Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1612 word(s) 1612 2021-05-11 08:13:44 |
2 format correct Meta information modification 1612 2021-05-11 09:43:49 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Galateanu, B. Organic Nanosized Materials for CRC. Encyclopedia. Available online: https://encyclopedia.pub/entry/9508 (accessed on 06 September 2024).
Galateanu B. Organic Nanosized Materials for CRC. Encyclopedia. Available at: https://encyclopedia.pub/entry/9508. Accessed September 06, 2024.
Galateanu, Bianca. "Organic Nanosized Materials for CRC" Encyclopedia, https://encyclopedia.pub/entry/9508 (accessed September 06, 2024).
Galateanu, B. (2021, May 11). Organic Nanosized Materials for CRC. In Encyclopedia. https://encyclopedia.pub/entry/9508
Galateanu, Bianca. "Organic Nanosized Materials for CRC." Encyclopedia. Web. 11 May, 2021.
Organic Nanosized Materials for CRC
Edit

Globally, colorectal cancer (CRC) ranks as one of the most prevalent types of cancers at the moment, being the second cause of cancer-related deaths. The CRC chemotherapy backbone is represented by 5-fluorouracil, oxaliplatin, irinotecan, and their combinations, but their administration presents several serious disadvantages, such as poor bioavailability, lack of tumor specificity, and susceptibility to multidrug resistance. To address these limitations, nanomedicine has arisen as a powerful tool to improve current chemotherapy since nanosized carriers hold great promise in improving the stability and solubility of the drug payload and enhancing the active concentration of the drug that reaches the tumor tissue, increasing, therefore, the safety and efficacy of the treatment.

drug-delivery systems nanoparticles liposomes colorectal cancer

1. Introduction

Globally, colorectal cancer (CRC) ranks as one of the most prevalent types of cancers at the moment, being the second cause of cancer-related deaths [1][2]. According to World Health Organization (WHO), CRC is positioned worldwide as the third most common type of cancer as incidence, and the second as mortality, with 1.93 million new cases and 935,000 deaths reported in 2020 [2][3]. The CRC incidence is directly correlated with the economic status of the countries, the most affected countries being low- and middle-income, where lifestyle habits and dietary patterns sustain the development of this malignancy [4][5][6]. Despite the existence of screening programs that allow CRC diagnosis in early stages [7], CRC morbidity and mortality are steadily increasing even in developed countries. This trend is correlated with the disease stage at the time of diagnosis that holds a great impact on the 5-year relative survival rate of CRC patients. The 5-year relative survival rate decreases from ~90% for stage I CRC to ~10% for stage IV metastatic CRC (mCRC) [8], the disease stage being also a crucial factor in the establishment of a proper and effective therapeutic approach. Unfortunately, the onset of CRC is asymptomatic, a feature that is responsible for the large number of patients in advanced stages of the disease at initial diagnosis. While one-quarter of the CRC patients are diagnosed from the beginning with mCRC, more than half of the patients will develop metastasis along with CRC progression [1][9]. More, the initiation of the screening programs is typically recommended for adults with ages above 50 years, but the recent rising incidence among young adults could rewrite the current CRC screening guidelines [10]. For example, the American Cancer Society already lowered the age threshold to 45 years for individuals with risk for CRC development [11], but further adjustments need to be done at a global level to lower the CRC burden and ensure the detection of CRC in early stages.

At the moment, several therapeutic approaches are implemented in clinics for CRC management such as surgery [12][13][14], radiotherapy [15][16], chemotherapy [17][18][19], and targeted therapy if applicable [20]. The choice of the approach is dependent on the stage of CRC at the initial diagnosis. While CRC patients diagnosed in the early stages of the disease by colonoscopy or sigmoidoscopy, which present a clean tumor with well-delimited margins, qualify for surgical resection of the tumor, the major challenge is represented by patients in stage III/IV, where the disease is spread to the lymph nodes and distant organs. For these patients, adjuvant chemotherapy is mandatory to control CRC and metastatic invasion, followed by tumor and metastases surgical resection if allowed [21].

