GET THE APP

Targeting the PI3K/AKT Pathway for the Treatment of Gastric Cance
Chemotherapy: Open Access

Chemotherapy: Open Access
Open Access

ISSN: 2167-7700

+44 1223 790975

Review Article - (2014) Volume 3, Issue 1

Targeting the PI3K/AKT Pathway for the Treatment of Gastric Cancer

Yuyan Liu1, Sunrong Sun2, Juanjuan Li2 and De-Hua Yu1,2*
1Hygeianey Bioscience CO, LTD, Building A82-1, NO. 858 Gaoxin Boulevard, Biomedical Park Phase II, East Lake High-Tech Development Zone 430076, Wuhan, China
2Wuhan University Renmin Hospital, Wuhan, China
*Corresponding Author: De-Hua Yu, Hygeianey Bioscience CO, LTD, Building A82-1, NO. 858 Gaoxin Boulevard, Biomedical Park Phase II, East Lake High-Tech Development Zone 430076, Wuhan, China, Tel: 86-138-165-849 Email:

Abstract

Gastric Cancer (GC) is the fourth most common malignancy and the second leading cause of cancer deaths,accounting for 10% of global cancer mortalities. Despite the progress made in recent years, the prognosis forpatients with advanced-stage GC remains poor. In the metastatic setting, chemotherapy is the primary choice forpalliative therapy and results in Objective Response Rates (ORRs) of only 20-40% and median Overall Survivals(OSs) of 8–10 months. Emerging evidence suggests that the aberrant activation of phosphatidylinositol 3-kinase(PI3K)/AKT signaling is one of the most common molecular events involved in the resistance to current systemictherapies for GC.A number of small molecule inhibitors targeting the PI3K/AKT pathway are currently under clinicalevaluation for the treatment of various malignancies, including GC. In this paper, we review the current clinicalpractice and discuss the potential use of inhibitors targeting the PI3K/AKT pathway, either alone or in combinationwith current therapies for the treatment of advanced GC.

Keywords: Cancer stem cells; Chemoresistance; Targeted therapies

Introduction

Gastric Cancer (GC) is one of the most common malignancies and the second leading cause of cancer deaths [1]. The estimated global incidence and mortality for GC were 990,000 and 737,000, respectively, in 2011, which accounted for approximately 8% of total cancer cases and 10% of annual cancer deaths worldwide [2,3].Geographically, GC is more prevalent in developing countries, especially those in Eastern Asia, Central and Eastern Europe, and South America, than in the rest of world. The current treatments for GC include surgery, chemotherapy,radiotherapy, and targeted therapy against the HER-2-positive segment n GC [4]. Although the prognosis for GC has been improved significantly in certain nations, such as Japan and Korea, with national early screening, the five-year survival rate remains poor. The genome-wide profiling of genetic aberrations in primary tumor samples from GC patients revealed a number of potential targets, such as FGFR, Met, and PI3K, for the development of next-generation therapies for GC [5]. Selective inhibitors of these potential targets are being developed at various clinical stages for the treatment of cancers, including GC. In this paper, we review the current clinical practice for the systemic treatment of advanced GC, the role of the activation of the PI3K/AKT pathway, and the potential of targeting the PI3K/AKT pathway for treatment of advanced GC.

Current Treatments for Advanced GC

The treatment of GC requires multidisciplinary approaches that incorporate surgery, chemotherapy, targeted therapy, and radiotherapy. For patients with operable tumors, surgery and chemotherapy remain the primary curative treatments. Although the 5-year survival rate has been improved in certain nations, such as Japan (57%) and Korea (64.3%), it still remains poor globally, ranging between 20 and 25% [6,7].

In the United States, patients with GC are often diagnosed at advanced stages due to lack of routine endoscopic screening. Overall, 5-year survival rates are less than 10% [8]. The implementation of additional combined strategies, including neoadjuvant and adjuvant therapy (pre- or postoperative chemo/radiotherapy or perioperative chemotherapy), led to 5-year survival rates of only 30-35% and pathologically Complete Responses (pCRs) in no more than 20-30% of patients [8-10]. In metastatic GC, chemotherapy is the mainstay of palliative therapy and results in objective response rates (ORRs) of only 20–40% and a median Overall Survival (OS) of 8–10 months [11]. The recurrence of GC after undergoing surgical treatment has been reported in approximately 45% of cases in western countries and in about 22% of cases in Korea and Japan [12].

