Open Access

Research progress on common adverse events caused by targeted therapy for colorectal cancer (Review)

  • Authors:
    • Bo Zhang
    • Chenyan Fang
    • Dehou Deng
    • Liang Xia
  • View Affiliations

  • Published online on: May 7, 2018     https://doi.org/10.3892/ol.2018.8651
  • Pages: 27-33
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As targeted drug therapy is increasingly applied in the treatment of colon cancer, understanding and managing the adverse reactions of patients is becoming increasingly important. The present review examines the mechanisms of and adverse reactions to the most commonly used targeted drugs for colon cancer, and discusses methods of coping with these adverse reactions. Approved targeted drugs for metastatic colon cancer include monoclonal antibodies targeting vascular endothelial growth factor (VEGF), including bevacizumab, ziv‑aflibercept and regorafenib, and monoclonal antibodies targeting epithelial growth factor receptor (EGFR), including cetuximab and panitumumab. The present review assesses the major adverse effects of these drugs and methods of dealing with reactions to them. VEGF inhibitors primarily result in cardiovascular and kidney problems. Meanwhile, EGFR receptor inhibitors are frequently reported to cause rashes, diarrhea and hypertension, and are reviewed from the point of view of resulting electrolyte disturbances.

Introduction

Colorectal cancer is one of the most common malignancies globally (13). According to statistics by the World Health Organization, ~1.2 million new cases of colorectal cancer were identified globally in 2008 (3). Each year, >0.6 million patients succumb to mortality due to colorectal cancer, either directly or indirectly. The incidence of this malignancy is increased among males compared with females. Furthermore, the risk of colorectal cancer increases with age. For example, the median onset age for colorectal cancer in developed countries is 70 years. In the USA, colorectal cancer ranks third in terms of incidence and mortality (2). In China, colorectal cancer ranks fifth in terms of incidence (4,5).

Approximately 20% of all cases with colorectal cancer are stage IV (tumor, node and metastasis staging system; www.nccn.org/patients) at the first diagnosis, and the 5-year survival rate of these cases is only 13% (4,68). Chemotherapy is the main treatment for metastatic and local late-stage colorectal cancer; however, the toxicity and adverse side effects maybe intolerable for patients with a poor prognosis (9). In comparison, targeted therapy is associated with improved compliance, decreased toxicity and fewer side effects in addition to improved prognosis (10,11).

However, increasingly, adverse events associated with targeted therapy have been reported in previous years (1215). At present, the approved targeted drugs for metastatic colorectal cancer include monoclonal antibodies targeting vascular endothelial growth factor (VEGF) and those targeting the epidermal growth factor receptor (EGFR). The representatives of the first category include bevacizumab, ziv-aflibercept and regorafenib; and those of the second category include cetuximab and panitumumab. The present review assesses the adverse events and the corresponding treatments following the use of these two categories of targeted drugs.

VEGF inhibitors

VEGF promotes angiogenesis. There are five ligands of VEGF (Fig. 1), as follows: VEGFA, VEGFB, VEGFC, VEGFD and placental growth factor (PlGF). Additionally, there are three receptors of VEGF: VEGFR1, VEGFR2 and VEGFR3, and 2 common receptors, neuropillin 1 and neuropillin 2. VEGFR1 binds to VEGFA, VEGFB and PlGF; VEGFR2 binds to VEGFA, VEGFC and VEGFD. VEGFR2 is considered to be the receptor involved in regulating angiogenesis, whereas VEGFR1 and VEGFR3 are involved in the chemotaxis of monocytes, survival of hematopoietic stem cells and formation of lymphatic vessels (16). VEGF inhibitors mainly function by blocking VEGF ligands. For example, bevacizumab inhibits the binding of VEGFA to VEGFR2, while ziv-aflibercept functions by intercepting the binding of VEGFA, VEGFB and PlGF (1720).

The common adverse events arising from the use of VEGF inhibitors are outlined below.

Hypertension
Mechanism of occurrence

VEGF induces the synthesis of nitric oxide synthase via endothelial cells and results in the release of nitric oxide, a notable vasodilator (21). The blocking of VEGF-associated signaling pathways may decrease the secretion of nitric oxide synthase and result in hypertension (22,23). A previous study suggested that VEGF inhibitors induce hypertension by altering the rennin-angiotensin-aldosterone system (24). Another study hypothesized that VEGF inhibitors decrease the microvessel density of the internal organs in patients with hypertension, thus decreasing the blood flow rate and resulting in hypertension (25).

