Skip to main content

REVIEW article

Front. Pharmacol., 08 August 2023
Sec. Gastrointestinal and Hepatic Pharmacology
This article is part of the Research Topic Advances in Novel Drugs and Targets for Hepatic and Gastrointestinal Diseases View all 33 articles

Progress of research on molecular targeted therapies for colorectal cancer

Shilin Huang&#x;Shilin Huang1Jiazhou Ye&#x;Jiazhou Ye2Xing GaoXing Gao1Xi HuangXi Huang1Julu HuangJulu Huang2Lu LuLu Lu1Cheng LuCheng Lu2Yongqiang LiYongqiang Li1Min LuoMin Luo1Mingzhi XieMingzhi Xie1Yan Lin
&#x;Yan Lin1*Rong Liang
Rong Liang1*
  • 1Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
  • 2Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China

Colorectal cancer (CRC) is one of the most common malignancies, accounting for approximately 10% of global cancer incidence and mortality. Approximately 20% of patients with CRC present metastatic disease (mCRC) at the time of diagnosis. Moreover, up to 50% of patients with localized disease eventually metastasize. mCRC encompasses a complex cascade of reactions involving multiple factors and processes, leading to a diverse array of molecular mechanisms. Improved comprehension of the pathways underlying cancer cell development and proliferation, coupled with the accessibility of relevant targeted agents, has propelled advancements in CRC treatment, ultimately leading to enhanced survival rates. Mutations in various pathways and location of the primary tumor in CRC influences the efficacy of targeted agents. This review summarizes available targeted agents for different CRC pathways, with a focus on recent advances in anti-angiogenic and anti-epidermal growth factor receptor agents, BRAF mutations, and human epidermal growth factor receptor 2-associated targeted agents.

1 Introduction

Colorectal cancer (CRC) is the third most common cancer and the third leading cause of cancer-related death worldwide. The onset of CRC is subtle and challenging to detect in early stages, underscoring the importance of timely screening. Moreover, approximately 22% of patients are diagnosed with metastatic disease at the outset. The 5-year metastatic CRC (mCRC) survival rate is approximately 15% (Howlader N et al., 2020). Therefore, precise treatment of mCRC using appropriate drugs based on relevant molecular biosignatures is essential to prolong the survival time of patients.

The main treatment strategies for mCRC are chemotherapy using cytotoxic agents, molecular-targeted therapy, and immunotherapy. Combination chemotherapy regimens frequently involve the use of fluorouracil (5-FU)-based drugs together with oxaliplatin or irinotecan. These combinations have demonstrated significant improvements in patient survival, with survival durations exceeding 20 months (Tournigand et al., 2004; Van Cutsem et al., 2014; García-Alfonso et al., 2021; Glimelius et al., 2021). Rapid and significant advancement in the development of targeted agents for CRC has been observed since the approval of cetuximab for mCRC treatment in 2004 (Figure 1) Currently, more than 10 agents have been approved for mCRC treatment. Combined treatments utilizing targeted drugs and chemotherapy have been reported to increase the 5-year survival rate from 9% to 15% and survival to more than 30 months in mCRC patients (Van Cutsem et al., 2016; Siegel et al., 2022). As distant metastasis is the main cause of death in CRC, relevant review that comprehensively explores and summarizes the efficacy of molecularly targeted drugs in CRC and associated latest research progress does not exist, and this review highlights these. This review focuses on progress of research on targeted agents for treating mCRC patients, providing a reference for clinicians for precise treatment of CRC.

FIGURE 1
www.frontiersin.org

FIGURE 1. FDA-approved molecular targeted agents for mCRC and their associated clinical studies.

2 Classification and mechanism of common targets in CRC

2.1 Vascular endothelial growth factor (VEGF) target

VEGF family includes VEGFA–D, VEGF receptor (VEGFR), and placental growth factor. VEGFA, commonly known as VEGF or vascular permeability factor, is the main angiogenic feature of the VEGF family. The role of VEGFB in tumor angiogenesis has not yet been elucidated; however, VEGFC and VEGFD mainly regulate lymphatic endothelial cell growth (Dvorak, 2002; Karkkainen et al., 2002; Ferrara et al., 2003; Bry et al., 2014). VEGFR includes VEGFR1–3, with VEGFA capable of binding to VEGFR1 and 2, and VEGFC and VEGFD binding to VEGFR2 and 3, respectively. VEGFR activation can promote cell proliferation, migration, and growth via the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinases (PI3K) pathways, leading to angiogenesis and tumor angiogenesis (Ivy et al., 2009). VEGF inhibitors used in CRC treatment include monoclonal antibodies (mAbs) like bevacizumab, ramucirumab, Ziv-aflibercept, as well as tyrosine kinase inhibitors (TKIs), such as sunitinib, sorafenib, fruquintinib, and regorafenib. These drugs target the VEGF pathway to inhibit angiogenesis and tumor growth (Figure 2).

FIGURE 2
www.frontiersin.org

FIGURE 2. Common molecular targets and their mechanisms of action in CRC.

2.2 EGFR and HER2 target

Epidermal growth factor receptor (EGFR), also known as ERBB1 or human epidermal growth factor receptor (HER) 1, is a member of the HER family, which also includes HER2–4. EGFR promotes tumor cell proliferation, differentiation, growth, and distant metastasis by activating downstream signaling pathways, such as PI3K and MAPK (Roskoski, 2014; Kumagai et al., 2021). EGFR activation induces the secretion of angiogenic factors, such as VEGF, which stimulate the formation of new blood vessels. Increased angiogenesis ensures sufficient blood supply to the tumor, facilitating its expansion and providing nutrients for sustained growth. Additionally, EGFR signaling provides anti-apoptotic signals through the activation of AKT, which inhibits apoptosis by inactivating pro-apoptotic proteins (De Luca et al., 2008). This allows cancer cells to evade programmed cell death and survive in unfavorable conditions. Furthermore, HER2 overexpression, as a member of the EGFR family, inhibits the tumor suppressor gene, p53 (Ménard et al., 2003). Common EGFR inhibitors used in cancer treatment include anti-EGFR mAbs, such as cetuximab and panitumumab, EGFR TKIs like gefitinib, erlotinib, and afatinib, as well as HER2 mAbs trastuzumab and pertuzumab, and HER2 TKI lapatinib. The inhibitors target EGFR and HER2, blocking their signaling pathways and inhibiting tumor growth and progression.

2.3 RAS/RAF/MEK/ERK target

The RAS/RAF/MEK/ERK signaling cascade (MAPK) pathway is one of the downstream EGFR pathways, which mainly regulates cell proliferation and differentiation. RAS family members include KRAS, NRAS, and HRAS (Karnoub and Weinberg, 2008). KRAS mutation (MT) accounts for approximately 32%–40% of CRC cases, and BRAF V600E MT accounts for 5%–15% (De Roock et al., 2011; Pakneshan et al., 2013). MEK, or MAPK/ERK kinase, plays a pivotal role as a mediator in the downstream signaling of the MAPK pathway. ERK, also known as extracellular signal-regulated kinase, is a critical component of this pathway (Downward, 2003). Studies have confirmed that MEK/ERK inhibitors significantly enhance the treatment efficacy in patients with KRAS/BRAF-mutant tumors (Hatzivassiliou et al., 2013; Morris et al., 2013). Available inhibitors targeting the RAS/RAF/MEK/ERK pathway include dabrafenib and vemurafenib as BRAF inhibitors, and trametinib and encorafenib as MEK inhibitors. Additionally, there are ERK inhibitors such as ulixertinib and temuterkib.

2.4 PI3K/AKT/mTOR target

PI3K is an enzyme that plays a crucial role in cellular signaling pathways involved in cell growth, survival, and metabolism. It functions by phosphorylating the lipid phosphatidylinositol 4,5-bisphosphate (PIP2) to generate phosphatidylinositol 3,4,5-trisphosphate (PIP3), which in turn activates downstream signaling pathways. PIP3 acts as a docking site for AKT, facilitating its activation through phosphorylation. Notably, one of the significant downstream AKT targets the mammalian target of rapamycin (mTOR), which plays a critical role in protein synthesis, cellular growth, and metabolic regulation. Aberrant activation of PI3K can occur through various mechanisms, including genetic mutations, PI3K genes amplification, or upstream receptors activation such as EGFR. Dysregulation of the PI3K/AKT/mTOR pathway has been implicated in various diseases, including cancer, highlighting its significance as a therapeutic target for intervention and treatment strategies (Fruman and Rommel, 2014; Polivka and Janku, 2014; Janku et al., 2018). Several inhibitors have been developed to target the PI3K/AKT/mTOR pathway. These include PI3K inhibitors such as buparlisib and sonolisib, which specifically target the PI3K enzyme and AKT inhibitors like MK-2206 and ipatasertib that block the activity of the AKT protein, a downstream effector of PI3K. Sirolimus and everolimus are targeted towards mTOR, a key component of the pathway. Additionally, dual PI3K/mTOR inhibitors such as dactolisib and apitolisib simultaneously target PI3K and mTOR.

2.5 NOTCH target

NOTCH signaling involves the activation of NOTCH receptors by ligands such as JAG1, resulting in the release of the Notch intracellular domain (NICD). The NICD then translocates to the nucleus and forms a complex with transcriptional regulators to activate target genes, including the hairy and enhancer of split (HES) family genes. This signaling pathway plays a crucial role in cell fate determination, differentiation, and various cellular processes (Siebel and Lendahl, 2017; Li et al., 2023). NOTCH receptors mAbs such as tarextumab and demcizumab, have been developed and tested in preclinical and clinical studies.

2.6 Wnt target

The Wnt signaling pathway plays a critical role in various biological processes, including embryonic development, tissue homeostasis, and cell proliferation. Dysregulation of this pathway has been implicated in several diseases, particularly cancer. Recruitment and activation of Disheveled (Dvl) are initiated by the activation of the Wnt signaling pathway. Subsequently, a complex involving Adenomatous Polyposis Coli (APC), glycogen synthase kinase 3β (GSK-3β), Casein kinase 1 (CK1), and Axin forms, leading to phosphorylation and inhibition of GSK3β (Duchartre et al., 2016; Zhang and Wang, 2020). This, in turn, results in increased levels of β-catenin, a pivotal component of the Wnt signaling pathway, which drives cancer cell proliferation. WNT974 and CGX1321, inhibitors targeting the Wnt ligand/receptor interface, have shown promising efficacy in preclinical studies (Rodon et al., 2021a; Rodon et al., 2021b).

2.7 NTRK gene fusion target

The tropomyosin receptor kinase (TRK) family comprises TRK A–C, which is encoded by neurotrophic TRK (NTRK)1–3. NTRK gene fusion occurs when the 3′region of the NTRK gene and 5′end of the fusion chaperone gene are connected by intra-chromosome or inter-chromosome rearrangement. The protein encoded by the fusion gene can bind to TRK and activate the downstream PI3K and MAPK pathways, resulting in tumor growth, proliferation, and differentiation. NTRK gene fusion was first identified in patients with CRC and then in those with other tumors (Martin-Zanca et al., 1986; Vaishnavi et al., 2015; Amatu et al., 2019; Solomon et al., 2019). Entrectinib, which targets the NTRK fusion gene, has been shown to be highly therapeutic for patients with NTRK. (2019).