Although no standard adjuvant or palliative chemotherapy regimen has been internationally approved for patients with advanced GC, a number of chemo agents have been widely used alone or in combination as the first-line therapy, with demonstrated benefits. These include fluoropyrimidine (5-FU, S-1, or capecitabine), platinum (cisplatin or oxalipatin), taxane (docetaxel or paclitaxel), epirubicin, and irinotecan. A systemic meta-analysis based on aggregate data revealed that in patients with advanced GC, palliative chemotherapy is more beneficial in terms of the improvement of OS and the relief of symptoms than treatment with the best supportive care (BSC) [13]. The combination of docetaxel/cisplatin/5-FU (DCF), ECF, and fluoropyrimidine (5-FU or capecitabine), as well as cisplatin, as a category 1 treatment has been recommended by the NCCN guidelines. The response rates to this combination regimen range from 25% to 54%, suggesting intrinsic resistance to these therapies [13].

In January of 2010, trastuzumab was approved as the first targeted therapy for the treatment of patients with HER2-positive GC. The HER family consists of four members: HER-1 (epidermal growth factor receptor (EGFR), HER-2, HER-3, and HER-4. HER-1, HER-3, and HER-4 are all activated via ligand binding, whereas HER-2 does not require a ligand for activation. The activation of these receptors triggers phosphorylation cascades and the subsequent activation of a number of signaling transducers, thus activating both the PI3K/AKT and Ras/Raf pathways, which are important in cancer cell proliferation and survival [14,15]. Trastuzumab is a humanized recombinant monoclonal antibody that selectively binds to the extracellular domain of HER-2, thereby blocking its downstream signaling.

HER-2 amplification or over-expression is observed in about 15% to 25% of GC cases [16]. HER-2 over-expression is more common in intestinal-type and gastroesophageal junction (GEJ) tumors than in diffuse-type and gastric tumors [16,17]. The Phase III ToGA trial found that 22% of advanced and metastatic GC patients overexpressed HER-2 via an immunohistochemistry (IHC) and/or fluorescence in situ hybridization (FISH) methodology. The addition of trastuzumab to chemotherapy also led to a significantly higher ORR, 47% versus 35% (p=0017); significantly longer progression-free survival (PFS) intervals, 6.7 months versus 5.5 months (p=0002); and significantly longer OS duration, 13.8 months versus 11.1 months (p=0046) [18]. In Korea, a combination therapy of trastuzumab and CF is recommended in patients with HER-2-over-expressing adencarcinoma. The greatest benefit was seen in patients with high levels of HER-2 expression. To date, trastuzumab is the first and only targeted agent for GC approved by both U.S. and European authorities. However, a significant proportion patients withHER-2 positive breast cancer either do not respond or eventually become resistant to trastuzumab [19], suggesting the existence of both intrinsic and acquired resistance.

The PI3K/AKT Pathway

PI3K is a family of intracellular lipid kinases involved in the signaling network that regulates cell survival, proliferation, differentiation, migration, and metabolism [20]. PI3Ks can be categorized into three classes (I–III) according to their substrate preferences and sequence homologies. The activation of receptor tyrosine kinases (RTKs), such as EGFR, IGFR, and HER2, activates class IA PI3Ks. The binding of the p85 regulatory subunit of PI3K to phosphotyrosine residues on activated RTKs leads to a conformational change in p85, releasing the inhibition of the catalytic subunit p110 (a, ß, and d isoforms) of PI3K.PI3K localizes to the plasma membrane and catalyzes the formation of phosphatidylinositol 3,4,5-trisphosphate (PIP3) through the phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP2). PIP3 is a critical activator of the serine/threonine kinase AKT (also known as protein kinase B). The binding of PIP3 to AKT leads to the membrane recruitment of AKT and its subsequent phosphorylation by PDK1 (3-phosphoinositide-dependent kinase) and PDK2 [20,21]. Activated AKT translocates to the cytoplasm and nucleus and activates the downstream targets involved in cell survival, proliferation, cell cycle progression, growth, migration, and angiogenesis [21,22] (Figure 1). PIP3 levels are tightly controlled by stringent PI3K regulation and via PTEN PIP3 phosphatase and SHIP (SH2-containing inositol5-phosphatase), which converts PIP3 back to phosphatidylinosito 4, 5-bisphosphate.

chemotherapy-de-regulated-pathway

Figure 1: PI3K/AKT signaling pathwayAKT is a critical node on the most commonly de-regulated pathway in human cancer. AKT signaling can be activated by growth factor receptor tyrosine kinases signaling via RAS and PI3K, or by mutations in AKT1 and PI3KCA or loss of PTEN.

