Next Article in Journal
Hypersensitivity Reactions to Iodinated Contrast Media
Previous Article in Journal
De-Escalation of Anti-Tumor Necrosis Factor Alpha Agents and Reduction in Adverse Effects: A Systematic Review
Previous Article in Special Issue
ICG-Guided Lymphadenectomy during Surgery for Colon and Rectal Cancer—Interim Analysis of the GREENLIGHT Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Colorectal Cancer Heterogeneity and the Impact on Precision Medicine and Therapy Efficacy

by
Gerardo Rosati
1,*,
Giuseppe Aprile
2,
Alfredo Colombo
3,
Stefano Cordio
4,
Marianna Giampaglia
1,
Alessandro Cappetta
2,
Concetta Maria Porretto
3,
Alfonso De Stefano
5,
Domenico Bilancia
1 and
Antonio Avallone
5,*
1
Medical Oncology Unit, “S. Carlo” Hospital, 85100 Potenza, Italy
2
Department of Oncology, “San Bortolo” General Hospital, Azienda ULSS8 Berica, 36100 Vicenza, Italy
3
Medical Oncology Unit, CDC Macchiarella, 90138 Palermo, Italy
4
Medical Oncology Unit, “Maria Paternò Arezzo” Hospital, 97100 Ragusa, Italy
5
Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori IRCCS-Fondazione “G. Pascale”, 80121 Napoli, Italy
*
Authors to whom correspondence should be addressed.
Biomedicines 2022, 10(5), 1035; https://doi.org/10.3390/biomedicines10051035
Submission received: 16 February 2022 / Revised: 16 April 2022 / Accepted: 28 April 2022 / Published: 30 April 2022
(This article belongs to the Special Issue State-of-the-Art on Gastrointestinal Cancers in Italy)

Abstract

:
Novel targeted therapies for metastatic colorectal cancer are needed to personalize treatments by guiding specific biomarkers selected on the genetic profile of patients. RAS and BRAF inhibitors have been developed for patients who become unresponsive to standard therapies. Sotorasib and adagrasib showed promising results in phase I/II basket trial and a phase III trial was planned with a combination of these RAS inhibitors and anti-EGFR monoclonal antibodies. Encorafenib and binimetinib were administered in phase II clinical trials for BRAF mutated patients. Pembrolizumab is now recommended in patients exhibiting microsatellite instability. Larotrectinib and entrectinib showed a fast and durable response with few and reversible adverse events in cases with NTRK fusions. Trastuzumab and trastuzumab deruxtecan exhibited promising and durable activity in HER-2-positive patients. In this review, the reasons for an extension of the molecular profile of patients were assessed and placed in the context of the advancements in the understanding of genetics. We highlight the differential effect of new targeted therapies through an ever-deeper characterization of tumor tissue. An overview of ongoing clinical trials is also provided.

1. Introduction

Colorectal cancer (CRC) is a major global health issue, being the third most commonly diagnosed malignancy with an estimated number of more than 1.9 million new cases and about 935,000 deaths worldwide in 2020 [1,2]. In twenty percent of cases, the disease occurs at an advanced stage at diagnosis, while up to 50% of patients with early-stage disease relapse, despite curative surgery, adjuvant chemotherapy, and/or radiation therapy. Advances in multidisciplinary treatment and care have led to significant improvements in survival, but a cure is not possible for most of these patients [3].
Targeted therapies work on cancer cells by directly inhibiting cell proliferation, differentiation, and migration. The tumor microenvironment, including local blood vessels and immune cells, could also be altered by targeted drugs, so as to inhibit tumor growth. Various pathways that mediate CRC initiation, progression, and migration, as well as those that can activate signaling cascades, are ideal sites for these drugs [4].
In recent years, clinical outcomes in metastatic colorectal patients (mCRC) have thus improved significantly. International guidelines have now first of all mandated, as a standard of care, the identification of approximately 40% of patients without rat sarcoma virus (RAS) and B-rapidly accelerated fibrosarcoma (BRAF) oncogene mutations [5]. The anti-epidermal growth factor receptor (EGFR) monoclonal antibodies cetuximab and panitumumab, combined with chemotherapy, are standard treatments in these cases. The most important limitation of these drugs is in inducing resistance sooner or later [6,7], although evidence suggests that some patients may benefit from a rechallenge strategy in the course of their disease [8]. On the contrary, patients with mutated tumors benefit from anti-vascular endothelial growth factor (VEGF) agents (bevacizumab and aflibercept) when combined with chemotherapy [9,10].
The refinement of the knowledge of molecular biology and new agents capable of more specifically targeting previously unknown mutated genes change the therapeutic perspectives of many patients. Since 2013, the National Cancer Institute (NCI) has launched, with renewed interest, programs, and initiatives to deepen the function of RAS and learn about its biology to identify innovative drugs [11]. Although BRAF inhibitors as single agents have shown only modest activity in BRAF-mutated mCRC, several clinical trials have demonstrated that combination therapies with EGFR and mitogen-activated protein kinase (MEK) inhibitors overcome resistance mechanisms [12]. Tumors with deficiency in mismatch repair genes (dMMR) are highly responsive to immune checkpoint inhibitors both in first- and second-line settings [13]. Human Epidermal Growth Factor Receptor (HER-2) amplification has emerged as a promising therapeutic target for mCRC patients displaying this molecular abnormality. Patients with RAS wild-type (wt) and HER-2 overexpression have been successfully treated with anti-HER-2 antibodies [14]. Other emerging actionable molecular alterations include rare gene fusions of neurotrophic tyrosine kinase receptor, type 1 (NTRK1) that can be targeted by specific inhibitors [15].
Although new techniques such as next-generation sequencing (NGS) and the availability of tumor panels allow the identification of many predictive markers (Figure 1), their application in clinical practice is often difficult. It is quite evident that recommendations are needed to guide the physician in these cases and support therapeutic decision making for patients with mCRC.
This paper provides an overview of existing CRC-targeted agents (Table 1) and their underlying mechanisms, as well as a discussion of their limitations and future trends.

2. Methods

We carried out a fine literature research to find relevant publications from the last few years utilizing the PubMed database (www.ncbi.nlm.nih.gov/pubmed/, accessed on 31 January 2022) by entering the following terms: “metastatic colorectal cancer” and “biomarkers”. Full-text manuscripts reporting data on these items were searched and reviewed in detail. In addition, we performed a general excursus of the principal oncology congresses to identify abstracts reporting new mCRC biomarker data and published between 1 January 2016 and December 2021.

