- Review
- Open access
- Published:
Cancer combination therapies by angiogenesis inhibitors; a comprehensive review
Cell Communication and Signaling volume 20, Article number: 49 (2022)
Abstract
Abnormal vasculature is one of the most conspicuous traits of tumor tissue, largely contributing to tumor immune evasion. The deregulation mainly arises from the potentiated pro-angiogenic factors secretion and can also target immune cells' biological events, such as migration and activation. Owing to this fact, angiogenesis blockade therapy was established to fight cancer by eliminating the nutrient and oxygen supply to the malignant cells by impairing the vascular network. Given the dominant role of vascular-endothelium growth factor (VEGF) in the angiogenesis process, the well-known anti-angiogenic agents mainly depend on the targeting of its actions. However, cancer cells mainly show resistance to anti-angiogenic agents by several mechanisms, and also potentiated local invasiveness and also distant metastasis have been observed following their administration. Herein, we will focus on clinical developments of angiogenesis blockade therapy, more particular, in combination with other conventional treatments, such as immunotherapy, chemoradiotherapy, targeted therapy, and also cancer vaccines.
Video abstract
Introduction
Angiogenesis is a critical process that is needed for many physiological and pathological activities [1]. Angiogenesis is a heavily controlled process under physiological circumstances. It usually happens throughout embryonic development, wound repair, and the menstrual cycle [2]. Under physiological circumstances, angiogenesis relies on the equilibrium of positive and negative angiogenic modulators within the vascular microenvironment and necessitates the contribution of diverse molecules, such as pro-angiogenic factors, extracellular matrix (ECM) proteins, adhesion receptors, and also proteolytic enzymes [3]. Pathological diseases including psoriasis, diabetic retinopathy, as well as cancer exhibit unregulated angiogenesis. Angiogenesis is necessary during tumor development for appropriate feeding and elimination of metabolic waste products from tumor regions [4]. In reality, tumor development and metastasis are dependent on angiogenesis as well as lymphangiogenesis, which are initiated by chemical impulses from cancer cells in a fast-growing phase [5, 6]. Muthukkaruppan and colleagues previously investigated the dynamics of cancer cells injected into various areas of the same organs [7]. One part was the iris, which had blood circulation, and the other was the anterior chamber, which did not [7]. Cancer cells lacking blood circulation expanded 1–2 mm3 in diameter and afterward halted, but when put in a location where angiogenesis was feasible, they expanded to more than 2 mm3. Given that tumors become necrotic or even apoptotic in the absence of a circulatory supply [8], it has strongly been validated that angiogenesis is a critical component in cancer development.
Tumors differ significantly in the patterns and characteristics of the angiogenic vascular system, as well as their sensitivity to anti-angiogenic treatment [9]. Cancer cells control the angiogenic programming of neoplastic tissues through collaboration with a range of tumor-associated stromal cells as well as their bioactive products, which include cytokines and growth hormones, the extracellular matrix, as well as secreted microvesicles [10]. Apart from cancer immunotherapy or other pioneering approaches such as chemotherapy and radiotherapy, which have resulted in a significant advance in cancer treatment [11, 12], another potential treatment approach is anti-angiogenesis, which aims to impair the vasculature and deprive the tumor of oxygen and nutrition [13]. This is accomplished mostly by targeting the pro-angiogenic factors-induced signaling pathway, which is prominent in the tumor microenvironment under hypoxic conditions [14]. Pro-angiogenic factors are classified into two main subgroups: (1) classical, including vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), platelet-derived growth factor (PDGF), platelet-derived endothelial cell growth factor/thymidine phosphorylase (PD-ECGF/TP), angiopoietins (Ang), hepatocyte growth factor (HGF), insulin-like growth factors (IGFs), tumor necrosis factor (TNF), interleukin-6 (IL-6); (2) non-classical, including stem cell factor (SCF), tryptase and also chymase [15]. VEGF family members are the regulator of angiogenesis both under normal circumstances and in a disease condition. This family consists of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and placenta growth factor (PlGF), which binds with divergent affinities and specificities to tyrosine kinase receptors (VEGFR) 1,-2, and -3 [16, 17]. The interfaces between VEGF-A and VEGFR 2 exceed angiogenesis, while VEGF-C and D preferentially make connections with VEGFR-3 [18]. The improved expression of VEGF inspires tumourigenesis by potentiating the epithelial-mesenchymal transition (EMT) activation. In addition to VEGF receptor tyrosine kinases, the neuropilins (NRPs), potent co-receptors for class 3 semaphorins, are crucial for exerting the impacts of VEGF on cancer cells as a result of their capability to affect the activities of growth factor receptors and integrins [19]. VEGF/NRP axis adjusts the expression and action of important biological molecules, such as Rho family guanosine triphosphatases (GTPases) and transcription factors in malignant cells [20]. Respecting the pivotal role of the VEGF/VEGFR signaling axis in cancer angiogenesis, several anti-angiogenic medicines have been authorized for various types of cancer, such as anti-VEGF antibodies, anti-VEGFR antibodies, and VEGFR tyrosine kinase inhibitors (TKIs) (Fig. 1) [14, 21]. Meanwhile, multitargeted small-molecule TKI can target multiple receptor sites simultaneously. The main targets included vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), fibroblast growth factor receptor (FGFR), c-Kit, and c-Met. Anti-angiogenic TKIs block the kinase activity of receptors and transduction of downstream signals involved in the proliferation, migration, and survival [22]. However, monotherapy with an anti-angiogenic drug has shown minimal therapeutic advantages for most cancer patients [23]. Thereby, it has been suggested and also evidenced that combining anti-angiogenic medicines with other strategies, comprising immune checkpoint inhibitors (ICIs), chemotherapy, human epidermal growth factor receptor 2 (HER2)-targeted therapies, adoptive cell transfer (ACT), cancer vaccines, and also radiotherapy may have a synergistic anti-tumor impact [24]. This review highlights current knowledge and clinical developments of anti-angiogenesis combination treatment, either alone or in conjunction with other modalities, focusing on last decade in vivo reports.
Tumor angiogenesis mechanism
Several successive stages throughout tumor angiogenesis may be emphasized. The vessel wall of mature capillaries comprises an endothelial cell lining, a basement membrane, and a layer of cells termed pericytes that partly surround the endothelium [25]. Pericytes share the same basement membrane as endothelial cells and sometimes come into touch with them. Tumor-derived angiogenic agents attach to endothelial cell receptors, initiating the angiogenesis process. VEGF, fibroblast growth factors (FGF), tumour necrosis factor α (TNFα), transforming growth factor (TGF-β), and angiopoietin (Ang) are the most well-known angiogenic cytokines and growth factors [26, 27]. When endothelial cells are encouraged to develop, proteases, heparanase, as well as other digestive enzymes are secreted, which break down the underlying membrane that surrounds the artery [28, 29].
Matrix metalloproteinases (MMPs), a class of metalloendopeptidase produced by tumor cells and supportive cells, allow for the degradation of the basement membrane as well as the extracellular matrix surrounding pre-existing capillaries, typically postcapillary venules [30, 31]. The breakdown of the extracellular matrix also enables the discharge of pro-angiogenic factors out from the matrix. Endothelial cell connections change, cell extensions cross through the gap produced, and the recently created sprout develops towards the source of the stimulation [32]. Endothelial cells enter the matrix and start migrating and proliferating inside the tumor mass. Freshly created endothelial cells arrange into hollow tubes and produce a new basement membrane for vascular stability at this site [33]. The blood flow inside the tumor is formed by freshly shaped fused blood vessels. Significant interactions between cell-associated surface proteins and the extracellular matrix promote the development of the lumen during canalization. Hybrid oligosaccharides galectin-2, platelet endothelial cell adhesion molecule-1 (PECAM-1 or CD31), and VE-cadherin are among the surface proteins discovered in this interaction [34, 35]. Different circumstances, including metabolic and mechanical stressors, hypoxia, and genetic alterations or changed oncogene expression or tumor suppressor genes, may cause an imbalanced shift towards pro-angiogenic factors, while the mechanism behind this is yet unknown.
Microenvironment role in tumor angiogenesis
Numerous pro-angiogenic agents, such as VEGF, platelet-derived growth factor (PDGF), and FGF are found in the tumor microenvironment. These compounds are produced by cancer cells or tumor-infiltrating lymphocytes or macrophages and can trigger pro-angiogenic signaling pathways, promoting tumor angiogenesis, development, invasion, and metastasis [36]. Furthermore, inflammatory cytokines in the tumor microenvironment have a significant role in tumor angiogenesis. Prior studies have shown that interferon’s (IFNs), TGF-β, and TNF may all have anticancer effects [37]. However, a few investigations have shown that these factors may promote angiogenesis and tumor development. These findings suggest that cytokines have a variety of roles in tumorigenesis as well as development. Numerous interleukin 1 (IL-1) family members stimulate tumor angiogenesis [38]. Through the activity of nuclear factor-kappa B (NF-κB), p38 mitogen-activated protein kinase (MAPK) signaling, and Janus kinase (JAK), IL-1 signaling stimulates angiogenesis by upregulating VEGF as well as angiogenesis-related molecules [39, 40].
IL-6, IL-8, and IL-22 may also increase tumor angiogenesis by modulating angiogenic factor expression [41]. A hypoxic microenvironment may also encourage tumor development, invasion, metastasis, immune evasion, and angiogenesis. As a result, co-targeting hypoxic, as well as anti-angiogenic factors, may enhance tumor outcomes. Researchers discovered that co-treatment with hypoxia-inducible factor 1(HIF-1) inhibitors and bevacizumab had a greater anticancer impact than therapy with bevacizumab separately in glioma xenografts [42]. HIF-1 is an upstream regulator of many angiogenic factors that may directly stimulate angiogenic factor transcription to enhance tumor angiogenesis [43]. Furthermore, various hypoxia-induced lncRNAs may enhance tumor angiogenesis by influencing angiogenic factor expression [44]. As angiogenic factors abound in the tumor microenvironment, treating cancer cells with medicines that target several angiogenic agents may result in improved outcomes. Moreover, type 1 T helper (Th1) CD4+ and also CD8 + cells polarize innate immune cells versus tumor regression, for instance by M1 macrophages polarization of tumor-associated macrophages (TAMs) [45]. In contrast, tumor-secreted cytokines largely stimulate a proangiogenic and protumorigenic phenotype of the tumor-associated inflammatory infiltrate. Inducing the type 2 T helper (Th2) CD4 + cells along with regulatory T cells (Tregs) can, in turn, elicit protumoral reactions, comprising M2 polarization of TAMs, culminating proangiogenic microenvironment (Fig. 2) [45]. Recently, Wang et al. showed intra- and inter-tumoral heterogeneities between TAM subpopulations and their functions, with CD86 + TAMs playing a crucial role in tumor progression [46].
FDA approved anti-angiogenic agents
Upon successful preclinical studies (Table 1), a myriad of clinical trials have been accomplished or are ongoing to determine the safety, feasibility, and efficacy of anti-angiogenic agents therapy in cancer patients alone or in combination with other therapeutic means (Table 2). The present era of anti-angiogenic treatment for cancer research started in 1971 with the publishing of Folkman's creative hypothesis [47], although it would take 33 years for the FDA to authorize the first drug produced as a blocker of angiogenesis. Bevacizumab, a humanized monoclonal antibody targeted against VEGF, was coupled with standard chemotherapy in a randomly selected phase 3 study of first-line therapy of metastatic colorectal cancer (CRC) [48]. When utilized in conjunction with conventional chemotherapy, bevacizumab therapy improved overall survival (OS) in the first-line treatments of advanced non–small-cell lung cancer (NSCLC) [49]. The FDA of the United States has authorized a variety of angiogenesis inhibitors for the treatment of cancer. Most of them are targeted treatments created to target VEGF, its receptor, or other angiogenesis-related molecules. Bevacizumab, axitinib, everolimus, cabozantinib, lenalidomide, lenvatinib, pazopanib, ramucirumab, regorafenib, sorafenib, sunitinib, thalidomide, Ziv-aflibercept and vandetanib are most famous accepted angiogenesis inhibitors, which have been approved for human advanced tumors [50].
