Previous Article in Journal
EUS-Guided Pancreaticobiliary Ablation: Is It Ready for Prime Time?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Etiopathogenesis and Treatment of Colorectal Cancer

by
Mayara Bocchi
1,
Eduardo Vignoto Fernandes
2,*,
Nathália de Sousa Pereira
3 and
Marla Karine Amarante
4
1
Department of Medicine, Federal University of Jataí, Jataí 75801-615, Brazil
2
Postgraduate Program in Bioscience and One Health, Federal University of Jataí, Jataí 75801-615, Brazil
3
Department of Microbiology, Biological Sciences Center, Londrina State University, Londrina 86055-900, Brazil
4
Oncology Laboratory, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, Londrina State University, Londrina 86055-900, Brazil
*
Author to whom correspondence should be addressed.
Immuno 2025, 5(3), 31; https://doi.org/10.3390/immuno5030031
Submission received: 25 June 2025 / Revised: 27 July 2025 / Accepted: 1 August 2025 / Published: 4 August 2025
(This article belongs to the Section Cancer Immunology and Immunotherapy)

Abstract

Human colorectal cancer (CRC) encompasses tumors affecting a segment of the large intestine (colon) and rectum. It is the third most commonly diagnosed malignancy and the second leading cause of cancer deaths worldwide. It is a multifactorial disease, whose carcinogenesis process involves genetic and epigenetic alterations in oncogenes and tumor suppressor genes, including genes related to DNA repair. The pathogenic mechanisms are described based on the pathways of chromosomal instability, microsatellite instability, and CpG island methylator phenotype. When detected early, CRC is potentially curable, and its treatment is based on the pathological characteristics of the tumor and factors related to the patient, as well as on drug efficacy and toxicity studies. Therefore, the aim of this study was to review the pathogenesis and molecular subtypes of CRC and to describe the main targets of disease-directed therapy used in patients refractory to current treatments.

1. Introduction

Human colorectal cancer (CRC) encompasses tumors that affect a segment of the large intestine (the colon) and the rectum [1]. The most recent global estimate shows around 1.066 million new cases of CRC in men, being the third most common tumor among all cancers, with an estimated risk of 23.4/100 thousand. For women, there were around 865 thousand new cases, being the second most common tumor, with an incidence rate of 16.2/100 thousand [2].
The main risk factors associated with the development of the disease include a family history of CRC, age, sedentary lifestyle habits, obesity, and the presence of some inflammatory bowel diseases, such as Crohn’s disease and chronic ulcerative colitis, as well as hereditary intestinal diseases, such as familial adenomatous polyposis (FAP) and hereditary non-polyposis colorectal cancer (HNPCC) [3].
CRC carcinogenesis occurs through genetic and epigenetic alterations in oncogenes and tumor suppressor genes, including genes related to DNA repair mechanisms. The pathogenic mechanisms involve chromosomal instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP) [4] and, depending on the origin of the mutation, colorectal carcinomas can be classified as sporadic, hereditary, and familial [5].
CRC is treatable and, in most cases, is curable when detected early [1]. Assessment of the anatomical extent of the disease through clinicopathological staging is an important prognostic factor for defining CRC treatment, which involves surgical resection for initial tumors, radiotherapy, and chemotherapy [6]. Furthermore, new therapies have been introduced, such as the use of monoclonal antibodies and immunotherapy, aiming at a more targeted treatment, with an improvement in the prognosis and overall survival of patients with CRC [7]. Therefore, the aim of the present study was to review the pathogenesis and molecular subtypes of CRC and describe the main treatments used in the disease.

1.1. Etiology

CRC exhibits diverse etiologies, broadly categorized by mutation origin into sporadic, hereditary, and familial forms [8]. The majority, approximately 70%, are sporadic, emerging in individuals lacking a familial disease history or inherited genetic mutations. Their molecular pathogenesis is complex and multifactorial, stemming from somatic genetic mutations and epigenetic alterations, often linked to modifiable risk factors. Sporadic CRC disproportionately affects older adults, a phenomenon likely attributed to extended temporal exposure to environmental, dietary, and aging-related influences [4,9].
The consumption of foods rich in saturated fats and refined sugars, for instance, predisposes individuals to the development of CRC. This association has been observed as visceral fat induces inflammation in the colorectal region, stimulated by pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), which are released by adipocytes. This inflammatory environment leads to DNA damage, increased cell proliferation, angiogenesis, and immunosuppression. Furthermore, the intake of ultra-processed foods triggers the release of reactive oxygen and nitrogen species in response to inflammation, also culminating in DNA damage to intestinal epithelial cells and neurons of the enteric nervous system [10,11,12].
Comprising around 5% of all CRC cases, hereditary tumors arise from an inherited mutated allele, with a subsequent point mutation in the second allele being critical for carcinoma initiation [5]. These neoplasms are further differentiated based on the presence or absence of associated polyposis. CRC linked to polyposis is predominantly exemplified by FAP, a condition marked by the proliferation of numerous potentially malignant polyps within the colon. FAP is an autosomal dominant disorder, triggered by a mutation in the adenomatous polyposis coli (APC) tumor suppressor gene, situated on chromosome 5q21. Such mutations impair APC protein function, culminating in overactivation of the Wnt/β-catenin signaling pathway, which in turn promotes heightened cell growth, differentiation, dissemination, and adhesion in colorectal cells [13].
Concerning HNPCC, Lynch syndrome is the primary underlying cause, resulting from germline mutations in one of several mismatch repair (MMR) genes (e.g., MSH2, MLH1, MSH6, and PSM2). The inactivation of these genes compromises DNA repair mechanisms, leading to alterations in DNA’s tandem repetitive sequences, specifically microsatellites. This process gives rise to a phenotype termed MSI [14]. It is crucial to note that while characteristic of Lynch syndrome, MSI is not exclusive to it, as approximately 12% of sporadic CRC cases also exhibit this instability [15].
The third category is familial CRC, accounting for an estimated 20–30% of all disease occurrences. The etiology of this CRC form is inherently diverse, with multiple genetic contributors implicated in its development. This is presumed to stem from alterations in a spectrum of less penetrant susceptibility genes, distinct from those underlying established hereditary syndromes [16].

1.2. Pathophysiology of CRC

Colorectal carcinogenesis initiates within the stem cells residing in colonic crypts. This process involves the sequential accumulation of genetic and epigenetic alterations, which progressively inactivate tumor suppressor genes and activate oncogenes. Consequently, these stem cells transform into malignant entities, leading to the formation of an aberrant crypt. This aberrant structure then evolves into a neoplastic precursor lesion, commonly known as a polyp, eventually progressing to an invasive carcinoma [17].
Two principal pathways describe the development of precursor lesions in CRC: the adenoma–carcinoma sequence (also termed the classical or chromosomal instability pathway), responsible for 70–90% of CRC, and the serrated neoplasia pathway, accounting for 10–20% of reported cases [8].
Initially elucidated by Fearon and Vogelstein in 1990, the adenoma–carcinoma sequence (Figure 1) establishes the adenoma as a direct precursor to CRC. Within this pathway, an early adenomatous polyp (typically less than 1 cm in size and exhibiting tubular or tubulovillous histology) undergoes gradual progression. This evolution leads to an advanced adenoma (defined by a size exceeding 1 cm and/or the presence of villous histology), culminating in the development of carcinoma. This progression is fundamentally driven by the cumulative acquisition of genetic and epigenetic alterations [5,18].
The etiology of this pathway is complex and multifaceted, involving the contribution of several genes to disease progression. Key genetic alterations include loss-of-function mutations in the APC tumor suppressor gene, gain-of-function mutations in the Kirsten rat sarcoma oncogene (KRAS), and inactivation of other tumor suppressor genes like deleted in colorectal cancer (DCC) and tumor protein p53 (TP53). These cumulative changes facilitate the crucial transition from advanced adenoma to adenocarcinoma [18,19].
Beyond the adenoma–carcinoma sequence, an estimated 10–20% of colorectal tumors originate through the serrated pathway [20]. Serrated polyps represent a diverse collection of colorectal lesions, encompassing hyperplastic polyps (HPs), sessile serrated adenomas/polyps (SSA/P), traditional serrated adenomas (TSA), and formerly categorized mixed polyps (MP), which exhibit a combination of two or more of these features [21].
Serrated CRC develops via two distinct molecular routes: the sessile serrated pathway and the traditional serrated pathway (Figure 2). The sessile serrated pathway predominantly occurs in the proximal colon and is often initiated by a BRAF gene mutation [22]. This mutation drives the constitutive activation of the mitogen-activated protein kinase (MAPK) pathway, leading to uncontrolled cellular proliferation. Consequently, normal mucosa undergoes transformation into HPs. Further hypermethylation of the p16 tumor suppressor gene and the gene encoding insulin-like growth factor binding protein 7 (IGFBP7) then facilitates their evolution into SSA/P [23].
The progression to sessile serrated adenoma with high-grade dysplasia (SSAD) is primarily driven by epigenetic alterations in the MLH1 gene, leading to subsequent microsatellite instability-high (MSI-high) amplification. Less frequently, this transformation can result from hypermethylation in other genes, such as O(6)-Methylguanine-DNA Methyltransferase (MGMT), maintaining a microsatellite-stable (MSS) profile. Tumors exhibiting MLH1 methylation typically present with a phenotype characterized by BRAF gene mutations, high CpG island methylator phenotype (CIMP-high), and MSI-high. Conversely, when hypermethylation affects other gene promoters, the tumor is generally marked by BRAF mutations, CIMP-high, and an MSS status [24].
The traditional serrated pathway typically manifests in the distal colon and rectum. Its carcinogenic mechanism, leading to malignant transformation, involves mutations in KRAS or BRAF genes within the normal colonic epithelium. These genetic alterations, in conjunction with MGMT or other epigenetic methylation changes, drive the malignant progression to traditional serrated adenoma (TSA), high-grade dysplasia, and ultimately, serrated adenocarcinoma [25].
Beyond the well-established adenoma–carcinoma sequence and the serrated pathway, some researchers suggest an alternative mechanism for MSI [8]. As described by Raskov et al. [26], this pathway is characterized by germline mutations in MMR genes such as MLH1, MSH2, MSH6, and PMS2. It also involves epigenetic silencing, primarily via MLH1 gene hypermethylation, and somatic mutations in the BRAF gene.

