, Cabecadas J, Da Silva MG,
et al. Characterization and clinical relevance of circulating and biopsy-
derived endothelial progenitor cells in lymphoma patients. Haematologica
(2007). doi: 10.3324/haematol.10723
208. Wiessman M, Leshem D, Yeshurun M, Yavin H, Iakobishvilli Z, Raanani P,
et al. Dysfunctional endothelial progenitor cells in patients with Hodgkins
lymphoma in complete remission. Cancer Med (2019). doi: 10.1002/
cam4.1914
209. Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, Chudakov
DM. B cells, plasma cells and antibody repertoires in the tumour
microenvironment. Nat Rev Immunol (2020). doi: 10.1038/s41577-019-
0257-x
210. Yang C, Lee H, Pal S, Jove V, Deng J, Zhang W, et al. B Cells Promote Tumor
Progression via STAT3 Regulated-Angiogenesis. PloS One (2013). doi:
10.1371/journal.pone.0064159
211. Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, et al. FcRg
Activation Regulates Inammation-Associated Squamous Carcinogenesis.
Cancer Cell (2010). doi: 10.1016/j.ccr.2009.12.019
212. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of
angiogenesis. Nature (2011). doi: 10.1038/nature10144
213. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes,
consequences, challenges and opportunities. Cell Mol Life Sci (2020). doi:
10.1007/s00018-019-03351-7
214. Ganjoo K, An C, Robertson M, Gordon L, Sen J, Weisenbach J, et al.
Rituximab, Bevacizumab and CHOP (RA-CHOP) in untreated diffuse large
B-cell lymphoma: Safety, biomarker and pharmacokinetic analysis. Leuk
Lymphoma (2006). doi: 10.1080/10428190600563821
215. Seymour JF, Pfreundschuh M, TrnenyM, Sehn LH, Catalano J, Csinady E,
et al. R-CHOP with or without bevacizumab in patients with previously
untreated diffuse large B-cell lymphoma: Final MAIN study outcomes.
Haematologica (2014). doi: 10.3324/haematol.2013.100818
216. Vacca A, Ribatti D, Roncali L, Dammacco F. Angiogenesis in b cell
lymphoproliferative diseases. Biological and clinical studies. Leuk
Lymphoma (1995). doi: 10.3109/10428199509054750
217. Jiang L, Li N. B-cell non-Hodgkin lymphoma: importance of angiogenesis
and antiangiogenic therapy. Angiogenesis (2020). doi: 10.1007/s10456-020-
09729-7
218. Hattori H. Caution should be taken in using CD31 for distinguishing the
vasculature of lymph nodes. J Clin Pathol (2003). doi: 10.1136/jcp.56.8.638-a
219. Rakocevic J, Orlic D, Mitrovic-Ajtic O, Tomasevic M, Dobric M, Zlatic N,
et al. Endothelial cell markers from clinicians perspective. Exp Mol Pathol
(2017). doi: 10.1016/j.yexmp.2017.02.005
220. Asabella AN, Di Palo A, Altini C, Ferrari C, Rubini G. Multimodality
imaging in tumor angiogenesis: Present status and perspectives. Int J Mol
Sci (2017). doi: 10.3390/ijms18091864
221. Yang J, Li W, He X, Zhang G, Yue L, Chai Y. VEGF overexpression is a
valuable prognostic factor for non-Hodgkins lymphoma evidence from a
systemic meta-analysis. Dis Markers (2015). doi: 10.1155/2015/786790
222. Paydas S, Seydaoglu G, Ergin M, Erdogan S, Yavuz S. The prognostic
signicance of VEGF-C and VEGF-A in non-Hodgkin lymphomas. Leuk
Lymphoma (2009). doi: 10.1080/10428190802706665
223. Koster A, Van Krieken JHJM, MacKenzie MA, Schraders M, Borm GF, Van
Der Laak JAWM, et al. Increased vascularization predicts favorable outcome
in follicular lymphoma. Clin Cancer Res (2005).
224. Chen H, Treweeke AT, West DC, Till KJ, Cawley JC, Zuzel M, et al. In vitro
and in vivo production of vascular endothelial growth factor by chronic
lymphocytic leukemia cells. Blood (2000). doi: 10.1182/blood.V96.9.
3181.h8003181_3181_3187
225. Salven P, Orpana A, Teerenhovi L, Joensuu H. Simultaneous elevation in the
serum concentrations of the angiogenic growth factors VEGF and bFGF is an
independent predictor of poor prognosis in non-Hodgkin lymphoma: A
single-institution study of 200 patients. Blood (2000). doi: 10.1182/
blood.V96.12.3712.h8003712_3712_3718
226. Wang ES, Teruya-Feldstein J, Wu Y, Zhu Z, Hicklin DJ, Moore MAS.
Targeting autocrine and paracrine VEGF receptor pathways inhibits human
lymphoma xenografts in vivo.Blood (2004). doi: 10.1182/blood-2004-01-
0226
227. Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R,
et al. Consensus guidelines for the use and interpretation of angiogenesis
assays. Angiogenesis (2018). doi: 10.1007/s10456-018-9613-x
228. Leukemia & Lymphoma Society. Mantle Cell Lymphoma Facts. Leuk
Lymphoma Soc (2014).
229. Maddocks K. Update on mantle cell lymphoma. Blood (2018). doi: 10.1182/
blood-2018-03-791392
230. Chiron D, Bellanger C, Papin A, Tessoulin B, Dousset C, Maiga S, et al.
Rational targeted therapies to overcome microenvironment-dependent
expansion of mantle cell lymphoma. Blood (2016). doi: 10.1182/blood-
2016-06-720490
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174121
231. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H TJ. WHO
Classication of Tumours of Haematopoietic and Lymphoid Tissues.in
WHO Classication of Tumours,Revised 4th Edition, vol. 2. (2017).
doi:10.1038/jhg.2010.42
232. Petrakis G, Veloza L, Clot G, Gine E, Gonzalez-Farre B, Navarro A, et al.
Increased tumour angiogenesis in SOX11-positive mantle cell lymphoma.
Histopathology (2019). doi: 10.1111/his.13935
233. Annese T, Ingravallo G, Tamma R, De Giorgis M, Maiorano E, Perrone T,
et al. Inammatory Inltrate and Angiogenesis in Mantle Cell Lymphoma.
Transl Oncol (2020). doi: 10.1016/j.tranon.2020.100744
234. Palomero J, Vegliante MC, Rodrıguez M, Planas E, Ribera I, Cid MC, et al.
SOX11 Regulates Angiogenesis In Mantle Cell Lymphoma. Blood (2013). doi:
10.1182/blood.V122.21.246.246
235. Palomero J, Vegliante MC, Rodrıguez ML, Eguileor A
, Castellano G, Planas-
Rigol E, et al. SOX11 promotes tumor angiogenesis through transcriptional
regulation of PDGFA in mantle cell lymphoma. Blood (2014). doi: 10.1182/
blood-2014-04-569566
236. Kuo PY, Jatiani SS, Rahman AH, Edwards D, Jiang Z, Ahr K, et al. Sox11
augments bcr signaling to drive mcl-like tumor development. Blood (2018).
doi: 10.1182/blood-2018-02-832535
237. Vegliante MC, Palomero J, Perez-Galan P, RoueG, Castellano G, Navarro A,
et al. SOX11 regulates PAX5 expression and blocks terminal B-cell
differentiation in aggressive mantle cell lymphoma. Blood (2013). doi:
10.1182/blood-2012-06-438937
238. Balsas P, Palomero J, Eguileor A
, Rodriguez ML, Vegliante MC,Planas-Rigol E,
et al. SOX11 promotes tumor protective microenvironment interactions
through CXCR4 and FAK regulation in mantle cell lymphoma. Blood (2017).
doi: 10.1182/blood-2017-04-776740
239. Koh YW, Shin SJ, Park C, Yoon DH, Suh C, Huh J. Absolute monocyte count
predicts overall survival in mantle cell lymphomas: Correlation with tumour-
associated macrophages. Hematol Oncol (2014). doi: 10.1002/hon.2106
240. Song K, Herzog BH, Sheng M, Fu J, McDaniel M, Ruan J, et al. Lenalidomide
inhibits lymphangiogenesis in preclinical models of mantle cell lymphoma.
