Current Approaches, Challenges, and Future Perspectives in Colorectal Cancer Therapeutics and Integrated Care
Download
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed malignancies and a key reason for the increase in the number of cancer-related deaths experienced all over the world, making it a serious global health concern. The approach for CRC is multidimensional managing the factors such as the tumor burden, molecular markers, and factors that are specific to the patient. The primary treatment for early-stage CRC is still the surgical removal of the tumor, and the adjuvant chemotherapy is recommended on the basis of the pathological staging, lymph node involvement, and the features that are high-risk. When it comes to mCRC, the treatment’s initial objectives are to extend the patient life and to maintain the quality of life with the help of systemic treatments, localized interventions, and personalized strategies. In this review, we will present the cutting-edge technologies that have emerged and the contention that came up in CRC treatment, including the use of surgical methods, chemotherapy, radiotherapy, targeted therapy (e.g., anti-EGFR, anti-VEGF agents), immunotherapy (particularly for mismatch repair-deficient tumors), and the introduction of gene therapy and novel combinations of drugs which are still at the research stage are also discussed.
Introduction
Colorectal cancer (CRC) is a prominent health issue being the second most diagnosed and the second cause of death in cancer in the world. The latest projections announced 2 million fresh cases and 612,000 deaths in the United States in 2024 [1]. Of special interest is the fact that the mortality rates have been decreasing continuously since 1991, which has “prevented” over 4 million deaths, mainly due to the introduction of tobacco control policies, screening advancements, and therapeutic breakthroughs. Through the help of newer screening technologies and novel treatments, CRC patient management has improved tremendously. With the diagnosis being the primary guide, the treatment of modern CRC requires a widespread method that combines surgery, systemic therapies (chemotherapy, targeted agents, and immunotherapy), and radiotherapy, along with the addition of molecular characteristics and individualized patient factors [2, 3]. One of the main types of CRC not associated with metastasis is surgical resection, which is also the key type of curative treatment [4]. Following neoadjuvant chemotherapy and/or RT (in rectal cancer) can downstage tumors, improve resectability, and decrease local recurrence. Adjuvant therapy is recommended with reference to pathological findings, such as lymph node involvement, extra-mural vascular invasion, or high-risk features. In metastatic CRC (mCRC), systemic therapy-inclusive chemotherapy, biologics (anti-EGFR or anti-VEGF agents), and immunotherapy (for mismatch repair-deficient tumors)-is the premier approach, while surgery is admitted in select cases with oligometastatic disease or palliative intent [5, 6]. The latest breakthroughs in the targeted therapies (e.g., KRAS/BRAF inhibitors), immunotherapy (e.g., checkpoint inhibitors), and other new modalities like cancer vaccines, adoptive cell therapies (CAR-T), and gene editing have furnished the treatment options for both localized and advanced CRC [7, 8]. This publication recaps the prevailing standards, newly developing surgical solutions, and opposing views about cutting-edge surgical techniques in CRC, hence giving clinicians fact-supported recommendations for the best treatment of patients.
Surgery in Colorectal Cancer
Surgical Management of CRC has undergone major changes and is now treated with individually tailored strategies that are based on tumor location, stage, and molecular characteristics and incorporate oncologic treatment effectiveness with preservation of quality of life. In the case of localized disease, total surgical resection, which is still the main method for treating them, is completed by segmental colectomy and good lymphadenectomy (harvesting ≥12 nodes) as the standard for colon cancer and by total mesorectal excision (TME) in the case of rectal tumor to get the best possible oncological outcomes and avoid local recurrence [9-11]. The introduction of minimally invasive surgical methods (laparoscopic and robotic ones) due to the rearrangement of the area, in turn, brought similar oncological results apart from the advantages concerning postoperative morbidity and recovery but their strong application in the fight with advanced rectal cancers is still under discussion [12, 13]. Neoadjuvant therapy is one of the major players in the process, especially in the case of rectal cancer, in which chemoradiation is able to downstage tumors, promote resectability, and also increase the rate of sphincter preservation, the new found “watch-and- wait” strategy that provides organ preservation for a group of selected patients obtaining clinical complete response, the age long data of which though are not yet completely developed [6]. Management of lymph nodes still pertains to the classical lymphadenectomy concurrent with resection, sentinel node mapping is still in trial, and according to some studies, lateral pelvic lymph node dissection could be performed on certain rectal carcinoma patients [14]. In cases with metastatic settings, surgery still remains important in the case of oligometastatic disease where the combination of resection or ablation of metastases in the liver/lung, followed by perioperative chemotherapy treatment could provide patients with long-term results. Peritoneal metastases have been studied with controversial approaches like cytoreductive surgery with HIPEC although these ideas are still in the process of being officially declared [15]. The locoregional recurrences can be checked by radical re-resection which is possible in some cases that is required for R0 resection but decision-making will also be influenced by functional aspects such as sphincter preservation and nerve-sparing approaches that are now being more and more included [11, 16]. As a progressive step, the interconnection of liquid biopsies (especially ctDNA for recurrence risk stratification) and advanced technologies like augmented reality and AI-assisted surgery should further sharpen the precision of lymph node dissection and margin assessment, which may change the whole surgery [17]. With this area developing and expanding, the multidisciplinary team still remains the key to achieving a fine line between eliminating cancer to the max degree and function and quality of life of the patient, with the research still going on in the context of the rules and the types of surgery associated with the entire sector of CRC management which get better with each passing modification of the patient selection process.