1.3. CRC Molecular Pathways

The process of colorectal carcinogenesis involves genetic alterations, described through the CIN and MSI pathways, and epigenetic alterations, such as the CIMP pathway [27]. Initially, these pathways were described as independent mechanisms, however, recent evidence indicates a crosstalk between them [28].

1.3.1. Chromosomal Instability (CIN)

The CIN pathway is defined by alterations in chromosome copy numbers, which, in turn, lead to the activation of pro-growth signaling and/or a reduction in apoptotic pathway activity [29]. This genomic imbalance promotes the development of tumors exhibiting aneuploidy (an abnormal chromosome number) and loss of heterozygosity (LOH) [30]. Key mechanisms within this pathway encompass defects in chromosomal segregation, telomere dysfunction, and altered DNA damage response, all of which can inactivate tumor suppressor genes and activate oncogenes [28].
Inactivating mutations in the APC tumor suppressor gene are commonly observed, leading to constitutive hyperactivation of the Wnt/β-catenin signaling pathway. This pathway plays a pivotal role in stem cell differentiation and cellular proliferation; consequently, its dysregulation can drive uncontrolled cell growth and tumor initiation [31]. Another significant mutation involves the KRAS oncogene. This gene encodes small (21 kDa) guanosine triphosphate (GTP)-binding proteins; when mutated, KRAS becomes constitutively active, promoting unchecked cellular proliferation [32,33].
LOH at chromosome 18q is detected in over 70% of advanced-stage colorectal tumors. This alteration primarily affects the DCC gene and genes encoding proteins involved in the transforming growth factor-beta (TGF-β) pathway, notably SMAD2/4. Under normal physiological conditions, TGF-β signaling inhibits the cell cycle, influencing processes like cell growth, differentiation, and apoptosis. Hence, the inactivation or loss of these genes can result in unconstrained cellular proliferation and evasion of programmed cell death [34]. TGF-β signaling exhibits a paradoxical role in CRC. While it functions as a tumor suppressor in the early stages of tumorigenesis—primarily by inducing cytostasis and apoptosis—in later stages, TGF-β switches to a pro-tumorigenic role. This includes the promotion of epithelial-to-mesenchymal transition (EMT), angiogenesis, immunosuppression, and maintenance of cancer stemness [35,36]. Notably, the immunomodulatory effects of TGF-β within the tumor microenvironment have been implicated in resistance to immunotherapy, and recent findings suggest its activity may serve as a biomarker for therapeutic responsiveness, particularly in immune checkpoint blockade therapies [37].
Additionally, the inactivation of the TP53 tumor suppressor gene frequently contributes to CRC progression [38]. Located on the short arm of chromosome 17 (17p13.1), this gene encodes a 53 kDa phosphoprotein critical for cell cycle regulation, either by inducing DNA repair or initiating apoptosis [39]. Mutations in TP53 compromise DNA repair mechanisms, thereby permitting uncontrolled proliferation of tumor cells and the accumulation of further genetic alterations [40].
From a clinical perspective, colorectal tumors exhibiting CIN are typically linked to a distal anatomical location and are associated with a poorer prognosis [41]. While CIN-positive CRC is predominantly connected to the adenoma–carcinoma sequence, the precise causal relationship remains undefined: it is currently unclear whether CIN drives the accumulation of mutations in key tumor suppressor genes and oncogenes, or if the reverse is true [42].

1.3.2. Microsatellite Instability (MSI)

CRC carcinogenesis is intricately linked to the DNA repair system. This vital system functions during DNA replication, tasked with correcting incorrectly paired nucleotides. When this system is inactivated, it can result in the erroneous accumulation of mismatched nucleotides, thereby predisposing cells to carcinogenesis. Specifically, mutations in MMR genes lead to a deficiency in this critical system, culminating in MSI [43].
Microsatellites are short, repetitive nucleotide sequences (typically 1 to 6 base pairs in length) dispersed across the genome. MSI is defined as any discrepancy in the number of these nucleotide repeats when compared to normal tissue. This instability arises from a failure of the mismatch repair mechanism to accurately correct DNA base errors [26].
MSI is observed in approximately 15% of CRC patients. Of these, 3% are linked to Lynch syndrome, recognized as the most prevalent form of hereditary CRC. Lynch syndrome is an autosomal dominant condition, displaying incomplete yet high penetrance, and is caused by germline mutations in MLH1, MSH2, MSH6, and PMS2 genes [15]. The remaining 12% of patients present with sporadic CRC exhibiting MSI, typically characterized by BRAF gene mutations and MLH1 gene hypermethylation, often associated with the CIMP [44].
The National Comprehensive Cancer Network (NCCN) guidelines for colon cancer recommend assessing MSI or MMR status for all newly diagnosed patients with CRC [45]. The European Society for Medical Oncology (ESMO) guidelines recommend determining MSI status for all patients at the time of diagnosis of metastatic CRC [46]. This assessment can be performed using immunohistochemistry, next-generation sequencing (NGS), or polymerase chain reaction (PCR) techniques. The panel used in the PCR technique comprises five mononucleotide markers: BAT-25, BAT-26, MONO-27, NR-21, and NR-24 [47]. Based on this analysis, MSI status in CRCs is characterized as MSI-high when instability is blocked in at least two markers, MSI-low when instability is present in a single marker, and MSS when no marker instability is observed [28].
Studies have elucidated that the microsatellite-stable (MSS)/MSI-low and MSI-high groups exhibit distinct genomic profiles, accompanied by varying phenotypes and prognoses. Clinically, the MSI-high group typically presents with a predominance in the proximal colon, often characterized by poor histological differentiation and/or mucin production, along with an increase in tumor-infiltrating lymphocytes [48]. Moreover, MSI-high tumors are frequently diagnosed at a younger age, commonly originate from sessile serrated adenomas, and are identified at an earlier stage compared to MSS CRCs, most frequently at stage II [49].
In addition to the analysis of MSI status, the NCCN and ESMO recommend determining tumor gene status for KRAS/NRAS and BRAF mutations, as well as HER2 amplifications, for all patients with metastatic CRC, due to their relevance in prognosis and selection of targeted therapy [45,46].

1.3.3. CpG Island Methylation Phenotype (CIMP)

Epigenetic instability, which underlies the CIMP, is a notable feature in CRC. This pathway is defined by molecular epigenetic alterations that profoundly impact gene expression or function [50]. DNA methylation stands as one of the most prevalent modifications, involving the enzymatic addition of a methyl group (CH3) to the C5 position of the cytosine ring by DNA methyltransferases (DNMTs), yielding 5-methylcytosine. This specific modification typically occurs at CpG dinucleotides, which are DNA regions where a cytosine residue is immediately followed by a guanine residue, linked by a phosphodiester bond [29].
DNA hypermethylation frequently targets CpG islands located within gene promoters. Consequently, this promoter CpG hypermethylation is intrinsically linked to the transcriptional silencing of tumor suppressor genes in malignant cells. In the context of CRC, this phenomenon has been identified within the promoter regions of critical tumor suppressor genes, such as APC and MLH1 [51].
CIMP is identified in approximately 35% of colorectal tumors, often demonstrating associations with proximal anatomical location, female gender, advanced age, mucinous histological features, and an advanced disease stage [29]. Moreover, CIMP exhibits a strong correlation with the serrated pathway of colorectal carcinogenesis and with mutations in the BRAF gene [52]. The mutation of this oncogene is considered a foundational event in the genesis of hyperplastic polyps, as it drives uncontrolled cellular proliferation through the constitutive activation of the MAPK pathway. Following the formation of hyperplastic polyps, CIMP then facilitates their progression to sessile serrated adenomas and subsequent malignant transformation [53].