Incidence

Kabbinavar et al (26) reported the results of a phase II clinical trial. The incidence of stage III hypertension was 2.9% following intravenous administration of fluorouracil alone. When fluorouracil was combined with bevacizumab, the incidence of hypertension increased to 60%. It was noteworthy that all recruited patients were aged >65 which may have affected drug metabolism. However, for the same population, the combined use of bevacizumab and S-1 resulted in an incidence of hypertension of 11% (27). The most notable limitation of this previous study was the small cohort size, in that only 56 patients were included.

As to the choice of targeted drugs, a previous meta-analysis (28) revealed that the overall incidence of hypertension was 42.4% in patients receiving ziv-aflibercept treatment; among them, the incidence of advanced hypertension was 17.4%. The overall incidence of hypertension was 23.6% following the use of bevacizumab, and the incidence of advanced hypertension was 7.9%. The overall incidence of hypertension was 44.4% for patients who were administered regorafenib, and the incidence of advanced hypertension was 12.5%. Another previous meta-analysis study (29), focusing on the adverse events occurring subsequent to bevacizumab treatment for non-small cell lung carcinoma, indicated that the incidence of hypertension was 19.55%, while that of advanced hypertension was 6.95%. This discrepancy between these previous studies may be due to the differing cancer types. Overall, VEGF inhibitors result in hypertension, particularly amongst elderly patients (30). Another population-based study revealed that the risk of hypertension induced by VEGF inhibitors was higher among those with a previous history of hypertension (31). Thus, VEGF inhibitors maybe used, but with caution, for patients with a previous history of hypertension.

Clinical treatment

The following principles should be followed in order to prevent and treat hypertension induced by VEGF inhibitors (32,33): i) Blood pressure monitoring should be performed for patients treated using VEGF inhibitors at least once every 2–3 weeks, and frequency of monitoring should be increased during treatment. ii) VEGF inhibitors should not be administered unless blood pressure is properly controlled. iii) If hypertension was once induced or aggravated by VEGF inhibitors for the patient, blood pressure monitoring should be continued even subsequent to the cessation of VEGF inhibitor treatment. iv) Any antihypertensive drugs may be used, however, the angiotensin converting enzyme inhibitor is considered to be the superior drug, as it may prevent or treat other side effects arising from treatment with VEGF inhibitors, namely, proteinuria.

Proteinuria
Mechanism of occurrence

Proteinuria is another side effect resulting from the use of VEGF inhibitors. If the protein content in the urine is >300 mg/dl, this usually indicates proteinuria (3439). Proteinuria caused by the use of VEGF inhibitors is asymptomatic (34) without obvious pathological changes of the kidney (35). As to the mechanism of occurrence of proteinuria, a previous study (36) proposed the intervention of a podocyte-derived VEGF signal axis. However, the glomerular podocytes may constitutively express VEGF and activate VEGFR2 on glomerular vascular endothelial cells, thus establishing and maintaining basic liver functions (36,37).

Incidence

The incidence of proteinuria appears to be dependent on the dose of VEGF inhibitors and the severity of hypertension (38,39). Generally speaking, VEGF inhibitors are more likely to induce hypertension compared with proteinuria. As demonstrated by a previous meta-analysis (40,41), the relative risk (RR) caused by VEGF inhibitors was 3.46 and that of proteinuria was 2.51 compared with the control group. Another meta-analysis included 6,882 cases from a total of 33 clinical trials, and the results revealed that the incidence of proteinuria was 18.7% among patients receiving VEGF inhibitor treatment and the incidence of advanced proteinuria (grade 3 or above) was 2.4% (42).

Clinical treatment

Prior to the use of VEGF inhibitors, screening for proteinuria should be performed. For patients that are negative for proteinuria, only screening is required prior to each treatment; for patients that are positive for proteinuria, evaluation by physicians in nephrology is required if the treatment with VEGF inhibitors is to be administered and the treatment should be highly individualized (43,44).

However, no standards have been established so far for the treatment of proteinuria caused by VEGF inhibitors. According to US Food and Drug Administration guidelines (44), anti-angiogenic drugs should be disused if protein content in the urine >2 g/24 h. Furthermore, if hypertension is induced by VEGF inhibitors and complicated by proteinuria, ACEI and angiotensin receptor blockers are often used for the effect of decreasing the level of protein in the urine and protecting the blood vessels (34).

