Angiogenesis Inhibitors for Colorectal Cancer: Comparison
Please note this is a comparison between Version 1 by Torben Hansen and Version 3 by Rita Xu.

Targeting angiogenesis, the formation of new blood vessels, is an integral part of many cancer treatments, including colorectal cancer. The overall clinical benefit is well documented but modest. It has been an ongoing task for the last decade to isolate patient and tumor characteristics instrumental in identifying the subgroups to truly benefit; so far with limited success. The introduction of immunotherapy has opened a new era for anti-angiogenic treatment, as these two therapeutic strategies seem to work in synergy. This review will highlight the clinical achievements of anti-angiogenic treatment of colorectal cancer since 2004 and elaborate on the perspectives of combining it with immunotherapy.  

  • Angiogenesis
  • colorectal cancer
  • monoclonal antibodies
  • small molecule tyrosine kinase inhibitors

1. Introduction

1.1. Colorectal Cancer

Worldwide, 1.8 million new patients are diagnosed each year with colorectal cancer (CRC). Approximately half of the patients will be diagnosed with metastatic CRC (mCRC), at either the time of diagnosis (synchronous) or due to later recurrence (metachronous) [1]. Almost half the number of new cases, 0.86 million, die each year.

1.2. Treatment Overview

For several years, the armamentarium of standard treatment for patients with mCRC have included combination chemotherapy with either 5-FU, oxaliplatin, irinotecan, or two classes of targeted therapies [2][3][2,3]. These therapies inhibit the signaling pathways related to the epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) receptors. The monoclonal antibodies cetuximab and panitumumab, targeting the EGF receptor (EGFR) and bevacizumab, targeting the VEGF-A ligand, are the most commonly used in the field of mCRC.

It is well known that the benefit of anti-EGFR is restricted to around 40% of the patients who are RAS and BRAF wild type (wt) [2][3][2,3]. The common treatment approach has changed from single agent chemotherapy to a doublet regimen, or occasionally triplet chemotherapy regimen, often in combination with bevacizumab, cetuximab, or panitumumab based on the RAS mutational status. Typically, anti-EGFR therapy improves major efficacy parameters (response rate, PFS and OS) when added to doublet regimens like 5-FU + irinotecan (FOLFIRI) or 5-FU + oxaliplatin (FOLFOX) but results were more equivocal when bevacizumab was added to modern infusional doublet regimens (Table 1). Nerveless, the optimal combination of chemotherapy and targeted therapy for first line therapy have been debated for many years. Three randomized trials have directly compared efficacy of EGFR inhibitors and bevacizumab in patients with RASwt mCRC, but with a very heterogeneous picture and no well-founded conclusion. Prior studies have shown that left-sided mCRC are dependent on EGFR related pathways and when investigators from the major cooperative groups pooled data in patients with left-sided tumors, it became evident, and the efficacy data became much more homogenous, showing a clear advantage of EGFR inhibitors with higher overall response rates (ORR) and prolonged overall survival (OS) in patients with left-sided primaries [4][5][4,5]. However, there is currently no solid evidence indicating that RAS mutations should render anti-VEGF-A therapy obsolete in the setting of mCRC.

Table 1. Principal trials comparing first line combination chemotherapy with or without bevacizumab in unselected patients with metastatic colorectal cancer (mCRC).