Chemotherapy in Colorectal Cancer
CRC has a prominent area in the area of chemotherapy and chemotherapy stands in front in the multidisciplinary efforts for dealing with CRC where it finds applications in preoperative, postoperative, and palliative cases [18, 19]. In the case of locally advanced rectal cancer, use of neoadjuvant chemoradiation (usually 5-FU or capecitabine-based regimens along with radiotherapy) has turned out to be the standard situation and it has now been established as such, it is the standard procedure for significant tumor downstaging, improvement in sphincter preservation rates, and the reduction of the local recurrence compared to surgery alone. In the case of colon cancer, the adjuvant regimen with FOLFOX (5- FU, leucovorin plus oxaliplatin) or CAPOX (capecitabine plus oxaliplatin) will be recommended for a stage III or a high-risk stage II disease, as it will reduce the potential recurrence by about 30-40% [20, 21]. The initiation of adjuvant chemotherapy is the most important factor in time, where it begins within 6-8 weeks after surgery associated with the best possible outcome, but newly released data results of circulating tumor DNA (ctDNA) could help to determine the decision of patient selection [22-25]. Chemotherapy is the main form of treatment in the metastatic state, including the possibility of using FOLFOX, FOLFIRI (5-FU, leucovorin, and irinotecan), or the combinations with targeted agents (anti-EGFR or anti-VEGF therapies) as first-line options [26]. In the latest breakthroughs, molecular profiling takes centre stage in chemotherapy decision-making, where deficient mismatch repair (dMMR)/microsatellite instability-high (MSI-H) tumors, which rarely benefit the use of conventional 5-FU-based adjuvant therapy but heal remarkably well to immunotherapy [27, 28]. In cases with proficient MMR (pMMR) tumors, studies propose the combinación of irinoteça in adjuvant regimens and temporary strategies maintenance (like 5-FU/bevacizumab after the starting induction therapy) in the case of the evolving disease. The ideal length of adjuvant therapy keeps circling the table, with debates arising whether three months are as effective as six for some low-risk patients, particularly in preventing oxaliplatin-related neurotoxicity. New biomarkers such as the concept of conversion therapy have gained traction, where aggressive chemotherapy regimens (usually FOLFOXIRI - 5-FU, leucovorin, oxaliplatin, and irinotecan) together with biologics make it possible, in a limited number of patients, to make surgically unresectable liver metastases operable. The thymidylate synthase expression and dihydropyrimidine dehydrogenase activity levels may be used to further tailor chemotherapy programs and the dosing of the drugs, while innovations in drug forms and transit systems will always boost treatment quality and convenience.
Radiotherapy in Colorectal Cancer
Radiotherapy (RT) remains a paramount fixture in the interdisciplinary management of CRC (CRC), especially in rectal cancer cases where the neoadjuvant chemoradiation (nCRT) is currently the therapy of choice for the stage locally advanced disease (T3-4 or N+) [29, 30]. Multiple large randomized trials have identified consistently the effect of the preoperative RT in reducing effectively the local recurrence rates with respect to surgery alone, with two proven approaches being long- course chemoradiation (45-50 Gy over 5 weeks with concurrent 5-FU or capecitabine) and short-course RT (25 Gy in 5 fractions). Short-course RT, on the other hand, not only demonstrates practical advantages but also has similar oncological outcomes for most patients, while long-course chemoradiation still remains the favored treatment for cases, in which the downstaging of the tumor may assist in the sphincter preservation operation [31, 32]. The concept of total neoadjuvant therapy (TNT), which includes administering systemic chemotherapy prior to or after the nCRT, has been examined in recent studies and the results are quite promising showing improved rates of the pathologic complete response and distant control of the disease [6, 33]. Compared to the role of RT in colon cancer where it is not that extensive, it can be taken into consideration in particular cases, which possess high-risk features (T4 disease, positive margins, or resectable recurrence). Intraoperative radiotherapy (IORT) is the boost that can be used in cases where the close or positive margins are concerned particularly in the local recurrent rectal cancer. The advent of such new technology as intensity-modulated radiotherapy (IMRT) and image-guided radiotherapy (IGRT) has led to the advancement in precision and dose escalation to target volumes while toxicity to adjacent organs (e.g., small bowel, bladder, and sexual function structures) could be minimized. The topic of nodal irradiation in rectal cancer is still a frontier area of research. While typically the fields will include the mesorectum and internal iliac nodes, it is ambiguous whether the use of additional proximal nodal coverage (common iliac or para-aortic) would actually benefit high-risk patients. The best way to deal with pelvic lateral lymph nodes in rectal cancer is also changing with time, and some institutions favour the approach of unstabilizing the nodes with a selective nodal irradiation followed by a surgical excision in node-positive cases. Postoperative RT is in the main experimental with patients with positive margins or other high-risk features not treated preoperatively, although the benefit it produces has to be offset by the increased toxicity risks to the previously irradiated tissues [34, 35]. Novel strategies embody the concept of organ preservation for those who respond very well clinically after nCRT (“watch-and-wait”), however, a proper selection of patients and following exact guidelines are imperative [36, 37]. Progressions in the field of radiomics and functional imaging bring about the better assessment of response and planning of RT, notwithstanding the novelty of techniques such as proton therapy being applied to lessen toxicity of treatment even further. Ongoing clinical trials are still making progress in fine-tuning the combination of RT with systemic therapies as well as optimizing the application of the entire spectrum of CRC management [38-41].
