Next Article in Journal
Red Blood Cell-Associated Features of Adenoviral Vector-Linked Venous Thrombosis
Previous Article in Journal
Small-Molecule Detection in Biological Fluids: The Emerging Role of Potentiometric Biosensors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Current Role of Antiangiogenics in Colorectal Cancer

1
Unit of Medical Oncology, San Giovanni Di Dio Hospital-Crotone, 88900 Crotone, Italy
2
Department of Medical Oncology, Centro di Riferimento Oncologico (CRO), IRCCS, 33081 Aviano, Italy
3
Unit of Medical Oncology, Giovanni Paolo II Hospital–Lamezia, ASP Catanzaro, 88046 Lamezia Terme, Italy
4
Department of Oncology, Mater Salutis Hospital, Az. ULSS 9 Scaligera, 37045 Legnago, Italy
5
Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(23), 11605; https://doi.org/10.3390/ijms262311605
Submission received: 16 October 2025 / Revised: 14 November 2025 / Accepted: 20 November 2025 / Published: 29 November 2025

Abstract

Colorectal carcinoma (CRC) represents the third most common cancer worldwide. Approximately 20% of patients present with metastatic disease at diagnosis, and 30–50% experience disease recurrence over time. For metastatic CRC (mCRC), the standard treatment consists of chemotherapy combined with a targeted agent based on molecular profile, such as RAS, BRAF, and MSI status. Anti-angiogenic drugs, which inhibit the formation of new blood vessels, have an established role in the management of mCRC. Mounting evidence highlights the critical interplay among angiogenesis, hypoxia, and the immune response in tumor progression. These insights have paved the way for testing novel combinations and molecules to control cancer progression. In particular, combining anti-angiogenic agents with immune checkpoint inhibitors has shown promise in improving outcomes for mCRC patients. Among emerging therapies, the novel anti-angiogenic agent fruquintinib has recently demonstrated clinical efficacy in the treatment of mCRC. Based on the data discussed in the present narrative review, the therapeutic landscape of mCRC appears poised for significant evolution in the near future. While numerous challenges and unanswered questions remain, the emergence of innovative therapeutic combinations and agents provides a promising opportunity for improving patient outcomes in mCRC.

1. Introduction

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of death by cancer worldwide [1]. For unresectable, advanced, or metastatic CRC (mCRC), the backbone of any therapeutic strategy is often represented by systemic treatment—chemotherapy associated with a targeted agent. There are multiple options for first-line therapy, depending on the biological molecular profile of the cancer (such as RAS, BRAF, and MSI status) and the patient’s individual characteristics.
Due to their increased metabolism, cancer cells require an increased availability of nutrients and oxygen, which is dependent on the generation of new blood vessels (angiogenesis); solid tumors have, therefore, acquired the ability to induce angiogenesis (angiogenic switch); among other mechanisms key to this process is the increased secretion of pro-angiogenic factors, such as VEGFs [2,3]. Hence, the development of angiogenic-targeting drugs has been increasingly relevant in the past few decades.
The first available anti-angiogenetic drug was bevacizumab, which inhibits the activation of VEGF signaling pathways through binding VEGF-A. In 2004, Hurwitz et al. demonstrated survival improvement by adding bevacizumab to FOLFIRI in the treatment of mCRC; hence, bevacizumab was approved for the treatment of mCRC in 2004 and 2005 in the United States (US) and the European Union (EU), respectively [4]. In August 2012, following the results from the VELOUR trial, the FDA approved ziv-aflibercept; this is a humanized recombinant fusion protein that functions as a VEGF inhibitor [5]. In September of the same year, regorafenib, a multi-kinase inhibitor that also has antiangiogenetic activity, was approved for advanced mCRC [6]. In 2013, the EMA also granted approval first for ziv-aflibercept and then for regorafenib. Lastly, in April 2014, ramucirumab, a recombinant human monoclonal IgG1 antibody that binds to the VEGF-R2, was also approved by the FDA (and by the EMA in December of the same year), based on the results of the RAISE trial [7].
Since then, no new antiangiogenetic drug has been approved, possibly owing to immunotherapy’s rise in prominence over the last few years. Still, the relevance of antiangiogenetic drugs in current clinical practice for the treatment of mCRC is undeniable; in this review, we will try to summarize the role of angiogenesis in pathophysiology, the clinical role of antiangiogenetic drugs, and potential developments for the future in mCRC.

2. From Benchside to Bedside

2.1. Role of Angiogenesis in Pathophysiology of CRC

The term “angiogenesis” was first proposed by Folkman over 50 years ago [8]. Angiogenesis is the formation of new capillaries out of pre-existing blood vessels under the regulation of growth and inhibitor factors [9]. This process is a multistep mechanism involving the interplay between many biological components, such as several cell types such as endothelial cells, tumor cells, stromal cells, immune-infiltrating cells (e.g., neutrophils and tumor-associated macrophages, “TAMs”), soluble angiogenic factors, and extracellular matrix (ECM) components [10,11,12]. Angiogenesis is intricate and primarily consists of four distinct consequential steps: (I) degradation of basement membrane glycoproteins and other components of the ECM surrounding the blood vessels by proteolytic enzymes; (II) endothelial cell (EC) activation and migration; (III) EC proliferation; and (IV) ECs transforming into tube-like structures, forming capillary tubes, and developing into novel basement membranes [12]. This process occurs in many physiological conditions through a balance between pro- and anti-angiogenic factors, as well as between multiple signal transduction pathways [9]; however, aberrant angiogenesis is a key process in cancer growth, invasion, and metastasis [12].
Neo-angiogenesis is primarily involved in CRC tumorogenesis, with two main regulators, hypoxia factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). HIF-1α is secreted by the cancer cell under hypoxic conditions and synergically acts with other pro-angiogenic molecules, such as VEGF, placental growth factor (PIGF), or angiopoietins. This factor affects a great variety of signal transduction pathways, including the up-regulation of the VEGF signaling cascade [11,13]. VEGF family includes a group of glycoproteins (VEGF-A, VEGF-B, VEGF-C, and VEGF-D) that, together with PlGF, interact with three VEGF receptors (VEGFR-1, -2, and -3) and two neuropilin co-receptors (NRP1, NRP2). VEGFA is produced by most cells in the body, but is up-regulated in hypoxic conditions [14]. In tumors, VEGF is produced by hypoxic tumor cells, ECs, and infiltrating myeloid cells, also known as the aforementioned TAMs [14,15].
The VEGF-A gene consists of eight exons on chromosome 6, with splice variants forming different isoforms, among which VEGFA165 is the most biologically active [16]. Most targeted cancer therapies act by inhibiting VEGF-A splice isoforms that promote microvessel growth, which is responsible for the most advanced and aggressive forms of the disease. Notably, the VEGF-A isoform balance, which is controlled by messenger ribonucleic acid (mRNA) splicing, coordinates angiogenesis [16].
VEGF-B and VEGF-A share the same architecture and activity, with a key role in carcinogenesis and blood vessels persistence, rather than an “angiogenic” factor, under stress conditions [17].
The VEGF pathway is up-regulated by several growth factors, including epidermal growth factor (EGF), platelet-derived growth factors (PDGFs), hepatocyte growth factor (HGF), and other cytokines [18].
VEGFRs are tyrosine kinase receptors (RTKs) found primarily in vascular endothelial cells [19]. The binding of the glycoprotein to its receptor results in the initiation of a sequence of events that ultimately leads to the formation of new vessels. The ligation of VEGF-A with VEGFR-2 is the most important step in the activation of angiogenesis in CRC [11,20]. This binding triggers multiple signaling networks that result in endothelial cell survival, migration, mitogenesis, differentiation, vascular permeability and vascular inflammation, vasodilatation, alteration of gene expression, and activation of the Ras pathway [9,14,18,20].
The role of VEGFR-1 is more complex and not fully understood. A soluble form of VEGFR-1 can prevent the binding between VEGF-A and VEGFR-2, which, in turn, inhibits the activation of the downstream pathway. However, VEGFR-1 is also involved in tumor-associated angiogenesis [21]. The third receptor, VEGFR-3, is involved in lymphangiogenesis, binding VEGF-C and VEGF-D [20]. Regarding VEGF-D expression, it has been associated with regional lymph node metastasis [9].
Likewise, PlGF acts by regulating endothelial and mural cell proliferation, as well as by engaging other pro-angiogenic cells and molecules. Moreover, it has a structural homology with VEGF-A, and thus, it interacts with VEGFR-1 downstream signaling [9]. This binding unleashes TAMs into the tumor bed, where they exert their immune-suppressive and pro-angiogenic function [22].
Microvascular density (MVD) is another important indicator used as a surrogate marker of tumoral angiogenesis and has been proposed to identify patients at a high risk of recurrence. MVD assessment is the most used technique to quantify the degree of neovascularization of the tumor [11,23]. MVD appears to increase during the evolutionary events in the sequence from normal mucosa to adenoma and from adenoma to cancer in CRC patients [24]. Since MVD is a biomarker for the quantification of angiogenesis, the question arises whether it can be used as a predictor biomarker for treatment with the antiangiogenic agents [25,26].

2.2. The Crosstalk Between Angiogenesis and Immune System

Tumor-associated blood vessels show an abnormal structure and a dysfunctional vascular network that modulate the expression of pro-inflammatory and co-stimulatory molecules; thus, they contribute to the permeability of immune-suppressing cells. Hence, cancer cells, through the overexpression of VEGFA, acquire the ability to affect, in turn, the immune and blood texture, shifting the tumor microenvironment (TME) towards an immune-suppressed microenvironment [11,27]. Within TME, immune and endothelial cells interact continuously with each other in a tight and mutual crosstalk, developing a dynamic context. Immune cells directly affect the phenotypes and functions of cancer vessels through various cytokines [28].
Innate immune cells, such as mature dendritic cells and M1-tumor-associated macrophages (TAM), produce cytokines (IFN-α, IL-12, IL-18, or TNF-α) and chemokines (CXCL9, CXCL10, or CCL21) that suppress tumor angiogenesis. Meanwhile, adaptive immune cells, such as CD8+ T cells and T helper 1 cells (TH1), secrete IFN-γ, a potent cytokine that inhibits angiogenesis and induces vascular normalization in TME [28].
The increased production of VEGF inhibits differentiation and maturation of monocytes into dendritic cells (DCs), impacting the antigen presentation, inhibiting nuclear factor (NF-kB), up-regulating programmed death-ligand 1 (PD-L1) on DCs, and, finally, inducing T cell suppression. Namely, VEGF inhibits progenitor cell differentiation into CD4+ and CD8+ lymphocytes, T cells’ proliferation, and increases T cells’ exhaustion by up-regulating PD-L1, cytotoxic T-lymphocytes associated protein 4 (CTLA4), TIM3, and LAG3 in T lymphocytes [29,30,31].
Neo-angiogenesis enhances the intra-tumoral pressure and decreases the endothelial intracellular adhesion molecule-1, such as the vascular cell adhesion molecule (VCAM-1) [32,33,34]. Likewise, hypoxia and acidosis induce the up-regulation of immune-suppressive chemokines that attract regulatory T cells (Treg), myeloid-derived suppressor cells (MDSCs) at the tumor site, polarize macrophages towards the M2-like phenotype, and reduce the extravasation of tumor-infiltrating cells (TILs). Moreover, pro-tumorigenic triggers are enhanced by the expression of FasL on tumor endothelial cells, which in turn, inhibits the proliferation of TCD8+ effector cells’ bed, allowing tumor spreading and resistance to anti-cancer treatment [35,36,37].
The use of anti-angiogenetic treatments reverses the immunosuppressive effect created by VEGFA. In fact, bevacizumab can normalize blood vessels restoring DCs maturation and reducing Treg recruitment in CRC cells and MDSCs in renal cell carcinoma [38,39,40,41] (Figure 1).

