ANGIOGENESIS
ANGIOGENESIS
ANGIOGENESIS
com/sigtrans
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic
molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology,
various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been
elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising
strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase
inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still
been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated
biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the
combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling
pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose
several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and
1234567890();,:
1
Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009 Nanjing, China
Correspondence: Li-Ping Sun ([email protected]) or Lei Shi ([email protected])
Fig. 1 The progression of the canceration through angiogenesis. The rapid expansion of tumor results in a reduction in the oxygen supply.
The consequent hypoxic tumor microenvironment stimulates excessive angiogenesis via increasing various angiogenic pro-factors including
VEGF, PDGF, FGF, and angiopoietin. Later, new blood vessels facilitate the transportation of oxygen and nutrients to further support the
survival, growth and proliferation of tumor cells. When tumor cells develop a more aggressive phenotype, they continue to proliferate, spread
and induce angiogenesis, with the invasion and metastasis of tumor cells into distant tissues through blood circulation
stimulation, inflammatory response, oncogene mutation, and drug efficacy may be caused by compensatory angiogenesis induced
treatment to aggravate tumor angiogenesis and further promote by alternative pro-factors, vessel co-option and other abnormal
tumor invasion and metastasis.26 modes. Hence, great efforts have been devoted to further
Up to now, although a significant number of research has been improving the therapeutic efficacy and mitigating drug resistance.
devoted to anti-cancer therapy to overcome this incurable and For example, a number of multi-targeted angiogenic inhibitors
lethal disease, none of them has achieved persistent clinical have been developed for cancer treatment. Additionally, the
efficacy.27,28 For example, chemotherapy is a form of systemic combination of angiogenic inhibitors with other conventional
treatment, which has been utilized for the treatment of cancer for cancer treatment including chemotherapy, radiotherapy, immune
over 70 years and remains a cornerstone in the treatment of many therapy, adoptive cell therapy, and cancer vaccines has been
types of cancers including BC, CRC, and NSCLC by directly killing evidently demonstrated through many pivotal clinical trials
or inhibiting the growth and reproduction of tumor cells under among patients with different types of cancer.47 With the in-
the administration of various cytotoxic agents such as cis- depth exploration of the tumor angiogenesis and drug resistance,
platinum, 5-fluorouracil, cyclophosphamide, methotrexate and great progress has been made in anti-tumor therapy in recent
doxorubicin.29,30 These cytotoxic chemotherapeutics have indis- years.
criminating cell lethality, poor tissue selectivity, and severe In the present review, we highlight the potent effects of
systemic adverse effects, resulting in poor tolerance and prognosis angiogenesis in tumor growth, proliferation, carcinogenesis,
of patients. Even so, tumor cells are not entirely killed, drug invasion and metastasis, summarize multiple signaling pathways
resistance rises unavoidably.31,32 Prior works have demonstrated in tumor angiogenesis and outline the development of anti-
that congenital and acquired drug resistance can be derived from angiogenic therapies, as well as classic anti-angiogenic drugs and
tumor genetic and phenotypic mutations.33–36 Furthermore, some potential clinical candidates. Moreover, we discuss the
cancerous tumors can escape into remote normal organizations challenges of anti-angiogenic treatment and some emerging
through blood and lymphatic circulation to invalidate the drugs therapeutic strategies to exploit the great advantages of anti-
and worsen the condition of patients.37–40 As an emerging angiogenic therapy.
treatment, anti-angiogenic therapy fights cancer by normalizing
tumor blood vessels, alleviating hypoxia of microenvironment,
increasing tissue concentration of drugs, and limiting distant PATHOPHYSIOLOGY
invasion and metastasis of tumors.41,42 Despite the ever-growing Blood circulation is a basis of cell metabolism, which flows in a
list of FDA-approved drugs, the clinical benefits of anti-angiogenic closed circuit from the heart to arteries, capillaries, veins, and
monotherapies are not long-lasting. Some limitations in che- finally back to the heart. In normal tissue, tight pericyte coverage
motherapy like acquired drug resistance and tumor recurrence and vascular endothelial cell junction ensure regular blood
have also been found in anti-angiogenic therapy.43–46 The limited circulation, forming a mature vascular structure.48 However, in
Fig. 2 Most common modes in tumor angiogenesis. a Sprouting angiogenesis: main way in both physiological and pathological
angiogenesis, which is induce by proliferation and migration of endothelial tip cells. b Intussusception: the existing blood vessel is divided
into two vessels under mediation of cell reorganization. c Vasculogenesis: bone-marrow-derived endothelial progenitor cells differentiate into
endothelial cells, participating in the formation of new vascular lumen. d Vessel co-option: tumor cells approach and hijack the existing blood
vessels. e Vessel mimicry: tumor cells form a vessel-like channel around normal blood vessels to direct the transport of oxygen and nutrients
into tumor tissue. f Trans-differentiation of cancer cells: cancer stem-like cells differentiate into endothelial cells, which participate in the
formation of new blood vessels. (Modified from Carmeliet, P. & Jain, R. K. Molecular mechanisms and clinical applications of angiogenesis.
Nature 473, 298–307 (2011).)
tumor tissue, more mechanical stress from the hypertrophic tumor lesions.68,69 Because of the tenacious viability, various pro-
tissue results in an uneven thickness and deformed architecture of angiogenic factors are secreted by tumor cells to stimulate
tumor vessels, which exhibit intensive sprouting orchestrated in endothelial cells proliferation and migration, promote vessel
an irregular convoluted manner that tends to hinder blood formation, increase blood circulation to meet the requirements
flow.49–51 Mechanical stress also disrupts lymphatic channels and of the tumor, and mitigate metabolic stress.
prevents lymphatic drainage of excess interstitial fluid. Besides, Tumor angiogenesis occurs mainly through any of the following
fragile and highly permeable tumor vessels, which have an modes described in Fig. 2. Among them, sprouting angiogenesis is
irregular arrangement of endothelial cells and thinly covered the most typical process in physiological and pathological
pericytes, lead to blood leakage and incoherent perfusion.52–54 angiogenesis. The patterns of vessel co-option and vessel mimicry
This spatially anomalous structure is manifested in low blood flow, are significantly related to tumor invasion, metastasis, and
which decreases the supply of oxygen and nutrient, causing therapeutic resistance in conventional anti-angiogenic therapy.