Other adverse events

Other side effects caused by VEGF inhibitors include hemorrhage (45), diarrhea (46), cardiovascular events including myocardial infarction, thromboembolism, stroke and heart failure (47), gastrointestinal perforation (48), hand-foot syndrome (49) and reversible encephalopathy (50). Symptomatic treatments are usually adopted for these events. However, these events experience low incidence rates (41).

EGFR tyrosine kinase inhibitors

The EGFR signaling pathway is one of the first pathways discovered to be involved in the targeted therapy of tumors (Fig. 2). This pathway affects the proliferation, differentiation, migration and apoptosis of cells (51,52) and usually demonstrates abnormal expression and activation in various solid tumor types (5254). The common adverse events associated with EGFR tyrosine kinase inhibitors (TKIs) are outlined below.

Hypomagnesemia
Mechanism of occurrence

EGF is a hormone that regulates Mg2+ reabsorption in the kidney by activating the Mg2+ channel transient receptor potential cation channel subfamily M member 6 (55). EGFR TKIs antagonize the reabsorption, leading to hypomagnesemia (55).

Incidence

A previous meta-analysis study (56) revealed that the incidence of hypomagnesemia due to the use of cetuximab or panitumumab was 17%, and that of advanced hypomagnesemia was 3.5%. Compared with cetuximab, the risk of hypomagnesemia with the use of panitumumab was increased (57). The incidence of hypomagnesemia was positively correlated with the treatment duration. Another previous study (58) revealed that the incidence of hypomagnesemia due to the use of cetuximab for metastatic colorectal cancer was 6–47% (with a treatment duration of 3–6 months). Another two meta-analyses (59) focusing on cetuximab revealed that the incidence of grade 3 and 4 hypomagnesemia were 3.9 and 5.6% with cetuximab, respectively. The relative risk of grade 3 and 4 hypomagnesemia with combined chemotherapy was 8 and 4.75, respectively (60).

Clinical treatment

Hypomagnesemia is negatively associated with age, potentially due to the easier loss of Mg2+ (61,62). Furthermore, severe hypomagnesemia may lead to changes in muscle strength (including cramps, muscle weakness and ataxia), heart lesions (including coronary spasms, arrhythmia and long Q-T syndrome) and psychotic symptoms (including epilepsy, insanity, depression and anxiety) (63). These symptoms are easily confused with paraneoplastic syndrome (64). Thus, an electrolyte test is recommended prior to treatment, particularly for elderly patients, together with reexamination once every 2–4 weeks.

For grade 1 hypomagnesemia, which is usually asymptomatic, no interventions are recommended clinically (65). For grade 2 hypomagnesemia or above, intravenous infusion of Mg2+ is usually required as oral administration, which may induce diarrhea (61).

Hypopotassemia
Mechanism of occurrence

At present, the underlying mechanism of hypopotassemia occurrence caused by EGFR TKI is not well understood. It is a generally held opinion that EGFR TKIs may cause nephrotoxicity (66). A previous study (67) revealed the function of inhibited Mg2+ channel TRPM-6 in this process. When hypomagnesemia occurs, increased K+ is required for the repair of Na-K-adenosine triphosphatase. This may result in decreased renal potassium conservation and hence hypopotassemia.

Incidence

A previous meta-analysis (68) indicated that the incidence of hypopotassemia due to EGFR TKIs for patients with tumors was 14.5%. Notably, the incidence of grade 3/4 hypopotassemia in colorectal cancer treated by cetuximab and panitumumab was increased compared with that in other tumor types (RR for cetuximab, 2.19; RR for panitumumab, 3.30). Cisplatin is the preferred chemotherapeutic drug for many tumor types, but it may cause strong nephrotoxicity (69,70). Thus, this result requires a more specific explanation.

Clinical treatment

The treatment of hypopotassemia is not difficult clinically. Regular potassium tests are necessary during medication, and potassium may be infused if necessary. If hypopotassemia is complicated by hypomagnesemia, Mg2+ infusion is necessary (71).

Other adverse events

The most common side effect of EGFR TKIs is a skin rash, which was one of the first identified side effects (7276). A number of studies and literature reviews are focused on the topic of the skin rash caused by EGFR TKIs (7779).

Other adverse events associated with EGFR TKIs are diarrhea (80), hypertension (81,82), transfusion reactions (83) and hepatotoxicity (84).