3. Current Landscape of Anti-Angiogenetic Treatments in Metastatic CRC

3.1. Anti-Angiogenetic in First-Line Therapy

Anti-angiogenetic agents have an established role in the treatment of mCRC. In 2004, the phase 3 AVF2107g study proved that the addition of bevacizumab to backbone chemotherapy led to an improvement in both overall survival (OS) (20.3 vs. 15.6 months, HR 0.66; p < 0.001) and progression-free survival (PFS) (10.6 vs. 6.2 months, HR 0.66; p < 0.001) [4]; thus, bevacizumab was approved as the first targeted therapy for patients with mCRC. A subsequent systematic review and pooled analysis of 29 clinical trials investigating the efficacy of the combination FOLFIRI-bevacizumab as first-line treatment yielded similar results, with a RR of 51.4% (22 publications), a median PFS (25 publications) of 10.8 months (95% C.I. 8.9–12.8), and a median OS (20 publications) of 23.7 months (95% C.I., 18.1–31.6) [42].
Adding bevacizumab to fluorouracil/leucovorin, Folfox, and Xelox as first-line therapy proved similarly effective [43,44,45,46]. Kabbinavar et al. randomly assigned 104 previously untreated mCRC patients to fluorouracil/leucovorin alone or associated with bevacizumab at 5 mg/kg or 10 mg/kg [44]. The combination treatment with bevacizumab resulted in better response rates (RR) (17% in the control arm vs. 40% and 8% in the two bevacizumab arms, low and high doses), with longer mPFS (5.2 months vs. 9.0 and 7.2 months) and mOS (13.8 vs. 21.5 and 16.1 months, respectively) [44]. A subsequent trial comparing FU/LV plus bevacizumab/placebo and a combined analysis of three clinical studies [4,5] validated these data. The BECOME study assessed the effects of the addition of bevacizumab to mFOLFOX6 as a first-line treatment of the RAS mutant mCRC with unresectable liver metastases; a significant benefit was demonstrated for mOS (25.7 vs. 20.5 months; p = 0.03), mPFS (9.5 vs. 5.6 months; p = 0.01), ORR (54.5% vs. 36.7%; p < 0.01), and R0 resection rates for liver metastases (22.3% vs. 5.8%, p < 0.01) [45]. In a different randomized phase III trial, bevacizumab/placebo was evaluated associated with either FOLFOX4 or XELOX; mPFS was 9.4 months in the bevacizumab group and 8.0 months in the placebo group (HR 0.83; 97.5% C.I. 0.72–0.95; p = 0.0023), and median OS was 21.3 vs. 19.9 months (HR 0.89; 97.5% C.I., 0.76 to 1.03; p = 0.077); RR was similar across groups of treatment [46]. Different meta-analyses have, since then, confirmed a benefit in RR, PFS, and OS by adding bevacizumab to a combination chemotherapy [47,48,49,50,51,52,53].
With the approval, over the years, of different targeted therapies (most commonly anti-EGFR antibodies for RAS wild-type cancers), the need to define the optimal strategy led to different trials; the FIRE-3 trial is well known, which compared FOLFIRI-bevacizumab with FOLFIRI-cetuximab in the first-line setting for RAS wild-type patients, demonstrating a similar mPFS (10.0 vs. 10.3 months in the cetuximab and in the bevacizumab group, respectively; HR 1.06, 95% C.I. 0.88–1.26; p = 0.55) but a mOS in favor of the cetuximab group (28.7 vs. 25.0 months, HR 0.77, 95% C.I. 0.62–0.96; p = 0.017) [54]. The Calgb/Swog 80405 trial compared either FOLFOX or FOLFIRI plus bevacizumab or cetuximab, with a subsequent amendment to include only RAS wild-type patients; it showed an equivalence between regimens in terms of OS (29.0 vs. 30.0 months for the chemotherapy/bevacizumab and chemotherapy/cetuximab combinations, respectively; HR 0.88; 95% C.I., 0.77–1.01; p = 0.08) and PFS (10.6 vs. 10.5 months, respectively) [55]. The PEAK trial compared the addition of either panitumumab or bevacizumab in a population of patients with KRAS exon 2 wild-type cancers; PFS was similar, and OS was improved (41.3 vs. 28.9 months, HR, 0.63; 95% C.I., 0.39 to 1.02; p = 0.058) in the panitumumab compared to the bevacizumab arm [56]. The PARADIGM trial compared mFOLFOX6 with either panitumumab to bevacizumab in a KRAS/NRAS wild-type population; the trial is currently completed and pending results [57].
Moreover, bevacizumab has shown efficacy in association with a non-fluoropyrimidine-based chemotherapy regimen, such as in the phase III trial TRICOLORE, which compared an association of S-1 and irinotecan plus bevacizumab to a standard oxaliplatin-based chemotherapy regimen (mFOLFOX6 or CAPOX) associated with bevacizumab; results of the trial showed the non-inferiority of the trial regimen in regard to PFS (10.8 months in the control group and 14.0 months in the experimental group; HR 0.84, 95% C.I. 0.70–1.02; p < 0.0001 for non-inferiority, and p = 0.0815 for superiority) [58].
Bevacizumab has also been proven effective in association with the chemotherapy triplet FOLFOXIRI, in particular for the BRAF-mutant population [59,60,61]. In the phase II OLIVIA study, the triplet chemotherapy associated with bevacizumab was found to induce better response rates (81% vs. 62%) and improved mPFS (18.6 vs. 11.5 months); the TRIBE study confirmed these results [59].
In the elderly population, the AVEX study showed that bevacizumab in association with capecitabine induces a significantly longer PFS than capecitabine alone (9.1 vs. 5.1 months; HR 0.53 [0.41–0.69]; p < 0.0001) [62].
Finally, Holch et al. demonstrated the relevance of primary tumor location; in right-sided mCRC, bevacizumab appears to be the targeting agent of choice [63]. Even in left-sided RAS wild-type cancers, a combination of a triplet with bevacizumab appears at least non-inferior to FOLFOX-panitumumab [64].
Other antiangiogenetic agents, such as aflibercept, have been investigated in the first-line setting; in the phase III trial AFFIRM, patients with mCRC were randomized to receive first-line therapy with mFOLFOX6 plus aflibercept (4 mg/kg) or mFOLFOX6 alone; however, the results showed no difference in PFS between treatment arms [65] (Table 1).

3.2. Maintenance

The role of maintenance therapy in CRC has been recognized, although, especially in RAS wild-type patients, there is still a lack of consensus regarding the optimal choice of targeted agent. The SAKK 41/06 trial explored the non-inferiority of continuation of single-agent bevacizumab versus no treatment, after a first-line chemotherapy combined with bevacizumab. The trial failed to demonstrate non-inferiority and showed increased treatment costs with no clinically meaningful benefit for the bevacizumab arm [66]. The PRODIGE-9 trial, similarly, explored bevacizumab maintenance versus no treatment in chemotherapy-free intervals after first-line induction treatment with FOLFIRI-bevacizumab; bevacizumab maintenance did not improve chemotherapy-free intervals, PFS, or OS [67].
More promising results were obtained with a strategy of de-escalation of treatment, rather than chemotherapy-free intervals; maintenance treatments with a combination of fluoropyrimidine and bevacizumab yielded more favorable results. The CAIRO-3 trial explored the combination of capecitabine and bevacizumab as maintenance after six cycles of treatment with a combination of capecitabine, oxaliplatin, and bevacizumab [68]. Patients in the treatment arm had a significant improvement of time to second progression, which was the primary endpoint of the study (11.4 months in the observation group vs. 13.9 months in the maintenance group; HR 0.63, 95% C.I. 0.52–0.76, p < 0.0001). The non-inferiority phase 3 trial AIO 0207 compared no treatment and treatment with bevacizumab alone to a maintenance with capecitabine and bevacizumab. Regarding the primary endpoint, which was time to failure of strategy, bevacizumab alone was non-inferior to standard fluoropyrimidine plus bevacizumab (HR 1.08, 95% C.I. 0.85–1.37; p = 0.53; upper limit of the one-sided 99.8% CI 1.42), whereas no treatment was not (HR 1.26, 95% C.I. 0.99–1.60, p = 0.056; upper limit of the one-sided 99.8% C.I. 1.65); to note, only 36% of patients received a re-induction as per protocol, thus limiting the clinical value of the chosen primary endpoint. However, in terms of PFS, pairwise comparison between treatment groups showed a significant improvement for the more active treatment group for each comparison, thus favoring the combination of capecitabine/bevacizumab [69] (Table 2).

3.3. Second Line

The E3200 trial is a phase III trial that explored the efficacy of bevacizumab as a second-line treatment in patients with previously treated mCRC; in the trial, patients who received a first-line treatment with fluoropyrimidines and irinotecan were randomized to treatment with either FOLFOX4 alone, FOLFOX4 in combination with bevacizumab, or bevacizumab alone [70]. The median OS was 12.9 months for the combination arm, compared with 10.8 months for the FOLFOX4 group and 10.2 months for those treated with bevacizumab alone. The median PFS was 7.3 months for the combination group, compared with 4.7 months for the FOLFOX4 group and 2.7 months for the bevacizumab group. Overall RR were, respectively, 22.7%, 8.6%, and 3.3% (p < 0.0001 for FOLFOX4/bevacizumab vs. FOLFOX4) [70].
Continuation of bevacizumab beyond progression has been investigated by different trials. The ML18147 trial assessed the efficacy of continuation of bevacizumab associated with a second-line chemotherapy after progression on a standard first-line of chemotherapy associated with bevacizumab [71]. mOS was 11.2 months for the bevacizumab arm, compared to 9.8 months for chemotherapy alone (HR 0.81, 95% C.I. 0.69–0.94; p = 0.0062) [71]. The BEBYP trial, which investigated the same therapeutic strategy [34], confirmed these findings, showing a significant improvement in PFS for the bevacizumab group (5.0 vs. 6.8 months, adjusted HR 0.70; 95% C.I. 0.52–0.95; stratified log-rank p = 0.010) and a trend toward an improved OS (adjusted HR 0.77; 95% C.I. 0.56–1.06; stratified log-rank p = 0.043); the study was prematurely stopped in consideration of the results of the ML18147 trial [72]. The EAGLE trial explored two different doses of bevacizumab (5 or 10 mg/kg) associated with FOLFIRI as second-line therapy after progression on a oxaliplatin/bevacizumab-based first line, finding no benefits associated with the use of a higher dose [73]. Analysis from the ARIES observational cohort confirmed the observed improvement in PFS (14.4 months vs. 10.6 months) [74].
The use of different antiangiogenetic agents has been explored by the VELOUR and the RAISE trials. The VELOUR phase III study examined the addition of aflibercept to a FOLFIRI regimen after progression in an oxaliplatin-based first-line treatment; the study allowed for bevacizumab as part of the first-line regimen [5]. The experimental arm demonstrated an improvement of mOS (13.5 vs. 12.06 months, HR 0.8; 95.34% C.I., 0.71–0.937; p = 0.0032), mPFS (6.9 vs. 4.67 months; HR, 0.758; 95% C.I. 0.66–0.86; p < 0.0001), and RR (19.8% vs. 11.1%, p = 0.0001); this benefit was consistent across subgroups, including the patients who received bevacizumab as part of their first-line therapy [5]. The RAISE trial explored the combination of FOLFIRI with either ramucirumab of placebo after a first-line treatment with bevacizumab, oxaliplatin, and a fluoropyrimidine; results demonstrated an improvement in mOS for the ramucirumab group over the placebo group (13.3 months vs. 11.7 months, HR 0.84, 95% C.I. 0.73–0.97; log-rank p = 0.0219) [7] (Table 3).

3.4. Beyond Second-Line Treatment

Use of antiangiogenetics beyond second-line treatment has been proven effective by several clinical trials. In a phase II study, the addition of bevacizumab to a standard treatment with TAS-102 in chemo-refractory patients induced an improvement in PFS (4.6 vs. 2.6 months; HR 0.45; p = 0.0015); these results were recently confirmed by the phase III SUNLIGHT trial, where the combination treatment showed an improvement of both mOS (10.8 vs. 7.5 months; HR, 0.61; 95% C.I., 0.49–0.77; p < 0.001) and mPFS (5.6 vs. 2.4 months; HR 0.44; 95% C.I., 0.36–0.54; p < 0.001) [75,76].
Regorafenib, a multi-kinase inhibitor, targets angiogenic (VEGFR-1-3, TIE2), stromal (PDGFR-β, FGFR), and oncogenic receptor tyrosine kinases (KIT, RET, and RAF). Following the results of the CORRECT trial, which showed an improvement in OS (6.4 vs. 5.0 months, HR 0.77; 95% C.I. 0.64–0.94; p = 0.0052) for treatment with regorafenib in chemo-resistant patients when compared to placebo, this drug was approved as a monotherapy for mCRC patients who progressed on previous lines of treatment [6]. Similar results were shown in the Asian population in the CONCUR trial (mOS 8.8 vs. 6.3 months; HR 0.55, 95% C.I. 0.40–0.77, p = 0.00016) [77] (Table 4).

4. Looking to the Future

4.1. New Treatment Goals in CRC

Mounting evidence in tumor biology have shown that understanding the various mechanisms behind tumor growth and progression requires a comprehensive investigation of the complex interactions between tumor cells and their TME in order to also use them as targets for active and effective therapies to be used in clinical practice.
In addition, TME harbors multiple actors that practically play a significant role in tumor initiation, progression, and metastatization as the genetic and epigenetic changes in cancer cells. As already written, there is an important connection between angiogenesis, hypoxia, and immune response. These premises have paved the way for testing new combinations and new molecules in order to control cancer progression.