Endocrine therapy in Colorectal Cancer
Endocrine therapy, of course, is an essential treatment in hormone receptor-positive breast cancer (42, 43], but at present, it has no defined role in CRC (CRC) management, even though many hormonal pathways have been studied as possible influencers in CRC pathogenesis and progression [44]. Observations from epidemiology imply the potential protective effects of Strogen in CRC, mainly the emphasis on the diminished number of cases that occur in postmenopausal women under the hormone replacement therapy, whereas preclinical studies showed different patches of Strogen receptors (ER-α and ER-β) in CRC tissues, of which ER-β was suggested to inhibit the growth of the tumor via anti-proliferative and pro-differentiation pathways, yet these results have not been reflected in the clinical application of anti-Strogen therapies [45]. The vitamin D endocrine system has also been investigated as an area of interest with extensive preclinical supporting data for the vitamin D receptor (VDR) signaling’s proposition of CRC progression modulation through cell differentiation, apoptosis, and Wnt/β-catenin pathway regulation leading to ongoing clinical trials of high-dose vitamin D supplementation as adjuvant therapy; moreover, the VDR expression may also be the prognostic biomarker for some CRC subtypes [46-49]. The insulin-like growth factor (IGF) pathway has also been carried into the conversation when it comes to CRC progression, where the IGF-1/IGF-1R axis was found to play a major role in tumor growth and metastasis, but the early clinical trials testing IGF-1R inhibitors on their own did not show any successful results leading to todayHw exploration of combination with cytotoxic or targeted agents, particularly in the context of metabolic comorbidities like diabetes and obesity that affect CRC risk and treatment outcomes through hormonal mechanisms [50, 51]. The recently emerging studies have distinguished the bile acid metabolism and the Farnesoid X receptor (FXR) signaling as the most likely therapeutic targets since they take part in the inflow and the intestine’s homeostasis, while gut hormone receptors like GLP-1 and GIP are being studied for their physiological role in CRC biology through metabolic influences on the tumor microenvironment [52-56]. Future directions in the field of research need to cover development of selective ER-β agonists, the full realization of vitamin D/VDR-targeted strategies, exploration of hormonal-metabolic combination therapies, and investigation of endocrine-immune system crosstalk in CRC, thereby, these progressive changes signal the slowly developing but growing acknowledgment that subclinical endocrine therapy in CRC care is the stepping stone for far-reaching implications.
Targeted therapy in colorectal cancer
The field of targeted therapy in CRC has begun a new era with the introduction of molecularly targeted therapies that have completely changed the way treatments for CRC (CRC) are offered, particularly through agents directed against the pathways of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) [57-60]. Anti-EGFR monoclonal antibodies cetuximab and panitumumab have become significant therapies for RAS/BRAF wild-type mCRC; their introduction, as a result, has shown a significant increase in the overall survival (OS) and progression-free survival (PFS) when they are combined with chemotherapy in first-line settings [61]. The CRYSTAL trial set the point for treating RAS wild-type mCRC with FOLFIRI plus cetuximab as a standard regimen, achieving it with a median OS increase from 20.0 to 23.5 months, while the PRIME trial demonstrated similar benefits for FOLFOX plus panitumumab. The mentioned agents have decreased the number of deaths from metastatic left-sided tumors by more than 60% of patients in the initial treatment phase. The inhibition of VEGF, the first anti-angiogenic therapy with a survival benefit in mCRC, was the result of the combination of bevacizumab with chemotherapy (FOLFOX or FOLFIRI). The AVF2107 trial announced the incorporation of this strategy along with the 20.3 months median OS of bevacizumab plus chemotherapy, compared to 15.6 months median OS achieved by IFL chemotherapy alone. The attempt to treat patients with mCRC through a combination of different drugs has got initial success with this. Subsequent studies confirmed the effectiveness of bevancizumab in various treatment lines, including the option to continue after disease progression. The increment of resistance mechanisms with respect to EGFR inhibitors has prompted the study of RAS (KRAS/NRAS) mutations as adverse predictive biomarkers. Thus, the fundamental switch in treatment algorithms allows anti-EGFR therapy only for RAS wild- type tumors. In the case of BRAF V600E-mutant mCRC (8-10% of disease cases), encorafenib in conjunction with cetuximab was the scheme approved through the BEACON trial, whose result was the median OS of 9.3 months as opposed to 5.9 months in the case of standard chemotherapy. One of the recent milestones achieved is the dual EGFR inhibition technique; according to the PARADIGM trial where the phase III study was conducted, panitumumab plus mFOLFOX6 improved OS significantly in the left-sided RAS wild-type mCRC as compared to the use of bevacizumab (p=0.007, 37.9 vs 34.3 months). The discovery of new agents with HE2 amplifications (2-3% of CRC), still at the experimental stage, has been praiseworthy, as per the MyPathway trial that demonstrated 52% response rates for pertuzumab/ trastuzumab in HER2-amplified mCRC, leading to the FDA approval of trastuzumab deruxtecan for HER2- positive mCRC based on the DESTINY-CRC01 trial results. The newly discovered objectives also include NTRK fusions (entrectinib/larotrectinib), RET fusions (selpercatinib), and KRAS G12C mutations (sotorasib/ adagrasib); however, their prevalence in CRC is negligible. Even though these improvements have been made, acquired resistance is still a major obstacle, where the focus of ongoing research is on the combination treatment of (EGFR/MET or EGFR/BRAF inhibition) antibody-drug conjugates, and newer mechanisms like Claudin 18.2 targeting. Studies that are taking place also look into the possible sequencing of targeted the therapies apart from RAS/RAF status, which would further personalize CRC treatment with the implementation of rechallenge strategies and the development of sophisticated biomarkers. These strategies, on the other hand, will bring radical changes in the approach to therapy, leading to the higher number of remissions among CRC patients with specific molecular characteristics [61] (Table 1).