, a crucial and initial step in the process of immunity, and lead to an upregulation of programmed death-ligand 1 PD–L1 on DCs further suppressing T cell function [26][81]. Leaky blood vessels increase the intra-tumoral pressure and together with the down-regulation of cell-adhesion molecules complicates extravasation of tumor infiltrating lymphocytes (TILs). Hypoxia itself lead to upregulation of PD–L1 and the simultaneous up-regulation of VEGF-A which furthermore impairs the function of the antigen presenting cells. Finally, the balance of TILs shifts towards increased infiltration of regulatory T cells (Treg) on behalf of cytotoxic effector cells (CD8+) due to regulatory changes in the ECs [27][82]. This is primarily due to an increased expression of FAS ligand on tumor-associated ECs that prevents effector T cells from crossing the EC barrier by inducing apoptosis. Treg are resistant to FAS ligand creating a relative overexpression of Treg in the tumor compared to the effector cells. Treg furthermore inhibit the antigen presenting cells in the tumor enhancing the immune suppressive environment. Tumor vasculature normalization thus creates an immune friendly microenvironment and may turn a “cold” tumor into a “warm”. Added to this is the recent discovery of how stimulated immune cells themselves lead to vascular normalization, partly through CD8+ T–cells and interferon gamma (IFN–Ƴ), creating a beneficial immune-vasculature crosstalk and a rationale for combining these two classes of therapeutics with a potential synergistic benefit [28][83]. The optimal dosing and timing of these treatment combinations will likely differ between individual tumor types and represent an essential key for unlocking their full potential.

2.2. Anti-angiogenetic Therapy and Immunotherapy in Colorectal Cancer

This potential synergism between anti-angiogenic and immune checkpoint inhibitor drugs has resulted in numerous clinical trials testing the combination of PD–1/PD–L1 antibodies with anti–VEGF drugs. By now, a number of randomized trials have shown remarkable results which have led to approvals by the FDA and/or EMA in renal cell carcinoma (axitinib plus pembrolizumab, and axitinib plus avelumab), endometrial carcinoma (pembrolizumab plus lenvatinib), non-squamous NSCLC (bevacizumab and atezolizumab), and hepatocellular carcinoma (bevacizumab plus atezolizumab). This broad clinical activity suggest that a combination strategy may also be of benefit in colorectal cancer [29][84].

The first clinical data in CRC were presented by Bendell et al. at the ASCO–GI conference in 2015 [30][85]. Among 13 patients with treatment refractory disease, they demonstrated one objective tumor response by adding bevacizumab to MPDL3280A (atezolizumab). The following year, at the AACR 107th annual meeting, Wallin et al. presented the results from 23 patients with mCRC treated with first-line FOLFOX, bevacizumab, and atezolizumab [31][86]. They demonstrated promising efficacy data, with a median PFS of 14.1 months and parallel translational research argued for immune-related activity by this combination. The corresponding papers to these two initial abstracts are so far not published.

In 2017, Yoshida et al. published the results of their pilot study (COMVI study) in anticancer research [32][87]. Six patients with previously untreated mCRC were included in a prospective single arm study. All patients received, as standard therapy, oxaliplatin (130 mg/m2) on day one, capecitabine (1000 mg/m2) twice daily on the days 1–14, and bevacizumab (7.5 mg/kg) on day one. To this backbone, they added cultured αβ T–lymphocytes (>5 × 109) combined with interleukin–2 and anti–CD3 on day 18. Two patients achieved a complete response, three a partial response, and one demonstrated stable disease as the best outcome. The median progression free and overall survival was 567 and 966 days, respectively. Adverse events were mild to moderate. Although a small pilot study, these published results, for the first time in patients with CRC, demonstrated that combining chemotherapy and anti-angiogenesis with immune-modulating therapies were feasible and efficacy data were promising.

An additional two abstracts were presented the following years but they did not quite meet the initial expectations. In 2018, at the ESMO congress, Grothey et al. presented a late-breaking abstract from cohort 2 of the MODUL trial [33][88]. After induction therapy with the FOLFOX + bevacizumab regimen, 445 patients were randomized to maintenance fluoropyrimidine and bevacizumab ± atezolizumab. The updated analyses revealed no difference in median PFS and OS between the two strategies. Mettu et al. presented an abstract at the poster discussion session at the same congress the following year [34][89]. In this study, 133 patients with mCRC, with treatment resistant disease, were randomized to receive capecitabine and bevacizumab plus placebo or atezolizumab as last line treatment. The study reached its primary endpoint demonstrating a significant improvement of PFS by the addition of atezolizumab, although the numerical difference was only one month. The corresponding manuscripts have not been published either.