4.2. Immunotherapy and Anti-Angiogenics

Over the last years, immunotherapy has reshaped the landscape in several tumors, both as a single agent and combined with chemotherapy, like melanoma, lung, and urothelial cancer, improving survival outcomes.
Based on these premises, many clinical trials have been designed to revert to the immune-suppressive microenvironment driven by vasculopathy, exploiting the interplay between angiogenesis and the immune system. In fact, the restoration of immune responsiveness induced by combining anti-angiogenic therapy with immunotherapy could reverse the TME balance towards an immune-activated system (Figure 1).
Current clinical practice mainly includes three types of immune checkpoint inhibitors (ICIs) targeting CTLA4, PD-1, or PD-L1 [78]. Unfortunately, these treatments as single agents did not achieve sufficient data in CRC, except for microsatellite instability (MSI) tumors [79].
Moreover, in more recent years, as the immune-suppressive TME is induced in part by the abnormal vasculature, the combination with antiangiogenetics has been tested in several tumors like NSCLC (atezolizumab plus bevacizumab), renal cell carcinoma (cabozantinib plus nivolumab or axitinib plus pembrolizumab), HCC (atezolizumab plus bevacizumab), and endometrial cancer (lenvatinib plus pembrolizumab), improving survival outcomes [80,81,82,83].
This broad efficacy has been the key premise for transferring this combination treatment also in CRC. Firstly, Bendel et al. in 2015 presented an improved objective tumor response among patients receiving bevacizumab with atezolizumab at the ASCO-GI conference [84]. Likewise, bevacizumab plus atezolizumab was added to chemotherapy as a first-line treatment administered to 23 CRC patients and showed a median PFS of 14.1 months [85].
The first large trials were presented by Grothey et al. Overall, 445 BRAF wild-type CRC patients included in the MODUL trial were randomly assigned to receive maintenance therapy with fluoropyrimidine and bevacizumab with or without atezolizumab after induction first-line treatment with FOLFOX plus bevacizumab. However, no differences between the two arms were reported in terms of PFS and OS [86,87]. Biomarker analyses are still ongoing.
Mettu et al. presented at ESMO 2019 results derived from the BACCI phase II trial conducted on 133 CRC patients treated at last-line with capecitabine plus bevacizumab and atezolizumab or placebo, meeting the primary endpoint; however, the benefit was only 1 month [88]. In the atezolizumab arm, mPFS was 4.4 months vs. 3.6 months in the control arm. An exploratory analysis observed a higher ORR in patients without liver disease (23.1% vs. 5.8%), accompanied by a better PFS and OS [88].
The updated results from the phase Ib trial REGONIVO provide an encouraging antitumor activity in the enrolled CRC patients receiving nivolumab 3 mg/kg every two weeks combined with regorafenib. Median PFS was 7.8 months, 1y-PFS was 41.7%, and 1y-OS was 68% [89]. These findings paved the way for other clinical trials investigating the combination of anti-VEGF with immune checkpoint blockade.
From 2018 to 2020, 218 CRC patients were randomized 1:2, in the phase II ATEZOTRIBE trial, to receive first-line FOLFOXIRI plus bevacizumab with or without atezolizumab, followed by maintenance treatment (fluorouracil and leucovorin plus bevacizumab with or without atezolizumab), irrespective of RAS and BRAF mutational status [90]. Median PFS was 13.1 months in the experimental arm compared to 11.5 months in the control one (HR 0.69, 80% C.I. 0.56–0.85, p = 0.012). OS data are still immature. The most, 3–4, adverse events were neutropenia (42% vs. 36% in the experimental and control group, respectively), diarrhea (15% vs. 13%), and febrile neutropenia (10% vs. 10%) [90]. This trial suggested that a synergic role is played by combining atezolizumab with first-line chemotherapy plus anti-VEGF. Additionally, in the translation exploratory analysis, patients with high tumor mutational burden (TMB) and high immunoscore demonstrated a longer PFS [90].
The CheckMate 9X8 investigated the combined first-line FOLFOX plus bevacizumab, added or not to nivolumab, regardless of mutational status [91]. The primary endpoint was not achieved. Median PFS was 11.9 months in both groups. However, authors described a higher ORR (60% vs. 46%) and prolonged duration of response (DOR) in patients treated with nivolumab compared to standard treatment. Similarly, the NIVACOR phase II single-arm study tested first-line FOLFOXIRI plus bevacizumab and nivolumab, followed by a maintenance strategy in RAS or BRAF mutant CRC. ORR was 76.7% in the whole population and 78.9% in the MSS mCRC patients, while median PFS was 9.8 months [92].
In the NCT03050814 phase II trial, patients with MSS mCRC were randomized to FOLFOX plus bevacizumab ± avelumab with a CEA-targeted vaccine. No positive results have been obtained in terms of PFS [93]. Furthermore, toripalimab (a PD-1 antibody) added to regorafenib showed a good response and OS in a phase 2 study [94].
Main clinical trials evaluating anti-angiogenic drugs associated with ICIs are reported in Table 5.

4.3. Targeting Angiogenesis Through a New Generation Molecule

In recent years, a new generation molecule, fruquintinib, highly selective for VEGFR-1, 2, and 3, has been developed based on the great antitumor activity demonstrated both in in vitro and in vivo models [95]. It exerts its properties by suppressing endothelial cell proliferation and tubule development in a dose-dependent manner. The phase III FRESCO trial randomly enrolled Chinese patients after two lines of therapy to receive fruquintinib at 5 mg administered on days 1–21 every 4 weeks plus BSC, or placebo plus BSC [96]. The experimental arm significantly improved the survival outcome, gaining a longer OS (9.3 vs. 6.6 months; HR 0.65, 95% C.I. 0.51–0.83, p < 0.001) and PFS (3.71 vs. 1.84 months; HR 0.26, 95% C.I. 0.21–0.34, p < 0.001) with an acceptable safety profile [96]. Based on these data, fruquintinb received its approval in China on 4 September 2018. However, given geographic differences between China and the Western population, a global phase III study has been developed: the FRESCO-2 trial [97].
Heavily pre-treated patients (at least two lines) were randomized 2:1 to receive fruquintinib plus BSC or placebo plus BSC. Consistently, the FRESCO-2 trial reached its primary and secondary endpoints, confirming the OS (7.4 vs. 4.8 months; HR 0.66, 95% C.I. 0.55–0.80, p < 0.001) and PFS (3.7 vs. 1.8 months; HR 0.32, 95% C.I. 0.27–0.39, p < 0.001) improvement.
The OS benefit has been demonstrated across all subgroups, including patients treated with TAS-102, regorafenib, or both (HR 0.60) [97].
Ongoing clinical trials are exploring the optimal strategy to use fruquintinib in mCRC patients, in first-line (NCT01975077), second-line (NCT05634590, NCT05555901, NCT05522738, and NCT05447715), or third-line (NCT05447715) combined with chemotherapy or with TAS-102 (NCT05004831). Further, in chemo-refractory patients, fruquintinb added to immunotherapy or raltitrexed is being tested (NCT04695470, NCT04582981, and NCT04866862) (Table 6).

5. Identification of Prognostic and Predictive Biomarkers During Anti-Angiogenic Treatment

The key role played by angiogenesis in tumor growth and spreading is well established. However, not all patients seem to benefit from anti-angiogenic treatment. Henceforth, the identification of prognostic and predictive biomarkers is necessary to better refine mCRC patients who will be more likely to benefit from these therapies.
Several potential biomarkers have been investigated as implicated in angiogenesis. First of all, circulating VEGFR analysis has been conducted in many trials with contrasting results. A subgroup of 59 patients enrolled in the BEBYP trial has been tested for VEGFR-2 levels, showing that in the group with high VEGFR-2 (>median value, 6.3 ng/mL), the prosecution of bevacizumab was associated with a better PFS (median PFS: 10.4 vs. 3.4 months; HR 0.24, 95% C.I. 0.10–0.58, p = 0.002). However, the benefit from the prosecution of bevacizumab was not reported in patients with low VEGFR-2 levels (mPFS 5.4 vs. 5.0; HR 0.98, 95% C.I. 0.45–2.11, p = 0.956) [98].
VEGF-D has been dosed in the MAX, CAIRO-2, and RAISE studies. In the first one, low expression of VEGF-D was associated with efficacy of bevacizumab-based therapy regarding PFS (HR 0.21, 95% C.I. 0.08–0.55) and OS (HR 0.35; 95% C.I. 0.13–0.90). Less efficacy was observed in the population with low VEGF-D levels. In CAIRO-2, no differences have been reported according to VEGF-D levels [99]. In the RAISE trial, ramucirumab added to chemotherapy resulted in more effective outcomes in the group with high VEGF-D expression, both for PFS and OS [100]. VEGF-A showed no predictive role in different retrospective and prospective studies, while VEGF-A splice isoform 165b and 121 seem to be a potential predictive biomarker of response to bevacizumab [101,102,103]. Preclinical studies observed an important role played by FGF-2 in restoring sensitivity during bevacizumab treatment. Giampieri et al. observed a significant association of high FGF2 expression with a longer PFS and OS, despite high FGF-2 levels at baseline representing a poor prognostic factor [104].
Further, preclinical studies observed a correlation between serum LDH levels with VEGF-A and VEGFR-1 expression [105]. High serum LDH levels seem to be associated with benefit from VEGF-A inhibition. However, the analysis conducted on the BEBYP trial revealed contrasting results; bevacizumab beyond progression was effective in the group with low LDH levels (HR: 0.39, 95% C.I.: 0.23–0.65), while no difference was reported in the group with high LDH levels HR: 1.10, (95% C.I.: 0.74–1.64) [106,107,108].
Many biomarkers have been identified as predictive factors during anti-angiogenic treatment, such as the transcription factor homeobox 9, hepatocyte growth factor, and markers of vascular immaturity [100,103,109,110,111]. The loss of chromosome 18q11.2–q12 [112] has been studied in a non-randomized trial and, then, in a post hoc analysis of the 256 AGIT-MAX trial. This loss was detected in 71% of mCRC patients and was associated with a bevacizumab benefit in terms of PFS (p = 0.009) and not in patients without (p = 0.67). However, a statistical significance for marker–treatment interaction was not yielded (p = 0.28) [113]. Moreover, several miRNAs have been tested to demonstrate their association with outcomes during anti-angiogenic treatment. High miR-664-3p levels were significantly associated with better survival in mCRC patients treated with bevacizumab compared to the placebo group [114]. Hamaguchi et al. conducted an exploratory analysis evaluating 78 potential prognostic and predictive biomarkers in 62 Japanese patients receiving aflibercept plus chemotherapy. Among all biomarkers, low levels of an extracellular newly identified receptor for advanced glycation end-products binding protein (EN-RAGE), tissue inhibitor of metalloproteinases 1 (TIMP-1), and interleukin-8 (IL-8) allowed for a beneficial gain in terms of OS [115,116]. Nonetheless, these biomarkers have not demonstrated sufficient prognostic and predictive data to warrant their translation into current clinical practice, even if promising. Hence, further prospective analyses should be conducted to better identify a class of patients that could benefit from any anti-angiogenic treatment.

6. Cost-Effectiveness of Anti-Angiogenic Treatment

Due to the lack of predictive biomarkers, the antiangiogenic-based treatments are broadly used, at least once, during the treatment history of mCRC patients. Hence, since it is administered in an unselected population and only some fraction of patients will really benefit from it with a considerable economic burden, a cost-effectiveness balance is mandatory.
The high variability of economic policies and pricing in individual countries is relevant for the cost-effectiveness analysis. However, the addition of anti-angiogenic therapy in mCRC is not cost-effective in most countries. In the analysis conducted by Goldstein et al. in 2017, the addition of bevacizumab to first-line chemotherapy in mCRC consistently failed to be cost-effective in all five countries (the U.S., the U.K., Australia, Canada, and Israel) evaluated, with the highest incremental cost-effectiveness ratio for the U.S. (USD 571,000 per quality-adjusted life years) [117]. Similar results are reported for bevacizumab in the maintenance strategy. Moreover, the introduction of anti-angiogenic drugs in second-line treatment was linked with a significant increase in costs. An analysis conducted on four randomized clinical trials, including 3938 mCRC patients treated with anti-angiogenic second-line therapy, combined the pharmacological costs of the drug with the benefit measured according to PFS. FOLFIRI plus aflibercept was the most cost-effective treatment with the lowest cost per month of PFS (EUR 4581) [118]. Further analyses will need to be conducted to establish the full balance between costs and benefits deriving from the newly developed molecule and, particularly, from the combination of anti-angiogenic therapies with immunotherapy.