Author (Year) | Patients | Intervention | Key Outcomes |
Van Cutsem et al. (2009) CRYSTAL Trial | 1,198 Mcrc (KRAS wild-type) | FOLFIRI + cetuximab vs FOLFIRI | Median OS: 23.5 vs 20.0 mo (HR 0.80; p=0.009) |
| Douillard et al. (2010) PRIME Trial | 1,183 mCRC (KRAS wild-type) | FOLFOX + panitumumab vs FOLFOX | Median PFS: 9.6 vs 8.0 mo (HR 0.80; p=0.02) |
Kopetz et al. (2019) BEACON Trial | 665 BRAF V600E-mutant mCRC | Encorafenib + cetuximab vs control | Median OS: 9.3 vs 5.9 mo (HR 0.60; p<0.001) |
Yoshino et al. (2022) PARADIGM Trial | 823 RAS wild-type mCRC | Panitumumab + mFOLFOX6 vs bevacizumab + mFOLFOX6 | Left-sided: Median OS 37.9 vs 34.3 mo (HR 0.82) |
Siena et al. (2021) DESTINY-CRC01 | 86 HER2+ mCRC | Trastuzumab deruxtecan | ORR: 45.3% in HER2 IHC3+ cohort |
André et al. (2020) KEYNOTE-177 | 307 MSI-H/dMMR mCRC | Pembrolizumab vs chemotherapy | Median PFS: 16.5 vs 8.2 mo (HR 0.60; p=0.0002) |
Fakih et al. (2022) CodeBreaK 101 | 40 KRAS G12C-mutant mCRC | Sotorasib + panitumumab | ORR: 30%, median PFS: 5.7 mo |
Gene therapy in colorectal cancer
Gene therapy has emerged as a new focus area in the treatment of CRC through the use of genetic material conceptually delivered via either viral or non-viral vectors to modulate gene expression, restore, tumor suppressor, functionality, or by introducing these two mechanisms at the same time lead to apoptosis of only the designated cancer cells [62, 63]. They currently remain entirely experimental, and the majority of approaches researched are in the preclinical and early-phase clinical trial periods. CRISPR-Cas9 and other gene-editing technologies are being pursued to create gene abnormalities that CRC (CRC) cells cannot cope with (e.g., KRAS, APC, or TP53 mutations) or to increase T cells’ immune response to these changes by lymphocyte editing for adoptive cell therapies, Suicide gene therapy, such as ganciclovir which is given with herpes simplex virus thymidine kinase (HSV-TK) is showing tumor-specific cytotoxicity in CRC models. Early trials were done to see if it could be safely administered via adenoviral vectors. The second strategy is about the deployment of the oncolytic viruses types, which are especially designed to reproduce CRC cells that are already under the attack of immune system like modified adenoviruses (e.g., ONCOS-102) and vaccinia viruses (e.g. Pexa-Vec), both of them are in the test phase, being performed with the immune checkpoint inhibitors [64, 65]. MicroRNA interference (RNAi) and microRNA therapies deal primarily with oncogenes being silent or the inclining tissue miRNAs (e.g., miR-143/145, which target KRAS and ERK5). Liabilities of systemic delivery notwithstanding, nanoparticle-encapsulated siRNAs and miRNA mimics are finding their applications in preclinical CRC models [66]. DNA vaccines concentrated on CRC-related antigens (e.g., CEACAM5, GUCY2C, or neoantigens) are also being developed with pilot trials that investigate their capacity of strengthening antitumor immunity. Furthermore, CAR-T cell therapy that is being predominantly used for blood cancers, is also in preparation for colon cancer adaptation based on new targets like GUCY2C or TAG-72. Nonetheless, the protection from tumor microenvironment immunosuppression remains the major obstacle to such development [67-69]. Besides the aforementioned breakthroughs, clinical translation runs into problems like the safety of delivery, the possibility of unintended consequences, and immune vector eviction. Yet the forward movement may very well be embodied by the latest experiments those that involve KRAS-targeted CRISPR editing (NCT05381454) or CEACAM5-directed CAR-T cells (NCT05341492) for example which are testimonials of the state of the field, fast progress. It is plausible that the field of CRC diathesis through genetic therapy will be fragmented and include various other combinations like with immunotherapy or chemotherapy, as the accuracy of gene-editing is upgraded. Organic endeavors will be spotlighted not just on the establishment of better transmission systems, recognition of superior molecular targets, inclusion of gene therapy in the “multimodal treatment regimens” as well [70-73] (Table 2).
Identifier | Patient Population | Phase | Intervention | Primary Endpoints | Target/Mechanism |
NCT05381454 | KRAS-mutant mCRC | I/II | CRISPR-edited KRAS T cells | Safety, MTD | KRAS G12D/V-specific CAR-T |
NCT05341492 | CEACAM5+ mCRC | I | CEACAM5-directed CAR-T | Dose-limiting toxicity (DLT), ORR | CEACAM5 antigen |
NCT05241331 | MSS/pMMR mCRC | II | GUCY2C-targeted CAR-T + PD-1 inhibitor | PFS, immune response | GUCY2C tumor antigen |
NCT05076682 | MSI-H/dMMR mCRC | I/II | Personalized neoantigen DNA vaccine + pembrolizumab | Safety, immunogenicity | Tumor-specific neoantigens |
NCT05141721 | Refractory mCRC | I | Oncolytic vaccinia virus (Pexa-Vec) + nivolumab | Safety, viral shedding | Oncolytic immunotherapy |
NCT03563326 | Metastatic CRC | I/II | HER2-targeted CAR-NK cells | MTD, ORR | HER2 amplification |
NCT05433181 | Advanced GI cancers (CRC) | I | Claudin 18.2 CAR-T (CT041) | Safety, DLTs | Claudin 18.2 overexpression |
NCT04853017 | MSS mCRC | II | | Tumor RNA-loaded dendritic cell vaccine + regorafenib | Immune response, PFS | Autologous DC vaccination |
NCT04503278 | TP53-mutant CRC | I | TP53 mRNA-loaded lipid nanoparticles | Safety, TP53 restoration | TP53 tumor suppressor |
NCT05137275 | Locally advanced rectal cancer | I | CRISPR-Cas9 PD-1 knockout TILs | Feasibility, pCR rate | PD-1 disruption in TILs |
In conclusion, as the treatment landscape for CRC has experienced radical changes [31], it is now common to use combined multimodal techniques, which include surgery, chemotherapy, radiotherapy, and targeted therapies. In the case of localized CRC, curative surgery is the mainstay of treatment, which is transformed into a neoadjuvant and adjuvant precision method designed according to tumor biology and anatomical factors. The total neoadjuvant therapy (TNT) has revolutionized treatment pathways in rectal cancer by elevating the rates of pathologic complete response and giving the opportunity to organ preservation to rightly chosen patients. For mCRC, the growth of median survival in the case of the population with the wild-type RAS gene beyond 30 months is a result of the combining of cytotoxic regimens (FOLFOX/FOLFIRI) with biological agents (anti-EGFR, anti-VEGF). Nevertheless, with these achievements, the problem remains unresolved since the effectiveness of traditional treatments has ceased to increase, which indicates the strong necessity for new therapeutic measures.