3 Common targeted agents for CRC therapy

3.1 Bevacizumab

Bevacizumab specifically binds to VEGF, blocking its interaction with the receptor, degrading existing tumor blood vessels, normalizing surviving ones, and inhibiting tumor neovascularization to exert antitumor effects.

3.1.1 Adjuvant therapy

The QUASAR 2 study showed that CRC patients treated with capecitabine (Cap) + bevacizumab or only Cap after radical (R0) resection had 3-year disease-free survival (DFS) rates of 75.4% and 78.4%, respectively (Kerr et al., 2016). Similarly, the NSABP protocol C-08 and other studies (Allegra et al., 2011; de Gramont et al., 2012; André et al., 2020) confirmed that neoadjuvant chemotherapy with bevacizumab in combination with oxaliplatin did not increase DFS in patients compared to that in patients treated with chemotherapy alone. Overall, none of these trials showed a significant survival benefit for CRC patients treated with adjuvant bevacizumab; therefore, none of the current guidelines recommend the use of bevacizumab as adjuvant therapy. Adjuvant and neoadjuvant treatment trials in mCRC are summarized in Table 1.

TABLE 1
www.frontiersin.org

TABLE 1. Adjuvant and neoadjuvant treatment trials in mCRC.

3.1.2 Neoadjuvant therapy

A previous study showed that six cycles of bevacizumab in combination with chemotherapy, followed by chemoradiotherapy (CRT) achieved an objective response rate (ORR) of 88.9% in locally advanced colorectal patients with T4 or high-risk T3, with R0 resection rate of 97.8% (Masi et al., 2019). Similarly, the CRAB study reported a 95% R0 resection rate in patients with stage II/III rectal cancer treated with neoadjuvant bevacizumab + CRT (Velenik et al., 2020). Although these studies reported positive outcomes, available evidence is insufficient to support the adoption of bevacizumab as a standard neoadjuvant (Table 1).

3.1.3 First-line treatment

The AVF2107 study showed the administration of FOLFIRI + bevacizumab as a first-line treatment significantly improved (p < 0.001) the median overall survival (mOS; 20.3 vs. 15.6 months) and median progression-free survival (mPFS; 10.6 vs. 6.2 months) of mCRC patients compared to those of the mCRC patients treated with chemotherapy alone (Hurwitz et al., 2004). This finding facilitated the approval of bevacizumab by the United States Food and Drug Administration (FDA) as a first-line mCRC treatment in 2004. Additionally, the administration of bevacizumab + chemotherapy significantly increased (p = 0.0023) the mPFS of patients compared to that of the patients treated with chemotherapy alone (9.4 vs. 8.0 months) but did not affect mOS (Saltz et al., 2008). Moreover, both the MAVERICC (Parikh et al., 2019) and ARIES studies (Bendell et al., 2012) confirmed that FOLFIRI or FOLFOX + bevacizumab had similar PFS and OS.

However, the TRIBE2 study (Cremolini et al., 2020) showed that FOLFOXIRI + bevacizumab achieved higher mOS (27.4 vs. 22.5 months) and mPFS (19.2 vs. 16.4 months) than mFOLFOX6 + bevacizumab. The treatment benefit was independent of RAS and BRAF mutation status but was better in patients with right-sided tumors, and the same benefit was achieved in patients who progressed after treatment with FOLFOXIRI + bevacizumab (Loupakis et al., 2015; Cremolini et al., 2018b). Moreover, both the AVEX (Cunningham et al., 2013) and PRODIGE20 (Aparicio et al., 2018a) studies confirmed that bevacizumab provided increased treatment benefits in elderly patients and did not induce adverse events. Key trials of anti-VEGF agents in the treatment of mCRC are illustrated in Table 2.

TABLE 2
www.frontiersin.org

TABLE 2. Key trials of anti-VEGF agents in the treatment of mCRC.

3.1.4 Maintenance treatment

In the CAIRO3 study (Simkens et al., 2015), bevacizumab + Cap was used as maintenance treatment for mCRC patients after first-line treatment with chemotherapy + bevacizumab that led to improved mPFS (8.5 vs. 4.1 months, p < 0.0001); however, the maintenance treatment did not affect OS. Similarly, both the PRODIGE9 (Aparicio et al., 2018b) and AIO0207 (Hegewisch-Becker et al., 2015) studies confirmed bevacizumab-induced improvement in PFS after induction chemotherapy. Moreover, the 2016 ESMO guidelines state that Cap + bevacizumab can be used for maintenance therapy after first-line treatment, but bevacizumab alone is not recommended for maintenance therapy. Additionally, the National Comprehensive Cancer Network (NCCN) guidelines do not recommend bevacizumab as maintenance therapy.

3.1.5 Second-line treatment

The E3200 study (Giantonio et al., 2007) showed that the administration of bevacizumab as a second-line treatment in mCRC patients achieved mOS of 12.9, 10.2, and 10.8 months and mPFS of 7.3, 2.7, and 4.7 months in patients subjected to FOLFOX + bevacizumab, bevacizumab only, or chemotherapy only first-line therapies, respectively. Thus, bevacizumab was approved in 2006 as a second-line treatment for patients with mCRC. Additionally, the ML18147 study showed that the administration of chemotherapy + bevacizumab or chemotherapy only as a second-line treatment achieved mOS of 11.2 vs. 9.6 months (p = 0.0062) and mPFS of 5.7 vs. 4.1 months (p < 0.0001), respectively, in mCRC patients subjected to bevacizumab as a first-line therapy (Bennouna et al., 2013). Moreover, the ML18147 and several other studies confirmed that retreatment with bevacizumab-containing regimens exerted significant therapeutic effects in mCRC patients who received bevacizumab-containing regimens as first-line therapy (Grothey et al., 2008; Hurwitz et al., 2014). Based on these findings, the FDA approved bevacizumab as a cross-line therapy in 2013.

3.1.6 Third-line treatment and beyond

A previous study showed that trifluridine and tipiracil (TAS-102) + bevacizumab was more effective than TAS-102 alone as a third-line treatment in patients with anti-EGFR positive and chemotherapy-resistant mCRC, with considerable increase in mOS (9.4 vs. 6.7 months, p = 0.028) and mPFS (4.6 vs. 2.6 months, p = 0.0010) (Pfeiffer et al., 2020). Similarly, the TAS-CC3 study confirmed the positive effects of TAS-102 + bevacizumab as a third-line treatment in Asian patients with mCRC (Yoshida et al., 2021). Accordingly, the NCCN guidelines recommend the use bevacizumab in combination with TAS-102 as a third-line treatment in patients with mCRC (Table 2).

3.2 Ziv-aflibercept

Ziv-aflibercept is an anti-VEGF agent that inhibits neovascularization by tightly binding to VEGF and reducing vascular permeability. The AFFIRM study showed (Folprecht et al., 2016) that abatacept did not achieve promising outcomes as a first-line treatment for mCRC. However, considerable therapeutic benefit was obtained with Ziv-aflibercept as a second-line treatment (Van Cutsem et al., 2012). Notably, FOLFIRI + Ziv-aflibercept was more effective than FOLFIRI only as a second-line treatment in mCRC patients, with improved mOS (13.5 vs. 12.06 months, p = 0.0032) and mPFS (6.90 vs. 4.67 months, p < 0.0001). Accordingly, Ziv-aflibercept was approved by the FDA as a second-line treatment for mCRC patients who progressed or were resistant to first-line oxaliplatin therapy in 2012 (Table 2).

3.3 Cetuximab

Cetuximab binds specifically to EGFR and competitively blocks VEGF and other receptors, inhibiting intracellular signaling pathways, thereby suppressing the proliferation of cancer cells and inducing apoptosis.

3.3.1 Adjuvant therapy

In the N0147 [NCCTG] study (Alberts et al., 2012), mFOLFOX + cetuximab did not show significant health-promoting effects as an adjuvant compared to those of chemotherapy alone, with no statistical difference in 3-year DFS in patients with KRAS wild-type (WT). Moreover, the PETACC-8 (Taieb et al., 2014) study confirmed that cetuximab adjuvant therapy had no survival benefit in patients with KRAS WT. Current guidelines do not recommend the use of cetuximab as an adjuvant therapy (Table 1).

3.3.2 Neoadjuvant treatment

The EXPERT-C study (Dewdney et al., 2012) showed that CAPOX + cetuximab was more effective than CAPOX only (four cycles each) in neoadjuvant therapy, with significantly higher response rate (RR) and OS in the cetuximab group. However, some studies, including the COIN study (Maughan et al., 2011; Tveit et al., 2012), showed that cetuximab treatment did not improve patient survival; in contrast, the TAILOR study (Qin et al., 2018) confirmed cetuximab OS benefit. The 2012 NCCN guidelines do not recommend cetuximab as a neoadjuvant therapy because of insufficient evidence (Table 1).

3.3.3 First-line treatment

The CRYSTAL study (Van Cutsem et al., 2009) showed that treatment with FOLFIRI + cetuximab significantly increased mOS (24.9 vs. 21.0 months) and mPFS (9.9 vs. 8.7 months) in patients with KRAS WT mCRC compared to those in patients with KRAS WT treated with FOLFIRI treatment alone. Accordingly, cetuximab was approved by the FDA in 2012 as a first-line treatment in patients with KRAS WT mCRC. In contrast, the COIN (Maughan et al., 2011) and NORDIC-VII (Tveit et al., 2012) studies showed insignificant differences in OS and PFS between oxaliplatin + cetuximab and oxaliplatin treated patients with KRAS WT mCRC. However, the TAILOR study (Qin et al., 2018) showed that cetuximab + FOLFOX increased mPFS (9.2 vs. 7.4 months, p = 0.004) and mOS (20.7 vs. 17.8 months, p = 0.02) in patients with RAS WT mCRC compared to those in FOLFOX alone treated patients with RAS WT mCRC. Moreover, previous studies have shown that cetuximab was as effective as bevacizumab in patients with RAS WT (Heinemann et al., 2014; Venook et al., 2017). Additionally, the NCT00208546 study showed insignificant difference in efficacy between cetuximab + bevacizumab and cetuximab only (Tol et al., 2009); therefore, cetuximab + bevacizumab is not recommended for patients with mCRC.

The CALGB/SWOG80405 (Alliance) study (Venook et al., 2017) found that cetuximab achieved significantly higher OS and PFS in patients with left-sided primary tumors than in those with right-sided primary tumors. Several studies confirmed that anti-EGFR antibody in combination with chemotherapy exerted the best effects in patients with left-sided primary tumors and RAS WT mCRC (Arnold et al., 2017; Tejpar et al., 2017). Pivotal trials of anti-EGFR monoclonal antibody treatment in mCRC are summarized in Table 3.

TABLE 3
www.frontiersin.org

TABLE 3. Pivotal trials of anti-EGFR mAbs treatment in mCRC.