subsequent acidosis and hypoxia within tumor microenvironment Sprouting angiogenesis is so-called angiogenesis, in which new
and high interstitial hypertension.55 Highly permeable tumor vascular branches form in existing blood vessels and finally
blood vessels facilitate plasma and proteins into the surrounding infiltrate into tumor tissue through the migration of tip cells and
interstitium, increasing blood viscosity and interstitial pressure in the proliferation of stem cells (Fig. 2a).70,71 Intussusceptive
tumor microenvironment, further impeding blood flow.56–58 All angiogenesis involves the formation of a double lumen, which
these factors result in chaotic function and abnormal architecture splits into two vessels, infiltrating into tumor tissue (Fig. 2b).72,73
of tumor blood vessels, further aggravating acid and hypoxic Vasculogenesis refers to recruiting bone marrow-derived or vessel
tumor microenvironment, which contributes to tumor angiogen- wall resident endothelial progenitor cells, which differentiate into
esis, invasion, and metastasis.59,60 endothelial cells to form new blood vessels (Fig. 2c).71,74 In
Studies have shown that 50–60% of solid tumors are hypoxic, addition to the above three models, tumors can achieve
which disrupts the expression of multiple tumor genes profiles angiogenesis through vessel co-option, vessel mimicry, lymphan-
and causes tumor necrosis, stimulating the spread and metas- giogenesis, and rare stromal-sharing modes.3,24,75 Vessel co-
tasis of tumor.61 Since tumor growth and reproduction require option, in which tumor cells migrate around pre-existing blood
substantial energy, the tumor cells in a hypoxic environment are vessels or infiltrate into surrounding tissue space, eventually
forced to release energy through glycolysis and secrete wrapping the blood vessels and leading them into tumor tissue to
considerable acidic substances, aggravating the acidity of the supply nutrients for tumor cells (Fig. 2d).76 Vessel mimicry is a
microenvironment (pH is usually between 6.5–7.2, or even process that tumor cells extend to form a simulated vascular
lower).62,63 Furthermore, the interstitial pressure in normal lumen and then insert into the pre-existing blood vessels,
tissues is only 0–3 mmHg.64 In tumor tissue, high interstitial transporting the erythrocyte and oxygen into tumor tissue (Fig.
pressure (5–40 mmHg, even 75–130 mmHg in some cases) 2e).77 Researchers believe that vessel mimicry is closely connected
hinders the transport of blood and drugs,65 which is caused by with hypoxia, which stimulates the secretion of matrix metallo-
blood leakage of tumor vessels and the increase of interstitial proteinases (MMPs) and periodic acid Schiff-positive substances to
fluid, thus the tumor cannot obtain sufficient oxygen and irritate the formation of vascular mimicry.77 Another mode is that
nutrients.66,67 These factors affect the drug treatment and trans-differentiation of cancer stem-like cells (which obtain the
benefit tumor proliferation, adhesion, invasion, and metastasis, endothelial phenotype) into endothelial-like cells via epithelial-
eventually leading to tumor resistance and malignant endothelial transformation (Fig. 2f).
Fig. 3 Schematic diagram showing crosstalk of multiple signaling pathways during tumor angiogenesis. Pointed arrows indicate activation
whereas flat arrows indicate inhibition
Monoclonal antibodies
Bevacizumab (Avastin®) VEGF-A CRC in 2004, Genentech/Roche Arterial or venous, back pain, dry skin,
NSCLC in 2006, exfoliative dermatitis gastrointestinal
RCC in 2009, perforation, headache, hemorrhage,
GBM in 2009, hypertension, lacrimation disorder,
CC in 2014 poor wound healing, proteinuria,
rhinitis, and taste alteration, thrombosis
Ranibizumab (Lucentis®, RG- VEGF-A wAMD in 2006, Genentech/Roche Conjunctival hemorrhage,
6321) DME in 2015, endophthalmitis, eye infection, eye
DR in 2017, pain, floaters, increased intraocular
Myopic choroidal pressure, rhegmatogenous retinal
neovascularization in 2017 detachment, and retinal hemorrhage
Ramucirumab (Cyramza®) VEGFR-2 NSCLC in 2014, Advanced GC in Genentech and Eli Abdominal pain, thrombocytopenia,
2014, GEJ adenocarcinoma in Lilly anorexia, arthralgia, constipation,
2014, metastatic CRC in 2015 cough, diarrhea, dyspnea, epistaxis,
fatigue, headache, hypertension,
leucopenia, nausea, neutropenia,
peripheral edema, proteinuria, upper
respiratory tract infection, and vomiting
Olaratumab (Lartruvo®) PDGFR-α STS in 2016 ImClone/Eli Lilly Appetite, abdominal pain, alopecia,
diarrhea, decreased fatigue, headache,
neuropathy, musculoskeletal pain,
mucositis, nausea, and vomiting
Bevacizumab-awwb (Mvasi®) VEGF CRC, NSCLC, RCC, GBM, and CC Amgen Altered taste, arterial and venous
in 2017 thromboembolic events, bleeding, dry
skin, epistaxis, exfoliative dermatitis,
headache, hypertension, hypertension,
infusion-related reactions, lacrimation
disorders, ovarian failure, perforation or
fistula, post-reversible encephalopathy
syndrome, proteinuria, proteinuria, and
rhinitis
Oligonucleotide aptamers
Pegaptanib (Macugen®) VEGF-A165 wAMD in 2004 Eyetech/Pfizer Endophthalmitis and retinal
detachment
Recombinant fusion proteins
Aflibercept (Eylea®) VEGF-A, VEGF-B, PlGF wAMD in 2011 Regeneron Cataracts, conjunctival hemorrhage,
CRC in 2012 decreased vision, eye pain, floaters,
DME in 2015 increased intraocular pressure, and
DR in 2019 vitreous detachment
ziv-Aflibercept (Zaltrap®) VEGF-A, VEGF-B, PlGF CRC in 2012 Sanofi and Abdominal pain, bleeding, decreased
Regeneron appetite, decreased ejection fraction,
diarrhea, dyspnea, epigastric pain,
fatigue, fatigue, gastrointestinal
perforation, headache, heart failure,
hypertension, impaired wound healing,
infection, leukopenia, nephrotic
syndrome, neutropenia, osteonecrosis
of the lower jaw, proteinuria, severe
diarrhea, stomatitis, thrombocytopenia,
and weight loss
mTOR inhibitors
Temsirolimus (Torisel®) mTOR RCC in 2007 Wyeth Acute renal failure, asthenia, edema,
elevated aspartate aminotransferases,
hyperlipidemia, hypersensitivity,
interstitial pneumonia, intestinal
perforation, lymphopenia, mucositis,
nausea, rash, thrombocytopenia
Everolimus (RAD001, mTOR RCC in 2009 Novartis Canker sores, increased heart rate,
Afinitor®) SEGA in 2010 paronychia, rash, swollen and painful
pNET in 2011 gums, tiredness, and tongue ulcers
HER2- BC
Immunomodulatory agents
Thalidomide (Thalomid®) VEGF-A, TNF, NF-κB MM in 2006 Celgene Abdominal pain, constipation,
dizziness, drowsiness, dry oral mucosa,
facial puffiness, nausea, rash,
teratogenic, and tiredness
Lenalidomide (Revlimid®) VEGF-A, TNF, NF-κB MM in 2006 Celgene Anemia, diarrhea, fatigue, headache,
MCL in 2013 loss of appetite, low back pain, neo-
malignant neoplasms, neutropenia,
rash, renal insufficiency,
thrombocytopenia, and thrombotic
complications
