Next Article in Journal
GRP94 Inhabits the Immortalized Porcine Hepatic Stellate Cells Apoptosis under Endoplasmic Reticulum Stress through Modulating the Expression of IGF-1 and Ubiquitin
Next Article in Special Issue
Strategies to Overcome Resistance to Immune-Based Therapies in Osteosarcoma
Previous Article in Journal
Whole Genome Analysis of SLs Pathway Genes and Functional Characterization of DlSMXL6 in Longan Early Somatic Embryo Development
Previous Article in Special Issue
Potential Role of Selenium in the Treatment of Cancer and Viral Infections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Curcumin: A Novel Way to Improve Quality of Life for Colorectal Cancer Patients?

by
Laura Layos
1,2,3,
Eva Martínez-Balibrea
3,4 and
Vicenç Ruiz de Porras
2,3,*
1
Medical Oncology Department, Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
2
Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
3
Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les escoles s/n, 08916 Badalona, Spain
4
ProCURE Program, Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les escoles s/n, 08916 Badalona, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(22), 14058; https://doi.org/10.3390/ijms232214058
Submission received: 17 October 2022 / Revised: 7 November 2022 / Accepted: 11 November 2022 / Published: 14 November 2022

Abstract

:
Colorectal cancer (CRC) is the third most common cancer in men and the second most common in women. Treatment of metastatic CRC consists of highly toxic chemotherapeutic drug combinations that often negatively affect patient quality of life (QoL). Moreover, chemotherapy-induced toxicity and chemotherapy resistance are among the most important factors limiting cancer treatment and can lead to the interruption or discontinuation of potentially effective therapy. Several preclinical studies have demonstrated that curcumin acts through multiple cellular pathways and possesses both anti-cancer properties against CRC and the capacity to mitigate chemotherapy-related side effects and overcome drug resistance. In this review article, we suggest that the addition of curcumin to the standard chemotherapeutic treatment for metastatic CRC could reduce associated side-effects and overcome chemotherapy resistance, thereby improving patient QoL.

1. First-Line Treatment of Metastatic Colorectal Cancer: An Overview

Colorectal cancer (CRC) is the third most common tumor and the second leading cause of cancer death worldwide [1,2]. Metastases, the greatest cause of cancer-related mortality, are present in nearly 25% of newly diagnosed CRC patients, up to 40% of whom will relapse during follow-up after curative primary tumor surgery. Over the last ten years, new combinations of cytotoxic agents and targeted therapies have improved the prognosis of metastatic CRC (mCRC), and median overall survival is now 30 months, highlighting the importance of a “continuum of care” approach in advanced disease [3]. Approximately one-third of mCRC patients have limited-liver metastatic disease and could be candidates for surgery with curative intent after systemic treatment [4].
The backbone of first-line treatment of mCRC consists of doublets or triplets of fluoropyrimidines (thymidylate synthase (TS) inhibitors) such as 5-fluorouracil or capecitabine in combination with oxaliplatin (a platinum DNA damage agent) and/or irinotecan (a topoisomerase I inhibitor). The addition of targeted therapies such as anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (cetuximab or panitumumab) or antiangiogenic therapy (bevacizumab or aflibercept) in first or later lines improves both overall survival and response rates. The clinical benefit of EGFR inhibitors is limited to RAS-wild-type tumors (KRAS exons 3-4 and NRAS exons 2-3-4), which are present in nearly 50% of newly diagnosed patients, and the response seems to be superior in left-side primary tumors [5,6,7,8,9,10]. In addition, 4–5% of mCRC patients harbor BRAF mutations (most frequently V600E), in which the combination of specific inhibitors, such as encorafenib, with anti-EGFR therapy has been shown to increase overall survival [11]. Other targeted therapies such as regorafenib (a multitargeted kinase-inhibitor) and ramucirumab (angiogenesis inhibitor) have also shown good results in chemorefractory mCRC [12,13]. In addition to agents blocking pathways involved in tumor growth and spread, accumulating evidence has shown that targeting pathways involved in immunomodulation is also effective. Immune checkpoint inhibitors stop the tumor from escaping T cell detection and thus stimulate immune surveillance and clearance [14,15]. However, the efficacy of immune checkpoint inhibitors is basically limited to tumors with deficient Mismatch Repair (dMMR) and a high mutational burden, which comprise only 4–8% of all mCRC cases [16,17]. The programmed death-ligand 1 (PD-1) blocker pembrolizumab has indeed shown promising results as a monotherapy in this subgroup of patients. The KEYNOTE-177 study comparing pembrolizumab with chemotherapy in dMMR patients reported a progression-free survival of 16.5 months with pembrolizumab compared to 8.2 months for the chemotherapy group. Moreover, the duration of response at 24 months was 83% compared to 35%, respectively [18]. Equally encouraging results have been achieved with another PD-1 inhibitor nivolumab and the cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitor ipilimumab [19].

2. Chemotherapy-Associated Toxicity in mCRC

The most common first-line treatment regimens in unresectable mCRC patients are FOLFOX (folinic acid + 5-fluorouracil + oxaliplatin) and FOLFIRI (folinic acid + 5-fluorouracil + irinotecan) in combination with either an anti-EGFR antibody (in RAS-wild-type tumors) or with bevacizumab. Unfortunately, all these combinations are extremely toxic and can compromise patient quality of life (QoL), which is increasingly being recognized as both a crucial outcome in clinical practice and an endpoint in randomized clinical trials [20,21]. In fact, chemotherapy-induced toxicity is one of the most influential factors limiting cancer treatment and is often associated with the interruption or even discontinuation of potentially effective anti-cancer therapy.
Approximately 30% of patients treated with fluoropyrimidines develop severe toxicities (≥Grade III Common Terminology Criteria for Adverse Events), including myelosuppression, severe diarrhea, vomiting, stomatitis (inflammation of the mucus lining in the mouth), mucositis, hand-foot syndrome (palmar-plantar erythrodysesthesia), and neuropathy. Moreover, fluoropyrimidine-related toxicity leads to death in 0.5–1% of patients [22,23,24,25]. Oxaliplatin also has severe side effects, such as gastrointestinal upset (nausea and vomiting, diarrhea, and mucositis), hematological disorders (anemia, thrombocytopenia, and neutropenia), peripheral neuropathy, and hepatotoxicity [26]. In fact, neuropathy, the major problem associated with oxaliplatin treatment, occurs in up to 70% of patients and leads to dose limitation and treatment discontinuation [27,28]. It has been suggested that the development of neurotoxicity, hepatotoxicity, and nephrotoxicity is at least partly due to oxaliplatin-induced oxidative damage to mitochondria and to the inhibition of sodium pumps by the chelating action of oxalate on calcium and magnesium molecules [29,30]. Management of these adverse effects is based on the administration of calcium gluconate and/or magnesium sulphate [31]. Irinotecan is also often associated with severe toxicities, especially neutropenia and diarrhea, generally caused by the insufficient glucuronidation of the irinotecan active metabolite SN-38 by the UDP-glucuronosyltransferase (UGT) 1A1 (UGT1A) enzyme. The resulting elevated SN-38 plasma concentration is responsible for the often life-threatening hematological and gastrointestinal toxicities associated with irinotecan [32]. Finally, targeted therapies such as bevacizumab have been associated with thromboembolic events and the occurrence of grade 3/4 hypertension and bleeding in 2% to 4% of patients [33]. Fortunately, the addition of bevacizumab to FOLFOX seems to be well tolerated and does not markedly change the overall chemotherapy-related toxicity [23].