3.3.4 Maintenance treatment

The MACRO2TTD study (Aranda et al., 2018) confirmed insignificant differences in mPFS and mOS between continuation of the original regimen and maintenance treatment with cetuximab alone after induction therapy with mFOLFOX + cetuximab. Moreover, the MACBETH study (Cremolini et al., 2018a) suggested that cetuximab could be used for maintenance therapy in patients with RAS/RAF WT mCRC. However, current guidelines do not recommend the use of cetuximab for maintenance therapy.

3.3.5 Second-line treatment

The BOND study (Cunningham et al., 2004) confirmed that cetuximab in combination with chemotherapy achieved a better therapeutic benefit than cetuximab alone in mCRC treatment, with increase in mOS (8.6 vs. 6.9 months, p = 0.48) and mPFS (4.1 vs. 1.5 months, p < 0.001). Cetuximab was approved by the FDA in 2004 as a second-line treatment for patients with mCRC. Subsequent studies (Sobrero et al., 2008; Ciardiello et al., 2016) have confirmed the therapeutic benefit of cetuximab in combination with chemotherapy over chemotherapy alone. Moreover, the UNICANCERPRODIGE18 study (Bennouna et al., 2019) showed that cetuximab had therapeutic benefits comparable to those of bevacizumab as second-line treatment.

3.3.6 Third-line treatment

The NCT00079066 study (Jonker et al., 2007) demonstrated that cetuximab monotherapy resulted in higher OS and PFS in patients with refractory mCRC than the best supportive care (BSC) alone. Vincenzi et al. (2006) treated patients with mCRC using cetuximab + irinotecan as third-line chemotherapy, with an mPFS of 4.7 months and mOS of 9.8 months.

Rechallenge therapy means the reintroduction of targeted agent to which a tumor has already proven to be resistant (Tonini et al., 2013). Santini et al. (2012) performed cetuximab rechallenge as a third-line treatment for refractory mCRC and achieved promising results, with ORR of 53.8% [partial response (PR), 48.7%; complete response (CR), 5.1%] and mPFS of 6.6 months. Similarly, patients with KRAS WT mCRC rechallenged with cetuximab as third-line treatment exhibited positive results, with mPFS and OS of 2.4 and 8.2 months, respectively (Masuishi et al., 2020). A meta-analysis conducted by Mauri et al. (2019) showed that anti-EGFR rechallenge therapy yielded better therapeutic benefits than sequential and dose escalation therapies. Therefore, cetuximab could be used as a third-line treatment (Table 3).

3.4 Panitumumab

Panitumumab is an IgG2 monoclonal antibody that binds to EGFR, blocking the binding of VEGFR and VEGF and inhibiting cancer cell growth.

3.4.1 First-line treatment

The PRIME study (Douillard et al., 2010) showed that treatment with FOLFOX4 + panitumumab significantly improved mPFS in patients with KRAS WT mCRC compared to that in patients with KRAS WT mCRC treated with FOLFOX4 alone (9.6 vs. 8.0 months, p = 0.02) but did not affect OS and was not significant in patients with KRAS MT. Moreover, the ASPECCT trial (Price et al., 2014) confirmed that panitumumab achieved results comparable to those of cetuximab in patients with KRAS WT mCRC. Based on these findings, panitumumab was approved by the FDA as a first-line treatment for patients with KRAS WT mCRC in 2014. A retrospective analysis of the PRIME study (Douillard et al., 2013) showed that all patients with RAS WT benefited from panitumumab treatment. Interestingly, the PEAK study (Schwartzberg et al., 2014) showed that panitumumab had better efficacy than bevacizumab in patients with KRAS/NRAS WT mCRC. Therefore, the FDA included NRAS WT mCRC as an indication for panitumumab treatment in 2017. Additionally, studies (Arnold et al., 2017; Boeckx et al., 2017; Peeters et al., 2018) have shown that panitumumab was more effective against RAS WT tumors located on the left side than those on the right side.

Furthermore, although treatment with mFOLFOXIRI + panitumumab did not significantly affect mOS and mPFS in patients with RAS WT mCRC, there was a significant increase (p = 0.004) in ORR (87.3% vs. 60.6%, p = 0.004) and metastasis resection-free recurrence survival (7.9 vs. 4.0 months) in the mFOLFOXIRI + panitumumab group compared to those in the mFOLFOXIRI group (Modest et al., 2019a). Similar results were found in the GONO study (Fornaro et al., 2013). Overall, panitumumab + FOLFOXIRI could be used for patients with RAS WT mCRC with metastases that require surgical resection.

3.4.2 Maintenance treatment

The NCT02476045 study (Pietrantonio et al., 2019) showed that 5-Fu + panitumumab was more effective than 5-Fu only as maintenance treatment in patients with RAS WT mCRC, with 10-month PFS of 59.9% and 49.0% (p = 0.01) in the 5-Fu + panitumumab and 5-Fu groups, respectively. Additionally, there were no significant differences in PFS, OS, and RR between panitumumab + mFOLFOX6-and 5-Fu + panitumumab-treated patients with RAS WT mCRC (Munemoto et al., 2019). Moreover, the PANAMA study (Modest et al., 2022) confirmed that 5-Fu + panitumumab was a better maintenance treatment than 5-Fu alone. A retrospective analysis of the PRIME and PEAK studies confirmed that panitumumab was comparable to bevacizumab as a maintenance therapy (Modest et al., 2019b), indicating that panitumumab could be combined with other agents as a maintenance therapy for patients with RAS WT mCRC.

3.4.3 Second-line treatment

The PICCOLO study (Seymour et al., 2013) showed that irinotecan + panitumumab was effective as a second-line treatment against KRAS WT mCRC than irinotecan only, as evidenced by a significant increase in PFS and RR in the panitumumab group; however, the OS was not significantly affected. Moreover, panitumumab had limited effects on patients with RAS MT. Similar results were obtained in the 20050181 study (Peeters et al., 2010; Peeters et al., 2014); therefore, panitumumab and cetuximab are sometimes used interchangeably as second-line treatments.

3.4.4 Third-line treatment and beyond

Cutsem Eric Van Cutsem et al. (2007) reported that panitumumab monotherapy as a third-line treatment significantly increased (p < 0.0001) mPFS to 8 weeks in patients with mCRC compared to that in patients with mCRC treated with BSC only (7.3 weeks) but did not affect OS. Accordingly, panitumumab was approved by the FDA as a third-line treatment for patients with EGFR-positive mCRC in 2006. Similar results were observed in patients retreated with panitumumab + BSC (Van Cutsem et al., 2008). Overall, several studies have confirmed the efficacy of panitumumab monotherapy as a third-line treatment for mCRC (Hecht et al., 2007; Kim et al., 2016; Kim et al., 2018). Moreover, the GERCOR (André et al., 2013) and WJOG6510G (Sakai et al., 2020) studies both confirmed that panitumumab + irinotecan improved PFS in patients with KRAS WT mCRC but did not affect OS (Table 3).

3.5 Ramucirumab

Ramucirumab exerts its antitumor effects by specifically binding to VEGFR-2 and inhibiting tumor angiogenesis. The RAISE study (Tabernero et al., 2015) showed that treatment with FOLFIRI + ramucirumab increased mOS (p = 0.0219) and mPFS (p < 0.0005) to 13.3 and 5.7 months, respectively, compared with 11.7 and 4.5 months, respectively, in FOLFIRI-treated patients. Moreover, the treatment benefit of ramucirumab was superior to chemotherapy-only for both OS and PFS in any subgroup. Ramucirumab was approved as a second-line treatment for patients with mCRC by the FDA in 2015 (Table 3).

3.6 Fruquintinib

Fruquintinib is a VEGFR inhibitor that blocks neointimal growth associated with tumor proliferation and is a potent and highly selective small-molecule inhibitor of VEGFR1–3. The FRESCO study (Li et al., 2018) showed that fruquintinib + BSC treatment achieved considerable therapeutic effects in mCRC patients, irrespective of whether the patients were previously treated with anti-VEGF or anti-EGFR drugs, as evidenced by an increase (p < 0.001) in mOS (9.3 months) and mPFS (3.7 months) compared to those in mCRC patients treated with the BSC only (mOS, 6.6 months; mPFS, 1.8 months). Accordingly, fruquintinib was approved by the China National Medical Products Administration as a third-line treatment for patients with mCRC in 2018; however, fruquintinib has not been approved for use in other countries. Similarly, the results of FRESCO-2 study showed that fruquintinib treatment significantly improved (p < 0.001) mOS (7.4 months) and mPFS (3.7 months) compared to those of BSC treatment (mOS, 4.8 months; mFPS, 1.8 months) (Dasari et al., 2022). Key trials of HER2 targeted agents and TKIs in mCRC are summarized in Table 4.

TABLE 4
www.frontiersin.org

TABLE 4. Major trials of HER2 mAbs and all TKIs in mCRC.

3.7 Regorafenib

Regorafenib is a TKI that acts on multiple targets, such as VEGFR and BRAF V600E. The CORRECT study (Grothey et al., 2013) showed that regorafenib + BSC improved mOS (6.4 months) and mPFS (1.9 months) in patients with mCRC compared to patients with mCRC treated with BSC only (mOS, 5 months; mPFS, 1.7 months). Accordingly, regorafenib was approved as a third-line treatment for patients with mCRC by the FDA in 2012. The CONCUR study (Li et al., 2015) demonstrated the benefits of regorafenib in Asian population. Regorafenib is the only monotherapy recommended by the NCCN guidelines for third-line therapy and further treatment (Table 4).

3.8 Encorafenib

Encorafenib primarily targets the BRAFV600E MT, which is commonly found in certain types of cancer. Additionally, it has inhibitory effects on JNK1–3, LIMK1–4, and STK36. The BEACON study (Kopetz et al., 2019) showed that encorafenib + binimetinib + cetuximab triple-agent treatment exerted considerable therapeutic effects in mCRC patients compared to those in the mCRC patients treated with FOLFIRI + cetuximab (control). However, subsequent studies showed insignificant differences in OS and mPFS between encorafenib + binimetinib + cetuximab and encorafenib + cetuximab groups (Tabernero et al., 2021). Accordingly, the NCCN guidelines recommend the use of encorafenib in combination with anti-EGFR as a second-line treatment in patients with BRAFV600E MT mCRC (Table 4).

3.9 Dabrafenib and trametinib

Dabrafenib specifically targets BRAF mutations (V600E and V600K), while trametinib selectively inhibits MEK1 and MEK2, key components of the RAS/RAF/MEK/ERK signaling pathway, resulting in the tumor growth suppression. The NCT01072175 study (Corcoran et al., 2015) showed that combined second-line treatment with dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor) increased mPFS to 3.5 months in patients with BRAF V600E MT mCRC, with PR observed in 12% of the patients and CR in one patient. Similarly, the NCT01750918 study (Corcoran et al., 2018) showed that panitumumab + dabrafenib + trametinib treatment increased RR (21%), mPFS (4.2 months), and mOS (13.2 months) in patients with BRAFV600E MT mCRC compared to those in patients with BRAFV600E MT mCRC treated with panitumumab + dabrafenib or panitumumab + trametinib. Therefore, the 2020 NCCN guidelines recommend that dabrafenib + trametinib could be combined with cetuximab or panitumumab as a second-line treatment for BRAFV600E MT mCRC. However, the 2021 and 2022 NCCN guidelines do not include dabrafenib and trametinib in combination with anti-EGFR for mCRC treatment (Table 4).