Cancer Res (2013). doi: 10.1158/0008-5472.CAN-13-0750
241. Papin A, Tessoulin B, Bellanger C, Moreau A, Le Bris Y, Maisonneuve H,
et al. CSF1R and BTK inhibitions as novel strategies to disrupt the dialog
between mantle cell lymphoma and macrophages. Leukemia (2019). doi:
10.1038/s41375-019-0463-3
242. Ek S, Björck E, Högerkorp CM, Nordenskjöld M, Porwit-MacDonald A,
Borrebaeck CAK. Mantle cell lymphomas acquire increased expression of
CCL4, CCL5 and 4-1BB-L implicated in cell survival. Int J Cancer (2006).
doi: 10.1002/ijc.21579
243. Nygren L, Wasik AM, Baumgartner-Wennerholm S, Jeppsson-Ahlberg Å,
Klimkowska M, Andersson P, et al. T-cell levels are prognostic in mantle cell
lymphoma. Clin Cancer Res (2014). doi: 10.1158/1078-0432.CCR-14-0889
244. Ruan J, Coleman M, Furman RR, Glynn P, Joyce M, Ketas J, et al. Targeting
Angiogenesis in Mantle Cell Lymphoma: Clinical Efcacy and Correlative
Studies of a Phase II Trial of RT-PEPC (Rituximab, Thalidomide and
Metronomic Oral Chemotherapy with Prednisone, Etoposide,
Procarbazine and Cyclophosphamide) in Relapsed/Re. Blood (2006). doi:
10.1182/blood.V108.11.2751.2751
245. Ruan J, Martin P, Shah B, Schuster SJ, Smith SM, Furman RR, et al.
Lenalidomide plus rituximab as initial treatment for mantle-cell
Lymphoma. N Engl J Med (2015). doi: 10.1056/NEJMoa1505237
246. Wang M, Fayad L, Wagner-Bartak N, Zhang L, Hagemeister F, Neelapu SS,
et al. Lenalidomide in combination with rituximab for patients with relapsed
or refractory mantle-cell lymphoma: A phase 1/2 clinical trial. Lancet Oncol
(2012). doi: 10.1016/S1470-2045(12)70200-0
247. Monga N, Nastoupil L, Garside J, Quigley J, Hudson M, ODonovan P, et al.
Burden of illness of follicular lymphoma and marginal zone lymphoma. Ann
Hematol (2019). doi: 10.1007/s00277-018-3501-8
248. Muppidi JR, Schmitz R, Green JA, Xiao W, Larsen AB, Braun SE, et al. Loss
of signalling via Ga13 in germinal centre B-cell-derived lymphoma. Nature
(2014). doi: 10.1038/nature13765
249. Gonzalez-RinconJ,Me
ndez M, Gomez S, Garcıa JF, Martın P, Bellas C, et al.
Unraveling transformation of follicular lymphoma to diffuse large B-cell
lymphoma. PloS One (2019). doi: 10.1371/journal.pone.0212813
250. Aguzzi A, Kranich J, Krautler NJ. Follicular dendritic cells: Origin,
phenotype, and function in health and disease. Trends Immunol (2014).
doi: 10.1016/j.it.2013.11.001
251. Pikor NB, Mörbe U, Lütge M, Gil-Cruz C, Perez-Shibayama C, Novkovic M,
et al. Remodeling of light and dark zone follicular dendritic cells governs
germinal center responses. Nat Immunol (2020). doi: 10.1038/s41590-020-
0672-y
252. He B, Chadburn A, Jou E, Schattner EJ, Knowles DM, Cerutti A. Lymphoma
B Cells Evade Apoptosis through the TNF Family Members BAFF/BLyS and
APRIL. J Immunol (2004). doi: 10.4049/jimmunol.172.8.5128-c
253. Heinig K, Gätjen M, Grau M, Stache V, Anagnostopoulos I, Gerlach K, et al.
Access to follicular dendritic cells is a pivotal step in murine chronic
lymphocytic leukemia b-cell activation and proliferation. Cancer Discovery
(2014). doi: 10.1158/2159-8290.CD-14-0096
254. Kurshumliu F, Sadiku-Zehri F, Qerimi A, Vela Z, Jashari F, Bytyci S, et al.
Divergent immunohistochemical expression of CD21 and CD23 by follicular
dendritic cells with increasing grade of follicular lymphoma. World J Surg
Oncol (2019). doi: 10.1186/s12957-019-1659-8
255. Matas-Cespedes A, Rodriguez V, Kalko SG, Vidal-Crespo A, Rosich L,
Casserras T, et al. Disruption of follicular dendritic cells-follicular
lymphoma cross-talk by the pan-PI3K inhibitor BKM120 (buparlisib). Clin
Cancer Res (2014). doi: 10.1158/1078-0432.CCR-14-0154
256. Serrat N, Guerrero-Hernandez M, Matas-Cespedes A, Yahiaoui A, Valero
JG, Nadeu F, et al. PI3Kdinhibition reshapes follicular lymphoma-immune
microenvironment cross talk and unleashes the activity of venetoclax. Blood
Adv (2020) 4(17):421731. doi: 10.1182/bloodadvances.2020001584
257. Ame-Thomas P, Le Priol J, Yssel H, Caron G, Pangault C, Jean R, et al.
Characterization of intratumoral follicular helper T cells in follicular
lymphoma: Role in the survival of malignant B cells. Leukemia (2012). doi:
10.1038/leu.2011.301
258. Amin R, Mourcin F, Uhel F, Guirriec M, Lamy T, Tarte K. DC-SIGN Binds
Preferentially Highly Glycosylated IgM to Trigger Classical BCR Signaling in
Follicular Lymphoma. Blood (2014). doi: 10.1182/blood.V124.21.2968.2968
259. Kridel R, Xerri L, Gelas-Dore B, Tan K, Feugier P, Vawda A, et al. The
prognostic impact of CD163-positive macrophages in follicular Lymphoma:
A study from the BC cancer agency and the lymphoma study association.
Clin Cancer Res (2015). doi: 10.1158/1078-0432.CCR-14-3253
260. Arias V, Soares FA. Vascular density (tumor angiogenesis) in non-Hodgkins
lymphomas and orid follicular hyperplasia: A morphometric study. Leuk
Lymphoma (2001). doi: 10.3109/10428190009054893
261. Crivellato E, Nico B, Vacca A, Ribatti D. B-cell non-Hodgkins lymphomas
express heterogeneous patterns of neovascularization. Haematologica (2003).
262. Ribatti D, Vacca A, Nico B, Fanelli M, Roncali L, Dammacco F. Angiogenesis
spectrum in the stroma of B-cell non-Hodgkinslymphomas.An
immunohistochemical and ultrastructural study. Eur J Haematol (2009).
doi: 10.1111/j.1600-0609.1996.tb00293.x
263. Shahini L, Gasparov S, Petrusevska G, Kerliu SM, Veselaj F, Kurshumliu F,
et al. Clinical signicance of VEGF-A and microvessel density in diffuse large
B-cell lymphoma and low-grade follicular lymphoma. Acta Clin Croat
(2017). doi: 10.20471/acc.2017.56.04.02
264. Passalidou E, Stewart M, Trivella M, Steers G, Pillai G, Dogan A, et al.
Vascular patterns in reactive lymphoid tissue and in non-Hodgkins
lymphoma. Br J Cancer (2003). doi: 10.1038/sj.bjc.6600742
265. Hainsworth JD, Greco FA, Raefsky EL, Thompson DS, Lunin S, Reeves J,
et al. Rituximab with or without bevacizumab for the treatment of patients
with relapsed follicular lymphoma. Clin Lymphoma Myeloma Leuk (2014).
doi: 10.1016/j.clml.2014.02.010
266. Abramson JS, Shipp MA. Advances in the biology and therapy of diffuse
large B-cell lymphoma: Moving toward a molecularly targeted approach.
Blood (2005). doi: 10.1182/blood-2005-02-0687
267. Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H, et al. Stromal gene
signatures in large-B-cell lymphomas. N Engl J Med (2008). doi: 10.1056/
NEJMoa0802885
268. Abdou AG, Asaad N, Kandil M, Shabaan M, Shams A. Signicance of
stromal-1 and stromal-2 signatures and biologic prognostic model in diffuse
large B-cell lymphoma. Cancer Biol Med (2017). doi: 10.20892/j.issn.2095-
3941.2017.0007
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174122
269. Perry AM, Cardesa-Salzmann TM, Meyer PN, Colomo L, Smith LM, Fu K,
et al. A new biologic prognostic model based on immunohistochemistry
predicts survival in patients with diffuse large B-cell lymphoma. Blood
(2012). doi: 10.1182/blood-2012-05-430389
270. Marinaccio C, Ingravallo G, Gaudio F, Perrone T, Nico B, Maoirano E,
et al. Microvascular density, CD68 and tryptase expression in human
Diffuse Large B-Cell Lymphoma. Leuk Res (2014). doi: 10.1016/
j.leukres.2014.09.007
271. Cardesa-Salzmann TM, Colomo L, Gutierrez G, Chan WC, Weisenburger D,
Climent F, et al. High microvessel density determines a poor outcome in
patients with diffuse large B-cell lymphoma treated with rituximab plus
chemotherapy. Haematologica (2011). doi: 10.3324/haematol.2010.037408
272. Tzankov A, Heiss S, Ebner S, Sterlacci W, Schaefer G, Augustin F, et al.
Angiogenesis in nodal B cell lymphomas: A high throughput study. J Clin
Pathol (2007). doi: 10.1136/jcp.2006.038661
273. Gomez-Gelvez JC, Salama ME, Perkins SL, Leavitt M, Inamdar KV.
Prognostic impact of tumor microenvironment in diffuse large B-cell
lymphoma uniformly treated with R-CHOP chemotherapy. Am J Clin
Pathol (2016). doi: 10.1093/ajcp/aqw034
274. Hazar B, Paydas S, Zorludemir S, Sahin B, Tuncer I. Prognostic signicance
of microvessel density and vascular endothelial growth factor (VEGF)
expression in non-Hodgkinslymphoma.Leuk Lymphoma (2003). doi:
10.1080/1042819031000123519
275. Foss HD, Araujo I, Demel G, Klotzbach H, Hummel M, Stein H. Expression
of vascular endothelial growth factor in lymphomas and Castlemans disease.