3. Mutational Status of RAS

RAS is a family of proteins expressed on all cells and responsible for transmitting signals through which proliferation, adhesion, migration, and cell differentiation as well as the apoptosis process are stimulated and controlled. When these proteins mutate, the cells acquire properties of invasion and metastatization. The main mutations concern Kirsten rat sarcoma virus (KRAS) and neuroblastoma ras viral oncogene homolog (NRAS) and consist in the change of a single nucleotide or its deletion or insertion into a deoxyribonucleic acid (DNA) sequence [23].
KRAS mutations are found in approximately 40% of patients with mCRC, mainly in exon 2, codons 12 (70–80%) and 13 (15–20%), less frequently in exons 3 and 4. The most common KRAS mutations of exon 2 (codons 12 and 13) include G12D (32.4%), G13D (14.1%), G12V (11.3%), G12S (9.9%), G12C (8.5%), and G12A (2.8%) [24]. Regardless of type and location, KRAS mutations play a prognostic role and, when grouped together, patients with KRAS mutated metastatic disease have a higher mortality rate (18.5% vs. 34%) and a shorter survival (14 months vs. 23.5 months) than wt ones [25]. Moreover, the various mutations of KRAS are not equal to each other and a pooled analysis of five randomized trials showed that they were associated with heterogeneous outcomes [26]. While patients harboring the KRAS G12C-variant correlated with inferior overall survival (OS) compared with unmutated tumors and a similar trend for OS was seen in the KRAS G13D-variant, more frequent KRAS exon 2 variants like G12D and G12V did not have a significant impact on OS. Although the reasons are not clear, these biological differences could be explained by a separate activation process for each individual variant of the KRAS-depending pathways.
NRAS mutations in codons 2, 3, and 4 are rare and found in 3–5% of metastatic patients, more frequently in left-side colon and mainly in women. An Italian study has shown that NRAS and KRAS mutated tumors did not show significant differences in terms of clinical and pathological characteristics, except for a lower prevalence of mucinous histology and lung metastases among NRAS mutated tumors. In uni- and multivariate analysis, NRAS mutations were associated with shorter OS than in all wt patients (median OS 25.6 vs. 42.7 months) [27]. NRAS mutations recorded at exon 3 identify patients with markedly lower OS not only compared to wt ones (HR 2.85; p < 0.01), but also to those with mutations in exon 2 (HR 2.0; p = 0.039) [28].
EGFR is a membrane receptor tyrosine kinase and is a key target for monoclonal antibodies which bind on the extracellular domain of the receptor. Several phase II and III trials indicate that an increased gene copy number of EGFR or mutations of KRAS and NRAS, responsible for downstream signalling, are important determinants of response to cetuximab and panitumumab [29,30,31,32]. While an improvement of treatment efficacy is proven only in wt patients, RAS mutated patients either had no benefit from the addition of anti-EGFRs or even showed a worse outcome than their comparators [7,33]. Thus, since 2013 extended RAS analysis is recommended at the time of diagnosis in metastatic disease for all patients [5,34].
The best treatment for RAS mutated patients is not sufficiently standardized due to the lack of clinical trials specifically designed for these patients. Although the major international guidelines report that chemotherapy plus bevacizumab should be the preferred first-line therapy for patients with RAS mutation, the evidence is debatable [35,36]. First of all, there are no prospective randomized trials for this specific setting of patients. Secondly, the benefit of the addition of bevacizumab to first-line treatment significantly prolonged progression-free survival (PFS), while in RAS mutated patients it led to a relatively modest reduction in risk of death of 12%. Thirdly, data from randomized phase III studies are confusing as they include patients with RAS wt and RAS mutated disease. Only recently, a Chinese randomized study specifically enrolled patients with RAS mutation and with metastases limited exclusively to the liver showing that bevacizumab plus chemotherapy versus chemotherapy alone results in a higher conversion rate in liver surgery, an increase in response rate (RR), and an extension of PFS and OS [37]. The strategy of employing triplet chemotherapy [leucovorin, oxaliplatin, irinotecan, and fluorouracil (FOLFOXIRI)] plus bevacizumab also appears to confirm that mutated RAS patients have no improvement in OS, although this is partially confirmed by the TRIBE2 study including many patients with these characteristics [38,39].
Only in recent years the clinical research focused on the possibility of its direct inhibition with specific target therapies. Few molecules have reached clinical development more directly and many have instead been studied in the preclinical phase [40].
Focusing only on the former, sotorasib (AMG 510) and adagrasib appear to be very promising. The KRAS p.G12C mutation occurs in approximately 1 to 3% of CRC and increases the proliferation and survival of cancer cells. The small molecules sotorasib and adagrasib specifically and irreversibly inhibit KRASG12C. A phase I study has shown that in 42 patients with heavily pretreated mCRC, sotorasib, administered orally once a day at a dose of 960 mg, was tolerable as only 15 patients (11.6%) reported grade 3 or 4 treatment-related adverse events (AEs) [16]. The most common events were diarrhea (29.5%), fatigue (23.3%), and nausea (20.9%). The median duration of treatment was 3.9 months and, although a confirmed partial response (PR) was observed in only 7.1% of patients, 28 of them (66.7%) had stable disease (SD). The median PFS was 4.0 months. In the same way, adagrasib was tested in a study involving 17 patients, mostly non-small cell lung cancer or mCRC. The reported toxicity was low once again and 12 patients were evaluated for the response: four had a PR and eight had SD. All responders received a dose of 600 mg twice a day; one patient with mCRC had a 47% reduction, six weeks after the start of treatment [41].
However, irreversible inhibition of KRASG12C may result in another benefit in patients due to the signaling pathways that determine a susceptibility to KRAS feedback reactivation. EGFR signaling is involved in this reactivation, providing a rational co-targeting strategy for KRAS-mutant mCRC.
KRYSTAL-1 (NCT03785249), presented at ESMO 2021, is a multicohort study evaluating adagrasib in patients with KRASG12C-mutant advanced solid tumors [17]. The cohort of mCRC patients was treated with adagrasib 600 mg twice a day as monotherapy or in association with cetuximab. Forty-six patients received adagrasib monotherapy and, among the 45 evaluable for clinical activity, the RR was 22% and the disease control rate (DCR) was 87%. Median duration of response was 4.2 months and median PFS was 5.6 months. Thirty-two patients were treated with adagrasib + cetuximab. Among the 28 patients evaluable for clinical activity, the RR was 43% and DCR was 100%. AEs of any grade occurred in 91% and grade 3–4 events in 30% of patients treated with adagrasib monotherapy, while AEs of any grade occurred in 100% and grade 3–4 events in 16% of patients with combination therapy. Adagrasib + cetuximab is currently being evaluated as second-line therapy in a phase III study in this setting of patients (NCT04793958). Sotorasib and panitumumab will also be evaluated in another phase III study (EudraCT Number: 2021-004008-16) in which the combination of the two drugs will be compared versus regorafenib or trifluridine-tipiracil (TAS-102) in pretreated patients with the same mutation.
RAS mutations in mCRC are associated with aberrations in DNA replication and this would make tumors sensitive to inhibition of WEE1, which is a tyrosine protein kinase that regulates G2/M checkpoints in the cell cycle in response to DNA damage. A randomized phase II trial (Focus4-C) has tested the hypothesis that adavosertib (AZD1775), a potent oral WEE1 inhibitor, could increase PFS in patients who had either a clinical response or a SD after at least 16 weeks of chemotherapy administration compared to those undergoing active monitoring [42]. Adavosertib was associated with a PFS improvement over active monitoring (median 3.61 vs. 1.87 months; HR = 0.35; 95% CI, 0.18 to 0.68; p = 0.0022), while the OS did not show substantial differences (median 14.0 vs. 12.8 months; HR = 0.92; 95% CI, 0.44 to 1.94; p = 0.93). In prespecified subgroup analysis, adavosertib activity was greater in left-sided tumors (HR = 0.24; 95% CI, 0.11 to 0.51), versus right-sided (HR = 1.02; 95% CI, 0.41 to 2.56; interaction p = 0.043). Adavosertib was well-tolerated, and few patients reported grade 3 toxicities such as diarrhea (9%), nausea (5%), and neutropenia (7%).
Immunotherapy may be an option for RAS mutated patients. RAS mutation influences the tumor microenvironment, the reduction in the number of tumor-infiltrating lymphocytes (TILs), mismatch-repair defects, and the increase in the expression of programmed cell death ligand 1 (PD-L1) [11]. A preclinical study has demonstrated the potential of therapy with TILs capable of recognizing tumor cells harboring the KRASG12D mutation resulting in tumor regression and a PR lasting 9 months in a patient with mCRC [43].
Another way to achieve RAS inhibition may be to antagonize other proteins of its pathway, as in the case of mitogen-activated protein kinase (MAPK). It has been shown in preclinical models that the combination of MEK inhibitors with an anti-PD-L1 results in a synergistic and lasting tumor regression [44]. Consequently, the feasibility of a combination of atezolizumab and the MEK inhibitor cobimetinib has been evaluated in a phase Ib study [45]. The primary aims were safety and tolerability. In most patients, cobimetinib was administered once daily orally for 21 days on, 7 days off, while atezolizumab was dosed at 800 mg intravenously every 2 weeks. The most common AEs related to the treatment were diarrhea (67%), rash (48%), and fatigue (40%), without significant differences with single-agent cobimeitinib and atezolizumab. Confirmed responses were obtained in 7 out of 84 patients (8%), a SD in 19 cases (23%). The 12-month PFS and OS rates were 11% and 43%. Nevertheless, this potential synergistic activity was not confirmed in a subsequent phase III study designed to compare atezolizumab plus cobimetinib or atezolizumab monotherapy versus standard therapy in the third-line setting [46]. In fact, IMblaze370 did not meet its primary endpoint of improved OS with experimental arms versus regorafenib.
The association of cytotoxics and bevacizumab may promote the sensitivity to immunotherapy increasing the exposure of neoantigens, inducing immunogenic cell death, and increasing the immune infiltration in tumor microenvironment [47]. AtezoTRIBE, a prospective, open label, phase II, comparative trial, randomized 218 initially unresectable mCRC patients (over 70% RAS mutated), irrespective of mismatch repair (MMR) status, to receive up to 8 cycles of FOLFOXIRI/bevacizumab (arm A) or FOLFOXIRI/bevacizumab/atezolizumab (arm B), followed by maintenance with 5-FU/bevacizumab or 5FU/bevacizumab/atezolizumab until disease progression [48]. The primary endpoint was PFS. A significant advantage with the addition of atezolizumab was observed in PFS (13.1 vs. 11.5 mos, HR 0.69, p = 0.012), but not in ORR (59% vs. 64%, p = 0.412). No safety issues were reported.

4. Mutational Status of BRAF

The routine molecular characterization of mCRC patients includes, beyond RAS, the tumor BRAF mutational testing, according to the recommendations provided by the international clinical guidelines [49,50,51]. The BRAF gene encodes a serine-threonine protein kinase that is part of the MAPK pathway. BRAF mutations occur in about 10% of patients with mCRC and are usually mutually exclusive with RAS mutations. They are most frequently caused (>90%) by the replacement of valine with glutamic acid inside the 600 codon (BRAFV600E), leading to an overactive MAPK pathway [12,52].
The presence of somatic BRAFV600E alteration mostly characterizes a subgroup of mCRC patients associated with the female sex, right-sided colonic cancer, mucinous histology, microsatellite instability (MSI)/dMMR profile and metastatic spread mainly to lymph nodes and peritoneum [12,52,53]. BRAFV600E mutant mCRC patients show a shorter OS and achieve a very modest benefit from standard chemotherapy, highlighting their poor prognosis [12]. Moreover, the benefit of anti-EGFRs in these cases remains unclear and two meta-analyses have not been able to provide more clarity to the issue [54,55]. With the advent of next-generation sequencing, non-BRAFV600E mutations have been increasingly identified in clinical practice, more often observed in younger patients, males and showing fewer peritoneal metastases compared to BRAFV600E mutants [56]. The expected survival of this subgroup of patients is not negatively influenced, as happens for BRAFV600. Of note, most of the non-BRAFV600E mutations, in particular those belonging to class 3, retain a sensitivity to anti-EGFRs based treatments [57].
The optimal treatment of BRAFV600E mutant mCRC patients has been matter of active clinical research and controversial debate. In recent years, FOLFOXIRI plus bevacizumab was introduced as a standard of care for initial treatment of this subgroup of mCRC patients [49,50,58]. The use of this intensive combination was mainly supported by a subgroup analysis of 28 BRAFV600E mCRC patients enrolled in the TRIBE trial, which showed a median OS of 19.0 months in patients treated with FOLFOXIRI/bevacizumab, whereas patients treated with leucovorin, irinotecan and fluorouracil (FOLFIRI)/bevacizumab had a shorter median OS of 10.7 months [59]. However, the evidence of benefit from the intensified approach was not confirmed in the TRIBE 2 trial, in which patients were randomized to FOLFOXIRI plus bevacizumab or to leucovorin, oxaliplatin and fluorouracil (FOLFOX) plus bevacizumab [38]. In addition, a recent meta-analysis of five randomized trials comparing FOLFOXIRI plus bevacizumab to a doublet combination plus bevacizumab confirmed the absence of any advantage of FOLFOXIRI plus bevacizumab in BRAF mutated cases [60]. Based on this evidence, an intensification of treatment does not offer a clear benefit in the frontline treatment of BRAFV600E mutated mCRC patients. However, patients with BRAFV600E mutation tumors appear to benefit from anti-VEGF therapy, unlike that with anti-EGFRs, similarly to patients with BRAF wt tumors [12].
A major efficacy of an antiangiogenic agent in combination with chemotherapy has also been reported in the second line treatment of BRAFV600E mutant mCRC patients. Ramucirumab-a highly specific antiangiogenic agent directed against the extracellular domain of the VEGF receptor-2-may block the activating phosphorylation of the proangiogenic receptor. In the VELOUR trial, a subgroup analysis showed that 11 patients treated with FOLFIRI plus aflibercept had a median PFS and OS compared with 19 patients receiving only chemotherapy for 5.5 and 10.3 months vs. 2.2 and 5.5 months, respectively [61]. Similar results were also observed with FOLFIRI plus ramucirumab in the subgroup analysis of the RAISE study [62]. However, the value of these post-hoc analyses should be carefully considered given the small number of patients included.
Unlike the favorable results observed in melanoma patients, treatment with BRAF inhibitors alone has yielded low clinical activity in mCRC due to feedback reactivation of EGFR [63]. Therefore, BRAF600E inhibitors have been combined with anti-EGFR inhibitors and other targeted agents, such as MEK inhibitors, in doublet and triplet regimens with the aim of overcoming resistance to BRAF inhibitor monotherapy and improving its clinical activity [64,65,66,67,68].
This strategy was evaluated in the phase III BEACON study in which 665 patients with BRAFV600E mutated mCRC and disease progression after one or two prior treatments were randomized to receive the BRAF inhibitor encorafenib in combination with the anti-EGFR antibody cetuximab plus the MEK inhibitor binimetinib (triplet therapy) versus encorafenib and cetuximab (doublet therapy) or cetuximab in combination with irinotecan or FOLFIRI as standard therapy [69]. The primary endpoints of the study were OS and RR. In the primary analysis, the triplet and doublet therapy compared with standard therapy resulted in a significantly longer median OS (9.0 and 8.4 months vs. 5.4 months, respectively, p < 0.001) and higher RR (26% and 20% vs. 2%, respectively, p < 0.001). The PFS, secondary endpoint, was also superior in the three-drugs and doublet-drugs regimen when compared to the standard group (4.3 and 4.2 months vs. 1.5 months, respectively, p < 0.001). In addition, safety and tolerability were not significantly different between the groups, resulting in grade 3 or higher AEs in 58% of cases in the triplet regimen, in 50% of cases in the doublet regimen, and in 61% of patients treated with standard therapy, respectively. The updated results of this study, with additional six months of follow up, showed that an OS of 9.3 months was observed for both triplet and doublet regimens compared to 5.9 months of the control therapy [18]. These results strongly support the combination of encorafenib plus cetuximab as a new standard of care for pretreated mCRC BRAFV600E mutated patients. A single arm phase II trial (ANCHOR study), evaluating the strategy with triplet regimen in the first-line setting, which has recently been completed, reported findings similar to the BEACON study [70]. Finally, on the basis of these results, the ongoing phase III study BREAKWATER (NCT04607421) aims to evaluate the efficacy in terms of PFS (primary endpoint) and OS of the combination of encorafenib plus cetuximab with or without chemotherapy compared with a standard anti-VEGF based treatment in the 1st line setting.