Future Frontiers in CRC Therapeutics
Frontiers of the CRC in Therapy is indeed innovative. CRC crisscrossing the intravenous management will be mainly on finding ways to break the intrinsic and acquired resistance softening, fine-tuning the sequencing and promotion of the biomarker-driven therapies. The main focuses of the research include Immunotherapy revolutions for microsatellite-stable (MSS) tumors, using new checkpoints combination (e.g., PD-1/LAG- 3 inhibition) and microenvironment modulation (e.g., TGF-β blockade), Dual-targeted approaches (e.g., EGFR/ MET co-inhibition, BRAF/MEK suppression) to disrupt parallel signaling pathways and circumvent resistance. As these innovations step off the premises of clinical trials and enter into the real-life scene, CRC management will embrace the model of precision oncology more and more, which will be the result of all this: molecular profiling, real-time monitoring, and adaptive therapeutic strategies will be combined as one. In addition to the extra survival benefits, the concentration of the emphasis will also switch to enhancing the patient-centric aspects to the highest level including the improvement of the life quality and functional preservation. The next great problem in CRC is necessitating a double assault: on the one hand, it is to demolish the tumor’s molecular fortifications; on the other hand, it is to demolish the immunosuppressive stronghold of the metastasis niche. The advent of this reformatory stage will be powered through groundbreaking collaboration; bench scientists, doctors, and patients will work together to design clinical trials that will blur the boundary between research and real-world practice. In the upcoming days, we are at the crossroads; CRISPR-engineered microbes could outsmart chemoresistance, while the community-based spatial transcriptomics may decrypt the niche-driven relapse path within the tumor, and the AI-curated combined treatment will make stage IV cancer a chronic condition. The breakthrough, of course, brings up the question of equity: democratizing the dendritic access to these tools on the part of people through decentralized trials and global molecular tumor boards; hence, a more disciplined route to progress is clear:
i. Precision Overhaul: Liquid biopsy-guided adaptive therapies that evolve with the tumor’s genomic drift
ii. Niche Domination: Stroma-targeting nano-therapies and engineered immune cells to break metastatic sanctuaries
iii. Systems Warfare: Fusing deep learning with multi- omics to predict and PREEMPT treatment escape routes.
This isn’t not just minor changes but a fundamental re-conceptualization of the whole architecture of cancer care. The tools exist. The science is ripe. Now we must build the ecosystem to deliver cures, not just care
Acknowledgments
Statement of Transparency and Principals:
• Author declares no conflict of interest
• Study was approved by Research Ethic Committee of author affiliated Institute.
• Study’s data is available upon a reasonable request.
• All authors have contributed to implementation of this research.
References
- Erratum to "Cancer statistics, 2024" Siegel RLGANJA . CA: a cancer journal for clinicians.2024;74(2). CrossRef
- The tumor microenvironment and dendritic cells: Developers of pioneering strategies in colorectal cancer immunotherapy? Ghorbaninezhad F, Nour MA , Farzam OR , Saeedi H, Vanan AG , Bakhshivand M, Jafarlou M, Hatami-Sadr A, Baradaran B. Biochimica Et Biophysica Acta. Reviews on Cancer.2025;1880(2). CrossRef
- Immunotherapy in colorectal cancer: Statuses and strategies Li Y, Cheng Z, Li S, Zhang J. Heliyon.2025;11(1). CrossRef
- Colorectal cancer recurrence and its impact on survival after curative surgery: An analysis based on multistate models Balboa-Barreiro V, Pértega-Díaz S, García-Rodríguez T, González-Martín C, Pardeiro-Pértega R, Yáñez-González-Dopeso L, Seoane-Pillado T. Digestive and Liver Disease: Official Journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver.2024;56(7). CrossRef
- Adjuvant Therapy for Stage II Colon Cancer: ASCO Guideline Update Baxter NN , Kennedy EB , Bergsland E, Berlin J, George TJ , Gill S, Gold PJ , et al . Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology.2022;40(8). CrossRef
- Modern rectal cancer management: A review of total neoadjuvant therapy and current practices Karahan SN , Gorgun E. American Journal of Surgery.2025;241. CrossRef
- Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR , Duttaroy AK , Pathak S. International Journal of Colorectal Disease.2024;40(1). CrossRef
- Switching It up with New KRAS Inhibitors Waterson AG . Journal of Medicinal Chemistry.2025;68(9). CrossRef
- Surgical treatment of rectal cancer: local resection Perretta S, Guerrero V, Garcia-Aguilar J. Surgical Oncology Clinics of North America.2006;15(1). CrossRef
- Surgery for locally recurrent rectal cancer in the era of total mesorectal excision: is there still a chance for cure? Rahbari NN , Ulrich AB , Bruckner T, Münter M, Nickles A, Contin P, Löffler T, et al . Annals of Surgery.2011;253(3). CrossRef
- Curative-intent surgery for isolated locoregional recurrence of colon cancer: Review of the literature and institutional experience Wisselink D. D., Klaver C. E. L., Hompes R., Bemelman W. A., Tanis P. J.. European Journal of Surgical Oncology: The Journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology.2020;46(9). CrossRef
- Advancements in Robotic Surgery: A Comprehensive Overview of Current Utilizations and Upcoming Frontiers Reddy K, Gharde P, Tayade H, Patil M, Reddy LS , Surya D. Cureus.2023;15(12). CrossRef
- A New Era of Minimally Invasive Surgery: Progress and Development of Major Technical Innovations in General Surgery Over the Last Decade Siddaiah-Subramanya M, Tiang KW , Nyandowe M. Surgery Journal (New York, N.Y.).2017;3(4). CrossRef
- Sentinel lymph node mapping in the modern management of gynaecological malignancy Collins A, Phillips A. The Obstetrician & Gynaecologist.2023;25(3):210-219.