As the first VEGF-targeted agent approved by FDA, bevacizumab, is used since February 2004, for the treatment of patients suffering from metastatic (m) CRC in combination with the standard chemotherapy treatment (as first-line treatment) [51]. In June 2006, it was approved with fluorouracil (5-FU)-based therapy for second-line mCRC. Also, it has been indicated for NSCLC (plus chemotherapy), breast cancer, glioblastoma, ovarian cancer (plus chemotherapy), and also cervical cancer [51]. Another well-known angiogenesis inhibitor, axitinib, has gained approval from FDA for use as a treatment for renal cell carcinoma (RCC) since January 2012 and also has shown promising outcomes in pancreatic cancer (plus gemcitabine) [52, 53]. In March 2009, everolimus was firstly approved for RCC therapy, and after that was indicated for breast cancer patient’s therapy [54]. Moreover, since 2016, it is used for neuroendocrine tumors (NET) of gastrointestinal (GI) or lung origin with unresectable, locally advanced, or metastatic disease [55]. In November 2012, cabozantinib, a small molecule inhibitor of the tyrosine kinases c-Met and VEGFR2, was approved for thyroid cancer [56] and also in April 2016 was accepted as second-line treatment for RCC [57]. Lenalidomide, a 4-amino-glutamyl analogue of thalidomide, is used to treat multiple myeloma (MM) [58] and myelodysplastic syndromes (MDS) [59], and also lenvatinib, which acts as a multiple kinase inhibitor against the VEGFR1, VEGFR2, and VEGFR3 kinases, is applied for the treatment of thyroid cancer [60]. In 2016, lenvatinib was also approved in combination with everolimus for the treatment of advanced RCC [61]. Since 2009, pazopanib, a potent and selective multi-targeted receptor tyrosine kinase inhibitor, is utilized for metastatic RCC and advanced soft tissue sarcomas therapy [62]. Besides, since April 2014, the ramucirumab, a direct VEGFR2 antagonist, is indicated as a single-agent treatment for advanced gastric cancer or gastro-esophageal junction (GEJ) adenocarcinoma after treatment with fluoropyrimidine- or platinum-containing chemotherapy [63]. Further, ramucirumab in combination with docetaxel has gained approval for treatment of metastatic NSCLC [64]. Ramucirumab also is used for mCRC (since 2015) [65] and HCC (since 2019) [66] therapy. Also, regorafenib, an orally-administered inhibitor of multiple kinases, has been indicated for the treatment of patients with advanced HCC who were previously treated with sorafenib [67]. Moreover, sorafenib as another type of kinase inhibitor is used since 2007 for RCC and HCC therapy, and since 2013 for thyroid cancer [68]. Multi-targeted receptor tyrosine kinase inhibitor sunitinib also is applied for gastrointestinal stromal tumor (GIST) and RCC therapy [69]. In addition, since 2006, thalidomide as a type of biological therapy in combination with dexamethasone has been approved for the treatment of newly diagnosed MM patients [70]. Also, Ziv-aflibercept in combination with 5-fluorouracil, leucovorin, irinotecan (FOLFIRI) are used to treat patients with metastatic CRC [71]. Finally, tyrosine kinase inhibitor vandetanib is employed to treat medullary thyroid cancer in adults who are ineligible for surgery [72, 73].
Resistance to anti-angiogenic therapies
Despite their total tumor growth reduction, therapeutic anti-angiogenic agents were linked to enhanced local invasiveness as well as distant metastasis. These events seem to be significant factors to resistance to anti-angiogenesis treatments. They were originally reported in various preclinical models by Paez-Ribes and coworkers [74]. Based on the literature, anti-angiogenic treatment may increase tumor invasiveness. RCC cells, for example, showed increased proliferation and an invasive character after being treated with bevacizumab [75]. Likewise, glioblastoma cells in mice models were more invasive after VEGF suppression [74]. Sunitinib treatment also has been found to cause vascular alterations such as decreased adherens junction protein expression, reduced basement membrane, pericyte coverage, and increased leakiness [76, 77]. These phenotypic alterations were found in both normal and tumor organ arteries, indicating that they promote tumor cell local intravasation and extravasation, culminating in metastatic colonization [78].
Angiogenesis blockade therapy may lead to vascular regression and resultant intra-tumoral hypoxia. Various investigations have been fulfilled to assess an enhancement in hypoxic areas in primary tumors upon angiogenesis blockade therapy [76, 79]. Further investigation also exposed an attendant augmentation in HIF-1a expression during treatment. HIF-1a and hypoxia are recognized drivers of epithelial-mesenchymal transition (EMT), a process that induced tumor metastasis. Significant improvement in the expression and activities of EMT-related genes (e.g., Twist and Snail) has been observed upon anti-angiogenic treatment and thereby may dampen treatment efficacy [80]. Moreover, loss of the epithelial marker, E-cadherin, and the stimulation of the mesenchymal marker, vimentin, has been evidenced following anti-angiogenic treatment [80]. Hypoxic milieu also largely promotes VEGF expression by the upstream transcription factor HIF-1a [81]. HIF-1, in turn, inspires tumors to achieve more angiogenic and invasive competencies, culminating in metastasis [82]. In fact, hypoxia and EMT bring about increased invasiveness and metastasis of tumors mainly caused by up-regulation of c-Met, Twist, and HIF-1a [83, 84]. Conversely, semaphorin 3A (Sema3A), a well-known endogenous anti-angiogenic molecule, is substantially down-regulated in tumors, ensuring provoked invasiveness and metastasis [85].
Ang-Tie signaling system is a vascular-specific receptor tyrosine kinases (RTK) pathway complicated in modifying the vascular permeability and blood vessel formation and remodeling by potent angiogenic growth factors, Ang-1 and Ang-2 [86]. Molecular analysis has confirmed that activation of the Ang-Tie pathway as a result of the connection between Ang-1 and Tie2 receptor on the M2 subpopulation of monocytes, hematopoietic stem cells (HSCs), and endothelial cells (ECs) of blood and lymphatic vessels elicits maturation or stabilization of blood vessels [80]. Besides, Ang-2 suppresses this pathway, eventually sustaining remodeling or generation of vascular sprouts upon exposure to VEGF [87]. Ang-2 up-regulation has been noticed in multiple types of tumors and is likely involved in resistance versus anti-VEGF therapy [88, 89]. For instance, there is clear evidence signifying that enhanced serum Ang-2 levels are in association with an undesired response to bevacizumab therapy in CRC patients [90]. Studies in lung adenocarcinoma patients revealed that elevated levels of VEGFA and Ang-2 is valued prognostic biomarkers and double targeting of VEGFA and Ang-2 can improve therapeutic outcome [91]. As well, up-regulation and compensatory mechanisms of other growth factors, in particular basic fibroblast growth factor (bFGF), are thought to contribute to the stimulation of the resistance to VEGF targeted therapies. Improved level of the bFGF has strongly been evidenced in the chronic inflammation area, after tissue injury, as well as human cancers bevacizumab [92]. The classical FGF axis can be transduced by RAS/MAPK, PI3K/Akt, Src tyrosine kinase, and STAT pathways, consisting of potent targets for current anti-cancer strategies [93]. Upon bevacizumab treatment in glioblastoma tumor models, Okamoto et al. showed the increased levels of the bFGF and PDGF expression in the endothelial cells, pericytes, and also tumor cells, in turn, caused robust resistance to bevacizumab [94]. Other results indicate that co-targeting of the VEGF and FGF pathways can potentiate tumor cells' sensitivity to bevacizumab, thereby suggesting that the upregulation of the FGF/FGFR autocrine axis plays an indispensable role in eliciting resistance to anti-VEGF/VEGFR therapies [92]. Also, cancer patients with up-regulated bFGF in serum usually show no desired response to sunitinib, indicating the necessity of co-targeting VEGF and bFGF pathways concurrently [95, 96].
Increased metastasis and invasiveness in response to anti-angiogenesis therapy vary according to treatment type, dosage, and schedule. Sunitinib and anti-VEGF antibody monotherapy showed varied effects on mice tumor models, according to Singh et al. reports [77]. While sunitinib therapy increased tumor cell aggressiveness, anti-VEGF antibody treatment did not [77]. Chung et al. also corroborated these findings by comparing the effectiveness of several RTK inhibitors and antibody treatments in mouse models [97]. Though imatinib, sorafenib, or sunitinib increased lung metastasis after 66c14 cell injection, employing an anti-VEGFR2 antibody reduced the development of lung nodules [97]. Overall, reports show that the increased metastasis and invasiveness caused by angiogenesis blockade therapy depend highly on the treatment type.
Anti-angiogenic drug dosage and delivery schedules may also potentially cause resistance. Indeed, short-term and high-dose sunitinib (120 mg/kg per day) therapy before and after intravenous breast tumor cell injection into severe combination immune-deficient animals exhibited the greatest detrimental effects [98]. Sunitinib at high doses accelerated tumor development and facilitated metastasis to the lung and liver, resulting in decreased survival [74, 98]. Although sorafenib had comparable outcomes, sunitinib produced conflicting findings in various trials. High-dose sunitinib therapy before systemic injection of tumor cells enhanced the metastatic potential of lung cancer cells, but not RCC cells. In contrast, low-dose sunitinib (30 and 60 mg/kg per day) had no supportive effect on metastases [78].
Combination therapy with anti-angiogenic agents
Anti-angiogenic agents plus ICIs
Recently, scientists have concentrated on the role of immune checkpoint molecules, such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1), largely participating in tumor cell escape from immune surveillance as their capacity to obstruct T cell activation [99, 100]. Hence, immune checkpoint inhibitors (ICIs) have been evolved for suppressing these immune checkpoint molecules [101]. FDA-approved ICIs comprise the nivolumab, cemiplimab, and pembrolizumab, atezolizumab, avelumab, durvalumab, and also ipilimumab [102]. Atezolizumab has been approved for use in combination with bevacizumab, paclitaxel, and carboplatin as the first-line treatment of patients with NSCLC [103]. Based on literature, only a subset of PD-L1 positive patients benefits from PD-1/PD-L1 targeted therapies [104]. PD-L1 expression is regulated by various factors, such as inflammatory and oncogenic signaling, leading to the varied significances of PD-L1 positivity. Such alterations in PD-L1 expression lead to the divergent response to PD-1/PD-L1 targeted therapies and may elicit resistance to the PD1/PD-L1 blockade therapies [105].
Recent reports exhibited that combination therapy with anti-angiogenic agents and ICIs could elicit synergistic anti-tumor effects in preclinical models as well as humans (Table 3). Meanwhile, co-administration of anti-PD-1 and anti-VEGFR2 monoclonal antibodies (mAbs) in the Colon-26 adenocarcinoma mice model gave rise to the potent inhibition of tumor growth synergistically without overt toxicity [106]. VEGFR2 blockade therapy negatively regulated tumor neovascularization, as evidenced by the attenuated frequencies of microvessels, whereas PD-1 inhibition exerted no effect on tumor angiogenesis. PD-1 mAbs improved T cell infiltration into tumors and promoted local immune response, as documented via the improvement in various proinflammatory cytokine expressions. Such events signified that concurrent suppression of PD-1 and VEGFR2 might inspire synergistic in vivo anti-tumor influences by dissimilar mechanisms [106]. Further, in a mouse model of small-cell lung cancer (SCLC), co-administration of anti-VEGF and anti-PD-L1 mAbs resulted in a more prominent therapeutic outcome than mono therapy with each agent [107]. Mice that received anti-PD-L1 mAbs alone relapsed after 3 weeks accompanied with a tumor-associated PD-1/ T-cell immunoglobulin domain and mucin domain 3 (TIM-3) double-positive depleted T-cell phenotype. Notably, the depleted T-cell phenotype following anti-PD-L1 therapy was revoked through the addition of anti-VEGF blockade therapy. Analysis revealed that VEGFA expression improves the expression of the inhibitory receptor TIM-3 on T cells, representative of an immunosuppressive action of VEGF in patients with SCLC during PD-1 blockade therapy. Thereby, it seems that VEGFA inhibition may entice T cell activation at higher levels, facilitating T cell-mediated anti-tumor immunity [107]. Similarly, combination therapy with sunitinib and PD-L1 blocked therapy prolonged overall survival (OS) of treated RCC mice models in comparison to mono therapy with either drug [108]. Besides, in the triple-negative breast cancer (TNBC) mice model, PD-L1 blocking was highly effective as an adjuvant monotherapy. However, its co-administration with paclitaxel chemotherapy (with or without VEGF blocked therapy) showed superiority over neoadjuvant therapy [108]. Studies also in VEGFA-overexpressed human tumors and mouse tumor models revealed that apatinib plus PD-1/PD-L1 blockade therapy could alleviate hyperangiogenesis and hypoxia in TME and also alter the immunosuppressive TME into an immunostimulatory microenvironment [109]. Consequently, it was suggested that anti-angiogenesis treatments could potentiate the efficiency of PD-1/PD-L1 blockade therapy in VEGFA-overexpressed tumors [109]. In 2017, Schmittnaegel et al. also noticed that dual Ang-2 and VEGFA inhibition induced antitumor immunity that was promoted by PD-1 blockade therapy in breast cancer, pancreatic neuroendocrine tumor, and melanoma [110]. They showed that Ang-2 and VEGFA blockade by a bispecific antibody (A2V) caused vascular regression, tumor necrosis along with improved antigen presentation by intratumoral phagocytes [110]. The combination therapy also enhanced the presence and activation of interferon-γ (IFNγ)-expressing CD8 + cytotoxic T lymphocytes (CTLs) in tumor tissue, supporting tumor regression [110]. Moreover, anti-VEGFR-1 mAb D16F7 enhanced the antitumor impacts of the anti-CTLA-4 and anti-PD-1 mAbs in B16F10 melanoma cell bearing mice most potently by augmented M1/M2 and CTLs/Tregs ratios, which offer an antitumor and immunostimulating TME [111].