5. Microsatellite Instability and Immunotherapy

Immunotherapy in cancer treatment arises from the concept that a condition of immunoevasion exists caused by neoplastic cells in the tumor microenvironment. Tumor cells, through the production of cytokines, stimulate suppressor myeloid cells and regulatory T cells (Treg) to inhibit the CD4+ and to increase CD8+ lymphocytes, braking immune responses. Furthermore, a loss of restricted major histocompatibility complex molecules has also been observed, resulting in an inability of the host to recognize non-self-antigens [71].
Microsatellites are repetitive sequences of coding, and non-coding DNA [72]. MSI results from the inability of the MMR gene to repeat DNA errors that occurred during the replication process. Gene insertions and deletions lead to somatic mutations in these repetitive DNA sequences resulting in genomic instability and production of immunogenic antigens and neoantigens, conditioning a response to checkpoint inhibitors [23]. Inactivation of MMR genes is the result of hypermethylation of the MLH1 promoter or of germline mutations of MLH1, MSH2, MSH6, and PMS2 [73].
Furthermore, MSI germline abnormalities also represent the molecular basis of Lynch syndrome [74]. It represents the most common hereditary form of this cancer. Latham et al. reported that dMMR is common in these patients, so those with MSI or dMMR tumors could predict Lynch syndrome through MSI related tests [75].
MSI is found in approximately 5% of patients with mCRC; only 3% of cases are associated with Lynch syndrome and the other 12% are caused by sporadic hypermethylation of the MLH1 gene. CRC with MSI are most frequently localized on the right and in women over 70 years, are poorly differentiated, and have mucinous histology [76]. MSI tumor status could be a prognostic marker for a more favorable outcome. A large study reported that the percentage of mCRC patients with this characteristic was only 3.5% suggesting that these tumors have a lower probability of metastasizing [77].
The incidence of MSI in stage II and III is about 16%. Some studies have shown that dMMR or MSI tumor status are predictors of reduced benefit from adjuvant chemotherapy and that fluoropyrimidines given alone may even have a detrimental effect in patients with stage II disease [78,79,80]. Conversely, regarding patients with MSI and stage III, ACCENT, a pooled analysis of 12 adjuvant studies, has demonstrated that adding oxaliplatin to fluoropyrimidines improves DFS and OS of patients compared to those of stable microsatellite tumors (MSS) [81]. In particular, this study found a close relationship between the number of positive lymph nodes on histological examination and OS, documenting better outcomes in the N1 group, while data were similar in the N2 group.
In the metastatic setting, a phase II study demonstrated that tumors with a high mutation load benefit most from the use of pembrolizumab. The primary endpoints were immune-related ORR and PFS at 20 weeks. Forty-one patients with progressive disease had ORR and PFS at 20 weeks of 40% and 78% for MSI tumors and 0% and 11% for MSS ones, respectively [82].
CheckMate-142, another phase II study, reported efficacy and safety results in 119 patients with MSI status and previously treated. They received nivolumab 3 mg/kg plus ipilimumab 1 mg/kg once every 3 weeks (four doses) followed by nivolumab 3 mg/kg once every 2 weeks. Primary end point was investigator-assessed ORR. At the median follow-up of 13.4 months, investigator-assessed ORR was 55% (95% CI, 45.2 to 63.8), and DCR for ≥ 12 weeks was 80%. Regarding the median duration of response, more than half of the patients were still responding, so the median is unknown, while PFS rates were 76% (9 months) and 71% (12 months); respective OS rates were 87% and 85%. Grade 3 or 4 AEs were manageable and occurred in 32% of patients [19]. Nivolumab plus ipilimumab could represent a promising new treatment option in these patients.
Finally, in the phase III Keynote 177 trial, pembrolizumab resulted in significantly longer PFS than chemotherapy when received as first-line therapy for MSI mCRC [13]. Co-primary endpoints were PFS and OS. Three hundred and seven patients were assigned to receive pembrolizumab at a dose of 200 mg every 3 weeks or investigator’s choice standard chemotherapy every 2 weeks. Patients undergoing chemotherapy could receive pembrolizumab after disease progression. After a median follow-up of 32.4 months, pembrolizumab was superior to chemotherapy with respect to PFS (median, 16.5 vs. 8.2 months; HR, 0.60; p = 0.0002). At the cutoff date, 56 patients in the pembrolizumab group and 69 in the chemotherapy group had died. OS data were not yet mature after final data evaluation and are pending. An ORR was observed in 43.8% of the patients in the pembrolizumab group and in 33.1% in the chemotherapy group. In addition, the responses appeared to be more prolonged over time with immunotherapy. The AEs of grade 3 or higher occurred in 22% of the patients in the pembrolizumab group, as compared with 66% in the chemotherapy group. A subsequent analysis evaluating the impact of therapy on quality of life showed clinically meaningful improvement in EORTC QLQ-C30 GHS /QOL scores with pembrolizumab compared to chemotherapy [83].