Despite the recent advances in cytotoxic chemotherapy and targeted therapy, the prognosis of CRC remains unsatisfactory, especially for mCRC patients [22]. The severe side effects associated with chemotherapy and the development of multidrug resistance are critical issues that hinder the proper care of CRC patients by limiting treatment efficacy and leading to chemotherapy failure. In this view, nanomedicine has emerged as a powerful tool to improve the existing drug-based strategies for CRC treatment. Nanomedicines are nanosized carrier biomaterials that are used as shuttles to deliver drug cargos to the tumor tissue. These nanosized drug-delivery systems possess the potential to improve the stability and solubility of the drug payload and to enhance the active concentration of the drug that reaches the tumor tissue, increasing the safety and efficacy of the treatment [23][24]. In this context, the present review offers an overview of the most recent advances in the development of organic nanosized drug-delivery systems as smart therapeutic tools in CRC management.

2. Current Pharmacotherapy Available for CRC

The existing pharmacotherapy available for CRC treatment relies on the administration of cytotoxic agents, targeted therapies, and their combinations. Among cytotoxic agents, CRC chemotherapeutic regiments are based on the administration of capecitabine or 5-fluorouracil (5-FU), oxaliplatin (OXP), and irinotecan (IRI) (Figure 1), anticancer agents that are usually combinatorically administrated to amplify their antineoplastic potential: 5-FU/LV/OXP (FOLFOX), 5-FU/LV/IRI (FOLFIRI), and 5-FU/LV/IRI/OXA (FOLFIRINOX) [25][26]. These anticancer agents act on both DNA or RNA synthesis to exert their cytotoxic effects [27][28][29], and although they still represent the backbone of CRC chemotherapy, their administration presents several serious disadvantages, such as poor bioavailability [27][30][31], lack of tumor specificity, and susceptibility to multidrug resistance [28][32][33].

Figure 1. Structure of conventional chemotherapeutic agents administrated in CRC.

One such example is 5-FU, a fluoropyrimidine analogue and a first-line drug in colorectal cancer chemotherapeutic regimens. The conversion of 5-FU into its inactive metabolite, 5,6-dihydro-5-fluorouracil (DHFU), is directly dependent on the dihydropyrimidine dehydrogenase (DPYD) enzyme, which is encoded by the DPYD gene [34]. Several genotypic variants of DPYD were identified in the population that can be translated into individual variations in the drug’s clearance [35], leading to 5-FU systemic toxicity and drug resistance [36]. Consequently, 5-FU administration is associated with side effects that can vary from mild to severe, depending on the patient’s tolerance [37][38], severely limiting the dose of the drug and leading to poor antineoplastic results.

For a more specific approach to CRC pathology, the strategy relies on targeting molecules that play a central role in tumor development and progression, generally by using monoclonal antibodies [39]. In this respect, anti-angiogenic drugs targeting the vascular endothelial growth factor (VEGF) pathway hold a key role in treating patients with metastatic CRC. Bevacizumab [40][41][42], ramucirumab [43], and aflibercept [44][45] are used for targeting angiogenesis, a culprit in tumor development and progression [46][47]. For CRC patients that do not harbor KRAS or NRAS mutations, cetuximab and panitumumab can be administrated as anti-EGFR antibodies [48][49][50]. Epidermal growth factor receptor (EGFR) modulates CRC initiation and progression due to its key role in activating downstream signaling pathways that control tumor cell growth, differentiation, and proliferation [51][52][53][54]. More recently, immune checkpoint blockade agents such as pembrolizumab, nivolumab, and ipilimumab have been exploited as targeted therapy in CRC [20]. Promising clinical studies raise the hope of survival improvement in CRC patients when administering bevacizumab and standard chemotherapy combined approach [55][56].