Recent clinical trials have also shown that bevacizumab plus atezolizumab could induce synergistic influence on the median OS of patients with RCC [112], and also in combination with nivolumab could elicit modest efficacy in ovarian cancer patients [113]. Also, co-administration of PD-L1 inhibitor avelumab with axitinib resulted in improved objective response rate (ORR) in HCC [114] and also RCC [115] patients, with acceptable safety profile. Also, combination therapy with axitinib and pembrolizumab enhanced median progression-free survival (PFS) in sarcoma patients more evidently than axitinib or pembrolizumab monotherapy. The most common treatment-related unwanted events were autoimmune colitis, pneumothorax, transaminitis, seizures, hemoptysis, and hypertriglyceridemia [116]. Besides, co-administration of regorafenib plus nivolumab resulted in significant antitumor impacts in patients with gastric cancer and CRC [117]. The objective response rate (ORR) was 44% in gastric cancer and 36% in CRC, and also median PFS was 5.6 in gastric cancer and 7.9 months in CRC patients [117]. Moreover, co-administration of nivolumab plus sunitinib or pazopanib showed a significant anti-tumor effect in advanced RCC patients [118, 119]. Conversely, other trials revealed that combined use of regorafenib plus nivolumab [120] and also ramucirumab plus pembrolizumab [121] had no remarkable therapeutic merits in CRC patients [120] and patients with advanced biliary tract cancer (BTC) [121], respectively.
Anti-angiogenic agents plus cancer vaccines
Therapeutic cancer vaccines ease tumor regression, remove minimal residual disease (MRD), entice durable antitumor memory, and also averts non-specific or adverse events [122, 123]. Till, FDA has approved three cancer vaccines, comprising Bacillus Calmette-Guérin (BCG) lives, sipuleucel-T, and also talimogene laherparepvec (T-VEC) respectively for patients with early-stage bladder cancer, prostate cancer as well as melanoma [124].
In the melanoma mice model, Bose and coworkers found that a treatment regimen comprising a 7-day course of axitinib (0.5 mg/day provided orally) in combination with a vaccine (ovalbumin (OVA) peptide-pulsed syngenic dendritic cells (DCs) adenovirally-engineered to produce anti-angiogenic cytokine IL-12p70) caused remarkable protection versus melanoma progress and prolonged OS when compared to mice receiving each agent alone [125]. These desired outcomes are probably exerted by a decrease in myeloid-derived suppressor cells (MDSC) and Treg frequencies in the tumor concomitant with induction and recruitment of CTLs in TME [125]. Also, addition of the axitinib to oncolytic herpes simplex virus (oHSV) expressing murine IL12 (G47Δ-mIL12) triggered improved OS in both immunodeficient and immunocompetent orthotopic glioblastoma mice models than mice receiving monotherapy [126]. Notably, the addition of the ICI did not promote efficacy in mice models [126]. As well, combination therapy with sunitinib and vesicular stomatitis virus (VSV) brought about the eradication of prostate, breast, and kidney malignant tumors in mice, while monotherapy with VSV or sunitinib did not [127]. Importantly, enhancement in median viral titers by 23-fold following combination therapy indicated that this regimen could potentiate oncolytic virotherapy permitting the recovery of tumor-bearing animals.
In RCC and NSCLC mice model, co-injection of reovirus and sunitinib more potently attenuated tumor burden supporting improved OS, and also reduced the population of immune suppressor cells in tumors compared with monotherapy with reovirus [128]. Thereby, it appears that this regimen can be a rational and effective strategy ready for clinical testing against RCC and NSCLC. Also, Tan and coworkers showed that the bevacizumab improved viral distribution and also tumor hypoxia and promoted the population of apoptotic cells and thus stimulated a synergistic antitumor impact when used in combination with oHSV in TNBC murine models [129]. Combining bevacizumab with OHSV expressing vasculostatin (RAMBO) also demonstrated great anti-tumor capacities in glioma xenografts [130]. Correspondingly, intratumorally administration of RAMBO 1 week after tumor inoculation, and intraperitoneally administration of bevacizumab twice a week reduced migration as well as invasion of glioma cells [130]. Co-treated mice also experienced improved OS and dampened tumor invasion than those treated with bevacizumab alone [130]. In another study, combining tumor antigen-loaded DCs vaccination and anti-angiogenic molecule lenalidomide synergistically potentiated antitumor immunity in the mice colon cancer model, largely provided by suppressing the establishment of immune suppressive cells and also activation of effector cells, such as natural killer (NK) cells [131]. As combination therapy convinced superior polarization of Th1/Th2 ratio in favor of Th1 immune response, it was signified that the applied combination method with DCs and lenalidomide could offer an innovative therapeutic alternative for the amelioration of colon cancer therapy [131]. This regimen similarly caused a robust reduction in tumor growth and malignant cell spread in lymphoma [132] and also myeloma [133] xenografts by similar mechanisms. Further, lenalidomide in combination with a fusion DNA lymphoma vaccine reduced the systemic population of MDSC and Treg in tumor-bearing mice and also led to the decreased tumor burden [134]. In addition, the combination therapy supported the incidence of the higher rates of the antitumor T cells, providing further rationale for clinical application [134].
Currently, a clinical trial was conducted to address the safety and efficacy of combination therapy with sipuleucel-T as a cellular prostate cancer vaccine with bevacizumab in 22 prostate cancer patients [135]. Combination therapy persuaded immune reactions and also alleviated prostate-specific antigen (PSA) in participants with biochemically recurrent prostate cancer [135]. In contrast, co-administration of bevacizumab plus MA950 multi-peptide vaccine adjuvanted with poly-ICLC (polyinosinic-polycytidylic acid stabilized with polylysine and carboxymethylcellulose) did not show superiority over monotherapy with each agent in terms of alteration in OS and PFS in glioblastoma patients [136]. However, a phase II study evaluating the safety and efficacy of bevacizumab in combination with ERC1671, advanced immunotherapy based on freshly extracted tumor cells and lysates, revealed that this regimen could prolong the OS in patients who received ERC1671 plus bevacizumab compared to bevacizumab monotherapy (12 months versus 7.5 months) [137]. Also, there was a tight positive association between the CD4 + T-lymphocyte count and OS in treated patients [137]. Besides, evaluation of the safety, tolerability, and anti-myeloma activity of the PVX-410, a novel tetra-peptide vaccine with 3 of the 4 antigens (XBP1 [2 splice variants] and CD138) with or without lenalidomide was accomplished in MM patients by Nooka et al. [138]. They showed that the PVX-410 vaccine was well tolerated, accompanied by mild injection site reactions and constitutional symptoms. Meanwhile, 5 of 12 patients showed clinical response to combination therapy [138]. The therapeutic values of combination therapy also were verified by an enhancement in frequency tetramer-positive cells as well as IFN-γ cells in the CD3 + CD8 + cell population [138]. Importantly, CRC patients presented complete pathological remission following treatment with bevacizumab, oxaliplatin plus leucovorin and 5-fluorouracil (FOLFOX-4), surgery, and the oncolytic virus Rigvir [139]. In consistence with previous findings, it appears that angiogenesis blockade therapy could promote viral delivery through targeting the TME [139].
Anti-angiogenic agents plus adoptive cell transfer (ACT)
Adoptive cell therapy (ACT) with using TILs or genetically-modified T cells expressing novel T cell receptors (TCR) or chimeric antigen receptors (CAR) T cells or CAR-NK cells is another plan to convince the immune system to stimulate recognition of the maligned cells and then their eradication [140, 141]. ACT-based immunotherapies can elicit significant tumor regression in animal models and also up to 70% of metastatic melanoma patients. Notwithstanding, tumor vasculature usually obstructs the tumor-specific T cells infiltration, averting anti-tumor immunity. Recent studies delivered proof of the notion that disrupting VEGF/VEGFR-2 signaling could improve the effectiveness of the ACT in tumor model [142]. In the B16 melanoma mice model, co-administration of anti-VEGF mAb to ACT abrogated tumor progress and improve OS [142]. Importantly, anti-VEGF, but not anti-VEGFR-2, antibody considerably augmented infiltration of injected cells into the tumor, suggesting that normalization of tumor vasculature by suppressing VEGF/VEGFR-2 axis could upsurge extravasation of administrated T cells into the tumor [142]. Similarly, anti-angiogenic therapy could also improve the antitumor functions of cytokine-induced killer cells (CIK cells) cells by normalizing tumor vasculature and alleviating the hypoxic TME, as shown in NSCLC xenografts [143]. Meanwhile, Shi et al. evaluated the therapeutic benefits of combination therapy with recombinant human endostatin (rh-endostatin) and CIK cells in NSCLC murine model. They exhibited that rh-endostatin normalized tumor vasculature and attenuated hypoxic regions in the TME [143]. The rh-endostatin markedly potentiated the administrated CIK cells homing and also reduced immune suppressive cells frequency in the tumor tissue. On the other hand, the used regimen instigated a higher level of TILs in tumor tissue [143]. Further, GD2-redirected CAR T cells plus bevacizumab displayed a remarkable anti-tumor effect in an orthotopic xenograft model of human neuroblastoma [144]. Co-administration of bevacizumab or ganglioside GD2-CAR T cells or both by single systemic injection supported higher rates of CAR T cells infiltration into tumor tissue accompanied with improved IFN-γ levels in TME. Additionally, the analysis presented that PD-L1 blockade therapy might augment the efficacy of this regimen [144]. Likewise, epithelial cell adhesion molecule (EpCAM) redirected CAR NK-92 cells injection resulted in CRC cell regression in animal models, which was potentiated when used in combination with regorafenib [145]. These findings delivered a novel plan for the treatment of CRC and also other solid tumors.
Anti-angiogenic agents plus chemotherapy
Anti-angiogenic agents as noticed can transiently stimulate a functional normalization of the disorganized labyrinth of vessels, sustaining the therapeutic efficacy of coadministered chemotherapeutic agents. Notwithstanding, durable angiogenesis suppression usually fences tumor uptake of chemotherapeutic drugs, and so accomplishment of further studies in this context are urgently required [146]. Correspondingly, designing intermittent treatment schedules is of paramount significance [147].
A study in 9L glioma cell-bearing rats showed that coadministration of axitinib with metronomic cyclophosphamide potently suppressed tumor progress, whereas multiple treatment cycles were needed by monotherapy with metronomic cyclophosphamide to abrogate tumor growth [148]. Importantly, axitinib had no impact on hepatic activation of cyclophosphamide, while it significantly attenuated 9L tumor uptake of cyclophosphamide activated metabolite, 4-hydroxy-cyclophosphamide (4-OH-CPA), by 30–40% [148]. Unfortunately, the abridged tumor infiltration of 4-OH-CPA resulted in a reduction in cyclophosphamide-mediated 9L cell elimination [148]. Such events in turn underlined lacking tumor complete regression by applied combined regimen, reflecting the importance of the optimization of drug scheduling and dosages. In another study, co-administration of the bevacizumab plus cisplatin and paclitaxel concurrently also induced reduced tumor growth as well as improved OS in ovarian cancer xenografts [149]. Also, monotherapy with bevacizumab suppressed ascites formation, accompanied by the partial impact on tumor burden [149]. TNP-470, an angiogenesis inhibitor, plus cisplatin inhibited the liver metastasis of human pancreatic carcinoma [150]. Indeed, liver metastasis percentages reduced from 81.8% in the cisplatin group and 73.3% in the TNP-470 group to 40% in TNP-470 plus cisplatin group. While monotherapy with each agent did not modify tumor growth in vivo, the addition of TNP-470 to cisplatin strikingly reduced tumor growth [150]. Of course, it seems that TNP-470 may entice a decrease in glioma tumor uptake of some chemotherapeutic drugs, such as temozolomide, by affecting the tumor vasculature as a result of its pharmacodynamic effect [151]. As cited, more comprehensive studies are required to define how these combinations can efficiently be utilized. Another study also exhibited that the addition of the TNP-470 to cisplatin chemotherapy reduced the microvascular density of bladder cancer in a murine model [152]. Nonetheless, TNP-470 has no significant influence on the cisplatin impact versus bladder cancer as determined by apoptosis and cell proliferation [152]. Besides, Bow and coworkers demonstrated that local delivery of angiogenesis-inhibitor minocycline could potentiate the anti-tumor efficacy of radiotherapy (RT) and oral temozolomide, as evidenced by enhanced OS in a rodent glioma model [153]. These findings offered further evidence for the idea that angiogenesis inhibitors in combination with conventional therapeutic modalities could promote OS in glioblastoma patients [153].