Sunitinib (Sutent®, SU11248) VEGFR-1/-2/-3, Flt-3, RCC in 2006 Sugen/Pfizer Abdominal pain, anorexia, asthenia,
c-Kit, Ret, PDGFR-α/- GIST in 2006 diarrhea, dysgeusia, dyspepsia, fatigue,
β, CSF-1R pNET in 2011 hypertension, mucositis, nausea, skin
discoloration, stomatitis, and
thrombocytopenia
Vandetanib (Caprelsa®, VEGFR-2, VEGFR-3, MTC in 2011 AstraZeneca Diarrhea, headache, rash, hypertension,
ZD6474) EGFR, Ret nausea, and QTc prolongation
Regorafenib (Stivarga®) VEGFR-1/-2/-3, c-Kit, CRC in 2012 Bayer Anorexia, diarrhea, fatigue, hand-foot
PDGFR-β, Ret, Raf-1, GIST in 2013 skin reaction, hypertension, and oral
bRaf, FGFR-1, Tie-2 HCC in 2017 mucositis
Axitinib (Inlyta®, AG013736) VEGFR-1/-2/3, c-Kit, RCC in 2012 Pfizer Asthenia, constipation, decreased
PDGFR-α, PDGFR-β appetite, diarrhea, dysphonia, fatigue,
hand-foot syndrome, hypertension,
nausea, vomiting, and weight
decreased
Cabozantinib (Cometriq®, VEGFR-2, c-Met, c-Kit, MTC in 2013 Exelixis Abdominal pain, constipation,
BMS-907351) Ret, Flt-3, Tie-2, AXL, RCC in 2016 decreased appetite, decreased weight,
RON HCC in 2019 diarrhea, dysgeusia, fatigue, hair color
changes, hypertension, nausea, oral
pain, palmar-plantar erythrodysesthesia
syndrome, and stomatitis
Apatinib (Aitan®) VEGFR-2, Src, c-Kit GC in 2014 Hengrui Medicine Fatigue, gastrointestinal bleeding,
granulocytopenia, hand-foot syndrome,
hoarseness, hypertension, leukopenia,
proteinuria, and thrombocytopenia
Nintedanib (Ofev®, BIBF1120) VEGFRs, FGFRs, NSCLC in 2015 Boehringer Bleeding, decreased appetite, diarrhea,
PDGFRs, Flt-3, LCK, and Ingelheim electrolyte imbalance, mucositis,
LYN, Src nausea, neutropenia, peripheral
neuropathy, rash, and vomiting
ALL acute lymphoblastic leukemia, BC breast cancer, BTC biliary tract cancer, CC cervical cancer, CML chronic myeloid leukemia, CRC colorectal cancer, CSF
colony-stimulating factor, DME diabetic macular edema, DR diabetic retinopathy, DTC differentiated thyroid cancer, EC esophageal cancer, GEJ
gastroesophageal junction, GBM glioblastoma, GC gastric cancer, GIST gastrointestinal stromal tumor, HCC hepatocellular carcinoma, HER2 human epidermal
growth factor receptor 2, HNSCC head and neck squamous cell carcinoma, MCL mantle cell lymphoma, MM multiple myeloma, MTC medullary thyroid cancer,
mTOR mammalian target of rapamycin, NSCLC non-small cell lung cancer, Ph+ AML Philadelphia chromosome-positive acute myeloid leukemia, pNET pancreas
neuroendocrine tumor, RCC renal cell carcinoma, SEGA subependymal giant cell astrocytoma, STS soft tissue sarcoma, TC thyroid cancer, TNBC triple-negative
breast cancer, wAMD wet age-related macular degeneration
FGFR is a transmembrane receptor family with five members of demonstrated that the exon 14 mutation of c-Met promotes the
FGFR1–5 (only FGFR5 lacks an intracellular kinase domain),163 metastasis of advanced cancer, like lung adenocarcinoma, RCC,
whose genes are proto-oncogenes with tumorigenic potential and brain glioma.189 Besides, drug resistance in treatment with
after gene amplification, chromosomal translocation or point EGFR kinase inhibitors is partly attributed to signaling crosstalk
mutation.166–168 FGFR mediates the survival, multiplication, and between similar EGFR and c-Met.155,190 All of these functions
migration, angiogenesis, and drug resistance in target cells above are achieved through activation of downstream signaling
through autophosphorylation and activating downstream Src pathways including JAK/STAT,191 Ras/MAPK,192 PI3K/AKT,192 Wnt/
family kinases,169 PLCγ/DAG/PKC,157,163 Ras/Raf-MAPK, and PI3K/ β-catenin, or others (Fig. 3).193,194 As the role of the HGF/c-Met
AKT pathways activated by bFGF, playing a pro-angiogenic role in system in pathological and physiological angiogenesis and drug
the human body (Fig. 3).159,170,171 In tumor angiogenesis, FGF/ resistance continues to be revealed, HGF/c-Met axis becomes an
FGFR signaling plays a key role in stimulating the secretion of attractive target for anti-tumor therapy.
MMPs and regulates the proliferation, differentiation, migration,
morphological changes, and vascular maturation of endothelial IGF/IGFRs. Insulin-like growth factor (IGF) is a peptide growth
cells.171 Aberrant activations of bFGF/FGFR are essential alter- factor that regulates human growth, development, and energy
native angiogenic pathways that induce drug resistance in anti- metabolism, which participates in physiological circulation
VEGFR therapy.133,172 through autocrine, paracrine, and endocrine.195 IGF modulates
the survival, proliferation, and differentiation of multiplicate cells
HGF/c-Met. The hepatocyte growth factor (known as the scatter- and the physiological process of the blood system under different
ing factor) is a multi-effect precursor protein and a mitogen of physiological conditions.196 IGF1 and IGF2 are the two main
mature rat hepatocytes,173 mainly derived from mesenchymal subtypes mediated by insulin receptors IGF1R, IGF2R and
cells and activated by extracellular protease cleavage.174 As a IGFBPs.197,198 Highly expressed IGF1 and IGF2 induces VEGF
soluble heterodimer, HGF can be cleaved into α chain and β chain. synthesis and up-regulates the expression of HIF-1α and VEGF to
α chain is responsible for binding receptors while β chain can promote angiogenesis. Besides, autocrine IGF2 induces drug
trigger receptors and transduce signals.175 Transmembrane helical resistance in anti-tumor therapy.195 Moreover, studies have shown
receptor c-Met is a 170 kDa cell-stroma-epithelium transition that over-expression of IGF fosters the progression of diabetic
factor usually expressed on endothelial cells, epithelial cells, and retinopathy (DR),199,200 retinopathy of prematurity,201–203 athero-
melanocytes in the pathological liver, kidney, lung and other sclerosis, and cancer.204–207
organs.155,173,176 c-Met was firstly discovered as a proto-oncogene IGFBPs are high-affinity receptors of IGF, with six subtypes of
in 1984 and later identified as a specific receptor for HGF in IGFBP1–6, secreted by endothelial cells living in macro-vessels and
1991.177,178 Owing to instinctively actuate cell growth, differentia- capillaries.198 Pro-angiogenic IGFBP2 induces chemotaxis and
tion, morphogenesis and suppress apoptosis, HGF/c-Met is a migration of ECs by increasing VEGF transcription and IGF
crucial signaling pathway in wound healing, tissue regeneration levels.208,209 IGFBP3 up-regulates the expression of VEGF, MMP2,
and embryogenesis.155,173,175 Inhibition of this pathway will and MMP9 and promotes tube formation.210 IGFBP4, IGFBP5, and
seriously affect the self-repair of patients with myocardial IGFBP6 appear to inhibit angiogenesis indirectly.196 More studies
ischemia,179 diabetic retinopathy,180 liver damage,181 and arthri- are expected to dissect the roles and mechanisms of IGF family in
tis.182 Nevertheless, abnormal HGF/c-Met signals such as amplifi- tumor angiogenesis.