7. Expert Opinion and Future Directions

Angiogenesis has long been recognized as a fundamental hallmark of cancer progression, especially in mCRC. Since the first approval in 2004 of bevacizumab, with encouraging results, several anti-angiogenic agents have been developed for the treatment of mCRC in the last few decades: ziv-aflibercept and regorafenib in 2012, and ramucirumab in 2014. Although most pivotal trials on antiangiogenic therapy in mCRC were conducted before 2018, they remain the key evidence supporting current treatment strategies. More recent studies have focused on biomarker identification, treatment sequencing, and combinations with immunotherapy, rather than introducing new antiangiogenic agents. More recently, a new molecule that is highly selective for VEGFR-1, 2, and 3, fruquintinb, showed its efficacy in improving mCRC prognosis. It exerts its properties by reducing endothelial cell proliferation and tubule growth. The FRESCO-2 trial showed the PFS and OS benefit in heavily pre-treated CRC patients.
Interestingly, the abnormal architecture of tumor blood vessels enhances the intra-tumoral pressure and decreases the endothelial intracellular adhesion molecules. Likewise, hypoxia and acidosis induce the up-regulation of immune-suppressive chemokines that recruit several immune-suppressive cytokines, molecules, and cells, such as Treg and MDSCs, into the tumor bed, polarize macrophages towards the M2-like phenotype, and reduce the extravasation of TILs. The use of anti-angiogenetic agents could reverse the immunosuppressive microenvironment. Thus, as the abnormal vessels can affect TME, combination treatments based on antiangiogenics and immune checkpoint inhibitors have also been tested in mCRC. In fact, the first clinical data with immunotherapy only became available in 2015, with initial limitation to MSI-high mCRC. Mounting data have recently demonstrated the synergic role played by the combination of immunotherapy with anti-VEGF.
According to the data discussed in the present narrative review, the therapeutic landscape of mCRC is likely to change in the near future.
However, it raises several challenges and some open questions.
The first is “positioning”: What will be the optimal strategy in the treatment of mCRC? Many molecules and combination treatments, in addition to anti-angiogenic drugs, are being tested in the mCRC treatment. Furthermore, ongoing clinical studies are evaluating the best strategy for the use of fruquintinib in mCRC patients, in first-, second-, or third-line combined with chemotherapy or immunotherapy. Moreover, many clinical trials are evaluating the best treatment combinations between anti-angiogenic drugs and immune checkpoint inhibitors. So, how will these therapy options fit into future mCRC scenarios?
Secondly, how do we identify patients who can benefit from anti-angiogenic agents?
Many doubts persist regarding patient selection, optimal sequencing, and the integration of antiangiogenics within a modern treatment algorithm that increasingly emphasizes molecular stratification and immunologic modulation.
In fact, not all CRC patients benefit from treatment with angiogenic inhibitors alone or combined with immunotherapy. However, none of the biomarkers tested (VEGFR-2, VEGF-D, FGF-2, LDH, EN-RAGE, TIMP-1, IL-8, transcription factor homeobox 9, the loss of chromosome 18q11.2–q12, VEGFR-1, and VEGF-A) have demonstrated sufficient prognostic and predictive information to warrant their use in current clinical practice. Hence, further prospective analyses should be conducted to better identify how, when, and the class of mCRC patients that could more likely benefit from these therapies.
Thirdly, what is the balance between cost and efficacy of the new proposed strategy? Different cost-effective analyses have been conducted regarding the use of bevacizumab and aflibercept. However, further analyses will need to be conducted to establish the full balance between costs and benefits deriving from this newly developed molecule and, particularly, from the combination of anti-angiogenic therapies with immunotherapy.
Antiangiogenic therapy remains an important player in mCRC treatment. However, to preserve its role in the evolving treatment landscape, a paradigm shift is needed. Combinations with immunotherapy seem to be a great opportunity for mCRC, but understanding the interplay of the tumor immune microenvironment and angiogenic dynamics is necessary to better define responsive patients. Despite extensive available data, no predictive biomarkers have yet demonstrated sufficient reliability or validation for routine clinical application in mCRC patients treated with antiangiogenic agents. Hence, ongoing research should prioritize biomarker discovery, rational combinations, and patient stratification to optimize the clinical impact and economic viability of antiangiogenic agents in mCRC.

Author Contributions

D.B.: Conceptualization, writing—original draft preparation, and supervision. P.D.N.: Methodology, data curation, and writing—review and editing. M.G.D.: Literature review and writing—original draft preparation. C.C.: Visualization and review of clinical trials. J.G.: Methodological support, original draft preparation, and critical revision. G.A.: Conceptualization, supervision, writing—review and editing, and final approval of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

BRAFv-Raf murine sarcoma viral oncogene homolog B
BSCBest supportive care
CEACarcinoembryonic antigen
CIConfidence interval
CRCColorectal cancer
CTLA-4Cytotoxic T-lymphocyte–associated protein 4
CXCL9/10C-X-C motif chemokine ligand 9/10
DCsDendritic cells
DORDuration of response
ECMExtracellular matrix
ECsEndothelial cells
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
EMAEuropean Medicines Agency
EN-RAGEExtracellular newly identified receptor for advanced glycation end-products binding protein
ESMOEuropean Society for Medical Oncology
FDAU.S. Food and Drug Administration
FGF-2Fibroblast growth factor-2
FGFRFibroblast growth factor receptor
FOLFIRI5-Fluorouracil + leucovorin + irinotecan
FOLFOX5-Fluorouracil + leucovorin + oxaliplatin
FOLFOXIRI5-Fluorouracil + leucovorin + oxaliplatin + irinotecan
FU/LV5-Fluorouracil/leucovorin
HCCHepatocellular carcinoma
HGFHepatocyte growth factor
HIF-1αHypoxia-inducible factor-1 alpha
HRHazard ratio
ICI(s)Immune checkpoint inhibitor(s)
IL-8/IL-12/IL-18Interleukin-8/-12/-18
INF-α/IFN-γInterferon-alpha/interferon-gamma
KD(not used)
KITKIT proto-oncogene receptor tyrosine kinase
KRAS/NRASKirsten rat sarcoma/neuroblastoma RAS viral oncogene homolog
LAG-3Lymphocyte activation gene-3
LDHLactate Dehydrogenase
mCRCMetastatic colorectal cancer
mFOLFOX6Modified FOLFOX6 regimen
mOSMedian overall survival
mPFSMedian progression-free survival
MDSCsMyeloid-derived suppressor cells
MVDMicrovascular density
MSSMicrosatellite atable
MSI/MSI-HMicrosatellite instability/microsatellite instability-high
mRNAMessenger ribonucleic acid
NRP1/NRP2Neuropilin-1/neuropilin-2
NSCLCNon–small-cell lung cancer
ORROverall response rate
OSOverall survival
PD-1Programmed cell death-1
PD-L1Programmed death-ligand 1
PDGF(s)Platelet-derived growth factor(s)
PDGFR-βPlatelet-derived growth factor receptor-beta
PFSProgression-free survival
PlGFPlacental growth factor
RAFRapidly accelerated fibrosarcoma kinase family
RASRat sarcoma viral oncogene family (KRAS/NRAS)
R0Microscopically margin-negative resection
RETRET proto-oncogene
RRResponse rate
RTKsReceptor tyrosine kinases
S-1Tegafur/gimeracil/oteracil (oral fluoropyrimidine)
TAM(s)Tumor-associated macrophage(s)
TAS-102Trifluridine/Tipiracil
TCE(not used)
TIE2TEK tyrosine kinase endothelial receptor 2
TILsTumor-infiltrating lymphocytes
TIMP-1Tissue inhibitor of metalloproteinases-1
TMBTumor mutational burden
TMETumor microenvironment
TNF-αTumor necrosis factor-alpha
TregRegulatory T cell
USUnited States
VCAM-1Vascular cell adhesion molecule-1
VEGFVascular endothelial growth factor
VEGF-A/-B/-C/-DVEGF isoforms A/B/C/D
VEGFR-1/-2/-3Vascular endothelial growth factor receptor-1/-2/-3
XELOX/CAPOXCapecitabine + oxaliplatin