In 2007, a clinical trial on 25 patients with advanced CRC documented the safety and well-tolerability of combining angiogenesis inhibitor vatalanib, an inhibitor of VEGFR tyrosine kinases, with oxaliplatin/5-FU/leucovorin (FOLFOX4) chemotherapy [154]. Moreover, the addition of the novel anti-angiogenic agent, SU5416, to paclitaxel supported improved PFS accompanied with some mild to modest adverse events (e.g., headache, facial flushing, and fatigue) in patients with head and neck cancer [155]. However, the regimen led to the occurrences of thromboembolic events and prophylactic anticoagulation, suggesting that careful consideration must be taken. Besides, TSU-68 when used plus carboplatin and paclitaxel showed a manageable safety profile in NSCLC patients [156]. Likewise, the addition of the angiogenesis inhibitor ABT-510 (50 mg and 100 mg) as a peptide mimetics of thrombospondin-1 with chemotherapeutic agents (gemcitabine/cisplatin) demonstrated acceptable safety as well as feasibly in patients with NSCLC [157]. Furthermore, combining TNP-470 and paclitaxel was well tolerated with no significant pharmacokinetic interaction between them in NSCLC patients [158]. Further, several clinical trials have verified the efficacy of combination therapy with anti-angiogenic agent and conventional therapy in patients with ovarian cancer [159, 160], CRC [161, 162], NSCLC [163], MCL [164] and also MM [165]. For instance combination therapy with bevacizumab and paclitaxel plus carboplatin prolonged the median OS in participants with platinum-sensitive recurrent ovarian cancer [159]. Also, bevacizumab in combination with low-dose RT and concurrent FOLFIRI induced remarkable objective response (about 39%) in CRC patients [161]. Finally, axitinib combined with cisplatin and gemcitabine [166] and also bevacizumab plus paclitaxel and carboplatin [163] induced significant anti-tumor effect in NSCLC patients, as documented by improved OS and PFS. In addition, the Ziv-aflibercept in combination with 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI) significantly promoted OS in a phase III study of patients with metastatic CRC previously treated with an oxaliplatin-based regimen [167]. However, Ziv-aflibercept in combination with cisplatin and pemetrexed did not significantly affect OS and PFS in patients with previously untreated NSCLC cancer [168].
A list of trials based on combination therapy with angiogenesis inhibitors plus chemotherapy or chemoradiotherapy has been offered (Table 4).
Anti-angiogenic agents plus radiotherapy (RT)
RT crucially contributes to the multimodality treatment of cancer. Current evolving in RT have chiefly complicated improvements in dose delivery [169]. Upcoming developments in tumor therapeutics will probably include the combination of RT with targeted therapies. Meanwhile, preliminary results of anti-angiogenic agents in combination with RT have produced encouraging consequences [170]. Further, there are clear proofs that suggest that well-vascularized and perfused tumors mainly exhibit desired response to RT [171, 172].
Studies have shown that the addition of the angiogenesis-inhibitor minocycline to radiotherapy and oral temozolomide could result in prolonged OS in a murine glioma model [153]. Minocycline plus RT enhanced OS by about 140% compared with treatment with RT, while minocycline plus temozolomide improved OS by about 38% compared with monotherapy with temozolomide [153]. Anti-angiogenesis therapy using anginex in combination with RT also supported tumor control in squamous cell carcinoma (SCC) xenografts accompanied by reducing oxygen levels in tumor tissue [173]. Observation showed that the applied regimen modified the amount of functional vasculature in tumors and also augmented radiation-elicited tumor eradication [173]. Likewise, robust hindrance of tumor proliferation was achieved from the addition of the angiogenesis inhibitor TNP-470 to RT in SCC xenografts more evidently than monotherapy with each approach [174]. Also, it was speculated that exclusive investigation of each tumor neovascularization competence can be imperative before deciding the angiogenesis blockade treatment [174]. In contrast, the addition of TNP-470 to RT attenuated the tumor control probability in murine mammary carcinoma [175]. Such unanticipated consequence could be ensured from the partial reserve of reoxygenation by TNP-470, as no remarkable alteration was shown between the RT plus TNP-470 and RT alone under hypoxic conditions [175]. Also, another anti-angiogenic agent, vandetanib (ZD6474) (50 mg/kg), as a potent VEGFR2 tyrosine kinase inhibitor, showed a synergistic effect with RT (3 × 2 Gy) in the NSCLC mice model [176]. Also, vandetanib plus RT strikingly diminished tumor volume by 86% in comparison to the control group in anaplastic thyroid carcinoma (ATC) xenografts [177]. A potent anti-angiogenesis agent, liposomal honokiol, also elicited significant anti-tumor influence by stimulating apoptosis and also suppressing angiogenesis when used plus RT in Lewis lung cancer (LLC) xenografts [178]. Liposomal honokiol, in fact, could ameliorate tumor cell radiosensitivity in vivo, offering that RT plus liposomal honokiol can engender better anti-tumor efficacy in a myriad of tumors, such as lung cancer, SCC, and CRC [178,179,180].
In 2021, Yang et al. evaluated the safety and efficacy of that combination therapy with axitinib plus RT in advanced HCC patients. They exhibited that the regimen was well tolerated with an axitinib MTD of 3 mg twice daily [181]. Also, the intervention resulted in an ORR of about 66%, comprising 3 complete responses and 3 partial responses among 9 total participants [181]. Besides, the addition of the bevacizumab to adjuvant radiotherapy was associated with the manageable safety profile in breast cancer patients [182]. Meanwhile, grade 3 acute dermatitis was shown in about 10% of patients undergoing combination therapy and 5% of patients undergoing monotherapy with RT without significant modification. Also, pain (18%), fibrosis (8%), and telangiectasia (5%) were the most mutual grade 1–2 side adverse events during 1 years follow-up [182]. Likewise, erlotinib in combination with bevacizumab as well as capecitabine-based definitive chemoradiation (CRT) showed acceptable safety in unresectable pancreatic cancer patients [183]. While 33% of patients showed a grade 3 acute toxicity (including 2 diarrhea, 1 rash), no grade 4 or 5 toxicities were observed during 10 months follow-up. As well 2 of 9 participants showed complete response to intervention [183]. Too, the study of the therapeutic effects of combining RT with FOLFIRI regimen, comprising leucovorin calcium (calcium folinate), 5-fluorouracil, and irinotecan, plus bevacizumab in metastatic CRC also noted objective response in 10/10 patients (3 partial response and 7 complete response) [184]. Similarly, the same regimen caused a partial response in 15/18 or complete response in 4/18 CRC patients, whereas grade 3–4 adverse events toxicity were 2/18 patients [161]. Of course, large-scale trials on this newer therapeutic mean seem justified. Albeit there are some reports which show that combining anti-angiogenic therapy with RT had no therapeutic advantages. For instance, in rectal carcinoma patients, combination therapy with bevacizumab and capecitabine plus RT revealed no merits in terms of improved PFS or OS in the short or long term during a phase 2 clinical trial (NCT01043484) [185].
Response biomarkers for anti-angiogenic therapy
As a result of some divergences results related to anti-angiogenic agents as well as their modest responses, we must determine and categorize a spectrum of biomarkers, screening the patients of possible responders [186]. Additionally, such biomarkers are urgently required to can monitor disease development and angiogenic actions of tumors following exposure with treatment angiogenesis inhibitors. There are some reports showing that angiogenesis inhibitors could not support therapeutic effect in previously treated metastatic breast cancer [187]. These undesired events are likely related to the secretion of pro-angiogenic factors from resistant malignant tissue [188]. The finding outlines the importance of determining biomarkers to predict the efficacy of VEGF-targeted therapies. Much effort has been spent in this regard and resulted in the finding several biomarkers comprising dynamic measurements (such as variations in systemic blood pressure), circulating markers (such as VEGF serum levels), genotypic markers (such as VEGF polymorphism), blood cells frequencies (such as progenitor cells), tissue markers (such as IFP) and also imaging parameters [such as estimating capillary permeability employing magnetic resonance imaging (MRI)] [189]. Recent studies have revealed that there is a negative correlation between OS with serum lactate dehydrogenase (LDH) and neutrophil levels in CRC patients who received bevacizumab plus standard chemotherapy [190]. Besides, enhanced IL-8 levels were associated with shorter PFS, while low Ang-2 serum levels were related to improved OS in tumor patients undergoing angiogenesis blockade therapy [90]. Circulating endothelial cells (CEC) also has been determined as a robust indicator for the outcome of treatment with bevacizumab. Correspondingly, patients with less than 65 CEC/4 mL blood at baseline mainly experienced prolonged OS and PFS [191]. Besides, patients with IL-6 G-174C and P53 codon 72, MMP9 C-1562T, and CXCR-1 G + 2607C polymorphism may exhibit the favored response to anti-angiogenic agents [191]. On the other hand, greater intra-tumoral expression of VEGFR-3 may predict better response, while overexpression of VEGFR1 mainly indicates poor survival [192]. Other studies in RCC patients upon treatment with sorafenib also revealed that high baseline levels of VEGF were related to poor prognosis [193], while serum levels of circulating neutrophil gelatinase-associated lipocalin (NGAL) and VEGF were powerfully supported prolonged PFS in RCC patients receiving sunitinib [194].
Conclusion and prospect
In contrast to the classical hypothesis of vascular regression, the central aim of conventional anti-angiogenic treatments is tumor vascular normalization and maturity. This event, in turn, offered enhanced tumor access to chemotherapeutic drugs and underlays more efficient cancer immunotherapy. As cited, survival benefits of angiogenesis blockade therapy are compromised by cancer resistance to theses agent, and thereby provoke interest in evolving more effective means to combine anti-angiogenic drugs with other conventional therapeutics. To date, a large number of clinical trials have evaluated the safety and therapeutic merits of angiogenesis blockade therapy alone or in combination with other modalities in cancer panties (Fig. 3). Although combination therapy regimen mainly caused significant efficacy in cancer patients, intervention-related toxicities hurdle their application in clinic. For instance, bevacizumab therapy could sustain ischemic heart disease. Indeed, CRC patients receiving bevacizumab may experience considerably augmented possibility of cardiac ischemia [195]. In addition, it has been proved that combination therapy with angiogenesis inhibitors and chemotherapeutic agents may attenuate antitumor effects of chemotherapy. Hence, further rigorous investigations are warranted to circumvent the cited problems. Moreover, determining the suitable dose and sequence is of paramount importance to optimize the effectiveness, toxicity, and tolerability of the combination therapy. Thanks to the involvement of a myriad of cytokines and growth factors and the resultant interplay and compensation among them, co-targeting various growth factors is urgently required. The recognition and potent suppression of downstream kinases and strategic signaling biomolecules where several angiogenic pathways converge may defeat current difficulties motivated via the variety of angiogenic ligands and receptors and should be the emphasis of upcoming investigations. For instance, dual EGFR inhibition (erlotinib and cetuximab) combined with bevacizumab is a safe and well-tolerated combination, demonstrating antitumor activity in patients with solid tumors [196]. BQ13esides, continued treatment with conventional anti-angiogenic agents is related to toxicity and drug resistance. These conditions offer a robust justification for novel plans to improve the efficacy of mAbs targeting tumor vasculature, such as antibody–drug conjugates (ADCs) and peptide-drug conjugates (PDCs), offering a new avenue to exert anti-angiogenic effects on cancerous cells.