cation or secondary mutation of c-Met genes, transcription
dysregulation, and abnormally autocrine or paracrine of HGF TGF-β. In 1978, a signaling protein with multiple biological
caused by over-expression of c-Met, encourages the spread, effects, named transforming growth factor-β (TGF-β), was
invasion and angiogenesis of cancerous tissues,183–185 drug discovered by scientists in mouse fibroblasts. TGF-β is a secreted
resistance, and poor prognoses of patients.175,186–188 It has been cytokine that is concerned with body homeostasis, tissue repair,
Fig. 4 The transduction of HIF-1α in normal and hypoxic conditions. Under normal conditions, HIF-1α is degraded by protease and loses
transcription function. In hypoxic environment, lack of enzyme degradation leads to efficient transcription of HIF-1α, resulting in over-
expression of pro-angiogenic factors including VEGF, PDGF, and MMPs
inflammation, and immune responses,211 which is also involved in unsatisfactory clinical outcomes. Accordingly, TGF-β simulta-
cell growth, differentiation, proliferation, autophagy, apoptosis, neously promotes tumorigenesis and induces angiogenesis to
and tumor angiogenesis.212 There are three types of single-pass nourish tumors. Perhaps TGF-β is the next breakthrough to fight
transmembrane receptors specifically interact with TGF-β, named against tumor angiogenesis and drug resistance.
type I (TβRI), type II (TβRII) and type III (TβRIII). Under the co-
transduction of these receptors, the downstream Smad- Transcription factors
dependent pathways, and non-Smad pathways (involves classical Hypoxia-inducible factor-1. Hypoxia is the most typical feature of
MAPK, JNK/p38 MAPK, PI3K/AKT, TAK1, and ERKs) are alternately the tumor microenvironment and is always associated with drug
activated to exert the physiological and pathological effects of resistance, tumor angiogenesis, aggressiveness, and recurrence.239
TGF-β.213–215 Apart from various physiological processes, TGF-β The hypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcrip-
involves in multiple atherosclerosis and fibrotic diseases like tion factor that regulates cell adaptation to hypoxia, energy
cirrhosis and pulmonary fibrosis and affects cancer progres- metabolism, erythropoiesis, and tissue perfusion balance and
sion.216,217 During the initial stage of tumorigenesis, TGF-β acts as involves in cell survival, proliferation, migration, adhesion,
a suppressor that induces apoptosis and confines pre-cancerous apoptosis, erythropoiesis, and glucose metabolism.240,241 HIF-1 is
cells.216,218 But in mature tumor tissue, the aggressiveness of the composed of the constitutive nuclear protein HIF-1β and
tumor is awakened by TGF-β, which encourages various pro- environment-dependent isomer HIF-1α,151 which is an oxygen
cancer activities, including epithelial-mesenchymal transition regulator that increases oxygen delivery, reduces oxygen con-
(EMT), metastasis, invasion, fibrosis, angiogenesis, and immune sumption and maintains oxygen balance.240–242 HIF-2α and HIF-3α
suppression of carcinomas.219–221 are the analogs of HIF-1α which are not well understood.241,243
The tumorigenic effects of TGF can be manifested in various Under normoxic conditions, the proline residues in HIF-1α are
modes. Firstly, TGF-β induces the migration of endothelial cells to hydroxylated by the proline hydroxylase domain (PHD), which can
impel vessel sprouting.222,223 Secondly, TGF-β encourages infiltra- stabilize HIF-1α. Subsequently, HIF-1α is degraded by proteasomes
tion and invasion of the tumor through EMT.224 Thirdly, TGF-β up- after ubiquitination mediated by E3 ubiquitin ligase and ρVHL.
regulates the expression of MMP-2 and MMP-9 to mobilize tumor Besides, hydroxylation of asparagine residues, which regulates
invasiveness and angiogenesis.224–226 Last but not least, TGF-β HIF-1α transcriptional activity and specificity, disrupts the interac-
induces the expression of connective tissue growth factor tion between HIF-1α and co-activation factor p300 to inhibit the
(CTGF),227 VEGF, bFGF, and interleukin-1, which are essential for transcriptional activity of HIF-1α, consequently inhibiting the
tumor angiogenesis.228,229 Many studies have demonstrated that expression of VEGF and angiogenesis (Fig. 4).151,239 However, since
TGF-β is closely related to the tumorigenesis and poor prognosis the hydroxylation under hypoxic conditions can be limited by
of patients in multifarious human organizations. For example, high oxygen concentration, HIF-1α constitutes a dimerized complex
tissue concentrations of TGF-β have been detected in human with HIF-1β through nuclear translocation. This complex binds the
pancreatic cancer,230–233 NSCLC,234 HCC,235–237 and BC,238 which hypoxia response element (HRE) (located on the HIF target) after
motivates tumor progression and angiogenesis, leading to interacting with the coactivator p300, subsequently activating the
Fig. 5 MMP-expressing stromal cells and functions of MMPs in tumor microenvironment. MMP precursors which are secreted by endothelial
cells, fibroblasts, and lymphocytes et al. converted into active MMPs through enzymolysis. Subsequently, active MMPs participate in different
biological processes including angiogenesis and tissue invasion by degrading specific extracellular matrix components
endogenous angiogenesis, angiostatin is a partial fragment of (NOS),351 pleiotrophin (PTN),352 steroid hormones,353 thrombos-
plasminogen that potently inhibits ECs proliferation.337 Coinci- pondin (TSP),354,355 and many other molecules also involve tumor
dentally, before the 2000s, scientists found that MMP-7 could angiogenesis to encourage tumor progression. The specific roles
hydrolyze the Pro (466)-Val (467) peptides bond, and MMP-9 could and mechanisms of these biomolecules in angiogenesis and
hydrolyze the Pro (465)-Pro (466) bond between cyclic domains 4 tumorigenesis will gradually be explored by researchers.