3. Targeting Strategies for Drug-Delivery Systems in CRC

An ideal drug-delivery system for anticancer agents should be designed and developed to significantly improve the efficacy of the drug-free traditional treatment by functioning as a protective shuttle for avoiding drug degradation, thus ensuring an increased drug concentration that reaches the tumor. More, in the preparation phase, the nanoparticles should be tailored to enhance their tumor selectivity and ensure accumulation to the tumor site, reducing therefore the cytotoxic effects on normal healthy tissues.

The physicochemical proprieties of the nanocarriers impact their effectiveness as drug-delivery systems in CRC management. The size and shape of the nanoparticles, as well as the charge of their surface, have a non-specific effect, which is determined by the interaction with the cell membrane [57]. Since the surface of the cell membrane is negatively charged, positively charged particles have the highest penetration rate, while the lowest penetration rate is observed at nanoparticles with a negative surface charge [58]. It has been shown that a high positive charge on the surface of particles leads to enhanced cellular uptake, while a negative charge contributes to reduced absorption of nanoparticles [59]. It is noteworthy that the immobilization of some proteins on the surface of charged nanoparticles has a leveling effect on the charge factor, contributing to a similar rate of absorption of positively and negatively charged particles [60].

The size of nanoparticles has a significant impact on the rate and mechanism of their absorption. The rate of nanoparticle absorption by cells increases with particle size [61]. For particles with a diameter of more than 200 nm, absorption generally occurs through clathrin-dependent endocytosis. The mechanism of smaller particle absorption remains a subject of discussion, being probably associated with passive transport through the pores of the cell membrane or directly through membrane fusion [62]. Lastly, the nanoparticles smaller than 100 nm may be absorbed through the nuclear membrane, enabling the transport of drugs into the cell nucleus. Thus, the size of the carrier particles determines the rate and mechanism of its absorption by the cell, as well as the character of drug distribution between the cell organelles [63].

The penetration rate of nanoparticles into the cell also depends on their geometric shape. It has been shown that spherical particles are absorbed by cells at the highest rate [64]. The interpretation of data on the effect of the size and shape of particles on the nature of their absorption by the cell should be carried out with caution since nanoparticles are simultaneously involved in at least three processes: diffusion, sedimentation, and agglomeration [61].

Although these factors are common for the therapy of all types of cancers, CRC of various etiology [65][66] has special requirements for ensuring the bioavailability of pharmacologically active substances delivered through nanoparticles [67]. The problem of increasing the bioavailability of drugs can be solved, at least partially, by introducing CRC-specific ligands on nanoparticle surfaces [68][69]. Based on these observations, there are two strategies by which nanosized drug-delivery systems reach tumor cells (Figure 2): (i) passive targeting, where nanoparticles take advantage of the abnormalities of tumor vasculature to accumulate at the tumor site, and (ii) active targeting, where nanoparticles are functionalized with moieties that directly target the tumor cells.