JPathol(1997). doi: 10.1002/(SICI)1096-9896(199709)183:1<44::AID-
PATH1103>3.0.CO;2-I
276. Ribatti D, Vacca A, Bertossi M, De Benedictis G, Roncali L, Dammacco F.
Angiogenesis induced by B-cell non-Hodgkins lymphomas. Lack of
correlation with tumor malignancy and immunologic phenotype.
Anticancer Res (1990).
277. Stewart M, Talks K, Leek R, Turley H, Pezzella F, Harris A, et al. Expression
of angiogenic factors and hypoxia inducible factors HIF 1, HIF 2 and CA IX
in non-Hodgkins lymphoma. Histopathology (2002). doi: 10.1046/j.1365-
2559.2002.01357.x
278. Suhasini AN, Wang L, Holder KN, Lin AP, Bhatnagar H, Kim SW, et al. A
phosphodiesterase 4B-dependent interplay between tumor cells and the
microenvironment regulates angiogenesis in B-cell lymphoma. Leukemia
(2016). doi: 10.1038/leu.2015.302
279. Broseus J, Mourah S, Ramstein G, Bernard S, Mounier N, Cuccuini W, et al.
VEGF121, is predictor for survival in activated B-cell-like diffuse large B-cell
lymphoma and is related to an immune response gene signature conserved in
cancers. Oncotarget (2017). doi: 10.18632/oncotarget.19385
280. Fiore F, von Bergwelt-Baildon MS, Drebber U, Beyer M, Popov A, Manzke O,
et al. Dendritic cells are signicantly reduced in non-Hodgkins lymphoma and
express less CCR7 and CD62L. Leuk Lymphoma (2006). doi: 10.1080/
10428190500360971
281. Cai QC, Liao H, Lin SX, Xia Y, Wang XX, Gao Y, et al. High expression of
tumor-inltrating macrophages correlates with poor prognosis in patients
with diffuse large B-cell lymphoma. Med Oncol (2012). doi: 10.1007/s12032-
011-0123-6
282. Li YL, Shi ZH, Wang X, Gu KS, Zhai ZM. Tumor-associated macrophages
predict prognosis in diffuse large B-cell lymphoma and correlation with
peripheral absolute monocyte count. BMC Cancer (2019). doi: 10.1186/
s12885-019-6208-x
283. Hasselblom S, Hansson U, Sigurdardottir M, Nilsson-Ehle H, Ridell B,
Andersson PO. Expression of CD68+ tumor-associated macrophages in
patients with diffuse large B-cell lymphoma and its relation to prognosis.
Pathol Int (2008). doi: 10.1111/j.1440-1827.2008.02268.x
284. Marchesi F, Cirillo M, Bianchi A, Gately M, Olimpieri OM, Cerchiara E, et al.
High density of CD68+/CD163+ tumour-associated macrophages (M2-
TAM) at diagnosis is signicantly correlated to unfavorable prognostic
factors and to poor clinical outcomes in patients with diffuse large B-cell
lymphoma. Hematol Oncol (2015). doi: 10.1002/hon.2142
285. Fukushima H, Ohsawa M, Ikura Y, Naruko T, Sugama Y, Suekane T, et al.
Mast cells in diffuse large B-cell lymphoma; their role in brosis.
Histopathology (2006). doi: 10.1111/j.1365-2559.2006.02534.x
286. Hedström G, Berglund M, Molin D, Fischer M, Nilsson G, Thunberg U, et al.
Mast cell inltration is a favourable prognostic factor in diffuse large B-cell
lymphoma. Br J Haematol (2007). doi: 10.1111/j.1365-2141.2007.06612.x
287. Ribatti D, Vacca A, Marzullo A, Nico B, Ria R, Roncali L, et al. Angiogenesis
and mast cell density with tryptase activity increase simultaneously with
pathological progression in B-cell non-Hodgkins lymphomas. Int J Cancer
(2000). doi: 10.1002/(SICI)1097-0215(20000115)85:2%3C171::AID-IJC4%
3E3.0.CO;2-W
288. Wroblewski M, Bauer R, Cubas Cordova M, Udonta F, Ben-Batalla I, Legler
K, et al. Mast cells decrease efcacy of anti-angiogenic therapy by secreting
matrix-degrading granzyme B. Nat Commun (2017). doi: 10.1038/s41467-
017-00327-8
289. Ribatti D, Crivellato E. Mast cells, angiogenesis, and tumour growth. Biochim
Biophys Acta - Mol Basis Dis (2012). doi: 10.1007/978-1-4614-5857-9_7
290. Valtola R, Salven P, Heikkilä P, Taipale J, Joensuu H, Rehn M, et al. VEGFR-
3 and its ligand VEGF-C are associated with angiogenesis in breast cancer.
Am J Pathol (1999). doi: 10.1016/S0002-9440(10)65392-8
291. Nowakowski GS, LaPlant B, Macon WR, Reeder CB, Foran JM, Nelson GD,
et al. Lenalidomide combined with R-CHOP overcomes negative prognostic
impact of non-germinal center B-cell phenotype in newly diagnosed diffuse
large B-cell lymphoma: A phase II study. J Clin Oncol (2015). doi: 10.1200/
JCO.2014.55.5714
292. Hartert KT, Wenzl K, Krull JE, Manske M, Sarangi V, Asmann Y, et al.
Targeting of inammatory pathways with R2CHOP in high-risk DLBCL.
Leukemia (2020). doi: 10.1038/s41375-020-0766-4
293. Kalisz K, Alessandrino F, Beck R, Smith D, Kikano E, Ramaiya NH, et al. An
update on Burkitt lymphoma: a review of pathogenesis and multimodality
imaging assessment of disease presentation, treatment response, and
recurrence. Insights Imaging (2019). doi: 10.1186/s13244-019-0733-7
294. Teruya-Feldstein J, Jaffe ES, Burd PR, Kanegane H, Kingma DW, Wilson
WH, et al. The role of Mig, the monokine induced by interferon-g, and IP-10,
the interferon-g-inducible protein-10, in tissue necrosis and vascular damage
associated with Epstein-Barr virus-positive lymphoproliferative disease.
Blood (1997). doi: 10.1182/blood.V90.10.4099
295. Becker J, Covelo-Fernandez A, Von Bonin F, Kube D, Wilting J. Specic
tumor-stroma interactions of EBV-positive Burkitts lymphoma cells in the
chick chorioallantoic membrane. Vasc Cell (2012). doi: 10.1186/2045-824X-
4-3
296. Vacca A, Ribatti D, Ruco L, Giacchetta F, Nico B, Quondamatteo F, et al.
Angiogenesis extent and macrophage density increase simultaneously with
pathological progression in B-cell non-Hodgkins lymphomas. Br J Cancer
(1999). doi: 10.1038/sj.bjc.6690154
297. Ribatti D, Nico B, Vacca A, Marzullo A, Calvi N, Roncali L, et al. Do mast
cells help to induce angiogenesis in B-cell non-Hodgkins lymphomas? Br J
Cancer (1998). doi: 10.1038/bjc.1998.316
298. Brandvold KA, Neiman P, Ruddell A. Angiogenesis is an early event in the
generation of myc-induced lymphomas. Oncogene (2000). doi: 10.1038/
sj.onc.1203589
299. Ruan J, Hyjek E, Kermani P, Christos PJ, Hooper AT, Coleman M, et al.
Magnitude of stromal hemangiogenesis correlates with histologic subtype of
non-Hodgkinslymphoma.Clin Cancer Res (2006). doi: 10.1158/1078-
0432.CCR-06-1204
300. Ruan J, Hyjek E, Hooper AT, Vincent L, Kermani P, Christos PJ, et al.
Stromal Incorporation of VEGFR-1+, CD68+ and a-SMA+ Hemangiogenic
Cells Correlates with Histologic Subtype in Non-Hodgkins Lymphoma.