Availability of data and materials
Not applicable.
References
Folkman J. Angiogenesis. Annu Rev Med. 2006;57:1–18.
Senger DR, Davis GE. Angiogenesis. Cold Spring Harb Perspect Biol. 2011;3(8):a005090.
Aguilar-Cazares D, Chavez-Dominguez R, Carlos-Reyes A, Lopez-Camarillo C, Hernadez de la Cruz ON, Lopez-Gonzalez JS. Contribution of angiogenesis to inflammation and cancer. Front Oncol. 2019;9:1399.
Kerbel RS. Tumor angiogenesis. N Engl J Med. 2008;358(19):2039–49.
Reinmuth N, Parikh AA, Ahmad SA, Liu W, Stoeltzing O, Fan F, et al. Biology of angiogenesis in tumors of the gastrointestinal tract. Microsc Res Tech. 2003;60(2):199–207.
Rajendran JG, Krohn KA. Imaging hypoxia and angiogenesis in tumors. Radiol Clin. 2005;43(1):169–87.
Muthukkaruppan VR, Kubai L, Auerbach R. Tumor-induced neovascularization in the mouse eye23. J Natl Cancer Inst. 1982;69(3):699–708.
Parangi S, O’Reilly M, Christofori G, Holmgren L, Grosfeld J, Folkman J, et al. Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc Natl Acad Sci U S A. 1996;93(5):2002–7.
Teleanu RI, Chircov C, Grumezescu AM, Teleanu DM. Tumor angiogenesis and anti-angiogenic strategies for cancer treatment. J Clin Med. 2020;9(1):84.
Al-Abd AM, Alamoudi AJ, Abdel-Naim AB, Neamatallah TA, Ashour OM. Anti-angiogenic agents for the treatment of solid tumors: potential pathways, therapy and current strategies—a review. J Adv Res. 2017;8(6):591–605.
Dudzinski SO, Cameron BD, Wang J, Rathmell JC, Giorgio TD, Kirschner AN. Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer. J Immunother Cancer. 2019;7(1):1–8.
Lee JJ, Chu E. Adjuvant chemotherapy for stage II colon cancer: the debate goes on. J Oncol Pract. 2017;13(4):245–6.
Shahneh FZ, Baradaran B, Zamani F, Aghebati-Maleki L. Tumor angiogenesis and anti-angiogenic therapies. Hum Antib. 2013;22(1–2):15–9.
Sharma PS, Sharma R, Tyagi T. VEGF/VEGFR pathway inhibitors as anti-angiogenic agents: present and future. Curr Cancer Drug Targets. 2011;11(5):624–53.
De Rosa L, Di Stasi R, D’Andrea LD. Pro-angiogenic peptides in biomedicine. Arch Biochem Biophys. 2018;660:72–86.
Li X, Eriksson U. Novel VEGF family members: VEGF-B, VEGF-C and VEGF-D. Int J Biochem Cell Biol. 2001;33(4):421–6.
Tomanek RJ, Holifield JS, Reiter RS, Sandra A, Lin JJ. Role of VEGF family members and receptors in coronary vessel formation. Dev Dyn. 2002;225(3):233–40.
Shibuya M. Vascular Endothelial Growth Factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer. 2011;2(12):1097–105.
Zachary I. Neuropilins: role in signalling, angiogenesis and disease. Chem Immunol Allergy. 2014;99:37–70.
Mercurio AM. VEGF/neuropilin signaling in cancer stem cells. Int J Mol Sci. 2019;20(3):490.
Finley SD, Popel AS. Predicting the effects of anti-angiogenic agents targeting specific VEGF isoforms. AAPS J. 2012;14(3):500–9.
Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J Hematol Oncol. 2019;12(1):27.
Dey N, De P, Brian L-J. Evading anti-angiogenic therapy: resistance to anti-angiogenic therapy in solid tumors. Am J Transl Res. 2015;7(10):1675.
Liang P, Ballou B, Lv X, Si W, Bruchez MP, Huang W, et al. Monotherapy and combination therapy using anti-angiogenic nanoagents to fight cancer. Adv Mater. 2021;33(15):2005155.
Ribatti D, Nico B, Crivellato E. The role of pericytes in angiogenesis. Int J Dev Biol. 2011;55(3):261–8.
Wang X, Ma W, Han S, Meng Z, Zhao L, Yin Y, et al. TGF-β participates choroid neovascularization through Smad2/3-VEGF/TNF-α signaling in mice with Laser-induced wet age-related macular degeneration. Sci Rep. 2017;7(1):1–13.
Kim JH, Kim S-K, Wang K-C. Ischemia/angiogenesis-related molecules and cells. Moyamoya disease update. Tokya: Springer; 2010. p. 73–81.
Jayatilleke KM, Hulett MD. Heparanase and the hallmarks of cancer. J Transl Med. 2020;18(1):1–25.
Vempati P, Popel AS, Mac GF. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev. 2014;25(1):1–19.
Rundhaug JE. Matrix metalloproteinases, angiogenesis, and cancer: commentary re: AC Lockhart et al., reduction of wound angiogenesis in patients treated with BMS-275291, a broad spectrum matrix metalloproteinase inhibitor. Clin. Cancer Res., 9: 00–00, 2003. Clin Cancer Res. 2003;9(2):551–4.
Rundhaug JE. Matrix metalloproteinases and angiogenesis. J Cell Mol Med. 2005;9(2):267–85.
Tang Y, Nakada MT, Kesavan P, McCabe F, Millar H, Rafferty P, et al. Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases. Can Res. 2005;65(8):3193–9.
Lamalice L, Le Boeuf F, Huot J. Endothelial cell migration during angiogenesis. Circ Res. 2007;100(6):782–94.
Colomb F, Wang W, Simpson D, Zafar M, Beynon R, Rhodes JM, et al. Galectin-3 interacts with the cell-surface glycoprotein CD146 (MCAM, MUC18) and induces secretion of metastasis-promoting cytokines from vascular endothelial cells. J Biol Chem. 2017;292(20):8381–9.
Tatum JL, Hoffman JM. Angiogenesis imaging methodology: AIM for clinical trials. The role of imaging in clinical trials of anti-angiogenesis therapy in oncology program, http:== imaging cancer gov= reportsandpublications= AngiogenesisImagingMethodsfor ClinicalTrials. 2005.
Voron T, Marcheteau E, Pernot S, Colussi O, Tartour E, Taieb J, et al. Control of the immune response by pro-angiogenic factors. Front Oncol. 2014;4:70.
Sheu B-C, Chang W-C, Cheng C-Y, Lin H-H, Chang D-Y, Huang S-C. Cytokine regulation networks in the cancer microenvironment. Front Biosci. 2008;13(13):6255–68.
Fahey E, Doyle SL. IL-1 family cytokine regulation of vascular permeability and angiogenesis. Front Immunol. 2019;10:1426.
Mountain DJ, Singh M, Menon B, Singh K. Interleukin-1β increases expression and activity of matrix metalloproteinase-2 in cardiac microvascular endothelial cells: role of PKCα/β1 and MAPKs. Am J Physiol Cell Physiol. 2007;292(2):C867–75.
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;183(7):4705–14.
Huang Q, Duan I, Qian X, Fan J, Lv Z, Zhang X, et al. IL-17 promotes angiogenic factors IL-6, IL-8, and Vegf production via Stat1 in lung adenocarcinoma. Sci Rep. 2016;6(1):36551.
Rapisarda A, Hollingshead M, Uranchimeg B, Bonomi CA, Borgel SD, Carter JP, et al. Increased antitumor activity of bevacizumab in combination with hypoxia inducible factor-1 inhibition. Mol Cancer Ther. 2009;8(7):1867–77.
Yang Y, Sun M, Wang L, Jiao B. HIFs, angiogenesis, and cancer. J Cell Biochem. 2013;114(5):967–74.
Niu Y, Bao L, Chen Y, Wang C, Luo M, Zhang B, et al. HIF2-induced long noncoding RNA RAB11B-AS1 promotes hypoxia-mediated angiogenesis and breast cancer metastasis. Can Res. 2020;80(5):964–75.
Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel). 2014;6(3):1670–90.
Wang L, He T, Liu J, Tai J, Wang B, Chen Z, et al. Pan-cancer analysis reveals tumor-associated macrophage communication in the tumor microenvironment. Exp Hematol Oncol. 2021;10(1):31.
Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182–6.
McCormack PL, Keam SJ. Bevacizumab. Drugs. 2008;68(4):487–506.
Sandler A. Bevacizumab in non-small cell lung cancer. Clin Cancer Res. 2007;13(15):4613s-s4616.
Wu H-C, Huang C-T, Chang D-K. Anti-angiogenic therapeutic drugs for treatment of human cancer. J Cancer Mol. 2008;4(2):37–45.
Mukherji S. Bevacizumab (avastin). Am J Neuroradiol. 2010;31(2):235–6.
Kelly RJ, Rixe O. Axitinib (AG-013736). Small Mol Oncol. 2010. https://doi.org/10.1007/978-3-642-01222-8_3.
Spano JP, Chodkiewicz C, Maurel J, Wong R, Wasan H, Barone C, et al. Efficacy of gemcitabine plus axitinib compared with gemcitabine alone in patients with advanced pancreatic cancer: an open-label randomised phase II study. Lancet. 2008;371(9630):2101–8.
Houghton PJ. Everolimus. Clin Cancer Res. 2010;16(5):1368–72.
Singh S, Carnaghi C, Buzzoni R, Pommier RF, Raderer M, Tomasek J, et al. Everolimus in neuroendocrine tumors of the gastrointestinal tract and unknown primary. Neuroendocrinology. 2018;106(3):211–20.
Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, et al. Cabozantinib in progressive medullary thyroid cancer. J Clin Oncol. 2013;31(29):3639.
Chanzá NM, Xie W, Bilen MA, Dzimitrowicz H, Burkart J, Geynisman DM, et al. Cabozantinib in advanced non-clear-cell renal cell carcinoma: a multicentre, retrospective, cohort study. Lancet Oncol. 2019;20(4):581–90.
Armoiry X, Aulagner G, Facon T. Lenalidomide in the treatment of multiple myeloma: a review. J Clin Pharm Ther. 2008;33(3):219–26.
Talati C, Sallman D, List A, editors. Lenalidomide: myelodysplastic syndromes with del (5q) and beyond. Seminars in hematology. Amsterdam: Elsevier; 2017.
Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55.
Leonetti A, Leonardi F, Bersanelli M, Buti S. Clinical use of lenvatinib in combination with everolimus for the treatment of advanced renal cell carcinoma. Ther Clin Risk Manag. 2017;13:799.
Keisner SV, Shah SR. Pazopanib. Drugs. 2011;71(4):443–54.
Poole RM, Vaidya A. Ramucirumab: first global approval. Drugs. 2014;74(9):1047–58.
Garon EB, Cao D, Alexandris E, John WJ, Yurasov S, Perol M. A randomized, double-blind, phase III study of docetaxel and ramucirumab versus docetaxel and placebo in the treatment of stage IV non–small-cell lung cancer after disease progression after 1 previous platinum-based therapy (REVEL): Treatment Rationale and Study Design. Clin Lung Cancer. 2012;13(6):505–9.
Verdaguer H, Tabernero J, Macarulla T. Ramucirumab in metastatic colorectal cancer: evidence to date and place in therapy. Ther Adv Med Oncol. 2016;8(3):230–42.
Syed YY. Ramucirumab: a review in hepatocellular carcinoma. Drugs. 2020;80(3):315–22.
Bruix J, Qin S, Merle P, Granito A, Huang Y-H, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2017;389(10064):56–66.
Abdelgalil AA, Alkahtani HM, Al-Jenoobi FI. Sorafenib. Profiles of drug substances, excipients and related methodology, vol. 44. Amsterdam: Elsevier; 2019. p. 239–66.
Le Tourneau C, Raymond E, Faivre S. Sunitinib: a novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors (GIST). Ther Clin Risk Manag. 2007;3(2):341.
Palumbo A, Facon T, Sonneveld P, Blade J, Offidani M, Gay F, et al. Thalidomide for treatment of multiple myeloma: 10 years later. Blood J Am Soc Hematol. 2008;111(8):3968–77.
Patel A, Sun W. Ziv-aflibercept in metastatic colorectal cancer. Biol Targets Ther. 2014;8:13.