- Role of Surgery in Oligometastatic Disease. In: Metastatic Bone Disease Calvert GT . Cham: Springer International Publishing.2024;:279-295.
- Long-term outcomes of resection for locoregional recurrence of colon cancer: A retrospective descriptive cohort study Chesney TR , Metz JJ , Nadler A, Quereshy FA , Ashamalla S, Acuna SA , Swallow CJ . European Journal of Surgical Oncology: The Journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology.2021;47(9). CrossRef
- Liquid biopsy in cancer management: Integrating diagnostics and clinical applications Pandey S, Yadav P. Practical Laboratory Medicine.2025;43. CrossRef
- Principles of cancer treatment by chemotherapy Dickens E, Ahmed S. Surgery (Oxford).2021;39(4):215-20.
- The Multidisciplinary Management of Colorectal Cancer: Present and Future Paradigms Sievers CK , Kratz JD , Zurbriggen LD , LoConte NK , Lubner SJ , Uboha N, Mulkerin D, Matkowskyj KA , Deming DA . Clinics in Colon and Rectal Surgery.2016;29(3). CrossRef
- Capecitabine Versus Continuous Infusion Fluorouracil for the Treatment of Advanced or Metastatic Colorectal Cancer: a Meta-analysis Wu Z, Deng Y. Current Treatment Options in Oncology.2018;19(12). CrossRef
- Capecitabine in the management of colorectal cancer Hirsch BR , Zafar SY . Cancer Management and Research.2011;3. CrossRef
- Timing of Adjuvant Chemotherapy and Survival in Colorectal, Gastric, and Pancreatic Cancer. A Systematic Review and Meta-Analysis Petrelli F, Zaniboni A, Ghidini A, Ghidini M, Turati L, Pizzo C, Ratti M, Libertini M, Tomasello G. Cancers.2019;11(4). CrossRef
- The optimal time of starting adjuvant chemotherapy after curative surgery in patients with colorectal cancer Yang Y, Lu Y, Tan H, Bai M, Wang X, Ge S, Ning T, et al . BMC cancer.2023;23(1). CrossRef
- Targeting Nanog expression increased Cisplatin chemosensitivity and inhibited cell migration in Gastric cancer cells Vasefifar P, Najafi S, Motafakkerazad R, Amini M, Safaei S, Najafzadeh B, Alemohammad H, Jafarlou M, Baradaran B. Experimental Cell Research.2023;429(2). CrossRef
- Nicotinic Acetylcholine Receptor Subunit Alpha-7 Mediates PD-L1 and CTLA-4 Expression in HepG2 Cells Nour MA , Kheradmand F, Rasmi Y, Asadzadeh Z, Doustvandi MA , Jafarlou M, Hajiasgharzadeh K, Baradaran B. ImmunoAnalysis.2021;1(1). CrossRef
- Systemic Therapy for Metastatic Colorectal Cancer: From Current Standards to Future Molecular Targeted Approaches Atreya CE , Yaeger R, Chu E. American Society of Clinical Oncology Educational Book. American Society of Clinical Oncology. Annual Meeting.2017;37. CrossRef
- Evolution of Precision Oncology, Personalized Medicine, and Molecular Tumor Boards Fujiwara Y, Kato S, Kurzrock R. Surgical Oncology Clinics of North America.2024;33(2). CrossRef
- Molecular profiling for precision cancer therapies Malone ER , Oliva M, Sabatini PJB , Stockley TL , Siu LL . Genome Medicine.2020;12(1). CrossRef
- The multidisciplinary management of locally advanced rectal cancer De Felice F, Miccini M, Botticelli A, Roberto M, Petrucciani N. Expert Review of Anticancer Therapy.2024;24(7). CrossRef
- Radiation therapy for rectal cancer Tseng M, Soon YY , Vellayappan B, Ho F, Tey J. Journal of Gastrointestinal Oncology.2019;10(6). CrossRef
- The evolution of rectal cancer treatment: the journey to total neoadjuvant therapy and organ preservation Iv AA , Koprowski MA , Nabavizadeh N, Tsikitis VL . Annals of Gastroenterology.2022;35(3). CrossRef
- Preoperative radiotherapy or chemoradiotherapy in rectal cancer - Is survival improved? An update of the "Nordic" LARC study in non-resectable cancers Brændengen M, Glimelius B. Radiotherapy and Oncology: Journal of the European Society for Therapeutic Radiology and Oncology.2018;127(3). CrossRef
- Total Neoadjuvant Therapy for Locally Advanced Rectal Cancer: Induction or Consolidation Chemotherapy? Aref A, Abdalla A. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology.2022;40(23). CrossRef
- Management of lateral pelvic lymph nodes in rectal cancer Chang G, Halabi WJ , Ali F. Journal of Surgical Oncology.2023;127(8). CrossRef
- Rectal cancer lexicon: consensus statement from the society of abdominal radiology rectal & anal cancer disease-focused panel Hope TA , Gollub MJ , Arya S, Bates DDB , Ganeshan D, Harisinghani M, Jhaveri KS , et al . Abdominal Radiology (New York).2019;44(11). CrossRef