Blood (2005). doi: 10.1182/blood.V106.11.1930.1930
301. De Palma M, Coukos G, Hanahan D. A New Twist on Radiation Oncology:
Low-Dose Irradiation Elicits Immunostimulatory Macrophages that Unlock
Barriers to Tumor Immunotherapy. Cancer Cell (2013). doi: 10.1016/
j.ccr.2013.10.019
302. Marinaccio C, Nico B, Maiorano E, Specchia G, Ribatti D. Insights in
Hodgkin Lymphoma angiogenesis. Leukemia Res (2014). doi: 10.1016/
j.leukres.2014.05.023
303. Korkolopoulou P, Thymara I, Kavantzas N, Vassilakopoulos TP,
Angelopoulou MK, Kokoris SI, et al. Angiogenesis in Hodgkins
lymphoma: A morphometric approach in 286 patients with prognostic
implications. Leukemia (2005). doi: 10.1038/sj.leu.2403690
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174123
304. Niitsu N, Okamoto M, Nakamine H, Yoshino T, Tamaru JI, Nakamura S,
et al. Simultaneous elevation of the serum concentrations of vascular
endothelial growth factor and interleukin-6 as independent predictors of
prognosis in aggressive non-Hodgkins lymphoma. Eur J Haematol (2002).
doi: 10.1034/j.1600-0609.2002.01609.x
305. Ben Arush MW, Ben Barak A, Maurice S, Livne E. Serum VEGF as a
signicant marker of treatment response in Hodgkin lymphoma. Pediatr
Hematol Oncol (2007). doi: 10.1080/08880010601052381
306. Kuittinen O, Soini Y, Turpeenniemi-Hujanen T. Diverse role of MMP-2 and
MMP-9 in the clinicopathological behavior of Hodgkins lymphoma. Eur J
Haematol (2002). doi: 10.1034/j.1600-0609.2002.02751.x
307. Bardelli M, Leucci E, Schürfeld K, Bellan C, Passiatore G, Rocchigiani M,
et al. VEGF-D is expressed in activated lymphoid cells and in tumors of
hematopoietic and lymphoid tissues. Leuk Lymphoma (2007). doi: 10.1080/
10428190701540975
308. Linke F, Harenberg M, Nietert MM, Zaunig S, Von Bonin F, Arlt A, et al.
Microenvironmental interactions between endothelial and lymphoma cells:
A role for the canonical WNT pathway in Hodgkin lymphoma. Leukemia
(2017). doi: 10.1038/leu.2016.232
309. Celegato M, Borghese C, Casagrande N, Mongiat M, Kahle XU, Paulitti A,
et al. Preclinical activity of the repurposed drug auranon in classical
Hodgkin lymphoma. Blood (2015). doi: 10.1182/blood-2015-07-660365
310. Fhu CW, Graham AM, Yap CT, Al-Salam S, Castella A, Chong SM, et al.
Reed-Sternberg cell-derived lymphotoxin-a activates endothelial cells to
enhance T-cell recruitment in classical Hodgkin lymphoma. Blood (2014).
doi: 10.1182/blood-2014-05-576140
311. Werner L, Dreyer JH, Hartmann D, Barros MHM, Büttner-Herold M,
Grittner U, et al. Tumor-associated macrophages in classical Hodgkin
lymphoma: hormetic relationship to outcome. Sci Rep (2020). doi:
10.1038/s41598-020-66010-z
312. Al Sayed Ahmed H, Raslan WF, Deifalla AHS, Fathallah MD. CD163 is a
predictive biomarker for prognosis of classical Hodgkins lymphoma in
Saudi patients. Mol Clin Oncol (2019). doi: 10.3892/mco.2019.1850
313. Koh YW, Park CS, Yoon DH, Suh C, Huh J. CD163 Expression was
associated with angiogenesis and shortened survival in patients with
uniformly treated classical Hodgkin lymphoma. PloS One (2014). doi:
10.1371/journal.pone.0087066
314. Locatelli SL, Careddu G, Serio S, Consonni FM, Maeda A, Viswanadha S,
et al. Dual PI3K/inhibition suppresses M2 macrophage polarization in
hodgkin lymphoma through PKM2 downregulation. Haematologica
(2018). doi: 10.1158/1078-0432.CCR-18-1133
315. Locatelli SL, Careddu G, Serio S, Consonni FM, Maeda A, Viswanadha S,
et al. Targeting cancer cells and tumor microenvironment in preclinical and
clinical models of Hodgkin lymphoma using the dual PI3Kd/G inhibitor
RP6530. Clin Cancer Res (2019). doi: 10.1158/1078-0432.CCR-18-1133
316. Cao Y, Langer R. A review of Judah Folkmans remarkable achievements in
biomedicine. Proc Natl Acad Sci U S A (2008). doi: 10.1073/pnas.
0806582105
317. Sherwood LM, Parris EE, Folkman J. Tumor Angiogenesis: Therapeutic
Implications. New Engl J Med (1971). doi: 10.1056/NEJM197111182852108
318. Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, et al.
Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. Front Oncol
(2020). doi: 10.3389/fonc.2020.00221
319. Rosen LS, Jacobs IA, Burkes RL. Bevacizumab in Colorectal Cancer: Current
Role in Treatment and the Potential of Biosimilars. Target Oncol (2017). doi:
10.1007/s11523-017-0518-1
320. Summers J, Cohen MH, Keegan P, Pazdur R. FDA Drug Approval Summary:
Bevacizumab plus Interferon for Advanced Renal Cell Carcinoma. Oncologist
(2010). doi: 10.1634/theoncologist.2009-0250
321. Winkler F, Osswald M, Wick W. Anti-Angiogenics: Their Role in the
Treatment of Glioblastoma. Oncol Res Treat (2018). doi: 10.1159/000488258
322. Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in
oncology: current status and future directions. Lancet (2016). doi: 10.1016/
S0140-6736(15)01088-0
323. Shaked Y, Henke E, Roodhart JML, Mancuso P, Langenberg MHG, Colleoni
M, et al. Rapid Chemotherapy-Induced Acute Endothelial Progenitor Cell
Mobilization: Implications for Antiangiogenic Drugs as Chemosensitizing
Agents. Cancer Cell (2008). doi: 10.1016/j.ccr.2008.08.001
324. Shaked Y, Ciarrocchi A, Franco M, Lee CR, Man S, Cheung AM, et al.
Therapy-induced acute recruitment of circulating endothelial progenitor
cells to tumors. Science (2006). doi: 10.1126/science.1127592
325. Park JS, Kim IK, Han S, Park I, Kim C, Bae J, et al. Normalization of Tumor
Vessels by Tie2 Activation and Ang2 Inhibition Enhances Drug Delivery and
Produces a Favorable Tumor Microenvironment. Cancer Cell (2016). doi:
10.1016/j.ccell.2016.10.018
326. Mattheolabakis G, Mikelis CM. Nanoparticle Delivery and Tumor Vascular
Normalization: The Chicken or The Egg? Front Oncol (2019). doi: 10.3389/
fonc.2019.01227
327. Wang L, Peng S, Sun W, Liu X. Bevacizumab synergises with the BCL 2
inhibitor venetoclax to effectively treat B-cell non-Hodgkins lymphoma. Eur
J Haematol (2019). doi: 10.1111/ejh.13279
328. Zhang Q, Cao J, Xue K, Liu X, Ji D, Guo Y, et al. Recombinant human
endostatin in combination with CHOP regimen for peripheral T cell
lymphoma. Onco Targets Ther (2017). doi: 10.2147/OTT.S117007
329. Yang J, Yan J, Liu B. Targeting VEGF/VEGFR to modulate antitumor
immunity. Front Immunol (2018). doi: 10.3389/mmu.2018.00978
330. Azad AK, Zhabyeyev P, Vanhaesebroeck B, Eitzen G, Oudit GY, Moore RB,
et al. Inactivation of endothelial cell phosphoinositide 3-kinase binhibits
tumor angiogenesis and tumor growth. Oncogene (2020). doi: 10.1038/
s41388-020-01444-3
331. Lannutti BJ, Meadows SA, Herman SEM, Kashishian A, Steiner B, Johnson
AJ, et al. CAL-101, a p110dselective phosphatidylinositol-3-kinase inhibitor
for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular
viability. Blood (2011). doi: 10.1182/blood-2010-03-275305
332. Ali K, Soond DR, Piñeiro R, Hagemann T, Pearce W, Lim EL, et al.
Inactivation of PI(3)K p110dbreaks regulatory T-cell-mediated immune
tolerance to cancer. Nature (2014). doi: 10.1038/nature13444
333. Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt Signaling
Pathway in Macrophage Activation and M1/M2 Polarization. J Immunol
(2017). doi: 10.4049/jimmunol.1601515
334. von Keudell G, Moskowitz AJ. The Role of PI3K Inhibition in Lymphoid
Malignancies. Curr Hematol Malig Rep (2019). doi: 10.1007/s11899-019-
00540-w
335. Wullenkord R, Friedrichs B, Erdmann T, Lenz G. Therapeutic potential of
PI3K signaling in distinct entities of B-cell lymphoma. Expert Rev Hematol
(2019). doi: 10.1080/17474086.2019.1676716
336. Yang Q, Modi P, Newcomb T, Queva C, Gandhi V. Idelalisib: First-in-class
PI3K delta inhibitor for the treatment of chronic lymphocytic leukemia,
small lymphocytic leukemia, and follicular lymphoma. Clin Cancer Res
(2015). doi: 10.1158/1078-0432.CCR-14-2034
337. Sapon-Cousineau V, Sapon-Cousineau S, Assouline S. PI3K Inhibitors and
Their Role as Novel Agents for Targeted Therapy in Lymphoma. Curr Treat
Options Oncol (2020). doi: 10.1007/s11864-020-00746-8
338. Tang H, Zhu M, Qiao J, Fu Y-X. Lymphotoxin signalling in tertiary
lymphoid structures and immunotherapy. Cell Mol Immunol (2017) 14
(10):80918. doi: 10.1038/cmi.2017.13
339. Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, et al. Low-Dose
Irradiation Programs Macrophage Differentiation to an iNOS+/M1
Phenotype that Orchestrates Effective T Cell Immunotherapy. Cancer Cell
(2013). doi: 10.1016/j.ccr.2013.09.014
340. Ruan J. Antiangiogenic Therapies in Non-Hodgkins Lymphoma. Curr
Cancer Drug Targets (2011). doi: 10.2174/156800911798073014
341. Vieth B, Parekh S, Ziegenhain C, Enard W, Hellmann I. A systematic
evaluation of single cell RNA-seq analysis pipelines. Nat Commun (2019).