3. Curcumin Attenuates Chemotherapy-Related Toxicity

For many years, curcumin (diferuloylmethane)—the “golden spice”—has been widely studied because of its pleiotropic effects in cancer. Curcumin, a hydrophobic polyphenol, is derived from the rhizome of the herb Curcuma longa and constitutes the major curcuminoid in the spice turmeric (77% curcumin, 17% demethoxycurcumin, 3% bis-demethoxycurcumin). Curcumin is “generally recognized as safe” (GRAS) as a dietary supplement by the U.S Food and Drug Administration (FDA) and the European Food Safety Authority (EFSA) and has been catalogued with the E100 code of the European Union. One of the clinical benefits of curcumin is the improvement of QoL in several health conditions [34], including cancer [35,36].
Curcumin is a pleiotropic agent that acts through multiple cellular pathways and has been shown to possess anti-cancer properties against CRC in vitro and in vivo [37,38]. Many of its anti-cancer properties have been attributed to its role as an anti-inflammatory and antioxidant, as well as to its ability to modulate the cell cycle and the pathways involved in proliferation, apoptosis, migration, invasion, angiogenesis, and metastasis [39], which are typically targeted by the drugs used to treat CRC. Mechanistically, curcumin modulates several CRC molecular targets at the same time—either by altering their gene expression, activation, or signaling pathways, or by direct interaction [37,38,39]. Importantly, in addition to its well-known anti-cancer properties, curcumin can also alleviate some of the chemotherapy-related side effects [40]. For example, curcumin attenuates the liver injury induced by oxaliplatin through activation of the nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling, a key regulator pathway of cellular defense against oxidative and electrophilic stresses [41], as well as the nerve damage and the oxidative damage to mitochondria caused by oxaliplatin [42]. In fact, curcumin has been shown to not only hinder mitochondrial damage but also to protect mitochondria and induce activity of mitochondrial complex enzymes [36,42,43]. Interestingly, similar effects of curcumin on cisplatin-related toxicity have been observed in several tumor types [44,45,46,47,48]. Additionally, curcumin protects against irinotecan-induced intestinal injury by inhibiting nuclear factor kappa B (NF-κB) transcription factor activation [49], and it is also active against FOLFIRI-related cardiovascular toxicity [50] and capecitabine-induced hand-foot syndrome [51]. Recently, it has been shown that curcumin attenuates bevacizumab-associated cardiotoxicity by suppressing oxidative stress and preventing mitochondrial dysfunction in heart mitochondria [52].
In a study of curcumin’s effects in cancer patients, Belcaro and colleagues looked at the side effects of chemotherapy in several tumor types, including colon, ovarian, lung, liver, kidney, and stomach cancers. Of 80 patients treated with chemotherapy, 40 simultaneously received 500 mg of curcumin. Chemotherapy-related nausea, diarrhea, constipation, weight loss, neutropenia, and cardiotoxicity were significantly lower in the patients receiving curcumin than in the control group. Moreover, patients receiving curcumin also required fewer medications for treating these side effects [53]. In the same vein, turmeric supplementation for 21 days resulted in a clinically relevant and statistically significant improvement in global health status, symptom scores (fatigue, nausea, vomiting, pain, appetite loss, insomnia), and hematological parameters of breast cancer patients treated with paclitaxel [54]. Taken together, these findings lead us to suggest that the addition of curcumin to the standard treatment of CRC could not only attenuate chemotherapy-associated side effects but also improve the QoL of patients (Figure 1).

4. Curcumin Reverts Chemotherapy Resistance in mCRC

In addition to chemotherapy-related toxicity, chemoresistance remains one of the main problems hindering treatment success. Tumor cells can be intrinsically resistant or acquire resistance during a treatment. Resistance to chemotherapy is a complex and multifactorial process involving several mechanisms, including drug influx/efflux modifications, alterations in DNA damage repair (DDR), decreased cell death activation, autocrine survival signaling, and high detoxification activity [55,56]. One of these mechanisms with consequences in mCRC is the hyperactivation of the NF-κB signaling pathway [57], which promotes the expression of several target genes involved in inflammation, cell proliferation, apoptosis, angiogenesis, invasion, metastasis, and chemoresistance [58,59]. In fact, most of the anti-inflammatory and anti-cancer properties of curcumin are believed to be due to its ability to inhibit NF-κB activation through interaction with the IκB kinase complex (IKK) by inhibiting the phosphorylation and degradation of IκBα, a NF-κB inhibitor, and thereby blocking the nuclear translocation of this transcription factor [37,60,61]. Along with other studies [62,63,64], our group has demonstrated that curcumin can overcome oxaliplatin resistance by inhibiting the activity of the CXC-chemokines/NF-κB axis and, consequently, the expression of genes involved in anti-apoptosis and proliferation [57]. Additionally, in CRC preclinical models, curcumin was shown to enhance the effect of 5-fluorouracil [65,66] and capecitabine [67] by inhibiting AKT and NF-κB activity, and consequently, NF-κB-regulated gene products. In the same vein, Pattel and colleagues reported that curcumin sensitizes CRC cells to FOLFOX by inhibiting EGFR family receptors and insulin-like growth factor-1 receptor (IGF-1R) [68,69,70], the overexpression of which has been related to chemoresistance in CRC [71,72].
Chemotherapy resistance is also related to the specific mechanism of action of the drug. An example of such a specific mechanism is gene amplification in TS in 5-fluorouracil treated patients [73] and upregulation of genes involved in DDR pathways, such as ERCC1 in oxaliplatin treated patients [74]. Interestingly, Rajitha and colleagues demonstrated that the inhibition of NF-κB translocation by curcumin or its analogs induces cell cycle arrest and downregulates TS in CRC cells [61]. Furthermore, curcumin was found to inhibit ERCC1 through its ability to modulate miR-409-3p, thereby overcoming oxaliplatin resistance in CRC cells [75].
Curcumin can also promote the activation of apoptotic pathways by increasing the generation of reactive oxygen species (ROS) [76]. In a recent work, Li and colleagues demonstrated that curcumin can reverse Nicotinamide N-methyltransferase-induced cell proliferation and 5-fluorouracil resistance through ROS generation and cell cycle arrest [77].
On the other hand, the drug-resistant phenotype is associated with the acquisition of mesenchymal features, and epithelial-to-mesenchymal transition (EMT) plays a key role in chemoresistance in CRC, mainly through the activation of the NF-κB and transforming growth factor β (TGF-β) pathways [78,79,80]. In fact, EMT was observed in chemotherapy-resistant CRC cell lines [57,81,82], while curcumin was able to revert this chemoresistance by downregulating EMT markers [83] through TGF-β/Smad2/3 signaling attenuation [84], by upregulating EMT suppressive miRNAs [85] or by downregulating the TET1-NKD2-WNT signaling pathway [86]. In addition, several studies have demonstrated that curcumin can sensitize colon cancer stem cells (CSC), a small subpopulation of cells within tumors capable of self-renewal, differentiation, and tumorigenicity [87], to 5-fluorouracil, FOLFOX and irinotecan, thereby preventing the emergence of chemoresistant CRC cells [70,88,89,90,91]. In this regard, a recent study has demonstrated that treatment of CRC organoids with a combination of amorphous curcumin (a compound with improved solubility and bioavailability) and oxaliplatin, 5-fluoroouracil, or irinotecan showed a synergistic activity through the inhibition of proliferation-related signals and CSC marker expression, in addition to arresting the ERK signaling pathway [92]. Along the same lines, Zheng and colleagues showed that low doses of curcumin promoted the sensitivity of CRC cells to 5-fluorouracil by downregulating phospho-ERK signaling [93].
Finally, several studies have shown that curcumin can increase the intracellular accumulation of oxaliplatin and 5-fluorouracil in CRC cells by downregulating the P-gp [75,94] and ATP-binding cassette transporter G2 (ABCG2) [70] drug-efflux transporters both at the mRNA and protein levels. Preclinical data have suggested that the expression of ATP-binding cassette (ABC) transporters, such as ABCC2 [95], ABCB4 [96], as well as the multidrug resistance protein 1 (MDR1, also known as P-glycoprotein or P-gp), which is encoded by ABCB1 [97,98], can confer resistance to chemotherapy. However, evidence that these transporters contribute to drug resistance in human tumors is sorely lacking [99] and the development of MDR1 as a therapeutic target has been unsuccessful [100]. It is important to highlight that although several studies have related the ABC transporters’ overexpression to platinum resistance [55,101,102], the association between oxaliplatin resistance and the MDR1 expression has shown unconvincing results. For instance, Ekblad and colleagues described an overexpression of this membrane transporter as a consequence of oxaliplatin resistance acquisition in vitro, although functional tests did not show any increase in ABCB1 transport activity in the oxaliplatin-resistant models compared with its parental cell lines [103]. Other studies have reported no association between these drug efflux pumps and the sensitivity to oxaliplatin in CRC clinical samples [104]. In the same vein, the ability of MDR1 to confer resistance to 5-fluorouracil and irinotecan has been demonstrated in different CRC cell lines transfected with this carrier. However, its clinical relevance in CRC refractoriness to antitumor chemotherapy remains to be established [105,106]. Taken together, these results highlight the necessity of further investigation into the role of MDR1 and curcumin in oxaliplatin and 5-fluorouracil resistance in CRC patients.
Most clinical data on curcumin come from early phase clinical trials, with results showing that oral curcumin can achieve efficacious levels in the colon with negligible distribution outside the gut [107,108]. Moreover, curcumin was shown to be safe in advanced CRC patients when administered for up to four months [109]. In addition, a study by James and colleagues found that curcumin at doses up to 2 gms daily was highly tolerable when added to a FOLFOX regimen in mCRC patients with liver metastases [110]. More recently, the same group performed a phase IIa randomized trial of first-line treatment for mCRC patients comparing FOLFOX +/−bevacizumab with the same regimen plus curcumin 2 gms/day (CUFOX) in mCRC patients. In the intention-to-treat population, patients in the CUFOX arm achieved longer overall survival (HR 0.34; p = 0.02) but there was no difference in progression-free survival (HR 0.57) [111].
In conclusion, a further improvement in outcomes for mCRC highly depends on identifying and targeting mechanisms of drug resistance. Taken together, these findings offer compelling evidence that combining curcumin with conventional chemotherapy may be effective in overcoming drug resistance in mCRC (Table 1).