- Organ Preservation: Current Concepts and New Strategies for the Next Decade Guibert EE , Petrenko AY , Balaban CL , Somov AY , Rodriguez JV , Fuller BJ . Transfusion Medicine and Hemotherapy: Offizielles Organ Der Deutschen Gesellschaft Fur Transfusionsmedizin Und Immunhamatologie.2011;38(2). CrossRef
- Organ Preservation Strategies After Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancer Yoo RN , Kim HJ . Annals of Coloproctology.2019;35(2). CrossRef
- Imaging for Response Assessment in Radiation Oncology: Current and Emerging Techniques Stieb S, Kiser K, Dijk L, Livingstone NR , Elhalawani H, Elgohari B, McDonald B, et al . Hematology/Oncology Clinics of North America.2020;34(1). CrossRef
- Functional imaging for radiotherapy treatment planning: current status and future directions-a review Thorwarth D.. The British Journal of Radiology.2015;88(1051). CrossRef
- Application of Scaffold-Based Drug Delivery in Oral Cancer Treatment: A Novel Approach Saberian E, Jenča A, Petrášová A, Zare-Zardini H, Ebrahimifar M. Pharmaceutics.2024;16(6). CrossRef
- Success in Tooth Bud Regeneration: A Short Communication Sadrabad MJ , Saberian E, Izadi A, Emami R, Ghadyani F. Journal of Endodontics.2024;50(3). CrossRef
- An emerging generation of endocrine therapies in breast cancer: a clinical perspective Patel R, Klein P, Tiersten A, Sparano JA . NPJ breast cancer.2023;9(1). CrossRef
- Preparation, Characterization and Cytotoxic Studies of Cisplatin-containing Nanoliposomes on Breast Cancer Cell Lines Mohammadinezhad F, Talebi A, Allahyartorkaman M, Nahavandi R, Vesal M, Khiyavi AA , Velisdeh ZJ , et al . Asian Pacific Journal of Cancer Biology.2023;8(2). CrossRef
- A Prospective Evaluation of Endogenous Sex Hormone Levels and Colorectal Cancer Risk in Postmenopausal Women Murphy N, Strickler HD , Stanczyk FZ , Xue X, Wassertheil-Smoller S, Rohan TE , Ho GY , et al . Journal of the National Cancer Institute.2015;107(10). CrossRef
- Expression of oestrogen receptor β and prognosis of colorectal cancer Rudolph A., Toth C., Hoffmeister M., Roth W., Herpel E., Jansen L., Marx A., Brenner H., Chang-Claude J.. British Journal of Cancer.2012;107(5). CrossRef
- Vitamin D Receptor Genotype, Vitamin D3 Supplementation, and Risk of Colorectal Adenomas: A Randomized Clinical Trial Barry EL , Peacock JL , Rees JR , Bostick RM , Robertson DJ , Bresalier RS , Baron JA . JAMA oncology.2017;3(5). CrossRef
- Vitamin D and Colorectal Cancer: Current Perspectives and Future Directions Na S, Kim KB , Lim YJ , Song HJ . Journal of Cancer Prevention.2022;27(3). CrossRef
- High Cholesterol Diet Increases Expression of Cholesterol 24-Hydroxylase and BACE1 in Rat Hippocampi: Implications for the Effect of Diet Cholesterol on Memory Nikasa M, Karimi P, Rajavand H, Afshari F, Jafarlou M, Soltanali M. Iranian Red Crescent Medical Journal (IRCMJ).2016;18(12). CrossRef
- Regulation of Dendritic Cell Functions by Vitamins as Promising Therapeutic Strategy for Immune System Disorders Ghahramanipour Z, Alipour S, Masoumi J, Rostamlou A, Hatami-Sadr A, Heris JA , Naseri B, Jafarlou M, Baradaran B. Advanced Biology.2023;7(12). CrossRef
- IGF-1R, IGF-1 and IGF-2 expression as potential prognostic and predictive markers in colorectal-cancer Peters G, Gongoll S, Langner C, Mengel M, Piso P, Klempnauer J, Rüschoff J, Kreipe H, Wasielewski R. Virchows Archiv: An International Journal of Pathology.2003;443(2). CrossRef
- The Insulin/IGF System in Colorectal Cancer Development and Resistance to Therapy Vigneri PG , Tirrò E, Pennisi MS , Massimino M, Stella S, Romano C, Manzella L. Frontiers in Oncology.2015;5. CrossRef
- GLP-1R signaling modulates colonic energy metabolism, goblet cell number and survival in the absence of gut microbiota Greiner TU , Koh A, Peris E, Bergentall M, Johansson MEV , Hansson GC , Drucker DJ , Bäckhed F. Molecular Metabolism.2024;83. CrossRef
- GLP-1, GIP/GLP-1, and GCGR/GLP-1 receptor agonists: Novel therapeutic agents for metabolic dysfunction-associated steatohepatitis Singh A, Sohal A, Batta A. World Journal of Gastroenterology.2024;30(48). CrossRef
- FXR agonists for colorectal and liver cancers, as a stand-alone or in combination therapy Yu D, Lu Z, Wang R, Xiang Y, Li H, Lu J, Zhang L, et al . Biochemical Pharmacology.2023;212. CrossRef
- The Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide Attenuates Colon Cancer Development by Regulating Glucose Metabolism Zhang Y, Xie Y, Xia S, Ge X, Li J, Liu F, Jia F, et al . Advanced Science (Weinheim, Baden-Wurttemberg, Germany).2025;12(19). CrossRef