Figure 2. Schematic representation of drug-delivery systems targeting strategies.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2019. Cancer J. Clin. 2019, 69, 7–34.
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Cancer J. Clin. 2020.
  3. Ferlizza, E.; Solmi, R.; Sgarzi, M.; Ricciardiello, L.; Lauriola, M. The Roadmap of Colorectal Cancer Screening. Cancers 2021, 13, 1101.
  4. Fidler, M.M.; Soerjomataram, I.; Bray, F. A Global View on Cancer Incidence and National Levels of the Human Development Index. Int. J. Cancer 2016, 139, 2436–2446.
  5. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Cancer J. Clin. 2018, 68, 394–424.
  6. Thanikachalam, K.; Khan, G. Colorectal Cancer and Nutrition. Nutrients 2019, 11, 164.
  7. Bevan, R.; Rutter, M.D. Colorectal Cancer Screening-Who, How, and When? Clin. Endosc. 2018, 51, 37–49.
  8. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of Colorectal Cancer: Incidence, Mortality, Survival, and Risk Factors. Przeglad Gastroenterol. 2019, 14, 89.
  9. Riihimäki, M.; Hemminki, A.; Sundquist, J.; Hemminki, K. Patterns of Metastasis in Colon and Rectal Cancer. Sci. Rep. 2016, 6, 1–9.
  10. Vuik, F.E.; Nieuwenburg, S.A.; Bardou, M.; Lansdorp-Vogelaar, I.; Dinis-Ribeiro, M.; Bento, M.J.; Zadnik, V.; Pellisé, M.; Esteban, L.; Kaminski, M.F. Increasing Incidence of Colorectal Cancer in Young Adults in Europe over the Last 25 Years. Gut 2019, 68, 1820–1826.
  11. Wolf, A.M.; Fontham, E.T.; Church, T.R.; Flowers, C.R.; Guerra, C.E.; LaMonte, S.J.; Etzioni, R.; McKenna, M.T.; Oeffinger, K.C.; Shih, Y.T. Colorectal Cancer Screening for Average-risk Adults: 2018 Guideline Update from the American Cancer Society. Cancer J. Clin. 2018, 68, 250–281.
  12. Nagtegaal, I.D.; Quirke, P. What Is the Role for the Circumferential Margin in the Modern Treatment of Rectal Cancer? J. Clin. Oncol. 2008, 26, 303–312.
  13. Heald, R.; Ryall, R. Recurrence and Survival after Total Mesorectal Excision for Rectal Cancer. Lancet 1986, 327, 1479–1482.
  14. Trastulli, S.; Cirocchi, R.; Listorti, C.; Cavaliere, D.; Avenia, N.; Gulla, N.; Giustozzi, G.; Sciannameo, F.; Noya, G.; Boselli, C. Laparoscopic vs Open Resection for Rectal Cancer: A Meta-analysis of Randomized Clinical Trials. Colorectal Dis. 2012, 14, e277–e296.
  15. Kapiteijn, E.; Marijnen, C.A.; Nagtegaal, I.D.; Putter, H.; Steup, W.H.; Wiggers, T.; Rutten, H.J.; Pahlman, L.; Glimelius, B.; van Krieken, J.H.J. Preoperative Radiotherapy Combined with Total Mesorectal Excision for Resectable Rectal Cancer. N. Engl. J. Med. 2001, 345, 638–646.
  16. Bujko, K.; Nowacki, M.; Nasierowska-Guttmejer, A.; Michalski, W.; Bebenek, M.; Kryj, M. Long-term Results of a Randomized Trial Comparing Preoperative Short-course Radiotherapy with Preoperative Conventionally Fractionated Chemoradiation for Rectal Cancer. Br. J. Surg. 2006, 93, 1215–1223.
  17. Kim, J.H. Chemotherapy for Colorectal Cancer in the Elderly. World J. Gastroenterol. 2015, 21, 5158.