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Carmeliet, P. Angiogenesis in health and disease. Nat. Med. Nat. Publ. Group 2003, 9, 653–660. [Google Scholar] [CrossRef] [PubMed]
  3. Dvorak, H.F. Vascular permeability factor/vascular endothelial growth factor: A critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J. Clin. Oncol. 2002, 20, 4368–4380. [Google Scholar] [CrossRef] [PubMed]
  4. Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E.; et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef] [PubMed]
  5. Van Cutsem, E.; Tabernero, J.; Lakomy, R.; Prenen, H.; Prausová, J.; Macarulla, T.; Ruff, P.; Van Hazel, A.; Moiseyenko, V.; Ferry, D.; et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J. Clin. Oncol. 2012, 30, 3499–3506. [Google Scholar] [CrossRef]
  6. Grothey, A.; Van Cutsem, E.; Sobrero, A.; Siena, S.; Falcone, A.; Ychou, M.; Humblet, Y.; Bouché, O.; Mineur, L.; Barone, C.; et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): An international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013, 381, 303–312. [Google Scholar] [CrossRef]
  7. Tabernero, J.; Takayuki, Y.; Cohn, A.L. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): A randomised, double-blind, multicentre, phase 3 study. Lancet Oncol. 2015, 16, e262, Erratum in Lancet Oncol. 2015, 16, 499–508. [Google Scholar]
  8. Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar]
  9. Giordano, G.; Febbraro, A.; Venditti, M.; Campidoglio, S.; Olivieri, N.; Raieta, K.; Parcesepe, P.; Imbriani, G.C.; Remo, A.; Pancione, M. Targeting angiogenesis and tumor microenvironment in metastatic colorectal cancer: Role of aflibercept. Gastroenterol. Res. Pract. 2014, 2014, 526178. [Google Scholar] [CrossRef]
  10. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef]
  11. Kampoli, K.; Foukas, P.G.; Ntavatzikos, A.; Arkadopoulos, N.; Koumarianou, A. Interrogating the interplay of angiogenesis and immunity in metastatic colorectal cancer. World J. Methodol. 2022, 12, 43–53. [Google Scholar] [CrossRef]
  12. Li, T.; Kang, G.; Wang, T.; Huang, H. Tumor angiogenesis and anti-angiogenic gene therapy for cancer. Oncol. Lett. 2018, 16, 687–702. [Google Scholar]
  13. Zimna, A.; Kurpisz, M. Hypoxia-Inducible Factor-1 in Physiological and Pathophysiological Angiogenesis: Applications and Therapies. Biomed. Res. Int. 2015, 2015, 549412. [Google Scholar] [CrossRef]
  14. Claesson-Welsh, L.; Welsh, M. VEGFA and tumour angiogenesis. J. Intern. Med. 2013, 273, 114–127. [Google Scholar] [CrossRef]
  15. Wang, H.; Tian, T.; Zhang, J. Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): From Mechanism to Therapy and Prognosis. Int. J. Mol. Sci. 2021, 22, 8470. [Google Scholar] [CrossRef]
  16. Harper, S.J.; Bates, D.O. VEGF-A splicing: The key to anti-angiogenic therapeutics? Nat. Rev. Cancer 2008, 8, 880–887. [Google Scholar] [CrossRef] [PubMed]
  17. Zheng, L.; Zhao, C.; Du, Y.; Lin, X.; Jiang, Y.; Lee, C.; Tian, G.; Mi, J.; Li, X.; Chen, Q.; et al. PDGF-CC underlies resistance to VEGF-A inhibition and combinatorial targeting of both suppresses pathologyical angiogenesis more efficiently. Oncotarget 2016, 7, 77902–77915. [Google Scholar] [CrossRef] [PubMed]
  18. Lopez, A.; Harada, K.; Vasilakopoulou, M.; Shanbhag, N.; Ajani, J.A. Targeting Angiogenesis in Colorectal Carcinoma. Drugs 2019, 79, 63–74. [Google Scholar] [CrossRef] [PubMed]
  19. Yancopoulos, G.D.; Davis, S.; Gale, N.W.; Rudge, J.S.; Wiegand, S.J.; Holash, J. Vascular-specific growth factors and blood vessel formation. Nature 2000, 407, 242–248. [Google Scholar] [CrossRef]
  20. Mousa, L.; Salem, M.E.; Mikhail, S. Biomarkers of Angiogenesis in Colorectal Cancer. Biomark. Cancer 2015, 7, 13–19. [Google Scholar]
  21. Ferrara, N.; Gerber, H.-P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef] [PubMed]
  22. Park, J.E.; Chen, H.H.; Winer, J.; Houck, K.A.; Ferrara, N. Placenta growth factor. Potentiation of vascular endothelial growth factor bioactivity, in vitro and in vivo, and high affinity binding to Flt-1 but not to Flk-1/KDR. J. Biol. Chem. 1994, 269, 25646–25654. [Google Scholar] [CrossRef] [PubMed]
  23. Des Guetz, G.; Uzzan, B.; Nicolas, P.; Cucherat, M.; Morere, J.-F.; Benamouzig, R.; Breau, J.L.; Perret, G.Y. Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br. J. Cancer 2006, 94, 1823–1832. [Google Scholar] [CrossRef]
  24. Hasan, J.; Byers, R.; Jayson, G.C. Intra-tumoural microvessel density in human solid tumours. Br. J. Cancer 2002, 86, 1566–1577. [Google Scholar] [CrossRef]
  25. Bais, C.; Mueller, B.; Brady, M.F.; Mannel, R.S.; Burger, R.A.; Wei, W.; Marien, K.M.; Kockx, M.M.; Husain, A.; Birrer, M.J. Tumor Microvessel Density as a Potential Predictive Marker for Bevacizumab Benefit: GOG-0218 Biomarker Analyses. J. Natl. Cancer Inst. 2017, 109, djx066. [Google Scholar] [CrossRef]
  26. Jubb, A.M.; Hurwitz, H.I.; Bai, W.; Holmgren, E.B.; Tobin, P.; Guerrero, A.S.; Kabbinavar, F.; Holden, S.N.; Novotny, W.F.; Frantz, G.D.; et al. Impact of vascular endothelial growth factor-A expression, thrombospondin-2 expression, and microvessel density on the treatment effect of bevacizumab in metastatic colorectal cancer. J. Clin. Oncol. 2006, 24, 217–227. [Google Scholar] [CrossRef]
  27. Kim, H.J.; Ji, Y.R.; Lee, Y.M. Crosstalk between angiogenesis and immune regulation in the tumor microenvironment. Arch. Pharm. Res. 2022, 45, 401–416. [Google Scholar] [CrossRef]
  28. Lee, W.S.; Yang, H.; Chon, H.J.; Kim, C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp. Mol. Med. Nat. Publ. Group 2020, 52, 1475–1485. [Google Scholar] [CrossRef]
  29. Curiel, T.J.; Wei, S.; Dong, H.; Alvarez, X.; Cheng, P.; Mottram, P.; Krzysiek, R.; Knutson, K.L.; Daniel, B.; Zimmermann, M.C.; et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 2003, 9, 562–567. [Google Scholar] [CrossRef]
  30. Oyama, T.; Ran, S.; Ishida, T.; Nadaf, S.; Kerr, L.; Carbone, D.P.; Gabrilovich, I. Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-kappa B activation in hemopoietic progenitor cells. J. Immunol. 1998, 160, 1224–1232. [Google Scholar] [CrossRef] [PubMed]
  31. Ohm, J.E.; Gabrilovich, D.I.; Sempowski, G.D.; Kisseleva, E.; Parman, K.S.; Nadaf, S.; Carbone, D.P. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood 2003, 101, 4878–4886. [Google Scholar] [CrossRef]
  32. Yi, M.; Jiao, D.; Qin, S.; Chu, Q.; Wu, K.; Li, A. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer 2019, 18, 60. [Google Scholar] [CrossRef] [PubMed]
  33. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef]
  34. Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.-P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef]
  35. Wada, J.; Suzuki, H.; Fuchino, R.; Yamasaki, A.; Nagai, S.; Yanai, K.; Koga, K.; Nakamura, M.; Tanaka, M.; Morisaki, T.; et al. The contribution of vascular endothelial growth factor to the induction of regulatory T-cells in malignant effusions. Anticancer Res. 2009, 29, 881–888. [Google Scholar] [PubMed]
  36. Huang, Y.; Chen, X.; Dikov, M.M.; Novitskiy, S.V.; Mosse, C.A.; Yang, L.; Carbone, D.P. Distinct roles of VEGFR-1 and VEGFR-2 in the aberrant hematopoiesis associated with elevated levels of VEGF. Blood 2007, 110, 624–631. [Google Scholar] [CrossRef] [PubMed]
  37. Varney, M.L.; Johansson, S.L.; Singh, R.K. Tumour-associated macrophage infiltration, neovascularization and aggressiveness in malignant melanoma: Role of monocyte chemotactic protein-1 and vascular endothelial growth factor-A. Melanoma Res. 2005, 15, 417–425. [Google Scholar] [CrossRef] [PubMed]
  38. Jain, R.K. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nat. Med. 2001, 7, 987–989. [Google Scholar] [CrossRef]
  39. Osada, T.; Chong, G.; Tansik, R.; Hong, T.; Spector, N.; Kumar, R.; Hurwitz, H.I.; Dev, I.; Nixon, A.B.; Lyerly, H.K.; et al. The effect of anti-VEGF therapy on immature myeloid cell and dendritic cells in cancer patients. Cancer Immunol. Immunother. 2008, 57, 1115–1124. [Google Scholar] [CrossRef]
  40. Terme, M.; Pernot, S.; Marcheteau, E.; Sandoval, F.; Benhamouda, N.; Colussi, O.; Dubreuil, O.; Carpentier, A.F.; Tartour, E.; Taieb, J. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 2013, 73, 539–549. [Google Scholar] [CrossRef]
  41. Kusmartsev, S.; Eruslanov, E.; Kübler, H.; Tseng, T.; Sakai, Y.; Su, Z.; Kaliberov, S.; Heiser, A.; Rosser, C.; Dahm, P.; et al. Oxidative stress regulates expression of VEGFR1 in myeloid cells: Link to tumor-induced immune suppression in renal cell carcinoma. J. Immunol. 2008, 181, 346–353. [Google Scholar] [CrossRef]
  42. Petrelli, F.; Borgonovo, K.; Cabiddu, M.; Ghilardi, M.; Lonati, V.; Maspero, F.; Sauta, M.G.; Beretta, G.D.; Barni, S. FOLFIRI-bevacizumab as first-line chemotherapy in 3500 patients with advanced colorectal cancer: A pooled analysis of 29 published trials. Clin. Color. Cancer 2013, 12, 145–151. [Google Scholar] [CrossRef]
  43. Kabbinavar, F.F.; Schulz, J.; McCleod, M.; Patel, T.; Hamm, J.T.; Hecht, J.R.; Mass, R.; Perrou, B.; Nelson, B.; Novotny, W.F. Addition of bevacizumab to bolus fluorouracil leucovorin in first-line metastatic colorectal cancer: Results of a randomized phase IItrial. J. Clin. Oncol. 2005, 23, 3697–3705. [Google Scholar] [CrossRef] [PubMed]
  44. Kabbinavar, F.; Hurwitz, H.I.; Fehrenbacher, L.; Meropol, N.J.; Novotny, W.F.; Lieberman, G.; Griffing, S.; Bergsland, E. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J. Clin. Oncol. 2003, 21, 60–65. [Google Scholar] [CrossRef]
  45. Tang, W.; Ren, L.; Liu, T.; Ye, Q.; Wei, Y.; He, G.; Lin, Q.; Wang, X.; Wang, M.; Liang, F.; et al. Bevacizumab Plus mFOLFOX6 Versus mFOLFOX6 Alone as First-Line Treatment for RAS Mutant Unresectable Colorectal Liver-Limited Metastases: The BECOME Randomized Controlled Trial. J. Clin. Oncol. 2020, 38, 3175–3184. [Google Scholar] [CrossRef] [PubMed]
  46. Saltz, L.B.; Clarke, S.; Díaz-Rubio, E.; Scheithauer, W.; Figer, A.; Wong, R.; Koski, S.; Lichinitser, M.; Yang, T.S.; Rivera, F.; et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: A randomized phase III study. J. Clin. Oncol. 2008, 26, 2013–2019. [Google Scholar] [CrossRef]
  47. Botrel, T.E.A.; Clark, L.G.d.O.; Paladini, L.; Clark, O.A.C. Efficacy and safety of bevacizumab plus chemotherapy compared to chemotherapy alone in previously untreated advanced or metastatic colorectal cancer: A systematic review and meta-analysis. BMC Cancer 2016, 16, 677. [Google Scholar] [CrossRef]
  48. Cao, Y.; Tan, A.; Gao, F.; Liu, L.; Liao, C.; Mo, Z. A meta-analysis of randomized controlled trials comparing chemotherapy plus bevacizumab with chemotherapy alone in metastatic colorectal cancer. Int. J. Color. Dis. 2009, 24, 677–685. [Google Scholar] [CrossRef]
  49. Hu, W.; Xu, W.S.; Liao, X.F.; He, H.J. Bevacizumab in combination with first-line chemotherapy in patients with metastatic colorectal cancer: A meta-analysis. Minerva Chir. 2015, 70, 451–458. [Google Scholar] [PubMed]
  50. Hurwitz, H.I.; Tebbutt, N.C.; Kabbinavar, F.; Giantonio, B.J.; Guan, Z.-Z.; Mitchell, L.; Waterkamp, D.; Tabernero, J. Efficacy and Safety of Bevacizumab in Metastatic Colorectal Cancer: Pooled Analysis From Seven Randomized Controlled Trials. Oncologist 2013, 18, 1004–1012. [Google Scholar] [CrossRef]
  51. Loupakis, F.; Bria, E.; Vaccaro, V.; Cuppone, F.; Milella, M.; Carlini, P.; Cremolini, C.; Salvatore, L.; Falcone, A.; Muti, P.; et al. Magnitude of benefit of the addition of bevacizumab to first-line chemotherapy for metastatic colorectal cancer: Meta-analysis of randomized clinical trials. J. Exp. Clin. Cancer Res. 2010, 29, 58. [Google Scholar] [CrossRef] [PubMed]