- The regulatory role of autophagy-related miRNAs in lung cancer drug resistance Shahverdi M, Hajiasgharzadeh K, Sorkhabi AD , Jafarlou M, Shojaee M, Jalili Tabrizi N, Alizadeh N, et al . Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie.2022;148. CrossRef
- Vascular endothelial growth factor and epidermal growth factor signaling pathways as therapeutic targets for colorectal cancer Winder T, Lenz H. Gastroenterology.2010;138(6). CrossRef
- Comprehensive review of targeted therapy for colorectal cancer Xie Y, Chen Y, Fang J. Signal Transduction and Targeted Therapy.2020;5(1). CrossRef
- CTLA-4 silencing in dendritic cells loaded with colorectal cancer cell lysate improves autologous T cell responses in vitro Ghorbaninezhad F, Masoumi J, Bakhshivand M, Baghbanzadeh A, Mokhtarzadeh A, Kazemi T, Aghebati-Maleki L, et al . Frontiers in Immunology.2022;13. CrossRef
- The Effects of siRNA-Mediated Gene Silencing of Alpha-7 Nicotinic Acetylcholine Receptors on Drug Resistance to Oxaliplatin in Colorectal Cancer Cells Hajiasgharzadeh K, Doustvandi MA , Khiabani NA , Mohammadi M, Dastmalchi N, Jafarlou M, et al . Biology Bulletin.2022;49.
- Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology Benson AB , Venook AP , Adam M, Chang G, Chen Y, Ciombor KK , Cohen SA , et al . Journal of the National Comprehensive Cancer Network: JNCCN.2024;22(2 D). CrossRef
- An overview of the history, applications, advantages, disadvantages and prospects of gene therapy Jafarlou M., Baradaran B., Saedi T. A., Jafarlou V., Shanehbandi D., Maralani M., Othman F.. Journal of Biological Regulators and Homeostatic Agents.2016;30(2).
- Advancements of gene therapy in cancer treatment: A comprehensive review Mohd Abas MD , Mohd Asri MF , Yusafawi NAS , Rosman NAZ , Baharudin NAZ , Taher M, Susanti D, Khotib J. Pathology, Research and Practice.2024;261. CrossRef
- Efficacy of HSV-TK/GCV system suicide gene therapy using SHED expressing modified HSV-TK against lung cancer brain metastases Oishi T, Ito M, Koizumi S, Horikawa M, Yamamoto T, Yamagishi S, Yamasaki T, et al . Molecular Therapy. Methods & Clinical Development.2022;26. CrossRef
- Precision oncolytic viral therapy in colorectal cancer: Genetic targeting and immune modulation for personalized treatment (Review) Sultan MH , Zhan Q, Wang Y, Xia Y, Jia X. International Journal of Molecular Medicine.2025;56(1). CrossRef
- Silencing of myeloid cell leukemia-1 by small interfering RNA improves chemosensitivity to etoposide in u-937 leukemic cells Jafarlou M., Baradaran B., Shanehbandi D., Saedi T. A., Jafarlou V., Karimi P., Othman F.. Journal of Biological Regulators and Homeostatic Agents.2016;30(1). CrossRef
- The Combination of PD-L1 and CTLA-4 Suppression Significantly Decreased the Expression Levels of Cancer Stem Cell Factors in the Pancreatic Cancer Cell Line Alizadeh N, Kazemi T, Hemmat N, Jafarlou M, Baradaran B. ImmunoAnalysis.2023 Jun 26;3:6.
- Dual silencing of tumor-intrinsic VISTA and CTLA-4 stimulates T-cell mediated immune responses and inhibits MCF7 breast cancer development Hosseinkhani N, Hemmat N, Baghbani E, Baghbanzadeh A, Kazemi T, Mokhtarzadeh A, Jafarlou M, Amin Doustvandi M, Baradaran B. Gene.2024;896. CrossRef
- MicroRNA-Based Therapeutics for Drug-Resistant Colorectal Cancer Jung E, Choi J, Kim J, Han T. Pharmaceuticals (Basel, Switzerland).2021;14(2). CrossRef
- CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future Uddin F, Rudin CM , Sen T. Frontiers in Oncology.2020;10. CrossRef
- Clinical translation of nanomedicines: Challenges, opportunities, and keys Younis MA , Tawfeek HM , Abdellatif AAH , Abdel-Aleem JA , Harashima H. Advanced Drug Delivery Reviews.2022;181. CrossRef
- Applications of artificial intelligence in regenerative dentistry: promoting stem cell therapy and the scaffold development Saberian E, Jenča A, Jenča A, Zare-Zardini H, Araghi M, Petrášová A, Jenčová J. Frontiers in Cell and Developmental Biology.2024;12. CrossRef
- Regulatory Effects of Long Non-coding RNAs on Th17/Treg Differentiation and Imbalance [Internet] Dabbaghipour R, Ahmadi E, Entezam M, Farzam OR , Baghbanzadeh A, Sichani AS , et al . Available from: http://ijaai.tums.ac.ir..
License

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
Copyright
© Asian Pacific Journal of Cancer Care , 2025
Author Details
How to Cite
- Abstract viewed - 0 times
- PDF (FULL TEXT) downloaded - 0 times
- XML downloaded - 0 times