3.10 Vemurafenib

Vemurafenib is a targeted therapy that specifically inhibits the mutated BRAF form, BRAFV600E. A combination of vemurafenib and panitumumab has been shown to have a 100% tumor shrinkage rate, mPFS of 3.2 months, and mOS of 7.6 months in patients with BRAFV600E MT mCRC (Yaeger et al., 2015). Similarly, RR of 35% and mPFS of 7.7 months was obtained in patients with BRAFV600E MT mCRC treated with a combination of irinotecan, vemurafenib, and cetuximab (Hong et al., 2016). Additionally, the SWOGS1406 study (Kopetz et al., 2021) showed that irinotecan + cetuximab + vemurafenib treatment significantly increased PFS in patients with BRAFV600E MT mCRC compared to patients with BRAFV600E MT mCRC administered treatment regimens without vemurafenib. Moreover, FOLFIRI + cetuximab + vemurafenib treatment achieved an ORR of 81%, mPFS of 9.7 months, and mOS of 15.4 months in patients with BRAFV600E MT mCRC (Wang et al., 2022). Overall, these results confirmed that vemurafenib plus anti-EGFR can achieve significant efficacy in patients with BRAF MT mCRC; however, vemurafenib is yet to be approved for use in mCRC patients owing to shortage of relevant trials and limited number of enrolled patients (Table 4).

3.11 Sotorasib and adagrasib

Sotorasib and adagrasib are potent inhibitors of KRASG12C, specifically designed to target this mutation and act as antineoplastic agents. These agents selectively bind to and inhibit the mutant KRASG12C protein, offering potential therapeutic options for patients with KRASG12C MT cancers. In the phase 1 CodeBreaK100 study (Fakih et al., 2022) involving KRASG12C MT solid tumors, sotorasib treatment resulted in a median mPFS of 4.0 months and an ORR of 7.1% in the mCRC group. In the phase 2 study specifically conducted with mCRC patients, the ORR was 12.9%, with a mOS of 10.6 months and mPFS of 4.0 months (Hong et al., 2020). MRTX849 study (Klempner et al., 2022) showed that adagrasib + cetuximab mPFS was 6.9 vs. 5.6 months in adagrasib only group, and ORR was 46% vs. 19. The aforementioned studies have provided evidence of sotorasib and adagrasib efficacy in the treatment of mCRC. Nevertheless, additional clinical validation is required to further substantiate their practical clinical application.

3.12 Trastuzumab, pertuzumab, lapatinib and tucatinib

Trastuzumab and pertuzumab are monoclonal antibodies that specifically bind to different epitopes of the HER2 receptor, inhibiting HER2 signaling and enhancing immune-mediated destruction of tumor cells. In contrast, lapatinib is a TKI that targets both HER2 and EGFR receptors, effectively blocking their activation and downstream signaling pathways. The HERACLES study (Sartore-Bianchi et al., 2016) showed that trastuzumab + lapatinib treatment achieved an ORR of 30% in patients with KRAS WT and HER2-positive mCRC who previously underwent anti-EGFR therapy. Moreover, trastuzumab + pertuzumab treatment (HER2 antibodies) achieved an ORR of 32%, mPFS of 2.9 months, and mOS of 1.5 months in patients with HER2 positive mCRC (Meric-Bernstam et al., 2019) and an mPFS of 5.3 months in RAS WT subgroup. The HERACLES-B study (Sartore-Bianchi et al., 2020) showed that trastuzumab + panitumumab treatment achieved an ORR of 9.7% and mPFS of 4.1 months in patients with RAS WT and HER2-positive mCRC who had previously been treated with a third-line regimen.

Trastuzumab deruxtecan (DS-8201) is a novel antibody-drug conjugate with a humanized anti-HER2 antibody, cleavable peptide linker, and potent topoisomerase I inhibitor payload that has been confirmed to be effective in multiple solid tumors, including CRC (Tsurutani et al., 2020). The DESTINY-CRC01 study (Siena et al., 2021; Yoshino et al., 2023) treated HER2-positive mCRC patients with DS-8201 after two or more prior regimens. The patients were divided into three cohorts based on HER2 expression levels: cohort A (HER2-positive, IHC 3+ or IHC 2+/ISH+), cohort B (HER2 IHC 2+/ISH-), and cohort C (HER2 IHC 1+). The mOS in cohorts A, B, and C was 15.5, 7.3, and 7.7 months, respectively, while the mPFS was 6.9, 2.1, and 1.4 months, respectively. Notably, the ORR was observed only in cohort A, with a rate of 24%.

Tucatinib blocks proliferation and the phosphorylation of HER2 and its downstream effector, which is a TKI. MOUNTAINEER Trial (Strickler et al., 2022) rolled mCRC patients with RAS WT and HER2-positive which was treatment before but without anti-HER2, mOS was 24.1 months and mPFS was 8.2 months in the tucatinib + trastuzumab. As a result of the remarkable breakthrough in this study, the FDA granted accelerated approval in 2023 for the use of tucatinib + trastuzumab as a second-line treatment in HER2+ and RAS WT mCRC patients.

In light of the research findings mentioned above, the NCCN guidelines recommend trastuzumab in combination with either pertuzumab, lapatinib, tucatinib, or DS-8201 as second-line and beyond treatment options for patients with mCRC who have RAS WT and HER2-positive (Table 4).

4 Conclusion

Recent years have witnessed remarkable advancements in tumor research, driven by breakthroughs in sequencing technologies. These advancements have enhanced our understanding of tumors, including their genetic and genomic alterations. The knowledge provided a basis for personalized treatments, identification of new therapeutic targets, and improved diagnostic methods for different types of cancer. Particularly, the rapid progress in single-cell sequencing technology has revolutionized the field by enabling investigations at individual tumor cell level. This approach has provided unprecedented insights into tumor heterogeneity, allowing us to gain a better understanding of the diverse cellular composition within tumors and identify potential therapeutic targets. Furthermore, it has enhanced our understanding of the tumor microenvironment, including the distinct subpopulations and functions of different cells, as well as their intricate interactions. These advancements have particularly highlighted the crucial role of the tumor microenvironment in studying mechanisms of drug resistance.

In the context of targeted therapy for tumors, significant improvements have been made in terms of CRC survival rates. The current strategies for targeted therapy in mCRC are summarized in Figure 3. However, several challenges remain. Drug resistance remains an inescapable obstacle, as the development of resistance in patients often goes unnoticed until disease progression occurs. Additionally, economic costs pose a major concern, as the need for testing multiple target markers further amplifies the financial burden on patients. Moreover, adverse drug reactions are a significant consideration, especially among vulnerable populations such as the elderly and children, given the varying tolerances to drug dosages. Striking a balance between achieving optimal efficacy and minimizing adverse effects remains an ongoing area of research.

FIGURE 3
www.frontiersin.org

FIGURE 3. The current strategies for molecular targeted therapy in mCRC.

Furthermore, the integration of immunotherapy and molecular targeted therapy has shown potential for further improvement of the survival rates of patients with tumors, including those with mCRC. However, combination of these therapies presents its own set of challenges requiring careful consideration. Co-administration of multiple drugs can potentially intensify adverse reactions, underscoring the importance of identifying the most effective combination regimen through thorough evaluation.

In summary, scientific progress has significantly enhanced our ability to combat CRC and other tumors. While obstacles and unanswered questions remain, the field of targeted therapy continues to advance, and it is anticipated that future clinical trials and research efforts will yield major breakthroughs, further increasing the survival rates and overall outcomes for cancer patients.

Author contributions

SH and JY drafted the manuscript, tables and figures. XG, XH, CL assisted the literature search. RL and YaL assisted the revision. ML, LL, YoL, and JH advised on the manuscript. YaL and RL supervised and finalized the work. All authors contributed to the article and approved the submitted version.

Funding

This research was supported by Guangxi Key Research and Development Program (NO. GUIKEAB19245002), National Natural Science Foundation of China (NO.82103297, NO.82060427), Advanced Innovation Teams and Xinghu Scholars Program of Guangxi Medical University, Guangxi Medical and health key cultivation discipline construction project, Training Program for Thousands of Young and Middle-aged Backbone Teachers in Guangxi Colleges and Universities, Guangxi Medical University Outstanding Young Talents Training Program, Nanning Qingxiu District Science and Technology Project (NO. 2020037, NO. 2020038, NO. 2021007, NO. 2021010, NO. 2021012), Guangxi Medical and health key discipline construction project.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

BSC, best supportive care; CRC, colorectal cancer; CRT, chemoradiotherapy; CR, complete response; Cap, capecitabine; DFS, disease-free survival; EGFR, epidermal growth factor receptor; FDA, United States Food and Drug Administration; HES, hairy and enhancer of split; HER, human epidermal growth factor receptor; mAbs, monoclonal antibodies; MAPK, mitogen-activated protein kinase; mCRC, metastasis colorectal cancer; mOS, median overall survival; mPFS, median progression-free survival; MT, mutation; mTOR, mammalian target of rapamycin; NCCN, National Comprehensive Cancer Network; NICD, Notch intracellular domain; ORR, objective response rate; OS, overall survival; PR, partial response; PI3K, phosphoinositide 3-kinases; RR, response rate; TKI, tyrosine kinase inhibitor; TRK, tropomyosin receptor kinase; TRK, tropomyosin receptor kinase; VEGF, vascular endothelial growth factor; WT, wild-Type; 5-FU, fluorouracil.