6. HER2 Inhibition

The HER family plays a crucial role in the development and progression of several gastrointestinal tumors, including colorectal, gastric, and biliary adenocarcinomas; its aberrant activation-mainly due to overexpression via HER-2 gene amplification or to alternative genetic mechanisms-has been reported consistently in 5–20% of cancer patients [84,85]. The possibility of inhibiting HER-2 to tackle the progression of the disease is certainly not new, and pivotal randomized trials have shown that the use of trastuzumab either alone or combined with another HER-2 blockade agent has significantly extended survival in molecularly selected cases [86,87,88]. In addition, it stimulated the need for specific classifications and scoring systems to establish HER-2 positivity [20], which is usually scored with immunohistochemistry (IHC), and then confirmed with in situ hybridization or innovative, more sensitive techniques [89]. The IHC scoring system for HER-2 positivity in CRC was established by experienced pathologists involved in the HERACLES project. In more detail, IHC staining judged as intense (3+) in more than 10% of cancer cells with circumferential, basolateral, or lateral pattern was defined as positive; the expert panel recommended to confirm the positivity if the percentage of positive cells was inferior to 50% [20]. As an outstanding example for gastrointestinal oncology, in the open-label, multicenter, international, phase III ToGA trial the combination of standard chemotherapy and trastuzumab was compared to chemotherapy alone [86]. In patients with IHC 3+ HER-2-positive advanced gastric cancers treated with trastuzumab the reported median OS was about 4 months longer that that reported for patients treated with standard therapy (16.0 versus 11.8 months, HR 0.65), and the drug gained accelerated Food and Drug administrative approval. In mCRC, HER-2 has been shown to represent a notable therapeutic target, regardless of its primary or acquired resistance to EGFR inhibition [90,91], although prognostic impact of HER-2 overexpression/amplification has not yet been fully elucitated [92]. HERACLES, a proof-of-concept phase II academic trial, enrolled 35 HER-2-positive, RAS wt, mCRC patients refractory to standard therapies (including cetuximab or panitumumab), with 32 patients evaluable for response. Enrolled patients received intravenous trastuzumab at 4 mg/kg loading dose followed by 2 mg/kg once per week, and oral lapatinib at 1000 mg per day until evidence of disease progression. A RR of 28% was reported with one case of complete response lasting over 7 years, a DCR of 69%, a median PFS of 4.7 months (95% CI 3.7–6.1), and a median OS of 10 months (95% CI 7.9–15.8) [14,89]. Interestingly, progression in the central nervous system occurred in 6 (19%) out of 32 patients, suggesting that the evaluation of HER-2 expression in brain metastases from CRC is important [93]. More recently, the improved understanding of HER-2-driven disease pathology in specific gastrointestinal cancers [94], the comprehension of complex primary or acquired resistance mechanisms [95,96,97,98], the development of novel, more potent HER-2 inhibitors [99], and the possibility to synergistically combine HER-2 inhibition with immunomodulating treatment strategies [100,101], have all contributed to a significant evolution of the treatment scope. In fact, the ultimate frontier for modern HER-2 inhibition in CRC includes three distinctive therapeutic roads.
Firstly, the use of specific antibody-drug conjugates, one of the foremost being trastuzumab deruxtecan (T-DXd), a humanized anti-HER-2 IgG1 monoclonal antibody covalently linked to a topoisomerase I inhibitor payload via a tetrapeptide-based cleavable linker. In the Destiny-CRC01 trial [21], this novel drug demonstrated promising and durable activity in patients with HER-2-positive mCRC refractory to available standard treatments, with an ORR of 45% and a median PFS of almost 7 months. Since antibody-related lung toxicity (i.e., interstitial lung disease/pneumonitis) may affect up to 10% of treated patients [102], this worrying side-effect should be considered and promptly assessed at least in symptomatic patients [100]. Secondly, the study of new specific monoclonal antibodies is providing novel treatment opportunities. Margetuximab, a next-generation Fc-modified HER-2 monoclonal antibody that binds with high affinity to CD16A, has activity in HER-2 positive gastrointestinal malignancies [103]. Lastly, while evidence from studies combining two different antibodies in pretreated patients is rapidly accumulating [104,105,106,107], bi-specific antibodies that may simultaneously target two different parts of HER-2 could potentiate their effect. In HERACLES-B, a single-arm, phase II trial, enrolling highly pretreated patients with RAS/BRAF wt and HER-2 positive mCRC, the combination of pertuzumab (840 mg intravenous load followed by 420 mg intravenous every 3 weeks) and T-DM1 (3.6 mg/kg every 3 weeks) was evaluated until disease progression or toxicity [104]. Unfortunately, the trial did not reach its primary endpoint, with a disappointing RR of 9.1% and a median PFS of 4.8 months. Three studies–namely My Pathway, TRIUMPH, and TAPUR-tested trastuzumab in combination with pertuzumab, with a RR ranging from 25% to 40% and median time to progression (MTP) of 4 to 5 months. In MOUNTAINEER, the combination of trastuzumab and tucatinib produced a RR of 52% and a MTP of 8.1 months, suggesting a synergistic value for the combination. In the meantime, zanidatamab (ZW25), a novel biparatropic targeting drug that can bind to two different HER-2 epitopes, showed high antitumoral activity in a phase I trial gastrointestinal adenocarcinomas with HER-2 overexpression [108]. Confirmatory trials have already started. In this evolving scenario towards the concept of precision oncology, there is the need of appropriate patient selection. A more profound molecular characterization established upfront [109] or at the time of disease progression [110], a better understanding of the optimal treatment sequences [111], the possibility to drive targeted treatments with easily repeatable tests [112], and the development of novel, potent inhibitors [113], will shape the near future and lead forthcoming clinical improvements in HER-2 positive CRC tumors.

7. Targeting NTRK, ALK, and ROS1 Fusions

Among novel actionable targets in mCRC, gene fusions such as NTRK rearrangements or fusions of anaplastic lymphoma kinase (ALK) or Proto-Oncogene 1 Receptor Tyrosine Kinase (ROS1) are of growing importance [114]. While several pathogenic alterations have been reported for such genes, including point mutations, amplifications, and splice variants, fusions are the most common genetic aberrations linked to cancer and cause constitutive gene activations and hyper-activation of the kinase domain. In mCRC, these fusions/rearrangements are rare (0.5–2%) and most frequently occur in elderly patients with right-sided, lymph-node positive, RAS wt, MSI cancers. They may suggest resistance to EGFR-inhibitors, have a negative prognostic survival impact and may be targeted with specific agents [115]. Larotrectinib and entrectinib are oral tropomyosin receptor kinases (TRK). Upon administration, these agents bind to TRK, preventing neurotrophin-TRK interaction and TRK activation, which results in both cellular apoptosis and the inhibition of cell growth in tumors that overexpress TRK. Based on the impressive results of agnostically testing larotrectinib and entrectinib in cancer patients with NTRK rearrangements, with a very high ORR in molecularly selected cases [116,117,118] and improvements in cancer-specific quality of life [115], both agents gained the Food and Drug Administration and the European Medicine Agency approval. In the phase II NAVIGATE study, larotrectinib produced an ORR of 50%, with a median duration of response of 15.5 months, and median OS of almost 30 months [22]. The possibility to use third-generation ALK inhibitors in mCRC has been suggested [119], but the rarity of this gene alteration makes it difficult to conduct large comparative trials. Novel NTRK/ROS1 inhibitors, including selitrectinib, repotrectinib, and belizatinib, are under investigation in early clinical trials [120].

8. Conclusions

As with other cancers, genetic heterogeneity and the consequent stratification of patients is now a reality in the treatment of mCRC. Using in-depth knowledge of the molecular asset, each patient therefore has the opportunity to receive a therapy, even more targeted than was possible until a few years ago through the simple determination of the status of RAS and BRAF. To treat mCRC properly, personalized oncology will soon include many tests useful for the identification of novel putative targets to further enrich the growing toolbox of agents for mCRC patients. It is to be hoped that the progress of molecular biology will be even more tangible in the coming years, even if this presupposes the establishment of adequately equipped laboratories in order to optimize the economic resources of each national health service.