and 5 of human plasminogen, finally producing angiostatin
fragments with potential anti-angiogenic effects.81 Later, in
2002, studies reported that MMP-2/-3/-12 could cleave plasmino- ANTI-ANGIOGENIC THERAPY: A VALUABLE STRATEGY FOR
gen to create angiostatin fragments, and MMP-3/-9/-13/-20 were CANCER TREATMENT
related to the production of endostatin.338 The physiological and The concept of angiogenesis has been proposed for more than 50
pathological functions of the MMPs family are significantly specific years, and the initial understanding is only “angiogenesis in
to different internal environments and a comprehensive study of tumor”: the growth, survival and proliferation of tumor rely on
their processes will be long-term research. angiogenesis after the tumor beyond a certain volume. At present,
In an intricate angiogenic system, almost all biomolecules act in this theory has been extended to various non-neoplastic diseases
interrelated manners to activate the proliferation, survival, such as cardiovascular disease, rheumatoid arthritis (RA), and
migration, and morphogenesis of target cells to excite tumor diabetic retinopathy.
angiogenesis. Apart from the factors above and downstream The formation of new blood vessels has been observed since
pathways shown in Fig. 3, Apelin/APLNR family,339 Slit/Robo the earliest time, especially wound healing. But this process has
family,340 adrenomedullin,341,342 COX-2,343,344 CXC chemo- only ever been regarded as a simple pathological or physiological
kines,345,346 interleukins,347 interferons,348–350 nitric oxide synthase process unrelated to malignancies. In the 1860s, some researchers
Fig. 6 Diagramatic illustrations of the relationship between tumor blood vessels, pro-angiogenic and anti-angiogenic factors. a Blood vessels
with regularity and completeness depend on dynamic balance of pro-factors and anti- factors in normal tissues. b Abnormal vessels with
chaos, leakage and feeble blood circulation are caused by imbalance of mediators in tumor tissue. c Blood vessels are repaired through
neutralizing abundant pro-factors or increasing anti-factors under the guidance of angiogenic inhibitors. d Blood vessels in tumor tissue are
destroyed by excessive inhibitors, which aggravates hypoxia within tumor tissue and hinders drug transportation
44.8%, and the median duration of response increased 3.3 months combining multiple chemotherapy drugs to treat recurrent or
owing to the addition of bevacizumab.380 Hence, bevacizumab metastatic malignant tumors. For example, the combination of
was approved by FDA for patients with CRC in 2004. In addition to bevacizumab, carboplatin, and paclitaxel (or gemcitabine) was
the first indication, bevacizumab has been approved for a variety approved by FDA for later treatment after bevacizumab mono-
of other cancers as monotherapy, as a surgical adjuvant, or in therapy in platinum-sensitive recurrent epithelial ovarian cancer in
combination with chemotherapy, and more potential in anti- 2016.44,75 Then in 2020, bevacizumab was approved for untreated
angiogenic therapy is being tested through clinical trials.381 locally advanced or metastatic RCC in combination with mono-
Moreover, because of excellent anti-angiogenic activity, bevaci- clonal antibody atezolizumab, an immune checkpoint inhibitor of
zumab has achieved satisfactory results in several clinical trials by PD-L1.75 Bevacizumab is an anti-angiogenic drug with excellent
Antibodies
Bemarituzumab (FPA144) FGFR-2 I Not yet Solid tumors (unspecified) NCT05325866
recruiting
III Recruiting GC or CEJ adenocarcinoma NCT05052801
JY-025 VEGFR-2 II/III Not yet NSCLC with EGFR 19 exon deletion or 21 exon mutation NCT04874844
recruiting
BAT-5906 VEGFR III Not yet wAMD NCT05439629
recruiting
II Completed wAMD NCT05141994
I/II Recruiting DME NCT04772105
Olinvacimab VEGFR-2 II Recruiting Metastatic TNBC NCT04986852
I/II Not yet Metastatic CRC who failed two prior standard NCT04751955
recruiting chemotherapies
Ak109 VEGFR-2 I/II Recruiting Advanced solid tumor NCT05142423
I/II Recruiting Advanced gastric adenocarcinoma or GEJ adenocarcinoma NCT04982276
I Unknown Advanced solid tumors NCT04547205
CTX-009 (ABL001) VEGF-A, Dll-4 I/II Recruiting Advanced or metastatic solid tumors; unresectable NCT04492033
advanced, metastatic or recurrent BTC
NOV-1105 (YYB-101) HGFR I/II Recruiting Metastatic or recurrent CRC NCT04368507
MCLA-129 c-Met, EGFR I/II Recruiting Advanced NSCLC or other solid tumors NCT04930432
I/II Recruiting Metastatic or advanced NSCLC, HNSCC or other solid tumors NCT04868877
SYD-3521 (BYON3521) c-Met I Recruiting Locally advanced or metastatic solid tumors NCT05323045
VRDN-001 IGF-1 I/II Recruiting TED NCT05176639
Oligonucleotide agents
IGV-001 – II Not yet GBM or GBM multiforme NCT04485949
recruiting
Anti-angiogenic fusion proteins
9MW-0813 VEGFR III Recruiting DME NCT05324774
I Completed DME NCT05324592
Tyrosine kinase inhibitors
Surufatinib VEGFR-1/-2/-3, II Recruiting Advanced CRC who failed front-line anti-angiogenic TKI NCT05372198
CSF1R, FGFR-1 therapy
II Recruiting Advanced HCC NCT05171439
II Recruiting HR+ unresectable metastatic BC refractory to endocrine NCT05186545
therapy
II Recruiting High-grade advanced-neuroendocrine neoplasm NCT05165407
II Not yet Inoperable or metastatic advanced intrahepatic NCT05236699
recruiting cholangiocarcinoma (ICC)
II Not yet OC with platinum-resistance and received prior PARP NCT05494580
recruiting inhibitors
II Not yet Advanced gastric adenocarcinoma or GEJ adenocarcinoma NCT05235906
recruiting
Olverembatinib (GZD824) Bcr-Abl, c-Kit III Recruiting CML in chronic phase who are resistant and/or intolerant to NCT05311943
at least two second-generation tyrosine kinase inhibitors
II Recruiting Myeloproliferative neoplasms, ALL or AML with FGFR1 NCT05521204
rearrangement
II Recruiting Advanced CML NCT05376852
II Not yet Ph+ ALL NCT05466175
recruiting
I Not yet Relapsed or refractory Ph+ ALL NCT05495035
recruiting
Pemigatinib FGFR-1/-2/-3 III Recruiting Unresectable or metastatic cholangiocarcinoma with FGFR2 NCT03656536
