Next Article in Journal
DNA Double Strand Break and Response Fluorescent Assays: Choices and Interpretation
Next Article in Special Issue
Renin–Angiotensin Inhibitor, Captopril, Attenuates Growth of Patient-Derived Colorectal Liver Metastasis Organoids
Previous Article in Journal
Brain Plasticity in Patients with Spinal Cord Injuries: A Systematic Review
Previous Article in Special Issue
Bafilomycin A1 Molecular Effect on ATPase Activity of Subcellular Fraction of Human Colorectal Cancer and Rat Liver
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Novel Therapeutic Approaches for Colorectal Cancer Treatment

by
Athanasios G. Papavassiliou
1 and
Donatella Delle Cave
2,*
1
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’, CNR, 80131 Naples, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(4), 2228; https://doi.org/10.3390/ijms25042228
Submission received: 1 February 2024 / Accepted: 7 February 2024 / Published: 13 February 2024
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Colorectal Cancer 2.0)
According to GLOBOCAN 2020 data, colorectal cancer (CRC) represents the third most common malignancy and the second most deadly cancer worldwide [1,2,3]. In a clinical setting, despite advances in diagnosis and surgical procedures, 20% of patients with CRC present with metastasis at the time of diagnosis due to residual tumor cells that have spread to distant organs prior to surgery, affecting the patient’s survival rate [4]. Standard systemic chemotherapy, alternative therapies targeting mechanisms in cancer progression and metastasis, immunotherapy, and combination therapies are the primary strategies for treating CRC [5,6]. Unfortunately, these treatment strategies are expensive and often lack selectivity in targeting cancer cells, leading to severe toxicity in normal tissues and various side effects [7]. The main purpose of this Editorial is to provide a concise and state-of-the-art overview of novel therapeutic approaches for CRC treatment.
Mutations in different signaling pathways contribute to the initiation, progression, and chemoresistance of CRC, and among them, the overactivation of the phosphoinositide 3-kinase (PI3K)/AKT/mechanistic target of rapamycin (mTOR) signaling axis is of pivotal importance for tumorigenicity [8,9,10]. Although there are conflicting data regarding the effectiveness of agents directed against the PI3K axis in CRC, several studies have shown favorable results for these drugs, whether used in primary or metastatic cases. Among them, Pictilisib, a potent small-molecule class I PI3K inhibitor (PI3Ki), has shown promising results in reducing mucinous colorectal adenocarcinoma (MCA) progression; however, its effectiveness as single-agent therapy is limited due to the potential development of drug-induced resistance [11,12,13]. Kuracha et al. demonstrated that, in MCA cells, Pictilisib-induced adaptive resistance is regulated by the forkhead box O (FOXO)-dependent rebound activity of receptor tyrosine kinases (RTKs) [14]. The results revealed that pictilisib treatment led to an increased accumulation of nuclear FOXO1 compared to vehicle-treated CRC cells, and the authors proposed FOXO1 as a putative co-target to rescue PI3Ki single-agent resistance in MCA therapy. In CRC, as well as for the majority of tumors, cancer stem cells (CSCs) are recognized as a primary contributor to drug resistance, tumor progression, and metastasis [15,16,17,18]. Several signaling pathways are implicated in maintaining cancer stemness; consequently, targeting these pathways emerges as a feasible strategy for eliminating CSCs and potentially tumor eradication [19]. Recently, some studies have shown that CD44, a cell surface glycoprotein, and its isoforms generated from alternative splicing involving standard and variant exons (CD44v) play a crucial role in the progression of CRC [20,21]. Notably, overexpression of CD44v6 is associated with an unfavorable prognosis in CRC patients, influencing