Conclusions

Targeted therapy has unique advantages in treating colorectal cancer, and the progression of this therapy is fueled by an enhanced understanding of the tumor types it aims to target at a genetic level (8,8587). However, as targeting remains imprecise, certain adverse events are consistently reported (2630,39,41,4551). This is particularly true when the targeted therapy is combined with chemotherapy (27,28) Therefore, gaining a deeper understanding of the underlying mechanisms of the side effects occurring as a result of targeted therapy and identifying methods to treat them are highly prioritized. According to a previous study, the optimal method for coping with the side effects associated with targeted therapy is not to decrease the dosage, but through symptomatic treatment, which is capable of avoiding toxicity and adverse side effects (33). For example, VEGFR TKIs may cause hypertension, which may be prevented by proper preventive measures. The active control of blood pressure during targeted therapy can avoid damage of relevant target organs caused by hypertension and prevent progression of hypertension, as a medical consensus, ordinary patients with hypertension also require active control of blood pressure.

In conclusion, proficient understanding of the underlying molecular mechanisms of targeted drugs and the potential adverse events in addition to the proper treatments for these adverse effects is crucial for improving the prognosis of patients with cancer.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81502147) and the Youth Scientific Innovation Foundation of Zhejiang Cancer Hospital (grant no. QN201402).

Competing interests

The authors declare that they have no competing interests.

References

1 

Kumar B, Bhat ZI, Bansal S, Saini S, Naseem A, Wahabi K, Burman A, Kumar GT, Saluja SS and Rizvi MMA: Association of mitochondrial copy number variation and T16189C polymorphism with colorectal cancer in North Indian population. Tumour Biol. 39:10104283177402962017. View Article : Google Scholar : PubMed/NCBI

2 

Zhang J, Wang TY and Niu XC: Increased plasma levels of pentraxin 3 are associated with poor prognosis of colorectal carcinoma patients. Tohoku J Exp Med. 240:39–46. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Edge SB and Compton CC: The American Joint Committee on cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Kumar SK, Callander NS, Alsina M, Atanackovic D, Biermann JS, Chandler JC, Costello C, Faiman M, Fung HC, Gasparetto C, et al: Multiple myeloma, version 3.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 15:230–269. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Benson AB III, Venook AP, Cederquist L, Chan E, Chen YJ, Cooper HS, Deming D, Engstrom PF, Enzinger PC, Fichera A, et al: Colon cancer, version 1.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 15:370–398. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Gibson RJ, Keefe DM, Lalla RV, Bateman E, Blijlevens N, Fijlstra M, King EE, Stringer AM, van der Velden WJ, Yazbeck R, et al: Systematic review of agents for the management of gastrointestinal mucositis in cancer patients. Support Care Cancer. 21:313–326. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al: Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 369:1023–1034. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chien Chang CR, Makhson A, D'Haens G, Pintér T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 360:1408–1417. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Isobe T, Uchino K, Makiyama C, Ariyama H, Arita S, Tamura S, Komoda M, Kusaba H, Shirakawa T, Esaki T, et al: Analysis of adverse events of bevacizumab-containing systemic chemotherapy for metastatic colorectal cancer in Japan. Anticancer Res. 34:2035–2040. 2014.PubMed/NCBI

13 

Folprecht G, Pericay C, Saunders MP, Thomas A, Lopez Lopez R, Roh JK, Chistyakov V, Höhler T, Kim JS, Hofheinz RD, et al: 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. 27:1273–1279. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Calcagno F, Lenoble S, Lakkis Z, Nguyen T, Limat S, Borg C, Jary M, Kim S and Nerich V: Efficacy, safety and cost of regorafenib in patients with metastatic colorectal cancer in French clinical practice. Clin Med Insights Oncol. 10:59–66. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Zhang D, Ye J, Xu T and Xiong B: Treatment related severe and fatal adverse events with cetuximab in colorectal cancer patients: A meta-analysis. J Chemother. 25:170–175. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Ferrara N: Vascular endothelial growth factor as a target for anticancer therapy. Oncologist. 9 Suppl 1:S2–S10. 2004. View Article : Google Scholar

17 

Correale P, Botta C, Ciliberto D, Pastina P, Ingargiola R, Zappavigna S, Tassone P, Pirtoli L, Caraglia M and Tagliaferri P: Immunotherapy of colorectal cancer: New perspectives after a long path. Immunotherapy. 8:1281–1292. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Klein A and Loewenstein A: Therapeutic monoclonal antibodies and fragments: Bevacizumab. Dev Ophthalmol. 55:232–245. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Syed YY and McKeage K: Aflibercept: A review in metastatic colorectal cancer. Drugs. 75:1435–1445. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Sun W: Angiogenesis in metastatic colorectal cancer and the benefits of targeted therapy. J Hematol Oncol. 5:632012. View Article : Google Scholar : PubMed/NCBI