References

Alberts, S. R., Sargent, D. J., Nair, S., Mahoney, M. R., Mooney, M., Thibodeau, S. N., et al. (2012). Effect of oxaliplatin, fluorouracil, and leucovorin with or without cetuximab on survival among patients with resected stage III colon cancer: A randomized trial. JAMA 307, 1383–1393. doi:10.1001/jama.2012.385

PubMed Abstract | CrossRef Full Text | Google Scholar

Allegra, C. J., Yothers, G., O'connell, M. J., Sharif, S., Petrelli, N. J., Colangelo, L. H., et al. (2011). Phase III trial assessing bevacizumab in stages II and III carcinoma of the colon: Results of NSABP protocol C-08. J. Clin. Oncol. 29, 11–16. doi:10.1200/JCO.2010.30.0855

PubMed Abstract | CrossRef Full Text | Google Scholar

Amatu, A., Sartore-Bianchi, A., Bencardino, K., Pizzutilo, E. G., Tosi, F., and Siena, S. (2019). Tropomyosin receptor kinase (TRK) biology and the role of NTRK gene fusions in cancer. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 30, viii5–viii15. doi:10.1093/annonc/mdz383

CrossRef Full Text | Google Scholar

André, T., Blons, H., Mabro, M., Chibaudel, B., Bachet, J. B., Tournigand, C., et al. (2013). Panitumumab combined with irinotecan for patients with KRAS wild-type metastatic colorectal cancer refractory to standard chemotherapy: A GERCOR efficacy, tolerance, and translational molecular study. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 24, 412–419. doi:10.1093/annonc/mds465

CrossRef Full Text | Google Scholar

André, T., Vernerey, D., Im, S. A., Bodoky, G., Buzzoni, R., Reingold, S., et al. (2020). Bevacizumab as adjuvant treatment of colon cancer: Updated results from the S-avant phase III study by the GERCOR group. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 31, 246–256. doi:10.1016/j.annonc.2019.12.006

CrossRef Full Text | Google Scholar

Aparicio, T., Bouché, O., Taieb, J., Maillard, E., Kirscher, S., Etienne, P. L., et al. (2018a). Bevacizumab+chemotherapy versus chemotherapy alone in elderly patients with untreated metastatic colorectal cancer: A randomized phase II trial-PRODIGE 20 study results. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 29, 2270–3138. doi:10.1093/annonc/mdx808

CrossRef Full Text | Google Scholar

Aparicio, T., Ghiringhelli, F., Boige, V., Le Malicot, K., Taieb, J., Bouché, O., et al. (2018b). Bevacizumab maintenance versus No maintenance during chemotherapy-free intervals in metastatic colorectal cancer: A randomized phase III trial (PRODIGE 9). J. Clin. Oncol. official J. Am. Soc. Clin. Oncol. 36, 674–681. doi:10.1200/JCO.2017.75.2931

CrossRef Full Text | Google Scholar

Aranda, E., Garcia-Alfonso, P., Benavides, M., Sanchez Ruiz, A., Guillen-Ponce, C., Safont, M. J., et al. (2018). First-line mFOLFOX plus cetuximab followed by mFOLFOX plus cetuximab or single-agent cetuximab as maintenance therapy in patients with metastatic colorectal cancer: Phase II randomised MACRO2 TTD study. Eur. J. Cancer 101, 263–272. doi:10.1016/j.ejca.2018.06.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Arnold, D., Lueza, B., Douillard, J. Y., Peeters, M., Lenz, H. J., Venook, A., et al. (2017). Prognostic and predictive value of primary tumour side in patients with RAS wild-type metastatic colorectal cancer treated with chemotherapy and EGFR directed antibodies in six randomized trials. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 28, 1713–1729. doi:10.1093/annonc/mdx175

CrossRef Full Text | Google Scholar

Bendell, J. C., Bekaii-Saab, T. S., Cohn, A. L., Hurwitz, H. I., Kozloff, M., Tezcan, H., et al. (2012). Treatment patterns and clinical outcomes in patients with metastatic colorectal cancer initially treated with FOLFOX–bevacizumab or FOLFIRI–bevacizumab: Results from ARIES, a bevacizumab observational cohort study. Oncol. 17, 1486–1495. doi:10.1634/theoncologist.2012-0190

CrossRef Full Text | Google Scholar

Bennouna, J., Hiret, S., Bertaut, A., Bouche, O., Deplanque, G., Borel, C., et al. (2019). Continuation of bevacizumab vs cetuximab plus chemotherapy after first progression in KRAS wild-type metastatic colorectal cancer: The UNICANCER PRODIGE18 randomized clinical trial. JAMA Oncol. 5, 83–90. doi:10.1001/jamaoncol.2018.4465

PubMed Abstract | CrossRef Full Text | Google Scholar

Bennouna, J., Sastre, J., Arnold, D., Österlund, P., Greil, R., Van Cutsem, E., et al. (2013). Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): A randomised phase 3 trial. Lancet Oncol. 14, 29–37. doi:10.1016/S1470-2045(12)70477-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Boeckx, N., Koukakis, R., Op De Beeck, K., Rolfo, C., Van Camp, G., Siena, S., et al. (2017). Primary tumor sidedness has an impact on prognosis and treatment outcome in metastatic colorectal cancer: Results from two randomized first-line panitumumab studies. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 28, 1862–1868. doi:10.1093/annonc/mdx119

CrossRef Full Text | Google Scholar

Bry, M., Kivelä, R., Leppänen, V.-M., and Alitalo, K. (2014). Vascular endothelial growth factor-B in physiology and disease. Physiol. Rev. 94, 779–794. doi:10.1152/physrev.00028.2013

PubMed Abstract | CrossRef Full Text | Google Scholar

Ciardiello, F., Normanno, N., Martinelli, E., Troiani, T., Pisconti, S., Cardone, C., et al. (2016). Cetuximab continuation after first progression in metastatic colorectal cancer (CAPRI-GOIM): A randomized phase II trial of FOLFOX plus cetuximab versus FOLFOX. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 27, 1055–1061. doi:10.1093/annonc/mdw136

CrossRef Full Text | Google Scholar

Corcoran, R. B., Andre, T., Atreya, C. E., Schellens, J. H. M., Yoshino, T., Bendell, J. C., et al. (2018). Combined BRAF, EGFR, and MEK inhibition in patients with BRAF(V600e)-mutant colorectal cancer. Cancer Discov. 8, 428–443. doi:10.1158/2159-8290.CD-17-1226

PubMed Abstract | CrossRef Full Text | Google Scholar

Corcoran, R. B., Atreya, C. E., Falchook, G. S., Kwak, E. L., Ryan, D. P., Bendell, J. C., et al. (2015). Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAF V600-mutant colorectal cancer. J. Clin. Oncol. 33, 4023–4031. doi:10.1200/JCO.2015.63.2471

PubMed Abstract | CrossRef Full Text | Google Scholar

Cremolini, C., Antoniotti, C., Lonardi, S., Aprile, G., Bergamo, F., Masi, G., et al. (2018a). Activity and safety of cetuximab plus modified FOLFOXIRI followed by maintenance with cetuximab or bevacizumab for RAS and BRAF wild-type metastatic colorectal cancer: A randomized phase 2 clinical trial. JAMA Oncol. 4, 529–536. doi:10.1001/jamaoncol.2017.5314

PubMed Abstract | CrossRef Full Text | Google Scholar

Cremolini, C., Antoniotti, C., Lonardi, S., Bergamo, F., Cortesi, E., Tomasello, G., et al. (2018b). Primary tumor sidedness and benefit from FOLFOXIRI plus bevacizumab as initial therapy for metastatic colorectal cancer. Retrospective analysis of the TRIBE trial by GONO. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 29, 1528–1534. doi:10.1093/annonc/mdy140

CrossRef Full Text | Google Scholar

Cremolini, C., Antoniotti, C., Rossini, D., Lonardi, S., Loupakis, F., Pietrantonio, F., et al. (2020). Upfront FOLFOXIRI plus bevacizumab and reintroduction after progression versus mFOLFOX6 plus bevacizumab followed by FOLFIRI plus bevacizumab in the treatment of patients with metastatic colorectal cancer (TRIBE2): A multicentre, open-label, phase 3, randomised, controlled trial. Lancet. Oncol. 21, 497–507. doi:10.1016/S1470-2045(19)30862-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunningham, D., Humblet, Y., Siena, S., Khayat, D., Bleiberg, H., Santoro, A., et al. (2004). Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N. Engl. J. Med. 351, 337–345. doi:10.1056/NEJMoa033025

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunningham, D., Lang, I., Marcuello, E., Lorusso, V., Ocvirk, J., Shin, D. B., et al. (2013). 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. 14, 1077–1085. doi:10.1016/S1470-2045(13)70154-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Dasari, N. A., Lonardi, S., Garcia-Carbonero, R., Fernandez, M. E. E., Yoshino, T., Sobrero, A. F., et al. (2022). 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. 33, S1391–S1392. doi:10.1016/j.annonc.2022.08.021

CrossRef Full Text | Google Scholar

De Gramont, A., Van Cutsem, E., Schmoll, H.-J., Tabernero, J., Clarke, S., Moore, M. J., et al. (2012). Bevacizumab plus oxaliplatin-based chemotherapy as adjuvant treatment for colon cancer (AVANT): A phase 3 randomised controlled trial. Lancet. Oncol. 13, 1225–1233. doi:10.1016/S1470-2045(12)70509-0

PubMed Abstract | CrossRef Full Text | Google Scholar

De Luca, A., Carotenuto, A., Rachiglio, A., Gallo, M., Maiello, M. R., Aldinucci, D., et al. (2008). The role of the EGFR signaling in tumor microenvironment. J. Cell. Physiology 214, 559–567. doi:10.1002/jcp.21260

CrossRef Full Text | Google Scholar

De Roock, W., De Vriendt, V., Normanno, N., Ciardiello, F., and Tejpar, S. (2011). KRAS, BRAF, PIK3CA, and PTEN mutations: Implications for targeted therapies in metastatic colorectal cancer. Lancet. Oncol. 12, 594–603. doi:10.1016/S1470-2045(10)70209-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Dewdney, A., Cunningham, D., Tabernero, J., Capdevila, J., Glimelius, B., Cervantes, A., et al. (2012). Multicenter randomized phase II clinical trial comparing neoadjuvant oxaliplatin, capecitabine, and preoperative radiotherapy with or without cetuximab followed by total mesorectal excision in patients with high-risk rectal cancer (EXPERT-C). J. Clin. Oncol. official J. Am. Soc. Clin. Oncol. 30, 1620–1627. doi:10.1200/JCO.2011.39.6036

CrossRef Full Text | Google Scholar

Douillard, J. Y., Oliner, K. S., Siena, S., Tabernero, J., Burkes, R., Barugel, M., et al. (2013). Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N. Engl. J. Med. 369, 1023–1034. doi:10.1056/NEJMoa1305275

PubMed Abstract | CrossRef Full Text | Google Scholar

Douillard, J. Y., Siena, S., Cassidy, J., Tabernero, J., Burkes, R., Barugel, M., et al. (2010). Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: The PRIME study. J. Clin. Oncol. 28, 4697–4705. doi:10.1200/JCO.2009.27.4860

PubMed Abstract | CrossRef Full Text | Google Scholar

Downward, J. (2003). Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer 3, 11–22. doi:10.1038/nrc969

PubMed Abstract | CrossRef Full Text | Google Scholar

Duchartre, Y., Kim, Y.-M., and Kahn, M. (2016). The Wnt signaling pathway in cancer. Crit. Rev. Oncology/hematology 99, 141–149. doi:10.1016/j.critrevonc.2015.12.005

CrossRef Full Text | Google Scholar

Dvorak, H. F. (2002). Vascular permeability factor/vascular endothelial growth factor: A critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J. Clin. Oncol. official J. Am. Soc. Clin. Oncol. 20, 4368–4380. doi:10.1200/JCO.2002.10.088

CrossRef Full Text | Google Scholar

Fakih, M. G., Kopetz, S., Kuboki, Y., Kim, T. W., Munster, P. N., Krauss, J. C., et al. (2022). Sotorasib for previously treated colorectal cancers with KRASG12C mutation (CodeBreaK100): A prespecified analysis of a single-arm, phase 2 trial. Lancet. Oncol. 23, 115–124. doi:10.1016/S1470-2045(21)00605-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferrara, N., Gerber, H.-P., and Lecouter, J. (2003). The biology of VEGF and its receptors. Nat. Med. 9, 669–676. doi:10.1038/nm0603-669