Commander H, Whiteside G, Perry C. Vandetanib. Drugs. 2011;71(10):1355–65.
Wells SA Jr, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. J Clin Oncol. 2012;30(2):134.
Pàez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15(3):220–31.
Grepin R, Guyot M, Jacquin M, Durivault J, Chamorey E, Sudaka A, et al. Acceleration of clear cell renal cell carcinoma growth in mice following bevacizumab/Avastin treatment: the role of CXCL cytokines. Oncogene. 2012;31(13):1683–94.
Maione F, Capano S, Regano D, Zentilin L, Giacca M, Casanovas O, et al. Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J Clin Investig. 2012;122(5):1832–48.
Singh M, Couto SS, Forrest WF, Lima A, Cheng JH, Molina R, et al. Anti-VEGF antibody therapy does not promote metastasis in genetically engineered mouse tumour models. J Pathol. 2012;227(4):417–30.
Welti JC, Powles T, Foo S, Gourlaouen M, Preece N, Foster J, et al. Contrasting effects of sunitinib within in vivo models of metastasis. Angiogenesis. 2012;15(4):623–41.
Cooke VG, LeBleu VS, Keskin D, Khan Z, O’Connell JT, Teng Y, et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell. 2012;21(1):66–81.
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;10:221.
Jiang B-H, Agani F, Passaniti A, Semenza GL. V-SRC induces expression of hypoxia-inducible factor 1 (HIF-1) and transcription of genes encoding vascular endothelial growth factor and enolase 1: involvement of HIF-1 in tumor progression. Can Res. 1997;57(23):5328–35.
Pereira ER, Frudd K, Awad W, Hendershot LM. Endoplasmic reticulum (ER) stress and hypoxia response pathways interact to potentiate hypoxia-inducible factor 1 (HIF-1) transcriptional activity on targets like vascular endothelial growth factor (VEGF). J Biol Chem. 2014;289(6):3352–64.
Luo D, Wang Z, Wu J, Jiang C, Wu J. The role of hypoxia inducible factor-1 in hepatocellular carcinoma. BioMed Res Int. 2014. https://doi.org/10.1155/2014/409272.
Kitajima Y, Miyazaki K. The critical impact of HIF-1a on gastric cancer biology. Cancers. 2013;5(1):15–26.
Maione F, Molla F, Meda C, Latini R, Zentilin L, Giacca M, et al. Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor growth and normalizes tumor vasculature in transgenic mouse models. J Clin Investig. 2009;119(11):3356–72.
Fagiani E, Christofori G. Angiopoietins in angiogenesis. Cancer Lett. 2013;328(1):18–26.
Hanahan D. Signaling vascular morphogenesis and maintenance. Science. 1997;277(5322):48–50.
Eklund L, Saharinen P. Angiopoietin signaling in the vasculature. Exp Cell Res. 2013;319(9):1271–80.
Cook KM, Figg WD. Angiogenesis inhibitors: current strategies and future prospects. CA Cancer J Clin. 2010;60(4):222–43.
Goede V, Coutelle O, Neuneier J, Reinacher-Schick A, Schnell R, Koslowsky T, et al. Identification of serum angiopoietin-2 as a biomarker for clinical outcome of colorectal cancer patients treated with bevacizumab-containing therapy. Br J Cancer. 2010;103(9):1407–14.
Qin S, Yi M, Jiao D, Li A, Wu K. Distinct roles of VEGFA and ANGPT2 in lung adenocarcinoma and squamous cell carcinoma. J Cancer. 2020;11(1):153–67.
Gyanchandani R, Alves MVO, Myers JN, Kim S. A proangiogenic signature is revealed in FGF-mediated bevacizumab-resistant head and neck squamous cell carcinoma. Mol Cancer Res. 2013;11(12):1585–96.
Zhou Y, Wu C, Lu G, Hu Z, Chen Q, Du X. FGF/FGFR signaling pathway involved resistance in various cancer types. J Cancer. 2020;11(8):2000.
Okamoto S, Nitta M, Maruyama T, Sawada T, Komori T, Okada Y, et al. Bevacizumab changes vascular structure and modulates the expression of angiogenic factors in recurrent malignant gliomas. Brain Tumor Pathol. 2016;33(2):129–36.
Schmidinger M. Third-line dovitinib in metastatic renal cell carcinoma. Lancet Oncol. 2014;15(3):245–6.
Welti J, Gourlaouen M, Powles T, Kudahetti S, Wilson P, Berney D, et al. Fibroblast growth factor 2 regulates endothelial cell sensitivity to sunitinib. Oncogene. 2011;30(10):1183–93.
Chung AS, Kowanetz M, Wu X, Zhuang G, Ngu H, Finkle D, et al. Differential drug class-specific metastatic effects following treatment with a panel of angiogenesis inhibitors. J Pathol. 2012;227(4):404–16.
Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell. 2009;15(3):232–9.
Grasselly C, Denis M, Bourguignon A, Talhi N, Mathe D, Tourette A, et al. The antitumor activity of combinations of cytotoxic chemotherapy and immune checkpoint inhibitors is model-dependent. Front Immunol. 2018;9:2100.
Pérez-Ruiz E, Melero I, Kopecka J, Sarmento-Ribeiro AB, García-Aranda M, De Las Rivas J. Cancer immunotherapy resistance based on immune checkpoints inhibitors: targets, biomarkers, and remedies. Drug Resist Updates. 2020;53:100718.
Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18(1):155.
Darvin P, Toor SM, Nair VS, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):1–11.
Reck M, Shankar G, Lee A, Coleman S, McCleland M, Papadimitrakopoulou VA, et al. Atezolizumab in combination with bevacizumab, paclitaxel and carboplatin for the first-line treatment of patients with metastatic non-squamous non-small cell lung cancer, including patients with EGFR mutations. Expert Rev Respir Med. 2020;14(2):125–36.
Nowicki TS, Hu-Lieskovan S, Ribas A. Mechanisms of resistance to PD-1 and PD-L1 blockade. Cancer J (Sudbury, Mass). 2018;24(1):47.
Yi M, Niu M, Xu L, Luo S, Wu K. Regulation of PD-L1 expression in the tumor microenvironment. J Hematol Oncol. 2021;14(1):10.
Yasuda S, Sho M, Yamato I, Yoshiji H, Wakatsuki K, Nishiwada S, et al. Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR 2) induces synergistic anti-tumour effect in vivo. Clin Exp Immunol. 2013;172(3):500–6.
Meder L, Schuldt P, Thelen M, Schmitt A, Dietlein F, Klein S, et al. Combined VEGF and PD-L1 blockade displays synergistic treatment effects in an autochthonous mouse model of small cell lung cancer. Can Res. 2018;78(15):4270–81.
Wu FT, Xu P, Chow A, Man S, Krüger J, Khan KA, et al. Pre-and post-operative anti-PD-L1 plus anti-angiogenic therapies in mouse breast or renal cancer models of micro-or macro-metastatic disease. Br J Cancer. 2019;120(2):196–206.
Wang Q, Gao J, Di W, Wu X. Anti-angiogenesis therapy overcomes the innate resistance to PD-1/PD-L1 blockade in VEGFA-overexpressed mouse tumor models. Cancer Immunol Immunother. 2020;69(9):1781–99.
Schmittnaegel M, Rigamonti N, Kadioglu E, Cassará A, Rmili CW, Kiialainen A, et al. Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade. Sci Transl Med. 2017. https://doi.org/10.1126/scitranslmed.aak9670.
Lacal PM, Atzori MG, Ruffini F, Scimeca M, Bonanno E, Cicconi R, et al. Targeting the vascular endothelial growth factor receptor-1 by the monoclonal antibody D16F7 to increase the activity of immune checkpoint inhibitors against cutaneous melanoma. Pharmacol Res. 2020;159:104957.
McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med. 2018;24(6):749–57.
Liu JF, Herold C, Gray KP, Penson RT, Horowitz N, Konstantinopoulos PA, et al. Assessment of combined nivolumab and bevacizumab in relapsed ovarian cancer: a phase 2 clinical trial. JAMA Oncol. 2019;5(12):1731–8.
Kudo M, Motomura K, Wada Y, Inaba Y, Sakamoto Y, Kurosaki M, et al. First-line avelumab+ axitinib in patients with advanced hepatocellular carcinoma: results from a phase 1b trial (VEGF Liver 100). Am Soc Clin Oncol. 2019;37:4072.
Choueiri TK, Larkin JM, Oya M, Thistlethwaite FC, Martignoni M, Nathan PD, et al. First-line avelumab+ axitinib therapy in patients (pts) with advanced renal cell carcinoma (aRCC): results from a phase Ib trial. Am Soc Clin Oncol. 2017.
Wilky BA, Trucco MM, Subhawong TK, Florou V, Park W, Kwon D, et al. Axitinib plus pembrolizumab in patients with advanced sarcomas including alveolar soft-part sarcoma: a single-centre, single-arm, phase 2 trial. Lancet Oncol. 2019;20(6):837–48.
Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38(18):2053–61.
Amin A, Plimack ER, Ernstoff MS, Lewis LD, Bauer TM, McDermott DF, et al. Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma: the CheckMate 016 study. J Immunother Cancer. 2018;6(1):1–12.
Amin A, Plimack ER, Infante JR, Ernstoff MS, Rini BI, McDermott DF, et al. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combination with sunitinib or pazopanib in patients (pts) with metastatic renal cell carcinoma (mRCC). Am Soc Clin Oncol. 2014.
Li J, Cong L, Liu J, Peng L, Wang J, Feng A, et al. The efficacy and safety of regorafenib in combination with anti-PD-1 antibody in refractory microsatellite stable metastatic colorectal cancer: a retrospective study. Front Oncol. 2020;10:2449.
Arkenau HT, Martin-Liberal J, Calvo E, Penel N, Krebs MG, Herbst RS, et al. Ramucirumab plus pembrolizumab in patients with previously treated advanced or metastatic biliary tract cancer: nonrandomized, open-label, phase I trial (JVDF). Oncologist. 2018;23(12):1407.
Pardoll DM. Cancer vaccines. Nat Med. 1998;4(5):525–31.
Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3(8):630–41.
Gatti-Mays ME, Redman JM, Collins JM, Bilusic M. Cancer vaccines: enhanced immunogenic modulation through therapeutic combinations. Hum Vaccin Immunother. 2017;13(11):2561–74.
Bose A, Lowe DB, Rao A, Storkus WJ. Combined vaccine+ axitinib therapy yields superior anti-tumor efficacy in a murine melanoma model. Melanoma Res. 2012;22(3):236.
Saha D, Wakimoto H, Peters CW, Antoszczyk SJ, Rabkin SD, Martuza RL. Combinatorial effects of VEGFR kinase inhibitor axitinib and oncolytic virotherapy in mouse and human glioblastoma stem-like cell models. Clin Cancer Res. 2018;24(14):3409–22.
Jha BK, Dong B, Nguyen CT, Polyakova I, Silverman RH. Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy. Mol Ther. 2013;21(9):1749–57.
Lawson KA, Mostafa AA, Shi ZQ, Spurrell J, Chen W, Kawakami J, et al. Repurposing sunitinib with oncolytic reovirus as a novel immunotherapeutic strategy for renal cell carcinoma. Clin Cancer Res. 2016;22(23):5839–50.
Tan G, Kasuya H, Sahin TT, Yamamura K, Wu Z, Koide Y, et al. Combination therapy of oncolytic herpes simplex virus HF10 and bevacizumab against experimental model of human breast carcinoma xenograft. Int J Cancer. 2015;136(7):1718–30.
Tomita Y, Kurozumi K, Yoo JY, Fujii K, Ichikawa T, Matsumoto Y, et al. Oncolytic herpes virus armed with vasculostatin in combination with bevacizumab abrogates glioma invasion via the CCN1 and AKT signaling pathways. Mol Cancer Ther. 2019;18(8):1418–29.
Vo M-C, Nguyen-Pham T-N, Lee H-J, Lakshmi TJ, Yang S, Jung S-H, et al. Combination therapy with dendritic cells and lenalidomide is an effective approach to enhance antitumor immunity in a mouse colon cancer model. Oncotarget. 2017;8(16):27252.
Lapenta C, Donati S, Spadaro F, Lattanzi L, Urbani F, Macchia I, et al. Lenalidomide improves the therapeutic effect of an interferon-α-dendritic cell-based lymphoma vaccine. Cancer Immunol Immunother. 2019;68(11):1791–804.