adhesion, proliferation, stemness, invasiveness, and chemoresistance [22]. Accordingly, CD44v6 emerges as a promising target for both cancer diagnosis and therapy in CRC. Ejima et al. established a novel anti-CD44mAb, C44Mab-5 (IgG1, kappa), and C44Mab-46 (IgG1, kappa), and they evaluated their applicability through enzyme-linked immunosorbent assay, flow cytometry, western immunoblotting, and immunohistochemical analyses on several CRC cells [23]. Another widely recognized CSCs marker is ATP-binding cassette superfamily G member 2 (ABCG2), a multidrug transporter that mediates the translocation of diverse physiological and xenobiotic substrates across cellular membranes in an ATP-dependent manner [24]. The expression of the ABCG2 gene has demonstrated negative prognostic implications in various malignancies, while in CRC, its prognostic significance remains undefined [25,26,27]. By analyzing publicly available datasets, Sałagacka-Kubiak et al. demonstrated that ABCG2 is downregulated in colon and rectum adenocarcinomas, exhibiting lower expression levels compared to both adjacent non-malignant tissues [28]. This deregulation is suggested to be associated with the methylation level of specific sites within the ABCG2 gene and correlated with microsatellite instability (MSI), weight, and age, whilst in rectum adenocarcinoma patients, it was linked to tumor localization, population type, and age. Furthermore, an ABCG2-centered protein—protein interaction network, constructed using STRING, revealed that the associated proteins are involved in leukotriene, organic anion, xenobiotic transport, endodermal cell-fate specification, as well as histone methylation and ubiquitination. Therefore, the downregulation of ABCG2 may serve as a marker of the activity of specific signaling pathways or protein interactors crucial for colorectal carcinogenesis. Another protein engaged in CRC progression is the muscarinic acetylcholine receptor M3 (M3R) [29,30,31]. Analyzing 754 surgical CRC tissue samples, Lobbes et al. demonstrated that high expression of M3R correlated with enhanced survivability, particularly in cases with lower tumor grade and a non-mucinous subtype. This association was linked to a more favorable outcome compared to cases with low M3R expression, where survival significantly decreased and higher tumor grade and mucinous subtype were prevalent [32]. Genomic instability is a hallmark of CRC, and metastatic CRC (mCRC) characterized by deficient mismatch repair (dMMR) and MSI can effectively be treated using immune checkpoint inhibitors (ICI) such as pembrolizumab and nivolumab, approved by both the FDA and EMA [12,33,34]. Alternatively, combinations of programmed cell death protein-1 (PD-1) inhibitors with ipilimumab, an antibody targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), have also demonstrated efficacy in this context [35,36,37]. Krekeler et al. described the case of a 63-year-old male with microsatellite instability (MSI-H) and mCRC associated with Lynch syndrome [38]. The patient experienced rapid normalization of tumor markers and achieved a complete metabolic remission (CMR) that has persisted for ten months. This notable outcome was observed through a sequential ICI treatment approach involving the combination of nivolumab and ipilimumab, followed by nivolumab maintenance therapy after progression on single-agent PD-1 ICI therapy. This represents the first reported instance of sustained metabolic complete remission in an MSI-H mCRC patient who initially showed progression on single-agent anti-PD-1 therapy, suggesting that individuals with dMMR mCRC may have benefited from sequential immune checkpoint regimens, even exhibiting long-term responses.
In conclusion, personalized therapeutic regimens represent the cutting edge in CRC treatment. Strategies focused on targeting patient-specific markers have the potential to augment standard chemotherapy efficacy and mitigate tumor progression.