  18. Gill, S.; Loprinzi, C.L.; Sargent, D.J.; Thomé, S.D.; Alberts, S.R.; Haller, D.G.; Benedetti, J.; Francini, G.; Shepherd, L.E.; Francois Seitz, J. Pooled Analysis of Fluorouracil-Based Adjuvant Therapy for Stage II and III Colon Cancer: Who Benefits and by How Much? J. Clin. Oncol. 2004, 22, 1797–1806.
  19. André, T.; Boni, C.; Navarro, M.; Tabernero, J.; Hickish, T.; Topham, C.; Bonetti, A.; Clingan, P.; Bridgewater, J.; Rivera, F. Improved Overall Survival with Oxaliplatin, Fluorouracil, and Leucovorin as Adjuvant Treatment in Stage II or III Colon Cancer in the MOSAIC Trial. J. Clin. Oncol. 2009, 27, 3109–3116.
  20. Xie, Y.-H.; Chen, Y.-X.; Fang, J.-Y. Comprehensive Review of Targeted Therapy for Colorectal Cancer. Signal Transduct. Target. Ther. 2020, 5, 1–30.
  21. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; Van Krieken, J.; Aderka, D.; Aguilar, E.A.; Bardelli, A.; Benson, A.; Bodoky, G. ESMO Consensus Guidelines for the Management of Patients with Metastatic Colorectal Cancer. Ann. Oncol. 2016, 27, 1386–1422.
  22. Wang, J.; Li, S.; Liu, Y.; Zhang, C.; Li, H.; Lai, B. Metastatic Patterns and Survival Outcomes in Patients with Stage IV Colon Cancer: A Population-based Analysis. Cancer Med. 2020, 9, 361–373.
  23. Blanco, E.; Shen, H.; Ferrari, M. Principles of Nanoparticle Design for Overcoming Biological Barriers to Drug Delivery. Nat. Biotechnol. 2015, 33, 941.
  24. Kou, L.; Bhutia, Y.D.; Yao, Q.; He, Z.; Sun, J.; Ganapathy, V. Transporter-Guided Delivery of Nanoparticles to Improve Drug Permeation across Cellular Barriers and Drug Exposure to Selective Cell Types. Front. Pharmacol. 2018, 9, 27.
  25. 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. Colorectal Cancer 2015, 14, 1–10.
  26. Gill, S.; Thomas, R.; Goldberg, R.M. Colorectal Cancer Chemotherapy. Aliment. Pharmacol. Ther. 2003, 18, 683–692.
  27. Alcindor, T.; Beauger, N. Oxaliplatin: A Review in the Era of Molecularly Targeted Therapy. Curr. Oncol. 2011, 18, 18–25.
  28. Zhang, N.; Yin, Y.; Xu, S.-J.; Chen, W.-S. 5-Fluorouracil: Mechanisms of Resistance and Reversal Strategies. Molecules 2008, 13, 1551–1569.
  29. Fujita, K.; Kubota, Y.; Ishida, H.; Sasaki, Y. Irinotecan, a Key Chemotherapeutic Drug for Metastatic Colorectal Cancer. World J. Gastroenterol. 2015, 21, 12234.
  30. Diasio, R.B.; Harris, B.E. Clinical Pharmacology of 5-Fluorouracil. Clin. Pharmacokinet. 1989, 16, 215–237.
  31. Kümler, I.; Sørensen, P.G.; Palshof, J.; Høgdall, E.; Skovrider-Ruminski, W.; Theile, S.; Fullerton, A.; Nielsen, P.; Jensen, B.V.; Nielsen, D. Oral Administration of Irinotecan in Patients with Solid Tumors: An Open-Label, Phase I, Dose Escalating Study Evaluating Safety, Tolerability and Pharmacokinetics. Cancer Chemother. Pharmacol. 2019, 83, 169–178.
  32. Hsu, H.; Chen, M.; Baskaran, R.; Lin, Y.; Day, C.H.; Lin, Y.; Tu, C.; Vijaya Padma, V.; Kuo, W.; Huang, C. Oxaliplatin Resistance in Colorectal Cancer Cells Is Mediated via Activation of ABCG2 to Alleviate ER Stress Induced Apoptosis. J. Cell. Physiol. 2018, 233, 5458–5467.