PubMed Abstract | CrossRef Full Text | Google Scholar

Folprecht, G., Pericay, C., Saunders, M. P., Thomas, A., Lopez Lopez, R., Roh, J. K., et al. (2016). 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. 27, 1273–1279. doi:10.1093/annonc/mdw176

PubMed Abstract | CrossRef Full Text | Google Scholar

Fornaro, L., Lonardi, S., Masi, G., Loupakis, F., Bergamo, F., Salvatore, L., et al. (2013). FOLFOXIRI in combination with panitumumab as first-line treatment in quadruple wild-type (KRAS, NRAS, HRAS, BRAF) metastatic colorectal cancer patients: A phase II trial by the gruppo oncologico nord ovest (GONO). Ann. Oncol. official J. Eur. Soc. Med. Oncol. 24, 2062–2067. doi:10.1093/annonc/mdt165

CrossRef Full Text | Google Scholar

Fruman, D. A., and Rommel, C. (2014). PI3K and cancer: Lessons, challenges and opportunities. Nat. Rev. Drug Discov. 13, 140–156. doi:10.1038/nrd4204

PubMed Abstract | CrossRef Full Text | Google Scholar

García-Alfonso, P., Muñoz Martín, A. J., Ortega Morán, L., Soto Alsar, J., Torres Pérez-Solero, G., Blanco Codesido, M., et al. (2021). Oral drugs in the treatment of metastatic colorectal cancer. Ther. Adv. Med. Oncol. 13, 17588359211009001. doi:10.1177/17588359211009001

PubMed Abstract | CrossRef Full Text | Google Scholar

Giantonio, B. J., Catalano, P. J., Meropol, N. J., O'dwyer, P. J., Mitchell, E. P., Alberts, S. R., et al. (2007). Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: Results from the eastern cooperative oncology group study E3200. J. Clin. Oncol. 25, 1539–1544. doi:10.1200/JCO.2006.09.6305

PubMed Abstract | CrossRef Full Text | Google Scholar

Glimelius, B., Stintzing, S., Marshall, J., Yoshino, T., and De Gramont, A. (2021). Metastatic colorectal cancer: Advances in the folate-fluoropyrimidine chemotherapy backbone. Cancer Treat. Rev. 98, 102218. doi:10.1016/j.ctrv.2021.102218

PubMed Abstract | CrossRef Full Text | Google Scholar

Grothey, A., Cutsem, E. V., Sobrero, A., Siena, S., Falcone, A., Ychou, M., et al. (2013). Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): An international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 381, 303–312. doi:10.1016/S0140-6736(12)61900-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Grothey, A., Sugrue, M. M., Purdie, D. M., Dong, W., Sargent, D., Hedrick, E., et al. (2008). Bevacizumab beyond first progression is associated with prolonged overall survival in metastatic colorectal cancer: Results from a large observational cohort study (BRiTE). J. Clin. Oncol. Official J. Am. Soc. Clin. Oncol. 26, 5326–5334. doi:10.1200/JCO.2008.16.3212

CrossRef Full Text | Google Scholar

Hatzivassiliou, G., Haling, J. R., Chen, H., Song, K., Price, S., Heald, R., et al. (2013). Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature 501, 232–236. doi:10.1038/nature12441

PubMed Abstract | CrossRef Full Text | Google Scholar

Hecht, J. R., Patnaik, A., Berlin, J., Venook, A., Malik, I., Tchekmedyian, S., et al. (2007). Panitumumab monotherapy in patients with previously treated metastatic colorectal cancer. Cancer 110, 980–988. doi:10.1002/cncr.22915

PubMed Abstract | CrossRef Full Text | Google Scholar

Hegewisch-Becker, S., Graeven, U., Lerchenmüller, C. A., Killing, B., Depenbusch, R., Steffens, C.-C., et al. (2015). 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. 16, 1355–1369. doi:10.1016/S1470-2045(15)00042-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Heinemann, V., Von Weikersthal, L. F., Decker, T., Kiani, A., Vehling-Kaiser, U., Al-Batran, S.-E., et al. (2014). 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. 15, 1065–1075. doi:10.1016/S1470-2045(14)70330-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, D. S., Fakih, M. G., Strickler, J. H., Desai, J., Durm, G. A., Shapiro, G. I., et al. (2020). KRASG12C inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med. 383, 1207–1217. doi:10.1056/NEJMoa1917239

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, D. S., Morris, V. K., El Osta, B., Sorokin, A. V., Janku, F., Fu, S., et al. (2016). Phase IB study of vemurafenib in combination with irinotecan and cetuximab in patients with metastatic colorectal cancer with BRAFV600E mutation. Cancer Discov. 6, 1352–1365. doi:10.1158/2159-8290.CD-16-0050

PubMed Abstract | CrossRef Full Text | Google Scholar

Howlader N, N. A., and Krapcho, M., Miller, D., Brest, A, , Yu, M., Ruhl, J., et al. (2020). SEER cancer Statistics review (Bethesda, MD: National Cancer Institute). https://seer.cancer.gov/csr/1975_2018/.based on November 2020 SEER data submission, posted to the SEER web site, April 2021. [Online]. [Accessed]

Google Scholar

Hurwitz, H., Fehrenbacher, L., Novotny, W., Cartwright, T., Hainsworth, J., Heim, W., et al. (2004). Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 350, 2335–2342. doi:10.1056/NEJMoa032691

PubMed Abstract | CrossRef Full Text | Google Scholar

Hurwitz, H. I., Bekaii-Saab, T. S., Bendell, J. C., Cohn, A. L., Kozloff, M., Roach, N., et al. (2014). Safety and effectiveness of bevacizumab treatment for metastatic colorectal cancer: Final results from the Avastin® registry – investigation of effectiveness and safety (ARIES) observational cohort study. Clin. Oncol. 26, 323–332. doi:10.1016/j.clon.2014.03.001

CrossRef Full Text | Google Scholar

Ivy, S. P., Wick, J. Y., and Kaufman, B. M. (2009). An overview of small-molecule inhibitors of VEGFR signaling. Nat. Rev. Clin. Oncol. 6, 569–579. doi:10.1038/nrclinonc.2009.130

PubMed Abstract | CrossRef Full Text | Google Scholar

Janku, F., Yap, T. A., and Meric-Bernstam, F. (2018). Targeting the PI3K pathway in cancer: Are we making headway? Nat. Rev. Clin. Oncol. 15, 273–291. doi:10.1038/nrclinonc.2018.28

PubMed Abstract | CrossRef Full Text | Google Scholar

Jonker, D. J., O'callaghan, C. J., Karapetis, C. S., Zalcberg, J. R., Tu, D., Au, H.-J., et al. (2007). Cetuximab for the treatment of colorectal cancer. N. Engl. J. Med. 357, 2040–2048. doi:10.1056/NEJMoa071834

PubMed Abstract | CrossRef Full Text | Google Scholar

Karkkainen, M. J., Mäkinen, T., and Alitalo, K. (2002). Lymphatic endothelium: A new frontier of metastasis research. Nat. Cell Biol. 4, E2–E5. doi:10.1038/ncb0102-e2

PubMed Abstract | CrossRef Full Text | Google Scholar

Karnoub, A. E., and Weinberg, R. A. (2008). Ras oncogenes: Split personalities. Nat. Rev. Mol. cell Biol. 9, 517–531. doi:10.1038/nrm2438

PubMed Abstract | CrossRef Full Text | Google Scholar

Kerr, R. S., Love, S., Segelov, E., Johnstone, E., Falcon, B., Hewett, P., et al. (2016). Adjuvant capecitabine plus bevacizumab versus capecitabine alone in patients with colorectal cancer (QUASAR 2): An open-label, randomised phase 3 trial. Lancet. Oncol. 17, 1543–1557. doi:10.1016/S1470-2045(16)30172-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, T. W., Elme, A., Kusic, Z., Park, J. O., Udrea, A. A., Kim, S. Y., et al. (2016). A phase 3 trial evaluating panitumumab plus best supportive care vs best supportive care in chemorefractory wild-type KRAS or RAS metastatic colorectal cancer. Br. J. Cancer 115, 1206–1214. doi:10.1038/bjc.2016.309

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, T. W., Elme, A., Park, J. O., Udrea, A. A., Kim, S. Y., Ahn, J. B., et al. (2018). Final analysis of outcomes and RAS/BRAF status in a randomized phase 3 study of panitumumab and best supportive care in chemorefractory wild type KRAS metastatic colorectal cancer. Clin. Colorectal Cancer 17, 206–214. doi:10.1016/j.clcc.2018.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Klempner, S. J., Weiss, J., Pelster, M., Spira, A., Barve, M., Ou, S. H. I., et al. (2022). LBA24 KRYSTAL-1: Updated efficacy and safety of adagrasib (MRTX849) with or without cetuximab in patients with advanced colorectal cancer (CRC) harboring a KRASG12C mutation. Ann. Oncol. 33, S1391. doi:10.1016/j.annonc.2022.08.020

CrossRef Full Text | Google Scholar

Kopetz, S., Grothey, A., Yaeger, R., Van Cutsem, E., Desai, J., Yoshino, T., et al. (2019). Encorafenib, binimetinib, and cetuximab in BRAF V600e-mutated colorectal cancer. N. Engl. J. Med. 381, 1632–1643. doi:10.1056/NEJMoa1908075

PubMed Abstract | CrossRef Full Text | Google Scholar

Kopetz, S., Guthrie, K. A., Morris, V. K., Lenz, H.-J., Magliocco, A. M., Maru, D., et al. (2021). Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG S1406). J. Clin. Oncol. Official J. Am. Soc. Clin. Oncol. 39, 285–294. doi:10.1200/JCO.20.01994

CrossRef Full Text | Google Scholar

Kumagai, S., Koyama, S., and Nishikawa, H. (2021). Antitumour immunity regulated by aberrant ERBB family signalling. Nat. Rev. Cancer 21, 181–197. doi:10.1038/s41568-020-00322-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Qin, S., Xu, R. H., Shen, L., Xu, J., Bai, Y., et al. (2018). Effect of fruquintinib vs placebo on overall survival in patients with previously treated metastatic colorectal cancer: The FRESCO randomized clinical trial. JAMA 319, 2486–2496. doi:10.1001/jama.2018.7855

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Qin, S., Xu, R., Yau, T. C. C., Ma, B., Pan, H., et al. (2015). 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. 16, 619–629. doi:10.1016/S1470-2045(15)70156-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Yan, X., Wang, Y., Kaur, B., Han, H., and Yu, J. (2023). The Notch signaling pathway: A potential target for cancer immunotherapy. J. Hematol. Oncol. 16, 45. doi:10.1186/s13045-023-01439-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Loupakis, F., Cremolini, C., Antoniotti, C., Lonardi, S., Ronzoni, M., Zaniboni, A., et al. (2015). FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as initial treatment for metastatic colorectal cancer (TRIBE study): Updated survival results and final molecular subgroups analyses. J. Clin. Oncol. 33, 3510. doi:10.1200/jco.2015.33.15_suppl.3510