Cell apoptosis is a normal function that controls excessive proliferation. Cancer cells utilize a variety of mechanisms to down-regulate cell apoptosis and prolong survival. An important function of the activated PI3K/AKT pathway in cells is the inhibition of apoptosis. AKT is a good candidate for mediating these PI3K-dependent cell-survival responses. A large number of AKT's direct downstream substrates have been identified, which include the pro-apoptotic Bcl-2 family members Bad, Bax, caspase-9, and GSK-3 and the forkhead family transcription factors (FoxO1).Activated AKT inhibits Bad's function through direct phosphorylation and suppresses the expression of Bim and FasL via the indirect phosphorylation of FoxO1 [23]. In addition, it is reported that AKT can phosphorylate iKKs, which promote cIAP expression for the inhibition of cell apoptosis. Recently, Jeong et al. proposed that AKT has a role in the activation of pro-survival pathways, possibly through NF-kB activation and the inhibition of p53 transcription activity [24].

Activation of the PI3K/AKT Pathway in GC

The dysregulation of the PI3K/AKT/mTOR pathway is a common phenomenon in many human cancers, including GC [25]. It can be triggered by a variety of mechanisms, including PI3KCA-activating mutations, loss of PTEN function, overexpression or activating mutations of AKT, and the overexpression of upstream receptors, such as IGFR, EGFR, and HER-2 [26]. PI3KCA is a gene that encodes for the p110-alpha subunit of PI3K.PI3KCA-activating mutations occur in exome 9 and 20 of the PI3K gene and have been reported in GC by several groups. Mutation frequencies range from 10.6% to 15.9% [27,28] in Caucasian and 4.3% to 7.1% in Asian GC cases [29,30]. Recently, we screened 127 GC samples from Chinese patients and detected PI3KCA hotspot mutations in 2.7% of cases (4/127) [31]. This apparent discrepancy between the western and eastern populations may be due to geographical differences or the sample size, or it could be related to disease stage and genomic instability status. PTEN phosphatase is a novel tumor suppressor that negatively regulates the PI3K/AKT signaling network [32]. Loss of PTEN function can be attributed to inactivating gene mutations, chromosomal deletions, and promoter methylation, which are implicated in multiple cancer types, including GC [33]. In comparison to PI3KCA mutations, the lack of detectable PTEN protein expression via IHC staining was more frequently observed in GC cases, with reported rates of between 20 and 36% [34,35]. Consistent with these observations, our recent data indicated that the loss of PTEN protein expression, detected via IHC, was found in 23% (14/61) of the samples from Chinese GC patients [31]. On the other hand, the increased phosphorylation of AKT and mTOR was observed in 80% [36] and 47%-64% of GC patients, respectively [37-39], suggesting that additional factors, such as activation of RTKs (e.g., HER2, IGFR), must be responsible for the activation of the PI3K/AKT pathway.

Activation of the PI3K/AKT Pathway and Resistance to Chemo and Anti-HER2 Therapies

Drug resistance is a major problem for the treatment of metastatic and recurrent GC. The involvement of the PI3K/AKT/mTOR pathway in the resistance to chemotherapies in GC has been documented by several studies [29,40]. When primary tumor tissues from GC patients were tested for their chemotherapeutic sensitivity in vitro, the association between activated AKT and increased resistance to multiple chemotherapeutic agents, including 5-fluorouracil, doxorubicin, mitomycin C, and cisplatin, was found [41]. Consistently, the reduction of basal AKT activity via the ectopic expression of PTEN sensitized GC cells to anti-cancer chemotherapy agents. These data suggest that the activation of the PI3K/AKT pathway has a direct role in resistance to chemotherapy in GC cases. The combination of PI3K/AKT pathway inhibitors with chemotherapy has successfully attenuated chemotherapeutic resistance in GC cell lines [42]. Recently, we tested the anti-tumor activity of a novel AKT kinase inhibitor, AZD5363, in a patient-derived GC xenograft (PDGCX) model with PTEN loss. This indicated that AZD5363 and taxotere monotherapies were ineffective, but significant anti-tumor activity was observed when AZD5363 was combined with taxotere. Similarly, Lin et al. showed that the administration of another AKT kinase inhibitor, GDC-0068, in combination with taxotere induced tumor regression in a PC-3 prostate xenograft model with a homozygous deletion of PTEN [43]. Because the doses used for each single agent only caused modest tumor growth delay in the study, these results suggest a new strategy to sensitize GC with PTEN loss to chemotherapy by targeting the PI3K/AKT pathway [31].