doi: 10.1101/583013
342. Tang X, Huang Y, Lei J, Luo H, Zhu X. The single-cell sequencing: New
developments and medical applications. Cell Biosci (2019). doi: 10.1186/
s13578-019-0314-y
343. Strzelecka PM, Ranzoni AM, Cvejic A. Dissecting human disease with single-
cell omics: Application in model systems and in the clinic. Dis Models Mech
(2018). doi: 10.1242/dmm.036525
344. Luecken MD, Theis FJ. Current best practices in single-cell RNA-
seq analysis: a tutorial. Mol Syst Biol (2019). doi: 10.15252/msb.20188746
345. Arnol D, Schapiro D, Bodenmiller B, Saez-Rodriguez J, Stegle O. Modeling
Cell-Cell Interactions from Spatial Molecular Data with Spatial Variance
Component Analysis. Cell Rep (2019). doi: 10.1101/265256
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174124
346. Solomon M, DeLay M, Reynaud D. Phenotypic Analysis of the Mouse
Hematopoietic Hierarchy Using Spectral Cytometry: From Stem Cell Subsets
to Early Progenitor Compartments. Cytometry A (2020). doi: 10.1002/
cyto.a.24041
347. Ferrer-Font L, Pellegues C, Mayer JU, Small SJ, Jaimes MC, Price KM. Panel
Design and Optimization for High-Dimensional Immunophenotyping
Assays Using Spectral Flow Cytometry. Curr Protoc Cytom (2020). doi:
10.1101/784884
348. Corey DM, Rinkevich Y, Weissman IL. Dynamic patterns of clonal evolution
in tumor vasculature underlie alterations in lymphocyte-endothelial
recognition to foster tumor immune escape. Cancer Res (2016). doi:
10.1158/0008-5472.CAN-15-1150
349. Huyghe L, Van Parys A, Cauwels A, Van Lint S, De Munter S, Bultinck J,
et al. Safe eradication of large established tumors using neovasculature-
targeted tumor necrosis factor-based therapies. EMBO Mol Med (2020). doi:
10.15252/emmm.201911223
350. Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, et al.
Convergence of acquired mutations and alternative splicing of CD19 enables
resistance to CART-19 immunotherapy. Cancer Discov (2015). doi: 10.1158/
2159-8290.CD-15-1020
351. Gardner R, Wu D, Cherian S, Fang M, HanaLA, Finney O, et al.
Acquisition of a CD19-negative myeloid phenotype allows immune escape
of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood (2016).
doi: 10.1182/blood-2015-08-665547
352. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev
Clin Oncol (2019). doi: 10.1038/s41571-019-0184-6
353. John LB, Devaud C, Duong CPM, Yong CS, Beavis PA, Haynes NM, et al.
Anti-PD-1 antibody therapy potently enhances the eradication of established
tumors by gene-modied T cells. Clin Cancer Res (2013). doi: 10.1158/1078-
0432.CCR-13-0458
354. Wei J, Luo C, Wang Y, Guo Y, Dai H, Tong C, et al. PD-1 silencing impairs
the anti-tumor function of chimeric antigen receptor modied T cells by
inhibiting proliferation activity. J Immunother Cancer (2019). doi: 10.1186/
s40425-019-0685-y
355. Kueberuwa G, Kalaitsidou M, Cheadle E, Hawkins RE, Gilham DE. CD19
CAR T Cells Expressing IL-12 Eradicate Lymphoma in Fully Lymphoreplete
Mice through Induction of Host Immunity. Mol Ther Oncolytics (2018). doi:
10.1016/j.omto.2017.12.003
356. Stach M, Musil J, Cetkovsky P, Otahal P. Interleukin 21 Enhances Survival
and Expansion of CAR T Cells Via Inhibition of Their Terminal
Differentiation during Interaction with Tumor Target Cells. Blood (2018).
doi: 10.1182/blood-2018-99-116294
357. Chmielewski M, Abken H. TRUCKs with IL-18 payload: Toward shaping the
immune landscape for a more efcacious CAR T-cell therapy of solid cancer.
Adv Cell Gene Ther (2018). doi: 10.1002/acg2.7
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2020 Menzel, Höpken and Rehm. This is an open-access article
distributed under the terms of the Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other forums is permitted, provided the
original author(s) and the copyright owner(s) are credited and that the original
publicationin this journalis cited, in accordance with accepted academic practice. No
use, distribution or reproduction is permitted which doesnot comply with these terms.
Menzel et al. Angiogenesis in Lymphoma
Frontiers in Immunology | www.frontiersin.org December 2020 | Volume 11 | Article 59174125
... In addition to the aforementioned cytokines, Reed-Sternberg cells secrete survival factors, such as B-cell-derived growth factor and vascular endothelial growth factor (VEGF). These factors promote the survival and proliferation of tumor cells, aiding their nutritional and oxygen supply [66,67]. ...
... It is instrumental in the organization of germinal centers within the lymphoid organs. The expression of CCL21 within the HL microenvironment may affect the organization of tumor and immune cells within the affected lymph nodes, potentially influencing disease progression and specific immune responses [66,99]. The other molecules associated with HL and vascular biology are listed in Table 1. ...
Article
Full-text available
Simple Summary Hodgkin’s lymphoma is a type of cancer that affects the lymphatic system, which is an important part of the body’s immune system. This type of cancer involves a set of molecules and cells that promote tumor survival. Some structures found in the tumor niche are associated with vascular biology, which integrates a network of molecules and cells that play a fundamental role in nutrition and oxygen supply to tissues, including tumors. Thus, the main objective of this study was to improve our knowledge of the relationship between vascular biology and Hodgkin’s lymphoma and to enable new studies on disease intervention. Here, we describe the main cells and molecules associated with tumors that play a role in vascular biology. Additionally, this study addresses the main models of disease experimentation. Abstract Hodgkin’s lymphoma (HL) is a lymphatic neoplasm typically found in the cervical lymph nodes. The disease is multifactorial, and in recent years, the relationships between various vascular molecules have been explored in the field of vascular biology. The connection between vascular biology and HL is intricate and the roles of several pathways remain unclear. This review summarizes the cellular and molecular relationships between vascular biology and HL. Proteins associated with various functions in vascular biology, including cytokines (TNF-α, IL-1, IL-13, and IL-21), chemokines (CXCL10, CXCL12, and CCL21), adhesion molecules (ELAM-1/VCAM-1), and growth factors (BDNF/NT-3, platelet-derived growth factor receptor-α), have been linked to tumor activity. Notable tumor activities include the induction of paracrine activation of NF-kB-dependent pathways, upregulation of adhesion molecule regulation, genome amplification, and effective loss of antigen presentation mediated by MHC-II. Preclinical study models, primarily those using cell culture, have been optimized for HL. Animal models, particularly mice, are also used as alternatives to complex biological systems, with studies primarily focusing on the physiopathogenic evaluation of the disease. These biomolecules warrant further study because they may shed light on obscure pathways and serve as targets for prevention and/or treatment interventions.
... Based on the pathophysiological background, angiogenesis has a vital role in lymphoma biology, and lymphoma subtypes differ signi cantly in terms of angiogenesis [7][8][9][10]. Menzel et al [11] found that the progression of invasive lymphoma is usually accompanied by a so-called "vascular phase," which represents extensive vascularization of the lymph nodes, with the exponential growth of the internal vessels and no intervals, while indolent lymphomas are well differentiated, with relatively normal blood ow. Hence, we focus on the advantages of ultrasonographic Vascular imaging for qualitative and quantitative assessment of lymph node blood ow. ...