5. Conclusions

Treatment for mCRC consists of highly toxic drug combinations that often negatively affect the QoL of patients. We believe that patient QoL must be recognized as an essential outcome in clinical practice; moreover, it is increasingly being reported as an endpoint in randomized clinical trials. Optimizing strategies to control chemotherapy-related toxicity will not only improve patient QoL but will also improve adherence to cancer treatment and thus improve patient survival. This is especially crucial for mCRC patients, in whom chemotherapy is prescribed as a neoadjuvant treatment before surgery for liver metastases, where it is critical to maintain an adequate dose intensity in order to proceed to curative surgical treatment. Furthermore, our own experience has taught us that the unprescribed use of several plant-derived supplements is very common among cancer patients even when their positive effect on QoL has not been demonstrated in clinical studies. One of the most commonly used herbal supplements is curcumin, which has been extensively studied in cancer prevention and treatment. In fact, a plethora of preclinical studies have demonstrated the anti-cancer properties of curcumin as well as its role as a chemosensitizer agent [37,38,39]. Several preclinical studies have demonstrated that the addition of curcumin to the standard treatment of CRC could decrease treatment-associated side effects and enhance chemotherapy efficacy [40,53]. Therefore, considering that therapy-induced toxicity is among the most important factors limiting cancer treatment and is usually associated with discontinuation of potentially effective therapy, we suggest that adding curcumin, a natural compound with a very low toxicity profile in humans [39], to current mCRC treatment regimens could be a potential synergistic strategy to reduce chemotherapy-related adverse effects, improve treatment efficacy, and decrease drug resistance.
Additionally, it is important to identify predictive biomarkers of response to curcumin-based treatment. To the best of our knowledge, only a few studies have focused on this question. However, in a previous study by our group, we found that treatment with oxaliplatin induces the expression of the CXCL1 chemokine that was repressed by the addition of curcumin—both in CRC cell lines and in patient-derived CRC liver metastasis explants treated with oxaliplatin or oxaliplatin + curcumin. Interestingly, the explants with the “best response” to oxaliplatin + curcumin were those with the highest baseline levels of CXCL1, suggesting that this chemokine could be a good predictive marker for this treatment [57]. Prompted by these observations, Howells’ group conducted a phase IIa trial in which they assessed CXCL1 plasma levels in patients receiving FOLFOX or CUFOX. Although there was no significant difference in plasma CXCL1 concentrations after curcumin treatment, mean baseline concentrations were 1.7-fold higher in FOLFOX patients than in CUFOX patients [111]. In the same vein, Lu and collaborators recently demonstrated that CRC patients with microsatellite-stable tumors and high baseline IκBα protein expression would benefit from curcumin treatment [112].
Finally, improved delivery strategies and new curcumin formulations (such as nanoparticles, liposomes, and synthetic analogues) will increase the absorption and bioavailability of curcumin [113].
Certainly, further research is warranted on the potential role of curcumin in reducing chemotherapy-induced toxicity and on predictive biomarkers to identify those patients most likely to benefit. Unfortunately, the few clinical trials of curcumin performed to date have often been limited by wide patient heterogeneity, small sample size, and the poor bioavailability of the curcumin formulations studied. For this reason, we strongly recommend that randomized, double-blind, placebo-controlled trials of bioavailable curcumin be carried out. The results of such trials will further elucidate the role of this polyphenol in overcoming chemoresistance and improving the QoL of mCRC patients.