Author Contributions

G.R. conceptualized, designed, and edited final draft of the study. G.R., G.A., A.C. (Alfredo Colombo), S.C., M.G., A.C. (Alessandro Cappetta), C.M.P., A.D.S., D.B., and A.A. conducted online research to find the latest related publications, and wrote, revised, and edited the draft of manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Italian Ministry of Health (RF-2016-02363314).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the writing of the manuscript or in the decision to publish the results.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  3. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
  4. Koehler, A.; Bataille, F.; Schmid, C.; Ruemmele, P.; Waldeck, A.; Blaszyk, H.; Hartmann, A.; Hofstaedter, F.; Dietmaier, W. Gene expression profiling of colorectal cancer and metastases divides tumours according to their clinicopathological stage. J. Pathol. 2004, 204, 65–74. [Google Scholar] [CrossRef]
  5. Sepulveda, A.R.; Hamilton, S.R.; Allegra, C.J.; Grody, W.; Cushman-Vokoun, A.M.; Funkhouser, W.K.; Kopetz, S.E.; Lieu, C.; Lindor, N.M.; Minsky, B.D.; et al. Molecular biomarkers for the evaluation of colorectal cancer: Guideline from the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical Oncology. J. Clin. Oncol. 2017, 35, 1453–1486. [Google Scholar] [CrossRef]
  6. Van Cutsem, E.; Lenz, H.J.; Kohne, C.H.; Heinemann, V.; Tejpar, S.; Melezinek, I.; Beier, F.; Stroh, C.; Rougier, P.; van Krieken, J.H.; et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J. Clin. Oncol. 2015, 33, 692–700. [Google Scholar] [CrossRef] [Green Version]
  7. Douillard, J.Y.; Oliner, K.S.; Siena, S.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.; et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N. Engl. J. Med. 2013, 369, 1023–1034. [Google Scholar] [CrossRef] [Green Version]
  8. Parseghian, C.M.; Napolitano, S.; Loree, J.M.; Kopetz, S. Mechanisms of Innate and Acquired Resistance to Anti-EGFR Therapy: A Review of Current Knowledge with a Focus on Rechallenge Therapies. Clin. Cancer Res. 2019, 25, 6899–6908. [Google Scholar] [CrossRef]
  9. Bang, Y.H.; Hong, Y.S.; Lee, J.S.; Lee, K.W.; Han, H.S.; Kim, S.Y.; Kim, J.W.; Kim, H.K.; Kim, J.W.; Eun, C.K.; et al. Effectiveness of Combining Bevacizumab With First-Line Chemotherapy Regimens for Metastatic Colorectal Cancer in Real-World Practice. Clin. Colorectal Cancer 2021, 20, 101–112. [Google Scholar] [CrossRef]
  10. Saoudi Gonzalez, N.; Salva, F.; Ros, J.; Baraibar, I.; Marmolejo, D.; Valdivia, A.; Cuadra-Urteaga, J.L.; Mulet, N.; Tabernero, J.; Elez, E. Up-to-date role of aflibercept in the treatment of colorectal cancer. Expert Opin. Biol. Ther. 2021, 21, 1315–1324. [Google Scholar] [CrossRef]
  11. Ryan, M.B.; Corcoran, R.B. Therapeutic strategies to target RAS-mutant cancers. Nat. Rev. Clin. Oncol. 2018, 15, 709–720. [Google Scholar] [CrossRef]
  12. Grothey, A.; Fakih, M.; Tabernero, J. Management of BRAF-mutant metastatic colorectal cancer: A review of treatment options and evidence-based guidelines. Ann. Oncol. 2021, 32, 959–967. [Google Scholar] [CrossRef]
  13. Andre, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N. Engl. J. Med. 2020, 383, 2207–2218. [Google Scholar] [CrossRef]
  14. Sartore-Bianchi, A.; Trusolino, L.; Martino, C.; Bencardino, K.; Lonardi, S.; Bergamo, F.; Zagonel, V.; Leone, F.; Depetris, I.; Martinelli, E.; et al. 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. 2016, 17, 738–746. [Google Scholar] [CrossRef]
  15. Kummar, S.; Lassen, U.N. TRK Inhibition: A New Tumor-Agnostic Treatment Strategy. Target Oncol. 2018, 13, 545–556. [Google Scholar] [CrossRef] [Green Version]
  16. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRAS(G12C) Inhibition with Sotorasib in Advanced Solid Tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef]
  17. Weiss, J.; Yaeger, R.D.; Johnson, M.L.; Spira, A.; Klempner, S.J.; Barve, M.A.; Christensen, J.G.; Chi, A.; Der-Torossian, H.; Velastegui, K.; et al. LBA6 KRYSTAL-1: Adagrasib (MRTX849) as monotherapy or combined with cetuximab (Cetux) in patients (Pts) with colorectal cancer (CRC) harboring a KRASG12C mutation. Ann. Oncol. 2021, 32, S1294. [Google Scholar] [CrossRef]
  18. Tabernero, J.; Grothey, A.; Van Cutsem, E.; Yaeger, R.; Wasan, H.; Yoshino, T.; Desai, J.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. 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. 2021, 39, 273–284. [Google Scholar] [CrossRef]
  19. Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef]
  20. Valtorta, E.; Martino, C.; Sartore-Bianchi, A.; Penaullt-Llorca, F.; Viale, G.; Risio, M.; Rugge, M.; Grigioni, W.; Bencardino, K.; Lonardi, S.; et al. Assessment of a HER2 scoring system for colorectal cancer: Results from a validation study. Mod. Pathol. 2015, 28, 1481–1491. [Google Scholar] [CrossRef] [Green Version]
  21. Siena, S.; Di Bartolomeo, M.; Raghav, K.; Masuishi, T.; Loupakis, F.; Kawakami, H.; Yamaguchi, K.; Nishina, T.; Fakih, M.; Elez, E.; et al. Trastuzumab deruxtecan (DS-8201) in patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): A multicentre, open-label, phase 2 trial. Lancet Oncol. 2021, 22, 779–789. [Google Scholar] [CrossRef]
  22. Chou, A.; Fraser, T.; Ahadi, M.; Fuchs, T.; Sioson, L.; Clarkson, A.; Sheen, A.; Singh, N.; Corless, C.L.; Gill, A.J. NTRK gene rearrangements are highly enriched in MLH1/PMS2 deficient, BRAF wild-type colorectal carcinomas-a study of 4569 cases. Mod. Pathol. 2020, 33, 924–932. [Google Scholar] [CrossRef]
  23. Afrăsânie, V.-A.; Marinca, M.V.; Alexa-Stratulat, T.; Gafton, B.; Păduraru, M.; Adavidoaiei, A.M.; Miron, L.; Rusu, C. KRAS, NRAS, BRAF, HER2 and microsatellite instability in metastatic colorectal cancer—Practical implications for the clinician. Radiol. Oncol. 2019, 53, 265–274. [Google Scholar] [CrossRef] [Green Version]
  24. Gong, J.; Cho, M.; Sy, M.; Salgia, R.; Fakih, M. Molecular profiling of metastatic colorectal tumors using next-generation sequencing: A single-institution experience. Oncotarget 2017, 8, 42198–42213. [Google Scholar] [CrossRef] [Green Version]
  25. Heinemann, V.; Stintzing, S.; Kirchner, T.; Boeck, S.; Jung, A. Clinical relevance of EGFR- and KRAS-status in colorectal cancer patients treated with monoclonal antibodies directed against the EGFR. Cancer Treat. Rev. 2009, 35, 262–271. [Google Scholar] [CrossRef]
  26. Modest, D.P.; Ricard, I.; Heinemann, V.; Hegewisch-Becker, S.; Schmiegel, W.; Porschen, R.; Stintzing, S.; Graeven, U.; Arnold, D.; von Weikersthal, L.F.; et al. Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: Pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group. Ann. Oncol. 2016, 27, 1746–1753. [Google Scholar] [CrossRef]
  27. Schirripa, M.; Cremolini, C.; Loupakis, F.; Morvillo, M.; Bergamo, F.; Zoratto, F.; Salvatore, L.; Antoniotti, C.; Marmorino, F.; Sensi, E.; et al. Role of NRAS mutations as prognostic and predictive markers in metastatic colorectal cancer. Int. J. Cancer 2015, 136, 83–90. [Google Scholar] [CrossRef]
  28. Matallanas, D.; Birtwistle, M.; Romano, D.; Zebisch, A.; Rauch, J.; von Kriegsheim, A.; Kolch, W. Raf family kinases: Old dogs have learned new tricks. Genes Cancer 2011, 2, 232–260. [Google Scholar] [CrossRef] [Green Version]
  29. Van Cutsem, E.; Kohne, C.H.; Hitre, E.; Zaluski, J.; Chang Chien, C.R.; Makhson, A.; D'Haens, G.; Pinter, T.; Lim, R.; Bodoky, G.; et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N. Engl. J. Med. 2009, 360, 1408–1417. [Google Scholar] [CrossRef] [Green Version]
  30. Bokemeyer, C.; Bondarenko, I.; Hartmann, J.T.; de Braud, F.; Schuch, G.; Zubel, A.; Celik, I.; Schlichting, M.; Koralewski, P. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: The OPUS study. Ann. Oncol. 2011, 22, 1535–1546. [Google Scholar] [CrossRef]
  31. Au, H.J.; Karapetis, C.S.; O’Callaghan, C.J.; Tu, D.; Moore, M.J.; Zalcberg, J.R.; Kennecke, H.; Shapiro, J.D.; Koski, S.; Pavlakis, N.; et al. Health-related quality of life in patients with advanced colorectal cancer treated with cetuximab: Overall and KRAS-specific results of the NCIC CTG and AGITG CO.17 Trial. J. Clin. Oncol. 2009, 27, 1822–1828. [Google Scholar] [CrossRef] [Green Version]
  32. Peeters, M.; Douillard, J.Y.; Van Cutsem, E.; Siena, S.; Zhang, K.; Williams, R.; Wiezorek, J. Mutant KRAS codon 12 and 13 alleles in patients with metastatic colorectal cancer: Assessment as prognostic and predictive biomarkers of response to panitumumab. J. Clin. Oncol. 2013, 31, 759–765. [Google Scholar] [CrossRef] [Green Version]
  33. Ciardiello, F.; Lenz, H.-J.; Kohne, C.-H.; Heinemann, V.; Tejpar, S.; Esser, R.; Beier, F.; Stroh, C.; Duecker, K.; Van Cutsem, E. Effect of KRAS and NRAS mutational status on first-line treatment with FOLFIRI plus cetuximab in patients with metastatic colorectal cancer (mCRC): New results from the CRYSTAL trial. J. Clin. Oncol. 2014, 32, LBA443. [Google Scholar] [CrossRef]
  34. Allegra, C.J.; Rumble, R.B.; Hamilton, S.R.; Mangu, P.B.; Roach, N.; Hantel, A.; Schilsky, R.L. Extended RAS Gene Mutation Testing in Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J. Clin. Oncol. 2016, 34, 179–185. [Google Scholar] [CrossRef]