Nguyen-Pham T-N, Jung S-H, Vo M-C, Thanh-Tran H-T, Lee Y-K, Lee H-J, et al. Lenalidomide synergistically enhances the effect of dendritic cell vaccination in a model of murine multiple myeloma. J Immunother. 2015;38(8):330–9.
Sakamaki I, Kwak LW, Cha S, Yi Q, Lerman B, Chen J, et al. Lenalidomide enhances the protective effect of a therapeutic vaccine and reverses immune suppression in mice bearing established lymphomas. Leukemia. 2014;28(2):329–37.
Rini BI, Weinberg V, Fong L, Conry S, Hershberg RM, Small EJ. Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (provenge) plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy. Cancer Interdiscip Int J Am Cancer Soc. 2006;107(1):67–74.
Boydell E, Marinari E, Migliorini D, Dietrich P-Y, Patrikidou A, Dutoit V. Exploratory study of the effect of ima950/poly-iclc vaccination on response to bevacizumab in relapsing high-grade glioma patients. Cancers. 2019;11(4):464.
Bota DA, Chung J, Dandekar M, Carrillo JA, Kong X-T, Fu BD, et al. Phase II study of ERC1671 plus bevacizumab versus bevacizumab plus placebo in recurrent glioblastoma: interim results and correlations with CD4+ T-lymphocyte counts. CNS Oncol. 2018;7(3):CNS22.
Nooka AK, Wang ML, Yee AJ, Kaufman JL, Bae J, Peterkin D, et al. Assessment of safety and immunogenicity of PVX-410 vaccine with or without lenalidomide in patients with smoldering multiple myeloma: a nonrandomized clinical trial. JAMA Oncol. 2018;4(12):e183267.
Tilgase A, Olmane E, Nazarovs J, Brokāne L, Erdmanis R, Rasa A, et al. Multimodality treatment of a colorectal cancer stage IV patient with FOLFOX-4, bevacizumab, Rigvir oncolytic virus, and surgery. Case Rep Gastroenterol. 2018;12(2):457–65.
Kato D, Yaguchi T, Iwata T, Morii K, Nakagawa T, Nishimura R, et al. Prospects for personalized combination immunotherapy for solid tumors based on adoptive cell therapies and immune checkpoint blockade therapies. Jpn J Clin Immunol. 2017;40(1):68–77.
Feros DL, Lane L, Ciarrochi J, Blackledge JT. Acceptance and Commitment Therapy (ACT) for improving the lives of cancer patients: a preliminary study. Psychooncology. 2013;22(2):459–64.
Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Can Res. 2010;70(15):6171–80.
Shi S, Wang R, Chen Y, Song H, Chen L, Huang G. Combining antiangiogenic therapy with adoptive cell immunotherapy exerts better antitumor effects in non-small cell lung cancer models. PLoS ONE. 2013;8(6):e65757.
Bocca P, Di Carlo E, Caruana I, Emionite L, Cilli M, De Angelis B, et al. Bevacizumab-mediated tumor vasculature remodelling improves tumor infiltration and antitumor efficacy of GD2-CAR T cells in a human neuroblastoma preclinical model. Oncoimmunology. 2018;7(1):e1378843.
Zhang Q, Zhang H, Ding J, Liu H, Li H, Li H, et al. Combination therapy with EpCAM-CAR-NK-92 cells and regorafenib against human colorectal cancer models. J Immunol Res. 2018. https://doi.org/10.1155/2018/4263520.
Lennernäs B, Albertsson P, Lennernäs H, Norrby K. Chemotherapy and antiangiogenesis. Acta Oncol. 2003;42(4):294–303.
Ma J, Waxman DJ. Combination of antiangiogenesis with chemotherapy for more effective cancer treatment. Mol Cancer Ther. 2008;7(12):3670–84.
Ma J, Waxman DJ. Modulation of the antitumor activity of metronomic cyclophosphamide by the angiogenesis inhibitor axitinib. Mol Cancer Ther. 2008;7(1):79–89.
Oliva P, Decio A, Castiglioni V, Bassi A, Pesenti E, Cesca M, et al. Cisplatin plus paclitaxel and maintenance of bevacizumab on tumour progression, dissemination, and survival of ovarian carcinoma xenograft models. Br J Cancer. 2012;107(2):360–9.
Shishido T, Yasoshima T, Denno R, Mukaiya M, Sato N, Hirata K. Inhibition of liver metastasis of human pancreatic carcinoma by angiogenesis inhibitor TNP-470 in combination with cisplatin. Jpn J Cancer Res. 1998;89(9):963–9.
Devineni D, Klein-Szanto A, Gallo JM. Uptake of temozolomide in a rat glioma model in the presence and absence of the angiogenesis inhibitor TNP-470. Cancer Res. 1996;56(9):1983–7.
Kong C, Zhu Y, Sun C, Li Z, Sun Z, Zhang X, et al. Inhibition of tumor angiogenesis during cisplatin chemotherapy for bladder cancer improves treatment outcome. Urology. 2005;65(2):395–9.
Bow H, Hwang LS, Schildhaus N, Xing J, Murray L, Salditch Q, et al. Local delivery of angiogenesis-inhibitor minocycline combined with radiotherapy and oral temozolomide chemotherapy in 9L glioma. J Neurosurg. 2014;120(3):662–9.
Thomas AL, Trarbach T, Bartel C, Laurent D, Henry A, Poethig M, et al. A phase IB, open-label dose-escalating study of the oral angiogenesis inhibitor PTK787/ZK 222584 (PTK/ZK), in combination with FOLFOX4 chemotherapy in patients with advanced colorectal cancer. Ann Oncol. 2007;18(4):782–8.
Cooney MM, Tserng K-Y, Makar V, McPeak RJ, Ingalls ST, Dowlati A, et al. A phase IB clinical and pharmacokinetic study of the angiogenesis inhibitor SU5416 and paclitaxel in recurrent or metastatic carcinoma of the head and neck. Cancer Chemother Pharmacol. 2005;55(3):295–300.
Okamoto I, Yoshioka H, Takeda K, Satouchi M, Yamamoto N, Seto T, et al. Phase I clinical study of the angiogenesis inhibitor TSU-68 combined with carboplatin and paclitaxel in chemotherapy-naive patients with advanced non-small cell lung cancer. J Thorac Oncol. 2012;7(2):427–33.
Gietema J, Hoekstra R, de Vos F, Uges D, van der Gaast A, Groen H, et al. A phase I study assessing the safety and pharmacokinetics of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 with gemcitabine and cisplatin in patients with solid tumors. Ann Oncol. 2006;17(8):1320–7.
Herbst RS, Madden TL, Tran HT, Blumenschein GR Jr, Meyers CA, Seabrooke LF, et al. Safety and pharmacokinetic effects of TNP-470, an angiogenesis inhibitor, combined with paclitaxel in patients with solid tumors: evidence for activity in non–small-cell lung cancer. J Clin Oncol. 2002;20(22):4440–7.
Coleman RL, Brady MF, Herzog TJ, Sabbatini P, Armstrong DK, Walker JL, et al. Bevacizumab and paclitaxel-carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017;18(6):779–91.
Bamias A, Gibbs E, Khoon Lee C, Davies L, Dimopoulos M, Zagouri F, et al. Bevacizumab with or after chemotherapy for platinum-resistant recurrent ovarian cancer: exploratory analyses of the AURELIA trial. Ann Oncol. 2017;28(8):1842–8.
Morganti AG, Cellini F, Mignogna S, Padula GD, Caravatta L, Deodato F, et al. Low-dose radiotherapy and concurrent FOLFIRI-bevacizumab: a Phase II study. Future Oncol. 2016;12(6):779–87.
Koukourakis MI, Giatromanolaki A, Sheldon H, Buffa FM, Kouklakis G, Ragoussis I, et al. Phase I/II trial of bevacizumab and radiotherapy for locally advanced inoperable colorectal cancer: vasculature-independent radiosensitizing effect of bevacizumab. Clin Cancer Res. 2009;15(22):7069–76.
Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355(24):2542–50.
Ruan J, Martin P, Christos P, Cerchietti L, Tam W, Shah B, et al. Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma. Blood. 2018;132(19):2016–25.
Hou J, Jin J, Xu Y, Wu D, Ke X, Zhou D, et al. Randomized, double-blind, placebo-controlled phase III study of ixazomib plus lenalidomide-dexamethasone in patients with relapsed/refractory multiple myeloma: China Continuation study. J Hematol Oncol. 2017;10(1):137.
Bondarenko IM, Ingrosso A, Bycott P, Kim S, Cebotaru CL. Phase II study of axitinib with doublet chemotherapy in patients with advanced squamous non-small-cell lung cancer. BMC Cancer. 2015;15:339.
Chau I, Fakih M, García-Alfonso P, Linke Z, Ruiz Casado A, Marques EP, et al. Safety and effectiveness of aflibercept+ fluorouracil, leucovorin, and irinotecan (FOLFIRI) for the treatment of patients with metastatic colorectal cancer (mCRC) in current clinical practice: OZONE study. Cancers. 2020;12(3):657.
Chen H, Modiano MR, Neal JW, Brahmer JR, Rigas JR, Jotte RM, et al. A phase II multicentre study of ziv-aflibercept in combination with cisplatin and pemetrexed in patients with previously untreated advanced/metastatic non-squamous non-small cell lung cancer. Br J Cancer. 2014;110(3):602–8.
Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol. 2009;10(7):718–26.
Wrona A, Dziadziuszko R, Jassem J. Combining radiotherapy with targeted therapies in non-small cell lung cancer: focus on anti-EGFR, anti-ALK and anti-angiogenic agents. Transl Lung Cancer Res. 2021;10(4):2032.
Overgaard J. Hypoxic radiosensitization: adored and ignored. J Clin Oncol. 2007;25(26):4066–74.
Gray LH, Conger AD, Ebert M, Hornsey S, Scott O. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol. 1953;26(312):638–48.
Amano M, Suzuki M, Andoh S, Monzen H, Terai K, Williams B, et al. Antiangiogenesis therapy using a novel angiogenesis inhibitor, anginex, following radiation causes tumor growth delay. Int J Clin Oncol. 2007;12(1):42–7.
Shintani S, Li C, Mihara M, Klosek SK, Terakado N, Hino S, et al. Anti-tumor effect of radiation response by combined treatment with angiogenesis inhibitor, TNP-470, in oral squamous cell carcinoma. Oral Oncol. 2006;42(1):66–72.
Murata R, Nishimura Y, Hiraoka M. An antiangiogenic agent (TNP-470) inhibited reoxygenation during fractionated radiotherapy of murine mammary carcinoma. Int J Radiat Oncol Biol Phys. 1997;37(5):1107–13.
Williams KJ, Telfer BA, Brave S, Kendrew J, Whittaker L, Stratford IJ, et al. ZD6474, a potent inhibitor of vascular endothelial growth factor signaling, combined with radiotherapy: schedule-dependent enhancement of antitumor activity. Clin Cancer Res. 2004;10(24):8587–93.
Chen Y, Gule M, Lemos R, Myers JN, Schwartz DL, Bankson JA, et al. Evaluation of vandetanib (ZD6474) and radiation therapy in an orthotopic xenograft model of anaplastic thyroid carcinoma. AACR. 2010.
Hu J, Chen LJ, Liu L, Chen X, Chen PL, Yang G, et al. Liposomal honokiol, a potent anti-angiogenesis agent, in combination with radiotherapy produces a synergistic antitumor efficacy without increasing toxicity. Exp Mol Med. 2008;40(6):617–28.
He Z, Subramaniam D, Ramalingam S, Dhar A, Postier RG, Umar S, et al. Honokiol radiosensitizes colorectal cancer cells: enhanced activity in cells with mismatch repair defects. Am J Physiol Gastrointest Liver Physiol. 2011;301(5):G929–37.
Wang X, Beitler JJ, Huang W, Chen G, Qian G, Magliocca K, et al. Honokiol radiosensitizes squamous cell carcinoma of the head and neck by downregulation of survivin. Clin Cancer Res. 2018;24(4):858–69.
Yang K-L, Chi M-S, Ko H-L, Huang Y-Y, Huang S-C, Lin Y-M, et al. Axitinib in combination with radiotherapy for advanced hepatocellular carcinoma: a phase I clinical trial. Radiat Oncol. 2021;16(1):1–9.
Pernin V, Belin L, Cottu P, Bontemps P, Lemanski C, De La Lande B, et al. Late toxicities and outcomes of adjuvant radiotherapy combined with concurrent bevacizumab in patients with triple-negative non-metastatic breast cancer. Br J Radiol. 2015;88(1048):20140800.