Author Contributions

Conceptualization, V.R.d.P.; literature review, L.L. and V.R.d.P.; writing—original draft, L.L. and V.R.d.P.; writing—review and editing, L.L., E.M.-B. and V.R.d.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge Renée Grupp for critical reading and language correction.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Keum, N.; Giovannucci, E. Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 713–732. [Google Scholar] [CrossRef] [PubMed]
  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. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Venook, A.P.; Niedzwiecki, D.; Lenz, H.J.; Innocenti, F.; Fruth, B.; Meyerhardt, J.A.; Schrag, D.; Greene, C.; O’Neil, B.H.; Atkins, J.N.; et al. Effect of First-Line Chemotherapy Combined with Cetuximab or Bevacizumab on Overall Survival in Patients with KRAS Wild-Type Advanced or Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 2017, 317, 2392–2401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. De Falco, V.; Napolitano, S.; Rosello, S.; Huerta, M.; Cervantes, A.; Ciardiello, F.; Troiani, T. How we treat metastatic colorectal cancer. ESMO Open 2020, 4 (Suppl. 2), e000813. [Google Scholar] [CrossRef] [PubMed]
  5. Bokemeyer, C.; Bondarenko, I.; Hartmann, J.T.; de Braud, F.; Schuch, G.; Zubel, A.; Celik, I.; Schlichting, M.; Koralewski, P. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: The OPUS study. Ann. Oncol. 2011, 22, 1535–1546. [Google Scholar] [CrossRef] [PubMed]
  6. Bokemeyer, C.; Kohne, C.H.; Ciardiello, F.; Lenz, H.J.; Heinemann, V.; Klinkhardt, U.; Beier, F.; Duecker, K.; van Krieken, J.H.; Tejpar, S. FOLFOX4 plus cetuximab treatment and RAS mutations in colorectal cancer. Eur. J. Cancer 2015, 51, 1243–1252. [Google Scholar] [CrossRef]
  7. Douillard, J.Y.; Oliner, K.S.; 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. 2013, 369, 1023–1034. [Google Scholar] [CrossRef] [Green Version]
  8. Douillard, J.Y.; Siena, S.; Cassidy, J.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.; et al. Final results from PRIME: Randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer. Ann. Oncol. 2014, 25, 1346–1355. [Google Scholar] [CrossRef]
  9. Van Cutsem, E.; Lenz, H.J.; Kohne, C.H.; Heinemann, V.; Tejpar, S.; Melezinek, I.; Beier, F.; Stroh, C.; Rougier, P.; van Krieken, J.H.; et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J. Clin. Oncol. 2015, 33, 692–700. [Google Scholar] [CrossRef] [Green Version]
  10. Arnold, D.; Lueza, B.; Douillard, J.Y.; Peeters, M.; Lenz, H.J.; Venook, A.; Heinemann, V.; Van Cutsem, E.; Pignon, J.P.; Tabernero, J.; et al. Prognostic and predictive value of primary tumour side in patients with RAS wild-type metastatic colorectal cancer treated with chemotherapy and EGFR directed antibodies in six randomized trials. Ann. Oncol. 2017, 28, 1713–1729. [Google Scholar] [CrossRef]
  11. Kopetz, S.; Grothey, A.; Yaeger, R.; Van Cutsem, E.; Desai, J.; Yoshino, T.; Wasan, H.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. N. Engl. J. Med. 2019, 381, 1632–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Adenis, A.; de la Fouchardiere, C.; Paule, B.; Burtin, P.; Tougeron, D.; Wallet, J.; Dourthe, L.M.; Etienne, P.L.; Mineur, L.; Clisant, S.; et al. Survival, safety, and prognostic factors for outcome with Regorafenib in patients with metastatic colorectal cancer refractory to standard therapies: Results from a multicenter study (REBECCA) nested within a compassionate use program. BMC Cancer 2016, 16, 412. [Google Scholar]
  13. 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.; et al. 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. [Google Scholar] [PubMed]
  14. Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Comprehensive review of targeted therapy for colorectal cancer. Signal. Transduct. Target. 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Seidel, J.A.; Otsuka, A.; Kabashima, K. Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Front. Oncol. 2018, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  16. Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit with Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef]
  17. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [Green Version]
  18. Diaz, L.A., Jr.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): Final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022, 23, 659–670. [Google Scholar] [CrossRef]
  19. Cohen, R.; Bennouna, J.; Meurisse, A.; Tournigand, C.; De La Fouchardiere, C.; Tougeron, D.; Borg, C.; Mazard, T.; Chibaudel, B.; Garcia-Larnicol, M.L.; et al. RECIST and iRECIST criteria for the evaluation of nivolumab plus ipilimumab in patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: The GERCOR NIPICOL phase II study. J. Immunother. Cancer 2020, 8, e001499. [Google Scholar] [CrossRef]
  20. Schuurhuizen, C.; Braamse, A.M.J.; Konings, I.; Sprangers, M.A.G.; Ket, J.C.F.; Dekker, J.; Verheul, H.M.W. Does severe toxicity affect global quality of life in patients with metastatic colorectal cancer during palliative systemic treatment? A systematic review. Ann. Oncol. 2017, 28, 478–486. [Google Scholar] [CrossRef]
  21. Schandelmaier, S.; Conen, K.; von Elm, E.; You, J.J.; Blümle, A.; Tomonaga, Y.; Saccilotto, R.; Amstutz, A.; Bengough, T.; Meerpohl, J.J.; et al. Planning and reporting of quality-of-life outcomes in cancer trials. Ann. Oncol. 2015, 26, 1966–1973. [Google Scholar] [CrossRef] [PubMed]
  22. Hoff, P.M.; Ansari, R.; Batist, G.; Cox, J.; Kocha, W.; Kuperminc, M.; Maroun, J.; Walde, D.; Weaver, C.; Harrison, E.; et al. Comparison of oral capecitabine versus intravenous fluorouracil plus leucovorin as first-line treatment in 605 patients with metastatic colorectal cancer: Results of a randomized phase III study. J. Clin. Oncol. 2001, 19, 2282–2292. [Google Scholar] [CrossRef] [PubMed]
  23. Hochster, H.S.; Hart, L.L.; Ramanathan, R.K.; Childs, B.H.; Hainsworth, J.D.; Cohn, A.L.; Wong, L.; Fehrenbacher, L.; Abubakr, Y.; Saif, M.W.; et al. Safety and efficacy of oxaliplatin and fluoropyrimidine regimens with or without bevacizumab as first-line treatment of metastatic colorectal cancer: Results of the TREE Study. J. Clin. Oncol. 2008, 26, 3523–3529. [Google Scholar] [CrossRef] [PubMed]
  24. Van Cutsem, E.; Twelves, C.; Cassidy, J.; Allman, D.; Bajetta, E.; Boyer, M.; Bugat, R.; Findlay, M.; Frings, S.; Jahn, M.; et al. Xeloda Colorectal Cancer Study, G., Oral capecitabine compared with intravenous fluorouracil plus leucovorin in patients with metastatic colorectal cancer: Results of a large phase III study. J. Clin. Oncol. 2001, 19, 4097–4106. [Google Scholar] [CrossRef]
  25. Vodenkova, S.; Buchler, T.; Cervena, K.; Veskrnova, V.; Vodicka, P.; Vymetalkova, V. 5-fluorouracil and other fluoropyrimidines in colorectal cancer: Past, present and future. Pharm. Ther. 2020, 206, 107447. [Google Scholar] [CrossRef]
  26. Andre, T.; Boni, C.; Mounedji-Boudiaf, L.; Navarro, M.; Tabernero, J.; Hickish, T.; Topham, C.; Zaninelli, M.; Clingan, P.; Bridgewater, J.; et al. Multicenter International Study of Oxaliplatin/5-Fluorouracil/Leucovorin in the Adjuvant Treatment of Colon Cancer, I., Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N. Engl. J. Med. 2004, 350, 2343–2351. [Google Scholar] [CrossRef] [Green Version]
  27. Beijers, A.J.; Mols, F.; Vreugdenhil, G. A systematic review on chronic oxaliplatin-induced peripheral neuropathy and the relation with oxaliplatin administration. Support Care Cancer 2014, 22, 1999–2007. [Google Scholar] [CrossRef] [Green Version]
  28. Cersosimo, R.J. Oxaliplatin-associated neuropathy: A review. Ann. Pharm. 2005, 39, 128–135. [Google Scholar] [CrossRef]
  29. Zheng, H.; Xiao, W.H.; Bennett, G.J. Functional deficits in peripheral nerve mitochondria in rats with paclitaxel- and oxaliplatin-evoked painful peripheral neuropathy. Exp. Neurol. 2011, 232, 154–161. [Google Scholar] [CrossRef] [Green Version]
  30. Ali, B.H.; Al Moundhri, M.S. Agents ameliorating or augmenting the nephrotoxicity of cisplatin and other platinum compounds: A review of some recent research. Food Chem. Toxicol. 2006, 44, 1173–1183. [Google Scholar] [CrossRef]
  31. Chay, W.Y.; Tan, S.H.; Lo, Y.L.; Ong, S.Y.; Ng, H.C.; Gao, F.; Koo, W.H.; Choo, S.P. Use of calcium and magnesium infusions in prevention of oxaliplatin induced sensory neuropathy. Asia Pac. J. Clin. Oncol. 2010, 6, 270–277. [Google Scholar] [CrossRef] [PubMed]
  32. Fujita, K.; Sparreboom, A. Pharmacogenetics of irinotecan disposition and toxicity: A review. Curr. Clin. Pharm. 2010, 5, 209–217. [Google Scholar] [CrossRef] [PubMed]
  33. Saltz, L.B.; Clarke, S.; Diaz-Rubio, E.; Scheithauer, W.; Figer, A.; Wong, R.; Koski, S.; Lichinitser, M.; Yang, T.S.; 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. 2008, 26, 2013–2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sadeghian, M.; Rahmani, S.; Jamialahmadi, T.; Johnston, T.P.; Sahebkar, A. The effect of oral curcumin supplementation on health-related quality of life: A systematic review and meta-analysis of randomized controlled trials. J. Affect. Disord 2021, 278, 627–636. [Google Scholar] [CrossRef] [PubMed]
  35. Panahi, Y.; Saadat, A.; Beiraghdar, F.; Sahebkar, A. Adjuvant therapy with bioavailability-boosted curcuminoids suppresses systemic inflammation and improves quality of life in patients with solid tumors: A randomized double-blind placebo-controlled trial. Phytother. Res. 2014, 28, 1461–1467. [Google Scholar] [CrossRef] [PubMed]
  36. Waseem, M.; Parvez, S.; Tabassum, H. Mitochondria as the Target for the Modulatory Effect of Curcumin in Oxaliplatin-induced Toxicity in Isolated Rat Liver Mitochondria. Arch. Med. Res. 2017, 48, 55–63. [Google Scholar] [CrossRef] [PubMed]
  37. Ruiz de Porras, V.; Layos, L.; Martinez-Balibrea, E. Curcumin: A therapeutic strategy for colorectal cancer? Semin. Cancer Biol. 2021, 73, 321–330. [Google Scholar] [CrossRef]
  38. Weng, W.; Goel, A. Curcumin and colorectal cancer: An update and current perspective on this natural medicine. Semin. Cancer Biol. 2022, 80, 73–86. [Google Scholar] [CrossRef]
  39. Giordano, A.; Tommonaro, G. Curcumin and Cancer. Nutrients 2019, 11, 2376. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, Z.; Huang, P.; Law, S.; Tian, H.; Leung, W.; Xu, C. Preventive Effect of Curcumin Against Chemotherapy-Induced Side-Effects. Front. Pharm. 2018, 9, 1374. [Google Scholar] [CrossRef] [Green Version]
  41. Lu, Y.; Wu, S.; Xiang, B.; Li, L.; Lin, Y. Curcumin Attenuates Oxaliplatin-Induced Liver Injury and Oxidative Stress by Activating the Nrf2 Pathway. Drug Des. Devel. 2020, 14, 73–85. [Google Scholar] [CrossRef] [PubMed]
  42. Al Moundhri, M.S.; Al-Salam, S.; Al Mahrouqee, A.; Beegam, S.; Ali, B.H. The effect of curcumin on oxaliplatin and cisplatin neurotoxicity in rats: Some behavioral, biochemical, and histopathological studies. J. Med. Toxicol. 2013, 9, 25–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Waseem, M.; Parvez, S. Neuroprotective activities of curcumin and quercetin with potential relevance to mitochondrial dysfunction induced by oxaliplatin. Protoplasma 2016, 253, 417–430. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, C.Y.; Zhang, Q.Y.; Zheng, G.J.; Feng, B. Phytochemicals: Current strategy to sensitize cancer cells to cisplatin. Biomed. Pharm. 2019, 110, 518–527. [Google Scholar] [CrossRef]
  45. Abadi, A.J.; Mirzaei, S.; Mahabady, M.K.; Hashemi, F.; Zabolian, A.; Hashemi, F.; Raee, P.; Aghamiri, S.; Ashrafizadeh, M.; Aref, A.R.; et al. Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytother. Res. 2022, 36, 189–213. [Google Scholar] [CrossRef] [PubMed]
  46. Hussain, Y.; Islam, L.; Khan, H.; Filosa, R.; Aschner, M.; Javed, S. Curcumin-cisplatin chemotherapy: A novel strategy in promoting chemotherapy efficacy and reducing side effects. Phytother. Res. 2021, 35, 6514–6529. [Google Scholar] [CrossRef] [PubMed]
  47. Fetoni, A.R.; Eramo, S.L.; Paciello, F.; Rolesi, R.; Podda, M.V.; Troiani, D.; Paludetti, G. Curcuma longa (curcumin) decreases in vivo cisplatin-induced ototoxicity through heme oxygenase-1 induction. Otol. Neurotol. 2014, 35, e169-77. [Google Scholar] [CrossRef]
  48. Mendonca, L.M.; da Silva Machado, C.; Teixeira, C.C.; de Freitas, L.A.; Bianchi Mde, L.; Antunes, L.M. Curcumin reduces cisplatin-induced neurotoxicity in NGF-differentiated PC12 cells. Neurotoxicology 2013, 34, 205–211. [Google Scholar] [CrossRef] [Green Version]
  49. Ouyang, M.; Luo, Z.; Zhang, W.; Zhu, D.; Lu, Y.; Wu, J.; Yao, X. Protective effect of curcumin against irinotecaninduced intestinal mucosal injury via attenuation of NFkappaB activation, oxidative stress and endoplasmic reticulum stress. Int. J. Oncol. 2019, 54, 1376–1386. [Google Scholar]
  50. Guclu, O.; Doganlar, O.; Yuksel, V.; Doganlar, Z.B. FOLFIRI-Mediated Toxicity in Human Aortic Smooth Muscle Cells and Possible Amelioration with Curcumin and Quercetin. Cardiovasc. Toxicol. 2020, 20, 139–154. [Google Scholar] [CrossRef]
  51. Scontre, V.A.; Martins, J.C.; de Melo Sette, C.V.; Mutti, H.; Cubero, D.; Fonseca, F.; Del Giglio, A. Curcuma longa (Turmeric) for Prevention of Capecitabine-Induced Hand-Foot Syndrome: A Pilot Study. J. Diet. Suppl. 2018, 15, 606–612. [Google Scholar] [CrossRef] [PubMed]
  52. Sabet, N.S.; Atashbar, S.; Khanlou, E.M.; Kahrizi, F.; Salimi, A. Curcumin attenuates bevacizumab-induced toxicity via suppressing oxidative stress and preventing mitochondrial dysfunction in heart mitochondria. Naunyn Schmiedebergs Arch. Pharm. 2020, 393, 1447–1457. [Google Scholar] [CrossRef] [PubMed]