21 

Shiojima I, Sato K, Izumiya Y, Schiekofer S, Ito M, Liao R, Colucci WS and Walsh K: Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the transition to heart failure. J Clin Invest. 115:2108–2118. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Al-Husseini A, Kraskauskas D, Mezzaroma E, Nordio A, Farkas D, Drake JI, Abbate A, Felty Q and Voelkel NF: Vascular endothelial growth factor receptor 3 signaling contributes to angioobliterative pulmonary hypertension. Pulm Circ. 5:101–116. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Voelkel NF and Gomez-Arroyo J: The role of vascular endothelial growth factor in pulmonary arterial hypertension. The angiogenesis paradox. Am J Respir Cell Mol Biol. 51:474–484. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Robinson ES, Khankin EV, Choueiri TK, Dhawan MS, Rogers MJ, Karumanchi SA and Humphreys BD: Suppression of the nitric oxide pathway in metastatic renal cell carcinoma patients receiving vascular endothelial growth factor-signaling inhibitors. Hypertension. 56:1131–1136. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Gurevich F and Perazella MA: Renal effects of anti-angiogenesis therapy: Update for the internist. Am J Med. 122:322–328. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Kabbinavar FF, Schulz J, McCleod M, Patel T, Hamm JT, Hecht JR, Mass R, Perrou B, Nelson B and Novotny WF: Addition of bevacizumab to bolus fluorouracil and leucovorin in first-line metastatic colorectal cancer: Results of a randomized phase II trial. J Clin Oncol. 23:3697–3705. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Yoshida M, Muro K, Tsuji A, Hamamoto Y, Yoshino T, Yoshida K, Shirao K, Miyata Y, Takahari D, Takahashi T and Ohtsu A: Combination chemotherapy with bevacizumab and S-1 for elderly patients with metastatic colorectal cancer (BASIC trial). Eur J Cancer. 51:935–941. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Qi WX, Shen Z, Tang LN and Yao Y: Risk of hypertension in cancer patients treated with aflibercept: A systematic review and meta-analysis. Clin Drug Investig. 34:231–240. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Chen J, Lu Y and Zheng Y: Incidence and risk of hypertension with bevacizumab in non-small-cell lung cancer patients: A meta-analysis of randomized controlled trials. Drug Des Devel Ther. 9:4751–4760. 2015.PubMed/NCBI

30 

Sclafani F and Cunningham D: Bevacizumab in elderly patients with metastatic colorectal cancer. J Geriatr Oncol. 5:78–88. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Hamnvik OP, Choueiri TK, Turchin A, McKay RR, Goyal L, Davis M, Kaymakcalan MD and Williams JS: Clinical risk factors for the development of hypertension in patients treated with inhibitors of the VEGF signaling pathway. Cancer. 121:311–319. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Hurwitz H and Saini S: Bevacizumab in the treatment of metastatic colorectal cancer: Safety profile and management of adverse events. Semin Oncol. 33 5 Suppl 10:S26–S34. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Pande A, Lombardo J, Spangenthal E and Javle M: Hypertension secondary to anti-angiogenic therapy: Experience with bevacizumab. Anticancer Res. 27:3465–3470. 2007.PubMed/NCBI

34 

Izzedine H, Massard C, Spano JP, Goldwasser F, Khayat D and Soria JC: VEGF signalling inhibition-induced proteinuria: Mechanisms, significance and management. Eur J Cancer. 46:439–448. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, Steinberg SM, Chen HX and Rosenberg SA: A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med. 349:427–434. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Eremina V, Sood M, Haigh J, Nagy A, Lajoie G, Ferrara N, Gerber HP, Kikkawa Y, Miner JH and Quaggin SE: Glomerular-specific alterations of VEGF-A expression lead to distinct congenital and acquired renal diseases. J Clin Invest. 111:707–716. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Dimke H, Sparks MA, Thomson BR, Frische S, Coffman TM and Quaggin SE: Tubulovascular cross-talk by vascular endothelial growth factor a maintains peritubular microvasculature in kidney. J Am Soc Nephrol. 26:1027–1038. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Zhu X, Wu S, Dahut WL and Parikh CR: Risks of proteinuria and hypertension with bevacizumab, an antibody against vascular endothelial growth factor: Systematic review and meta-analysis. Am J Kidney Dis. 49:186–193. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Zhang ZF, Wang T, Liu LH and Guo HQ: Risks of proteinuria associated with vascular endothelial growth factor receptor tyrosine kinase inhibitors in cancer patients: A systematic review and meta-analysis. PLoS One. 9:e901352014. View Article : Google Scholar : PubMed/NCBI