In addition to its involvement in resistance to chemotherapies, the role of the activation of the PI3K/AKT pathway in resistance to anti-HER-2 agents has been well-studied in HER-2-positive breast cancer [31,44]. In a HER-2-positive BT474 model, a combination of AZD5363 and taxotere led to complete tumor regression.A recent study by Linos et al. indicated that PTEN was lost in the majority of HER-2-positive GC cases [45]. These observations provide a possible explanation for the observed clinical resistance of HER-2-positive breast cancer patients to current anti-HER-2 therapies, including trastuzumab and lapatinib.

The mechanisms of the AKT pathway in chemo resistance are multifold.First, activated AKT suppresses apoptotic cell death via the inhibition of cytochrome c's release from the mitochondria or via its regulatory effect on various downstream effectors, e.g., NF-?B, Bcl-2-family proteins, FOXO transcription factors, and MDM2 [23,24]. In addition, AKT activation mediates cell cycle progression through the inhibition of GSK-3, opposing the action of p21WAF1 and p27Kip1, and via the phosphorylation of mTOR kinases [46]. Geng et al. showed that Bcl-2 expression was significantly associated with chemo resistance. The over-expression of Bcl-2 may predict decreased chemotherapy efficacy in patients with GC [47]. Chemotherapy promotes the activation of NFkB, which regulates various genes involved in angiogenesis, metastasis, and the suppression of apoptosis [48]. The PI3K/AKT pathway is involved in the activation of NF-kappa B via tumour necrosis factor [49,50] and plays a role in chemo resistance in GC [51]. The PI3K inhibitor LY294002 was able to decrease the expression of MDR1/Pgp, Bcl2, and XIAP and to up-regulate the expression of Bax and caspase3, thereby enhancing chemo sensitivity by inhibiting a drug pump and inducing apoptosis. These results suggest an approach to enhance chemo sensitivity by direct targeting PI3K in human GC [52].

Agents Targeting the PI3K/AKT Pathway in Clinical Development

A number of inhibitors of PI3K, AKT, and mTOR have being developed and are now at various stages of clinic evaluation (Table 1).Because mTOR is one of the downstream effectors of PI3K and AKT, it is expected that inhibitors of mTOR will have more selective effects and better safety profiles. At present, the mTOR inhibitor everolimus is the only one approved for the treatment of malignancies, including breast cancer, neuroendocrine tumors of pancreatic origin, and subependymal giant cell astrocytoma.

Agents Manufacturer Target Indications Stage
mTOR inhibitors        
Everolimus Novartis mTORC1 RCC, HCC, lymphoma, mBC, and GBM Approved for RCC, PNET and SEGA
Temsirolimus Pfizer mTORC1 Various tumors Approved for CRC
Ridaforolimus Merck mTORC1 Sarcoma, EC, mBC, prostate cancer, Her2+ mBC, NSCLC Phase III
OSI-027 OSI mTORC1/2 Solid tumors and lymphoma Phase I
AZD8055 AstraZeneca mTORC1/2 Gliomas, HCC and solid tumors Phase I
AZD2014 AstraZeneca mTORC1/2 mBC, RCC and solid tumors Phase I/II
INK128 Millennium mTORC1/2 mBC, NHL, MM, and solid tumors Phase I
CC-223 Celgene mTORC1/2 NHL, NSCLC and MM Phase I
DS-30786a Daiichi Sankyo mTORC1/2 Solid tumors and lymphoma Phase I
PI3K inhibitors        
BKM120 Novartis Pan-PI3K Solid tumors, prostate cancer, CRC, ER+ mBC, Her2+ mBC, GBM, hematologic malignancies, NSCLC, thyroid Cancers and EC Phase I/II/III
XL147 Sanofi-aventis Pan-PI3K GBM, EC, OC, NSCLC, Her2+ breast cancer, lymphoma, and solid tumors Phase I/ II
PX866 Oncothyreon Pan-PI3K GBM, NSCLC, CRC, HNSCC, prostate cancer, and solid tumors Phase II
GDC-0941 Genentech Pan-PI3K ER+ mBC, Her2+ mBC, NSCLC, NHL, and solid tumors Phase I/ II
GSK2126458 GSK Pan-PI3K Pulmonary fibrosis, lymphoma and solid tumors Phase I
BAY 80-6946 Bayer Pan-PI3K Solid tumors, lymphoma Phase I/II
BYL719 Novartis PI3K p110α Solid tumors, mBC, CRC, GC Phase I/II
GS-1101 (CAL-101), Gilead PI3K p110δ CLL, NHL, and hematologic malignancies Phase II
AMG319 Amgen PI3K p110δ CLL, NHL, and hematologic malignancies Phase I
BEZ235 Novartis PI3K/mTOR RCC, prostate cancer, Leukemia , solid tumors and mBC Phase I/II
XL765 Sanofi-aventis PI3K/mTOR GBM, BC, NSCLC, and solid tumors PhaseIb/II
GDC-0980 Genentech PI3K/mTOR mBC, NHL, RCC,EC, prostate cancer and solid tumors Phase I/ II
SF1126 Semafore PI3K/mTOR Solid tumors Phase I
AKT inhibitors        
Perifosine (KRX-0401) Keryx AKT CRC, MM, glioma, hematologic malignancies, lymphoma, OC, BC, NSCLC, RCC, sarcoma, and solid tumors Phase II/III
AZD5363 AstraZeneca AKT BC, OC, CC, prostate cancer, solid tumors Phase I/II
GDC-0068 Genentech AKT Prostate cancer, GC, Solid tumors Phase I/II
MK2206 Merck AKT BC, pancreatic cancer, GC, NSCLC, lymphoma, solid tumors Phase II
RX-0201 Rexahn AKT Pancreatic cancer Phase II
GSK2110183 GSK AKT MM, OC, CLL, hematologic malignancies, and Langerhans cell histiocytosis Phase I/II
GSK2141795 GSK AKT CC, CRC, melanoma, OC, lymphoma and solid tumors Phase I/II
LY2780301 Eli Lily P70S6K/AKT BC, NHL Phase I/II
PBI-05204 Phoenix Biotechnology AKT Solid tumors Phase I
VD-0002 VioQuest AKT Solid tumors and hematologic malignancies Phase I
BAY1125976 Bayer AKT Solid tumors Phase I
SR13668 NCI AKT   Phase I
MKC-1 EntreMed AKT/mTOR EC, OC, NSCLC, BC, pancreatic cancer, and CRC Phase II