  33. Xu, Y.; Villalona-Calero, M. Irinotecan: Mechanisms of Tumor Resistance and Novel Strategies for Modulating Its Activity. Ann. Oncol. 2002, 13, 1841–1851.
  34. Negrei, C.; Hudita, A.; Ginghina, O.; Galateanu, B.; Voicu, S.N.; Stan, M.; Costache, M.; Fenga, C.; Drakoulis, N.; Tsatsakis, A.M. Colon Cancer Cells Gene Expression Signature as Response to 5-Fluorouracil, Oxaliplatin, and Folinic Acid Treatment. Front. Pharmacol. 2016, 7, 172.
  35. Johnson, M.R.; Hageboutros, A.; Wang, K.; High, L.; Smith, J.B.; Diasio, R.B. Life-Threatening Toxicity in a Dihydropyrimidine Dehydrogenase-Deficient Patient after Treatment with Topical 5-Fluorouracil. Clin. Cancer Res. 1999, 5, 2006–2011.
  36. Nikolouzakis, T.K.; Stivaktakis, P.D.; Apalaki, P.; Kalliantasi, K.; Sapsakos, T.M.; Spandidos, D.A.; Tsatsakis, A.; Souglakos, J.; Tsiaoussis, J. Effect of Systemic Treatment on the Micronuclei Frequency in the Peripheral Blood of Patients with Metastatic Colorectal Cancer. Oncol. Lett. 2019, 17, 2703–2712.
  37. Oun, R.; Moussa, Y.E.; Wheate, N.J. The Side Effects of Platinum-Based Chemotherapy Drugs: A Review for Chemists. Dalton Trans. 2018, 47, 6645–6653.
  38. Braun, M.S.; Seymour, M.T. Balancing the Efficacy and Toxicity of Chemotherapy in Colorectal Cancer. Ther. Adv. Med. Oncol. 2011, 3, 43–52.
  39. Tariman, J.D. Changes in Cancer Treatment: Mabs, Mibs, Mids, Nabs, and Nibs. Nurs. Clin. 2017, 52, 65–81.
  40. Cremolini, C.; Loupakis, F.; Antoniotti, C.; Lupi, C.; Sensi, E.; Lonardi, S.; Mezi, S.; Tomasello, G.; Ronzoni, M.; Zaniboni, A. FOLFOXIRI plus Bevacizumab versus FOLFIRI plus Bevacizumab as First-Line Treatment of Patients with Metastatic Colorectal Cancer: Updated Overall Survival and Molecular Subgroup Analyses of the Open-Label, Phase 3 TRIBE Study. Lancet Oncol. 2015, 16, 1306–1315.
  41. Cunningham, D.; Lang, I.; Marcuello, E.; Lorusso, V.; Ocvirk, J.; Shin, D.B.; Jonker, D.; Osborne, S.; Andre, N.; Waterkamp, D. Bevacizumab plus Capecitabine versus Capecitabine Alone in Elderly Patients with Previously Untreated Metastatic Colorectal Cancer (AVEX): An Open-Label, Randomised Phase 3 Trial. Lancet Oncol. 2013, 14, 1077–1085.
  42. Simkens, L.H.; van Tinteren, H.; May, A.; ten Tije, A.J.; Creemers, G.-J.M.; Loosveld, O.J.; de Jongh, F.E.; Erdkamp, F.L.; Erjavec, Z.; van der Torren, A.M. Maintenance Treatment with Capecitabine and Bevacizumab in Metastatic Colorectal Cancer (CAIRO3): A Phase 3 Randomised Controlled Trial of the Dutch Colorectal Cancer Group. Lancet 2015, 385, 1843–1852.
  43. Tabernero, J.; Yoshino, T.; Cohn, A.L.; Obermannova, R.; Bodoky, G.; Garcia-Carbonero, R.; Ciuleanu, T.-E.; Portnoy, D.C.; Van Cutsem, E.; Grothey, A. Ramucirumab versus Placebo in Combination with Second-Line FOLFIRI in Patients with Metastatic Colorectal Carcinoma That Progressed during or after First-Line Therapy with Bevacizumab, Oxaliplatin, and a Fluoropyrimidine (RAISE): A Randomised, Double-Blind, Multicentre, Phase 3 Study. Lancet Oncol. 2015, 16, 499–508.