40 

Izzedine H, Rixe O, Billemont B, Baumelou A and Deray G: Angiogenesis inhibitor therapies: Focus on kidney toxicity and hypertension. Am J Kidney Dis. 50:203–218. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Faruque LI, Lin M, Battistella M, Wiebe N, Reiman T, Hemmelgarn B, Thomas C and Tonelli M: Systematic review of the risk of adverse outcomes associated with vascular endothelial growth factor inhibitors for the treatment of cancer. PLoS One. 9:e1011452014. View Article : Google Scholar : PubMed/NCBI

42 

Qi WX, Sun YJ, Tang LN, Shen Z and Yao Y: Risk of gastrointestinal perforation in cancer patients treated with vascular endothelial growth factor receptor tyrosine kinase inhibitors: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 89:394–403. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Cartwright TH: Adverse events associated with antiangiogenic agents in combination with cytotoxic chemotherapy in metastatic colorectal cancer and their management. Clin Colorectal Cancer. 12:86–94. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Grenon NN: Managing toxicities associated with antiangiogenic biologic agents in combination with chemotherapy for metastatic colorectal cancer. Clin J Oncol Nurs. 17:425–433. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Tabernero J, Takayuki Y and Cohn AL: Correction to Lancet Oncol 2015; 16: 499–508. 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. 16:e2622015.PubMed/NCBI

46 

Liu S and Kurzrock R: Toxicity of targeted therapy: Implications for response and impact of genetic polymorphisms. Cancer Treat Rev. 40:883–891. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Economopoulou P, Kotsakis A, Kapiris I and Kentepozidis N: Cancer therapy and cardiovascular risk: Focus on bevacizumab. Cancer Manag Res. 7:133–143. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Van Cutsem E, Rivera F, Berry S, Kretzschmar A, Michael M, DiBartolomeo M, Mazier MA, Canon JL, Georgoulias V, Peeters M, et al: Safety and efficacy of first-line bevacizumab with FOLFOX, XELOX, FOLFIRI and fluoropyrimidines in metastatic colorectal cancer: The BEAT study. Ann Oncol. 20:1842–1847. 2009. View Article : Google Scholar : PubMed/NCBI

49 

McLellan B, Ciardiello F, Lacouture ME, Segaert S and Van Cutsem E: Regorafenib-associated hand-foot skin reaction: Practical advice on diagnosis, prevention, and management. Ann Oncol. 26:2017–2026. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Elmalik HH, ElAzzazy S, Salem KS and Bujassoum S: A grave outcome of posterior reversible encephalopathy syndrome in a patient receiving avastin (Bevacizumab) for metastatic High-grade serous ovarian cancer. Case Rep Oncol. 8:290–294. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Ayuso-Sacido A, Moliterno JA, Kratovac S, Kapoor GS, O'Rourke DM, Holland EC, García-Verdugo JM, Roy NS and Boockvar JA: Activated EGFR signaling increases proliferation, survival, and migration and blocks neuronal differentiation in post-natal neural stem cells. J Neurooncol. 97:323–337. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Takahashi N, Yamada Y, Furuta K, Honma Y, Iwasa S, Takashima A, Kato K, Hamaguchi T and Shimada Y: Serum levels of hepatocyte growth factor and epiregulin are associated with the prognosis on anti-EGFR antibody treatment in KRAS wild-type metastatic colorectal cancer. Br J Cancer. 110:2716–2727. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Hasegawa Y, Ando M, Maemondo M, Yamamoto S, Isa S, Saka H, Kubo A, Kawaguchi T, Takada M, Rosell R, et al: The role of smoking status on the progression-free survival of non-small cell lung cancer patients harboring activating epidermal growth factor receptor (EGFR) mutations receiving first-line EGFR tyrosine kinase inhibitor versus platinum doublet chemotherapy: A meta-analysis of prospective randomized trials. Oncologist. 20:307–315. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Philip PA and Lutz MP: Targeting epidermal growth factor receptor-related signaling pathways in pancreatic cancer. Pancreas. 44:1046–1052. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Groenestege WM, Thébault S, van der Wijst J, van den Berg D, Janssen R, Tejpar S, van den Heuvel LP, van Cutsem E, Hoenderop JG, Knoers NV and Bindels RJ: Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia. J Clin Invest. 117:2260–2267. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Petrelli F, Borgonovo K, Cabiddu M, Ghilardi M and Barni S: Risk of anti-EGFR monoclonal antibody-related hypomagnesemia: Systematic review and pooled analysis of randomized studies. Expert Opin Drug Saf. 11 Suppl 1:S9–S19. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Maliakal P and Ledford A: Electrolyte and protein imbalance following anti-EGFR therapy in cancer patients: A comparative study. Exp Ther Med. 1:307–311. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Fakih MG, Wilding G and Lombardo J: Cetuximab-induced hypomagnesemia in patients with colorectal cancer. Clin Colorectal Cancer. 6:152–156. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Chen P, Wang L, Li H, Liu B and Zou Z: Incidence and risk of hypomagnesemia in advanced cancer patients treated with cetuximab: A meta-analysis. Oncol Lett. 5:1915–1920. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Cao Y, Liao C, Tan A, Liu L and Gao F: Meta-analysis of incidence and risk of hypomagnesemia with cetuximab for advanced cancer. Chemotherapy. 56:459–465. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Tejpar S, Piessevaux H, Claes K, Piront P, Hoenderop JG, Verslype C and Van Cutsem E: Magnesium wasting associated with epidermal-growth-factor receptor-targeting antibodies in colorectal cancer: A prospective study. Lancet Oncol. 8:387–394. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Streb J, Püsküllüoğlu M, Glanowska I, Ochenduszko S, Konopka K, Łupkowski R, Michalowska-Kaczmarczyk A, Bochenek-Cibor J, Majka M and Krzemieniecki K: Assessment of frequency and severity of hypomagnesemia in patients with metastatic colorectal cancer treated with cetuximab, with a review of the literature. Oncol Lett. 10:3749–3755. 2015. View Article : Google Scholar : PubMed/NCBI