CrossRef Full Text | Google Scholar

Martin-Zanca, D., Hughes, S. H., and Barbacid, M. (1986). A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature 319, 743–748. doi:10.1038/319743a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Masi, G., Vivaldi, C., Fornaro, L., Lonardi, S., Buccianti, P., Sainato, A., et al. (2019). Total neoadjuvant approach with FOLFOXIRI plus bevacizumab followed by chemoradiotherapy plus bevacizumab in locally advanced rectal cancer: The TRUST trial. Eur. J. Cancer 110, 32–41. doi:10.1016/j.ejca.2019.01.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Masuishi, T., Tsuji, A., Kotaka, M., Nakamura, M., Kochi, M., Takagane, A., et al. (2020). Phase 2 study of irinotecan plus cetuximab rechallenge as third-line treatment in KRAS wild-type metastatic colorectal cancer: JACCRO CC-08. Br. J. Cancer 123, 1490–1495. doi:10.1038/s41416-020-01042-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Maughan, T. S., Adams, R. A., Smith, C. G., Meade, A. M., Seymour, M. T., Wilson, R. H., et al. (2011). Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: Results of the randomised phase 3 MRC COIN trial. Lancet 377, 2103–2114. doi:10.1016/S0140-6736(11)60613-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Mauri, G., Pizzutilo, E. G., Amatu, A., Bencardino, K., Palmeri, L., Bonazzina, E. F., et al. (2019). Retreatment with anti-EGFR monoclonal antibodies in metastatic colorectal cancer: Systematic review of different strategies. Cancer Treat. Rev. 73, 41–53. doi:10.1016/j.ctrv.2018.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Ménard, S., Pupa, S. M., Campiglio, M., and Tagliabue, E. (2003). Biologic and therapeutic role of HER2 in cancer. Oncogene 22, 6570–6578. doi:10.1038/sj.onc.1206779

PubMed Abstract | CrossRef Full Text | Google Scholar

Meric-Bernstam, F., Hurwitz, H., Raghav, K. P. S., Mcwilliams, R. R., Fakih, M., Vanderwalde, A., et al. (2019). Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): An updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 20, 518–530. doi:10.1016/S1470-2045(18)30904-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Modest, D. P., Karthaus, M., Fruehauf, S., Graeven, U., Müller, L., König, A. O., et al. (2022). Panitumumab plus fluorouracil and folinic acid versus fluorouracil and folinic acid alone as maintenance therapy in RAS wild-type metastatic colorectal cancer: The randomized Panama trial (AIO KRK 0212). J. Clin. Oncol. 40, 72–82. doi:10.1200/JCO.21.01332

PubMed Abstract | CrossRef Full Text | Google Scholar

Modest, D. P., Martens, U. M., Riera-Knorrenschild, J., Greeve, J., Florschütz, A., Wessendorf, S., et al. (2019a). FOLFOXIRI plus panitumumab as first-line treatment of RAS wild-type metastatic colorectal cancer: The randomized, open-label, phase II VOLFI study (AIO KRK0109). J. Clin. Oncol. 37, 3401–3411. doi:10.1200/JCO.19.01340

PubMed Abstract | CrossRef Full Text | Google Scholar

Modest, D. P., Rivera, F., Bachet, J.-B., De Braud, F., Pietrantonio, F., Koukakis, R., et al. (2019b). Panitumumab-based maintenance after oxaliplatin discontinuation in metastatic colorectal cancer: A retrospective analysis of two randomised trials. Int. J. cancer 145, 576–585. doi:10.1002/ijc.32110

PubMed Abstract | CrossRef Full Text | Google Scholar

Morris, E. J., Jha, S., Restaino, C. R., Dayananth, P., Zhu, H., Cooper, A., et al. (2013). Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 3, 742–750. doi:10.1158/2159-8290.CD-13-0070

PubMed Abstract | CrossRef Full Text | Google Scholar

Munemoto, Y., Nakamura, M., Takahashi, M., Kotaka, M., Kuroda, H., Kato, T., et al. (2019). Sapphire: A randomised phase II study of planned discontinuation or continuous treatment of oxaliplatin after six cycles of modified FOLFOX6 plus panitumumab in patients with colorectal cancer. Eur. J. Cancer (Oxford, Engl. 1990) 119, 158–167. doi:10.1016/j.ejca.2019.07.006

CrossRef Full Text | Google Scholar

NTRK (2019). Entrectinib effective across NTRK fusion-positive cancers. Cancer Discov. 9, OF4. doi:10.1158/2159-8290.CD-NB2018-156

PubMed Abstract | CrossRef Full Text | Google Scholar

Pakneshan, S., Salajegheh, A., Smith, R. A., and Lam, A. K.-Y. (2013). Clinicopathological relevance of BRAF mutations in human cancer. Pathology 45, 346–356. doi:10.1097/PAT.0b013e328360b61d

PubMed Abstract | CrossRef Full Text | Google Scholar

Parikh, A. R., Lee, F.-C., Yau, L., Koh, H., Knost, J., Mitchell, E. P., et al. (2019). 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. official J. Am. Assoc. Cancer Res. 25, 2988–2995. doi:10.1158/1078-0432.CCR-18-1221

CrossRef Full Text | Google Scholar

Peeters, M., Price, T. J., Cervantes, A., Sobrero, A. F., Ducreux, M., Hotko, Y., et al. (2014). Final results from a randomized phase 3 study of FOLFIRI {+/-} panitumumab for second-line treatment of metastatic colorectal cancer. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 25, 107–116. doi:10.1093/annonc/mdt523

CrossRef Full Text | Google Scholar

Peeters, M., Price, T. J., Cervantes, A., Sobrero, A. F., Ducreux, M., Hotko, Y., et al. (2010). Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. J. Clin. Oncol. 28, 4706–4713. doi:10.1200/JCO.2009.27.6055

PubMed Abstract | CrossRef Full Text | Google Scholar

Peeters, M., Price, T., Taieb, J., Geissler, M., Rivera, F., Canon, J.-L., et al. (2018). Relationships between tumour response and primary tumour location, and predictors of long-term survival, in patients with RAS wild-type metastatic colorectal cancer receiving first-line panitumumab therapy: Retrospective analyses of the PRIME and PEAK clinical trials. Br. J. cancer 119, 303–312. doi:10.1038/s41416-018-0165-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Pfeiffer, P., Yilmaz, M., Möller, S., Zitnjak, D., Krogh, M., Petersen, L. N., et al. (2020). TAS-102 with or without bevacizumab in patients with chemorefractory metastatic colorectal cancer: An investigator-initiated, open-label, randomised, phase 2 trial. Lancet Oncol. 21, 412–420. doi:10.1016/S1470-2045(19)30827-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Pietrantonio, F., Morano, F., Corallo, S., Miceli, R., Lonardi, S., Raimondi, A., et al. (2019). Maintenance therapy with panitumumab alone vs panitumumab plus fluorouracil-leucovorin in patients with RAS wild-type metastatic colorectal cancer: A phase 2 randomized clinical trial. JAMA Oncol. 5, 1268–1275. doi:10.1001/jamaoncol.2019.1467

PubMed Abstract | CrossRef Full Text | Google Scholar

Polivka, J., and Janku, F. (2014). Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol. Ther. 142, 164–175. doi:10.1016/j.pharmthera.2013.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Price, T. J., Peeters, M., Kim, T. W., Li, J., Cascinu, S., Ruff, P., et al. (2014). Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): A randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol. 15, 569–579. doi:10.1016/S1470-2045(14)70118-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, S., Li, J., Wang, L., Xu, J., Cheng, Y., Bai, Y., et al. (2018). Efficacy and Tolerability of first-line cetuximab plus leucovorin, fluorouracil, and oxaliplatin (FOLFOX-4) versus FOLFOX-4 in patients with RAS wild-type metastatic colorectal cancer: The open-label, randomized, phase III TAILOR trial. J. Clin. Oncol. official J. Am. Soc. Clin. Oncol. 36, 3031–3039. doi:10.1200/JCO.2018.78.3183

CrossRef Full Text | Google Scholar

Rodon, J., Argilés, G., Connolly, R. M., Vaishampayan, U., De Jonge, M., Garralda, E., et al. (2021a). Phase 1 study of single-agent WNT974, a first-in-class Porcupine inhibitor, in patients with advanced solid tumours. Br. J. Cancer 125, 28–37. doi:10.1038/s41416-021-01389-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodon, J., Argilés, G., Connolly, R. M., Vaishampayan, U., De Jonge, M., Garralda, E., et al. (2021b). Phase 1 study of single-agent WNT974, a first-in-class Porcupine inhibitor, in patients with advanced solid tumours. Br. J. Cancer 125, 28–37. doi:10.1038/s41416-021-01389-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Roskoski, R. (2014). The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol. Res. 79, 34–74. doi:10.1016/j.phrs.2013.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Sakai, D., Taniguchi, H., Sugimoto, N., Tamura, T., Nishina, T., Hara, H., et al. (2020). Randomised phase II study of panitumumab plus irinotecan versus cetuximab plus irinotecan in patients with KRAS wild-type metastatic colorectal cancer refractory to fluoropyrimidine, irinotecan and oxaliplatin (WJOG 6510G). Eur. J. cancer 135, 11–21. doi:10.1016/j.ejca.2020.04.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Saltz, L. B., Clarke, S., Diaz-Rubio, E., Scheithauer, W., Figer, A., Wong, R., et al. (2008). Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: A randomized phase III study. J. Clin. Oncol. 26, 2013–2019. doi:10.1200/JCO.2007.14.9930

PubMed Abstract | CrossRef Full Text | Google Scholar

Santini, D., Vincenzi, B., Addeo, R., Garufi, C., Masi, G., Scartozzi, M., et al. (2012). Cetuximab rechallenge in metastatic colorectal cancer patients: How to come away from acquired resistance? Ann. Oncol. 23, 2313–2318. doi:10.1093/annonc/mdr623

PubMed Abstract | CrossRef Full Text | Google Scholar

Sartore-Bianchi, A., Lonardi, S., Martino, C., Fenocchio, E., Tosi, F., Ghezzi, S., et al. (2020). Pertuzumab and trastuzumab emtansine in patients with HER2-amplified metastatic colorectal cancer: The phase II HERACLES-B trial. ESMO Open 5, e000911. doi:10.1136/esmoopen-2020-000911

PubMed Abstract | CrossRef Full Text | Google Scholar

Sartore-Bianchi, A., Trusolino, L., Martino, C., Bencardino, K., Lonardi, S., Bergamo, F., et al. (2016). Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): A proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 17, 738–746. doi:10.1016/S1470-2045(16)00150-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwartzberg, L. S., Rivera, F., Karthaus, M., Fasola, G., Canon, J. L., Hecht, J. R., et al. (2014). 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. 32, 2240–2247. doi:10.1200/JCO.2013.53.2473

PubMed Abstract | CrossRef Full Text | Google Scholar

Seymour, M. T., Brown, S. R., Middleton, G., Maughan, T., Richman, S., Gwyther, S., et al. (2013). Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced colorectal cancer (PICCOLO): A prospectively stratified randomised trial. Lancet Oncol. 14, 749–759. doi:10.1016/S1470-2045(13)70163-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Siebel, C., and Lendahl, U. (2017). Notch signaling in development, tissue homeostasis, and disease. Physiol. Rev. 97, 1235–1294. doi:10.1152/physrev.00005.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Siegel, R. L., Miller, K. D., Fuchs, H. E., and Jemal, A. (2022). Cancer statistics, 2022. A Cancer J. Clin. 72, 7–33. doi:10.3322/caac.21708