The first publication based on a commercially available immuno-therapeutic, within this specific field in CRC, was published earlier this year, in April 2020, and revealed the results from the dose expansion phase Ib trial REGONIVO (EPOC1603) [35][90]. Fukuoka et al. included patients with gastric or CRC, 25 of each, who had progressed on a minimum of two previous lines of palliative treatment. All the patients with CRC had previously received anti-angiogenetic treatment, the cancer in one patient had deficient mismatch repair (dMMR) but the remaining 24 were all proficient (microsatellite stable), and six had RAS mutations. Patients were treated with nivolumab 3 mg/kg every two weeks and regorafenib once daily, day 1–21, in a four-week cycle. During the dosing-finding, part of the study regorafenib was reduced from initially 160 mg to the recommended 80 mg at which no patients experienced dose-limiting toxicity. Among the patients with CRC 9 (36%) achieved an objective tumor response and median PFS was 7.9 months. A trend towards better outcome for the patients with lung metastases, compared to liver metastases was presented, which could be due to a more immunosuppressive environment in the liver compared to the lung. This study provided real clinical evidence of synergy between the investigated drugs. Neither of the two would be expected to provide meaningful benefit as singe agents in this group (except the one patient with a dMMR tumor) but combined, one out of three responded.

These results, together with similar findings in other cancer types, have paved the way for multiple trials assessing the clinical benefit from combining immunotherapy and anti-angiogenetic treatment in CRC. An example is the recently published study protocol AtezoTRIBE by Antoniotti et al. [36][91]. In this randomized phase II trial untreated patients with unresectable mCRC will receive FOLFOXIRI and bevacizumab ± atezolizumab in four months followed by maintenance 5–fluoruracil, leucovorin, bevacizumab ± atezolizumab. The study is estimated to be completed in April 2021. A supplementary search at clinical.trials.gov for trials in CRC combining immunotherapy with an anti-angiogenetic drug revealed more than 20 ongoing clinical trials (Table 2Table 6). With a specific focus on CRC only, this level of clinical activity underlines the potential impact gained by combining these two classes of therapeutics.

Table 26. Ongoing clinical trials in metastatic colorectal cancer combining immunotherapy and anti-angiogenetic treatment.

 

 

A number of randomized studies, pioneered by Dr. Falcone’s group, have evaluated triplet chemotherapy (FOLFOXIRI) in patients unselected by the RAS mutational status. The FOLFOXIRI regimen does have a significant toxicity-profile, thus requiring patients to exhibit a good performance status. Consequently, patients included in the FOLFOXIRI trials are more often younger or more often in performance status 0 than usually in clinical trials. Two Italian phase III trials [15][16][15,16] showed that triplet chemotherapy was more effective than a doublet (either FOLFIRI or FOLFOX) in terms of ORR, PFS and OS. In the TRIBE–study, bevacizumab was added to both the triplet and the doublet combinations, and thus we can only conclude that a triplet chemotherapy can be safely combined with bevacizumab but whether bevacizumab adds to the efficacy of a triplet cannot be concluded from these studies [16]. In the randomized phase II OLIVIA trial [17], in which mCRC patients with liver-limited disease were included, a triplet chemotherapy with bevacizumab produced a very impressing ORR of 81%. Consistently, a high response rate of at least 60% was observed in all studies that evaluated FOLFOXIRI with or without bevacizumab [3].