Preprint
Full-text available
Objective This study aimed to explore whether superficial invasive lymphomas and indolent lymphomas could be identified by Ultrasonographic vascular imaging. Method A retrospectively study enrolled 82 lymphoma patients. According to proliferation rates and clinical course, the lymph nodes were classified as invasive and indolent lymphomatous lymph nodes. All patients underwent ultrasound (US) with three effective techniques: color Doppler flow imaging (CDFI), angio plus ultrasound imaging (AngioPLUS), and contrast-enhanced ultrasound (CEUS). Qualitative and quantitative parameters from the two groups were compared. Finally, the area under the receiver-operating characteristic (ROC) and regression analysis were used to compare the differences between the two groups and determine the diagnostic efficiency of the three techniques for differentiating invasive lymphoma from indolent lymphoma. Result The types of blood flow distribution between invasive and indolent lymphomatous lymph nodes were statistically different in all three Ultrasound techniques. In CDFI, invasive or indolent lymphomatous lymph nodes were determined by resistance index (RI) (p < 0.001). In CEUS, the differences between the two groups in necrosis and arrival time (ATM) (p = 0.026, 0.043) were statistically significant. Finally, CDFI combined with CEUS had the highest diagnostic sensitivity of 98.1%. Interobserver agreements for qualitative parameters were all excellent. Conclusion Ultrasonographic Vascular imaging is an aid in identifying invasive and indolent lymphomatous lymph nodes, and CDFI combined with CEUS had the highest diagnostic sensitivity, which can guide clinicians to make more accurate diagnosis and better treatment for patients.
... In addition, overexpression of the isozyme SPHK1, responsible for the production of sphingosine-1 phosphate, acts as a potent inducer of angiogenesis in DLBCL ( Figure 1) (13,14). However, it should be noted that follow up of patients treated with anti-angiogenic agents such as anti-VEGF-A antibodies, bevacizumab, and R-CHOP has shown that increased vascularity in DLBCL patients may have only minor importance for the disease course as these regimens did not show increased therapeutic efficacy on patients (15). ...
Article
Background and objective: The tumor microenvironment (TME) is known to exert a significant impact on disease biology and convey prognostic and therapeutic implications in several hematopoietic neoplasms, including diffuse large B-cell lymphoma (DLBCL). Recent discoveries have yielded new information regarding the genetic signatures of the TME. This study aims to review the updates on the cellular markers and genetics of various components of the TME in DLBCL influencing tumor behavior and patients' responses to treatment and discuss the novel treatment modalities available for the patients. Methods: We systematically reviewed the literature in Medline for DLBCL-related articles on gene expression studies of TME. Forty-seven articles were identified and included that were published between Apr 2006 and Apr 2023. Key content and findings: We review the key components of the TME including the endothelial cells, myofibroblasts and mast cells, and discuss their biologic roles, with a particular focus on elements of their crosstalk relevant to DLBCL. We also review the genetic changes in lymphocytes and macrophages in TME of DLBCL. Increased understanding of emergent molecular alterations may hopefully allow improved prognostication and translational discoveries which will benefit patients with aggressive B-cell lymphomas. Conclusions: Combining cell of origin and TME as a risk stratification/prognostication system could provide more effective targeted therapeutic regiments. Identifying TME-targeted therapies will happen after providing the TME markers in the DLBCLs.
... cDCs communicate with fibroblastic reticular cells and help adjust the stromal compartment during priming (39). They also produce VEGF or IL31 for endothelial and neuronal crosstalk (40,41). Thus, cDCs represent a central component in the interconnective immune and non-immune network during immune responses. ...
Article
Full-text available
Dendritic cells (DCs) are major antigen-presenting cells that connect innate and adaptive immunity. Hepatic DCs are less activated and contribute to maintain the tolerogenic environment of the liver under steady state. Several studies indicated DCs in metabolic dysfunction-associated steatohepatitis (MASH), representing a substantial burden on healthcare systems due to its association with liver-related morbidity and mortality. Studies highlighted the potential disease-promoting role of liver DCs in the development of MASH while other experimental systems suggested their protective role. This review discusses this controversy and the current understanding of how DCs affect the pathogenesis of MASH.
... Our study reinforces the conclusion that nIC, nZeff, and ECV could effectively identify lymph node activity, aligning with prior findings [7,18,[22][23][24]26,31,38]. An increase in the number of blood vessels and alterations in tumor-associated vascular patterns within malignant lymph nodes may contribute to the elevation of IC [39,40], resulting in higher Zeff values [38]. Although 18 FDG-PET/CT is central to lymphoma assessment, high-resolution contrast-enhanced CT may be required for precise measurements of lymph nodes or mass size. ...
Article
Full-text available
Dual-energy CT has shown promising results in determining tumor characteristics and treatment effectiveness through spectral data by assessing normalized iodine concentration (nIC), normalized effective atomic number (nZeff), normalized electron density (nED), and extracellular volume (ECV). This study explores the value of quantitative parameters in contrast-enhanced dual-layer spectral detector CT (SDCT) as a potential tool for detecting lymph node activity in lymphoma patients. A retrospective analysis of 55 lymphoma patients with 289 lymph nodes, assessed through 18FDG-PET/CT and the Deauville five-point scale, revealed significantly higher values of nIC, nZeff, nED, and ECV in active lymph nodes compared to inactive ones (p < 0.001). Generalized linear mixed models showed statistically significant fixed-effect parameters for nIC, nZeff, and ECV (p < 0.05). The area under the receiver operating characteristic curve (AUROC) values of nIC, nZeff, and ECV reached 0.822, 0.845, and 0.811 for diagnosing lymph node activity. In conclusion, the use of g nIC, nZeff, and ECV as alternative imaging biomarkers to PET/CT for identifying lymph node activity in lymphoma holds potential as a reliable diagnostic tool that can guide treatment decisions.
... Since AG in DLBCL depends on the secretion of several factors [29,30] and the final MVD scores may not correlate with proangiogenic protein levels [31], the AG variables assessed by IHC were analyzed alone and compiled together with equal weights. For the purposes of this study, we defined the compilation of different IHC variables as a unique scoring method, named as 'angiogenic score' (AGS), which ranged from 0 to 3. In this setting, an AGS = 0 encompassed cases that were negative for VEGFR2 and whose VEGFA expression and MVD were below the median values. ...
Article
Full-text available
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma subtype and dependent on angiogenesis (AG), whose main effectors are VEGFA and VEGFR2. Functional single nucleotide variants (SNVs) are described in VEGFA and KDR genes. However, it still unknown whether VEGFA - 2578C/A, -2489C/T, -1154G/A, -634G/C, -460C/T and KDR-604T/C, -271G/A, +1192G/A and +1719A/T SNVs act on DLBCL risk and angiogenic features. Genomic DNA from 168 DLBCL patients and 205 controls was used for SNV genotyping. Angiogenesis was immunohistochemically assessed in tumor biopsies, with reactions for VEGFA, VEGFR2, and CD34. VEGFA -1154GG genotype were associated with 1.6-fold higher DLBCL risk. KDR + 1192GG plus KDR + 1719 TT and KDR + 1192GG plus VEGFA - 2578CC combined genotypes are associated with 2.19- and 2.04-fold higher risks of DLBCL, respectively. VEGFA - 634GG or GC genotypes are associated with increased microvessel density and VEGFA levels. No relationship was observed between SNVs and cell-of-origin classification of DLBCL, but higher VEGFA and VEGFR2 were seen in non-germinal center tumors.
... To grow beyond a certain volume, a malignant tumor is dependent on blood supply via newly formed vessels. The initiation of blood vessel expansion into the tumor is referred to as the angiogenic switch (10). ...
Article
Full-text available
Background: Increased microvascular density correlates with more advanced disease and unfavorable overall survival in non-Hodgkin lymphoma (NHL), suggesting that angiogenesis is important for disease progression. However, studies of anti-angiogenic agents in NHL patients, have generally not shown favorable outcomes. The aim of this study was to investigate whether plasma levels of a subset of angiogenesis-associated proteins are increased in indolent B-cell derived NHL (B-NHL) and to investigate whether the levels differ between patients with asymptomatic versus symptomatic disease. Methods: Plasma levels of growth differentiation factor 15 (GDF15), endostatin, matrix metalloproteinase 9 (MMP9), neutrophil gelatinase-associated lipocalin (NGAL), long pentraxin 3 (PTX3), and galectin 3 (GAL-3) were measured by ELISA in 35 patients with symptomatic indolent B-NHL, 41 patients with asymptomatic disease, and 62 healthy controls. Bootstrap t-tests were used to assess the relative differences in biomarker levels between groups. Group differences were visualized using a principal component plot. Results: Mean plasma endostatin and GDF15 levels were significantly higher in symptomatic and asymptomatic lymphoma patients than in controls. Symptomatic patients had higher mean MMP9 and NGAL than controls. Conclusions: The finding of increased plasma endostatin and GDF15 in patients with asymptomatic indolent B-NHL suggests that increased angiogenic activity is an early event in indolent B-NHL disease progression.