1.4. Key Molecular Targets for CRC Targeted Therapy

1.4.1. RAS/MAPK/MEK Signaling

  • EGFR and KRAS:
The epidermal growth factor receptor (EGFR), initially isolated and sequenced by S. Cohen in 1980, functions as a transmembrane receptor tyrosine kinase (RTK) within the ErB family. This receptor is encoded by a gene located on the short arm of chromosome 7 and comprises 28 exons [54]. Structurally, EGFR consists of an N-terminal extracellular ligand-binding domain, a lipophilic transmembrane segment, and a C-terminal intracellular tyrosine kinase domain [55]. Upon specific ligand binding to its extracellular domain, EGFR undergoes homo- or heterodimerization. This dimerization induces a conformational change in the receptor, activating the tyrosine kinase domain and leading to the transphosphorylation of tyrosine residues within the C-terminal region [56].
Activation of EGFR initiates a signal transduction cascade, engaging critical pathways such as Ras/MAPK, PI3K/Akt, and JAK/STAT, which ultimately promote cell growth and proliferation. Genetic mutations impacting these pivotal signaling molecules can disrupt normal cellular processes, contributing to carcinogenesis by fostering uncontrolled proliferation, angiogenesis, apoptosis inhibition, cellular invasion, and metastasis [55].
Within the EGFR gene, the most prevalent somatic mutations are deletions in exon 19 (e.g., dels746-750) or a leucine-to-arginine substitution at codon 858 in exon 21 (L858R). Both types of mutations reside within the ATP-binding domain of the tyrosine kinase and are classified as EGFR-activating alterations. These changes frequently co-occur with enhanced production of EGFR ligands, often mediated by autocrine or paracrine feedback loops. Additionally, EGFR mutations can lead to aberrant receptor trafficking, further augmenting signaling and promoting tumor progression [57].
Recent advancements in antineoplastic drug development have focused on agents targeting specific receptors and oncogenic signaling pathways, demonstrating considerable efficacy in CRC management. Therapeutic strategies can involve monoclonal antibodies (mAbs) directed against the extracellular domain of EGFR, exemplified by panitumumab or cetuximab [58].
Cetuximab, a chimeric IgG1 monoclonal antibody, exerts its action by binding to the extracellular domain of EGFR. This interaction blocks receptor phosphorylation and the subsequent biochemical cascade that would otherwise stimulate cell proliferation [59]. For patients with metastatic CRC refractory to prior treatments and irinotecan, the inclusion of cetuximab has shown potential to overcome resistance to this chemotherapeutic agent [60]. Panitumumab, a humanized antibody, similarly targets EGFR and is a therapeutic option for patients with metastatic CRC who have become refractory to chemotherapy [58]. Notably, both these drugs have proven effective in treating CRC patients who lack a mutation in the KRAS gene [61].
The KRAS gene was first described by Scolnick et al. as part of a cancer-inducing virus, the Kirsten sarcoma virus [62]. By the 1980s, it was firmly established as an oncogene with a significant role in the pathogenesis of numerous cancers. KRAS is a member of the Ras protein superfamily, and alongside neuroblastoma rat sarcoma viral oncogene homolog (NRAS) and Harvey rat sarcoma viral oncogene homolog (HRAS) genes, it functions downstream of receptor tyrosine kinases, including EGFR. These proteins collectively regulate vital cellular processes such as proliferation, differentiation, adhesion, apoptosis, and migration [44].
Ras proteins inherently possess GTPase activity, meaning their activation and regulation hinge on guanosine triphosphate (GTP) binding and hydrolysis. In its wild-type state, KRAS protein activation is typically triggered by EGFR binding, leading to enzymatic events that ultimately support the neoplastic phenotype [63]. Deactivation of KRAS occurs when the GTP molecule is hydrolyzed to guanosine diphosphate (GDP) [64]. However, the presence of somatic mutations in this gene renders protein activation independent of growth factor stimulation, resulting in the constitutive activation of downstream signaling cascades, notably the RAF-MEK-ERK and PI3K pathways, thereby contributing to uncontrolled cellular proliferation [65].
Mutations within the KRAS gene are identified in approximately 30% to 40% of CRC patients [66], with the highest frequency observed in codons 12 and 13. Key mutations at codon 12 include glycine-to-aspartic acid (p.G12D), glycine-to-valine (p.G12V), or glycine-to-cysteine (p.G12C) substitutions. At codon 13, the most common mutation is c.38G > C, leading to a glycine-to-aspartic acid change (p.G13D) [67].
The glycine-to-cysteine mutation at position 12 (p.G12C) is located near a crucial region known as the switch-II pocket (S-IIP). This mutation stabilizes the active, GTP-bound form of the KRAS protein, promoting enhanced proliferation and survival in tumor cells. The identification of this allosteric site paved the way for the development of sotorasib, the first inhibitor specifically targeting KRAS p.G12C, approved by the Food and Drug Administration (FDA) in 2021. Sotorasib selectively and irreversibly binds to cysteine 12 (Cys12) within the S-IIP pocket, effectively locking the KRAS p.G12C protein into an inactive conformation [68,69].
According to studies, the use of sotorasib produces considerable clinical benefits and lower gastrointestinal and hepatic adverse effects. Furthermore, the majority of CRC patients presented disease control, with a median duration of stable disease of 5.4 months and a median progression-free survival of 4 months, with conventional therapies having a median progression-free survival rate of 1.9 to 2.1 months [28].
Two novel KRAS p.G12C inhibitors have been proposed in combination with cetuximab. In a phase 1b trial, divarasib showed an overall response rate of 62.5% and a median progression-free survival of 8.1 months in 24 patients with CRC harboring the KRAS p.G12C mutation [70]. Adagrasib (MRTX849) showed an overall response rate of 46% and a median progression-free survival of 6.9 months with combination therapy [71].
  • BRAF:
The BRAF gene encodes the B-Raf protein, a cytoplasmic serine-threonine kinase that plays a pivotal role in activating the MAPK pathway by acting downstream of the Ras protein [72]. Activation of this critical signaling cascade contributes to fundamental cellular processes, including growth, proliferation, differentiation, migration, survival, and angiogenesis [44]. Over 40 distinct missense mutations in BRAF, spanning 24 different codons, have been identified across various human cancers [73]. Approximately 90% of BRAF mutations result from a thymine-to-adenosine transversion at nucleotide T1799A in exon 15, leading to a valine-to-glutamate substitution at codon 600, designated as V600E [74]. This specific mutation drives the constitutive activation of the pro-proliferative MAPK pathway [75].
Approximately 10% of all CRC patients harbor a mutation in the BRAF gene [76], and research indicates a strong association between this mutation and several clinicopathological parameters in metastatic CRC. Tumors carrying BRAF mutations commonly originate from serrated adenomas, are predominantly located in the right colon, and exhibit a higher incidence in women over 60 years of age. Histologically, these tumors are frequently characterized by mucinous features and poor differentiation [77].
In recent years, BRAF V600E-mutated CRC has been recognized as a distinct biological entity. This subtype is typically refractory to standard chemotherapy regimens approved for metastatic CRC, and is consistently associated with a poorer prognosis [78]. Patients with BRAF V600E-mutated tumors demonstrate reduced overall survival and shorter progression-free survival when compared to those with BRAF wild-type CRC [79].
Consequently, targeted therapies against the BRAF gene mutation, including agents like vemurafenib, dabrafenib, and encorafenib, have been proposed. However, preclinical studies involving BRAF V600E-mutated CRC models revealed a rapid feedback activation via EGFR upon BRAF inhibition. This phenomenon indicates that treatment with BRAF inhibitors alone is insufficient to adequately suppress pathway signaling, thereby explaining the limited clinical efficacy observed with single-agent BRAF inhibition in this tumor type [80,81].
To circumvent this paradoxical activation, a combination approach has been proposed, integrating an EGFR inhibitor (e.g., cetuximab, panitumumab) with a MEK inhibitor (e.g., binimetinib, trametinib) [82]. The BEACON study [83] evaluated 665 patients with metastatic CRC with BRAF V600E mutation. Participants were assigned to receive the triple combination (encorafenib, cetuximab and binimetinib), the double combination (encorafenib and cetuximab), or one of the control regimens (irotecan or FOLFIRI).
The results demonstrated that the median overall survival was 9.3 months (95% CI, 8.2 to 10.8) for the triple and double doses and 5.9 months for the control. The confirmed objective response rate was 26.8% for the triple group, 19.5% for the double group, and 1.8% (95% CI, 0.5% to 4.6%) for the control group. Notably, the BEACON study demonstrated that a triplet regimen of encorafenib, cetuximab, and binimetinib yielded significantly prolonged overall survival and a superior response rate compared to standard therapy in patients with metastatic BRAF V600E-mutated CRC [83].

1.4.2. PD-1 and CTLA-4

Approximately 15% of CRC present MSI, a molecular indicator of defective DNA mismatch repair. The high tumor mutational burden and neoantigen load in MSI tumors favor the infiltration of immune effector cells, such as CD8+ tumor-infiltrating lymphocytes, CD4+ T helper 1 cells, and macrophages, resulting in antitumor immune responses. In order to promote immune escape, MSI tumor cells upregulate T cell inhibitory ligands, such as programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) (Figure 3) [84,85].
Immune checkpoint molecules regulate T cell function by activating (costimulatory molecules) or inhibiting signals (coinhibitory molecules). Costimulatory molecules, such as CD28 and CD40, can regulate multiple T cell functions, including activation, specialization, differentiation, and survival, through binding to their B7 ligands (B7-1/CD80 and B7-2/CD86) and CD40L. Coinhibitory molecules, such as programmed death 1 (PD-1) and CTLA-4, can attenuate or suppress the T cell response after initial activation, preventing excessive immune responses and tissue damage [86,87].
PD-1 is an immune checkpoint receptor expressed primarily on T cells, B cells, natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs). Its primary function involves regulating immune responses and preventing unwarranted immune reactions [88]. PD-1 binds to its ligand PD-L1, expressed on the outer membrane of numerous normal cells, such as antigen-presenting cells (APCs), as well as certain cancer cells. The interaction between PD-L1 on cancer cells and PD-1 on T cells transmits inhibitory signals to T cells, resulting in decreased activity and compromised antitumor immune responses, culminating in tumor immune evasion [89,90].
CTLA-4, a cell surface immunoglobulin receptor, is expressed on FoxP3+ Tregs and conventional T cells following activation by the T cell receptor (TCR). As a negative regulator of T cells, it structurally resembles the CD28 receptor and shares the ability to bind B7 ligands on APCs [91]. In the tumor environment, CTLA-4 higher affinity for B7 competes with and depletes CD28 at the immune synapse. Subsequent loss of the B7-CD28 costimulatory signal leads to T cell inactivation and hyporesponsiveness [88].
In this sense, blocking the PD-1/PD-L1 pathway and CTLA-4 may increase the antitumor activity of T cells and, thus, immune control and the ability to kill cancer cells. The presence of immune cells in MSI CRC supports the notion that a treatment based on immunotherapies should provide clinical benefits for this group of patients. Thus, the use of immunomodulatory monoclonal antibodies targeting immune control points has been studied and, in recent years, PD-1 inhibitors, such as pembrolizumab or nivolumab, associated or not with a CTLA-4 inhibitor, such as ipilimumab, were approved by the FDA for the treatment of patients with refractory or metastatic CRC with MSI, increasing progression-free survival and overall survival [92,93].
André et al. [94] reported the efficacy and safety of pembrolizumab versus chemotherapy in patients with MSI-high metastatic CRC from the KEYNOTE-177 trial after 5 years of follow-up. According to the authors, median progression-free survival was 16.5 months with pembrolizumab and 8.2 months with chemotherapy. The 5-year overall survival rates were 55% for the immunotherapy agent versus 44% for chemotherapy. Furthermore, median survival was improved with pembrolizumab (77.5 versus 36.7 months; HR, 0.73; 95% CI 0.53–0.99).
A randomized phase III clinical trial demonstrated that treatment of patients with MSI-high metastatic CRC with nivolumab together with ipilimumab resulted in improved progression-free survival compared to nivolumab alone (hazard ratio 0.62, 95% CI 0.48–0.81; p = 0.0003) [95]. Dual treatment with anti-PD-1/anti-PD-L1 and anti-CTLA-4 appears to reverse the upregulation of other immune checkpoints in T cells, which are induced as a compensatory effect by either drug alone [88].

1.5. Treatment

CRC treatment is based on factors related to the pathological characteristics of the tumor, such as disease staging and the presence of markers such as MSI, KRAS, and BRAF, as well as factors related to the patient (age and presence of comorbidities) and the efficacy and toxicity profiles of the medications. All of these factors help in decision-making regarding the use of adjuvant therapy for these patients [96].
Non-metastatic CRC is initially treated with surgery and, depending on the stage of the disease, adjuvant chemotherapy is recommended [5]. The treatment of high-risk stage II and III patients, i.e., patients with T4 tumors, poorly differentiated histology, lymphovascular and/or perineural invasion, the presence of intestinal obstruction or perforation, positive or indeterminate margins, and inadequate lymph node sampling (<12 dissected lymph nodes), is performed with combined regimens of 5-fluorouracil (5-FU), capecitabine, leucovorin (LV), oxaliplatin and irinotecan, such as the FOLFOX (LV + 5-FU + oxaplatin) and XELOX (capecitabine + oxaplatin) regimens [97].
Approximately 50 to 60% of patients diagnosed with CRC develop metastases [98]. Treatment of metastatic disease involves the administration of single or combined drugs, considering the goals of therapy, the type and timing of prior therapy, the mutational profile of the tumor, and the different toxicity profiles of the drugs. The different strategies are based on the use of chemotherapy regimens such as FOLFOX, XELOX, FOLFIRI (LV + 5-FU + irinotecan), FOLFIRINOX (5-FU + LV + oxaplatin + irinotecan) or trifluridine-tipiracil in combination with anti-EGFR (cetuximab and panitumunab), anti-VEGF (bevacizumab, aflibercept, ramucirumab, regorafenib and fruquintinib) and anti-HER2 (trastuzumab, tucatinib and trastuzumab deruxtecan) agents, KRAS p.G12C inhibitors (sotorasib and adagrasib), BRAF and MEK inhibitors (encorafenib and brinimetinid) and immune checkpoint inhibitors (pembrolizumab, nivolumab and ipilimumab) [6,45,46,99]. Table 1 demonstrates the treatment for unresectable stage IV metastatic CRC.
As can be seen, in addition to classic therapeutic regimens, immunotherapy and targeted therapy regimens are becoming an increasingly important part of the treatment landscape for metastatic CRC [100]. In this sense, a deeper understanding of cancer biology will reveal new targets, develop new therapies, and improve outcomes for patients with the disease. Figure 4 demonstrates the clinical applicability of the molecular classification of CRC in the selection of targeted therapies and prescription of immunotherapy.

2. Conclusions

CRC results from uncontrolled inspection of cells due to factors, whether environmental or genetic, which can culminate in tissue invasion close to the primary tumor or even the development of several metastases. Therefore, the research and review of the genetic and immunological aspects of molecules involved in this disease contribute to better understanding of the pathogenesis and evolution of CRC, providing clinically relevant data that could be of great value in evaluating the prognosis and in designing therapy for patients. Although research related to CRC has advanced, there remain significant gaps that need to be addressed in order to understand the etiology of the disease, improve treatment responses, and prolong patient survival while preserving their quality of life.

Author Contributions

Conceptualization, M.B. and N.d.S.P.; investigation, M.B. and N.d.S.P.; resources, E.V.F.; data curation, E.V.F. and M.K.A.; writing—original draft preparation, M.B. and N.d.S.P.; writing—review and editing, E.V.F. and M.K.A.; supervision, M.K.A.; project administration, M.K.A. All authors have read and agreed to the published version of the manuscript.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

Acknowledgments

The authors are grateful for Brazilian Coordination of Superior Level Staff Improvement (CAPES), for the scholarship granted to M.B.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

17p13.1Chromosome 17
5-FU5-fluorouracil
APCsAntigen-presenting cells
APCAdenomatous polyposis coli
CH3Methyl group
CIMPCpG island methylation phenotype
CINChromosomal instability
CRCColorectal cancer
CTLA-4Cytotoxic T-lymphocyte-associated protein 4
DCCDeleted in colorectal cancer
dels746-750EGFR gene are deletions in exon 19
DNMTsDNA methyltransferases
EGFREpidermal growth factor receptor
EMTEpithelial-to-mesenchymal transition
ESMOEuropean Society for Medical Oncology
FAPFamilial adenomatous polyposis
FDAFood and Drug Administration
FOLFIRILV + 5-FU + irinotecan
FOLFIRINOXLV + 5-FU + irinotecan + oxaplatin
FOLFOXLV + 5-FU + oxaplatin
GDPGuanosine diphosphate
GTPGuanosine triphosphate
HER2Human Epidermal growth factor Receptor-type 2
HNPCCHereditary non-polyposis colorectal cancer
HPsHyperplastic polyps
HRASHarvey rat sarcoma viral oncogene homolog
IGFBP7Insulin-like growth factor binding protein 7
IL-6Interleukin-6
INCANational Cancer Institute
KRASKirsten rat sarcoma
L858RLeucine for arginine in codon 858 of exon 21
LOHLoss of heterozygosity
LVLeucovorin
MAPKMitogen-activated protein kinase
MGMTMethylguanine-DNA Methyltransferase
MMRMismatch repair
MPMixed polyps
MSSMicrosatellites stability
MSIMicrosatellite instability
NCCNNational Comprehensive Cancer Network
NGSNext-generation sequencing
NRASNeuroblastoma rat sarcoma viral oncogene homolog
PCRPolymerase chain reaction
PD-1Programmed cell death protein 1
PD-L1Programmed death-ligand 1
PIK3Phosphatidylinositol 3-kinase
RTKReceptor with tyrosine kinase
S-IIPSwitch-II pocket
SSA/PSessile serrated adenomas/polyps
SSADSessile serrated adenoma with high-grade dysplasia
TCRT cell receptor
TGFβTransforming growth factor-beta
TNF-αTumor necrosis factor-alpha
TP53Gene that encodes the tumor protein p53
TSATraditional serrated adenomas
VEGFVascular endothelial growth factor
XELOXCapecitabine + oxaplatin

References

  1. Ministério da Saúde. Estimativa 2023: Incidência de Câncer no Brasil, 1st ed.; Instituto Nacional de Câncer: Rio de Janeiro, Brazil, 2022; pp. 1–162. Available online: https://www.inca.gov.br/sites/ufu.sti.inca.local/files/media/document/estimativa-2023.pdf (accessed on 20 June 2025).
  2. International Agency for Research on Cancer. Globocan 2020: Cancer Fact Sheets-Colorectal Cancer. Available online: https://gco.iarc.fr/en (accessed on 23 June 2025).
  3. Matsuda, T.; Fujimoto, A.; Igarashi, Y. Colorectal Cancer: Epidemiology, risk factors, and public health strategies. Digestion 2025, 106, 91–99. [Google Scholar] [CrossRef]
  4. Li, Q.; Geng, S.; Luo, H.; Wang, W.; Mo, Y.Q.; Luo, Q.; Wang, L.; Song, G.B.; Sheng, J.P.; Xu, B. Signaling pathways involved in colorectal cancer: Pathogenesis and targeted therapy. Signal Transduct. Target. Ther. 2024, 9, 266. [Google Scholar] [CrossRef]
  5. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal carcinoma: A general overview and future perspectives in colorectal cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef]
  6. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Arain, M.A.; Chen, Y.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Farkas, L.; et al. NCCN guidelines insights: Colon cancer, version 2.2021, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 329–359. [Google Scholar] [CrossRef]
  7. Singh, M.; Morris, V.K.; Bandey, I.N.; Hong, D.S.; Kopetz, S. Advancements in combining targeted therapy and immunotherapy for colorectal cancer. Trends Cancer 2024, 10, 598–609. [Google Scholar] [CrossRef]
  8. 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]
  9. Saha, S.; Ghosh, S.; Ghosh, S.; Nandi, S.; Nayak, A. Unraveling the complexities of colorectal cancer and its promising therapies —An updated review. Int. Immunopharmacol. 2024, 143, 113325. [Google Scholar] [CrossRef] [PubMed]
  10. Hibino, S.; Kawazoe, T.; Kasahara, H.; Itoh, S.; Ishimoto, T.; Sakata-Yanagimoto, M.; Taniguchi, K. Inflammation-Induced Tumorigenesis and Metastasis. Int. J. Mol. Sci. 2021, 22, 5421. [Google Scholar] [CrossRef]
  11. Jankowski, W.M.; Fichna, J.; Tarasiuk-Zawadzka, A. The interplay between diet and the enteric nervous system in the pathophysiology of colorectal cancer. Folia Med. Cracov. 2024, 64, 5–16. [Google Scholar] [CrossRef] [PubMed]
  12. Zhou, Z.; Kleis, L.; Depetris-Chauvin, A.; Jaskulski, S.; Damerell, V.; Michels, K.B.; Gigic, B.; Nöthlings, U.; Panagiotou, G. Beneficial microbiome and diet interplay in early-onset colorectal cancer. EMBO Mol. Med. 2025, 17, 9–30. [Google Scholar] [CrossRef] [PubMed]
  13. Esplin, E.D.; Hanson, C.; Wu, S.; Horning, A.M.; Barapour, N.; Nevins, S.A.; Jiang, L.; Contrepois, K.; Lee, H.; Guha, T.K.; et al. Multiomic analysis of familial adenomatous polyposis reveals molecular pathways associated with early tumorigenesis. Nat. Cancer 2024, 5, 1737–1753. [Google Scholar] [CrossRef]
  14. Dominguez-Valentin, M.; Sampson, J.R.; Seppälä, T.T.; Ten Broeke, S.W.; Plazzer, J.P.; Nakken, S.; Engel, C.; Aretz, S.; Jenkins, M.A.; Sunde, L.; et al. Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: Findings from the Prospective Lynch Syndrome Database. Genet. Med. 2020, 22, 1569. [Google Scholar] [CrossRef]
  15. Roht, L.; Laidre, P.; Tooming, M.; Tõnisson, N.; Nõukas, M.; Nurm, M.; Estonian Biobank Research Team; Roomere, H.; Rekker, K.; Toome, K.; et al. The prevalence and molecular landscape of lynch syndrome in the affected and general population. Cancers 2023, 15, 3663. [Google Scholar] [CrossRef] [PubMed]
  16. Schubert, S.A.; Morreau, H.; de Miranda, N.F.C.C.; van Wezel, T. The missing heritability of familial colorectal cancer. Mutagenesis 2020, 35, 221–231. [Google Scholar] [CrossRef]
  17. Nassar, D.; Blanpain, C. Cancer stem cells: Basic concepts and therapeutic implications. Annu. Rev. Pathol. 2016, 23, 47–76. [Google Scholar] [CrossRef]
  18. Fearon, E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61, 759–767. [Google Scholar] [CrossRef] [PubMed]
  19. Morán, A.; Ortega, P.; de Juan, C.; Fernández-Marcelo, T.; Frías, C.; Sánchez-Pernaute, A.; Torres, A.J.; Díaz-Rubio, E.; Iniesta, P.; Benito, M. Differential colorectal carcinogenesis: Molecular basis and clinical relevance. World J. Gastrointest. Oncol. 2010, 2, 151–158. [Google Scholar] [CrossRef]
  20. Müller, M.F.; Ibrahim, A.E.; Arends, M.J. Molecular pathological classification of colorectal cancer. Virchows Arch. 2016, 469, 125–134. [Google Scholar] [CrossRef]
  21. Mäkinen, M.J.; George, S.M.; Jernvall, P.; Mäkelä, J.; Vihko, P.; Karttunen, T.J. Colorectal carcinoma associated with serrated adenoma--prevalence, histological features, and prognosis. J. Pathol. 2001, 193, 286–294. [Google Scholar] [CrossRef] [PubMed]
  22. O’Brien, M.J.; Yang, S.; Mack, C.; Xu, H.; Huang, C.S.; Mulcahy, E.; Amorosino, M.; Farraye, F.A. Comparison of microsatellite instability, CpG island methylation phenotype, BRAF and KRAS status in serrated polyps and traditional adenomas indicates separate pathways to distinct colorectal carcinoma end points. Am. J. Surg. Pathol. 2006, 30, 1491–1501. [Google Scholar] [CrossRef] [PubMed]
  23. Kriegl, L.; Neumann, J.; Vieth, M.; Greten, F.R.; Reu, S.; Jung, A.; Kirchner, T. Up and downregulation of p16(Ink4a) expression in BRAF-mutated polyps/adenomas indicates a senescence barrier in the serrated route to colon cancer. Mod. Pathol. 2011, 24, 1015–1022. [Google Scholar] [CrossRef] [PubMed]
  24. De Palma, F.D.E.; D’Argenio, V.; Pol, J.; Kroemer, G.; Maiuri, M.C.; Salvatore, F. The molecular hallmarks of the serrated pathway in colorectal cancer. Cancers 2019, 11, 1017. [Google Scholar] [CrossRef]
  25. Nguyen, L.H.; Goel, A.; Chung, D.C. Pathways of colorectal carcinogenesis. Gastroenterology 2020, 158, 291–302. [Google Scholar] [CrossRef]
  26. Raskov, H.; Søby, J.H.; Troelsen, J.; Bojesen, R.D.; Gögenur, I. Driver gene mutations and epigenetics in colorectal cancer. Ann. Surg. 2020, 271, 75–85. [Google Scholar] [CrossRef]
  27. Hultcrantz, R. Aspects of colorectal cancer screening, methods, age and gender. J. Intern. Med. 2021, 289, 493–507. [Google Scholar] [CrossRef]
  28. Hong, S.N. Genetic and epigenetic alterations of colorectal cancer. Intestig. Res. 2018, 16, 327–337. [Google Scholar] [CrossRef]
  29. Jung, G.; Hernández-Illán, E.; Moreira, L.; Balaguer, F.; Goel, A. Epigenetics of colorectal cancer: Biomarker and therapeutic potential. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 111–130. [Google Scholar] [CrossRef]
  30. Aaltonen, L.A.; Peltomäki, P.; Mecklin, J.P.; Järvinen, H.; Jass, J.R.; Green, J.S.; Lynch, H.T.; Watson, P.; Tallqvist, G.; Juhola, M. Replication errors in benign and malignant tumors from hereditary nonpolyposis colorectal cancer patients. Cancer Res. 1994, 54, 1645–1648. [Google Scholar] [PubMed]
  31. Zhao, H.; Ming, T.; Tang, S.; Ren, S.; Yang, H.; Liu, M.; Tao, Q.; Xu, H. Wnt signaling in colorectal cancer: Pathogenic role and therapeutic target. Mol. Cancer 2022, 21, 144. [Google Scholar] [CrossRef]
  32. Jimeno, A.; Messersmith, W.A.; Hirsch, F.R.; Franklin, W.A.; Eckhardt, S.G. KRAS mutations and sensitivity to epidermal growth factor receptor inhibitors in colorectal cancer: Practical application of patient selection. J. Clin. Oncol. 2009, 27, 1130–1136. [Google Scholar] [CrossRef]
  33. Lièvre, A.; Bachet, J.B.; Boige, V.; Cayre, A.; Le Corre, D.; Buc, E.; Ychou, M.; Bouché, O.; Landi, B.; Louvet, C.; et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J. Clin. Oncol. 2008, 26, 374–379. [Google Scholar] [CrossRef]
  34. Jung, B.; Staudacher, J.J.; Beauchamp, D. Transforming growth factor β superfamily signaling in development of colorectal cancer. Gastroenterology 2017, 152, 36–52. [Google Scholar] [CrossRef]
  35. Li, X.; Wu, Y.; Tian, T. TGF-β Signaling in Metastatic Colorectal Cancer (mCRC): From Underlying Mechanism to Potential Applications in Clinical Development. Int. J. Mol. Sci. 2022, 23, 14436. [Google Scholar] [CrossRef]
  36. Calon, A.; Espinet, E.; Palomo-Ponce, S.; Tauriello, D.V.; Iglesias, M.; Céspedes, M.V.; Sevillano, M.; Nadal, C.; Jung, P.; Zhang, X.H.; et al. Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell 2012, 22, 571–584. [Google Scholar] [CrossRef] [PubMed]
  37. Usset, J.; Rosendahl Huber, A.; Andrianova, M.A.; Batlle, E.; Carles, J.; Cuppen, E.; Elez, E.; Felip, E.; Gómez-Rey, M.; Giacco, D.L.; et al. Five latent factors underlie response to immunotherapy. Nat. Genet. 2024, 56, 2112–2120. [Google Scholar] [CrossRef] [PubMed]
  38. Armaghany, T.; Wilson, J.D.; Chu, Q.; Mills, G. Genetic alterations in colorectal cancer. Gastrointest. Cancer. Res. 2012, 5, 19–27. [Google Scholar] [PubMed]
  39. Lane, D.; Levine, A. p53 Research: The past thirty years and the next thirty years. Cold Spring Harb. Perspect. Biol. 2010, 2, a000893. [Google Scholar] [CrossRef]
  40. Malki, A.; ElRuz, R.A.; Gupta, I.; Allouch, A.; Vranic, S.; Al Moustafa, A.E. Molecular mechanisms of colon cancer progression and metastasis: Recent insights and advancements. Int. J. Mol. Sci. 2020, 22, 130. [Google Scholar] [CrossRef]
  41. Lynch, H.T.; de la Chapelle, A. Hereditary colorectal cancer. N. Engl. J. Med. 2003, 348, 919–932. [Google Scholar] [CrossRef]
  42. Pino, M.S.; Chung, D.C. The chromosomal instability pathway in colon cancer. Gastroenterology 2010, 138, 2059–2072. [Google Scholar] [CrossRef]
  43. Wankhede, D.; Yuan, T.; Kloor, M.; Halama, N.; Brenner, H.; Hoffmeister, M. Clinical significance of combined tumour-infiltrating lymphocytes and microsatellite instability status in colorectal cancer: A systematic review and network meta-analysis. Lancet Gastroenterol. Hepatol. 2024, 9, 609–619. [Google Scholar] [CrossRef]
  44. 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]
  45. Benson, A.B.; Venook, A.P.; Adam, M.; Chang, G.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.A.; Cooper, H.S.; Deming, D.; Garrido-Laguna, I.; et al. Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2024, 22, e240029. [Google Scholar] [CrossRef] [PubMed]
  46. Cervantes, A.; Adam, R.; Roselló, S.; Arnold, D.; Normanno, N.; Taïeb, J.; Seligmann, J.; De Baere, T.; Osterlund, P.; Yoshino, T.; et al. Metastatic colorectal cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023, 34, 10–32. [Google Scholar] [CrossRef] [PubMed]
  47. Li, K.; Luo, H.; Huang, L.; Luo, H.; Zhu, X. Microsatellite instability: A review of what the oncologist should know. Cancer Cell Int. 2020, 20, 16. [Google Scholar] [CrossRef]
  48. Boland, C.R.; Thibodeau, S.N.; Hamilton, S.R.; Sidransky, D.; Eshleman, J.R.; Burt, R.W.; Meltzer, S.J.; Rodriguez-Bigas, M.A.; Fodde, R.; Ranzani, G.N.; et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: Development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998, 58, 5248–5257. [Google Scholar]
  49. Ryan, E.; Sheahan, K.; Creavin, B.; Mohan, H.M.; Winter, D.C. The current value of determining the mismatch repair status of colorectal cancer: A rationale for routine testing. Crit. Rev. Oncol. Hematol. 2017, 116, 38–57. [Google Scholar] [CrossRef]
  50. Vila, A.P.S.; Rodrigues, G.H.; Marzochi, L.L.; Oliveira-Cucolo, J.G.; Galbiatti-Dias, A.L.S.; Andrade, R.F.M.; de Santi Neto, D.; Gomes Netinho, J.; Castiglioni, L.; Pavarino, É.C.; et al. Epidemiological and molecular evaluation of BRAF, KRAS, NRAS genes and MSI in the development of colorectal cancer. Gene 2023, 20, 147395. [Google Scholar] [CrossRef] [PubMed]
  51. van Engeland, M.; Derks, S.; Smits, K.M.; Meijer, G.A.; Herman, J.G. Colorectal cancer epigenetics: Complex simplicity. J. Clin. Oncol. 2011, 29, 1382–1391. [Google Scholar] [CrossRef]
  52. Keum, N.; Giovannucci, E. Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 713–732. [Google Scholar] [CrossRef]
  53. Gallois, C.; Pernot, S.; Zaanan, A.; Taieb, J. Colorectal cancer: Why does side matter? Drugs 2018, 78, 789–798. [Google Scholar] [CrossRef]
  54. Li, J.; Ma, X.; Chakravarti, D.; Shalapour, S.; DePinho, R.A. Genetic and biological hallmarks of colorectal cancer. Genes Dev. 2021, 35, 787–820. [Google Scholar] [CrossRef]
  55. Kim, J.E.; Hong, Y.S.; Kim, H.J.; Kim, K.P.; Lee, J.L.; Park, S.J.; Lim, S.B.; Park, I.J.; Kim, C.W.; Yoon, Y.S.; et al. Defective mismatch repair status was not associated with DFS and OS in stage II colon cancer treated with adjuvant chemotherapy. Ann. Surg. Oncol. 2015, 22, S630–S6307. [Google Scholar] [CrossRef] [PubMed]
  56. Protásio, B.M.; Matutino, A.; Lage, L.V.; Santana, I.; Ramos, R.; Sabbaga, J.; Capareli, F.; Saragiotto, D.; Riechelmann, R.; Hoff, P.M. Safety and efficacy of a modified FLOX adjuvant regimen for patients with stage III colorectal cancer treated in the community. Clin. Color. Cancer 2017, 16, 65–72. [Google Scholar] [CrossRef]
  57. Van Cutsem, E.; Nordlinger, B.; Adam, R.; Köhne, C.H.; Pozzo, C.; Poston, G.; Ychou, M.; Rougier, P.; European Colorectal Metastases Treatment Group. Towards a pan-European consensus on the treatment of patients with colorectal liver metastases. Eur. J. Cancer 2006, 42, 2212–2221. [Google Scholar] [CrossRef]
  58. Morris, V.K.; Kennedy, E.B.; Baxter, N.N.; Benson, A.B., III; Cercek, A.; Cho, M.; Ciombor, K.K.; Cremolini, C.; Davis, A.; Deming, D.A.; et al. Treatment of Metastatic Colorectal Cancer: ASCO Guideline. J. Clin. Oncol. 2023, 41, 678–700. [Google Scholar] [CrossRef]
  59. Potocki, P.M.; Wójcik, P.; Chmura, Ł.; Goc, B.; Fedewicz, M.; Bielańska, Z.; Swadźba, J.; Konopka, K.; Kwinta, Ł.; Wysocki, P.J. Clinical Characterization of Targetable Mutations (BRAF V600E and KRAS G12C) in Advanced Colorectal Cancer—A Nation-Wide Study. Int. J. Mol. Sci. 2023, 24, 9073. [Google Scholar] [CrossRef]
  60. Riely, G.J.; Politi, K.A.; Miller, V.A.; Pao, W. Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clin. Cancer Res. 2006, 12, 7232–7241. [Google Scholar] [CrossRef]
  61. Segatto, O.; Anastasi, S.; Alemà, S. Regulation of epidermal growth factor receptor signalling by inducible feedback inhibitors. J. Cell Sci. 2011, 124, 1785–1793. [Google Scholar] [CrossRef]
  62. Mitsudomi, T.; Yatabe, Y. Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J. 2010, 277, 301–308. [Google Scholar] [CrossRef]
  63. Jurišić, V.; Obradovic, J.; Pavlović, S.; Djordjevic, N. Epidermal Growth Factor Receptor Gene in Non- Small-Cell Lung Cancer: The Importance of Promoter Polymorphism Investigation. Anal. Cell. Pathol. 2018, 14, 6192187. [Google Scholar] [CrossRef]
  64. Van Cutsem, E.; Peeters, M.; Siena, S.; Humblet, Y.; Hendlisz, A.; Neyns, B.; Canon, J.L.; Van Laethem, J.L.; Maurel, J.; Richardson, G.; et al. 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. 2007, 25, 1658–1664. [Google Scholar] [CrossRef]
  65. Baselga, J. The EGFR as a target for anticancer therapy-focus on cetuximab. Eur. J. Cancer 2001, 37, S16–S22. [Google Scholar] [CrossRef]
  66. Cunningham, D.; Humblet, Y.; Siena, S.; Khayat, D.; Bleiberg, H.; Santoro, A.; Bets, D.; Mueser, M.; Harstrick, A.; Verslype, C.; et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N. Engl. J. Med. 2004, 351, 337–345. [Google Scholar] [CrossRef]
