Open access peer-reviewed chapter

What Are the New Challenges of the Current Cancer Biomarkers?

Written By

Jie Chen, Liqiong Yang, Yuxi Duan, Tinging Pu, Sha Zheng, Fangfang Liu, Kun Huang, Greg Mirt and Fan Xu

Submitted: 08 July 2019 Reviewed: 30 September 2019 Published: 31 October 2019

DOI: 10.5772/intechopen.89976

From the Edited Volume

Current Cancer Treatment

Edited by Mirjana Rajer and Eva Segelov

Chapter metrics overview

768 Chapter Downloads

View Full Metrics

Abstract

Biomarkers are emerging research filed in the past decade. Even though numerous biomarkers were reported, the efficiency of cancer therapy remains low. So the question emerges as to how much can we trust the current biomarkers on cancer therapy? In this upcoming chapter, we would like to illustrate the outcomes of classical cancer therapies on advanced pancreatic cancer disclosed successful, neutral and failed in clinical trials. The analysis will be carried on the perspective interdisciplinary on the biomarkers including anatomy, physiology, pharmacology, biochemistry, history path and development of pharmacy. Particular in-depth insight may open a window for new researches and lighting the therapies.

Keywords

  • advanced pancreatic cancer
  • biomarker
  • clinical trials

1. Introduction

Advance pancreatic cancer (APC) is a highly lethal tumor. Most patients with APC remain asymptomatic until the disease reaches an advanced stage [1]. The incidence rate was 5.5 for men and 4.0 for women per 100,000 people. The mortality rate was 5.1 for men and 3.8 for women per 100,000 people around the world, according to data, from 2018 [2]. For the incidence rate, Asia is at 48.4%, Europe at 23.4%, and the Americas is at 21.0%. As for the mortality rate, Asia is at 57.3%, Europe at 20.3%, and the Americas is at 14.3% [3].

Our previous study disclosed that there were more than 19 chemotherapy regimens combinations in clinical practice available [4]. The advantages and disadvantages of each therapy regimens are clear. Briefly, to lengthen the overall survival and to reduce the treatment-related toxicity we must consider the outclass selection. There are more than 14 treatment-related toxicities in gastrointestinal, constitutional, skin, hepatotoxicity, infection, vascular, neuropathy, mental, pain, renal, electrolytes and pulmonary of human body in current dominant chemotherapy regimens. To broaden the balance requires expertise and professional medical training based on evidence.

Advertisement

2. Long period run in research and development in pharmacy

The development of drugs is based on the determination of new therapeutic targets, the pharmacological receptors. This concept was first proposed by Paul Ehrlich in 1908 [5]. Normal cells replicate their DNA with great accuracy, but cancer has a large number of mutations that show up in cancer cells that make them pharmacological targets [6]. From the initial concept of molecular targets, drug targets were discovered and validated, which successful translated the most drugs into practice [7].

Excellent and reliable targets identification and validation can increase the credibility of the relationship between intentions and diseases, this may strength the effectively of drugs. Drugs are usually developed only when specific drug target for the action of these drugs are analyzed and examined. Sufficient potential targets have been discovered rapidly for the drug discovery process.

Numerous data including identified gene and drug discovery cycles have been generated exponentially. This may forge the difficultness in decision making and becomes more and more difficult for drug R&D. Thanks to rapid bioinformatic discoveries, more and more biopharmaceutical targets can be identified and analyzed [8].

Validation from cross-species a bioeffect is performed after the drug target is determined and verified. Rodents and non-human primates provide appropriate animal models for screening and evaluation of a new drug. Most of current cancer in vivo experiments use rodent experimental animals such as mice and rats. Because they are small, rapid reproduction, clear genetic background and mature genetic modification technology can be done. However, due to the distant relationship between rodents and humans, many of the results obtained from rodent models cannot be reproduced in humans. Moreover, non-human primates are highly similar to humans in terms of genetic evolution, immunity, physiology and metabolism. They are theoretically more suitable for cancer researches [9].

Clinical trials are the best channel to tie up pharmaceutical targets to reliable drugs. The goal is to determine whether a candidate drug is safe and effective. There are four phases in clinical trials. More specific biomarker studies are based on data from prospective studies [10]. In the study of cancer biomarkers, retrospective studies and prospective studies help to identify potential biomarkers, which may be validated in the future studies, however, the reliability of evidence remains controversial.

Advertisement

3. Can we trust the current biomarkers of cancer?

Unfortunately, the overall survival of APC patients has not revised assuredly. There are too many choices in clinical practice and evidence-based medicine is a permanent challenge. Which of the modern biomarkers is reliable? Are we ever going to detect precise pharmaceutical targets on APC [11]?