Author Contributions

Conceptualization, D.D.C.; writing—original draft preparation, D.D.C. and A.G.P.; writing—review and editing, D.D.C. and A.G.P. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

D.D.C. was supported by Fondazione Umberto Veronesi (FUV) and Fondazione Italiana per la ricerca sulle Malattie del Pancreas (FIMP).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Xi, Y.; Xu, P. Global Colorectal Cancer Burden in 2020 and Projections to 2040. Transl. Oncol. 2021, 14, 101174. [Google Scholar] [CrossRef] [PubMed]
  2. Xie, Y.-H.; Chen, Y.-X.; Fang, J.-Y. Comprehensive Review of Targeted Therapy for Colorectal Cancer. Signal Transduct. Target. Ther. 2020, 5, 22. [Google Scholar] [CrossRef]
  3. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of Colorectal Cancer: Incidence, Mortality, Survival, and Risk Factors. Prz. Gastroenterol. 2019, 14, 89–103. [Google Scholar] [CrossRef] [PubMed]
  4. Vatandoust, S. Colorectal Cancer: Metastases to a Single Organ. WJG 2015, 21, 11767. [Google Scholar] [CrossRef]
  5. Gustavsson, B.; Carlsson, G.; Machover, D.; Petrelli, N.; Roth, A.; Schmoll, H.-J.; Tveit, K.-M.; Gibson, F. A Review of the Evolution of Systemic Chemotherapy in the Management of Colorectal Cancer. Clin. Color. Cancer 2015, 14, 1–10. [Google Scholar] [CrossRef]
  6. McQuade, R.M.; Stojanovska, V.; Bornstein, J.C.; Nurgali, K. Colorectal Cancer Chemotherapy: The Evolution of Treatment and New Approaches. CMC 2017, 24, 1537–1557. [Google Scholar] [CrossRef]
  7. Negarandeh, R.; Salehifar, E.; Saghafi, F.; Jalali, H.; Janbabaei, G.; Abdhaghighi, M.J.; Nosrati, A. Evaluation of Adverse Effects of Chemotherapy Regimens of 5-Fluoropyrimidines Derivatives and Their Association with DPYD Polymorphisms in Colorectal Cancer Patients. BMC Cancer 2020, 20, 560. [Google Scholar] [CrossRef] [PubMed]
  8. Hua, H.; Kong, Q.; Zhang, H.; Wang, J.; Luo, T.; Jiang, Y. Targeting mTOR for Cancer Therapy. J. Hematol. Oncol. 2019, 12, 71. [Google Scholar] [CrossRef]
  9. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in Cancer: Mechanisms and Advances in Clinical Trials. Mol. Cancer 2019, 18, 26. [Google Scholar] [CrossRef]
  10. Panwar, V.; Singh, A.; Bhatt, M.; Tonk, R.K.; Azizov, S.; Raza, A.S.; Sengupta, S.; Kumar, D.; Garg, M. Multifaceted Role of mTOR (Mammalian Target of Rapamycin) Signaling Pathway in Human Health and Disease. Signal Transduct. Target. Ther. 2023, 8, 375. [Google Scholar] [CrossRef]
  11. Yue, Q.; Khojasteh, S.C.; Cho, S.; Ma, S.; Mulder, T.; Chen, J.; Pang, J.; Ding, X.; Deese, A.; Pellet, J.D.; et al. Absorption, Metabolism and Excretion of Pictilisib, a Potent Pan-Class I Phosphatidylinositol-3-Kinase (PI3K) Inhibitor, in Rats, Dogs, and Humans. Xenobiotica 2021, 51, 796–810. [Google Scholar] [CrossRef]
  12. Banerji, U.; Stewart, A.; Coker, E.A.; Minchom, A.; Pölsterl, S.; Georgiou, A.; Al-Lazikani, B. Unravelling the Context Specificity of Signalling in KRAS Mutant Cancers: Implications for Design of Clinical Trials. Ann. Oncol. 2018, 29, iii7. [Google Scholar] [CrossRef]
  13. Vitiello, P.P.; Cardone, C.; Martini, G.; Ciardiello, D.; Belli, V.; Matrone, N.; Barra, G.; Napolitano, S.; Della Corte, C.; Turano, M.; et al. Receptor Tyrosine Kinase-Dependent PI3K Activation Is an Escape Mechanism to Vertical Suppression of the EGFR/RAS/MAPK Pathway in KRAS-Mutated Human Colorectal Cancer Cell Lines. J. Exp. Clin. Cancer Res. 2019, 38, 41. [Google Scholar] [CrossRef]