Table 1: Inhibitors targeting PI3K/AKT pathway in clinical development CC, Cervical cancer, CLL, chronic lymphocytic leukemia; CRC, colorectal carcinoma; EC, endometria cancer; ER, estrogen receptor; GC, gastric cancer, GBM, glioblastoma; HCC, hepatocellular carcinoma; Her2, human epidermal growth factor receptor-2; HNSCC, head and neck squamous cell carcinoma; HR, hormone receptor; mBC, metastatic breast cancer; MM, multiple myeloma; NHL, Non-Hodgkin's lymphoma; NSCLC, non-small cell lung cancer; OC, ovarian cancer; PI3K, phosphatidylinositol 3-kinase; PNET, pancreatic neuroendocrine tumors; RCC, renal cell carcinoma; SEGA, subependymal giant cell astrocytoma.

mTOR Inhibitors

Everolimus is a mTORC1 selective inhibitor and has been tested in GC patients in phase II and phase III trials. In the recent phase II trial, everolimus demonstrated anti-tumor activity with a response rate of 3.7% (2/44) and a disease control rate (DCR) of 38.9% (17/44) [53]. However, a everolimus phase III study failed to achieve a survival benefit in comparison with the best supportive care (BSC) in previously treated advanced GC cases [54]. Although the phase II trial failed to achieve its primary objective, the observed association between high levels of pS6 expression (Ser240/4) at baseline and higher DCR and prolonged PFS warrant further trials with a molecular-stratification-based patient selection strategy. Because eveolimus suppresses only mTORC1 and the feedback activation of MAPK may limit its anti-tumor potency [55], a few inhibitors of both mTORC1 and mTORC2 with improved potency (e.g., OSI-027, BEZ235, XL765, AZ8055, and Ink128) are being currently evaluated in phase I/II trials for patients with solid tumors.

PI3KCA Inhibitors

There are two types of PI3K inhibitors targeting the p110 catalytic subunit that are currently under clinical development: pan-PI3K inhibitors and isoform-specific PI3K inhibitors.Pan-PI3K inhibitors are active against all family members of PI3K, whereas isoform specific inhibitors selectively inhibit p110a, ß, or d. These include the pan-PI3K inhibitors BKM120 (Novartis), PX-886 (Oncothyreon), XL147 (SAR245408; Sanofi) and the p110a selective inhibitors BYL719, GDC0032, and INK1117 etc. [56]. Because the catalytic domains of p110 and mTOR are structurally similar and mTOR activation is the downstream event of the PI3K/AKT pathway, it is expected that the dual inhibition of these targets may be more effective. Some of the dual PI3K/mTOR inhibitors currently being investigated in clinical trials include BEZ235, XL765, GDC-0980, GDC0084, SF1126, and PF-46915 etc. [57]. The clinical anti-tumor activity of some of these inhibitors has been reported [58].