  52. Welch, S.; Spithoff, K.; Rumble, R.B.; Maroun, J.; Gastrointestinal Cancer Disease Site Group. Bevacizumab combined with chemotherapy for patients with advanced colorectal cancer: A systematic review. Ann. Oncol. 2010, 21, 1152–1162. [Google Scholar] [CrossRef]
  53. Qu, C.-Y.; Zheng, Y.; Zhou, M.; Zhang, Y.; Shen, F.; Cao, J.; Xu, L.M. Value of bevacizumab in treatment of colorectal cancer: Ameta-analysis World. J. Gastroenterol. 2015, 21, 5072–5080. [Google Scholar]
  54. Heinemann, V.; Weikersthal, L.F.; von Decker, T.; Kiani, A.; Vehling-Kaiser, U.; Al-Batran, S.-E.; Heintges, T.; Lerchenmüller, C.; Kahl, C.; Seipelt, G.; et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): A randomised, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 1065–1075. [Google Scholar] [CrossRef]
  55. Venook, A.P.; Niedzwiecki, D.; Lenz, H.-J.; Innocenti, F.; Fruth, B.; Meyerhardt, J.A.; Schrag, D.; Greene, C.; O’Neil, B.H.; Atkins, J.N.; et al. Effect of First-Line Chemotherapy Combined With Cetuximab or Bevacizumab on Overall Survival in Patients With KRAS Wild-Type Advanced or Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 2017, 317, 2392–2401. [Google Scholar] [CrossRef]
  56. Schwartzberg, L.S.; Rivera, F.; Karthaus, M.; Fasola, G.; Canon, J.-L.; Hecht, J.R.; Yu, H.; Oliner, K.S.; Go, W.Y. PEAK: A randomized, multicenter phase II study of panitumumab plus modified fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or bevacizumab plus mFOLFOX6 in patients with previously untreated, unresectable, wild-type KRAS exon 2 metastatic colorectal cancer. J. Clin. Oncol. 2014, 32, 2240–2247. [Google Scholar]
  57. Yoshino, T.; Uetake, H.; Tsuchihara, K.; Shitara, K.; Yamazaki, K.; Oki, E.; Sato, T.; Naitoh, T.; Komatsu, Y.; Kato, T.; et al. Rationale for and Design of the PARADIGM Study: Randomized Phase III Study of mFOLFOX6 Plus Bevacizumab or Panitumumab in Chemotherapy-naïve Patients With RAS (KRAS/NRAS) Wild-type, Metastatic Colorectal Cancer. Clin. Color. Cancer 2017, 16, 158–163. [Google Scholar] [CrossRef]
  58. Yamada, Y.; Denda, T.; Gamoh, M.; Iwanaga, I.; Yuki, S.; Shimodaira, H.; Nakamura, M.; Yamaguchi, T.; Ohori, H.; Kobayashi, K.; et al. S-1 and irinotecan plus bevacizumab versus mFOLFOX6 or CapeOX plus bevacizumab as first-line treatment in patients with metastatic colorectal cancer (TRICOLORE): A randomized, open-label, phase III, noninferiority trial. Ann. Oncol. 2018, 29, 624–631. [Google Scholar] [CrossRef] [PubMed]
  59. Gruenberger, T.; Bridgewater, J.; Chau, I.; García Alfonso, P.; Rivoire, M.; Mudan, S.; Lasserre, S.; Hermann, F.; Waterkamp, D.; Adam, R. Bevacizumab plus mFOLFOX-6 or FOLFOXIRI in patients with initially unresectable liver metastases from colorectal cancer: The OLIVIA multinational randomised phase II trial. Ann. Oncol. 2015, 26, 702–708. [Google Scholar] [CrossRef] [PubMed]
  60. Cremolini, C.; Loupakis, F.; Antoniotti, C.; Lupi, C.; Sensi, E.; Lonardi, S.; Mezi, S.; Tomasello, G.; Ronzoni, M.; Zaniboni, A.; et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: Updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol. 2015, 16, 1306–1315. [Google Scholar] [CrossRef]
  61. Cremolini, C.; Antoniotti, C.; Stein, A.; Bendell, J.; Gruenberger, T.; Rossini, D.; Masi, G.; Ongaro, E.; Hurwitz, H.; Falcone, A.; et al. Individual Patient Data Meta-Analysis of FOLFOXIRI Plus Bevacizumab Versus Doublets Plus Bevacizumab as Initial Therapy of Unresectable Metastatic Colorectal Cancer. J. Clin. Oncol. 2020, 38, 3314–3324. [Google Scholar] [CrossRef] [PubMed]
  62. Cunningham, D.; Lang, I.; Marcuello, E.; Lorusso, V.; Ocvirk, J.; Shin, D.B.; Jonker, D.; Osborne, S.; Andre, N.; Waterkamp, D.; et al. Bevacizumab plus capecitabine versus capecitabine alone in elderly patients with previously untreated metastatic colorectal cancer (AVEX): An open-label, randomised phase 3 trial. Lancet Oncol. 2013, 14, 1077–1085. [Google Scholar] [CrossRef]
  63. Holch, J.W.; Ricard, I.; Stintzing, S.; Modest, D.P.; Heinemann, V. The relevance of primary tumour location in patients with metastatic colorectal cancer: A meta-analysis of first-line clinical trials. Eur. J. Cancer 2017, 70, 87–98. [Google Scholar] [CrossRef]
  64. Pietrantonio, F.; Fucà, G.; Rossini, D.; Schmoll, H.-J.; Bendell, J.C.; Morano, F.; Antoniotti, C.; Corallo, S.; Borelli, B.; Raimondi, A.; et al. FOLFOXIRI-Bevacizumab or FOLFOX-Panitumumab in Patients with Left-Sided RAS/BRAF Wild-Type Metastatic Colorectal Cancer: A Propensity Score-Based Analysis. Oncologist 2021, 26, 302–309. [Google Scholar] [PubMed]
  65. Folprecht, G.; Pericay, C.; Saunders, M.P.; Thomas, A.; Lopez Lopez, R.; Roh, J.K.; Chistyakov, V.; Höhler, T.; Kim, J.S.; Hofheinz, R.D.; et al. Oxaliplatin and 5-FU/folinic acid (modified FOLFOX6) with or without aflibercept in first-line treatment of patients with metastatic colorectal cancer: The AFFIRM study. Ann. Oncol. 2016, 27, 1273–1279. [Google Scholar] [CrossRef]
  66. Koeberle, D.; Betticher, D.C.; von Moos, R.; Dietrich, D.; Brauchli, P.; Baertschi, D.; Matter, K.; Winterhalder, R.; Borner, M.; Anchisi, S.; et al. Bevacizumab continuation versus no continuation after first-line chemotherapy plus bevacizumab in patients with metastatic colorectal cancer: A randomized phase III non-inferiority trial (SAKK 41/06). Ann. Oncol. 2015, 26, 709–714. [Google Scholar] [CrossRef]
  67. Aparicio, T.; Ghiringhelli, F.; Boige, V.; Le Malicot, K.; Taieb, J.; Bouché, O.; Phelip, J.M.; François, E.; Borel, C.; Faroux, R.; et al. Bevacizumab Maintenance Versus No Maintenance During Chemotherapy-Free Intervals in Metastatic Colorectal Cancer: A Randomized Phase III Trial (PRODIGE 9). J. Clin. Oncol. 2018, 36, 674–681. [Google Scholar] [CrossRef]
  68. Goey, K.K.H.; Elias, S.G.; van Tinteren, H.; Laclé, M.M.; Willems, S.M.; Offerhaus, G.J.A.; de Leng, W.W.J.; Strengman, E.; Ten Tije, A.J.; Creemers, G.M.; et al. Maintenance treatment with capecitabine and bevacizumab versus observation in metastatic colorectal cancer: Updated results and molecular subgroup analyses of the phase 3 CAIRO3 study. Ann. Oncol. 2017, 28, 2128–2134. [Google Scholar] [CrossRef]
  69. Hegewisch-Becker, S.; Graeven, U.; Lerchenmüller, C.A.; Killing, B.; Depenbusch, R.; Steffens, C.-C.; Al-Batran, S.E.; Lange, T.; Dietrich, G.; Stoehlmacher, J.; et al. Maintenance strategies after first-line oxaliplatin plus fluoropyrimidine plus bevacizumab for patients with metastatic colorectal cancer (AIO 0207): A randomised, non-inferiority, open-label, phase 3 trial. Lancet Oncol. 2015, 16, 1355–1369, Erratum in Lancet Oncol. 2015, 16, 1355–1363. [Google Scholar] [CrossRef]
  70. Giantonio, B.J.; Catalano, P.J.; Meropol, N.J.; O’Dwyer, P.J.; Mitchell, E.P.; Alberts, S.R.; Schwartz, M.A.; Benson, A.B. Bevacizumab in combination with oxaliplatin fluorouracil leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: Results from the Eastern Cooperative Oncology Group Study E3200. J. Clin. Oncol. 2007, 25, 1539–1544. [Google Scholar] [CrossRef] [PubMed]
  71. Bennouna, J.; Sastre, J.; Arnold, D.; Österlund, P.; Greil, R.; Van Cutsem, E.; von Moos, R.; Viéitez, J.M.; Bouché, O.; Borg, C.; et al. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): A randomised phase 3 trial. Lancet Oncol. 2013, 14, 29–37. [Google Scholar] [CrossRef]
  72. Masi, G.; Salvatore, L.; Boni, L.; Loupakis, F.; Cremolini, C.; Fornaro, L.; Schirripa, M.; Cupini, S.; Barbara, C.; Safina, V.; et al. Continuation or reintroduction of bevacizumab beyond progression to first-line therapy in metastatic colorectal cancer: Final results of the randomized BEBYP trial. Ann. Oncol. 2015, 26, 724–730. [Google Scholar] [CrossRef]
  73. Iwamoto, S.; Takahashi, T.; Tamagawa, H.; Nakamura, M.; Munemoto, Y.; Kato, T.; Hata, T.; Denda, T.; Morita, Y.; Inukai, M.; et al. FOLFIRI plus bevacizumab as second-line therapy in patients with metastatic colorectal cancer after first-line bevacizumab plus oxaliplatin-based therapy: The randomized phase III EAGLE study. Ann. Oncol. 2015, 26, 1427–1433. [Google Scholar] [CrossRef]
  74. Grothey, A.; Flick, E.D.; Cohn, A.L.; Bekaii-Saab, T.S.; Bendell, J.C.; Kozloff, M.; Roach, N.; Mun, Y.; Fish, S.; Hurwitz, H.I. Bevacizumab exposure beyond first disease progression in patients with metastatic colorectal cancer: Analyses of the ARIES observational cohort study. Pharmacoepidemiol. Drug Saf. 2014, 23, 726–734. [Google Scholar] [CrossRef]
  75. Pfeiffer, P.; Yilmaz, M.; Möller, S.; Zitnjak, D.; Krogh, M.; Petersen, L.N.; Poulsen, L.Ø.; Winther, S.B.; Thomsen, K.G.; Qvortrup, C. TAS-102 with or without bevacizumab in patients with chemorefractory metastatic colorectal cancer: An investigator-initiated, open-label, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 412–420. [Google Scholar]
  76. Tabernero, J.; Prager, G.W.; Fakih, M.; Ciardiello, F.; Van Cutsem, E.; Elez, E.; Cruz, F.M.; Wyrwicz, L.; Stroyakovskiy, D.; Papai, Z.; et al. Trifluridine/tipiracil plus bevacizumab for third-line treatment of refractory metastatic colorectal cancer: The phase 3 randomized SUNLIGHTstudy. J. Clin. Oncol. 2023, 41, 4. [Google Scholar]
  77. Li, J.; Qin, S.; Xu, R.; Yau, T.C.C.; Ma, B.; Pan, H.; Xu, J.; Bai, Y.; Chi, Y.; Wang, L.; et al. Regorafenib plus best supportive care versus placebo plus best supportive care in Asian patients with previously treated metastatic colorectal cancer (CONCUR): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2015, 16, 619–629. [Google Scholar] [PubMed]
  78. Seidel, J.A.; Otsuka, A.; Kabashima, K. Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Front. Oncol. 2018, 8, 86. [Google Scholar]
  79. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. New England Journal of Medicine. Mass. Med. Soc. 2015, 372, 2509–2520. [Google Scholar]
  80. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar]
  81. Bedke, J.; Albiges, L.; Capitanio, U.; Giles, R.H.; Hora, M.; Lam, T.B.; Ljungberg, B.; Marconi, L.; Klatte, T.; Volpe, A.; et al. Updated European Association of Urology Guidelines on Renal Cell Carcinoma: Nivolumab plus Cabozantinib Joins Immune Checkpoint Inhibition Combination Therapies for Treatment-naïve Metastatic Clear-Cell Renal Cell Carcinoma. Eur. Urol. 2021, 79, 339–342. [Google Scholar] [CrossRef]
  82. Rini, B.I.; Plimack, E.R.; Stus, V.; Gafanov, R.; Hawkins, R.; Nosov, D.; Pouliot, F.; Alekseev, B.; Soulières, D.; Melichar, B.; et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1116–1127. [Google Scholar] [CrossRef]
  83. Kudo, M. Scientific Rationale for Combined Immunotherapy with PD-1/PD-L1 Antibodies and VEGF Inhibitors in Advanced Hepatocellular Carcinoma. Cancers 2020, 12, 1089. [Google Scholar] [CrossRef]
  84. Bendell, J.C.; Powderly, J.D.; Lieu, C.H.; Eckhardt, S.G.; Hurwitz, H.; Hochster, H.S. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) and/or FOLFOX in patients (pts) with metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2015, 33, 704. [Google Scholar] [CrossRef]
  85. Wallin, J.; Pishvaian, M.J.; Hernandez, G.; Yadav, M.; Jhunjhunwala, S.; Delamarre, L.; Xian, H.e.; Powderly, J.; Lieu, C.; Eckhardt, S.; et al. Abstract 2651: Clinical activity and immune correlates from a phase Ib study evaluating atezolizumab (anti-PDL1) in combination with FOLFOX and bevacizumab (anti-VEGF) in metastatic colorectal carcinoma. Cancer Res. 2016, 76, 2651. [Google Scholar] [CrossRef]
  86. Grothey, A.; Tabernero, J.; Arnold, D.; Gramont, A.D.; Ducreux, M.P.; O’Dwyer, P.J. Fluoropyrimidine (FP) + bevacizumab (BEV) + atezolizumab vs FP/BEV in BRAFwt metastatic colorectal cancer (mCRC): Findings from Cohort 2 of MODUL—A multicentre, randomized trial of biomarker-driven maintenance treatment following first-line induction therapy. Ann. Oncol. 2018, 29, viii714–viii715. [Google Scholar]