rearrangement
II Completed Advanced/Metastatic or surgically unresectable NCT02924376
cholangiocarcinoma with FGFR2 translocations who failed
previous therapy
II Recruiting Previously treated GBM or other primary central nervous NCT05267106
system tumors with FGFR1–3 alterations
II Recruiting GBM, or other primary CNS tumors, or adult-type diffuse NCT05267106
gliomas with FGFR mutation
II Recruiting Advanced NSCLC with FGFR alterations who have failed NCT05287386
standard therapy
II Recruiting Advanced GC or CRC with FGFR alterations who have failed NCT05202236
standard therapy
II Recruiting HER2 negative advanced BC with FGFR 1–3 alterations who NCT05560334
have failed standard therapy
II Recruiting Advanced gastrointestinal cancer (excluding BTC) with FGFR NCT05559775
1–3 alterations who have failed standard therapy
II Recruiting Relapsed or refractory advanced NSCLC with FGFR mutation NCT05253807
II Active, not Advanced or unresectable CRC with FGFR mutation NCT04096417
recruiting
II Active, not Advanced, metastatic or unresectable cholangiocarcinoma NCT04256980
recruiting
FGFR-1/-2/-3/-4 III Recruiting Advanced, metastatic, or recurrent unresectable NCT04093362
cholangiocarcinoma harboring FGFR2 gene rearrangements
III Recruiting Advanced or metastatic STS NCT03784014
II Recruiting Advanced or metastatic HCC with FGF19 positive NCT04828486
Rogaratinib FGFR-1/-2/-3/-4 II/III Completed Locally advanced or metastatic urothelial carcinoma with NCT03410693
FGFR-positive
II Completed Cancer (unspecified) NCT04125693
II Recruiting Advanced GIST, STS NCT04595747
II Active, not Pretreated advanced SQCLC NCT03762122
recruiting
I Completed FGFR positive refractory, locally advanced or metastatic solid NCT03788603
tumors
I Recruiting Metastatic FGFR1/2/3 positive, hormone receptor positive NCT04483505
BC
Telatinib PDGFR-β, II Unknown Advanced HER2 negative advanced gastric or GEJ NCT03817411
c-Kit, VEGFR adenocarcinoma
II Recruiting Advanced GC, GEJ adenocarcinoma, or HCC NCT04798781
Tepotinib c-Met II Recruiting Solid tumors with Met amplification or Met exon 14 skipping NCT04647838
mutation
II Recruiting Advanced or metastatic NSCLC with Met amplifications NCT03940703
I/II Recruiting Advanced NSCLC with Met mutation NCT04739358
I/II Recruiting advanced GC, GEJ cancer with Met amplified or Met exon 14 NCT05439993
alternated
I Completed Patients with hepatic impairment NCT03546608
I Not yet Brain tumors with Met alternations NCT05120960
recruiting
ALL acute lymphoblastic leukemia, AML acute myeloid leukemia, BC breast cancer, BTC biliary tract cancer, CML chronic myeloid leukemia, CRC colorectal cancer,
DME diabetic macular edema, GEJ gastroesophageal junction, GBM glioblastoma, GC gastric cancer, GIST gastrointestinal stromal tumor, HCC hepatocellular
carcinoma, HER2 human epidermal growth factor receptor 2, HNSCC head and neck squamous cell carcinoma, NSCLC non-small cell lung cancer, PARP poly
ADP-ribose polymerase, Ph+ AML Philadelphia chromosome-positive acute myeloid leukemia, RCC renal cell carcinoma, SQCLC squamous-cell non-small cell
lung cancer, STS soft tissue sarcoma, TNBC triple-negative breast cancer, TED thyroid eye disease, wAMD wet age-related macular degeneration
research and application value, which has great potential for Bevacizumab-awwb (Mvasi®) is the first anti-tumor biosimilar of
emerging combination therapies to synergy chemotherapeutic bevacizumab approved by FDA.388,389 Ranibizumab (Lucentis®) is a
48 kDa humanized anti-VEGF monoclonal antibody fragment,
drugs and immune checkpoint inhibitors.
which can bind all VEGF-A isoforms, including VEGF110, VEGF121,
Ramucirumab (Cyramza®) is a fully humanized IgG1 antibody
with a weight of 147 kDa, which targets the extracellular binding and VEGF165 (Table 1). Ranibizumab is a prevalent anti-angiogenic
domain of VEGFR-2 to disturb the potent VEGF signal in tumor agent in treating oculopathy (Table 1).
angiogenesis (Table 1).382,383 As the first antibody targeted VEGFR-
2, ramucirumab significantly improved the median OS (5.2 months Anti-angiogenic oligonucleotide derivatives. Oligonucleotides are
vs. placebo 3.8 months) and PFS (2.1 months vs. placebo nucleic acid polymers that regulates gene expression and have
1.3 months) rates of patients (adults with advanced or unresect- specially designed sequences, including antisense oligonucleo-
tides (ASOs), siRNA (small interfering RNA), microRNA and
able gastric and gastroesophageal junction adenocarcinoma) in a
prospective, double-blind and placebo-controlled a phase III aptamers. Pegaptanib (Macugen®) is a 50 kDa VEGF-A targeted
REGARD clinical trial. Furthermore, it prolonged the median OS RNA aptamer, which has been approved for angiogenic age-
related macular degeneration in December 2004, leading to good
(9.6 months vs. 7.4 months) and PFS (4.4 months vs. 2.86 months)
tolerability and negligible local adverse effects of AMD patients
rates of homogeneous patients in a randomized, double-blind and
placebo-controlled a phase III RAINBOW trial through a combina- through intravitreous injections (Table 1).390
tion with paclitaxel.382 In 2014, ramucirumab was approved by
Anti-angiogenic recombinant fusion proteins. Fusion proteins are
FDA for previously treated advanced gastric or gastroesophageal
junction (GEJ) adenocarcinoma. Additionally, the first-line therapy complexes from binding the Fc segment of immunoglobulin to a
for metastatic CRC is a combination of ramucirumab and a biologically active functional protein molecule through genetic
modified FOLFOX-6 regimen (mFOLFOX-6), which demonstrated engineering technology. Aflibercept (Eylea®) is a recombinant
decoy receptor targeted VEGF, which is combined of the
gratifying safety and efficacy in a phase II clinical trial
(NCT00862784).384 extracellular VEGFR domain (VEGFR-1 Ig2 region and VEGFR-2
Olaratumab (Lartruvo®) is a 154 kDa fully recombinant human Ig3 region) and the Fc segment of human immunoglobulin G1
IgG1 monoclonal antibody with high affinity to PDGFRα, which is (IgG1) and has long half-life in anti-angiogenesis (Table 1).