Advertisement

4. Validation method

In order to clarify this question, we collected the raw data source (http://clinicaltrials.gov) and searched all the drug treatments on APC. We refined all the data which had results and were published. Briefly, a total of 2726 recordings were found since May 2019. Hundred and fifteen recordings which finished clinical trials, further we ruled out irrelevant 32 recordings and 56 unclear results. Finally, 27 recordings kept comprising the following three tables. Raw data are free, please follow the link 10.6084/m9.figshare.8275190.

Advertisement

5. What can data tell us?

Total, there are 18 biomarkers used among these results. For details please see Table 1. The results from 27 clinical trials could be divided into three categories (a) rank of the effectiveness (b) rank of intervention and (c) quality of life improved.

5.1 Estimation of clinical trials with success outcome on advanced pancreatic cancer

Total, we found 10 publications where the author declares the successful outcome on the treatment of APC from 2011 to 2019, Table 2. These clinical trials recruited 1080 patients with APC (611 male and 469 female). The average overall survival month is 11.62 and progress-free survival month is 10.79. Briefly, Lutz et al. tests the GVAX pancreatic cancer vaccine via GPI biomarkers The OS and PFS approach got the highest point, 24.8 and 17.3, respectively Similarly, Phan et al. tested the pazopanib hydrochloride via VEGF biomarkers. This approach had a higher OS and PFS points. Survival months are 25 and 14.4. Furthermore, Hong et al. disclosed the capecitabine may put the OS and PFS to 17.3 and 10.4 survival months; the remain studies presented similar results, the OS and PFS were lower than 10 months.

BiomarkersAbbreviation
Vascular endothelial growth factor*VEGF
Thymidine phosphorylaseTP
Epidermal growth factor receptor*EGFR
Tumor necrosis factor αTNF-α
Topoisomerase I inhibitorTIH
Sonic hedgehogSHH
Severe hypoxia intracellular reductasesSHIR
Secreted protein acid rich in cysteineSPARC
Platelet-derived growth factorPDGF
MEK1/2-dependent effector proteinsERK1/2
Kirsten rat sarcoma viral oncogene homolog*KRAS
Interleukin 6/interleukin 8IL-6/IL-8
Heat shock protein 27Hsp27
Glycosyl-phosphatidylinositolGPI
Double-strand breaks in DNA
Checkpoint kinase 1CHK1
Microtubule-associated protein light chain 3- IILC3- II
Circulating free DNAcfDNA
Dihydropyrimidine dehydrogenaseDPD

Table 1.

Potential biomarkers used in advanced pancreatic cancer.

NCCN Recommend: National Comprehensive Cancer Network.

YearAuthorNMFDrugOSPFSBiomarkers
2011Hill [12]21813Capecitabine; docetaxel;7.45.8TP
gemcitabine
2011Lutz [13]603723GVAX pancreatic cancer24.817.3GPI
Vaccine
2011Raymond [14]864244Sunitinib930VEGF
854045Sunitinib2151
2013Hosein [15]19910Abraxane7.31.7SPARC
2014Soares [16]432122Capecitabine; docetaxel5.33.7TP
2014Ban [17]33303Belotaxel; belloxa10.93.6Double-strand breaks in DNA
2014Borad [18]694029TH-302 with gemcitabine;6.93.6Severe hypoxia
Intracellular reductases
Gemcitabine
714427TH-303 with gemcitabine;8.75.6
gemcitabine
744232TH-304 with gemcitabine;9.26
gemcitabine
2015Phan [19]322210Pazopanib hydrochloride2514.4VEGF
20128Pazopanib hydrochloride18.512.2
2019Wang-Gillam [20]1518764MM-3984.92.7Topoisomerase I inhibitor
14981685 Fluorouracil; leucovorin4.21.6
1176948MM-398; 5 fluorouracil;6.23.1
leucovorin

Table 2.

Clinical trials with a successful outcome on pancreatic cancer.

5.2 Estimation of clinical trials with the neutral outcome on advanced pancreatic cancer

Here are three studies clarified neural results in OS and PFS result, ranged from 2008 to 2016, totally recruited 229 APC patients (121 male and 108 female). For details please see Table 3. Averagely the OS is 16.6 months and PFS is 9 months in neutral studies. For example, Jean in 2008 reported gemcitabine plus AG-013736 achieved better OS and PFS (6.9 and 4.2) than gemcitabine single used.

YearAuthorNMFDrugOSPFSBiomarkers
2008Spano [21]693534Gemcitabine5.63.7VEGF
341618Gemcitabine; AG-0137366.94.2
2015Hobday [22]582929Bevacizumab; temsirolimus3413.2VEGF
2016Stein [23]372116MPC modified FOLFIRINOX10.26.1DPD
312011LAPC modified FOLFIRINOX26.617.8

Table 3.

Clinical trial with neutral outcomes on pancreatic cancer.