  14. Kuracha, M.R.; Govindarajan, V.; Loggie, B.W.; Tobi, M.; McVicker, B.L. Pictilisib-Induced Resistance Is Mediated through FOXO1-Dependent Activation of Receptor Tyrosine Kinases in Mucinous Colorectal Adenocarcinoma Cells. IJMS 2023, 24, 12331. [Google Scholar] [CrossRef]
  15. Cavo, M.; Delle Cave, D.; D’Amone, E.; Gigli, G.; Lonardo, E.; del Mercato, L.L. A Synergic Approach to Enhance Long-Term Culture and Manipulation of MiaPaCa-2 Pancreatic Cancer Spheroids. Sci. Rep. 2020, 10, 10192. [Google Scholar] [CrossRef]
  16. Cave, D.D.; Hernando-Momblona, X.; Sevillano, M.; Minchiotti, G.; Lonardo, E. Nodal-Induced L1CAM/CXCR4 Subpopulation Sustains Tumor Growth and Metastasis in Colorectal Cancer Derived Organoids. Theranostics 2021, 11, 5686–5699. [Google Scholar] [CrossRef]
  17. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ Drives Immune Evasion in Genetically Reconstituted Colon Cancer Metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed]
  18. Tauriello, D.V.F.; Calon, A.; Lonardo, E.; Batlle, E. Determinants of Metastatic Competency in Colorectal Cancer. Mol. Oncol. 2017, 11, 97–119. [Google Scholar] [CrossRef] [PubMed]
  19. Liu, Z.; Xu, H.; Weng, S.; Ren, Y.; Han, X. Stemness Refines the Classification of Colorectal Cancer with Stratified Prognosis, Multi-Omics Landscape, Potential Mechanisms, and Treatment Options. Front. Immunol. 2022, 13, 828330. [Google Scholar] [CrossRef]
  20. Wang, Z.; Tang, Y.; Xie, L.; Huang, A.; Xue, C.; Gu, Z.; Wang, K.; Zong, S. The Prognostic and Clinical Value of CD44 in Colorectal Cancer: A Meta-Analysis. Front. Oncol. 2019, 9, 309. [Google Scholar] [CrossRef] [PubMed]
  21. Todaro, M.; Gaggianesi, M.; Catalano, V.; Benfante, A.; Iovino, F.; Biffoni, M.; Apuzzo, T.; Sperduti, I.; Volpe, S.; Cocorullo, G.; et al. CD44v6 Is a Marker of Constitutive and Reprogrammed Cancer Stem Cells Driving Colon Cancer Metastasis. Cell Stem Cell 2014, 14, 342–356. [Google Scholar] [CrossRef]
  22. Ma, L.; Dong, L.; Chang, P. CD44v6 Engages in Colorectal Cancer Progression. Cell Death Dis. 2019, 10, 30. [Google Scholar] [CrossRef]
  23. Ejima, R.; Suzuki, H.; Tanaka, T.; Asano, T.; Kaneko, M.K.; Kato, Y. Development of a Novel Anti-CD44 Variant 6 Monoclonal Antibody C44Mab-9 for Multiple Applications against Colorectal Carcinomas. IJMS 2023, 24, 4007. [Google Scholar] [CrossRef]
  24. Robey, R.W.; Polgar, O.; Deeken, J.; To, K.W.; Bates, S.E. ABCG2: Determining Its Relevance in Clinical Drug Resistance. Cancer Metastasis Rev. 2007, 26, 39–57. [Google Scholar] [CrossRef]
  25. Wang, X.; Xia, B.; Liang, Y.; Peng, L.; Wang, Z.; Zhuo, J.; Wang, W.; Jiang, B. Membranous ABCG2 Expression in Colorectal Cancer Independently Correlates with Shortened Patient Survival. CBM 2013, 13, 81–88. [Google Scholar] [CrossRef]
  26. Cave, D.D.; Di Guida, M.; Costa, V.; Sevillano, M.; Ferrante, L.; Heeschen, C.; Corona, M.; Cucciardi, A.; Lonardo, E. TGF-Β1 Secreted by Pancreatic Stellate Cells Promotes Stemness and Tumourigenicity in Pancreatic Cancer Cells through L1CAM Downregulation. Oncogene 2020, 39, 4271–4285. [Google Scholar] [CrossRef]
  27. Cave, D.D.; Buonaiuto, S.; Sainz, B.; Fantuz, M.; Mangini, M.; Carrer, A.; Di Domenico, A.; Iavazzo, T.T.; Andolfi, G.; Cortina, C.; et al. LAMC2 Marks a Tumor-Initiating Cell Population with an Aggressive Signature in Pancreatic Cancer. J. Exp. Clin. Cancer Res. 2022, 41, 315. [Google Scholar] [CrossRef] [PubMed]
  28. Sałagacka-Kubiak, A.; Zawada, D.; Saed, L.; Kordek, R.; Jeleń, A.; Balcerczak, E. ABCG2 Gene and ABCG2 Protein Expression in Colorectal Cancer—In Silico and Wet Analysis. IJMS 2023, 24, 10539. [Google Scholar] [CrossRef] [PubMed]