Aflibercept inhibits the binding and activation of the VEGF family
the first-line drug approved by FDA for soft tissue sarcoma (STS)
(Table 1).385 STS is a relatively rare malignancy that occurs in and natural VEGFR by specifically blocking VEGF-A and most
connective tissue. In a randomized ANNOUNCE clinical trial among proangiogenic cytokines, thereby inhibiting division and prolifera-
tion of ECs, reducing vascular permeability, and is commonly used
509 patients, the addition of olaratumab did not significantly
improve the OS rate (doxorubicin plus olaratumab 20.4 months vs. in non-neoplastic angiogenic disease like AMD, DR, and
doxorubicin plus placebo 19.7 months) of advanced STS DME.380,391 Ziv-aflibercept is an adaptive variant of aflibercept,
patients.386,387 which has lower pH and higher osmolality (Table 1). It has been
472
2 VEGFR-2 2.6 nM
473
3 VEGFR-2 3.2 nM
474
4 VEGFR-2 3.2 nM
475
5 VEGFR-2 27 nM
476
6 VEGFR-2 66 nM
481
12 VEGFR-2 24.7 nM
PDGFR-β 16.1 nM
482
13 VEGFR-2 7 nM
FGFR-1 69 nM
PDGFR-β 31 nM
FGFR-1 0.23 μM
PDGFR-β 0.1 μM
481,483
15 VEGFR-2 435 nM
c-Met 654 nM
PDGFR-β 371 nM
484,485
16 VEGFR-2 1.05 nM
Tie-2 2.47 nM
EphB4 0.27 nM
486
17 VEGFR-2 1.85 nM
Tie-2 0.73 nM
EphB4 2.99 nM
19 VEGFR-2 5 μM 488
HDAC4 0.36 μM
489
20 FGFR-4 5.4 nM
490
21 FGFR-1 1.0 nM
FGFR-2 4.5 nM
VEGFR-2 2.9 nM
FGFR-2 1.3 μM
FGFR-3 4.1 μM
inhibitory activity targeted HIF-α. TME is a highly complex of other angiogenic modalities, genetic or phenotypic mutations,
ecosystem of cellular and noncellular components, which is stromal autophagy and induction of EMT.
broadly related to tumor invasion and recurrence.466,467 In recent
years, research on tumor microenvironment and targeted therapy Drug resistance
has indicated that limiting tumor deterioration and angiogenesis Drug resistance is a dominant difficulty that consistently limits the
by inhibiting HIF expression to downregulate the level of some clinical outcomes in targeted anti-angiogenic therapy, which can be
angiogenic factors may be a valuable strategy.468–470 Table 3 divided into congenital resistance and acquired resistance (Fig. 8).503
summarizes the structures of reported molecules and related Congenital drug resistance is defined as the inherent insensitivity to
references, which could be beneficial to the research on drugs of patients, which may be related to the genes of patients and
angiogenic inhibitors. tumors. Acquired drug resistance has been comprehensively
analyzed by researchers through cytological and molecular studies.
These unique mechanisms include: (a) upregulation of compensa-
LIMITATIONS AND CHALLENGES OF ANTI-ANGIOGENIC tory pro-angiogenic signaling pathways in tumor tissue (HGF, bFGF,
THERAPY VEGF-C, PlGF, angiopoietins, and Dll-4 have been widely testified
Angiogenic inhibitors used in cancer therapy by affecting the that upregulated in various tumors with drug resistance);133,504,505
formation of new blood vessels in tumors, which have expended a (b) recruiting bone marrow-derived endothelial progenitor cells,506
new field for the treatment of a wide range of solid tumors. pericyte progenitor cells,507 tumor-associated macrophages,508 and
However, there are still some shortcomings in anti-angiogenic immature monocytic cells, which can maintain the formation of
therapy due to the complex mechanisms of tumor angiogenesis blood vessels; (c) recruitment of perivascular cells (like pericytes),
and limited research, including tumor relapse,495 drug resis- which can cover immature tumor blood vessels to prevent
tance,496,497 lack of bio-markers,496 short-acting efficacy,27,28 and destruction by anti-angiogenic drugs;509 (d) unconventional angio-
several serious adverse events.498,499 genic processes like vessel co-option,510–513 vessel mimicry and
intussusceptive angiogenesis.77,514,515 Additionally, drug resistance
Limited therapeutic efficacy also involves high heterogeneity of tumor tissue and TME,
It was initially assumed that anti-angiogenic therapy might not be endothelial heterogeneity,516 autophagy of tumor cells, differentia-
toxic compared with other chemotherapeutic agents owing to tion of cancer stem cells,517 infiltration of stromal cells,518 tumor
genetic stability and quiescence of ECs under normal physiological types, gene mutations of tumors or targets, development stage of
conditions and the selectivity of targeted drugs. However, this was the tumor, medication history of patients, and other factors, all of
proved to be a miscalculation. Common serious adverse events which can affect the response and tolerance of patients to anti-
such as hypertension, proteinuria, lymphopenia, thrombocytope- tumor therapy.
nia, leukopenia, neutropenia, and some physical abnormalities
caused by different drugs have appeared in a number of different Lack of valid biomarkers
clinical treatments (Table 1), which may affect the tolerance of The application of biomarkers is a powerful adjuvant means which
patients and even lead to treatment termination.45,54,500 This are essential for disease identification, early diagnosis and
requires further optimization of the structures of angiogenic prevention, and drug treatment monitoring. Biomarkers refer to
inhibitors to better target selectivity, or other techniques to biochemical indicators of normal physiological or pathogenic
increase drug delivery to tumor tissue while bypassing normal processes to furnish the structural or functional changes of
tissue, such as nano-preparations. systems, organs, tissues, cells and subcells, and can also be used
In addition, angiogenic inhibitors have a result on controlling for disease diagnosis, disease stage, or evaluating the safety and
growth and spread of tumor in the short term by blocking the blood efficacy of a drug or regimen among targeted population.519 In
supply (which is manifested in clinical treatment as increased PFS), clinical practice, there are at least six categories of biomarkers
but the long-term result is an increased risk of tumor local invasion including diagnostic, predictive, prognostic, pharmacodynamic,
and distant metastasis induced by hypoxia, as well as the probability safety and monitoring biomarkers owing to different bio-functions.
of revascularization and tumor resurgence after discontinuation of For example, HER2 is a diagnostic indicator for breast cancer typing,
sustained treatment (which manifests as an insignificant or even and levels of PD-L1 is used to predict the efficacy of immune
unchanged increase in OS).501,502 Some molecular events under- checkpoint inhibitors (ICIs). Despite considerable efforts, there are
stood by researchers within the past few decades could be the few biomarkers responding to angiogenesis approved for clinical
activation of alternative pro-angiogenic molecules, the development application.172,519,520 Given the variable results from anti-angiogenic
Fig. 8 Mechanisms of drug resistance in anti-angiogenic therapy. Some patients are intrinsically non-responsive to anti-angiogenic therapy
while other patients who are initially responsive acquire adaptive resistance. The mechanisms that manifest acquired resistance to anti-
angiogenic therapy include: compensatory upregulation of alternative pro-angiogenic factors such as bFGF, PDGF, and PlGF within the tumor;
recruitment of bone marrow-derived endothelial progenitor cells to facilitate neovascularization; increased pericyte coverage protects tumor
blood vessels; autophagy helps tumor cells thrive in a hypoxic environment; increased invasiveness of the tumor promotes the distant
metastasis and invasion of tumor cells through blood and lymphatic circulation. In addition, genetic mutations, vessel mimicry, vessel co-
option, and intussusception angiogenesis also contribute to drug resistance
treatments in the clinic, there is a need to characterize changes in characteristically synergistic or an additive manner targeting
the blood vessels and tumor microenvironment to detect and important signaling pathway. Diversified methods in anti-cancer
prevent tumor escape, and to monitor the patients’ response to therapy provide more options for clinical treatment and make
drugs and the advances in treatment.521,522 It is an inevitable trend strong alliances possible.