  87. Tabernero, J.; Grothey, A.; Arnold, D.; de Gramont, A.; Ducreux, M.; O’Dwyer, P.; Tahiri, A.; Gilberg, F.; Irahara, N.; Schmoll, H.J.; et al. MODUL cohort 2: An adaptable, randomized, signal-seeking trial of fluoropyrimidine plus bevacizumab with or without atezolizumab maintenance therapy for BRAFwt metastatic colorectal cancer. ESMO Open 2022, 7, 100559. [Google Scholar] [CrossRef] [PubMed]
  88. Mettu, N.; Twohy, E.; Ou, F.-S.; Halfdanarson, T.; Lenz, H.; Breakstone, R.; Boland, P.M.; Crysler, O.; Wu, C.; Grothey, A.; et al. 533PDBACCI: A phase II randomized, double-blind, multicenter, placebo-controlled study of capecitabine (C) bevacizumab (B) plus atezolizumab (A) or placebo (P) in refractory metastatic colorectal cancer (mCRC): An ACCRU network study. Ann. Oncol. 2019, 30, v203. [Google Scholar] [CrossRef]
  89. Shitara, K.; Hara, H.; Takahashi, N.; Kojima, T.; Kawazoe, A.; Asayama, M.; Yoshii, T.; Kotani, D.; Tamura, H.; Mikamoto, Y.; et al. Updated results from a phase Ib trial of regorafenib plus nivolumab in patients with advanced colorectal or gastric cancer (REGONIVO, EPOC1603). J. Clin. Oncol. 2020, 38, 135. [Google Scholar] [CrossRef]
  90. Antoniotti, C.; Borelli, B.; Rossini, D.; Pietrantonio, F.; Morano, F.; Salvatore, L.; Lonardi, S.; Marmorino, F.; Tamberi, S.; Corallo, S.; et al. AtezoTRIBE: A randomised phase II study of FOLFOXIRI plus bevacizumab alone or in combination with atezolizumab as initial therapy for patients with unresectable metastatic colorectal cancer. BMC Cancer 2020, 20, 683. [Google Scholar] [CrossRef]
  91. Lenz, H.-J.; Parikh, A.R.; Spigel, D.R.; Cohn, A.L.; Yoshino, T.; Kochenderfer, M.D.; Elez, E.; Shao, S.H.; Deming, D.; Holdridge, R.; et al. Nivolumab (NIVO) + 5-fluorouracil/leucovorin/oxaliplatin (mFOLFOX6)/bevacizumab (BEV) versus mFOLFOX6/BEV for first-line (1L) treatment of metastatic colorectal cancer (mCRC): Phase 2 results from CheckMate 9X8. J. Clin. Oncol. 2022, 40, 8. [Google Scholar] [CrossRef]
  92. Damato, A.; Bergamo, F.; Antonuzzo, L.; Nasti, G.; Pietrantonio, F.; Tonini, G.; Romagnani, A.; Berselli, A.; Normanno, N.; Pinto, C. Phase II study of nivolumab in combination with FOLFOXIRI/bevacizumab as first-line treatment in patients with advanced colorectal cancer RAS/BRAFmutated (mut): NIVACORtrial (GOIRC-03-2018). J. Clin. Oncol. 2022, 40, 3509. [Google Scholar] [CrossRef]
  93. Redman, J.M.; Tsai, Y.-T.; Weinberg, B.A.; Donahue, R.N.; Gandhy, S.; Gatti-Mays, M.E.; Abdul Sater, H.; Bilusic, M.; Cordes, L.M.; Steinberg, S.M.; et al. A Randomized Phase II Trial of mFOLFOX6 + Bevacizumab Alone or with AdCEA Vaccine + Avelumab Immunotherapy for Untreated Metastatic Colorectal Cancer. Oncologist 2022, 27, 198–209. [Google Scholar] [CrossRef]
  94. Wang, F.; He, M.-M.; Yao, Y.-C.; Zhao, X.; Wang, Z.-Q.; Jin, Y.; Luo, H.Y.; Li, J.B.; Wang, F.H.; Qiu, M.Z.; et al. Regorafenib plus toripalimab in patients with metastatic colorectal cancer: A phase Ib/II clinical trial and gut microbiome analysis. Cell Rep. Med. 2021, 2, 100383. [Google Scholar] [CrossRef]
  95. Shirley, M. Fruquintinib: First Global Approval. Drugs 2018, 78, 1757–1761. [Google Scholar] [CrossRef] [PubMed]
  96. Li, J.; Qin, S.; Xu, R.-H.; Shen, L.; Xu, J.; Bai, Y.; Yang, L.; Deng, Y.; Chen, Z.D.; Zhong, H.; et al. Effect of Fruquintinib vs Placebo on Overall Survival in Patients with Previously Treated Metastatic Colorectal Cancer: The FRESCO Randomized Clinical Trial. JAMA 2018, 319, 2486–2496. [Google Scholar] [CrossRef]
  97. Dasari, N.A.; Lonardi, S.; Garcia-Carbonero, R.; Fernandez, M.E.E.; Yoshino, T.; Sobrero, A.F.; Yao, J.; García-Alfonso, P.; Kocsis, J.; Cubillo Gracian, A.; et al. LBA25 FRESCO-2: A global phase III multiregional clinical trial (MRCT) evaluating the efficacy and safety of fruquintinib in patients with refractory metastatic colorectal cancer. Ann. Oncol. 2022, 33, S1391–S1392. [Google Scholar] [CrossRef]
  98. Loupakis, F.; Cremolini, C.; Fioravanti, A.; Orlandi, P.; Salvatore, L.; Masi, G.; Di Desidero, T.; Canu, B.; Schirripa, M.; Frumento, P.; et al. Pharmacodynamic and pharmacogenetic angiogenesis-related markers of first-line FOLFOXIRI plus bevacizumab schedule in metastatic colorectal cancer. Br. J. Cancer 2011, 104, 1262–1269. [Google Scholar] [CrossRef] [PubMed]
  99. Weickhardt, A.J.; Williams, D.S.; Lee, C.K.; Chionh, F.; Simes, J.; Murone, C.; Wilson, K.; Parry, M.M.; Asadi, K.; Scott, A.M.; et al. Vascular endothelial growth factor D expression is a potential biomarker of bevacizumab benefit in colorectal cancer. Br. J. Cancer 2015, 113, 37–45. [Google Scholar] [CrossRef] [PubMed]
  100. Tabernero, J.; Hozak, R.R.; Yoshino, T.; Cohn, A.L.; Obermannova, R.; Bodoky, G.; Garcia-Carbonero, R.; Ciuleanu, T.E.; Portnoy, D.C.; Prausová, J.; et al. Analysis of angiogenesis biomarkers for ramucirumab efficacy in patients with metastatic colorectal cancer from RAISE, a global, randomized, double-blind, phase III study. Ann. Oncol. 2018, 29, 602–609. [Google Scholar] [CrossRef]
  101. Parikh, A.R.; Lee, F.-C.; Yau, L.; Koh, H.; Knost, J.; Mitchell, E.P.; Bosanac, I.; Choong, N.; Scappaticci, F.; Mancao, C.; et al. MAVERICC, a Randomized, Biomarker-stratified, Phase II Study of mFOLFOX6-Bevacizumab versus FOLFIRI-Bevacizumab as First-line Chemotherapy in Metastatic Colorectal Cancer. Clin. Cancer Res. 2019, 25, 2988–2995. [Google Scholar]
  102. Bates, D.O.; Catalano, P.J.; Symonds, K.E.; Varey, A.H.R.; Ramani, P.; O’Dwyer, P.J.; Giantonio, B.J.; Meropol, N.J.; Benson, A.B.; Harper, S.J.; et al. Association between VEGF Splice Isoforms and Progression-Free Survival in Metastatic Colorectal Cancer Patients Treated with Bevacizumab. Clin. Cancer Res. 2012, 18, 6384–6391. [Google Scholar] [CrossRef]
  103. Bai, L.; Wang, F.; Zhang, D.-S.; Li, C.; Jin, Y.; Wang, D.-S.; Chen, D.L.; Qiu, M.Z.; Luo, H.Y.; Wang, Z.Q.; et al. A plasma cytokine and angiogenic factor (CAF) analysis for selection of bevacizumab therapy in patients with metastatic colorectal cancer. Sci. Rep. 2015, 5, 17717. [Google Scholar] [CrossRef]
  104. Giampieri, R.; Ziranu, P.; Daniele, B.; Zizzi, A.; Ferrari, D.; Lonardi, S.; Zaniboni, A.; Cavanna, L.; Rosati, G.; Casagrande, M.; et al. From CENTRAL to SENTRAL (SErum aNgiogenesis cenTRAL): Circulating Predictive Biomarkers to Anti-VEGFR Therapy. Cancers 2020, 12, 1330. [Google Scholar]
  105. Harris, A.L. Hypoxia—A key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47. [Google Scholar] [PubMed]
  106. Scartozzi, M.; Giampieri, R.; Maccaroni, E.; Del Prete, M.; Faloppi, L.; Bianconi, M.; Galizia, E.; Loretelli, C.; Belvederesi, L.; Bittoni, A.; et al. Pre-treatment lactate dehydrogenase levels as predictor of efficacy of first-line bevacizumab-based therapy in metastatic colorectal cancer patients. Br. J. Cancer 2012, 106, 799–804. [Google Scholar]
  107. Passardi, A.; Scarpi, E.; Tamberi, S.; Cavanna, L.; Tassinari, D.; Fontana, A.; Pini, S.; Bernardini, I.; Accettura, C.; Ulivi, P.; et al. Impact of Pre-Treatment Lactate Dehydrogenase Levels on Prognosis and Bevacizumab Efficacy in Patients with Metastatic Colorectal Cancer. PLoS ONE 2015, 10, e0134732. [Google Scholar]
  108. Marmorino, F.; Salvatore, L.; Barbara, C.; Allegrini, G.; Antonuzzo, L.; Masi, G.; Loupakis, F.; Borelli, B.; Chiara, S.; Banzi, M.C.; et al. Serum LDH predicts benefit from bevacizumab beyond progression in metastatic colorectal cancer. Br. J. Cancer 2017, 116, 318–323. [Google Scholar] [CrossRef]
  109. Carbone, C.; Piro, G.; Simionato, F.; Ligorio, F.; Cremolini, C.; Loupakis, F.; Alì, G.; Rossini, D.; Merz, V.; Santoro, R.; et al. Homeobox B9 Mediates Resistance to Anti-VEGF Therapy in Colorectal Cancer Patients. Clin. Cancer Res. 2017, 23, 4312–4322. [Google Scholar] [CrossRef] [PubMed]
  110. Noonan, S.A.; Morrissey, M.E.; Martin, P.; Biniecka, M.; Ó’Meachair, S.; Maguire, A.; Tosetto, M.; Nolan, B.; Hyland, J.; Sheahan, K.; et al. Tumour vasculature immaturity, oxidative damage and systemic inflammation stratify survival of colorectal cancer patients on bevacizumab treatment. Oncotarget 2018, 9, 10536–10548. [Google Scholar] [CrossRef]
  111. Zhan, J.; Niu, M.; Wang, P.; Zhu, X.; Li, S.; Song, J.; He, H.; Wang, Y.; Xue, L.; Fang, W.; et al. Elevated HOXB9 expression promotes differentiation and predicts a favourable outcome in colon adenocarcinoma patients. Br. J. Cancer 2014, 111, 883–893. [Google Scholar] [CrossRef]
  112. van Dijk, E.; Biesma, H.D.; Cordes, M.; Smeets, D.; Neerincx, M.; Das, S.; Eijk, P.P.; Murphy, V.; Barat, A.; Bacon, O.; et al. Loss of Chromosome 18q11.2-q12.1 Is Predictive for Survival in Patients With Metastatic Colorectal Cancer Treated With Bevacizumab. J. Clin. Oncol. 2018, 36, 2052–2060. [Google Scholar] [CrossRef]
  113. van Dijk, E.; van Werkhoven, E.; Asher, R.; Mooi, J.K.; Espinoza, D.; van Essen, H.F.; Eijk, P.P.; Murphy, V.; Barat, A.; Bacon, O.; et al. Predictive value of chromosome 18q11.2-q12.1 loss for benefit from bevacizumab in metastatic colorectal cancer: A post hoc analysis of the randomized phase III-trial AGITG-MAX. Int. J. Cancer 2022, 151, 1166–1174. [Google Scholar] [CrossRef] [PubMed]
  114. Boisen, M.K.; Dehlendorff, C.; Linnemann, D.; Nielsen, B.S.; Larsen, J.S.; Østerlind, K.; Nielsen, S.E.; Tarpgaard, L.S.; Qvortrup, C.; Pfeiffer, P.; et al. Tissue MicroRNAs as Predictors of Outcome in Patients with Metastatic Colorectal Cancer Treated with First Line Capecitabine and Oxaliplatin with or without Bevacizumab. PLoS ONE 2014, 9, e109430. [Google Scholar] [CrossRef]
  115. Denda, T.; Sakai, D.; Hamaguchi, T.; Sugimoto, N.; Ura, T.; Yamazaki, K.; Fujii, H.; Kajiwara, T.; Nakajima, T.E.; Takahashi, S.; et al. Phase II trial of aflibercept with FOLFIRI as a second-line treatment for Japanese patients with metastatic colorectal cancer. Cancer Sci. 2019, 110, 1032–1043. [Google Scholar] [CrossRef] [PubMed]
  116. Hamaguchi, T.; Denda, T.; Kudo, T.; Sugimoto, N.; Ura, T.; Yamazaki, K.; Fujii, H.; Kajiwara, T.; Nakajima, T.E.; Takahashi, S.; et al. Exploration of potential prognostic biomarkers in aflibercept plus FOLFIRI in Japanese patients with metastatic colorectal cancer. Cancer Sci. 2019, 110, 3565–3572. [Google Scholar] [CrossRef]
  117. Goldstein, D.A.; Chen, Q.; Ayer, T.; Chan, K.K.W.; Virik, K.; Hammerman, A.; Brenner, B.; Flowers, C.R.; Hall, P.S. Bevacizumab for Metastatic Colorectal Cancer: A Global Cost-Effectiveness Analysis. Oncologist 2017, 22, 694–699. [Google Scholar] [CrossRef]
  118. Giuliani, J.; Bonetti, A. First-line therapies in metastatic colorectal cancer: Integrating clinical benefit with the costs of drugs. Int. J. Color. Dis. 2018, 33, 1505–1516. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Interplay between angiogenesis, the immune system, and vessel normalization in colorectal cancer (the Figure was originally created by the authors with Adobe Illustrator CC 2015-version 19.0.0. Abnormal tumor vasculature recruits immune-suppressive cells by altering the function of endothelial cells (ECs), macrophages, dendritic cells, myeloid-derived suppressor cells (MDSCs), CD8+ T cells, and regulatory T cells (Tregs). Anti-angiogenic agents can normalize tumor vasculature, reduce hypoxia, and reestablish the immune microenvironment, particularly of CD8 T cells, while decreasing immune-suppressive cells. When combined with immunotherapy, this synergy enhances vessel normalization, T cell activation, and antitumor immunity. Abbreviations: ANG2 = Angiopoietin-2; CCL28 = C-C motif chemokine ligand 28; CSF1 = colony-stimulating factor 1; CXCL8/12 = C-X-C motif chemokine ligand 8/12; DC = dendritic cell; FGF2 = fibroblast growth factor 2; FAsL = Fas Ligand; IL-10 = Interleukin 10; MDSC = myeloid-derived suppressor cell; MMP9 = matrix metalloproteinase 9; M2 = M2-like macrophage; PD-1 = programmed cell death protein 1; PD-L1 = programmed death-ligand 1; TCD4+ = CD4+ T cell; TCD8+ = CD8+ T cell; TGF-b = transforming growth factor beta; Treg = Regulatory T cell; and VEGF = vascular endothelial growth factor.