Article
Full-text available
Extranodal NK/T cell lymphoma (ENKTCL) is an extremely aggressive form of lymphoma and lacks of specific diagnostic markers. The study intended to unearth the role of interleukin-33 (IL-33) in ENKTCL. RT-qPCR was conducted to assess mRNA levels of ENKTCL tissues and cells, while western blot assay was performed for evaluating protein levels. Plate cloning experiment and transwell assay were employed to measure aggressiveness of ENKTCL. Tube formation assay was executed to determine the angiogenesis ability. Mice ENKTCL xenograft model was designed to probe the impacts of IL-33 in vivo. IL-33 and suppression of tumorigenicity 2 receptor (ST2, receptor of IL-33) were enhanced in ENKTCL. IL-33 inhibition suppressed viability, migration, and invasion of ENKTCL cells. Moreover, IL-33 knockdown restricted angiogenesis in human umbilical vein endothelial cells (HUVECs). Furthermore, Wnt/β-catenin pathway associated proteins (β-catenin, c-myc, and cyclin D1) were downregulated by loss of IL-33. However, these impacts were overturned by Wnt/β-catenin signaling agonist lithium chloride (LiCl). Additionally, IL-33 silencing exerted anti-tumor effect via Wnt/β-catenin pathway in vivo. Silencing of IL-33 inhibited ENKTCL tumorigenesis and angiogenesis by inactivating Wnt/β-catenin signaling pathway. As such, IL-33 might be a prospective treatment target for ENKTCL.
Chapter
Cancer is an illness wherein cells multiply and grow abnormally, invading other tissues and organs via the bloodstream and lymphatic systems (Thiagalingam, 2015). The human body contains trillions of cells, and cancer can manifest itself practically anywhere in the body of human beings. Cell division is how human cells grow and divide to form new cells (Weinberg & Weinberg, 2006). Old and damaged cells die and are replaced by new cells. Once this tightly controlled process fails, affected cells divide and grow when they are supposed to undergo cell death. Tumors are tissue lumps that are originated from these abnormal cells. Tumor cells can be cancerous or not (benign). By metastasis, the malignant cell can invade the neighboring tissue, enter the circulation, travel to distant organs, colonize them, and develop into new tumors (Chaffer & Weinberg, 2011). Malignant tumors invade surrounding tissues and enter into circulation and distant areas of the body to form new tumors. Many forms of cancer grow into solid tumors. However, a few cancer, namely, leukemia, do not generate solid tumors (Coghlin & Murray, 2010). Benign tumors, however, do not infiltrate or expand into the underlying tissue. Benign tumors rarely recur after their removal, whereas malignant tumors occasionally show recurrence. However, benign tumors, when grown in large, can be fatal. Some, like brain tumors of benign type, can cause serious illnesses or even death.
Article
Cancer-associated fibroblasts (CAFs) are abundant and important components of the tumour mesenchyme, and have been extensively studied for their role in primary tumours. CAFs provide biomechanical support for tumour cells and play key roles in immunosuppression and tumour metastasis. CAFs can promote epithelial-mesenchymal transition (EMT) of the primary tumour by secreting extracellular vesicles (EVs), increasing adhesion to tumour cells, remodelling the extracellular matrix (ECM) of the primary tumour, and changing its mechanical stiffness, which provides a pathway for tumour metastasis. Moreover, CAFs can form cell clusters with circulating tumour cells (CTCs) to help them resist blood shear forces and achieve colonisation of distant host organs. Recent studies have revealed their roles in pre-metastatic niche (PMN) formation and prevention. In this review, we discuss the role of CAFs in PMN formation and therapeutic interventions targeting PMN and CAFs to prevent metastasis.
Preprint
Full-text available
Technological advances allow for assaying multiplexed spatially resolved RNA and protein expression profiling of individual cells, thereby capturing physiological tissue contexts of single cell variation. While methods for the high-throughput generation of spatial expression profiles are increasingly accessible, computational methods for studying the relevance of the spatial organization of tissues on cell-cell heterogeneity are only beginning to emerge. Here, we present spatial variance component analysis (SVCA), a computational framework for the analysis of spatial molecular data. SVCA enables quantifying the effect of cell-cell interactions, as well as environmental and intrinsic cell features on the expression levels of individual genes or proteins. In application to a breast cancer Imaging Mass Cytometry dataset, our model allows for robustly estimating spatial variance signatures, identifying cell-cell interactions as a major driver of expression heterogeneity. Finally, we apply SVCA to high-dimensional imaging-derived RNA data, where we identify molecular pathways that are linked to cell-cell interactions.
Article
Full-text available
Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. However, only a fraction of patients respond, and most ultimately develop resistance to current angiogenesis inhibitor therapies. Activity of alternative pro-angiogenic growth factors, acting via RTK or G-protein coupled receptors (GPCR), may mediate VEGF inhibitor resistance. The phosphoinositide 3-kinase (PI3K)β isoform is uniquely coupled to both RTK and GPCRs. We investigated the role of endothelial cell (EC) PI3Kβ in tumor angiogenesis. Pro-angiogenic GPCR ligands were expressed by patient-derived renal cell carcinomas (PD-RCC), and selective inactivation of PI3Kβ reduced PD-RCC-stimulated EC spheroid sprouting. EC-specific PI3Kβ knockout (ΕC-βKO) in mice potentiated the sunitinib-induced reduction in subcutaneous growth of LLC1 and B16F10, and lung metastasis of B16F10 tumors. Compared to single-agent sunitinib treatment, tumors in sunitinib-treated ΕC-βKO mice showed a marked decrease in microvessel density, and reduced new vessel formation. The fraction of perfused mature tumor microvessels was increased in ΕC-βKO mice suggesting immature microvessels were most sensitive to combined sunitinib and PI3Kβ inactivation. Taken together, EC PI3Kβ inactivation with sunitinib inhibition reduces microvessel turnover and decreases heterogeneity of the tumor microenvironment, hence PI3Kβ inhibition may be a useful adjuvant antiangiogenesis therapy with sunitinib.
Article
Full-text available
Blood vascular endothelial cells (BECs) control the immune response by regulating blood flow and immune cell recruitment in lymphoid tissues. However, the diversity of BEC and their origins during immune angiogenesis remain unclear. Here we profile transcriptomes of BEC from peripheral lymph nodes and map phenotypes to the vasculature. We identify multiple subsets, including a medullary venous population whose gene signature predicts a selective role in myeloid cell (vs lymphocyte) recruitment to the medulla, confirmed by videomicroscopy. We define five capillary subsets, including a capillary resident precursor (CRP) that displays stem cell and migratory gene signatures, and contributes to homeostatic BEC turnover and to neogenesis of high endothelium after immunization. Cell alignments show retention of developmental programs along trajectories from CRP to mature venous and arterial populations. Our single cell atlas provides a molecular roadmap of the lymph node blood vasculature and defines subset specialization for leukocyte recruitment and vascular homeostasis. The origin and diversity of blood vascular endothelial cells (BEC) in lymphoid tissues is unclear. Here, the authors profile murine BECs from peripheral lymph nodes by single cell analysis and identify subsets of cells specialised for immune cell recruitment and vascular homeostasis.
Article
Full-text available
Commonly attributed to the prevalence of M2 macrophages, tumor-associated macrophages (TAM) are linked to poor outcome in Hodgkin lymphoma (HL). MYC is supposed to control the expression of M2-specific genes in macrophages, and deficiency in MYC-positive macrophages inhibits tumor growth in mouse models. To verify this hypothesis for HL, seventy-six samples were subjected to immunohistochemical double staining using CD68 or CD163 macrophage-specific antibodies and a reagent detecting MYC. For each cell population, labelled cells were grouped according to low, intermediate and high numbers and related to disease-free survival (DFS) and overall survival (OS). MYC+ cells accounted for 21% and 18% of CD68+ and CD163+ cells, respectively. Numbers of MYC− macrophages were significantly higher in EBV+ cases while no differences were observed for MYC+ macrophages between EBV+ and EBV− cases. Cases with highest numbers of macrophages usually showed worst DFS and OS. In most scenarios, intermediate numbers of macrophages were associated with better outcome than very low or very high numbers. Our observations are reminiscent of the “hormesis hypothesis” and suggest that a relative lack of TAM may allow HL growth while macrophages display an inhibitory effect with increasing numbers. Above a certain threshold, TAM may again support tumor growth.
Article
Full-text available
Angiogenesis is critical for the initiation and progression of solid tumors, as well as hematological malignancies. While angiogenesis in solid tumors has been well characterized, a large body of investigation is devoted to clarify the impact of angiogenesis on lymphoma development. B-cell non-Hodgkin lymphoma (B-NHL) is the most common lymphoid malignancy with a highly heterogeneity. The malignancy remains incurable despite that the addition of rituximab to conventional chemotherapies provides substantial improvements. Several angiogenesis-related parameters, such as proangiogenic factors, circulating endothelial cells, microvessel density, and tumor microenvironment, have been identified as prognostic indicators in different types of B-NHL. A better understanding of how these factors work together to facilitate lymphoma-specific angiogenesis will help to design better antiangiogenic strategies. So far, VEGF-A monoclonal antibodies, receptor tyrosine kinase inhibitors targeting VEGF receptors, and immunomodulatory drugs with antiangiogenic activities are being tested in preclinical and clinical studies. This review summarizes recent advances in the understanding of the role of angiogenesis in B-NHL, and discusses the applications of antiangiogenic therapies.