  29. Kuol, N.; Godlewski, J.; Kmiec, Z.; Vogrin, S.; Fraser, S.; Apostolopoulos, V.; Nurgali, K. Cholinergic Signaling Influences the Expression of Immune Checkpoint Inhibitors, PD-L1 and PD-L2, and Tumor Hallmarks in Human Colorectal Cancer Tissues and Cell Lines. BMC Cancer 2023, 23, 971. [Google Scholar] [CrossRef] [PubMed]
  30. Carroll, R.C. The M3 Muscarinic Acetylcholine Receptor Differentially Regulates Calcium Influx and Release through Modulation of Monovalent Cation Channels. EMBO J. 1998, 17, 3036–3044. [Google Scholar] [CrossRef] [PubMed]
  31. Cheng, K.; Xie, G.; Khurana, S.; Heath, J.; Drachenberg, C.B.; Timmons, J.; Shah, N.; Raufman, J.-P. Divergent Effects of Muscarinic Receptor Subtype Gene Ablation on Murine Colon Tumorigenesis Reveals Association of M3R and Zinc Finger Protein 277 Expression in Colon Neoplasia. Mol. Cancer 2014, 13, 77. [Google Scholar] [CrossRef] [PubMed]
  32. Lobbes, L.A.; Schütze, M.A.; Droeser, R.; Arndt, M.; Pozios, I.; Lauscher, J.C.; Hering, N.A.; Weixler, B. Muscarinic Acetylcholine Receptor M3 Expression and Survival in Human Colorectal Carcinoma—An Unexpected Correlation to Guide Future Treatment? IJMS 2023, 24, 8198. [Google Scholar] [CrossRef] [PubMed]
  33. Pino, M.S.; Chung, D.C. The Chromosomal Instability Pathway in Colon Cancer. Gastroenterology 2010, 138, 2059–2072. [Google Scholar] [CrossRef] [PubMed]
  34. Ferguson, L.R.; Chen, H.; Collins, A.R.; Connell, M.; Damia, G.; Dasgupta, S.; Malhotra, M.; Meeker, A.K.; Amedei, A.; Amin, A.; et al. Genomic Instability in Human Cancer: Molecular Insights and Opportunities for Therapeutic Attack and Prevention through Diet and Nutrition. Semin. Cancer Biol. 2015, 35, S5–S24. [Google Scholar] [CrossRef]
  35. Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse Effects of Immune-Checkpoint Inhibitors: Epidemiology, Management and Surveillance. Nat. Rev. Clin. Oncol. 2019, 16, 563–580. [Google Scholar] [CrossRef]
  36. Wong, S.K.; Beckermann, K.E.; Johnson, D.B.; Das, S. Combining Anti-Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) and -Programmed Cell Death Protein 1 (PD-1) Agents for Cancer Immunotherapy. Expert. Opin. Biol. Ther. 2021, 21, 1623–1634. [Google Scholar] [CrossRef]
  37. Yin, Q.; Wu, L.; Han, L.; Zheng, X.; Tong, R.; Li, L.; Bai, L.; Bian, Y. Immune-Related Adverse Events of Immune Checkpoint Inhibitors: A Review. Front. Immunol. 2023, 14, 1167975. [Google Scholar] [CrossRef]
  38. Krekeler, C.; Wethmar, K.; Mikesch, J.-H.; Kerkhoff, A.; Menck, K.; Lenz, G.; Schildhaus, H.-U.; Wessolly, M.; Hoffmann, M.W.; Pascher, A.; et al. Complete Metabolic Response to Combined Immune Checkpoint Inhibition after Progression of Metastatic Colorectal Cancer on Pembrolizumab: A Case Report. IJMS 2023, 24, 12056. [Google Scholar] [CrossRef]
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

Papavassiliou, A.G.; Delle Cave, D. Novel Therapeutic Approaches for Colorectal Cancer Treatment. Int. J. Mol. Sci. 2024, 25, 2228. https://doi.org/10.3390/ijms25042228

AMA Style

Papavassiliou AG, Delle Cave D. Novel Therapeutic Approaches for Colorectal Cancer Treatment. International Journal of Molecular Sciences. 2024; 25(4):2228. https://doi.org/10.3390/ijms25042228

Chicago/Turabian Style

Papavassiliou, Athanasios G., and Donatella Delle Cave. 2024. "Novel Therapeutic Approaches for Colorectal Cancer Treatment" International Journal of Molecular Sciences 25, no. 4: 2228. https://doi.org/10.3390/ijms25042228

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

Article Metrics

Back to TopTop