Figure 1. Interplay between angiogenesis, the immune system, and vessel normalization in colorectal cancer (the Figure was originally created by the authors with Adobe Illustrator CC 2015-version 19.0.0. Abnormal tumor vasculature recruits immune-suppressive cells by altering the function of endothelial cells (ECs), macrophages, dendritic cells, myeloid-derived suppressor cells (MDSCs), CD8+ T cells, and regulatory T cells (Tregs). Anti-angiogenic agents can normalize tumor vasculature, reduce hypoxia, and reestablish the immune microenvironment, particularly of CD8 T cells, while decreasing immune-suppressive cells. When combined with immunotherapy, this synergy enhances vessel normalization, T cell activation, and antitumor immunity. Abbreviations: ANG2 = Angiopoietin-2; CCL28 = C-C motif chemokine ligand 28; CSF1 = colony-stimulating factor 1; CXCL8/12 = C-X-C motif chemokine ligand 8/12; DC = dendritic cell; FGF2 = fibroblast growth factor 2; FAsL = Fas Ligand; IL-10 = Interleukin 10; MDSC = myeloid-derived suppressor cell; MMP9 = matrix metalloproteinase 9; M2 = M2-like macrophage; PD-1 = programmed cell death protein 1; PD-L1 = programmed death-ligand 1; TCD4+ = CD4+ T cell; TCD8+ = CD8+ T cell; TGF-b = transforming growth factor beta; Treg = Regulatory T cell; and VEGF = vascular endothelial growth factor.
Ijms 26 11605 g001
Table 1. First-line trials.
Table 1. First-line trials.
CitationTrialN° ptTreatmentType of TrialORRPFSOS
Hurwitz et al., NEJM 2004 [4]AVF2107g813Irinotecan/leucovorin/fluorouracil + Bevacizumab/placeboPhase III, blinded, randomized44.8% vs. 34.8% (p = 0.004)10.6 vs. 6.2 m
(p < 0.001)
20.3 vs. 15.6 m
(p < 0.001)
Kabbinavar et al., JCO 2003 [43]NA104Leucovorin/fluorouracil + placebo/Bevacizumab low dose/Bevacizumab high dosePhase II, open-label, randomized17% vs. 40% vs. 24%5.2 vs. 9.0 vs. 7.2 m13.8 vs. 21.5 vs. 16.1 m
Kabbinavar et al., JCO 2005 [44]NA209Leucovorin/fluorouracil + bevacizumab/placeboPhase II, blinded, randomized26% vs. 15.2%
(p = 0.055)
9.2 vs. 5.5 m
(p = 0.0002)
16.6 vs. 12.9 m
(p = 0.16)
Tang W et al., JCO 2020 [45]BECOME241mFOLFOX6 +/− BevacizumabPhase IV, open-label, randomized54.5% vs. 36.7%
(p < 0.01)
9.5 vs. 5.6 m
(p < 0.01)
25.7 vs. 20.5 m
(p = 0.03)
Saltz et al., JCO 2008 [46]NO169661401XELOX/FOLFOX4 + bevacizumab/placeboPhase III, blinded, randomized47% vs. 49%
(p = 0.31)
9.4 vs. 8 m
(p = 0.0023)
21.0 vs. 19.9 m
(p = 0.077)
Schmiegel et al.,
Ann Oncol 2013 [8]
NA255Bevacizumab + Capox/mCapIriPhase II, randomized, open-label53% vs. 56%10.4 vs. 12.1 m24.4 vs. 25.5 m
Heinemann V et al., Lancet Onc 2014 [54]FIRE 3592FOLFIRI + Cetuximab/BevacizumabPhase III, randomized, open-label62% vs. 58%
(p = 0.18)
10.0 vs. 10.3 m
(p = 0.55)
28.7 vs. 25.0 m
(p = 0.017)
Venook et al., JAMA 2017 [55]Calgb/Swog 804053058 (2334 KRAS WT)FOLFIRI/mFOLFOX6 + cetuximab/bevacizumabPhase III, randomized, open-label59.6% vs. 55.2%
(p = 0.13)
10.5 vs. 10.6 m
(p = 0.45)
30.0 vs. 29.0 m
(p = 0.08)
Schwartzberg et al., JCO 2014 [56]PEAK285mFOLFOX6 + panitumumab/bevacizumabPhase II, randomized, open-label57.8% vs. 53.5%10.9 vs. 10.1 m
(p = 0.353)
34.2 vs. 24.3 m
(p = 0.009)
Yamada et al., Ann Oncol 2018 [58]TRICOLORE487mFOLFOX6/Capox + bevacizumab vs. S1 + Irinotecan + BevacizumabPhase III, randomized, open-label, non-inferiority70.6% vs. 66.4%
(p = 0.34)
10.8 vs. 14.0 m
(p < 0.0001)
33.6 vs. 34.9 m
(p = 0.2841)
Gruenberger et al., Ann Oncol 2015 [59]OLIVIA80Bevacizumab + mFOLFOX6/FOLFOXIRIPhase II, randomized, open-label62% vs. 81%11.5 vs. 18.0 m
(HR 0.43)
32.2 vs. NR
(HR 0.35)
Cremolini et al., Lancet Oncol 2015 [60]TRIBE508Bevacizumab + FOLFIRI/FOLFOXIRIPhase III, randomized, open-label54% vs. 65%
(p = 0.013)
9.7 vs. 12.3 m
(p = 0.01)
25.8 vs. 29.8 m
(p = 0.03)
Cunningham et al., Lancet Oncol 2013 [62]AVEX280Capecitabine +/− BevacizumabPhase III, randomized, open-label 9.1 vs. 5.1 m
(p < 0.0001)
20.7 vs. 17.0 m
(p = 0.13)
Table 2. Maintenance trials.
Table 2. Maintenance trials.
CitationTrialN° ptTreatmentType of TrialPFSOS
Koeberle et al., Ann Oncol 2015 [66]SAKK 41/06262Bevacizumab vs. no treatmentPhase III, randomized, open-label4.1 vs. 2.9 m (TTP)25.4 vs. 23.8 m
(p = 0.2)
Aparicio et al., JCO 2018 [67]PRODIGE 9491Bevacizumab vs. no treatmentPhase III, randomized, open-label9.2 vs. 8.9 m
(p = 0.316)
21.7 vs. 22.0 m
(p = 0.5)
Goey et al., Ann Oncol 2017 [68]CAIRO 3558Capecitabine/Bevacizumab vs. no treatmentPhase III, randomized, open-label8.5 vs. 4.1 m
(p < 0.0001)
21.6 vs. 18.2
(p = 0.1)
Hegewisch-Becker et al., Lancet Oncol 2015 [69]AIO 02074725FU/Bevacizumab vs. Bevacizumab vs. no treatmentPhase III, randomized, open-label, non-inferiority6.3 vs. 4.6 vs. 3.5 m
(p < 0.0001)
20.2 vs. 21.9 vs. 23.1
(p = 0.77)
Table 3. Second-line trials.
Table 3. Second-line trials.
CitationTrialN° ptTreatmentType of TrialPFSOS
Giantonio et al., JCO 2007 [70]E3200829FOLFOX4/Bevacizumab vs. FOLFOX4 vs. Bevacizumab Phase III, open-label, randomized7.3 vs. 4.7 vs. 2.7 m
(p < 0.0001)
12.9 vs. 10.8 vs. 10.2
(p = 0.0011)
Bennouna et al., Lancet 2013 [71]ML18147409Chemotherapy +/− BevacizumabPhase III, open-label, randomized5.7 vs. 4.1 m
(p < 0.0001)
11.2 vs. 9.8 m
(p = 0.0062)
Masi et al., Ann Oncol 2015 [72]BEBYP185Chemotherapy +/− BevacizumabPhase III, open-label, randomized6.8 vs. 5.0 m
(p = 0.010)
15.5 vs. 14.1 m
(p = 0.043)
Iwamoto et al., Ann Oncol 2015 [73]EAGLE387FOLFIRI + Bevacizumab 5/10 mg/kgPhase III, open-label, randomized6.1 vs. 6.4 m
(p = 0.676)
16.3 vs. 17.0 m
(p = 0.667)
Van Cutsem et al., JCO 2012 [5]VELOUR1226FOLFIRI + Aflibercept/placeboPhase III, double-blind, randomized6.9 vs. 4.67 m
(p < 0.0001)
13.05 vs. 12.06 m
(p = 0.0032)
Tabernero et al., Lancet Oncol 2015 [7]RAISE1072FOLFIRI + Ramucirumab/placeboPhase III, double-blind, randomized5.7 vs. 4.5 m
(p = 0.0005)
13.3 vs. 11.7 m
(p = 0.0219)
Table 4. Beyond second-line trials.
Table 4. Beyond second-line trials.
CitationTrialN° ptTreatmentType of TrialPFSOS
Pfeiffer et al., Lancet Oncol 2020 [75]NA93TAS-102 +/− BevacizumabPhase II, open-label, randomized4.6 vs. 2.6 m
(p = 0.0010)
9.4 vs. 6.7 m
(p = 0.028)
Tabernero et al., JCO suppl 2023 [76]SUNLIGHT492TAS-102 +/− BevacizumabPhase III, open-label, randomized5.6 vs. 2.4 m
(p < 0.001)
10.8 vs. 7.5 m
(p < 0.001)
Grothey et al., Lancet 2013 [6]CORRECT760Regorafenib/placeboPhase III, quadruple masking, randomized1.9 vs. 1.7 m
(p < 0.0001)
6.4 vs. 5.0 m
(p = 0.0052)
Li et al., Lancet Oncol 2015 [77]CONCUR204BSC +/− RegorafenibPhase III,
double-blind,
randomized
3.2 vs. 1.7 m
(p < 0.0001)
8.8 vs. 6.3 m
(p = 0·00016)
Table 5. Summary of main clinical trials evaluating anti-angiogenic drugs plus ICIs.
Table 5. Summary of main clinical trials evaluating anti-angiogenic drugs plus ICIs.
TrialPhaseYearsN. of ptsTreatmentmPFSmOS
NCT01633970IbDecember 2014 through April 2017Arm A (pre-treated): 13
Arm B (naïve): 26
Arm A: atezolizumab + bevacizumab
Arm B: atezolizumab + FOLFOX + bevacizumab
NANA
MODUL (NCT02291289)IIApril 2015 through July 2019445 (naïve, BRAF wt)Maintenance bevacizumab +/− atezolizumab after FOLFOX + bevacizumabNot metNA
BACCI (NCT02873195)IISeptember 2017 through June 2018133 (pre-treated)Capecitabine and bevacizumab + atezolizumab/placebo4.4 vs. 3.6 moNA
NCT03239145Ib 18 (MSS, pre-treated)Pembrolizumab + trebananibNA9 mo
NCT03946917Ib/IIMarch 2019 through January 202042 (MSS pre-treated)Toripalimab + regorafenib2.1 mo15.5 mo
LEAP-005 (NCT03797326)II2019–202032 (MSS, pre-treated)Pembrolizumab + lenvatinib2.3 mo7.5 mo
NCT04126733IIOctober 2019 through January 202070 (MSS, pre-treated)Nivolumab + regorafenib15 weeks52 weeks
REGOMUNE (NCT03475953)IINovember 2018 through October 201948 (MSS, pre-treated)Avelumab + regorafenib3.6 mo10.8 mo
CheckMate 9X8
(NCT03414983)
IIFebruary 2018 through April 2019Experimental Arm: 127
Control Arm:68
Experimental Arm: FOLFOX + Bevacizumab + Nivolumab
Control Arm: FOLFOX + Bevacizumab
11.9 mo in both armsImmature data
Atezo TRIBE (NCT03721653)2November 2018 through February 2020218 (naïve)FOLFOXIRI + bevacizumab +/− atezolizumab13.1 vs. 11.5 mo (p = 0.012)NA
NIVACOR (NCT04072198)2October 2019 through March 202173 (naïve, RAS/BRAF mut)FOLFOXIRI + bevacizumab + nivolumab10.1 moNA
NCT033969262April 2018 through October 202144 (MSS, pre-treated)Pembrolizumab + capecitabine + bevacizumab4.3 mo9.6 mo
NCT030508142April 2017 through October 201926 (MSS, naïve)mFOLFOX6 + bevacizumab +/− avelumab + CEA-targeted vaccineNo diff.NA
NCT037129431bNovember 2018 through September 202051 (MSS, pre-treated)Nivolumab + regorafenib4.3 mo11.1 mo
NCT036576411/2July 2019 through July 202173 (MSS, pre-treated)Pembrolizumab + regorafenib2.8 mo9.6 mo
Legend: pts = patients; mPFS = median progression-free survival; mOS = median overall survival; MSS = microsatellite stability; MSI = microsatellite instability; mo = months.
Table 6. Summary of ongoing clinical trials with fruquintinb.
Table 6. Summary of ongoing clinical trials with fruquintinb.
TrialPhaseStudy StartSettingTreatmentPrimary Endpoint
NCT05004441II2021First-lineFOLFOX/FOLFIRI, fruquintinibORR
NCT04296019, NCT05016869,
NCT05451719, NCT04733963,
NCT05659290
II or I/II2021–2023MaintenanceFruquintinib or fruquintinib plus capecitabinePFS
NCT05634590II2022Second-lineFOLFOX/FOLFIRI, fruquintinibPFS
NCT05555901II2023Second-lineFOLFIRI plus fruquintinib vs. FOLFIRI plus bevacizumabPFS
NCT05522738Ib/II2022Second-lineFOLFIRI, fruquintinibORR
NCT05447715II2022Second-/Third-lineFruquintinib sequential bevacizumab plus FOLFIRI vs. bevacizumab plus FOLFIRI sequential fruquintinibPFS
NCT05004831II2022Third-lineFruquintinib, trifluridine/tipiracilPFS
NCT04695470II2020Pre-treatedFruquintinib, sintilimabPFS
NCT04582981II2020Pre-treatedFruquintinib plus raltitrexed vs. fruquintinibPFS
NCT04866862II2021Pre-treatedFruquintinib, camrelizumabORR
Legend: PFS = Progression-free survival; ORR = Overall response rate.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Basile, D.; Di Nardo, P.; Daffinà, M.G.; Cortese, C.; Giuliani, J.; Aprile, G. The Current Role of Antiangiogenics in Colorectal Cancer. Int. J. Mol. Sci. 2025, 26, 11605. https://doi.org/10.3390/ijms262311605

AMA Style

Basile D, Di Nardo P, Daffinà MG, Cortese C, Giuliani J, Aprile G. The Current Role of Antiangiogenics in Colorectal Cancer. International Journal of Molecular Sciences. 2025; 26(23):11605. https://doi.org/10.3390/ijms262311605

Chicago/Turabian Style

Basile, Debora, Paola Di Nardo, Maria Grazia Daffinà, Carla Cortese, Jacopo Giuliani, and Giuseppe Aprile. 2025. "The Current Role of Antiangiogenics in Colorectal Cancer" International Journal of Molecular Sciences 26, no. 23: 11605. https://doi.org/10.3390/ijms262311605

APA Style

Basile, D., Di Nardo, P., Daffinà, M. G., Cortese, C., Giuliani, J., & Aprile, G. (2025). The Current Role of Antiangiogenics in Colorectal Cancer. International Journal of Molecular Sciences, 26(23), 11605. https://doi.org/10.3390/ijms262311605

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop