Colorectal cancer (CRC) remains one of the most prevalent malignancies worldwide, ranking as the fourth most common cause of cancer-related mortality and morbidity17. Despite declining overall incidence rates, early-onset CRC (diagnosed before age 50) has been increasing, presenting a concerning epidemiological shift that demands attention from healthcare systems globally5. Approximately 20% of CRC cases are metastatic at diagnosis, making them particularly challenging to treat effectively and resulting in poor outcomes for affected patients2. The development of CRC involves complex interactions between genetic, environmental, and lifestyle factors, including diet, sedentary behavior, smoking, excessive alcohol consumption, obesity, gut microbiota imbalances, and diabetes17. This comprehensive review examines the molecular mechanisms underlying CRC pathogenesis, evaluates the key signaling pathways involved, and provides an evidence-based assessment of both established and emerging treatment approaches.
Molecular Mechanisms and Pathogenesis
Genetic Alterations and Key Mutations
The development of colorectal cancer follows a complex multistep process involving the accumulation of genetic and epigenetic alterations that transform normal colonic and rectal epithelium into invasive carcinoma14. These molecular changes affect several critical pathways that regulate cell growth, differentiation, and survival, ultimately leading to the malignant phenotype characteristic of CRC14. The p53 tumor suppressor gene is frequently mutated in CRC, with alterations observed in a significant proportion of cases6. Under normal conditions, p53 functions as a transcription factor that induces cell cycle arrest, senescence, and apoptosis under cellular stress, serving as a critical safeguard against cancer development6. However, when mutated, p53 fails to perform these protective functions, contributing to cancer progression and conferring resistance to current therapies, which typically results in poor prognosis for affected patients6.
RAS gene family mutations, particularly in KRAS and NRAS, represent another significant class of genetic alterations in CRC that have profound implications for treatment selection11. These mutations lead to constitutive activation of downstream signaling pathways, promoting uncontrolled cellular proliferation and survival even in the absence of appropriate growth signals1011. The identification of RAS mutation status has become standard practice in the molecular workup of CRC patients, as it strongly predicts resistance to anti-EGFR therapies and helps guide treatment decision-making in the metastatic setting11. Similarly, BRAF mutations, especially the V600E variant, have established clinical utility in CRC management and are associated with distinct clinicopathological features and therapeutic implications11. Patients harboring BRAF V600E mutations typically experience more aggressive disease with poorer outcomes compared to those with BRAF wild-type tumors, necessitating specific treatment considerations and often more intensive therapeutic approaches1011.
DNA mismatch repair (MMR) deficiency, which results in microsatellite instability (MSI), represents another important molecular characteristic with significant implications for treatment selection11. MMR-deficient tumors accumulate mutations at an accelerated rate, creating a hypermutated phenotype that generates numerous neoantigens and increased immunogenicity1118. This molecular feature has emerged as a key biomarker for immunotherapy response, with MMR-deficient tumors showing remarkable sensitivity to immune checkpoint inhibitors that can effectively harness the patient's immune system against the cancer1118.
Signaling Pathways in CRC Pathogenesis
Several interconnected signaling pathways drive CRC development and progression, creating a complex molecular network that regulates various aspects of tumor biology1417. The Wnt/β-catenin pathway is frequently dysregulated in CRC, leading to increased cellular proliferation and inhibition of apoptosis614. Aberrations in this pathway, often involving mutations in the APC gene or other key components, are typically early events in CRC carcinogenesis and set the stage for subsequent molecular alterations614. The PI3K/AKT/mTOR pathway, activated by various growth factors, plays a crucial role in regulating cell growth, proliferation, survival, and metabolism in CRC17. Mutations or alterations in components of this pathway, including PIK3CA, PTEN, and AKT, contribute significantly to cancer progression and resistance to therapy by promoting cell survival and metabolic adaptations favorable to tumor growth1417.
MAPK signaling, comprising three major subfamilies, is intricately involved in regulating diverse cellular activities including proliferation, differentiation, motility, and survival in the context of CRC development17. Dysregulation of MAPK signaling, often through upstream RAS or BRAF mutations, contributes significantly to CRC pathogenesis by promoting uncontrolled cell division and resistance to apoptotic signals617. Growth factors and their receptors, including Epidermal Growth Factor Receptor (EGFR), Insulin-like Growth Factor-1 Receptor (IGF-1R), and Vascular Endothelial Growth Factor-A (VEGF-A), are frequently overexpressed in CRC and serve as important therapeutic targets17. These growth factors sustain cancer cell proliferation, survival, motility, and invasion through activation of downstream signaling pathways including PI3K/AKT/mTOR and MAPK, creating a permissive environment for tumor progression and metastasis17.
Inflammation and Cancer Stem Cells
The link between chronic inflammation and cancer development is particularly strong in CRC, with systemic inflammatory responses playing a pivotal role in disease progression and metastasis1. Several inflammatory biomarkers, including neutrophil/lymphocyte ratio (NLR), lymphocyte/monocyte ratio (LMR), and platelet/lymphocyte ratio (PLR), have demonstrated prognostic value in CRC and can help identify patients at higher risk of poor outcomes1. These ratios, derived from routine blood tests, reflect the balance between host inflammatory and immune responses, providing valuable information about the tumor's interaction with the host immune system and the overall inflammatory state1. The evaluation of these inflammatory ratios, while showing promise as prognostic factors, currently demonstrates limited clinical utility when used individually, suggesting that integrated approaches combining multiple biomarkers may yield more robust predictive models1.
The heterogeneous nature of CRC is increasingly attributed to colorectal cancer stem cells (CCSCs), a small subpopulation with stemness behaviors responsible for tumor progression, recurrence, and therapy resistance2. These cells possess self-renewal and pluripotency capabilities, allowing them to regenerate tumor tissue and resist conventional treatments, thus serving as a reservoir for cancer recurrence and metastasis2. CCSCs are likely generated through genetic alterations in signaling pathways that control self-renewal and pluripotency in normal stem cells, including Wnt/β-catenin, Notch, and Hedgehog pathways2. Various deregulated microRNAs (miRNAs) have been identified that modulate stemness features by regulating cell cycle gene expression, epithelial-mesenchymal transition, metastasis, and drug-resistance mechanisms in CCSCs, presenting potential targets for novel therapeutic approaches2.
Evidence-Based Treatment Approaches
Conventional Treatment Strategies
Surgery remains the primary curative approach for localized CRC, with the specific technique determined by tumor location, stage, and patient-specific factors7. For early-stage lesions, endoscopic resection may be sufficient, while more advanced disease requires colectomy with appropriate lymph node dissection to ensure complete removal of the primary tumor and any potentially involved lymph nodes7. Surgical approaches are supported by robust evidence and represent the standard of care for resectable disease, with post-operative outcomes significantly improved by advances in surgical techniques and perioperative care7. Following surgery for higher-risk disease, adjuvant chemotherapy is often recommended to eradicate micrometastatic disease and reduce the risk of recurrence, with several evidence-based regimens established in clinical practice guidelines7.
Chemotherapy has historically been the conventional treatment modality for advanced CRC, particularly metastatic disease, with common regimens including combinations of fluoropyrimidines, oxaliplatin, and irinotecan8. Despite their widespread use and established efficacy, chemotherapy approaches face significant challenges, including systemic toxicity and the development of resistance mechanisms that ultimately limit their effectiveness8. Chemoresistance represents one of the major obstacles in CRC treatment, significantly limiting efficacy and influencing patient prognosis8. To overcome this resistance, numerous strategies are being investigated, including drug repurposing, gene therapy approaches, and novel drug delivery systems designed to enhance therapeutic efficacy while minimizing adverse effects8.
Targeted Therapies and Precision Medicine
Several targeted therapies have demonstrated efficacy in molecularly-defined subsets of CRC, representing significant advances in personalized medicine approaches1415. Anti-EGFR antibodies (cetuximab, panitumumab) have shown clinical benefit in RAS wild-type metastatic CRC, improving response rates and survival outcomes by blocking EGFR-mediated signaling that drives tumor growth and proliferation15. The efficacy of these agents is strongly dependent on the absence of RAS mutations, highlighting the importance of comprehensive molecular testing prior to treatment initiation to identify patients most likely to benefit from this therapeutic approach1115. Antiangiogenic agents targeting the VEGF pathway, such as bevacizumab, have demonstrated efficacy in metastatic CRC irrespective of molecular subtype by inhibiting tumor angiogenesis1517. These agents reduce blood supply to the tumor, thereby restricting growth and spread, and have become standard components of many first-line treatment regimens for metastatic disease15.
BRAF inhibitors, particularly in combination with other targeted agents, have shown promise in patients with BRAF V600E-mutated CRC, although the durability of response remains limited compared to their efficacy in other malignancies like melanoma10. The development of resistance to BRAF inhibition occurs relatively quickly in CRC, necessitating combination approaches that simultaneously target multiple nodes in the signaling network to achieve more durable responses1015. Ongoing research efforts are focused on understanding resistance mechanisms and developing more effective combination strategies to overcome the challenges associated with targeting BRAF-mutant CRC1015.