5.3 Estimation of clinical trials with the failed outcome on advanced pancreatic cancer

Regarding the failed outcomes, there are 14 studies ranged from 2011 to 2018, recruited 2448 APC patients (1385 male and 1063 female) for clinical trials. For details please see Table 1. Averagely the OS is 8.25 months and PFS is 4.39 months. It is evident that Faivre et al. in 2016 tested sunitinib malate to treat APC and achieved 38.6 months of OS and 12.6 months of PFS. However, Brian reported that hydroxychloroquine cures APC, unfortunately, there were 1.8 months both in OS and PFS negativity. Even though many combination tests, the benefit for APC patient is low.

Advertisement

6. Different perspectives on biomarkers

6.1 Vascular endothelial growth factor, VEGF

VEGF is a highly specific vascular endothelial cell mitogen and an angiogenic factor associated with platelet-derived growth factor (PDGF) structure. It is also known as vascular permeability factor (VPF), due to its permeabilization of blood vessels [24]. It is a subfamily of growth factors and belongs to a family of platelet-derived growth factors of cystine knot growth factor. VEGF is divided into the following groups: VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E and placental growth factor. VEGF binds to three transmembrane receptors (VEGFR1, VEGFR2 and VEGFR3). This receptor initiates downstream signaling through intracellular tyrosine kinase activity [25]. In fact, VEGF family members are playing an important role in the physiological angiogenesis of adults. Like wound healing, ovulation and pregnancy [26].

Activation of the VEGF/VEGF receptor (VEGFR) axis may trigger multiple signaling networks. Consequently, this may lead to endothelial cell survival, mitosis, migration and differentiation, vascular permeability and mobilization of endothelial progenitor cells (EPCs) from the bone marrow into the peripheral circulation [27]. On ligand binding, VEGFR-2 dimerization results in kinase activation and autophosphorylation of tyrosine residues. Activation of PKC may stimulate the Raf/MEK/ERK pathway, which accelerates the cell proliferation. Ca2+ mobilization and PKC activation are playing the key role in signaling events for VEGF-A-induced vascular permeability through activation of endothelial nitric oxide synthase activity [28].

The sword has double sides. VEGF participates in the pathogenesis of cancer, proliferative retinopathy and rheumatoid arthritis [29]. Its antibodies have shown therapeutic potential to inhibit tumor growth in vivo by inhibiting tumor-induced angiogenesis [30]. VEGF overexpression is associated with a variety of tumor progression and poor prognosis, including colorectal cancer [31], pancreatic cancer [32], gastric cancer [33], breast cancer [34], lung cancer [35], prostate cancer [36] and melanoma [37]. This unique protein aids tumors grow and can be used for cancer treatment if used properly.

Early in 1971, Folkman first proposed the idea of angiogenesis. He believed that tumor growth and proliferation are closely related to angiogenesis and could be used as a targeted tumor therapy procedure [38]. Subsequently, countless scientists gathered in the field of VEGF to study the molecular mechanism of VEGF in tumor angiogenesis. They have also used it as a drug target to block the formation of blood vessels, thereby inhibiting tumor growth [39]. Studies have shown that the mechanism of anti-VEGF inhibitors may involve a variety of signaling pathways, such as FGF, D114, PGF/VEGFR1 and VEGF-C/VEGFR2. At least some of these pathways can increase the efficacy of VEGF inhibitors [40]. The anti-tumor drugs were developed with VEGF as the main target area. Like anti-VEGF humanized monoclonal antibody, VEGF-targeted antibody, protein kinase inhibitor and tumor vaccine [41, 42]. However, in the clinical application of anti-antigenic drugs, reliable biomarkers have not been found to screen the target population before patient improvement.

Our data disclosed that there were two studies reporting successful outcomes [14, 19], two studies reported neutral outcomes [21, 22] and four studies reported negative outcomes [43, 44, 45, 46]. The results from the above eight trials remain controversial.

Regarding successful outcomes, Raymond et al. found that neuroendocrine tumors may be particularly sensitive to the combined inhibition of VEGFR and PDGFR. As for the neutral outcomes [14], Spano et al. used the gemcitabine + axitinib to treat the APC [21]. However, results differ from the results of Phase III trials in which erlotinib + gemcitabine confers the greatest survival advantage for patients with ECOG status 2 and metastatic disease, it possibly due to the mechanism of action between different EGFR and VEGFR inhibitors. Moreover, Hobday et al. found that the median PFS in the gefitinib trial was only 3.7 months which was even lower than the placebo group in the phase III trial [22].

With respect to the failed outcomes, Kindler et al. found that the effect of axitinib and gemcitabine on APC was limited to improve the survival period in patients with locally APC [43]. In fact, the results may be related to the gene locus of the VEGF receptor 1 tyrosine kinase domain. Furthermore, Ropugier et al. found that PFS was not significantly improved between the treatment arms. It indicates that blocking the VEGF/VEGFR axis does not lead to the survival of a patient with APC [45]. Nooan et al. study shows that pelareorep combination chemotherapy is not a sufficient solution to overcome the severe immunosuppression prevalent in PCA patients [46].