  35. Lakatos, G.; Kohne, C.H.; Bodoky, G. Current therapy of advanced colorectal cancer according to RAS/RAF mutational status. Cancer Metastasis Rev. 2020, 39, 1143–1157. [Google Scholar] [CrossRef]
  36. Stahler, A.; Heinemann, V.; Ricard, I.; von Einem, J.C.; Giessen-Jung, C.; Westphalen, C.B.; Michl, M.; Heinrich, K.; Miller-Phillips, L.; Jelas, I.; et al. Current treatment options in RAS mutant metastatic colorectal cancer patients: A meta-analysis of 14 randomized phase III trials. J. Cancer Res. Clin. Oncol. 2020, 146, 2077–2087. [Google Scholar] [CrossRef]
  37. Xu, J.; Liu, T.; Tang, W.; Chang, W.; Feng, Q.; Wei, Y.; Ren, L.; Ye, Q.; Cui, Y.; He, G.; et al. Bevacizumab plus chemotherapy versus chemotherapy alone as first-line treatment for patients with RAS mutant unresectable colorectal liver-limited metastases: A single center randomized control trial. Ann. Oncol. 2019, 30, v867. [Google Scholar] [CrossRef]
  38. Cremolini, C.; Loupakis, F.; Antoniotti, C.; Lupi, C.; Sensi, E.; Lonardi, S.; Mezi, S.; Tomasello, G.; Ronzoni, M.; Zaniboni, A.; et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: Updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol. 2015, 16, 1306–1315. [Google Scholar] [CrossRef]
  39. Cremolini, C.; Antoniotti, C.; Rossini, D.; Lonardi, S.; Loupakis, F.; Pietrantonio, F.; Bordonaro, R.; Latiano, T.P.; Tamburini, E.; Santini, D.; et al. 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. 2020, 21, 497–507. [Google Scholar]
  40. Rosati, G.; Aprile, G.; Basile, D.; Avallone, A. Perspectives in the Treatment of RAS or BRAF Mutated Metastatic Colorectal Cancer Patients. Front. Oncol. 2021, 11, 602596. [Google Scholar] [CrossRef]
  41. Romero, D. Two new agents target KRAS G12C. Nat. Rev. Clin. Oncol. 2020, 17, 6. [Google Scholar] [CrossRef] [PubMed]
  42. Seligmann, J.F.; Fisher, D.J.; Brown, L.C.; Adams, R.A.; Graham, J.; Quirke, P.; Richman, S.D.; Butler, R.; Domingo, E.; Blake, A.; et al. Inhibition of WEE1 Is Effective in TP53- and RAS-Mutant Metastatic Colorectal Cancer: A Randomized Trial (FOCUS4-C) Comparing Adavosertib (AZD1775) With Active Monitoring. J. Clin. Oncol. 2021, 39, 3705–3715. [Google Scholar] [CrossRef] [PubMed]
  43. Tran, E.; Robbins, P.F.; Lu, Y.C.; Prickett, T.D.; Gartner, J.J.; Jia, L.; Pasetto, A.; Zheng, Z.; Ray, S.; Groh, E.M.; et al. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N. Engl. J. Med. 2016, 375, 2255–2262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ebert, P.J.R.; Cheung, J.; Yang, Y.; McNamara, E.; Hong, R.; Moskalenko, M.; Gould, S.E.; Maecker, H.; Irving, B.A.; Kim, J.M.; et al. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity 2016, 44, 609–621. [Google Scholar] [CrossRef] [Green Version]
  45. Hellmann, M.D.; Kim, T.W.; Lee, C.B.; Goh, B.C.; Miller, W.H., Jr.; Oh, D.Y.; Jamal, R.; Chee, C.E.; Chow, L.Q.M.; Gainor, J.F.; et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Ann Oncol. 2019, 30, 1134–1142. [Google Scholar] [CrossRef]
  46. Eng, C.; Kim, T.W.; Bendell, J.; Argiles, G.; Tebbutt, N.C.; Di Bartolomeo, M.; Falcone, A.; Fakih, M.; Kozloff, M.; Segal, N.H.; et al. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): A multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2019, 20, 849–861. [Google Scholar] [CrossRef]
  47. Antoniotti, C.; Borelli, B.; Rossini, D.; Pietrantonio, F.; Morano, F.; Salvatore, L.; Lonardi, S.; Marmorino, F.; Tamberi, S.; Corallo, S.; et al. AtezoTRIBE: A randomised phase II study of FOLFOXIRI plus bevacizumab alone or in combination with atezolizumab as initial therapy for patients with unresectable metastatic colorectal cancer. BMC Cancer 2020, 20, 683. [Google Scholar] [CrossRef]
  48. Cremolini, C.; Rossini, D.; Antoniotti, C.; Pietrantonio, F.; Lonardi, S.; Salvatore, L.; Marmorino, F.; Borelli, B.; Ambrosini, M.; Barsotti, G. FOLFOXIRI plus bevacizumab (bev) plus atezolizumab (atezo) versus FOLFOXIRI plus bev as first-line treatment of unresectable metastatic colorectal cancer (mCRC) patients: Results of the phase II randomized AtezoTRIBE study by GONO. Ann. Oncol. 2021, 32, S1294–S1295. [Google Scholar] [CrossRef]
  49. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; Van Krieken, J.H.; Aderka, D.; Aranda Aguilar, E.; Bardelli, A.; Benson, A.; Bodoky, G.; et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. 2016, 27, 1386–1422. [Google Scholar] [CrossRef]
  50. Yoshino, T.; Arnold, D.; Taniguchi, H.; Pentheroudakis, G.; Yamazaki, K.; Xu, R.H.; Kim, T.W.; Ismail, F.; Tan, I.B.; Yeh, K.H.; et al. Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: A JSMO-ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann. Oncol. 2018, 29, 44–70. [Google Scholar] [CrossRef]
  51. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Cederquist, L.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Engstrom, P.F.; et al. Rectal Cancer, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2018, 16, 874–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Lebrun, H.; Turpin, A.; Zerbib, P. Therapeutic implications of B-RAF mutations in colorectal cancer. J. Visc. Surg. 2021, 158, 487–496. [Google Scholar] [CrossRef] [PubMed]
  53. Loupakis, F.; Moretto, R.; Aprile, G.; Muntoni, M.; Cremolini, C.; Iacono, D.; Casagrande, M.; Ferrari, L.; Salvatore, L.; Schirripa, M.; et al. Clinico-pathological nomogram for predicting BRAF mutational status of metastatic colorectal cancer. Br. J. Cancer 2016, 114, 30–36. [Google Scholar] [CrossRef] [PubMed]
  54. Rowland, A.; Dias, M.M.; Wiese, M.D.; Kichenadasse, G.; McKinnon, R.A.; Karapetis, C.S.; Sorich, M.J. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br. J. Cancer 2015, 112, 1888–1894. [Google Scholar] [CrossRef] [Green Version]
  55. Pietrantonio, F.; Petrelli, F.; Coinu, A.; Di Bartolomeo, M.; Borgonovo, K.; Maggi, C.; Cabiddu, M.; Iacovelli, R.; Bossi, I.; Lonati, V.; et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: A meta-analysis. Eur. J. Cancer 2015, 51, 587–594. [Google Scholar] [CrossRef]
  56. Jones, J.C.; Renfro, L.A.; Al-Shamsi, H.O.; Schrock, A.B.; Rankin, A.; Zhang, B.Y.; Kasi, P.M.; Voss, J.S.; Leal, A.D.; Sun, J.; et al. (Non-V600) BRAF Mutations Define a Clinically Distinct Molecular Subtype of Metastatic Colorectal Cancer. J. Clin. Oncol. 2017, 35, 2624–2630. [Google Scholar] [CrossRef]
  57. Yaeger, R.; Kotani, D.; Mondaca, S.; Parikh, A.R.; Bando, H.; Van Seventer, E.E.; Taniguchi, H.; Zhao, H.; Thant, C.N.; de Stanchina, E.; et al. Response to Anti-EGFR Therapy in Patients with BRAF non-V600-Mutant Metastatic Colorectal Cancer. Clin. Cancer Res. 2019, 25, 7089–7097. [Google Scholar] [CrossRef] [Green Version]
  58. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Cederquist, L.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Engstrom, P.F.; et al. NCCN Guidelines Insights: Colon Cancer, Version 2.2018. J. Natl. Compr. Cancer Netw. 2018, 16, 359–369. [Google Scholar] [CrossRef] [Green Version]
  59. Loupakis, F.; Cremolini, C.; Masi, G.; Lonardi, S.; Zagonel, V.; Salvatore, L.; Cortesi, E.; Tomasello, G.; Ronzoni, M.; Spadi, R.; et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N. Engl. J. Med. 2014, 371, 1609–1618. [Google Scholar] [CrossRef] [Green Version]
  60. Cremolini, C.; Antoniotti, C.; Stein, A.; Bendell, J.; Gruenberger, T.; Rossini, D.; Masi, G.; Ongaro, E.; Hurwitz, H.; Falcone, A.; et al. Individual Patient Data Meta-Analysis of FOLFOXIRI Plus Bevacizumab Versus Doublets Plus Bevacizumab as Initial Therapy of Unresectable Metastatic Colorectal Cancer. J. Clin. Oncol. 2020, 38, 3314–3324. [Google Scholar] [CrossRef]
  61. Wirapati, P.; Pomella, V.; Vandenbosch, B.; Kerr, P.; Maiello, E.; Jeffery, G.M.; Curca, R.-O.D.; Karthaus, M.; Bridgewater, J.A.; Mihailov, A.C.; et al. Velour trial biomarkers update: Impact of RAS, BRAF, and sidedness on aflibercept activity. J. Clin. Oncol. 2017, 35, 3538. [Google Scholar] [CrossRef]
  62. Yoshino, T.; Portnoy, D.C.; Obermannova, R.; Bodoky, G.; Prausova, J.; Garcia-Carbonero, R.; Ciuleanu, T.; Garcia-Alfonso, P.; Cohn, A.L.; Van Cutsem, E.; et al. Biomarker analysis beyond angiogenesis: RAS/RAF mutation status, tumour sidedness, and second-line ramucirumab efficacy in patients with metastatic colorectal carcinoma from RAISE-a global phase III study. Ann. Oncol. 2019, 30, 124–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kopetz, S.; Desai, J.; Chan, E.; Hecht, J.R.; O’Dwyer, P.J.; Maru, D.; Morris, V.; Janku, F.; Dasari, A.; Chung, W.; et al. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J. Clin. Oncol. 2015, 33, 4032–4038. [Google Scholar] [CrossRef] [PubMed]
  64. van Geel, R.; Tabernero, J.; Elez, E.; Bendell, J.C.; Spreafico, A.; Schuler, M.; Yoshino, T.; Delord, J.P.; Yamada, Y.; Lolkema, M.P.; et al. A Phase Ib Dose-Escalation Study of Encorafenib and Cetuximab with or without Alpelisib in Metastatic BRAF-Mutant Colorectal Cancer. Cancer Discov. 2017, 7, 610–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kopetz, S.; Grothey, A.; Yaeger, R.; Van Cutsem, E.; Desai, J.; Yoshino, T.; Wasan, H.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. N. Engl. J. Med. 2019, 381, 1632–1643. [Google Scholar] [CrossRef] [Green Version]