63 

de Baaij JH, Hoenderop JG and Bindels RJ: Magnesium in man: Implications for health and disease. Physiol Rev. 95:1–46. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Cosmai L, Gallieni M and Porta C: Renal toxicity of anticancer agents targeting HER2 and EGFR. J Nephrol. 28:647–657. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Arora N, Gupta A and Singh PP: Biological agents in gastrointestinal cancers: Adverse effects and their management. J Gastrointest Oncol. 8:485–498. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Cao Y, Liu L, Liao C, Tan A and Gao F: Meta-analysis of incidence and risk of hypokalemia with cetuximab-based therapy for advanced cancer. Cancer Chemother Pharmacol. 66:37–42. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Dimke H, Monnens L, Hoenderop JG and Bindels RJ: Evaluation of hypomagnesemia: Lessons from disorders of tubular transport. Am J Kidney Dis. 62:377–383. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Wang Q, Qi Y, Zhang D, Gong C, Yao A, Xiao Y, Yang J, Zhou F and Zhou Y: Electrolyte disorders assessment in solid tumor patients treated with anti-EGFR monoclonal antibodies: A pooled analysis of 25 randomized clinical trials. Tumour Biol. 36:3471–3482. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Geyikoglu F, Emir M, Colak S, Koc K, Turkez H, Bakir M, Hosseinigouzdagani M, Cerig S, Keles ON and Ozek NS: Effect of oleuropein against chemotherapy drug-induced histological changes, oxidative stress, and DNA damages in rat kidney injury. J Food Drug Anal. 25:447–459. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Oh CJ, Ha CM, Choi YK, Park S, Choe MS, Jeoung NH, Huh YH, Kim HJ, Kweon HS, Lee JM, et al: Pyruvate dehydrogenase kinase 4 deficiency attenuates cisplatin-induced acute kidney injury. Kidney Int. 91:880–895. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Abbas A, Mirza MM, Ganti AK and Tendulkar K: Renal toxicities of targeted therapies. Target Oncol. 10:487–499. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Qi WX, Sun YJ, Shen Z and Yao Y: Risk of anti-EGFR monoclonal antibody-related skin rash: An up-to-date meta-analysis of 25 randomized controlled trials. J Chemother. 26:359–368. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Vaubel J, Livingstone E, Schadendorf D and Zimmer L: Retarded low-dose doxycycline for EGFR or MEK inhibitor-induced papulopustular rash. J Eur Acad Dermatol Venereol. 28:1685–1689. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Singh N, Vishwanath G, Aggarwal AN and Behera D: Clinical experience on use of oral EGFR-TKIs as first-line treatment of advanced NSCLC from a tertiary care centre in North India and implications of skin rash. Indian J Chest Dis Allied Sci. 56:149–152. 2014.PubMed/NCBI