  67. Tveit, K.M.; Guren, T.; Glimelius, B.; Pfeiffer, P.; Sorbye, H.; Pyrhonen, S.; Sigurdsson, F.; Kure, E.; Ikdahl, T.; Skovlund, E.; et al. 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. 2012, 30, 1755–1762. [Google Scholar] [CrossRef] [PubMed]
  68. Malumbres, M.; Barbacid, M. RAS oncogenes: The first 30 years. Nat. Rev. Cancer 2003, 3, 459–465. [Google Scholar] [CrossRef] [PubMed]
  69. Porru, M.; Pompili, L.; Caruso, C.; Biroccio, A.; Leonetti, C. Targeting KRAS in metastatic colorectal cancer: Current strategies and emerging opportunities. J. Exp. Clin. Cancer Res. 2018, 37, 57. [Google Scholar] [CrossRef]
  70. McCormick, F. KRAS as a therapeutic target. Clin. Cancer Res. 2015, 21, 1797–1801. [Google Scholar] [CrossRef]
  71. Huang, D.; Sun, W.; Zhou, Y.; Li, P.; Chen, F.; Chen, H.; Xia, D.; Xu, E.; Lai, M.; Wu, Y.; et al. Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev. 2018, 37, 173–187. [Google Scholar] [CrossRef] [PubMed]
  72. Haigis, K.M. KRAS alleles: The devil is in the detail. Trends Cancer 2017, 3, 686–697. [Google Scholar] [CrossRef]
  73. Sameer, A.S.; ul Rehman, S.; Pandith, A.A.; Syeed, N.; Shah, Z.A.; Chowdhri, N.A.; Wani, K.A.; Siddiqi, M.A. Molecular gate keepers succumb to gene aberrations in colorectal cancer in Kashmiri population, revealing a high incidence area. Saudi J. Gastroenterol. 2009, 15, 244–252. [Google Scholar] [CrossRef] [PubMed]
  74. Huang, L.; Guo, Z.; Wang, F.; Fu, L. KRAS mutation: From undruggable to druggable in cancer. Signal Transduct. Target. Ther. 2021, 6, 386. [Google Scholar] [CrossRef] [PubMed]
  75. Zhu, C.; Guan, X.; Zhang, X.; Luan, X.; Song, Z.; Cheng, X.; Zhang, W.; Qin, J.J. Targeting KRAS mutant cancers: From druggable therapy to drug resistance. Mol. Cancer 2022, 21, 159. [Google Scholar] [CrossRef]
  76. Desai, J.; Alonso, G.; Kim, S.H.; Cervantes, A.; Karasic, T.; Medina, L.; Shacham-Shmueli, E.; Cosman, R.; Falcon, A.; Gort, E.; et al. Divarasib plus cetuximab in KRAS G12C-positive colorectal cancer: A phase 1b trial. Nat. Med. 2024, 30, 271–278. [Google Scholar] [CrossRef] [PubMed]
  77. Yaeger, R.; Weiss, J.; Pelster, M.S.; Spira, A.I.; Barve, M.; Ou, S.I.; Leal, T.A.; Bekaii-Saab, T.S.; Paweletz, C.P.; Heavey, G.A.; et al. Adagrasib with or without Cetuximab in Colorectal Cancer with Mutated KRAS G12C. N. Engl. J. Med. 2023, 388, 44–54. [Google Scholar] [CrossRef]
  78. Garcia-Carbonero, N.; Martinez-Useros, J.; Li, W.; Orta, A.; Perez, N.; Carames, C.; Hernandez, T.; Moreno, I.; Serrano, G.; Garcia-Foncillas, J. KRAS and BRAF Mutations as Prognostic and Predictive Biomarkers for Standard Chemotherapy Response in Metastatic Colorectal Cancer: A Single Institutional Study. Cells 2020, 9, 219. [Google Scholar] [CrossRef]
  79. Xing, M. BRAF mutation in thyroid cancer. Endocr. Relat. Cancer 2005, 12, 245–262. [Google Scholar] [CrossRef]
  80. Wan, P.T.; Garnett, M.J.; Roe, S.M.; Lee, S.; Niculescu-Duvaz, D.; Good, V.M.; Jones, C.M.; Marshall, C.J.; Springer, C.J.; Barford, D.; et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 2004, 116, 855–867. [Google Scholar] [CrossRef]
  81. Caputo, F.; Santini, C.; Bardasi, C.; Cerma, K.; Casadei-Gardini, A.; Spallanzani, A.; Andrikou, K.; Cascinu, S.; Gelsomino, F. BRAF-mutated colorectal cancer: Clinical and molecular insights. Int. J. Mol. Sci. 2019, 20, 5369. [Google Scholar] [CrossRef]
  82. Cohen, R.; Cervera, P.; Svrcek, M.; Pellat, A.; Dreyer, C.; de Gramont, A.; André, T. BRAF-mutated colorectal cancer: What is the optimal strategy for treatment? Curr. Treat. Options Oncol. 2017, 18, 9. [Google Scholar] [CrossRef]
  83. 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] [PubMed]
  84. Ursem, C.; Atreya, C.E.; Van Loon, K. Emerging treatment options for BRAF-mutant colorectal cancer. Gastrointest. Cancer 2018, 8, 13–23. [Google Scholar] [CrossRef] [PubMed]
  85. Bylsma, L.C.; Gillezeau, C.; Garawin, T.A.; Kelsh, M.A.; Fryzek, J.P.; Sangaré, L.; Lowe, K.A. Prevalence of RAS and BRAF mutations in metastatic colorectal cancer patients by tumor sidedness: A systematic review and meta-analysis. Cancer Med. 2020, 9, 1044–1057. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, X.; Chen, L.J.; Peng, X.F.; Deng, L.; Wang, Y.; Li, J.J.; Guo, D.L.; Niu, X.H. Anti-PD-1/PD-L1 therapy for colorectal cancer: Clinical implications and future considerations. Transl. Oncol. 2024, 40, 101851. [Google Scholar] [CrossRef]
  87. Edner, N.M.; Carlesso, G.; Rush, J.S.; Walker, L.S.K. Targeting co-stimulatory molecules in autoimmune disease. Nat. Rev. Drug Discov. 2020, 19, 860–883. [Google Scholar] [CrossRef] [PubMed]
  88. Lin, K.X.; Istl, A.C.; Quan, D.; Skaro, A.; Tang, E.; Zheng, X. PD-1 and PD-L1 inhibitors in cold colorectal cancer: Challenges and strategies. Cancer Immunol. Immunother. 2023, 72, 3875–3893. [Google Scholar] [CrossRef]
  89. Kuno, S.; Pakpian, N.; Muanprasat, C. The potential role of PD-1/PD-L1 small molecule inhibitors in colorectal cancer with different mechanisms of action. Eur. J. Pharmacol. 2025, 992, 177351. [Google Scholar] [CrossRef]
  90. Sangani, P.S.; Yazdani, S.; Khalili-Tanha, G.; Ghorbani, E.; Al-Hayawi, I.S.; Fiuji, H.; Khazaei, M.; Hassanian, S.M.; Kiani, M.; Ghayour-Mobarhan, M.; et al. The therapeutic impact of programmed death-1 in the treatment of colorectal cancer. Pathol. Res. Pract. 2024, 259, 155345. [Google Scholar] [CrossRef]
  91. Tai, X.; Van Laethem, F.; Pobezinsky, L.; Guinter, T.; Sharrow, S.O.; Adams, A.; Granger, L.; Kruhlak, M.; Lindsten, T.; Thompson, C.B.; et al. Basis of CTLA-4 function in regulatory and conventional CD4(+) T cells. Blood 2012, 119, 5155–5163. [Google Scholar] [CrossRef]
  92. Ciombor, K.K.; Strickler, J.H.; Bekaii-Saab, T.S.; Yaeger, R. BRAF-mutated advanced colorectal cancer: A rapidly changing therapeutic landscape. J. Clin. Oncol. 2022, 40, 2706–2715. [Google Scholar] [CrossRef]
  93. 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]
  94. André, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.J.A.; Smith, D.; Garcia-Carbonero, R.; Alcaide-Garcia, J.; Gibbs, P.; et al. Pembrolizumab versus chemotherapy in microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer: 5-year follow-up from the randomized phase III KEYNOTE-177 study. Ann. Oncol. 2025, 36, 277–284. [Google Scholar] [CrossRef]
  95. André, T.; Elez, E.; Lenz, H.J.; Jensen, L.H.; Touchefeu, Y.; Van Cutsem, E.; Garcia-Carbonero, R.; Tougeron, D.; Mendez, G.A.; Schenker, M.; et al. Nivolumab plus ipilimumab versus nivolumab in microsatellite instability-high metastatic colorectal cancer (CheckMate 8HW): A randomised, open-label, phase 3 trial. Lancet 2025, 405, 383–395. [Google Scholar] [CrossRef]
  96. 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]
  97. Frank, M.H.; Wilson, B.J.; Gold, J.S.; Frank, N.Y. Clinical implications of colorectal cancer stem cells in the age of single-cell omics and targeted therapies. Gastroenterology 2021, 160, 1947–1960. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, X.; Wu, T.; Cai, X.; Dong, J.; Xia, C.; Zhou, Y.; Ding, R.; Yang, R.; Tan, J.; Zhang, L.; et al. Neoadjuvant immunotherapy for MSI-H/dMMR locally advanced colorectal cancer: New strategies and unveiled opportunities. Front. Immunol. 2022, 17, 795972. [Google Scholar] [CrossRef]