6.2 Epidermal growth factor receptor, EGFR

EGFR is one of the transmembrane receptors of epidermal growth factor family members of extracellular protein ligands. Its main function is to regulate various cellular functions including proliferation, movement and differentiation. Its mechanism can be described as the binding of the ligand to EGFR leading to dimerization followed by autophosphorylation of EGFR and activation of downstream signaling pathways. Activation of EGFR triggers multiple signal cascades within the cell, ultimately leading to gene transcription and biological responses [47]. Recently, studies have shown that dimerization occurs even in the absence of ligands, particularly when EGFR is overexpressed, possibly limited to a subset of dimers. Moreover, overexpressed EGFR can dimerize and become tyrosine phosphorylated without ligand [48].

EGFR signal transduction pathway can directly participate in tumor pathogenesis and progression [49]. EGFR overexpression plays a major role in carcinogenesis in cancer development [50]. Overexpression of EGFR accounts for 90% in pancreatic cancer cells [51]. Specifically, the aberrant activity of EGFR may impact the development and growth of tumor cells [52].

Mutation of EGFR may induce the resistance of tyrosine kinase inhibitors (TKIs) [53]. Notably, in tumor angiogenesis, vascular endothelial growth factor receptor-2 (VEGFR-2) plays a key role, and inhibition of VEGFR-2 signaling pathway has become an attractive cancer treatment method [54]. The binding of VEGF to VEGFR-2 stimulates the signaling pathway (PI3K/Akt, p38MAPK) that mediates several cellular functions. Besides, glycoproteins, EGFR and VEGFR-2 are in close correlation. Inhibition of EGFR can also reduce VEGF expression, while VEGFR-2 targeting can enhance the anticancer activity of EGFR inhibitors. Therefore, the dual inhibition treatment of EGFR and VEGFR-2 has a good effect and represents a promising cancer treatment. Recently, several EGFR/VEGFR-2 dual inhibitors have been discovered, such as vandetanib showing effective inhibitory activity on EGFR and VEGFR-2 [55].

A receptor tyrosine kinase is associated with cell proliferation and survival. Epidermal Growth Factor Receptor, EGFR, is overactive in many epithelial-derived tumors. It has been reported that EGFR is not related to its kinase activity, but rather maintains basal intracellular glucose levels to prevent autophagic death of cells. Despite the presence of chemotherapeutic drugs and tyrosine kinase inhibitors, this function of EGFR allows tumor cells to have higher viability [56]. EGFR inhibitors for cancer therapy are rapidly evolving in the broad context of cancer therapy, and in those patients achieving significant tumor response to EGFR inhibitors. Most patients will eventually exhibit disease progression, suggesting acquired resistance. This reminds us that increasing our ability to recognize tumors that depend on EGFR signaling growth is critical for the best choice of treating patients [57].

Unfortunately, two clinical trials showed that the results failed via EGFR. Studies by Ko et al. showed that subjects treated by cetuximab, bevacizumab and gemcitabine had 5.41 months in OS and 4.17 months in PFS. Propper et al. in 2014 tested Erlotinib to treat APC, and only achieved 4.0 months of OS and 1.5 months of PFS, Table 4 [44, 58]. Ko reported the incidence of severe (grades 3–5) toxicity, comparable to the use of gemcitabine as a single agent in this patient population which may reflect a relatively short duration of treatment due to lack of efficacy. Moreover, it is difficult to explain the quality of life analysis based on the number of patients completing the series of questionnaires and the overall time the patients stayed in the study [44]. Propper concluded that there is limited evidence to support the use of predictive biomarkers for patients with pancreatic cancer who could benefit from targeted therapies [58].

YearAuthorNMFDrugOSPFSBiomarkers
2011Kindler [43]314191123Gemcitabine; AG-0137368.54.4VEGF
316188128Gemcitabine; placebo8.34.4
2012Ko [44]291811Cetuximab; bevacizumab
gemcitabine
5.414.17EGFR
VEGF
291415Cetuximab; bevacizumab3.551.91
2013Rougier [45]275157118Placebo; gemcitabine7.83.7VEGF
271160111Aflibercept; gemcitabine6.53.7
2013Wu [61]301614Etanercept; gemcitabine5.430.3TNF-α
835Gemcitabine8.11.8
2014Propper [58]1045945Erlotinib4.01.5EGFR
1035944Placebo3.11.5
2014Infante [62]803941Gemcitabine; GSK11202128.416.1cfDNA
804634Placebo; gemcitabine6.715.1
2014Wolpin [63]1055Hydroxychloroquine 400 mg1.81.8LC3-II
1064Hydroxychloroquine 600 mg3.01.6
2015Catenacci [64]532726Gemcitabine hydrochloride;
Placebo
6.12.5SHH
533122Gemcitabine hydrochloride;
vismodegib
6.94.0
2016Noonan [46]362214wild-type reovirus; carboplatin; paclitaxel7.314.94VEGF;
IL-6;
IL-8
371918Carboplatin; paclitaxel8.775.2
2017Chung [65]622240Fluorouracil; oxaliplatin6.72.0KRAS
protein
583523Akt inhibitor
MK2206; selumetinib
3.91.9
2017Faivre [66]864244Sunitinib malate38.612.6VEGF;
PDGF
854045Placebo29.15.8
2017Ko [67]663828OGX-4276.93.8Hsp27
663729Placebo5.32.7
2017Laquente [68]654223LY2603618; gemcitabine7.83.5CHK1
342014Gemcitabine8.35.6
2018Van Cutsem [69]442222Gemcitabine; placebo7.62.8ERK 1/2
442717Gemcitabine; pimasertib7.33.7