Chadha AS, Skinner HD, Gunther JR, Munsell MF, Das P, Minsky BD, et al. Phase i trial of consolidative radiotherapy with concurrent bevacizumab, erlotinib and capecitabine for unresectable pancreatic cancer. PLoS ONE. 2016;11(6):e0156910.
Morganti AG, Mignogna S, Caravatta L, Deodato F, Macchia G, Plantamura NM, et al. FOLFIRI-bevacizumab and concurrent low-dose radiotherapy in metastatic colorectal cancer: preliminary results of a phase I-II study. J Chemother. 2014;26(6):353–8.
Salazar R, Capdevila J, Manzano JL, Pericay C, Martínez-Villacampa M, López C, et al. Phase II randomized trial of capecitabine with bevacizumab and external beam radiation therapy as preoperative treatment for patients with resectable locally advanced rectal adenocarcinoma: long term results. BMC Cancer. 2020;20(1):1–11.
Abdalla AME, Xiao L, Ullah MW, Yu M, Ouyang C, Yang G. Current challenges of cancer anti-angiogenic therapy and the promise of nanotherapeutics. Theranostics. 2018;8(2):533–48.
Miller KD, Chap LI, Holmes FA, Cobleigh MA, Marcom PK, Fehrenbacher L, et al. Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol. 2005;23(4):792–9.
Relf M, LeJeune S, Scott PA, Fox S, Smith K, Leek R, et al. Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor β-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Can Res. 1997;57(5):963–9.
Jain RK, Duda DG, Willett CG, Sahani DV, Zhu AX, Loeffler JS, et al. Biomarkers of response and resistance to antiangiogenic therapy. Nat Rev Clin Oncol. 2009;6(6):327–38.
Cetin B, Kaplan MA, Berk V, Ozturk SC, Benekli M, Isikdogan A, et al. Prognostic factors for overall survival in patients with metastatic colorectal carcinoma treated with vascular endothelial growth factor-targeting agents. Asian Pac J Cancer Prev. 2012;13(3):1059–63.
Singh H, Pohl A, El-Khoueiry A, Lurje G, Zhang W, Yang D, et al. Use of genetic variants to predict clinical outcome in patients (pts) with metastatic colorectal cancer (mCRC) treated with first-line 5-FU or capecitabine in combination with oxaliplatin and bevacizumab (FOLFOX/BV or XELOX/BV). J Clin Oncol. 2009;27(15_suppl):4070.
Fountzilas G, Kourea HP, Bobos M, Televantou D, Kotoula V, Papadimitriou C, et al. Paclitaxel and bevacizumab as first line combined treatment in patients with metastatic breast cancer: the Hellenic Cooperative Oncology Group experience with biological marker evaluation. Anticancer Res. 2011;31(9):3007–18.
Eisen T, Bukowski R, Staehler M, Szczylik C, Oudard S, Stadler W, et al. Randomized phase III trial of sorafenib in advanced renal cell carcinoma (RCC): impact of crossover on survival. J Clin Oncol. 2006;24(18_suppl):4524.
Porta C, Paglino C, De Amici M, Quaglini S, Sacchi L, Imarisio I, et al. Predictive value of baseline serum vascular endothelial growth factor and neutrophil gelatinase-associated lipocalin in advanced kidney cancer patients receiving sunitinib. Kidney Int. 2010;77(9):809–15.
Chen X-L, Lei Y-H, Liu C-F, Yang Q-F, Zuo P-Y, Liu C-Y, et al. Angiogenesis inhibitor bevacizumab increases the risk of ischemic heart disease associated with chemotherapy: a meta-analysis. PLoS ONE. 2013;8(6):e66721.
Subbiah V, Dumbrava EI, Jiang Y, Thein KZ, Naing A, Hong DS, et al. Dual EGFR blockade with cetuximab and erlotinib combined with anti-VEGF antibody bevacizumab in advanced solid tumors: a phase 1 dose escalation triplet combination trial. Exp Hematol Oncol. 2020;9(1):7.
Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.
Hodi FS, Lawrence D, Lezcano C, Wu X, Zhou J, Sasada T, et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014;2(7):632–42.
Wu X, Li J, Connolly EM, Liao X, Ouyang J, Giobbie-Hurder A, et al. Combined anti-VEGF and anti-CTLA-4 therapy elicits humoral immunity to galectin-1 which is associated with favorable clinical outcomes. Cancer Immunol Res. 2017;5(6):446–54.
Moore KN, Bookman M, Sehouli J, Miller A, Anderson C, Scambia G, et al. Atezolizumab, bevacizumab, and chemotherapy for newly diagnosed stage III or IV Ovarian cancer: placebo-controlled randomized phase III trial (IMagyn050/GOG 3015/ENGOT-OV39). J Clin Oncol. 2021;39(17):1842–55.
Atkins MB, Plimack ER, Puzanov I, Fishman MN, McDermott DF, Cho DC, et al. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: a non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial. Lancet Oncol. 2018;19(3):405–15.
Amin A, Plimack ER, Ernstoff MS, Lewis LD, Bauer TM, McDermott DF, et al. Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma: the CheckMate 016 study. J Immunother Cancer. 2018;6(1):109.
Apolo AB, Nadal R, Girardi DM, Niglio SA, Ley L, Cordes LM, et al. Phase I study of cabozantinib and nivolumab alone or with ipilimumab for advanced or metastatic urothelial carcinoma and other genitourinary tumors. J Clin Oncol. 2020;38(31):3672–84.
Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, et al. Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2021;384(9):829–41.
Sakamuri D, Glitza IC, Betancourt Cuellar SL, Subbiah V, Fu S, Tsimberidou AM, et al. Phase I dose-escalation study of anti-CTLA-4 antibody ipilimumab and lenalidomide in patients with advanced cancers. Mol Cancer Ther. 2018;17(3):671–6.
Taylor MH, Lee CH, Makker V, Rasco D, Dutcus CE, Wu J, et al. Phase IB/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol. 2020;38(11):1154–63.
Lee CH, Shah AY, Rasco D, Rao A, Taylor MH, Di Simone C, et al. Lenvatinib plus pembrolizumab in patients with either treatment-naive or previously treated metastatic renal cell carcinoma (Study 111/KEYNOTE-146): a phase 1b/2 study. Lancet Oncol. 2021;22(7):946–58.
Bang YJ, Golan T, Dahan L, Fu S, Moreno V, Park K, et al. Ramucirumab and durvalumab for previously treated, advanced non-small-cell lung cancer, gastric/gastro-oesophageal junction adenocarcinoma, or hepatocellular carcinoma: an open-label, phase Ia/b study (JVDJ). Eur J Cancer. 2020;137:272–84.
Wakelee HA, Dahlberg SE, Keller SM, Tester WJ, Gandara DR, Graziano SL, et al. Adjuvant chemotherapy with or without bevacizumab in patients with resected non-small-cell lung cancer (E1505): an open-label, multicentre, randomised, phase 3 trial. Lancet Oncol. 2017;18(12):1610–23.
Yang KL, Chi MS, Ko HL, Huang YY, Huang SC, Lin YM, et al. Axitinib in combination with radiotherapy for advanced hepatocellular carcinoma: a phase I clinical trial. Radiat Oncol. 2021;16(1):18.
Chibaudel B, Henriques J, Rakez M, Brenner B, Kim TW, Martinez-Villacampa M, et al. Association of bevacizumab plus oxaliplatin-based chemotherapy with disease-free survival and overall survival in patients with stage II colon cancer: a secondary analysis of the AVANT trial. JAMA Netw Open. 2020;3(10):e2020425.
Jiang X, Guan W, Li M, Liang W, Qing Y, Dai N, et al. Endostatin combined with platinum-based chemo-radiotherapy for advanced non-small cell lung cancer. Cell Biochem Biophys. 2015;71(2):571–7.
Velenik V, Ocvirk J, Music M, Bracko M, Anderluh F, Oblak I, et al. Neoadjuvant capecitabine, radiotherapy, and bevacizumab (CRAB) in locally advanced rectal cancer: results of an open-label phase II study. Radiat Oncol. 2011;6:105.
Lan CY, Wang Y, Xiong Y, Li JD, Shen JX, Li YF, et al. Apatinib combined with oral etoposide in patients with platinum-resistant or platinum-refractory ovarian cancer (AEROC): a phase 2, single-arm, prospective study. Lancet Oncol. 2018;19(9):1239–46.
Yu X, Wang QX, Xiao WW, Chang H, Zeng ZF, Lu ZH, et al. Neoadjuvant oxaliplatin and capecitabine combined with bevacizumab plus radiotherapy for locally advanced rectal cancer: results of a single-institute phase II study. Cancer Commun (Lond). 2018;38(1):24.
Jiang X, Ding M, Qiao Y, Liu Y, Liu L. Recombinant human endostatin combined with radiotherapy in the treatment of brain metastases of non-small cell lung cancer. Clin Transl Oncol. 2014;16(7):630–6.
Crane CH, Ellis LM, Abbruzzese JL, Amos C, Xiong HQ, Ho L, et al. Phase I trial evaluating the safety of bevacizumab with concurrent radiotherapy and capecitabine in locally advanced pancreatic cancer. J Clin Oncol. 2006;24(7):1145–51.
Salazar R, Capdevila J, Manzano JL, Pericay C, Martínez-Villacampa M, López C, et al. Phase II randomized trial of capecitabine with bevacizumab and external beam radiation therapy as preoperative treatment for patients with resectable locally advanced rectal adenocarcinoma: long term results. BMC Cancer. 2020;20(1):1164.
Zhao Z, Li J, Ye R, Wu X, Gao L, Niu B. A phase II clinical study of combining FOLFIRI and bevacizumab plus erlotinib in 2nd-line chemotherapy for patients with metastatic colorectal cancer. Medicine (Baltimore). 2017;96(30):e7182.
Yu JP, Sun SP, Sun ZQ, Ni XC, Wang J, Li Y, et al. Clinical trial of thalidomide combined with radiotherapy in patients with esophageal cancer. World J Gastroenterol. 2014;20(17):5098–103.
Schneider BJ, Gadgeel SM, Ramnath N, Wozniak AJ, Dy GK, Daignault S, et al. Phase II trial of sunitinib maintenance therapy after platinum-based chemotherapy in patients with extensive-stage small cell lung cancer. J Thorac Oncol. 2011;6(6):1117–20.
Resch G, De Vries A, Öfner D, Eisterer W, Rabl H, Jagoditsch M, et al. Preoperative treatment with capecitabine, bevacizumab and radiotherapy for primary locally advanced rectal cancer–a two stage phase II clinical trial. Radiother Oncol. 2012;102(1):10–3.
Hansen TF, Nielsen BS, Sørensen FB, Johnsson A, Jakobsen A. Epidermal growth factor-like domain 7 predicts response to first-line chemotherapy and bevacizumab in patients with metastatic colorectal cancer. Mol Cancer Ther. 2014;13(9):2238–45.
Jones BS, Jerome MS, Miley D, Jackson BE, DeShazo MR, Reddy VV, et al. Pilot phase II study of metronomic chemotherapy in combination with bevacizumab in patients with advanced non-squamous non-small cell lung cancer. Lung Cancer. 2017;106:125–30.
Guan ZZ, Xu JM, Luo RC, Feng FY, Wang LW, Shen L, et al. Efficacy and safety of bevacizumab plus chemotherapy in Chinese patients with metastatic colorectal cancer: a randomized phase III ARTIST trial. Chin J Cancer. 2011;30(10):682–9.
Ohta T, Kato T, Kawakami H, Miyake Y, Goto M, Iwamoto S, et al. Phase II study of 5-fluorouracil-leucovorin plus bevacizumab for chemotherapy-naïve older or frail patients with metastatic colorectal cancer (OGSG 0802). Int J Clin Oncol. 2020;25(7):1291–8.
Acknowledgements
Not applicable.
Funding
No Funders.
Author information
Authors and Affiliations
Contributions
All authors contributed to the conception and the main idea of the work. MJA, DB, SC, AM, WS, NS, HSA, MNS, AZ, AM and ATJ drafted the main text, figures, and tables. MD supervised the work and provided the comments and additional scientific information. DB, MD and MJA also reviewed and revised the text. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
There is no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Ansari, M.J., Bokov, D., Markov, A. et al. Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell Commun Signal 20, 49 (2022). https://doi.org/10.1186/s12964-022-00838-y
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s12964-022-00838-y