1.3. Angiogenesis

Basically, the term vasculogenesis describes the process of the initial endothelial differentiation of angioblasts during embryogenesis [18], whereas angiogenesis refers to the formation of new blood vessels from existing endothelial cells [19]. The regulation of the angiogenic process is a complex balance between stimulating and inhibiting stimuli. The VEGF system consists of six ligands and three receptors (VEGFR). The VEGF-A ligand is the most important. It is secreted by multiple cell types including the malignant cells and stimulates endothelial cell (EC) differentiation, migration, growth and survival [20]. The receptor that is primarily responsible for transmitting this VEGF-A-mediated signal in the EC is VEGFR-2, whereas the role of VEGFR-1 probably is more regulatory and inhibitory [21]. The autonomic growth pattern that characterizes malignant neoplasms are contributing to the fact that malignant tumors are often hypoxic to varying degrees. This hypoxia leads to increased transcription of a large number of genes, including VEGF-A, with the common purpose of ensuring a more adequate oxygenation of the tumor [22]. It is the so-called hypoxia-inducible factors (HIF) that are the cause of this gene regulation. The three members are formed from the oxygen-sensitive subunits (HIF-1α, HIF-2α and HIF-3α) and the non-oxygen-sensitive HIF-1β subunit [23]. During hypoxia, HIF-1α (the best described) stabilizes and translocate to the cell nucleus to form the activated HIF-1 complex together with HIF-1β.

1.4. Anti-angiogenetics

The therapeutics targeting angiogenesis are divided into two main groups: the monoclonal antibodies (mAbs) and the small molecules, tyrosine kinase inhibitors (TKIs). The mAbs exert their action by either directly binding VEGF-A or blocking the extracellular binding domain of the corresponding receptor. Bevacizumab (Avastin®) binds to all isoforms of VEGF-A and aflibercept (Zaltrap®) a soluble decoy receptor binds VEGF, thereby preventing activation of their endogenous receptors whereas ramucirumab (Cyramza®) binds with high affinity to the VEGFR-2 extracellular domain, which prevents binding of VEGF ligands and thereby inhibiting receptor activation. The TKIs exert their anti-angiogenetic effect after internalization in the cell and binding to, and inhibiting, the kinase domain of the various receptors involved in the angiogenetic process (tyrosine kinase, serine/threonine kinase or dual protein kinase inhibitors).

1.5. Current Challenge

Inhibition of tumor-associated angiogenesis have been utilized for the treatment of patients with mCRC for more than 15 years [2][3][2,3]. Since the initial approval of bevacizumab in 2004, several other agents have been investigated within phase III trials, leading to several additional approvals.

2. Anti-angiogenesis and Immunotherapy

The limited benefit from anti-angiogenetic therapies in mCRC have paved the way for new combinations in order to sustain tumor control. The currently most promising approach is the combination with immunotherapy.

2.1. Tumor-Microenvironment

Tumor-associated blood vessels have a unique architecture and physiologic properties as previously addressed. These properties lead to the generation of an immune suppressive tumor microenvironment [24][79]. Malignant tumors have the ability to evade immune suppression through changes in the recruitment, trafficking and infiltration of effector T cells and their final recognition and killing of cancer cells. This is the consequence of several processes related to tumor-initiated angiogenesis. The initial up-regulation of VEGF-A inhibit the maturation of dendritic cells (DCs) [25][80]

1 Of investigators choice; Atezolizumab (anti-PD–L1); avelumab (anti-PD–L1); bevacizumab (anti-vascular endothelial growth factor A); BNC105 (a vascular disrupting agent); cabozantinib (tyrosine kinase inhibitor of c–MET and vascular endothelial growth factor receptor 2); cediranib (tyrosine kinase inhibitor of vascular endothelial growth factor receptors 1–3); durvalumab (anti-PD–1); ipilimumab (anti-CTLA–4); lenvatinib (tyrosine kinase inhibitor of vascular endothelial growth factor receptors 1–3); nivolumab (anti-PD–1); pembrolizumab (anti-PD–1); regorafenib (tyrosine kinase inhibitor of TIE2 and vascular endothelial growth factor 2); trebananib (anti-angiopoietin–2).

Video Production Service