Immune checkpoint inhibitors have demonstrated remarkable efficacy in MMR-deficient/MSI-high CRC, leading to regulatory approvals in this molecular subtype18. These agents work by disinhibiting the antitumoral T-cell response, allowing the immune system to recognize and attack cancer cells more effectively by removing the brakes that cancer cells place on immune function418. The success of immunotherapy in MMR-deficient tumors contrasts sharply with its limited efficacy in microsatellite stable (MSS) CRC, which represents approximately 85% of cases18. This discrepancy is attributed to the lower mutational burden and reduced immunogenicity of MSS tumors, highlighting the need for strategies to convert these "cold" tumors into more immunologically active lesions that might respond to immunotherapeutic approaches18.
Emerging Approaches with Developing Evidence
Numerous novel strategies are being investigated to overcome treatment resistance and improve outcomes in CRC, representing the frontier of research in this field81215. Photodynamic therapy (PDT) is emerging as a promising minimally invasive approach that utilizes photosensitizers and specific light wavelengths to induce cell death in targeted tumor tissues12. Recent advances in PDT techniques include the development of novel photosensitizers with improved tumor selectivity, enhanced light sources for more precise energy delivery, and innovative delivery methods to optimize treatment efficacy while minimizing damage to surrounding normal tissues12. While still evolving, PDT represents a potentially valuable addition to the therapeutic armamentarium for selected CRC cases, particularly in settings where conventional approaches may be limited by patient factors or disease characteristics12.
Gene therapy approaches, including ribozymes, RNA interference (RNAi), CRISPR/Cas9, and epigenetic therapy, target specific genetic alterations involved in CRC pathogenesis and offer the potential for highly selective anticancer effects8. These technologies enable precise manipulation of gene expression and function, potentially addressing the fundamental molecular drivers of CRC development and progression8. Drug repurposing strategies investigate the anticancer effects of medications developed for other indications, with agents such as nonsteroidal anti-inflammatory drugs (NSAIDs), metformin, dichloroacetate, enalapril, ivermectin, and melatonin showing promising activity in preclinical CRC models8. These approaches offer the advantage of established safety profiles and potentially accelerated clinical development timelines compared to novel compounds8.
Novel drug delivery systems, including nanocarriers, liposomes, exosomes, and hydrogels, aim to improve the efficacy and reduce the toxicity of existing therapies by enhancing targeted delivery to tumor tissues while sparing normal cells8. These technologies can overcome biological barriers, improve pharmacokinetic properties, and achieve higher local drug concentrations within tumors, potentially enhancing therapeutic index and overcoming resistance mechanisms8. Combination therapies are increasingly being explored to overcome resistance mechanisms and exploit synergistic effects between different treatment modalities815. These include combinations of targeted therapies targeting different nodes in the same or complementary pathways, immunotherapy with conventional or targeted agents, and novel combinations designed to convert immunologically "cold" tumors into more responsive lesions81518.
Screening and Prevention Strategies
Effective screening represents a critical approach to reducing CRC mortality through early detection and intervention when treatment is most effective913. Colonoscopy has demonstrated significant efficacy in reducing CRC incidence and mortality, with studies showing a 7 percentage point reduction compared to sigmoidoscopy screening, translating to approximately 30% additional benefit9. This superior performance is attributed to colonoscopy's ability to visualize the entire colon and rectum, allowing detection and removal of precancerous polyps throughout the colorectum rather than just the distal portions accessible by sigmoidoscopy9. Despite its demonstrated efficacy, colonoscopy screening faces challenges related to resource requirements, patient acceptability, and access disparities that limit its implementation in many healthcare settings913.
Stool-based tests, particularly the fecal immunochemical test (FIT), have shown high participation rates in structured screening programs (85.8% in Latin American studies) and represent effective first-line screening tools that can improve population coverage13. These non-invasive tests detect occult blood in stool, which may indicate the presence of CRC or advanced adenomas, and serve as a triage mechanism to identify individuals who would benefit most from diagnostic colonoscopy13. However, the effectiveness of stool-based screening depends critically on appropriate follow-up with colonoscopy for positive results, with studies showing that only 56% of patients receive such follow-up within one year of a positive stool test in the United States16. This substantial gap in follow-up represents a significant missed opportunity for early intervention and highlights the importance of developing systems to ensure complete diagnostic evaluation after positive screening results16.
The COVID-19 pandemic has further disrupted CRC screening programs and follow-up services, creating a backlog of delayed colonoscopies that threatens to reverse progress in reducing CRC mortality16. Addressing this backlog and implementing targeted interventions to improve follow-up rates, particularly among underserved populations with historically lower screening participation, will be essential to achieving the full potential benefits of CRC screening programs16. Factors associated with lower follow-up rates include race, ethnicity, insurance type, and healthcare organization characteristics, suggesting the need for multifaceted approaches to address barriers and reduce disparities in screening completion16.