AKT Inhibitors

AKT plays a central role in the activation of the PI3K/AKT pathway to facilitate cellular survival and suppress cell apoptosis. A number of AKT inhibitors have entered clinical trials. These include the allosteric inhibitors perifosine (Keryx Biopharmaceuticals) and MK-2206 (Merck) and the ATP competitive inhibitors AZD5363, GSK690693 and GDC0068 etc. Previously, we reported the development of AZD5363 and demonstrated its activity in GC cell lines [31,44]. Recent phase I data indicated that AZD5363, as a monotherapy, led to partial responses in two patients harboring tumor mutations in either AKT1 or PI3KCA [59].

Future Direction

For patients with metastatic or recurrent GC, the evidence supports the use of chemotherapy to prolong survival and maintain quality of life. However, the long-term outcomes of chemotherapy treatments are poor, suggesting the need for novel targeted agents that may confer a greater survival benefit. The critical role of the activation of the PI3K/AKT signaling pathway in both tumorigenesis and drug resistance has been well-documented. A number of small molecule inhibitors of PI3K, AKT, or mTOR are at various stages of drug development for the treatment of solid tumors, including GC (Table 1). Because the activations of AKT and mTOR have been commonly detected in human GC, it is expected that agents against the PI3K/AKT pathway alone or in combination with current therapies will provide viable options for the treatment of advanced GC.However, there are still challenges ahead in terms of their clinical application. First, the results of the phase III everolimus trial in GC cases without patient selection were disappointing. Second, the preclinical data indicated that only PI3KCA mutations were predictive of a response to AKT inhibitor (31).Thus, it is critical to validate and implement biomarkers and assays for the selection of patients with a predictive response to the therapy. Another approach for improving efficacy is to combine the inhibitors of the PI3K/AKT pathway with chemo or other targeted agents. Preclinical translational studies based on rational design and the safety profiles of the inhibitors will help in choosing the right agents and combinations.