to explore effective cancer-specific biomarkers responding to In recent several years, one of the prevalent research direction is
angiogenic system to enhance the efficacy of anti-angiogenic the combination of angiogenic inhibitors and immune checkpoint
regime and anti-cancer therapy. With the advancement in bio- inhibitors, in which better clinical benefits from HCC and RCC
analytical technology and clinical bio-chemistry, tissue and cell patients treated with programmed cell death 1 (PD-1) and VEGFR-
concentrations of some angiogenic mediators, circulating ECs, 2 inhibitors than with monotherapy.467,525,526 Tumors can induce
circulating progenitor cells, CT imaging of blood flow and blood immune tolerance and limit proliferation and activation of T cells
volume have been shown to have potential as biomarkers, but more during growth and metastasis by using immune checkpoints (ICs)
clinical trials are needed to validate their prospective. Developing produced on T cells to accomplish immune escape. Blockade with
efficient biomarkers for diagnosing the progression and stage of different immune checkpoint inhibitors may activate the body’s
cancer and identifying mechanisms of tumor angiogenesis and drug immune system and weaken immunosuppression in TME against
resistance, in order to benefit drug selection, balance efficacy and tumor cells by promoting the activation and proliferation of
toxicity, and simplify anti-cancer therapy. Actually, due to numerous T cells, including PD-1, programmed cell death-ligand 1 (PD-L1)
factors such as the complexity of tumor angiogenesis, heterogeneity and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibi-
and variability of tumors, the unpredictability of response or toxicity, tors.525 As mentioned before, the tumor microenvironment is
and limitations of preclinical and clinical trials, the development of composed of tumor cells, cancer stem cells, immune cells,
biomarkers will be a great challenge. fibroblasts and other cells and their secretions, as well as non-
cellular components such as extracellular matrix. High levels of
VEGF in TME are not only crucial factors in inducing abnormaliza-
EMERGING APPROCHES TO FURTHER IMPROVE ANTI- tion and increasing permeability of tumor vessels, but also weaken
ANGIOGENIC THERAPY the anti-tumor effect of immune cells through multiple pathways
Combination therapy including: a) immune-activating cells and immune effector cells
Since the first angiogenic inhibitor bevacizumab approved for can be effectively blocked by VEGF, such as inhibiting maturation
treatment, combination therapy based on anti-angiogenic agents of dendritic cells (DCs), and inducing the failure and apoptosis of
has infiltrated anti-tumor field.523 Combination therapy is a cytotoxic T cells; b) the aggregation and activity of immunosup-
modality aiming to enhance anti-tumor efficacy through combin- pressive cells including regulatory T cells (Tregs), myeloid-derived
ing two or more therapeutic agents, including anti-angiogenic suppressor cells (MDSCs), M2-like tumor-associated macrophages
therapy combined with surgery, immunotherapy, chemotherapy, (M2 TAM) can be up-regulated by intratumoral VEGF; c) by
radiotherapy, genetherapy or (and) other targeted anti-tumor elevating the production of endothelial adhesion molecules and
agents.524 Compared with monotherapy, the combination of anti- up-regulating immune checkpoints, VEGF can generate a selective
tumor drugs improves the therapeutic efficacy in a endothelial barrier for cytotoxic T cells to prevent infiltration while
Atezolizumab with or without cobimetinib vs. Metastatic CRC II Positive results in median OS: atezolizumab 7.10 months, atezolizumab NCT02788279
regorafenib plus cobimetinib 8.87 months, regorafenib 8.51 monthsa.
Unsuccessful or terminated clinical trials
Irinotecan and temozolomide plus bevacizumab Relapsed or refractory neuroblastoma II Expected and transient toxicities, but the addition of bevacizumab did NCT01114555
not improve response rates compared to irinotecan plus
temozolomidea.
FOLFOX6 plus bevacizumab Biliary system carcinoma II 1/8 patient with perforation of colon; impossible to get insurance NCT00881504
companies to cover bevacizumabc.
Ixabepilone plus bevacizumab Metastatic RCC II Well tolerated, with modest activity in second - or later-line mRCC, not NCT00923130
recommendeda.
Docetaxel plus sorafenib Advanced non-squamous cell NSCLC II Preliminary efficacy data was not encouraging, 4/5 patients with serious NCT00801801
adverse eventsc.
Temozolomide plus sorafenib Recurrent GBM II Well tolerated, but limited activity for recurrent GBMa. NCT00597493
Paclitaxel and carboplatin plus axitinib vs. plus Advanced lung cancer II Axitinib plus paclitaxel and carboplatin is worse than bevacizumab plus NCT00600821
bevacizumab paclitaxel and carboplatin: PFS 11.0 vs. 15.9 months; OS 18.1 vs.
21.6 monthsa.
Modified FOLFOX6 plus axitinib and/or bevacizumab Metastatic CRC II No improvements in combination with axitinib or axitinib/bevacizumab NCT00460603
compared to bevacizumab plus FOLFOX6: PFS 11.0 vs. 12.5 vs.15.9
months; OS 18.1 vs. 19.7 vs. 21.6 monthsa.
Chemotherapy (capecitabine or docetaxel) vs. TNBC II No improvements of sunitinib in median OS (9.4 vs. 10.5 months), NCT00246571
sunitinib objective response rates (3% vs. 7%)a.
AML acute myeloid leukemia, BC breast cancer, CRC colorectal cancer, FOLFOX6 oxaliplatin, calcium folinate and 5-fluorouracil, GBM glioblastoma, MDS myelodysplastic syndromes, NSCLC non-small cell lung
dant VEGF derived from tumor cells lead to disordered and leaky
vascular networks, which seriously affects the blood transport of
cytotoxic drugs and immunosuppressants.525,527,528 If immu-
notherapy is accompanied by anti-angiogenic drugs targeting
VEGF pathway, it reverses these immunosuppression caused by
VEGF and enhances the immune function of patients. At the same
time, it can neutralize excess VEGF, reconstruct the vascular
system of tumor tissue, normalize vascular network, promote the
No significant benefits from the addition of durvalumab and