  44. Folprecht, G.; Pericay, C.; Saunders, M.P.; Thomas, A.; Lopez, R.L.; Roh, J.; Chistyakov, V.; Höhler, T.; Kim, J.-S.; Hofheinz, R.-D. Oxaliplatin and 5-FU/Folinic Acid (Modified FOLFOX6) with or without Aflibercept in First-Line Treatment of Patients with Metastatic Colorectal Cancer: The AFFIRM Study. Ann. Oncol. 2016, 27, 1273–1279.
  45. Van Cutsem, E.; Tabernero, J.; Lakomy, R.; Prenen, H.; Prausová, J.; Macarulla, T.; Ruff, P.; Van Hazel, G.A.; Moiseyenko, V.; Ferry, D. Addition of Aflibercept to Fluorouracil, Leucovorin, and Irinotecan Improves Survival in a Phase III Randomized Trial in Patients with Metastatic Colorectal Cancer Previously Treated with an Oxaliplatin-Based Regimen. J. Clin. Oncol. 2012, 30, 3499–3506.
  46. Zuazo-Gaztelu, I.; Casanovas, O. Unraveling the Role of Angiogenesis in Cancer Ecosystems. Front. Oncol. 2018, 8, 248.
  47. Folkman, J. Role of Angiogenesis in Tumor Growth and Metastasis; Elsevier: Amsterdam, The Netherlands, 2002; Volume 29, pp. 15–18.
  48. Seymour, M.T.; Brown, S.R.; Middleton, G.; Maughan, T.; Richman, S.; Gwyther, S.; Lowe, C.; Seligmann, J.F.; Wadsley, J.; Maisey, N. Panitumumab and Irinotecan versus Irinotecan Alone for Patients with KRAS Wild-Type, Fluorouracil-Resistant Advanced Colorectal Cancer (PICCOLO): A Prospectively Stratified Randomised Trial. Lancet Oncol. 2013, 14, 749–759.
  49. Douillard, J.-Y.; Siena, S.; Cassidy, J.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J. Randomized, Phase III Trial of Panitumumab with Infusional Fluorouracil, Leucovorin, and Oxaliplatin (FOLFOX4) versus FOLFOX4 Alone as First-Line Treatment in Patients with Previously Untreated Metastatic Colorectal Cancer: The PRIME Study. J. Clin. Oncol. 2010, 28, 4697–4705.
  50. Van Cutsem, E.; Köhne, C.-H.; Hitre, E.; Zaluski, J.; Chang Chien, C.-R.; Makhson, A.; D’Haens, G.; Pintér, T.; Lim, R.; Bodoky, G. Cetuximab and Chemotherapy as Initial Treatment for Metastatic Colorectal Cancer. N. Engl. J. Med. 2009, 360, 1408–1417.
  51. Markman, B.; Javier Ramos, F.; Capdevila, J.; Tabernero, J. EGFR and KRAS in Colorectal Cancer. Adv. Clin. Chem. 2010, 51, 72.
  52. Wee, P.; Wang, Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers 2017, 9, 52.
  53. Roskoski, R., Jr. Small Molecule Inhibitors Targeting the EGFR/ErbB Family of Protein-Tyrosine Kinases in Human Cancers. Pharmacol. Res. 2019, 139, 395–411.
  54. Wang, Z. ErbB Receptors and Cancer. ErbB Recept. Signal. 2017, 3–35.
  55. Hubbard, J.M.; Grothey, A. Progress in Defining First-Line and Maintenance Therapies. Nat. Rev. Clin. Oncol. 2015, 12, 73–74.
  56. Loupakis, F.; Cremolini, C.; Salvatore, L.; Masi, G.; Sensi, E.; Schirripa, M.; Michelucci, A.; Pfanner, E.; Brunetti, I.; Lupi, C. FOLFOXIRI plus Bevacizumab as First-Line Treatment in BRAF Mutant Metastatic Colorectal Cancer. Eur. J. Cancer 2014, 50, 57–63.
  57. Zhang, J.; Tang, H.; Liu, Z.; Chen, B. Effects of Major Parameters of Nanoparticles on Their Physical and Chemical Properties and Recent Application of Nanodrug Delivery System in Targeted Chemotherapy. Int. J. Nanomed. 2017, 12, 8483.