75 

Liu HB, Wu Y, Lv TF, Yao YW, Xiao YY, Yuan DM and Song Y: Skin rash could predict the response to EGFR tyrosine kinase inhibitor and the prognosis for patients with non-small cell lung cancer: A systematic review and meta-analysis. PLoS One. 8:e551282013. View Article : Google Scholar : PubMed/NCBI

76 

Jaka A, Gutiérrez-Rivera A, Ormaechea N, Blanco J, La Casta A, Sarasqueta C, Izeta A and Tuneu A: Association between EGFR gene polymorphisms, skin rash and response to anti-EGFR therapy in metastatic colorectal cancer patients. Exp Dermatol. 23:751–753. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Ocvirk J, Heeger S, McCloud P and Hofheinz RD: A review of the treatment options for skin rash induced by EGFR-targeted therapies: Evidence from randomized clinical trials and a meta-analysis. Radiol Oncol. 47:166–175. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Gerber PA, Meller S, Eames T, Buhren BA, Schrumpf H, Hetzer S, Ehmann LM, Budach W, Bölke E, Matuschek C, et al: Management of EGFR-inhibitor associated rash: A retrospective study in 49 patients. Eur J Med Res. 17:42012. View Article : Google Scholar : PubMed/NCBI

79 

Eaby-Sandy B and Lynch K: Side effects of targeted therapies: Rash. Semin Oncol Nurs. 30:147–154. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Stremitzer S, Sebio A, Stintzing S and Lenz HJ: Panitumumab safety for treating colorectal cancer. Expert Opin Drug Saf. 13:843–851. 2014.PubMed/NCBI

81 

Lv ZC, Ning JY and Chen HB: Efficacy and toxicity of adding cetuximab to chemotherapy in the treatment of metastatic colorectal cancer: A meta-analysis from 12 randomized controlled trials. Tumour Biol. 35:11741–11750. 2014. View Article : Google Scholar : PubMed/NCBI

82 

André T and Dumont SN: Regorafenib approved in metastatic colorectal cancer. Bull Cancer. 100:1027–1029. 2013.(In French). PubMed/NCBI

83 

Saif MW, Syrigos KI, Hotchkiss S, Shanley J, Grasso J, Ferencz TM, Syrigos K and Shah MM: Successful desensitization with cetuximab after an infusion reaction to panitumumab in patients with metastatic colorectal cancer. Cancer Chemother Pharmacol. 65:107–112. 2009. View Article : Google Scholar : PubMed/NCBI

84 

Garcia-Foncillas J and Diaz-Rubio E: Progress in metastatic colorectal cancer: Growing role of cetuximab to optimize clinical outcome. Clin Transl Oncol. 12:533–542. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Emmanouilides C, Sfakiotaki G, Androulakis N, Kalbakis K, Christophylakis C, Kalykaki A, Vamvakas L, Kotsakis A, Agelaki S, Diamandidou E, et al: Front-line bevacizumab in combination with oxaliplatin, leucovorin and 5-fluorouracil (FOLFOX) in patients with metastatic colorectal cancer: A multicenter phase II study. BMC Cancer. 7:912007. View Article : Google Scholar : PubMed/NCBI

86 

Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al: 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. 28:4697–4705. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Saltz LB, Clarke S, Diaz-Rubio E, Scheithauer W, Figer A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F, et al: Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: A randomized phase III study. J Clin Oncol. 26:2013–2019. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 16 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang B, Fang C, Deng D and Xia L: Research progress on common adverse events caused by targeted therapy for colorectal cancer (Review). Oncol Lett 16: 27-33, 2018
APA
Zhang, B., Fang, C., Deng, D., & Xia, L. (2018). Research progress on common adverse events caused by targeted therapy for colorectal cancer (Review). Oncology Letters, 16, 27-33. https://doi.org/10.3892/ol.2018.8651
MLA
Zhang, B., Fang, C., Deng, D., Xia, L."Research progress on common adverse events caused by targeted therapy for colorectal cancer (Review)". Oncology Letters 16.1 (2018): 27-33.
Chicago
Zhang, B., Fang, C., Deng, D., Xia, L."Research progress on common adverse events caused by targeted therapy for colorectal cancer (Review)". Oncology Letters 16, no. 1 (2018): 27-33. https://doi.org/10.3892/ol.2018.8651