  99. Martianov, A.S.; Mitiushkina, N.V.; Ershova, A.N.; Martynenko, D.E.; Bubnov, M.G.; Amankwah, P.; Yanus, G.A.; Aleksakhina, S.N.; Tiurin, V.I.; Venina, A.R.; et al. KRAS, NRAS, BRAF, HER2 and MSI status in a large consecutive series of colorectal carcinomas. Int. J. Mol. Sci. 2023, 24, 4868. [Google Scholar] [CrossRef] [PubMed]
  100. Picard, E.; Verschoor, C.P.; Ma, G.W.; Pawelec, G. Relationships between immune landscapes, genetic subtypes and responses to immunotherapy in colorectal cancer. Front. Immunol. 2020, 6, 369. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Adenoma–carcinoma progression model in CRC. In the adenoma–carcinoma sequence, an initial adenomatous polyp slowly progresses to advanced adenoma and subsequent cancer through genetic and epigenetic alterations, such as loss of function of the APC gene, with changes in the Wnt signaling pathway; gain-of-function in the oncogene KRAS, through activation of the MAPK pathway; loss of heterozygosity on chromosome 18q in tumor suppressor genes, such as DCC, SMAD2, and SMAD4, activating the PIK3 and TGFβ signaling pathways. Furthermore, inactivation of the TP53 tumor suppressor gene may also occur. Alterations in the aforementioned signaling pathways contribute to uncontrolled proliferation and survival of cancer cells, culminating in the development of CRC. APC—adenomatous polyposis coli; KRAS—Kirsten rat sarcoma; MAPK—mitogen-activated protein kinase; LOH—loss of heterozygosity; DCC—deleted in colorectal cancer; PIK3—Phosphatidylinositol 3-kinase; TGF-β—transforming growth factor-beta; TP53—gene that encodes the tumor protein p53. Source: author’s own work (Created with BioRender.com).
Figure 1. Adenoma–carcinoma progression model in CRC. In the adenoma–carcinoma sequence, an initial adenomatous polyp slowly progresses to advanced adenoma and subsequent cancer through genetic and epigenetic alterations, such as loss of function of the APC gene, with changes in the Wnt signaling pathway; gain-of-function in the oncogene KRAS, through activation of the MAPK pathway; loss of heterozygosity on chromosome 18q in tumor suppressor genes, such as DCC, SMAD2, and SMAD4, activating the PIK3 and TGFβ signaling pathways. Furthermore, inactivation of the TP53 tumor suppressor gene may also occur. Alterations in the aforementioned signaling pathways contribute to uncontrolled proliferation and survival of cancer cells, culminating in the development of CRC. APC—adenomatous polyposis coli; KRAS—Kirsten rat sarcoma; MAPK—mitogen-activated protein kinase; LOH—loss of heterozygosity; DCC—deleted in colorectal cancer; PIK3—Phosphatidylinositol 3-kinase; TGF-β—transforming growth factor-beta; TP53—gene that encodes the tumor protein p53. Source: author’s own work (Created with BioRender.com).
Immuno 05 00031 g001
Figure 2. CRC serrated pathway. Transformation of normal colon mucosa to serrated adenocarcinoma by two different molecular pathways: the sessile serrated pathway and the traditional serrated pathway. KRAS—Kirsten rat sarcoma; p16—tumor suppressor gene p16; IGFB7—insulin-like growth factor-binding protein 7; HPs—hyperplastic polyps; SSA/P—sessile serrated adenoma/polyp; SSAD—sessile serrated adenoma with high-grade dysplasia; TSA—traditional serrated adenoma; TSA HGD—traditional serrated adenoma with high-grade dysplasia. Source: author’s own work.
Figure 2. CRC serrated pathway. Transformation of normal colon mucosa to serrated adenocarcinoma by two different molecular pathways: the sessile serrated pathway and the traditional serrated pathway. KRAS—Kirsten rat sarcoma; p16—tumor suppressor gene p16; IGFB7—insulin-like growth factor-binding protein 7; HPs—hyperplastic polyps; SSA/P—sessile serrated adenoma/polyp; SSAD—sessile serrated adenoma with high-grade dysplasia; TSA—traditional serrated adenoma; TSA HGD—traditional serrated adenoma with high-grade dysplasia. Source: author’s own work.
Immuno 05 00031 g002
Figure 3. Immune cell infiltration into RCC and tumor cell immune escape by upregulation of PD-L1 and CTLA-4. PD-L1—Programmed death-ligand 1; CTLA-4—cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Source: author’s own work (Created with BioRender.com).
Figure 3. Immune cell infiltration into RCC and tumor cell immune escape by upregulation of PD-L1 and CTLA-4. PD-L1—Programmed death-ligand 1; CTLA-4—cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Source: author’s own work (Created with BioRender.com).
Immuno 05 00031 g003
Figure 4. Overview of the major molecular pathways in CRC—CIN, MSI, and CIMP—and their respective associations with genetic alterations, tumor features, prognosis, and therapeutic strategies. APC—adenomatous polyposis coli; KRAS—Kirsten rat sarcoma; TP53—gene that encodes the tumor protein p53. Source: author’s own work (Created with BioRender.com).
Figure 4. Overview of the major molecular pathways in CRC—CIN, MSI, and CIMP—and their respective associations with genetic alterations, tumor features, prognosis, and therapeutic strategies. APC—adenomatous polyposis coli; KRAS—Kirsten rat sarcoma; TP53—gene that encodes the tumor protein p53. Source: author’s own work (Created with BioRender.com).
Immuno 05 00031 g004
Table 1. Treatment for unresectable stage IV metastatic CRC.
Table 1. Treatment for unresectable stage IV metastatic CRC.
Treatment LineTumor CharacteristicsTreatment
First-Line TreatmentRAS-wt, BRAF-wt, MSS:
  • Left colon
FOLFOX or FOLFIRI + Cetuximab or Panitumumab
  • Right colon
FOLFOX, FOLFIRI or FOLFOXIRI + Bevacizumab
RAS-mutFOLFOX, FOLFIRI or FOLFOXIRI + Bevacizumab
BRAF-mutFOLFOX + Cetuximab + Encorafenib
MMR/MSI-high or MMR/MSI-high + BRAF-mutPembrolizumab
Nivolumab + Ipilimumab
Second-Line TreatmentRAS-mut, Anti-EGFR naïve:
  • Left colon
FOLFIRI or Irinotecan + Cetuximab or Panitumumab
  • Right colon
5-FU or Capecitabine-based ChT + Bevacizumab, Ramucirumabe or Aflibercept
BRAF V600E-mutEncorafenib + Cetuximab
If not previously used
MMR/MSI-highNivolumab + Ipilimumab (Patients not receiving immunotherapy)
Pembrolizumab
KRAS p.G12CCetuximab + Adagradib
Panitumumab + Sotorasib
Third-Line TreatmentRAS-wt, BRAF-wt, HER2+Trastuzumab + Tucatinib
Trastuzumab deruxtecan
RAS-wt, BRAF-wtTrifluridine/Tipiracil + Bevacizumab
Cetuximab or Panitumumab (If not previously used)
Irinotecan + Cetuximab
Regorafenib
Trifluridine/Tipiracil or Fruquintinib
RAS-mutTrifluridine/Tipiracil + Bevacizumab
Regorafenib
Trifluridine/Tipiracil or Fruquintinib
KRAS p.G12CCetuximab + Adagradib
Panitumumab + Sotorasib
If not previously used
BRAF-mutEncorafenib + Cetuximab (If not previously used)
Trifluridine/Tipiracil + Bevacizumab
Regorafenib
Trifluridine/Tipiracil
Fruquintinib
Legend: mut—mutant; wt—wild type.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bocchi, M.; Fernandes, E.V.; Pereira, N.d.S.; Amarante, M.K. Etiopathogenesis and Treatment of Colorectal Cancer. Immuno 2025, 5, 31. https://doi.org/10.3390/immuno5030031

AMA Style

Bocchi M, Fernandes EV, Pereira NdS, Amarante MK. Etiopathogenesis and Treatment of Colorectal Cancer. Immuno. 2025; 5(3):31. https://doi.org/10.3390/immuno5030031

Chicago/Turabian Style

Bocchi, Mayara, Eduardo Vignoto Fernandes, Nathália de Sousa Pereira, and Marla Karine Amarante. 2025. "Etiopathogenesis and Treatment of Colorectal Cancer" Immuno 5, no. 3: 31. https://doi.org/10.3390/immuno5030031

APA Style

Bocchi, M., Fernandes, E. V., Pereira, N. d. S., & Amarante, M. K. (2025). Etiopathogenesis and Treatment of Colorectal Cancer. Immuno, 5(3), 31. https://doi.org/10.3390/immuno5030031

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop