Field Of Vision Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastrointest Surg. Dec 27, 2012; 4(12): 275-277
Published online Dec 27, 2012. doi: 10.4240/wjgs.v4.i12.275
On biomarkers and pathways in rectal cancer: What's the target?
Gabriele Zoppoli, Department of Internal Medicine, Istituti di Ricovero e Cura a Carattere Scientifico, Azienda Ospedaliera Universitaria San Martino, Istituto Nazionale per la Ricerca sul Cancro, Istituto Scientifico Tumori, 16137 Genova, Italy
Valter Ferrando, Stefano Scabini, Oncological Surgery and Implantable Systems Unit, Istituti di Ricovero e Cura a Carattere Scientifico, Azienda Ospedaliera Universitaria San Martino, Istituto Scientifico Tumori, 16137 Genova, Italy
Author contributions: Zoppoli G, Ferrando V and Scabini S discussed the topics, wrote and revised the manuscript.
Correspondence to: Gabriele Zoppoli, MD, PhD, Department of Internal Medicine, Istituti di Ricovero e Cura a Carattere Scientifico, Azienda Ospedaliera Universitaria San Martino, Istituto Nazionale per la Ricerca sul Cancro, Istituto Scientifico Tumori, Viale Benedetto XV, 6, 16137 Genova, Italy. zoppoli@gmail.com
Telephone: +39-10-3537968 Fax: +39-10-3537996
Received: June 14, 2012
Revised: October 2, 2012
Accepted: December 1, 2012
Published online: December 27, 2012

Abstract

In spite of tremendous progresses in surgical and chemo-radiotherapeutic regimens, rectal cancer still suffers from high relapse and mortality rates, and metastatic disease is incurable. Here we assess some of the most recent and validated biomarkers and potential targets studied in rectal cancer, and provide comments to a recent monographic topic covering several aspects of colorectal cancer, published in Current Cancer Drug Targets.

Key Words: Rectal cancer, Colorectal surgery, Chemotherapy, Targeted molecular therapy, Biological markers



COMMENTARY ON HOT ARTICLES

The management of rectal adenocarcinoma has undergone tremendous improvements in the last decade, especially through the advancement of surgical techniques combined with a better defined timing of medical treatment. Still, rectal adenocarcinoma affects about 140 000 new patients each year in Europe, and has a 5-year overall survival of 54%[1]. Currently, standard treatment for stage II and III rectal cancer includes pre-operative (neoadjuvant) treatment with 5-fluorouracyl or capecitabine, in combination with ionizing radiation (IR) therapy[1]. The introduction of biological drugs targeting receptor kinases like epidermal growth factor receptor (EGFR) (cetuximab, panitumumab[2,3]), or their ligands, like vascular endothelial growth factor (VEGF) (bevacizumab[4]), has shown promising results. Nonetheless, DNA damaging agents like capecitabine and IR have maintained their roles as the single, most effective modalities of treatment in locally advanced rectal cancer, together with surgery.

In spite of the above mentioned advancements, a large fraction of the patients who undergo regimens containing these agents does not respond to their action, strongly suggesting that rectal tumors can harbor resistance mechanisms ab initio, or are able to acquire them in the course of therapy. Microarray studies have been used in the effort of creating classifiers and predictors to treatment response, but the results are scarcely consistent and have not been validated extensively[5,6]. Small datasets, different technical and statistical approaches, and inhomogeneous treatment modalities might have contributed to suboptimal results in data interpretation. Among the best studied molecular markers of rectal cancer (p53[7], p21[8], Bax and Mib[9], p27[10], thymidylate synthase[11], EGFR[12] or VEGFR[13]) few have shown to hold some promise in rectal cancer prognostic assessment before standard regimens, and their advantage over conventional pathological staging procedures to predict outcome has not yet been validated in large, prospective studies.

Identifying new biomarkers in rectal cancer management could be advantageous for several reasons: (1) the a priori knowledge of tumor resistance to neoadjuvant treatment would spare patients useless and potentially toxic pharmacologic agents, and could lead to the choice of different strategies (e.g., immediate or more radical surgical intervention or shorter courses of adjuvant chemotherapy in complete responders to neoadjuvant treatment); (2) understanding the molecular alterations which constitute the ground of rectal cancer may allow the use of new biologically targeted agents, in combination with surgical resection; and (3) last but not least, the cost/effectiveness ratio of proposed management strategies could be better assessed, in a time when the economic burden of the health care system is steadily growing toward unmanageable dimensions.

The increasingly appreciated complexity of colorectal cancer systems biology is well addressed by the recently published monographic topic, published in Current Cancer Drug Targets (CCDT)[14-19]. Here, the contributing Authors deal with two of the mainstays of the new “smart weaponries” in colorectal cancer treatment: the EGFR pathway[15] and VEGF signaling[14]. Moreover, two new “hot topics” are covered: the concept of synthetic lethality[18] and the translational potential of mathematical simulations of signaling networks involved in the neoplastic process[19]. Synthetic lethality refers to the ideal situation where the inactivation of one protein product or another does not affect cancer viability, whereas the combined deficiency of both proteins is deadly for the cancer cell[20,21]. The first successful application of this model has been observed in the treatment of breast cancer 1 gene (BRCA1)-deficient breast cancer with poly (ADP-ribose) polymerase (PARP) inhibitors[22,23]. The difficulty of finding new synthetic lethal interactions lies in the combinatorial complexity of identifying pairs of protein products showing such properties. While high throughput silencing RNA based screenings may be of help in this field[24], a deepened understanding of how molecular networks interact with each other and dynamically react to internal and external stimuli[25] (such as chemotherapy, IR or targeted agents) is of the essence to generate plausible hypotheses before testing them in “real life”. This last issue is well addressed by Parodi[16] in the aforementioned topic.

Finally, an “old dog with potentially new tricks” is also presented in the above referenced CCDT issue: targeting DNA damage repair pathways and cell cycle checkpoints in colorectal cancer[17]. While the Reader could reasonably object that such targets are nothing else than those aimed at for the last fifty years by conventional chemotherapy, an essential and relatively overlooked concept is highlighted: since cancer is ontologically characterized, among other features, by genomic instability and mutations[26], intrinsic deficits must exist in tumors which hamper their ability to repair their own genetic information. As a consequence, cancer cells should be more prone than healthy tissues to be killed by DNA damaging agents. It is therefore likely that, with a better knowledge of “what’s wrong”, physicians could be able to predict “what would be right” in individual cases. Again, PARP inhibition in breast cancer with BRCA1 germline alterations has been the proof of principle, but it would be simplistic to assume that no other DNA damage repair genes are altered in somatic tumors, hence showing similar properties. This, in turn, leads directly back to one of the main questions in the management of rectal cancer, i.e., why do some cases exhibit exquisite sensitivity to neoadjuvant chemo-radiation, whereas others appear to be completely resistant?

In conclusion, the recent topic appeared in CCDT is an interesting and comprehensive reading, that covers several essential aspects of what is currently known about colorectal cancer, and provides the Reader with an updated overview of its biology and of the future roads that may potentially lead to the complete cure of most patients affected by rectal cancer. The greatest endeavor of research in rectal cancer remains that of combining big, well-conducted prospective clinical trials with large breadth ancillary biologic studies. Only this synergism between basic and clinical analytic efforts will lead to the discovery and validation of new biomarkers with a real impact in everyday oncological and surgical practice.

Footnotes

Peer reviewer: Tsuyoshi Konishi, MD, PhD, Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-10-6 Ariake, Koto-ku, Tokyo 135-8550, Japan