  53. Belcaro, G.; Hosoi, M.; Pellegrini, L.; Appendino, G.; Ippolito, E.; Ricci, A.; Ledda, A.; Dugall, M.; Cesarone, M.R.; Maione, C.; et al. A controlled study of a lecithinized delivery system of curcumin (Meriva(R)) to alleviate the adverse effects of cancer treatment. Phytother. Res. 2014, 28, 444–450. [Google Scholar] [CrossRef]
  54. Kalluru, H.; Kondaveeti, S.S.; Telapolu, S.; Kalachaveedu, M. Turmeric supplementation improves the quality of life and hematological parameters in breast cancer patients on paclitaxel chemotherapy: A case series. Complement. Clin. Pract. 2020, 41, 101247. [Google Scholar] [CrossRef]
  55. Martinez-Balibrea, E.; Martinez-Cardus, A.; Gines, A.; Ruiz de Porras, V.; Moutinho, C.; Layos, L.; Manzano, J.L.; Buges, C.; Bystrup, S.; Esteller, M.; et al. Tumor-Related Molecular Mechanisms of Oxaliplatin Resistance. Mol. Cancer 2015, 14, 1767–1776. [Google Scholar] [CrossRef] [Green Version]
  56. Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ruiz de Porras, V.; Bystrup, S.; Martinez-Cardus, A.; Pluvinet, R.; Sumoy, L.; Howells, L.; James, M.I.; Iwuji, C.; Manzano, J.L.; Layos, L.; et al. Curcumin mediates oxaliplatin-acquired resistance reversion in colorectal cancer cell lines through modulation of CXC-Chemokine/NF-kappaB signalling pathway. Sci. Rep. 2016, 6, 24675. [Google Scholar] [CrossRef] [Green Version]
  58. Wang, S.; Liu, Z.; Wang, L.; Zhang, X. NF-kappaB signaling pathway, inflammation and colorectal cancer. Cell Mol. Immunol. 2009, 6, 327–334. [Google Scholar] [CrossRef] [Green Version]
  59. Patel, M.; Horgan, P.G.; McMillan, D.C.; Edwards, J. NF-kappaB pathways in the development and progression of colorectal cancer. Transl. Res. 2018, 197, 43–56. [Google Scholar] [CrossRef] [Green Version]
  60. Jobin, C.; Bradham, C.A.; Russo, M.P.; Juma, B.; Narula, A.S.; Brenner, D.A.; Sartor, R.B. Curcumin blocks cytokine-mediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity. J. Immunol. 1999, 163, 3474–3483. [Google Scholar]
  61. Rajitha, B.; Belalcazar, A.; Nagaraju, G.P.; Shaib, W.L.; Snyder, J.P.; Shoji, M.; Pattnaik, S.; Alam, A.; El-Rayes, B.F. Inhibition of NF-kappaB translocation by curcumin analogs induces G0/G1 arrest and downregulates thymidylate synthase in colorectal cancer. Cancer Lett. 2016, 373, 227–233. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, N.; Hao, Y.; Liu, H.; Yu, Q.; Bo, B.; Liang, J. Combined anti-cancer effects of curcumin and oxaliplatin on colon carcinoma colo205 cells using transplanted nude mice. Pak. J. Pharm. Sci. 2021, 34, 2021–2025. [Google Scholar] [PubMed]
  63. Howells, L.M.; Sale, S.; Sriramareddy, S.N.; Irving, G.R.; Jones, D.J.; Ottley, C.J.; Pearson, D.G.; Mann, C.D.; Manson, M.M.; Berry, D.P.; et al. Curcumin ameliorates oxaliplatin-induced chemoresistance in HCT116 colorectal cancer cells in vitro and in vivo. Int. J. Cancer 2011, 129, 476–486. [Google Scholar] [CrossRef] [PubMed]
  64. Ozawa-Umeta, H.; Kishimoto, A.; Imaizumi, A.; Hashimoto, T.; Asakura, T.; Kakeya, H.; Kanai, M. Curcumin beta-D-glucuronide exhibits anti-tumor effects on oxaliplatin-resistant colon cancer with less toxicity in vivo. Cancer Sci. 2020, 111, 1785–1793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Shakibaei, M.; Mobasheri, A.; Lueders, C.; Busch, F.; Shayan, P.; Goel, A. Curcumin enhances the effect of chemotherapy against colorectal cancer cells by inhibition of NF-kappaB and Src protein kinase signaling pathways. PLoS ONE 2013, 8, e57218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Shakibaei, M.; Kraehe, P.; Popper, B.; Shayan, P.; Goel, A.; Buhrmann, C. Curcumin potentiates antitumor activity of 5-fluorouracil in a 3D alginate tumor microenvironment of colorectal cancer. BMC Cancer 2015, 15, 250. [Google Scholar] [CrossRef] [Green Version]
  67. Kunnumakkara, A.B.; Diagaradjane, P.; Anand, P.; Harikumar, K.B.; Deorukhkar, A.; Gelovani, J.; Guha, S.; Krishnan, S.; Aggarwal, B.B. Curcumin sensitizes human colorectal cancer to capecitabine by modulation of cyclin D1, COX-2, MMP-9, VEGF and CXCR4 expression in an orthotopic mouse model. Int. J. Cancer 2009, 125, 2187–2197. [Google Scholar] [CrossRef] [PubMed]
  68. Patel, B.B.; Gupta, D.; Elliott, A.A.; Sengupta, V.; Yu, Y.; Majumdar, A.P. Curcumin targets FOLFOX-surviving colon cancer cells via inhibition of EGFRs and IGF-1R. Anticancer Res. 2010, 30, 319–325. [Google Scholar]
  69. Patel, B.B.; Sengupta, R.; Qazi, S.; Vachhani, H.; Yu, Y.; Rishi, A.K.; Majumdar, A.P. Curcumin enhances the effects of 5-fluorouracil and oxaliplatin in mediating growth inhibition of colon cancer cells by modulating EGFR and IGF-1R. Int. J. Cancer 2008, 122, 267–273. [Google Scholar] [CrossRef]
  70. Kanwar, S.S.; Yu, Y.; Nautiyal, J.; Patel, B.B.; Padhye, S.; Sarkar, F.H.; Majumdar, A.P. Difluorinated-curcumin (CDF): A novel curcumin analog is a potent inhibitor of colon cancer stem-like cells. Pharm. Res. 2011, 28, 827–838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Codony-Servat, J.; Cuatrecasas, M.; Asensio, E.; Montironi, C.; Martinez-Cardus, A.; Marin-Aguilera, M.; Horndler, C.; Martinez-Balibrea, E.; Rubini, M.; Jares, P.; et al. Nuclear IGF-1R predicts chemotherapy and targeted therapy resistance in metastatic colorectal cancer. Br. J. Cancer 2017, 117, 1777–1786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chen, X.; Yeung, T.K.; Wang, Z. Enhanced drug resistance in cells coexpressing ErbB2 with EGF receptor or ErbB3. Biochem. Biophys. Res. Commun. 2000, 277, 757–763. [Google Scholar] [CrossRef] [PubMed]
  73. Watson, R.G.; Muhale, F.; Thorne, L.B.; Yu, J.; O’Neil, B.H.; Hoskins, J.M.; Meyers, M.O.; Deal, A.M.; Ibrahim, J.G.; Hudson, M.L.; et al. Amplification of thymidylate synthetase in metastatic colorectal cancer patients pretreated with 5-fluorouracil-based chemotherapy. Eur. J. Cancer 2010, 46, 3358–3364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bohanes, P.; Labonte, M.J.; Lenz, H.J. A review of excision repair cross-complementation group 1 in colorectal cancer. Clin. Color. Cancer 2011, 10, 157–164. [Google Scholar] [CrossRef]
  75. Han, W.; Yin, H.; Ma, H.; Wang, Y.; Kong, D.; Fan, Z. Curcumin Regulates ERCC1 Expression and Enhances Oxaliplatin Sensitivity in Resistant Colorectal Cancer Cells through Its Effects on miR-409–3p. Evid. Based Complement Altern. Med. 2020, 2020, 8394574. [Google Scholar] [CrossRef]