Future Directions and Research Priorities
Current research is focused on several key areas to improve CRC outcomes through enhanced understanding of disease biology and development of more effective therapeutic strategies141518. Developing a better understanding of the molecular networks in CRC enables more precise and personalized treatment approaches tailored to the specific genetic and epigenetic alterations present in individual tumors14. This precision medicine paradigm aims to maximize therapeutic efficacy while minimizing unnecessary toxicity by matching patients with the treatments most likely to benefit their particular molecular profile1415. Investigating novel combinations of existing therapies to overcome resistance mechanisms and enhance efficacy represents another important research priority, with particular emphasis on approaches that target complementary pathways or address known resistance mechanisms815.
Exploring strategies to make immunotherapy effective in MSS CRC, which represents the majority of cases, could potentially transform the treatment landscape for patients with this molecular subtype18. Approaches under investigation include combinations of immune checkpoint inhibitors with tyrosine kinase inhibitors and other targeted therapies that may create synergistic effects and enhance antitumor immune responses in the tumor microenvironment18. Addressing the rising incidence of early-onset CRC through research into its unique risk factors and biology represents an emerging priority, with particular focus on environmental exposures, dietary patterns, and the gut microbiome as potential contributors to this concerning trend520. Understanding the drivers of early-onset CRC could inform both prevention strategies and treatment approaches for this increasingly important patient population5.
Improving screening implementation and follow-up to maximize the impact of early detection remains a critical public health priority that could significantly reduce CRC mortality1316. Innovations in screening modalities, strategies to increase participation rates, and interventions to ensure appropriate follow-up after positive screening tests are all essential components of comprehensive CRC control programs1316. The development and validation of novel biomarkers for early detection, risk stratification, and treatment selection represents another promising avenue for improving CRC outcomes across the continuum from prevention to advanced disease management1214.
Conclusion
Colorectal cancer represents a complex, heterogeneous disease with multiple genetic alterations, dysregulated signaling pathways, and diverse clinical presentations that collectively determine its behavior and response to treatment. Significant progress has been made in understanding its molecular pathogenesis, from key mutations in genes like p53, RAS, and BRAF to dysregulation of critical signaling pathways including Wnt/β-catenin, PI3K/AKT/mTOR, and MAPK cascades. The identification of cancer stem cells and elucidation of the complex inflammatory processes involved in CRC progression have further advanced our understanding of this malignancy and revealed potential therapeutic targets.
Conventional approaches including surgery and chemotherapy remain the backbone of CRC treatment, while molecularly-guided targeted therapies and immunotherapy have transformed the management of specific CRC subtypes. The efficacy of these approaches varies considerably based on molecular features, highlighting the importance of comprehensive biomarker testing to guide personalized treatment strategies. Despite these advances, substantial challenges remain, particularly in overcoming treatment resistance and developing effective therapies for currently untreatable molecular subtypes such as microsatellite stable tumors that do not respond to immunotherapy.
The rising incidence of early-onset CRC presents a concerning trend that requires further investigation into potential environmental, dietary, and microbiome-related risk factors. Effective screening programs, with proper follow-up of positive results, represent the most promising strategy for reducing CRC mortality through early detection and intervention. As research continues to unravel the complex biology of CRC and develop novel therapeutic approaches, a multidisciplinary, precision medicine-based approach that integrates molecular profiling, targeted therapies, immunotherapy, and conventional treatment modalities offers the greatest promise for improving outcomes in this common but challenging malignancy.
Citations:
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9833126/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7915330/
- https://pubmed.ncbi.nlm.nih.gov/37451055/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5943429/
- https://pubmed.ncbi.nlm.nih.gov/36744311/
- https://pubmed.ncbi.nlm.nih.gov/25574081/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6433585/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10242875/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10905314/
- https://pubmed.ncbi.nlm.nih.gov/35486097/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5989850/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10418644/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10835514/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9018988/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9950860/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9856942/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8508474/
- https://pubmed.ncbi.nlm.nih.gov/35658496/
- https://pubmed.ncbi.nlm.nih.gov/31130830/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7314664/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10698107/
- https://pubmed.ncbi.nlm.nih.gov/36740959/
- https://pubmed.ncbi.nlm.nih.gov/23665275/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7834960/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6946738/
- https://www.semanticscholar.org/paper/a915daba4d9b23bc9379b225b5f302fb1946dc9e
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6634269/
- https://pubmed.ncbi.nlm.nih.gov/31792582/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10080378/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10341111/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10339147/
- https://www.semanticscholar.org/paper/7a40194a53e1adf477e9d9c7f9ffbc76eb848a7d
- https://pubmed.ncbi.nlm.nih.gov/24876728/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7250912/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9225678/
- https://pubmed.ncbi.nlm.nih.gov/33813632/
- https://pubmed.ncbi.nlm.nih.gov/33539739/
- https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7540970/
- https://www.semanticscholar.org/paper/6f0c525ffe637eaa93ce7fb8c0e17500cc823e35
- https://pubmed.ncbi.nlm.nih.gov/35852844/