Table 4.

Clinical trial failed outcome on pancreatic cancer.

6.3 Kirsten rat sarcoma viral oncogene homolog, KRAS

KRAS protein plays a key role in human cancer but has not yet succumbed to therapeutic attacks [59]. The search is now focused on targeting alternative pathways that are activated in mtKRAS cells, to circumvent or prevent drug resistance [60].

There are currently no therapeutic interventions for KRAS. Pharmacological agents that are speculated to inhibit KRAS include farnesyltransferase inhibitors that block the binding of KRAS to the cell membrane. These drugs have failed in clinical studies. Antisense oligonucleotides and engineered microRNAs (miRNAs) have been used as an alternative to targeted mutations in KRAS without disrupting the expression of non-mutant KRAS and have achieved some success in preclinical trials [70].

In recent years, many studies have suggested that the oncogene KRAS plays a major role in controlling cancer metabolism by coordinating multiple metabolic changes [71]. Furthermore, combined inhibition of therapeutic effects and feedback pathways is promising in KRAS mutant cancers. Moreover, it is unclear what specific pathways should be used to optimize treatment response [72].

Unfortunately, Chung et al. tested pharmaceutical target via KRAS protein, whereas the result was unsuccessful. The results showed that the OS and PFS of patients treated by Akt Inhibitor MK2206 and selumetinib were 3.9 and 1.9 months, respectively, see Table 4 [65]. The results indicated that the strategy of utilizing two or more kinase inhibitors is subject to the challenge of toxicity overlap. These toxicities will significantly block the delivery of effective inhibitory amounts of both drugs in vivo. However, a major disadvantage factor is the delay in toxicity-related treatment and the frequency of dose reduction in the experimental group through damaging sustained signal suppression [65].

Advertisement

7. Conclusion

APC reserves unpredictable mechanisms to maintain a highly resistant phenotype. The genetic and epigenetic alterations of the APC lead to the resistance of the chemotherapy.

Nowadays, many biomarkers have been on board to improve the clinical treatment outcome of advanced pancreatic cancer. Although these successful biomarkers have provided notable therapeutic effects on advanced pancreatic cancer, the outcomes remain unsatisfactory to the patients and health providers. With the development of the biology of advanced pancreatic cancer, we now expect better biomarkers and conduct therapy by unveiling the tumor microenvironment and the mechanism of the mutations (Figure 1).

Figure 1.

Difficulties in decision making both for R&D and doctors.

We can assume that with the development of truly effective treatments and clinically useful markers for early detection of the disease, better combination of markers to advanced pancreatic cancer. In the meanwhile, researchers are trying to detect magnificently predictive biomarkers to decide the treatment strategy and permit practitioners to adequately evaluate and propose individualized treatment protocols which would give a greater survival rate.

Clearly, there is a need to better understand the underlying signaling networks that drive pancreatic cancer progression and potential escape mechanisms. In addition, it is necessary to improve the role of preclinical models of pancreatic cancer and the optimal transformation of preclinical success into experimental design. The genetic and proteomic technologies show great potential to detect the novel biomarkers in cancer research. We place great expectations on these technologies to personalize treatment for advanced pancreatic cancer patients.

Advertisement

Acknowledgments

This work was supported by the Chengdu Medical College Natural Science Foundation (CYZ18-08, CYZ18-20, CYZ18-33), the Sichuan Provincial Education Department (17ZA0134), the Sichuan Medical Association (S18023), and Sichuan undergraduate innovation and startup program funding support (S201913705080, S201913705130, S201913705059).

Advertisement

Authors’ contribution

XF developed the general idea and drafted the manuscript with CJ; YLQ and DYX searched the reference, HK sort the clinical trials data, PTT and ZS participated in drafting process and double check the data; LFF drew the figure; GM proofread the manuscript and ensure the general quality.

Advertisement

Competing interests

The authors declare that they have no competing interests.

Ethics statement

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

All data will be made available upon reasonable request.