  76. Agarwal, A.; Kasinathan, A.; Ganesan, R.; Balasubramanian, A.; Bhaskaran, J.; Suresh, S.; Srinivasan, R.; Aravind, K.B.; Sivalingam, N. Curcumin induces apoptosis and cell cycle arrest via the activation of reactive oxygen species-independent mitochondrial apoptotic pathway in Smad4 and p53 mutated colon adenocarcinoma HT29 cells. Nutr. Res. 2018, 51, 67–81. [Google Scholar] [CrossRef]
  77. Li, G.; Fang, S.; Shao, X.; Li, Y.; Tong, Q.; Kong, B.; Chen, L.; Wang, Y.; Yang, J.; Yu, H.; et al. Curcumin Reverses NNMT-Induced 5-Fluorouracil Resistance via Increasing ROS and Cell Cycle Arrest in Colorectal Cancer Cells. Biomolecules 2021, 11, 1295. [Google Scholar] [CrossRef]
  78. Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [Green Version]
  79. Lu, W.; Kang, Y. Epithelial-Mesenchymal Plasticity in Cancer Progression and Metastasis. Dev. Cell 2019, 49, 361–374. [Google Scholar] [CrossRef]
  80. Cao, H.; Xu, E.; Liu, H.; Wan, L.; Lai, M. Epithelial-mesenchymal transition in colorectal cancer metastasis: A system review. Pathol. Res. Pract. 2015, 211, 557–569. [Google Scholar] [CrossRef]
  81. Yang, Y.; Wang, G.; Zhu, D.; Huang, Y.; Luo, Y.; Su, P.; Chen, X.; Wang, Q. Epithelial-mesenchymal transition and cancer stem cell-like phenotype induced by Twist1 contribute to acquired resistance to irinotecan in colon cancer. Int. J. Oncol. 2017, 51, 515–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Yang, A.D.; Fan, F.; Camp, E.R.; van Buren, G.; Liu, W.; Somcio, R.; Gray, M.J.; Cheng, H.; Hoff, P.M.; Ellis, L.M. Chronic oxaliplatin resistance induces epithelial-to-mesenchymal transition in colorectal cancer cell lines. Clin. Cancer Res. 2006, 12 Pt 1, 4147–4153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zhang, C.; Xu, Y.; Wang, H.; Li, G.; Yan, H.; Fei, Z.; Xu, Y.; Li, W. Curcumin reverses irinotecan resistance in colon cancer cell by regulation of epithelial-mesenchymal transition. Anticancer Drugs 2018, 29, 334–340. [Google Scholar] [CrossRef]
  84. Yin, J.; Wang, L.; Wang, Y.; Shen, H.; Wang, X.; Wu, L. Curcumin reverses oxaliplatin resistance in human colorectal cancer via regulation of TGF-beta/Smad2/3 signaling pathway. Onco. Targets 2019, 12, 3893–3903. [Google Scholar] [CrossRef] [PubMed]
  85. Toden, S.; Okugawa, Y.; Jascur, T.; Wodarz, D.; Komarova, N.L.; Buhrmann, C.; Shakibaei, M.; Boland, C.R.; Goel, A. Curcumin mediates chemosensitization to 5-fluorouracil through miRNA-induced suppression of epithelial-to-mesenchymal transition in chemoresistant colorectal cancer. Carcinogenesis 2015, 36, 355–367. [Google Scholar] [CrossRef] [Green Version]
  86. Lu, Y.; Zhang, R.; Zhang, X.; Zhang, B.; Yao, Q. Curcumin may reverse 5-fluorouracil resistance on colonic cancer cells by regulating TET1-NKD-Wnt signal pathway to inhibit the EMT progress. Biomed. Pharm. 2020, 129, 110381. [Google Scholar] [CrossRef]
  87. Najafi, M.; Mortezaee, K.; Majidpoor, J. Cancer stem cell (CSC) resistance drivers. Life Sci. 2019, 234, 116781. [Google Scholar] [CrossRef]
  88. Yu, Y.; Kanwar, S.S.; Patel, B.B.; Nautiyal, J.; Sarkar, F.H.; Majumdar, A.P. Elimination of Colon Cancer Stem-Like Cells by the Combination of Curcumin and FOLFOX. Transl. Oncol. 2009, 2, 321–328. [Google Scholar] [CrossRef] [Green Version]
  89. Shakibaei, M.; Buhrmann, C.; Kraehe, P.; Shayan, P.; Lueders, C.; Goel, A. Curcumin chemosensitizes 5-fluorouracil resistant MMR-deficient human colon cancer cells in high density cultures. PLoS ONE 2014, 9, e85397. [Google Scholar] [CrossRef] [Green Version]
  90. Su, P.; Yang, Y.; Wang, G.; Chen, X.; Ju, Y. Curcumin attenuates resistance to irinotecan via induction of apoptosis of cancer stem cells in chemoresistant colon cancer cells. Int. J. Oncol. 2018, 53, 1343–1353. [Google Scholar] [CrossRef] [Green Version]
  91. Yu, Y.; Sarkar, F.H.; Majumdar, A.P. Down-regulation of miR-21 Induces Differentiation of Chemoresistant Colon Cancer Cells and Enhances Susceptibility to Therapeutic Regimens. Transl. Oncol. 2013, 6, 180–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Elbadawy, M.; Hayashi, K.; Ayame, H.; Ishihara, Y.; Abugomaa, A.; Shibutani, M.; Hayashi, S.M.; Hazama, S.; Takenouchi, H.; Nakajima, M.; et al. Anti-cancer activity of amorphous curcumin preparation in patient-derived colorectal cancer organoids. Biomed. Pharm. 2021, 142, 112043. [Google Scholar] [CrossRef] [PubMed]
  93. Zheng, X.; Yang, X.; Lin, J.; Song, F.; Shao, Y. Low curcumin concentration enhances the anticancer effect of 5-fluorouracil against colorectal cancer. Phytomedicine 2021, 85, 153547. [Google Scholar] [CrossRef] [PubMed]
  94. He, W.T.; Zhu, Y.H.; Zhang, T.; Abulimiti, P.; Zeng, F.Y.; Zhang, L.P.; Luo, L.J.; Xie, X.M.; Zhang, H.L. Curcumin Reverses 5-Fluorouracil Resistance by Promoting Human Colon Cancer HCT-8/5-FU Cell Apoptosis and Down-regulating Heat Shock Protein 27 and P-Glycoprotein. Chin. J. Integr. Med. 2019, 25, 416–424. [Google Scholar] [CrossRef]
  95. Hoffmann, U.; Kroemer, H.K. The ABC transporters MDR1 and MRP2: Multiple functions in disposition of xenobiotics and drug resistance. Drug Metab. Rev. 2004, 36, 669–701. [Google Scholar] [CrossRef]
  96. Smith, A.J.; van Helvoort, A.; van Meer, G.; Szabo, K.; Welker, E.; Szakacs, G.; Varadi, A.; Sarkadi, B.; Borst, P. MDR3 P-glycoprotein, a phosphatidylcholine translocase, transports several cytotoxic drugs and directly interacts with drugs as judged by interference with nucleotide trapping. J. Biol. Chem. 2000, 275, 23530–23539. [Google Scholar] [CrossRef] [Green Version]
  97. Germann, U.A. P-glycoprotein—A mediator of multidrug resistance in tumour cells. Eur. J. Cancer 1996, 32A, 927–944. [Google Scholar] [CrossRef]
  98. Ruetz, S.; Gros, P. A mechanism for P-glycoprotein action in multidrug resistance: Are we there yet? Trends Pharm. Sci. 1994, 15, 260–263. [Google Scholar] [CrossRef]
  99. Borst, P. Looking back at multidrug resistance (MDR) research and ten mistakes to be avoided when writing about ABC transporters in MDR. FEBS Lett. 2020, 594, 4001–4011. [Google Scholar] [CrossRef]
  100. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef]
  101. Montazami, N.; Kheir Andish, M.; Majidi, J.; Yousefi, M.; Yousefi, B.; Mohamadnejad, L.; Shanebandi, D.; Estiar, M.A.; Khaze, V.; Mansoori, B.; et al. siRNA-mediated silencing of MDR1 reverses the resistance to oxaliplatin in SW480/OxR colon cancer cells. Cell Mol. Biol. 2015, 61, 98–103. [Google Scholar] [PubMed]
  102. Beretta, G.L.; Benedetti, V.; Cossa, G.; Assaraf, Y.G.; Bram, E.; Gatti, L.; Corna, E.; Carenini, N.; Colangelo, D.; Howell, S.B.; et al. Increased levels and defective glycosylation of MRPs in ovarian carcinoma cells resistant to oxaliplatin. Biochem. Pharm. 2010, 79, 1108–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ekblad, L.; Kjellstrom, J.; Johnsson, A. Reduced drug accumulation is more important in acquired resistance against oxaliplatin than against cisplatin in isogenic colon cancer cells. Anticancer Drugs 2010, 21, 523–531. [Google Scholar] [CrossRef] [PubMed]