  58. Pittella, F.; Zhang, M.; Lee, Y.; Kim, H.J.; Tockary, T.; Osada, K.; Ishii, T.; Miyata, K.; Nishiyama, N.; Kataoka, K. Enhanced Endosomal Escape of SiRNA-Incorporating Hybrid Nanoparticles from Calcium Phosphate and PEG-Block Charge-Conversional Polymer for Efficient Gene Knockdown with Negligible Cytotoxicity. Biomaterials 2011, 32, 3106–3114.
  59. Graf, C.; Gao, Q.; Schütz, I.; Noufele, C.N.; Ruan, W.; Posselt, U.; Korotianskiy, E.; Nordmeyer, D.; Rancan, F.; Hadam, S. Surface Functionalization of Silica Nanoparticles Supports Colloidal Stability in Physiological Media and Facilitates Internalization in Cells. Langmuir 2012, 28, 7598–7613.
  60. Tang, J.; Li, L.; Howard, C.B.; Mahler, S.M.; Huang, L.; Xu, Z.P. Preparation of Optimized Lipid-Coated Calcium Phosphate Nanoparticles for Enhanced in vitro Gene Delivery to Breast Cancer Cells. J. Mater. Chem. B 2015, 3, 6805–6812.
  61. Limbach, L.K.; Li, Y.; Grass, R.N.; Brunner, T.J.; Hintermann, M.A.; Muller, M.; Gunther, D.; Stark, W.J. Oxide Nanoparticle Uptake in Human Lung Fibroblasts: Effects of Particle Size, Agglomeration, and Diffusion at Low Concentrations. Environ. Sci. Technol. 2005, 39, 9370–9376.
  62. Herd, H.; Daum, N.; Jones, A.T.; Huwer, H.; Ghandehari, H.; Lehr, C.-M. Nanoparticle Geometry and Surface Orientation Influence Mode of Cellular Uptake. ACS Nano 2013, 7, 1961–1973.
  63. Sabourian, P.; Yazdani, G.; Ashraf, S.S.; Frounchi, M.; Mashayekhan, S.; Kiani, S.; Kakkar, A. Effect of Physico-Chemical Properties of Nanoparticles on Their Intracellular Uptake. Int. J. Mol. Sci. 2020, 21, 8019.
  64. Liu, Y.; Tan, J.; Thomas, A.; Ou-Yang, D.; Muzykantov, V.R. The Shape of Things to Come: Importance of Design in Nanotechnology for Drug Delivery. Ther. Deliv. 2012, 3, 181–194.
  65. Iftode, A.; Drăghici, G.A.; Macașoi, I.; Marcovici, I.; Coricovac, D.E.; Dragoi, R.; Tischer, A.; Kovatsi, L.; Tsatsakis, A.M.; Cretu, O.; et al. Exposure to Cadmium and Copper Triggers Cytotoxic Effects and Epigenetic Changes in Human Colorectal Carcinoma HT-29 Cells. Exp. Ther. Med. 2021, 21, 100.
  66. Krasanakis, T.; Nikolouzakis, T.K.; Sgantzos, M.; Mariolis-Sapsakos, T.; Souglakos, J.; Spandidos, D.A.; Tsitsimpikou, C.; Tsatsakis, A.; Tsiaoussis, J. Role of Anabolic Agents in Colorectal Carcinogenesis: Myths and Realities (Review). Oncol. Rep. 2019, 42, 2228–2244.
  67. Ashraf, N.; Mahipal, A.; Kim, R. Viral Vector Vaccines to Treat Colorectal Cancer. Curr. Colorectal Cancer Rep. 2013, 9, 398–405.
  68. Cheng, X.J.; Lin, J.C.; Ding, Y.F.; Zhu, L.; Ye, J.; Tu, S.P. Survivin Inhibitor YM155 Suppresses Gastric Cancer Xenograft Growth in Mice without Affecting Normal Tissues. Oncotarget 2016, 7, 7096–7109.
  69. Basak, S.; Eck, S.; Gutzmer, R.; Smith, A.J.; Birebent, B.; Purev, E.; Staib, L.; Somasundaram, R.; Zaloudik, J.; Li, W.; et al. Colorectal Cancer Vaccines: Antiidiotypic Antibody, Recombinant Protein, and Viral Vector. Ann. N. Y. Acad. Sci. 2000, 910, 237–253.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 424
Revisions: 2 times (View History)
Update Date: 11 May 2021
1000/1000
ScholarVision Creations