Article
Background: The leading cause of death after high-dose chemotherapy and autologous stem cell transplantation (ASCT) for relapsed / refractory (R/R) diffuse large B-cell lymphoma (DLBCL) remains disease relapse. Except for specific subsets, DLBCL appears to have relatively low susceptibility to single-agent PD-1 blockade. However, administering PD-1 blockade early after ASCT could leverage the remodeling immune landscape and minimal residual disease state to increase the therapeutic effect of PD-1 blockade and decrease the relapse rate after ASCT. A prior study with pidilizumab showed encouraging results in this setting (Armand et al. JCO 2013;31:4199-206), but the specificity of that antibody is still under investigation. We therefore conducted a phase 2 multi-center single-arm study of the anti-PD-1 monoclonal antibody pembrolizumab (pembro) in patients with chemosensitive DLBCL after ASCT. Another arm of this study enrolled pts with R/R classical Hodgkin lymphoma and will be presented separately. Methods: Patients ≥ 18y with R/R DLBCL who had received no more than 3 lines of prior therapy and had undergone ASCT with chemosensitive disease were enrolled on this study. In addition to meeting standard eligibility criteria for pembro treatment, pts had to have recovered from ASCT toxicities and had to begin study treatment within 60 days of stem cell infusion, with a goal of starting treatment within 21 days of hospital discharge. All patients received pembro 200mg IV every 3 weeks for 8 cycles. PET-CT scans were obtained at post-ASCT baseline, after 3 and 7 cycles, then at 12 and 18 months post-ASCT. The primary endpoint was the progression-free survival rate (PFS) at 18 months after ASCT, assessed using the International Harmonization Project 2007 criteria, with the approach considered promising if 22 patients were alive and in remission at 18 months. Results: 31 pts were enrolled and 2 withdrew consent before starting treatment. Among the 29 eligible pts, median age was 57 (22-76). Prior to ASCT, 18 (62%) were in CR and 11 (38%) were in PR. At study baseline (post-ASCT), 25 (86%) were in CR. 18 pts (62%) completed all 8 cycles of pembro per protocol. 11 pts (38%) stopped pembro early for pt choice (n=1, after febrile neutropenia), toxicity (n=6, including 3 pts with gr3 pneumonitis, 1 with g3 hepatitis, 1 with gr4 aplastic anemia) or progressive disease (n=4). 23 pts (79%) experienced a total of 57 gr3 or higher adverse events (AEs). The most common gr4 AE was neutropenia (n=6, 21%). Concerning related AEs, 9 pts (31%) experienced 14 gr3-4 AEs at least probably related to pembro including neutropenia (4 gr3, 1 gr 4) and pneumonitis (2 gr3). 10 pts (34%) experienced at least one immune-related AE of gr2 or higher severity including pneumonitis (n=1 gr2, n=2 gr3), transaminitis (n=1 gr2, n=1 gr3) and rash (n=1 gr2, n=1 gr3). There were no treatment-related deaths. Among the 29 eligible pts, 1 patient withdrew consent after cycle 1 and 1 pt was lost to follow-up after the 12m assessment (in CR). 27 pts (93%) were evaluable for the primary endpoint. 10 patients (34%, 95% CI: 18-54%) experienced disease relapse at a median of 5 months (3-18) after ASCT, and all other evaluable patients (n=17, 59%, 95% CI: 39-76%) were in CR at the 18m timepoint. 2 pts have died, both of whom had relapse at 3m after ASCT. Correlative studies including immune reconstitution and MRD analyses are ongoing. Conclusions: Pembrolizumab administered after ASCT in patients with R/R DLBCL is feasible with toxicity similar to its use in the R/R setting for other hematological malignancies. The high rate of neutropenia on this study, which is not a common AE of pembro in other settings, may be related to the burden of prior therapy or possibly to an accentuated toxicity of pembro in this specific patient population. The 18-month progression-free rate did not meet the protocol-specified primary objective, and therefore does not support a larger confirmatory study. Future studies in this setting should likely focus on specific subsets of DLBCL, e.g. primary mediastinal BCL, EBV+ DLBCL, T cell histiocyte rich LCL, which may be especially sensitive to PD1 blockade. Disclosures Chen: REGiMMUNE: Consultancy; Magenta Therapeutics: Consultancy; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda Pharmaceuticals: Consultancy. Armand:Pfizer: Consultancy; Adaptive: Research Funding; Merck: Consultancy, Research Funding; Infinity: Consultancy; Affimed: Consultancy, Research Funding; Otsuka: Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding; Roche: Research Funding; Tensha: Research Funding. Herrera:Seattle Genetics: Research Funding; KiTE Pharma: Consultancy, Research Funding; Gilead Sciences: Research Funding; Merck, Inc.: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding; Pharmacyclics: Consultancy, Research Funding; AstraZeneca: Research Funding; Genentech: Consultancy, Research Funding; Immune Design: Research Funding. LaCasce:Humanigen: Consultancy, Honoraria; Bristol-Myers Squibb: Other: Data safety and monitoring board; Research to Practice: Speakers Bureau; Seattle Genetics: Consultancy, Honoraria. Jacobson:Pfizer: Consultancy; Precision Bioscience: Consultancy; Kite: Consultancy; Novartis: Consultancy; Bayer: Consultancy; Humanigen: Consultancy. Jacobsen:Merck: Consultancy; Seattle Genetics: Consultancy. Rodig:Merck & Co., Inc.: Research Funding; Affimed Inc.: Research Funding; KITE Pharma: Research Funding; Bristol-Meyers-Squibb: Research Funding. Shipp:AstraZeneca: Honoraria; Merck: Research Funding; Bayer: Research Funding; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
Article
Significance Complex transcription factor networks regulate the development, maturation, and lineage commitment of dendritic cell (DC) subsets. Here, we demonstrate a previously unexpected role of the transcription factor C/EBPβ in murine DC maturation and immunogenic functionality under homeostatic and lymphoma-transformed conditions. Regulated expression of functional C/EBPβ isoforms enables a controlled maturation of DCs. In contrast, the presence of lymphoma cells leads to an up-regulation of C/EBPβ in DCs which transforms them into an immature and protumorigenic subtype. This study also shows that inhibition of the C/EBPβ/mTOR signaling axis abrogates the protumorigenic function of human DCs, suggesting that inhibitors regulating C/EBPβ activity can be used for blocking tumor-promoting functions of DCs in the treatment of hematological neoplasms.
Article
Despite idelalisib approval in relapsed follicular lymphoma (FL), a complete characterization of the immunomodulatory consequences of phosphatidylinositol 3-kinase δ (PI3Kδ) inhibition, biomarkers of response, and potential combinatorial therapies in FL remain to be established. Using ex vivo cocultures of FL patient biopsies and follicular dendritic cells (FDCs) to mimic the germinal center (n = 42), we uncovered that PI3Kδ inhibition interferes with FDC-induced genes related to angiogenesis, extracellular matrix formation, and transendothelial migration in a subset of FL samples, defining an 18-gene signature fingerprint of idelalisib sensitivity. A common hallmark of idelalisib found in all FL cases was its interference with the CD40/CD40L pathway and induced proliferation, together with the downregulation of proteins crucial for B-T-cell synapses, leading to an inefficient cross talk between FL cells and the supportive T-follicular helper cells (TFH). Moreover, idelalisib downmodulates the chemokine CCL22, hampering the recruitment of TFH and immunosupressive T-regulatory cells to the FL niche, leading to a less supportive and tolerogenic immune microenvironment. Finally, using BH3 profiling, we uncovered that FL-FDC and FL-macrophage cocultures augment tumor addiction to BCL-XL and MCL-1 or BFL-1, respectively, limiting the cytotoxic activity of the BCL-2 inhibitor venetoclax. Idelalisib restored FL dependence on BCL-2 and venetoclax activity. In summary, idelalisib exhibits a patient-dependent activity toward angiogenesis and lymphoma dissemination. In all FL cases, idelalisib exerts a general reshaping of the FL immune microenvironment and restores dependence on BCL-2, predisposing FL to cell death, providing a mechanistic rationale for investigating the combination of PI3Kδ inhibitors and venetoclax in clinical trials.
Article
Purpose of review: This article focuses on the immunosuppressive impact of myeloid-derived suppressor cells (MDSCs) and the potential clinical implications in hematological malignancies. Recent findings: MDSCs play a critical role in the regulation of the immune response in cancer. They inhibit activation of adaptive immune response and as a result foster the growth of the malignancy. Recent studies have shown that MDSCs serve as prognostic biomarkers and as targets for cancer immunotherapy. Preclinical and clinical studies have identified new approaches to deplete MDSC populations and inhibit MDSC function with combination immunomodulatory therapies including chemotherapeutic agents with immune checkpoint-directed treatment. Summary: A broad spectrum of publications indicate that direct targeting of MDSCs may abrogate their protumorigenic impact within the tumor microenvironment through activation of the adaptive immune response.