  104. Lee, J.H.; Um, J.W.; Lee, J.H.; Kim, S.H.; Lee, E.S.; Kim, Y.S. Can immunohistochemistry of multidrug-resistant proteins replace the histoculture drug response assay in colorectal adenocarcinomas? Hepatogastroenterology 2012, 59, 1075–1078. [Google Scholar] [PubMed]
  105. Marin, J.J.G.; Macias, R.I.R.; Monte, M.J.; Herraez, E.; Peleteiro-Vigil, A.; Blas, B.S.; Sanchon-Sanchez, P.; Temprano, A.G.; Espinosa-Escudero, R.A.; Lozano, E.; et al. Cellular Mechanisms Accounting for the Refractoriness of Colorectal Carcinoma to Pharmacological Treatment. Cancers 2020, 12, 2605. [Google Scholar] [CrossRef]
  106. Cao, D.; Qin, S.; Mu, Y.; Zhong, M. The role of MRP1 in the multidrug resistance of colorectal cancer. Oncol. Lett. 2017, 13, 2471–2476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Garcea, G.; Berry, D.P.; Jones, D.J.; Singh, R.; Dennison, A.R.; Farmer, P.B.; Sharma, R.A.; Steward, W.P.; Gescher, A.J. Consumption of the putative chemopreventive agent curcumin by cancer patients: Assessment of curcumin levels in the colorectum and their pharmacodynamic consequences. Cancer Epidemiol. Biomark. Prev. 2005, 14, 120–125. [Google Scholar] [CrossRef]
  108. Irving, G.R.; Howells, L.M.; Sale, S.; Kralj-Hans, I.; Atkin, W.S.; Clark, S.K.; Britton, R.G.; Jones, D.J.; Scott, E.N.; Berry, D.P.; et al. Prolonged biologically active colonic tissue levels of curcumin achieved after oral administration--a clinical pilot study including assessment of patient acceptability. Cancer Prev. Res. 2013, 6, 119–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Sharma, R.A.; Euden, S.A.; Platton, S.L.; Cooke, D.N.; Shafayat, A.; Hewitt, H.R.; Marczylo, T.H.; Morgan, B.; Hemingway, D.; Plummer, S.M.; et al. Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance. Clin. Cancer Res. 2004, 10, 6847–6854. [Google Scholar] [CrossRef] [Green Version]
  110. James, M.I.; Iwuji, C.; Irving, G.; Karmokar, A.; Higgins, J.A.; Griffin-Teal, N.; Thomas, A.; Greaves, P.; Cai, H.; Patel, S.R.; et al. Curcumin inhibits cancer stem cell phenotypes in ex vivo models of colorectal liver metastases, and is clinically safe and tolerable in combination with FOLFOX chemotherapy. Cancer Lett. 2015, 364, 135–141. [Google Scholar] [CrossRef] [Green Version]
  111. Howells, L.M.; Iwuji, C.O.O.; Irving, G.R.B.; Barber, S.; Walter, H.; Sidat, Z.; Griffin-Teall, N.; Singh, R.; Foreman, N.; Patel, S.R.; et al. Curcumin Combined with FOLFOX Chemotherapy Is Safe and Tolerable in Patients with Metastatic Colorectal Cancer in a Randomized Phase IIa Trial. J. Nutr. 2019, 149, 1133–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lu, L.; Przybylla, R.; Shang, Y.; Dai, M.; Krohn, M.; Kramer, O.H.; Mullins, C.S.; Linnebacher, M. Microsatellite Status and IkappaBalpha Expression Levels Predict Sensitivity to Pharmaceutical Curcumin in Colorectal Cancer Cells. Cancers 2022, 14, 1032. [Google Scholar] [CrossRef] [PubMed]
  113. Stohs, S.J.; Chen, O.; Ray, S.D.; Ji, J.; Bucci, L.R.; Preuss, H.G. Highly Bioavailable Forms of Curcumin and Promising Avenues for Curcumin-Based Research and Application: A Review. Molecules 2020, 25, 1397. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Potential use of curcumin to mitigate therapy-related side effects in mCRC. CURC: curcumin; NF-κB: nuclear factor kappa B; Nrf2: nuclear factor -erythroid 2- related factor 2; ROS: reactive oxygen species. Created with BioRender.com (accessed on 16 September 2022).
Figure 1. Potential use of curcumin to mitigate therapy-related side effects in mCRC. CURC: curcumin; NF-κB: nuclear factor kappa B; Nrf2: nuclear factor -erythroid 2- related factor 2; ROS: reactive oxygen species. Created with BioRender.com (accessed on 16 September 2022).
Ijms 23 14058 g001
Table 1. Main molecular mechanisms of action of the combination of curcumin and chemotherapeutic agents in preclinical models of CRC. ABCG2: ATP-binding cassette transporter G2; CSCs: cancer stem cells; EGFR: epidermal growth factor receptor; EMT: epithelial-to-mesenchymal transition; IGF-1R: growth factor-1 receptor; NF-κB: nuclear factor kappa B; P-gp: P-glycoprotein; ROS: reactive oxygen species; TGF-β: transforming growth factor β; TS: thymidylate synthase.
Table 1. Main molecular mechanisms of action of the combination of curcumin and chemotherapeutic agents in preclinical models of CRC. ABCG2: ATP-binding cassette transporter G2; CSCs: cancer stem cells; EGFR: epidermal growth factor receptor; EMT: epithelial-to-mesenchymal transition; IGF-1R: growth factor-1 receptor; NF-κB: nuclear factor kappa B; P-gp: P-glycoprotein; ROS: reactive oxygen species; TGF-β: transforming growth factor β; TS: thymidylate synthase.
Treatment RegimenMolecular Targets of CurcuminReferences
Oxaliplatin + CurcuminInhibition of NF-κB activation[57,62,63,64]
Downregulation of CXCL8, CXCL1 and CXCL2 chemokines[57]
Inhibition of AKT activation[57]
Inhibition of miR-409-3p/ERCC1 axis[75]
TGF-β/SMAD2/3 signaling attenuation[84]
P-gp downregulation[75,94]
5-fluorouracil + CurcuminInhibition of NF-κB activation[61,65,66]
Downregulation of TS[61]
P-gp downregulation[75,94]
ROS generation[77]
Downregulation of TET1-NK2-WNT pathway[86]
Elimination of CSCs[89]
Upregulation of EMT-suppressive miRNAs (miR-200b, miR-200c, miR-141, miR-429, miR-101t)[85]
Inhibition of ERK signaling pathway[92,93]
FOLFOX + CurcuminDownregulation of EGFR and IGF-1R[68,69,70]
Inhibition of NF-κB activation[70]
Inactivation of β-catenin, COX-2, c-Myc and Bcl-xL[70]
Elimination of CSCs[70,88]
Downregulation of miR-21[91]
Downregulation of ABCG2 drug-efflux transporter[70]
Irinotecan + CurcuminDownregulation of EMT markers (Vimentin and N-cadherin)[83]
Elimination of CSCs by apoptosis induction[90]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Layos, L.; Martínez-Balibrea, E.; Ruiz de Porras, V. Curcumin: A Novel Way to Improve Quality of Life for Colorectal Cancer Patients? Int. J. Mol. Sci. 2022, 23, 14058. https://doi.org/10.3390/ijms232214058

AMA Style

Layos L, Martínez-Balibrea E, Ruiz de Porras V. Curcumin: A Novel Way to Improve Quality of Life for Colorectal Cancer Patients? International Journal of Molecular Sciences. 2022; 23(22):14058. https://doi.org/10.3390/ijms232214058

Chicago/Turabian Style

Layos, Laura, Eva Martínez-Balibrea, and Vicenç Ruiz de Porras. 2022. "Curcumin: A Novel Way to Improve Quality of Life for Colorectal Cancer Patients?" International Journal of Molecular Sciences 23, no. 22: 14058. https://doi.org/10.3390/ijms232214058

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