References

  1. Wilkinson NW, Howe J, Gay G, Patel-Parekh L, Scott-Conner C, et al. (2008) Differences in the pattern of presentation and treatment of proximal and distal gastric cancer: results of the 2001 gastric patient care evaluation. Ann Surg Oncol 15: 1644-1650.
  2. Parkin DM, Bray F, Ferlay J, Pisani P (2005) Global cancer statistics, 2002. CA Cancer J Clin 55: 74-108.
  3. Jemal A, Bray F, Center MM, Ferlay J, Ward E, et al. (2011) Global cancer statistics. CA Cancer J Clin 61: 69-90.
  4. Takahashi T, Saikawa Y, Kitagawa Y (2013) Gastric cancer: current status of diagnosis and treatment. Cancers (Basel) 5: 48-63.
  5. Tan IB, Ng I, Tai WM, Tan P (2012) Understanding the genetic basis of gastric cancer: recent advances. Expert Rev Gastroenterol Hepatol 6: 335-341.
  6. Jung KW, Park S, Kong HJ, Won YJ, Lee JY, et al. (2012) Cancer Statistics in Korea: Incidence, Mortality, Survival, and Prevalence in 2009. Cancer Res Treat 44: 11-24.
  7. Strong VE, Song KY, Park CH, Jacks LM, Gonen M, et al. (2010) Comparison of Gastric Cancer survival following R0 resection in the United States and Korea using an internationally validated nomogram. Ann Surg 251: 640-646.
  8. Power DG, Kelsen DP, Shah MA (2010) Advanced gastric cancer--slow but steady progress. Cancer Treat Rev 36: 384-392.
  9. Ohtsu A, Yoshida S, Saijo N (2006) Disparities in gastric cancer chemotherapy between the East and West. J Clin Oncol 24: 2188-2196.
  10. Roviello F, Marrelli D, de Manzoni G, Morgagni P, Di Leo A, et al. (2003) Prospective study of peritoneal recurrence after curative surgery for gastric cancer. Br J Surg 90: 1113-1119.
  11. GASTRIC Group, Paoletti X, Oba K, Burzykowski T, Michiels S, Ohashi Y, et al.(2010) Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA 303:1729-1737.
  12. Wagner AD, Grothe W, Haerting J, Kleber G, Grothey A, et al. (2006) Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. J Clin Oncol 24: 2903-2909.
  13. Park SC, Chun HJ (2013) Chemotherapy for advanced gastric cancer: review and update of current practices. Gut Liver 7: 385-393.
  14. Dhanasekaran DN, Johnson GL (2007) MAPKs: function, regulation, role in cancer and therapeutic targeting. Oncogene 26: 3097-3099.
  15. Schlessinger J (2004) Common and distinct elements in cellular signaling via EGF and FGF receptors. Science 306: 1506-1507.
  16. De Vita F, Giuliani F, Silvestris N, Catalano G, Ciardiello F, et al. (2010) Human epidermal growth factor receptor 2 (HER2) in gastric cancer: a new therapeutic target. Cancer Treat Rev 36 Suppl 3: S11-15.
  17. Gravalos C, Jimeno A (2008) HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 19: 1523-1529.
  18. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, et al. (2010) Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376: 687-697.
  19. Okines AF, Cunningham D (2010) Trastuzumab in gastric cancer. Eur J Cancer 46: 1949-1959.
  20. Engelman JA, Luo J, Cantley LC (2006) The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet 7: 606-619.
  21. Yang ZZ1, Tschopp O, Baudry A, Dümmler B, Hynx D, et al. (2004) Physiological functions of protein kinase B/Akt. Biochem Soc Trans 32: 350-354.
  22. Engelman JA (2009) Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 9: 550-562.
  23. Kumar A, Rajendran V, Sethumadhavan R, Purohit R (2013) AKT kinase pathway: a leading target in cancer research. ScientificWorldJournal 2013: 756134.
  24. Jeong SJ, Pise-Masison CA, Radonovich MF, Park HU, Brady JN (2005) Activated AKT regulates NF-kappaB activation, p53 inhibition and cell survival in HTLV-1-transformed cells. Oncogene 24: 6719-6728.
  25. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, et al. (2004) High frequency of mutations of the PIK3CA gene in human cancers. Science 304: 554.
  26. Wong H, Yau T (2012) Targeted therapy in the management of advanced gastric cancer: are we making progress in the era of personalized medicine? Oncologist 17: 346-358.
  27. Velho S, Oliveira C, Ferreira A, Ferreira AC, Suriano G, et al. (2005) The prevalence of PIK3CA mutations in gastric and colon cancer. Eur J Cancer 41: 1649-1654.
  28. Barbi S, Cataldo I, De Manzoni G, Bersani S, Lamba S, et al. (2010) The analysis of PIK3CA mutations in gastric carcinoma and metanalysis of literature suggest that exon-selectivity is a signature of cancer type. J Exp Clin Cancer Res 29: 32.
  29. Li VS, Wong CW, Chan TL, Chan AS, Zhao W, et al. (2005) Mutations of PIK3CA in gastric adenocarcinoma. BMC Cancer 5: 29.
  30. Shi J, Yao D, Liu W, Wang N, Lv H, et al. (2012) Highly frequent PIK3CA amplification is associated with poor prognosis in gastric cancer. BMC Cancer 12: 50.
  31. Li J, Davies BR, Han S, Zhou M, Bai Y, et al. (2013) The AKT inhibitor AZD5363 is selectively active in PI3KCA mutant gastric cancer, and sensitizes a patient-derived gastric cancer xenograft model with PTEN loss to Taxotere. J Transl Med 11: 241.
  32. Georgescu MM (2010) PTEN Tumor Suppressor Network in PI3K-Akt Pathway Control. Genes Cancer 1: 1170-1177.