  66. Yaeger, R.; Cercek, A.; O’Reilly, E.M.; Reidy, D.L.; Kemeny, N.; Wolinsky, T.; Capanu, M.; Gollub, M.J.; Rosen, N.; Berger, M.F.; et al. Pilot trial of combined BRAF and EGFR inhibition in BRAF-mutant metastatic colorectal cancer patients. Clin. Cancer Res. 2015, 21, 1313–1320. [Google Scholar] [CrossRef] [Green Version]
  67. Corcoran, R.B.; Atreya, C.E.; Falchook, G.S.; Kwak, E.L.; Ryan, D.P.; Bendell, J.C.; Hamid, O.; Messersmith, W.A.; Daud, A.; Kurzrock, R.; et al. Combined BRAF and MEK Inhibition With Dabrafenib and Trametinib in BRAF V600-Mutant Colorectal Cancer. J. Clin. Oncol. 2015, 33, 4023–4031. [Google Scholar] [CrossRef] [Green Version]
  68. Huijberts, S.; Boelens, M.C.; Bernards, R.; Opdam, F.L. Mutational profiles associated with resistance in patients with BRAFV600E mutant colorectal cancer treated with cetuximab and encorafenib +/− binimetinib or alpelisib. Br. J. Cancer 2021, 124, 176–182. [Google Scholar] [CrossRef]
  69. Kopetz, S.; Grothey, A.; Tabernero, J. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. Reply. N. Engl. J. Med. 2020, 382, 877–878. [Google Scholar]
  70. Van Cutsem, E.; Taieb, J.; Yaeger, R.; Yoshino, T.; Maiello, E.; Elez, E.; Dekervel, J.; Ross, P.; Ruiz Casado, A.; Graham, J.; et al. O-10 ANCHOR CRC: Results from a single-arm, phase 2 study of encorafenib, binimetinib plus cetuximab in previously untreated BRAF V600E–mutant metastatic colorectal cancer. Ann. Oncol. 2021, 32, S222. [Google Scholar] [CrossRef]
  71. Tumeh, P.C.; Harview, C.L.; Yearley, J.H. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, L. Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part II. The utility of microsatellite instability testing. J. Mol. Diagn. 2008, 10, 301–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chang, S.C.; Li, A.F.; Lin, P.C.; Lin, C.C.; Lin, H.H.; Huang, S.C.; Lin, C.H.; Liang, W.Y.; Chen, W.S.; Jiang, J.K.; et al. Clinicopathological and Molecular Profiles of Sporadic Microsatellite Unstable Colorectal Cancer with or without the CpG Island Methylator Phenotype (CIMP). Cancer 2020, 12, 3487. [Google Scholar] [CrossRef] [PubMed]
  74. Tiwari, A.K.; Roy, H.K.; Lynch, H.T. Lynch syndrome in the 21st century: Clinical perspectives. QJM Int. J. Med. 2016, 109, 151–158. [Google Scholar] [CrossRef] [Green Version]
  75. Latham, A.; Srinivasan, P.; Kemel, Y. Microsatellite instability is associated with the presence of lynch syndrome pan-cancer. J. Clin. Oncol. 2018, 37, 286–297. [Google Scholar] [CrossRef]
  76. Ashktorab, H.; Ahuja, S.; Kannan, L.; Llor, X.; Ellis, N.A.; Xicola, R.M.; Laiyemo, A.O.; Carethers, J.M.; Brim, H.; Nouraie, M. A meta-analysis of MSI frequency and race in colorectal cancer. Oncotarget 2016, 7, 34546–34557. [Google Scholar] [CrossRef]
  77. Koopman, M.; Kortman, G.A.; Mekenkamp, L.; Ligtenberg, M.J.; Hoogerbrugge, N.; Antonini, N.F.; Punt, C.J.; van Krieken, J.H. Deficient mismatch repair system in patients with sporadic advanced colorectal cancer. Br. J. Cancer 2009, 100, 266–273. [Google Scholar] [CrossRef] [Green Version]
  78. Ribic, C.M.; Sargent, D.J.; Moore, M.J.; Thibodeau, S.N.; French, A.J.; Goldberg, R.M.; Hamilton, S.R.; Laurent-Puig, P.; Gryfe, R.; Shepherd, L.E.; et al. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N. Engl. J. Med. 2003, 349, 247–257. [Google Scholar] [CrossRef] [Green Version]
  79. Sinicrope, F.A.; Mahoney, M.R.; Smyrk, T.C.; Thibodeau, S.N.; Warren, R.S.; Bertagnolli, M.M.; Nelson, G.D.; Goldberg, R.M.; Sargent, D.J.; Alberts, S.R. Prognostic impact of deficient DNA mismatch repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J. Clin. Oncol. 2013, 31, 3664–3672. [Google Scholar] [CrossRef]
  80. Hutchins, G.; Southward, K.; Handley, K.; Magill, L.; Beaumont, C.; Stahlschmidt, J.; Richman, S.; Chambers, P.; Seymour, M.; Kerr, D.; et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J. Clin. Oncol. 2011, 29, 1261–1270. [Google Scholar] [CrossRef]
  81. Cohen, R.; Taieb, J.; Fiskum, J.; Yothers, G.; Goldberg, R.; Yoshino, T.; Alberts, S.; Allegra, C.; de Gramont, A.; Seitz, J.F.; et al. Microsatellite Instability in Patients With Stage III Colon Cancer Receiving Fluoropyrimidine With or Without Oxaliplatin: An ACCENT Pooled Analysis of 12 Adjuvant Trials. J. Clin. Oncol. 2021, 39, 642–651. [Google Scholar] [CrossRef] [PubMed]
  82. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [Green Version]
  83. Andre, T.; Amonkar, M.; Norquist, J.M.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.J.A.; Smith, D.; Garcia-Carbonero, R.; et al. Health-related quality of life in patients with microsatellite instability-high or mismatch repair deficient metastatic colorectal cancer treated with first-line pembrolizumab versus chemotherapy (KEYNOTE-177): An open-label, randomised, phase 3 trial. Lancet Oncol. 2021, 22, 665–677. [Google Scholar] [CrossRef]
  84. Fanotto, V.; Ongaro, E.; Rihawi, K.; Avallone, A.; Silvestris, N.; Fornaro, L.; Vasile, E.; Antonuzzo, L.; Leone, F.; Rosati, G.; et al. HER-2 inhibition in gastric and colorectal cancers: Tangible achievements, novel acquisitions and future perspectives. Oncotarget 2016, 7, 69060–69074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hyman, D.M.; Piha-Paul, S.A.; Won, H.; Rodon, J.; Saura, C.; Shapiro, G.I.; Juric, D.; Quinn, D.I.; Moreno, V.; Doger, B.; et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 2018, 554, 189–194. [Google Scholar] [CrossRef]
  86. Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010, 376, 687–697, Erratum in: Lancet 2010, 376, 1302. [Google Scholar] [CrossRef]
  87. Javle, M.; Borad, M.J.; Azad, N.S.; Kurzrock, R.; Abou-Alfa, G.K.; George, B.; Hainsworth, J.; Meric-Bernstam, F.; Swanton, C.; Sweeney, C.J.; et al. Pertuzumab and trastuzumab for HER2-positive, metastatic biliary tract cancer (MyPathway): A multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2021, 22, 1290–1300. [Google Scholar] [CrossRef]
  88. Meric-Bernstam, F.; Hurwitz, H.; Raghav, K.P.S.; McWilliams, R.R.; Fakih, M.; VanderWalde, A.; Swanton, C.; Kurzrock, R.; Burris, H.; Sweeney, C.; et al. 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. 2019, 20, 518–530. [Google Scholar] [CrossRef]
  89. Roviello, G.; Catalano, M.; Iannone, L.F.; Marano, L.; Brugia, M.; Rossi, G.; Aprile, G.; Antonuzzo, L. Current status and future perspectives in HER2 positive advanced gastric cancer. Clin. Transl. Oncol. 2022, in press. [Google Scholar] [CrossRef]
  90. Bertotti, A.; Migliardi, G.; Galimi, F.; Sassi, F.; Torti, D.; Isella, C.; Corà, D.; Di Nicolantonio, F.; Buscarino, M.; Petti, C.; et al. A molecularly annotated platform of patient-derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 2011, 1, 508–523. [Google Scholar] [CrossRef] [Green Version]
  91. Sartore-Bianchi, A.; Amatu, A.; Porcu, L.; Ghezzi, S.; Lonardi, S.; Leone, F.; Bergamo, F.; Fenocchio, E.; Martinelli, E.; Borelli, B.; et al. HER2 Positivity Predicts Unresponsiveness to EGFR-Targeted Treatment in Metastatic Colorectal Cancer. Oncologist 2019, 24, 1395–1402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tosi, F.; Sartore-Bianchi, A.; Lonardi, S.; Amatu, A.; Leone, F.; Ghezzi, S.; Martino, C.; Bencardino, K.; Bonazzina, E.; Bergamo, F.; et al. Long-term Clinical Outcome of Trastuzumab and Lapatinib for HER2-positive Metastatic Colorectal Cancer. Clin. Colorectal Cancer 2020, 19, 256–262.e2. [Google Scholar] [CrossRef] [PubMed]
  93. Aprile, G.; De Maglio, G.; Menis, J.; Casagrande, M.; Tuniz, F.; Pisa, E.F.; Fontanella, C.; Skrap, M.; Beltrami, A.C.; Fasola, G.; et al. HER-2 Expression in Brain Metastases from Colorectal Cancer and Corresponding Primary Tumors: A Case Cohort Series. Int. J. Mol. Sci. 2013, 14, 2370–2387. [Google Scholar] [CrossRef] [Green Version]
  94. Siena, S.; Sartore-Bianchi, A.; Marsoni, S.; Hurwitz, H.I.; McCall, S.J.; Penault-Llorca, F.; Srock, S.; Bardelli, A.; Trusolino, L. Targeting the human epidermal growth factor receptor 2 (HER2) oncogene in colorectal cancer. Ann. Oncol. 2018, 29, 1108–1119. [Google Scholar] [CrossRef] [PubMed]
  95. Vernieri, C.; Milano, M.; Brambilla, M.; Mennitto, A.; Maggi, C.; Cona, M.S.; Prisciandaro, M.; Fabbroni, C.; Celio, L.; Mariani, G.; et al. Resistance mechanisms to anti-HER2 therapies in HER2-positive breast cancer: Current knowledge, new research directions and therapeutic perspectives. Crit. Rev. Oncol. Hematol. 2019, 139, 53–66. [Google Scholar] [CrossRef] [PubMed]
  96. Piro, G.; Carbone, C.; Cataldo, I.; Di Nicolantonio, F.; Giacopuzzi, S.; Aprile, G.; Simionato, F.; Boschi, F.; Zanotto, M.; Mina, M.M.; et al. An FGFR3 Autocrine Loop Sustains Acquired Resistance to Trastuzumab in Gastric Cancer Patients. Clin. Cancer Res. 2016, 22, 6164–6175. [Google Scholar] [CrossRef] [Green Version]
  97. Pietrantonio, F.; Fucà, G.; Morano, F.; Gloghini, A.; Corso, S.; Aprile, G.; Perrone, F.; De Vita, F.; Tamborini, E.; Tomasello, G.; et al. Biomarkers of Primary Resistance to Trastuzumab in HER2-Positive Metastatic Gastric Cancer Patients: The AMNESIA Case-Control Study. Clin. Cancer Res. 2018, 24, 1082–1089. [Google Scholar] [CrossRef] [Green Version]