S- Editor Wen LL L- Editor A E- Editor Xiong L

References
1.  Glimelius B, Oliveira J. Rectal cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2009;20 Suppl 4:54-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
2.  Bengala C, Bettelli S, Bertolini F, Salvi S, Chiara S, Sonaglio C, Losi L, Bigiani N, Sartori G, Dealis C. Epidermal growth factor receptor gene copy number, K-ras mutation and pathological response to preoperative cetuximab, 5-FU and radiation therapy in locally advanced rectal cancer. Ann Oncol. 2009;20:469-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 74]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
3.  Stephenson JJ, Gregory C, Burris H, Larson T, Verma U, Cohn A, Crawford J, Cohen RB, Martin J, Lum P. An open-label clinical trial evaluating safety and pharmacokinetics of two dosing schedules of panitumumab in patients with solid tumors. Clin Colorectal Cancer. 2009;8:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
4.  Willett CG, Duda DG, di Tomaso E, Boucher Y, Ancukiewicz M, Sahani DV, Lahdenranta J, Chung DC, Fischman AJ, Lauwers GY. Efficacy, safety, and biomarkers of neoadjuvant bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a multidisciplinary phase II study. J Clin Oncol. 2009;27:3020-3026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 375]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
5.  Ghadimi BM, Grade M, Difilippantonio MJ, Varma S, Simon R, Montagna C, Füzesi L, Langer C, Becker H, Liersch T. Effectiveness of gene expression profiling for response prediction of rectal adenocarcinomas to preoperative chemoradiotherapy. J Clin Oncol. 2005;23:1826-1838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 260]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
6.  Watanabe T, Komuro Y, Kiyomatsu T, Kanazawa T, Kazama Y, Tanaka J, Tanaka T, Yamamoto Y, Shirane M, Muto T. Prediction of sensitivity of rectal cancer cells in response to preoperative radiotherapy by DNA microarray analysis of gene expression profiles. Cancer Res. 2006;66:3370-3374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 168]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
7.  Lopez-Crapez E, Bibeau F, Thézenas S, Ychou M, Simony-Lafontaine J, Thirion A, Azria D, Grenier J, Senesse P. p53 status and response to radiotherapy in rectal cancer: a prospective multilevel analysis. Br J Cancer. 2005;92:2114-2121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
8.  Rau B, Sturm I, Lage H, Berger S, Schneider U, Hauptmann S, Wust P, Riess H, Schlag PM, Dörken B. Dynamic expression profile of p21WAF1/CIP1 and Ki-67 predicts survival in rectal carcinoma treated with preoperative radiochemotherapy. J Clin Oncol. 2003;21:3391-3401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 105]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
9.  Huerta S, Hrom J, Gao X, Saha D, Anthony T, Reinhart H, Kapur P. Tissue microarray constructs to predict a response to chemoradiation in rectal cancer. Dig Liver Dis. 2010;42:679-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
10.  Hoos A, Nissan A, Stojadinovic A, Shia J, Hedvat CV, Leung DH, Paty PB, Klimstra D, Cordon-Cardo C, Wong WD. Tissue microarray molecular profiling of early, node-negative adenocarcinoma of the rectum: a comprehensive analysis. Clin Cancer Res. 2002;8:3841-3849.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Liersch T, Langer C, Ghadimi BM, Kulle B, Aust DE, Baretton GB, Schwabe W, Häusler P, Becker H, Jakob C. Lymph node status and TS gene expression are prognostic markers in stage II/III rectal cancer after neoadjuvant fluorouracil-based chemoradiotherapy. J Clin Oncol. 2006;24:4062-4068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
12.  Giralt J, de las Heras M, Cerezo L, Eraso A, Hermosilla E, Velez D, Lujan J, Espin E, Rosello J, Majó J. The expression of epidermal growth factor receptor results in a worse prognosis for patients with rectal cancer treated with preoperative radiotherapy: a multicenter, retrospective analysis. Radiother Oncol. 2005;74:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Zlobec I, Steele R, Compton CC. VEGF as a predictive marker of rectal tumor response to preoperative radiotherapy. Cancer. 2005;104:2517-2521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
14.  Bagnasco L, Piras D, Parodi S, Bauer I, Zoppoli G, Patrone F, Ballestrero A. Role of angiogenesis inhibitors in colorectal cancer: sensitive and insensitive tumors. Curr Cancer Drug Targets. 2012;12:303-315.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Ballestrero A, Garuti A, Cirmena G, Rocco I, Palermo C, Nencioni A, Scabini S, Zoppoli G, Parodi S, Patrone F. Patient-tailored treatments with anti-EGFR monoclonal antibodies in advanced colorectal cancer: KRAS and beyond. Curr Cancer Drug Targets. 2012;12:316-328.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Parodi S. EDITORIAL: Molecularly targeted treatments for colorectal cancer: advances and limitations. Curr Cancer Drug Targets. 2012;12:301-302.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Solier S, Zhang YW, Ballestrero A, Pommier Y, Zoppoli G. DNA damage response pathways and cell cycle checkpoints in colorectal cancer: current concepts and future perspectives for targeted treatment. Curr Cancer Drug Targets. 2012;12:356-371.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Soncini D, Caffa I, Patrone F, Ballestrero A, Nencioni A. Synthetic lethality-based therapeutics: perspectives for applications in colorectal cancer. Curr Cancer Drug Targets. 2012;12:329-338.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Tortolina L, Castagnino N, De Ambrosi C, Moran E, Patrone F, Ballestrero A, Parodi S. A multi-scale approach to colorectal cancer: from a biochemical- interaction signaling-network level, to multi-cellular dynamics of malignant transformation. Interplay with mutations and onco-protein inhibitor drugs. Curr Cancer Drug Targets. 2012;12:339-355.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Kaelin WG. The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer. 2005;5:689-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1064]  [Cited by in F6Publishing: 1002]  [Article Influence: 52.7]  [Reference Citation Analysis (0)]
21.  Iglehart JD, Silver DP. Synthetic lethality--a new direction in cancer-drug development. N Engl J Med. 2009;361:189-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 170]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
22.  Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361:123-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2792]  [Cited by in F6Publishing: 2724]  [Article Influence: 181.6]  [Reference Citation Analysis (0)]
23.  Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, Friedlander M, Arun B, Loman N, Schmutzler RK. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376:235-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1352]  [Cited by in F6Publishing: 1316]  [Article Influence: 94.0]  [Reference Citation Analysis (0)]
24.  Echeverri CJ, Perrimon N. High-throughput RNAi screening in cultured cells: a user's guide. Nat Rev Genet. 2006;7:373-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 261]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
25.  Kholodenko B, Yaffe MB, Kolch W. Computational approaches for analyzing information flow in biological networks. Sci Signal. 2012;5:re1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 147]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
26.  Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39812]  [Cited by in F6Publishing: 42993]  [Article Influence: 3307.2]  [Reference Citation Analysis (4)]