  33. Krohn A, Diedler T, Burkhardt L, Mayer PS, De Silva C, et al. (2012) Genomic deletion of PTEN is associated with tumor progression and early PSA recurrence in ERG fusion-positive and fusion-negative prostate cancer. Am J Pathol. 181: 401-412.
  34. Byun DS, Cho K, Ryu BK, Lee MG, Park JI, et al. (2003) Frequent monoallelic deletion of PTEN and its reciprocal associatioin with PIK3CA amplification in gastric carcinoma. Int J Cancer 104: 318-327.
  35. Kang YH, Lee HS, Kim WH (2002) Promoter methylation and silencing of PTEN in gastric carcinoma. Lab Invest 82: 285-291.
  36. Bellacosa A, Kumar CC, Di Cristofano A, Testa JR (2005) Activation of AKT kinases in cancer: implications for therapeutic targeting. Adv Cancer Res 94: 29-86.
  37. Murayama T, Inokuchi M, Takagi Y, Yamada H, Kojima K, et al. (2009) Relation between outcomes and localisation of p-mTOR expression in gastric cancer. Br J Cancer 100: 782-788.
  38. Lang SA, Gaumann A, Koehl GE, Seidel U, Bataille F, et al. (2007) Mammalian target of rapamycin is activated in human gastric cancer and serves as a target for therapy in an experimental model. Int J Cancer 120: 1803-1810.
  39. Yu G, Wang J, Chen Y, Wang X, Pan J, et al. (2009) Overexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of chinese patients with gastric cancer. Clin Cancer Res 15: 1821-1829.
  40. Oki E1, Baba H, Tokunaga E, Nakamura T, Ueda N, et al. (2005) Akt phosphorylation associates with LOH of PTEN and leads to chemoresistance for gastric cancer. Int J Cancer 117: 376-380.
  41. Oki E, Kakeji, E. Tokunaga, K. Nishida, T. Koga, et al. (2006) Impact of PTEN/AKT/PI3K signal pathway on the chemotherapy for gastric cancer. J Clin Oncol 24: 4034.
  42. Yu HG, Ai YW, Yu LL, Zhou XD, Liu J, et al. (2008) Phosphoinositide 3-kinase/Akt pathway plays an important role in chemoresistance of gastric cancer cells against etoposide and doxorubicin induced cell death. Int J Cancer 122: 433-443.
  43. Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, et al. (2013) Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models. Clin Cancer Res 19: 1760-1772.
  44. Davies BR, Greenwood H, Dudley P, Crafter C, Yu DH, et al.(2012) Preclinical pharmacology of AZD5363, an inhibitor of AKT: pharmacodynamics, antitumor activity, and correlation of monotherapy activity with genetic background. Mol Cancer Ther 11:873-887.
  45. Im SA, Lee KE, Nam E, Kim DY, Lee JH, et al. (2005) Potential prognostic significance of p185(HER2) overexpression with loss of PTEN expression in gastric carcinomas. Tumori 91: 513-521.
  46. Vadlakonda L, Dash A, Pasupuleti M, Anil Kumar K, Reddanna P (2013) The Paradox of Akt-mTOR Interactions. Front Oncol 3: 165.
  47. Geng M, Wang L, Li P (2013) Correlation between chemosensitivity to anticancer drugs and Bcl-2 expression in gastric cancer. Int J Clin Exp Pathol 6: 2554-2559.
  48. Rayet B, Gélinas C (1999) Aberrant rel/nfkb genes and activity in human cancer. Oncogene 18: 6938-6947.
  49. Madrid LV, Wang CY, Guttridge DC, Schottelius AJ, Baldwin AS Jr, et al. (2000) Akt suppresses apoptosis by stimulating the transactivation potential of the RelA/p65 subunit of NF-kappaB. Mol Cell Biol 20: 1626-1638.
  50. Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM, et al. (1999) NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature 401: 82-85.
  51. Xie X, Tang B, Zhou J, Gao Q, Zhang P (2013) Inhibition of the PI3K/Akt pathway increases the chemosensitivity of gastric cancer to vincristine. Oncol Rep 30: 773-782.
  52. Shin JY, Kim JO, Lee SK, Chae HS, Kang JH (2010) LY294002 may overcome 5-FU resistance via down-regulation of activated p-AKT in Epstein-Barr virus-positive gastric cancer cells. BMC Cancer 10: 425.
  53. Yoon DH, Ryu MH, Park YS, Lee HJ, Lee C, et al. (2012) Phase II study of everolimus with biomarker exploration in patients with advanced gastric cancer refractory to chemotherapy including fluoropyrimidine and platinum. Br J Cancer 106: 1039-1044.
  54. Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, eta al. (2013) Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol. 31: 3935-3943.
  55. Carracedo A, Ma L, Teruya-Feldstein J, Rojo F, Salmena L, et al. (2008) Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest 118: 3065-3074.
  56. Welker ME, Kulik G (2013) Recent syntheses of PI3K/Akt/mTOR signaling pathway inhibitors. Bioorg Med Chem 21: 4063-4091.
  57. Brana I, Siu LL (2012) Clinical development of phosphatidylinositol 3-kinase inhibitors for cancer treatment. BMC Med 10: 161.
  58. Akinleye A, Avvaru P, Furqan M, Song Y, Liu D1 (2013) Phosphatidylinositol 3-kinase (PI3K) inhibitors as cancer therapeutics. J Hematol Oncol 6: 88.
  59. Udai Banerji, Malcolm R, Jan HS, Taito Esaki, Emma D, et al. (2013) Results of two phase I multicenter trials of AZD5363, an inhibitor of AKT1, 2 and 3: Biomarker and early clinical evaluation in Western and Japanese patients with advanced solid tumors. 104th AACR Annual meeting LB-66.
Citation: Thomas ML, Coyle KM, Sultan M, Ahmad VK, Marcato P (2014) Chemoresistance in Cancer Stem Cells and Strategies to Overcome Resistance. Chemotherapy 3:125.

Copyright: © 2014 Thomas ML, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top