References

  1. 1. Kamisawa T, Wood LD, Itoi T, et al. Pancreatic cancer. Lancet. 2016;388:73-85
  2. 2. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;68:394-424
  3. 3. Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. International Journal of Cancer. 2019;144:1941-1953
  4. 4. Chen J, Chen L, Yu J, et al. Metaanalysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatmentassociated toxicities. Molecular Medicine Reports. 2019;19:477-489
  5. 5. Drews J. Paul Ehrlich: magister mundi. Nature Reviews. Drug Discovery. 2004;3:797-801
  6. 6. Prindle MJ, Fox EJ, Loeb LA. The mutator phenotype in cancer: Molecular mechanisms and targeting strategies. Current Drug Targets. 2010;11:1296-1303
  7. 7. Andrade EL, Bento AF, Cavalli J, et al. Non-clinical studies required for new drug development - part I: Early in silico and in vitro studies, new target discovery and validation, proof of principles and robustness of animal studies. Brazilian Journal of Medical and Biological Research. 2016;49:e5644
  8. 8. Katara P. Role of bioinformatics and pharmacogenomics in drug discovery and development process. Network Modeling and Analysis in Health Informatics and Bioinformatics. 2013;2:225-230
  9. 9. Xia HJ, Chen CS. Progress of non-human primate animal models of cancers. Dongwuxue Yanjiu. 2011;32:70-80
  10. 10. Ballman KV. Introduction to clinical trials, clinical trial designs, and statistical terminology used for predictive biomarker research and validation. In: Badve S, Kumar GL, editors. Predictive Biomarkers in Oncology: Applications in Precision Medicine. Cham: Springer International Publishing; 2019. pp. 19-36
  11. 11. Bharali D, Banerjee BD, Bharadwaj M, et al. Expression analysis of MicroRNA-21 and MicroRNA-122 in hepatocellular carcinoma. Journal of Clinical and Experimental Hepatology. 2019;9:294-301
  12. 12. Hill ME, Li X, Kim S, et al. A phase I study of the biomodulation of capecitabine by docetaxel and gemcitabine (mGTX) in previously untreated patients with metastatic adenocarcinoma of the pancreas. Cancer Chemotherapy and Pharmacology. 2011;67:511-517
  13. 13. Lutz E, Yeo CJ, Lillemoe KD, et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A phase II trial of safety, efficacy, and immune activation. Annals of Surgery. 2011;253:328-335
  14. 14. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. The New England Journal of Medicine. 2011;364:501-513
  15. 15. Hosein PJ, de Lima Lopes G Jr, Pastorini VH, et al. A phase II trial of nab-paclitaxel as second-line therapy in patients with advanced pancreatic cancer. American Journal of Clinical Oncology. 2013;36:151-156
  16. 16. Soares HP, Bayraktar S, Blaya M, et al. A phase II study of capecitabine plus docetaxel in gemcitabine-pretreated metastatic pancreatic cancer patients: CapTere. Cancer Chemotherapy and Pharmacology. 2014;73:839-845
  17. 17. Ban H, Kim KS, Oh IJ, et al. Efficacy and safety of docetaxel plus oxaliplatin as a first-line chemotherapy in patients with advanced or metastatic non-small cell lung cancer. Thoracic Cancer. 2014;5:525-529
  18. 18. Borad MJ, Reddy SG, Bahary N, et al. Randomized phase II trial of gemcitabine plus TH-302 versus gemcitabine in patients with advanced pancreatic cancer. Journal of Clinical Oncology. 2015;33:1475-1481
  19. 19. Phan AT, Halperin DM, Chan JA, et al. Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: A multicentre, single-group, phase 2 study. The Lancet Oncology. 2015;16:695-703
  20. 20. Wang-Gillam A, Hubner RA, Siveke JT, et al. NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. European Journal of Cancer. 2019;108:78-87
  21. 21. Spano JP, Chodkiewicz C, Maurel J, et al. Efficacy of gemcitabine plus axitinib compared with gemcitabine alone in patients with advanced pancreatic cancer: An open-label randomised phase II study. Lancet. 2008;371:2101-2108
  22. 22. Hobday TJ, Qin R, Reidy-Lagunes D, et al. Multicenter phase II trial of temsirolimus and bevacizumab in pancreatic neuroendocrine tumors. Journal of Clinical Oncology. 2015;33:1551-1556
  23. 23. Stein SM, James ES, Deng Y, et al. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. British Journal of Cancer. 2016;114:737-743
  24. 24. Neufeld G, Tessler S, Gitay-Goren H, et al. Vascular endothelial growth factor and its receptors. Progress in Growth Factor Research. 1994;5:89-97
  25. 25. Stewart W, Vascular Endothelial M. Growth factor (VEGF) biochemistry and development of inhibitory drugs. Current Drug Therapy. 2012;7:80-89
  26. 26. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. Journal of Clinical Oncology. 2005;23:1011-1027
  27. 27. Koch S, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harbor Perspectives in Medicine. 2012;2:a006502-a006502
  28. 28. Cross MJ, Dixelius J, Matsumoto T, et al. VEGF-receptor signal transduction. Trends in Biochemical Sciences. 2003;28:488-494
  29. 29. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. The New England Journal of Medicine. 1994;331:1480-1487
  30. 30. Kim KJ, Li B, Winer J, et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature. 1993;362:841-844
  31. 31. Lee JC, Chow NH, Wang ST, et al. Prognostic value of vascular endothelial growth factor expression in colorectal cancer patients. European Journal of Cancer. 2000;36:748-753
  32. 32. Fujimoto K, Hosotani R, Wada M, et al. Expression of two angiogenic factors, vascular endothelial growth factor and platelet-derived endothelial cell growth factor in human pancreatic cancer, and its relationship to angiogenesis. European Journal of Cancer. 1998;34:1439-1447
  33. 33. Takahashi Y, Cleary KR, Mai M, et al. Significance of vessel count and vascular endothelial growth factor and its receptor (KDR) in intestinal-type gastric cancer. Clinical Cancer Research. 1996;2:1679-1684
  34. 34. Berns EM, Klijn JG, Look MP, et al. Combined vascular endothelial growth factor and TP53 status predicts poor response to tamoxifen therapy in estrogen receptor-positive advanced breast cancer. Clinical Cancer Research. 2003;9:1253-1258
  35. 35. Fontanini G, Lucchi M, Vignati S, et al. Angiogenesis as a prognostic indicator of survival in non-small-cell lung carcinoma: A prospective study. Journal of the National Cancer Institute. 1997;89:881-886
  36. 36. George DJ, Halabi S, Shepard TF, et al. Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on cancer and leukemia group B 9480. Clinical Cancer Research. 2001;7:1932-1936
  37. 37. Gorski DH, Leal AD, Goydos JS. Differential expression of vascular endothelial growth factor-a isoforms at different stages of melanoma progression. Journal of the American College of Surgeons. 2003;197:408-418
  38. 38. Folkman J. Tumor angiogenesis: Therapeutic implications. The New England Journal of Medicine. 1971;285:1182-1186
  39. 39. Chou HF, Lin MF, Chen CY, et al. Three-dimensional brain images in preterm children with periventricular leukomalacia. Pediatrics and Neonatology. 2012;53:45-48
  40. 40. Ferrara N. Pathways mediating VEGF-independent tumor angiogenesis. Cytokine & Growth Factor Reviews. 2010;21:21-26
  41. 41. Semenza GL. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene. 2010;29:625-634
  42. 42. Hoeben A, Landuyt B, Highley MS, et al. Vascular Endothelial growth factor and angiogenesis. Pharmacological Reviews. 2004;56:549
  43. 43. Kindler HL, Ioka T, Richel DJ, et al. Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: A double-blind randomised phase 3 study. The Lancet Oncology. 2011;12:256-262
  44. 44. Ko AH, Youssoufian H, Gurtler J, et al. A phase II randomized study of cetuximab and bevacizumab alone or in combination with gemcitabine as first-line therapy for metastatic pancreatic adenocarcinoma. Investigational New Drugs. 2012;30:1597-1606
  45. 45. Rougier P, Riess H, Manges R, et al. Randomised, placebo-controlled, double-blind, parallel-group phase III study evaluating aflibercept in patients receiving first-line treatment with gemcitabine for metastatic pancreatic cancer. European Journal of Cancer. 2013;49:2633-2642
  46. 46. Noonan AM, Farren MR, Geyer SM, et al. Randomized phase 2 trial of the oncolytic virus Pelareorep (Reolysin) in upfront treatment of metastatic pancreatic adenocarcinoma. Molecular Therapy. 2016;24:1150-1158
  47. 47. Guo G, Gong K, Wohlfeld B, et al. Ligand-independent EGFR signaling. Cancer Research. 2015;75:3436-3441
  48. 48. Chung I, Akita R, Vandlen R, et al. Spatial control of EGF receptor activation by reversible dimerization on living cells. Nature. 2010;464:783-787
  49. 49. Habban Akhter M, Sateesh Madhav N, Ahmad J. Epidermal growth factor receptor based active targeting: A paradigm shift towards advance tumor therapy. Artificial Cells, Nanomedicine, and Biotechnology. 2018;46:1188-1198
  50. 50. Chakraborty S, Li L, Puliyappadamba VT, et al. Constitutive and ligand-induced EGFR signalling triggers distinct and mutually exclusive downstream signalling networks. Nature Communications. 2014;5:5811