  98. Mangiapane, L.R.; Nicotra, A.; Turdo, A.; Gaggianesi, M.; Bianca, P.; Di Franco, S.; Sardina, D.S.; Veschi, V.; Signore, M.; Beyes, S.; et al. PI3K-driven HER2 expression is a potential therapeutic target in colorectal cancer stem cells. Gut 2022, 71, 119–128. [Google Scholar] [CrossRef]
  99. Guarini, C.; Grassi, T.; Pezzicoli, G.; Porta, C. Beyond RAS and BRAF: HER2, a New Actionable Oncotarget in Advanced Colorectal Cancer. Int. J. Mol. Sci. 2021, 22, 6813. [Google Scholar] [CrossRef]
  100. Gall, V.A.; Philips, A.V.; Qiao, N.; Clise-Dwyer, K.; Perakis, A.A.; Zhang, M.; Clifton, G.T.; Sukhumalchandra, P.; Ma, Q.; Reddy, S.M.; et al. Trastuzumab Increases HER2 Uptake and Cross-Presentation by Dendritic Cells. Cancer Res. 2017, 77, 5374–5383. [Google Scholar] [CrossRef] [Green Version]
  101. Park, S.; Jiang, Z.; Mortenson, E.D.; Deng, L.; Radkevich-Brown, O.; Yang, X.; Sattar, H.; Wang, Y.; Brown, N.K.; Greene, M.; et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 2010, 18, 160–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Tarantino, P.; Modi, S.; Tolaney, S.M.; Cortés, J.; Hamilton, E.P.; Kim, S.B.; Toi, M.; Andrè, F.; Curigliano, G. Interstitial Lung Disease Induced by Anti-ERBB2 Antibody-Drug Conjugates: A Review. JAMA Oncol. 2021, 7, 1873–1881. [Google Scholar] [CrossRef] [PubMed]
  103. Bang, Y.J.; Giaccone, G.; Im, S.A.; Oh, D.Y.; Bauer, T.M.; Nordstrom, J.L.; Li, H.; Chichili, G.R.; Moore, P.A.; Hong, S.; et al. First-in-human phase 1 study of margetuximab (MGAH22), an Fc-modified chimeric monoclonal antibody, in patients with HER2-positive advanced solid tumors. Ann. Oncol. 2017, 28, 855–861. [Google Scholar] [CrossRef] [PubMed]
  104. Sartore-Bianchi, A.; Lonardi, S.; Martino, C.; Fenocchio, E.; Tosi, F.; Ghezzi, S.; Leone, F.; Bergamo, F.; Zagonel, V.; Ciardiello, F.; et al. Pertuzumab and trastuzumab emtansine in patients with HER2-amplified metastatic colorectal cancer: The phase II HERACLES-B trial. ESMO Open 2020, 5, e000911. [Google Scholar] [CrossRef]
  105. Nakamura, Y.; Okamoto, W.; Kato, T.; Esaki, T.; Kato, K.; Komatsu, Y.; Yuki, S.; Masuishi, T.; Nishina, T.; Ebi, H.; et al. Circulating tumor DNA-guided treatment with pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer: A phase 2 trial. Nat. Med. 2021, 27, 1899–1903. [Google Scholar] [CrossRef]
  106. Gupta, R.; Garrett-Mayer, E.; Halabi, S.; Mangat, P.K.; D’Andre, S.D.; Meiri, E.; Shrestha, S.; Warren, S.L.; Ranasinghe, S.; Schilsky, R.L. Pertuzumab plus trastuzumab (P+T) in patients (Pts) with colorectal cancer (CRC) with ERBB2 amplification or overexpression: Results from the TAPUR Study. J. Clin. Oncol. 2020, 38 (suppl. 4), 133. [Google Scholar] [CrossRef]
  107. Siena, S.; Elez, E.; Peeters, M.; André, T.; Van den Eynde, M.; Ng, K.; Cercek, A.; Fountzilas, C.; Sanchez, F.; Hubbard, J.; et al. PD-1 MOUNTAINEER: Open-label, phase 2 study of tucatinib combined with trastuzumab for HER2-positive metastatic colorectal cancer (SGNTUC-017, trial in progress). Ann. Oncol. 2021, 32, S199. [Google Scholar] [CrossRef]
  108. Meric-Bernstam, F.; Beeram, M.; Mayordomo, J.I.; Hanna, D.L.; Ajani, J.A.; Blum Murphy, M.A.; Murthy, R.K.; Piha-Paul, S.A.; Bauer, T.M.; Bendell, J.C.; et al. Single agent activity of ZW25, a HER2-targeted bispecific antibody, in heavily pretreated HER2-expressing cancers. J. Clin. Oncol. 2018, 36 (suppl. 15), 2500. [Google Scholar] [CrossRef]
  109. Nguyen, B.; Fong, C.; Luthra, A.; Smith, S.A.; DiNatale, R.G.; Nandakumar, S.; Walch, H.; Chatila, W.K.; Madupuri, R.; Kundra, R.; et al. Genomic characterization of metastatic patterns from prospective clinical sequencing of 25,000 patients. Cell 2022, 185, 563–575.e11. [Google Scholar] [CrossRef]
  110. Siravegna, G.; Lazzari, L.; Crisafulli, G.; Sartore-Bianchi, A.; Mussolin, B.; Cassingena, A.; Martino, C.; Lanman, R.B.; Nagy, R.J.; Fairclough, S.; et al. Radiologic and Genomic Evolution of Individual Metastases during HER2 Blockade in Colorectal Cancer. Cancer Cell 2018, 34, 148–162.e7. [Google Scholar] [CrossRef] [Green Version]
  111. Raghav, K.P. Sequencing strategies in the management of metastatic colorectal cancer with HER2 amplification. Clin. Adv. Hematol. Oncol. 2022, 20, 86–88. [Google Scholar] [PubMed]
  112. Manca, P.; Corallo, S.; Busico, A.; Lonardi, S.; Corti, F.; Antoniotti, C.; Procaccio, L.; Clavarezza, M.; Smiroldo, V.; Tomasello, G.; et al. The Added Value of Baseline Circulating Tumor DNA Profiling in Patients with Molecularly Hyperselected, Left-sided Metastatic Colorectal Cancer. Clin. Cancer Res. 2021, 27, 2505–2514. [Google Scholar] [CrossRef] [PubMed]
  113. Xu, J.; Meng, Q.; Sun, H.; Zhang, X.; Yun, J.; Li, B.; Wu, S.; Li, X.; Yang, H.; Zhu, H.; et al. HER2-specific chimeric antigen receptor-T cells for targeted therapy of metastatic colorectal cancer. Cell Death Dis. 2021, 12, 1109. [Google Scholar] [CrossRef] [PubMed]
  114. Armstrong, S.A.; Malley, R.; Weinberg, B.A. Molecular Profiling in Metastatic Colorectal Cancer. Oncology 2020, 34, 352–355. [Google Scholar] [PubMed]
  115. Pietrantonio, F.; Di Nicolantonio, F.; Schrock, A.B.; Lee, J.; Tejpar, S.; Sartore-Bianchi, A.; Hechtman, J.F.; Christiansen, J.; Novara, L.; Tebbutt, N.; et al. ALK, ROS1, and NTRK Rearrangements in Metastatic Colorectal Cancer. J. Natl. Cancer Inst. 2017, 109, djx089. [Google Scholar] [CrossRef] [Green Version]
  116. Drilon, A.; Laetsch, T.W.; Kummar, S.; DuBois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
  117. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  118. Paz-Ares, L.; Barlesi, F.; Siena, S.; Ahn, M.J.; Drilon, A.; Conley, A.; Rolfo, C.; Wolf, J.; Seto, T.; Doebele, R.; et al. Patient-reported outcomes from STARTRK-2: A global phase II basket study of entrectinib for ROS1 fusion-positive non-small-cell lung cancer and NTRK fusion-positive solid tumours. ESMO Open 2021, 6, 100113. [Google Scholar] [CrossRef]
  119. He, X.; Jiao, X.D.; Liu, K.; Qin, B.D.; Wu, Y.; Ling, Y.; Liu, J.; Xu, A.Q.; Song, K.; Zang, Y.S. Clinical Responses to Crizotinib, Alectinib, and Lorlatinib in a Metastatic Colorectal Carcinoma Patient With ALK Gene Rearrangement: A Case Report. JCO Precis. Oncol. 2021, 5, PO.20.00534. [Google Scholar] [CrossRef]
  120. Jiang, T.; Wang, G.; Liu, Y.; Feng, L.; Wang, M.; Liu, J.; Chen, Y.; Ouyang, L. Development of small-molecule tropomyosin receptor kinase (TRK) inhibitors for NTRK fusion cancers. Acta Pharm. Sin. B 2021, 11, 355–372. [Google Scholar] [CrossRef]
Figure 1. Therapeutic targets in metastatic colorectal cancer. The main oncogenic drivers. Signaling pathways and their prevalence in patients with metastatic colorectal cancer.
Figure 1. Therapeutic targets in metastatic colorectal cancer. The main oncogenic drivers. Signaling pathways and their prevalence in patients with metastatic colorectal cancer.
Biomedicines 10 01035 g001
Table 1. Selected trial data for emerging biomarkers in mCRC.
Table 1. Selected trial data for emerging biomarkers in mCRC.
BiomarkerReferencesNo. of PatientsTreatmentKey Findings
KRASG12C[16]42SotorasibMedian PFS 4.0 months; PR 7.1%; SD 66.7%
KRYSTAL-1 [17]28Adagrasib + cetuximabORR 43%; DCR 100%
BRAFBEACON [18]665Encorafenib + Cmab vs. Encorafenib + Cmab + binimetinib vs. Cmab + CTMedian OS (9.0 and 8.4 months vs. 5.4 months, p < 0.001); ORR (26% and 20% vs. 2%, p < 0.001)
MSICheckMate-142 [19]119Nivolumab + ipilimumabORR 55%; DCR > 12-week of 80%
Keynote 177 [13]307Pembrolizumab vs. CTPFS 16.5 vs. 8.2 months, p = 0.0002; ORR 43.8% vs. 33.1%
HER-2HERACLES [14,20]32Trastuzumab + lapatinibORR 28%; DCR 69%; median PFS 4.7 months
Destiny-CRC01 [21]53Trastuzumab deruxtecanORR 45%; median PFS 7 months
NTRKNAVIGATE [22]40LarotrectinibORR 50%; median OS 30 months
Cmab cetuximab, CT chemotherapy, DCR disease control rate, mCRC metastatic colorectal cancer, ORR overall response rate, OS overall survival, PFS progression-free survival, PR partial response, SD stable disease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rosati, G.; Aprile, G.; Colombo, A.; Cordio, S.; Giampaglia, M.; Cappetta, A.; Porretto, C.M.; De Stefano, A.; Bilancia, D.; Avallone, A. Colorectal Cancer Heterogeneity and the Impact on Precision Medicine and Therapy Efficacy. Biomedicines 2022, 10, 1035. https://doi.org/10.3390/biomedicines10051035

AMA Style

Rosati G, Aprile G, Colombo A, Cordio S, Giampaglia M, Cappetta A, Porretto CM, De Stefano A, Bilancia D, Avallone A. Colorectal Cancer Heterogeneity and the Impact on Precision Medicine and Therapy Efficacy. Biomedicines. 2022; 10(5):1035. https://doi.org/10.3390/biomedicines10051035

Chicago/Turabian Style

Rosati, Gerardo, Giuseppe Aprile, Alfredo Colombo, Stefano Cordio, Marianna Giampaglia, Alessandro Cappetta, Concetta Maria Porretto, Alfonso De Stefano, Domenico Bilancia, and Antonio Avallone. 2022. "Colorectal Cancer Heterogeneity and the Impact on Precision Medicine and Therapy Efficacy" Biomedicines 10, no. 5: 1035. https://doi.org/10.3390/biomedicines10051035

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

Article Metrics

Back to TopTop