CrossRef Full Text | Google Scholar

Siena, S., Di Bartolomeo, M., Raghav, K., Masuishi, T., Loupakis, F., Kawakami, H., et al. (2021). Trastuzumab deruxtecan (DS-8201) in patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): A multicentre, open-label, phase 2 trial. Lancet Oncol. 22, 779–789. doi:10.1016/S1470-2045(21)00086-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Simkens, L. H. J., Van Tinteren, H., May, A., Ten Tije, A. J., Creemers, G.-J. M., Loosveld, O. J. L., et al. (2015). Maintenance treatment with capecitabine and bevacizumab in metastatic colorectal cancer (CAIRO3): A phase 3 randomised controlled trial of the Dutch colorectal cancer group. Lancet 385, 1843–1852. doi:10.1016/S0140-6736(14)62004-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Sobrero, A. F., Maurel, J., Fehrenbacher, L., Scheithauer, W., Abubakr, Y. A., Lutz, M. P., et al. (2008). Epic: Phase III trial of cetuximab plus irinotecan after fluoropyrimidine and oxaliplatin failure in patients with metastatic colorectal cancer. J. Clin. Oncol. 26, 2311–2319. doi:10.1200/JCO.2007.13.1193

PubMed Abstract | CrossRef Full Text | Google Scholar

Solomon, J. P., Benayed, R., Hechtman, J. F., and Ladanyi, M. (2019). Identifying patients with NTRK fusion cancer. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 30, viii16–viii22. doi:10.1093/annonc/mdz384

CrossRef Full Text | Google Scholar

Strickler, J., Cercek, A., Siena, S., André, T., Ng, K., Van Cutsem, E., et al. (2022). LBA-2 primary analysis of MOUNTAINEER: A phase 2 study of tucatinib and trastuzumab for HER2-positive mCRC. Ann. Oncol. 33, S375–S376. doi:10.1016/j.annonc.2022.04.440

CrossRef Full Text | Google Scholar

Tabernero, J., Grothey, A., Van Cutsem, E., Yaeger, R., Wasan, H., Yoshino, T., et al. (2021). Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600e-mutant metastatic colorectal cancer: Updated survival results and Subgroup analyses from the BEACON study. J. Clin. Oncol. official J. Am. Soc. Clin. Oncol. 39, 273–284. doi:10.1200/JCO.20.02088

CrossRef Full Text | Google Scholar

Tabernero, J., Yoshino, T., Cohn, A. L., Obermannova, R., Bodoky, G., Garcia-Carbonero, R., et al. (2015). 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. 16, 499–508. doi:10.1016/S1470-2045(15)70127-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Taieb, J., Tabernero, J., Mini, E., Subtil, F., Folprecht, G., Van Laethem, J.-L., et al. (2014). Oxaliplatin, fluorouracil, and leucovorin with or without cetuximab in patients with resected stage III colon cancer (PETACC-8): An open-label, randomised phase 3 trial. Lancet Oncol. 15, 862–873. doi:10.1016/S1470-2045(14)70227-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Tejpar, S., Stintzing, S., Ciardiello, F., Tabernero, J., Van Cutsem, E., Beier, F., et al. (2017). Prognostic and predictive relevance of primary tumor location in patients with RAS wild-type metastatic colorectal cancer: Retrospective analyses of the CRYSTAL and FIRE-3 trials. JAMA Oncol. 3, 194–201. doi:10.1001/jamaoncol.2016.3797

PubMed Abstract | CrossRef Full Text | Google Scholar

Tol, J., Koopman, M., Cats, A., Rodenburg, C. J., Creemers, G. J. M., Schrama, J. G., et al. (2009). Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. N. Engl. J. Med. 360, 563–572. doi:10.1056/NEJMoa0808268

PubMed Abstract | CrossRef Full Text | Google Scholar

Tonini, G., Imperatori, M., Vincenzi, B., Frezza, A. M., and Santini, D. (2013). Rechallenge therapy and treatment holiday: Different strategies in management of metastatic colorectal cancer. J. Exp. Clin. cancer Res. CR 32, 92. doi:10.1186/1756-9966-32-92

PubMed Abstract | CrossRef Full Text | Google Scholar

Tournigand, C., André, T., Achille, E., Lledo, G., Flesh, M., Mery-Mignard, D., et al. (2004). FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: A randomized GERCOR study. J. Clin. Oncol. 22, 229–237. doi:10.1200/JCO.2004.05.113

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsurutani, J., Iwata, H., Krop, I., Janne, P. A., Doi, T., Takahashi, S., et al. (2020). Targeting HER2 with trastuzumab deruxtecan: A dose-expansion, phase I study in multiple advanced solid tumors. Cancer Discov. 10, 688–701. doi:10.1158/2159-8290.CD-19-1014

PubMed Abstract | CrossRef Full Text | Google Scholar

Tveit, K. M., Guren, T., Glimelius, B., Pfeiffer, P., Sorbye, H., Pyrhonen, S., et al. (2012). Phase III trial of cetuximab with continuous or intermittent fluorouracil, leucovorin, and oxaliplatin (nordic FLOX) versus FLOX alone in first-line treatment of metastatic colorectal cancer: The NORDIC-VII study. J. Clin. Oncol. 30, 1755–1762. doi:10.1200/JCO.2011.38.0915

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaishnavi, A., Le, A. T., and Doebele, R. C. (2015). TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 5, 25–34. doi:10.1158/2159-8290.CD-14-0765

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Cutsem, E., Cervantes, A., Adam, R., Sobrero, A., Van Krieken, J. H., Aderka, D., et al. (2016). ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. Official J. Eur. Soc. Med. Oncol. 27, 1386–1422. doi:10.1093/annonc/mdw235

CrossRef Full Text | Google Scholar

Van Cutsem, E., Cervantes, A., Nordlinger, B., and Arnold, D. (2014). Metastatic colorectal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 25, iii1–iii9. doi:10.1093/annonc/mdu260

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Cutsem, E., Köhne, C.-H., Hitre, E., Zaluski, J., Chang Chien, C.-R., Makhson, A., et al. (2009). Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N. Engl. J. Med. 360, 1408–1417. doi:10.1056/NEJMoa0805019

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Cutsem, E., Peeters, M., Siena, S., Humblet, Y., Hendlisz, A., Neyns, B., et al. (2007). Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J. Clin. Oncol. 25, 1658–1664. doi:10.1200/JCO.2006.08.1620

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Cutsem, E., Siena, S., Humblet, Y., Canon, J. L., Maurel, J., Bajetta, E., et al. (2008). An open-label, single-arm study assessing safety and efficacy of panitumumab in patients with metastatic colorectal cancer refractory to standard chemotherapy. Ann. Oncol. official J. Eur. Soc. Med. Oncol. 19, 92–98. doi:10.1093/annonc/mdm399

CrossRef Full Text | Google Scholar

Van Cutsem, E., Tabernero, J., Lakomy, R., Prenen, H., Prausova, J., Macarulla, T., et al. (2012). 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. 30, 3499–3506. doi:10.1200/JCO.2012.42.8201

PubMed Abstract | CrossRef Full Text | Google Scholar

Velenik, V., Zadnik, V., Omejc, M., Grosek, J., and Tuta, M. (2020). Influence of concurrent capecitabine based chemoradiotherapy with bevacizumab on the survival rate, late toxicity and health-related quality of life in locally advanced rectal cancer: A prospective phase II CRAB trial. Radiol. Oncol. 54, 461–469. doi:10.2478/raon-2020-0043

PubMed Abstract | CrossRef Full Text | Google Scholar

Venook, A. P., Niedzwiecki, D., Lenz, H. J., Innocenti, F., Fruth, B., Meyerhardt, J. A., et al. (2017). 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 317, 2392–2401. doi:10.1001/jama.2017.7105

PubMed Abstract | CrossRef Full Text | Google Scholar

Vincenzi, B., Santini, D., Rabitti, C., Coppola, R., Beomonte Zobel, B., Trodella, L., et al. (2006). Cetuximab and irinotecan as third-line therapy in advanced colorectal cancer patients: A single centre phase II trial. Br. J. Cancer 94, 792–797. doi:10.1038/sj.bjc.6603018

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Z., Qin, B. D., Ye, C. Y., Wang, M. M., Yuan, L. Y., Dai, W. P., et al. (2022). Cetuximab and vemurafenib plus FOLFIRI (5-fluorouracil/leucovorin/irinotecan) for BRAF V600E-mutated advanced colorectal cancer (IMPROVEMENT): An open-label, single-arm, phase II trial. Eur. J. Cancer 163, 152–162. doi:10.1016/j.ejca.2021.12.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Yaeger, R., Cercek, A., O'reilly, E. M., Reidy, D. L., Kemeny, N., Wolinsky, T., et al. (2015). Pilot trial of combined BRAF and EGFR inhibition in BRAF-mutant metastatic colorectal cancer patients. Clin. Cancer Res. 21, 1313–1320. doi:10.1158/1078-0432.CCR-14-2779

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshida, Y., Yamada, T., Kamiyama, H., Kosugi, C., Ishibashi, K., Yoshida, H., et al. (2021). Combination of TAS-102 and bevacizumab as third-line treatment for metastatic colorectal cancer: TAS-CC3 study. Int. J. Clin. Oncol. 26, 111–117. doi:10.1007/s10147-020-01794-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshino, T., Di Bartolomeo, M., Raghav, K., Masuishi, T., Loupakis, F., Kawakami, H., et al. (2023). Final results of DESTINY-CRC01 investigating trastuzumab deruxtecan in patients with HER2-expressing metastatic colorectal cancer. Nat. Commun. 14, 3332. doi:10.1038/s41467-023-38032-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., and Wang, X. (2020). Targeting the Wnt/β-catenin signaling pathway in cancer. J. Hematol. Oncol. 13, 165. doi:10.1186/s13045-020-00990-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: colorectal cancer, EGFR, HER2, BRAF, anti-angiogenic

Citation: Huang S, Ye J, Gao X, Huang X, Huang J, Lu L, Lu C, Li Y, Luo M, Xie M, Lin Y and Liang R (2023) Progress of research on molecular targeted therapies for colorectal cancer. Front. Pharmacol. 14:1160949. doi: 10.3389/fphar.2023.1160949

Received: 07 February 2023; Accepted: 26 July 2023;
Published: 08 August 2023.

Edited by:

Yong Gao, Guangzhou University of Chinese Medicine, China

Reviewed by:

Fan He, Washington University in St. Louis, United States
Yuanfeng Li, Beijing Proteome Research Center, China
Guillem Argiles Martinez, Memorial Sloan Kettering Cancer Center, United States

Copyright © 2023 Huang, Ye, Gao, Huang, Huang, Lu, Lu, Li, Luo, Xie, Lin and Liang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yan Lin, linyan@gxmu.edu.cn; Rong Liang, liangrong@gxmu.edu.cn

These authors have contributed equally to this work and share first authorship

Lead Contact

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.