  51. 51. Tuan Anh HL, Tran PT, Thao DT, et al. Degalactotigonin, a steroidal glycoside from Solanum nigrum, induces apoptosis and cell cycle arrest via inhibiting the EGFR signaling pathways in pancreatic cancer cells. BioMed Research International. 2018;2018:3120972
  52. 52. Seshacharyulu P, Ponnusamy MP, Haridas D, et al. Targeting the EGFR signaling pathway in cancer therapy. Expert Opinion on Therapeutic Targets. 2012;16:15-31
  53. 53. Wu K, Guo C, Li R. Clinical characterization of icotinib-induced chemoresistance in erlotinib-treated lung adenocarcinoma patient with EGFR mutations: A case report. Medicine (Baltimore). 2019;98:e15489
  54. 54. Sun W, Hu S, Fang S, et al. Design, synthesis and biological evaluation of pyrimidine-based derivatives as VEGFR-2 tyrosine kinase inhibitors. Bioorganic Chemistry. 2018;78:393-405
  55. 55. Mghwary AE, Gedawy EM, Kamal AM, et al. Novel thienopyrimidine derivatives as dual EGFR and VEGFR-2 inhibitors: Design, synthesis, anticancer activity and effect on cell cycle profile. Journal of Enzyme Inhibition and Medicinal Chemistry. 2019;34:838-852
  56. 56. Weihua Z, Tsan R, Huang WC, et al. Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell. 2008;13:385-393
  57. 57. Wheeler DL, Dunn EF, Harari PM. Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nature Reviews. Clinical Oncology. 2010;7:493-507
  58. 58. Propper D, Davidenko I, Bridgewater J, et al. Phase II, randomized, biomarker identification trial (MARK) for erlotinib in patients with advanced pancreatic carcinoma. Annals of Oncology. 2014;25:1384-1390
  59. 59. McCormick F. KRAS as a therapeutic target. Clinical Cancer Research. 2015;21:1797-1801
  60. 60. Charitou T, Srihari S, Lynn MA, et al. Transcriptional and metabolic rewiring of colorectal cancer cells expressing the oncogenic KRAS(G13D) mutation. British Journal of Cancer. 2019;121:37-50
  61. 61. Wu C, Fernandez SA, Criswell T, et al. Disrupting cytokine signaling in pancreatic cancer: A phase I/II study of etanercept in combination with gemcitabine in patients with advanced disease. Pancreas. 2013;42:813-818
  62. 62. Infante JR, Somer BG, Park JO, et al. A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. European Journal of Cancer. 2014;50:2072-2081
  63. 63. Wolpin BM, Rubinson DA, Wang X, et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. The Oncologist. 2014;19:637-638
  64. 64. Catenacci DV, Junttila MR, Karrison T, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. Journal of Clinical Oncology. 2015;33:4284-4292
  65. 65. Chung V, McDonough S, Philip PA, et al. Effect of selumetinib and MK-2206 vs oxaliplatin and fluorouracil in patients with metastatic pancreatic cancer after prior therapy: SWOG S1115 study randomized clinical trial. JAMA Oncology. 2017;3:516-522
  66. 66. Faivre S, Niccoli P, Castellano D, et al. Sunitinib in pancreatic neuroendocrine tumors: Updated progression-free survival and final overall survival from a phase III randomized study. Annals of Oncology. 2017;28:339-343
  67. 67. Ko AH, Murphy PB, Peyton JD, et al. A randomized, double-blinded, phase II trial of gemcitabine and nab-paclitaxel plus apatorsen or placebo in patients with metastatic pancreatic cancer: The RAINIER trial. The Oncologist. 2017;22:1427-e1129
  68. 68. Laquente B, Lopez-Martin J, Richards D, et al. A phase II study to evaluate LY2603618 in combination with gemcitabine in pancreatic cancer patients. BMC Cancer. 2017;17:137
  69. 69. Van Cutsem E, Hidalgo M, Canon JL, et al. Phase I/II trial of pimasertib plus gemcitabine in patients with metastatic pancreatic cancer. International Journal of Cancer. 2018;143:2053-2064
  70. 70. Gober MK, Flight RM, Lambert J, et al. Deregulation of a network of mRNA and miRNA genes reveals that CK2 and MEK inhibitors may synergize to induce apoptosis KRAS-active NSCLC. Cancer Informatics. 2019;18:1176935119843507
  71. 71. Kawada K, Toda K, Sakai Y. Targeting metabolic reprogramming in KRAS-driven cancers. International Journal of Clinical Oncology. 2017;22:651-659
  72. 72. Anderson GR, Winter PS, Lin KH, et al. A landscape of therapeutic cooperativity in KRAS mutant cancers reveals principles for controlling tumor evolution. Cell Reports. 2017;20:999-1015

Written By

Jie Chen, Liqiong Yang, Yuxi Duan, Tinging Pu, Sha Zheng, Fangfang Liu, Kun Huang, Greg Mirt and Fan Xu

Submitted: 08 July 2019 Reviewed: 30 September 2019 Published: 31 October 2019