Minireviews Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Nov 15, 2016; 8(11): 786-792
Published online Nov 15, 2016. doi: 10.4251/wjgo.v8.i11.786
Clinical impact of chemotherapy to improve tumor microenvironment of pancreatic cancer
Takahiro Tsuchikawa, Shintaro Takeuchi, Toru Nakamura, Toshiaki Shichinohe, Satoshi Hirano, Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
Author contributions: Tsuchikawa T wrote the manuscript; Takeuchi S and Hirano S contributed to the writing of the manuscript; Nakamura T and Shichinohe T designed the manuscript.
Conflict-of-interest statement: None of the authors have any commercial or financial involvements in connection with this study that represent or appear to represent any conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Takahiro Tsuchikawa, Associate Professor, Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, N-15 W-7, Sapporo 060-8638, Japan. tsuchi-t@med.hokudai.ac.jp
Telephone: +81-11-7067714 Fax: +81-11-7067158
Received: March 28, 2016
Peer-review started: April 1, 2016
First decision: May 23, 2016
Revised: July 19, 2016
Accepted: September 13, 2016
Article in press: September 18, 2016
Published online: November 15, 2016

Abstract

A perioperative multimodal strategy including combination chemotherapy and radiotherapy, in addition to surgical resection, has been acknowledged to improve patient prognosis. However chemotherapy has not been actively applied as an immunomodulating modality because of concerns about various immunosuppressive effects. It has recently been shown that certain chemotherapeutic agents could modify tumor microenvironment and host immune responses through several underlying mechanisms such as immunogenic cell death, local T-cell infiltration and also the eradication of immune-suppressing regulatory cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells. With the better understanding of the cell components in the tumor microenvironment and the effect of chemotherapy to improve tumor microenvironment, it has been gradually clear that the chemotherapeutic agents is two-edged sword to have both immune promoting and suppressing effects. The cellular components of the tumor microenvironment include infiltrating T lymphocytes, dendritic cells, regulatory T cells, tumor associated macrophages, myeloid derived suppressor cells and cancer associated fibroblasts. Based on the better understanding of tumor microenvironment following chemotherapy, the treatment protocol could be modified as personalized medicine and the prognosis of pancreas cancer would be more improved utilizing multimodal chemotherapy. Here we review the recent advances of chemotherapy to improve tumor microenvironment of pancreatic cancer, introducing the unique feature of tumor microenvironment of pancreatic cancer, interaction between anti-cancer reagents and these constituting cells and future prospects.

Key Words: Pancreas cancer, Microenvironment, Chemotherapy, Immune cells, Immunomodulation

Core tip: It has been gradually clear that the chemotherapeutic agents are two-edged sword to have both immune promoting and suppressing effects. The cellular components of the tumor microenvironment including infiltrating T lymphocytes, dendritic cells, regulatory T cells, tumor associated macrophages, myeloid derived suppressor cells and cancer associated fibroblasts could be improved. Based on the better understanding of tumor microenvironment following chemotherapy, the treatment protocol could be modified as personalized medicine and the prognosis of pancreas cancer would be more improved utilizing multimodal treatment strategy.



INTRODUCTION

Pancreatic carcinoma is an extremely aggressive malignant tumor and the fifth leading cause of death worldwide and is expected to be the second by 2030 in Western countries[1,2]. The only curative option is surgical resection, but the 5-year overall survival (OS) rate still needs to be improved from the current 10%-15% even after curative resection[1,3]. A perioperative multimodal strategy including combination chemotherapy and radiotherapy, in addition to surgical resection, has been acknowledged to improve patient prognosis. New cytotoxic agents such as gemcitabine, Tegafur-gimeracil-oteracil potassium (TS-1) and combination chemotherapy with 5-fluorouracil (5-FU), oxaliplatin, and irinotecan along with perioperative chemoradiotherapy before and after surgery have recently been widely investigated[4,5]. Chemotherapy, usually a standard treatment option for cancer, has not been actively applied as an immune-modulating modality because of concerns about various immunosuppressive effects. However, certain chemotherapeutic agents have recently been shown to improve host immune responses and even break immune tolerance[6]. Several underlying mechanisms have been clarified, including immunogenic cell death[7,8], local T-cell infiltration and also the eradication of immune-suppressing regulatory cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), all of which are associated with cells in the tumor microenvironment[9]. On the other hand, care must be taken that chemotherapy-induced cancer metastasis does occur during treatment through non-immunological pathways[10].

We review recent advances in chemotherapeutic regimens to improve the tumor microenvironment for pancreatic cancer, and introduce unique features of the tumor microenvironment for pancreatic cancer, interactions between anti-cancer reagents and the constituent cells, and future prospects.

OVERVIEW OF STANDARD CHEMOTHERAPY AND CHEMORADIATION THERAPY FOR PANCREATIC CANCER

Although the only curative option is surgical resection, with the advances in perioperative strategy for pancreatic carcinoma, many cytotoxic agents have proven effective in treating this disease. Chemoradiation therapy had been also adopted aiming at locoregional response and additional effects outside the field of irradiation (abscopal effects)[11].

Representative cytotoxic agents include historical 5-FU monotherapy, gemcitabine monotherapy, and gemcitabine-based combination therapies[4]. The following randomized controlled trials are investigations recently undertaken try to improve the chemotherapeutic strategy for pancreatic cancer. Burris et al[12] had shown for the first time that gemcitabine was superior to 5-FU in terms of overall survival, thus suggesting gemcitabine as a key drug in advanced pancreatic cancer. In 2011, FOLFILINOX (5-FU, leucovorin, irinotecan and oxaliplatin) had been shown to have survival benefit over gemcitabine alone in patients with metastatic pancreas cancer[13]. Recently Nab-paclitaxel plus gemcitabine have been reported to have superior efficacy compared with gemcitabine monotherapy in metastatic pancreas cancer (MPACT trial)[14]. However, MPACT trial, consisting of Gem + Nab-paclitaxel had OS of 8.5 mo compared to 6.7 mo in patients treated with gemcitabine alone) suggesting minimal improvement of survival by current chemotherapy regimens and requiring for further developments.

Taken together, overall survival of patients with metastatic disease extended to nearly 1 year from around 6 mo in the preceding two decades, thanks to recent therapeutic advances. However, although these reagents are promising, median progression-free survival remains limited and the 5-year survival rate of patients is still unsatisfactory, at around 15%-20%, even with these multimodal treatment strategies. This is due in part to dose limiting toxicity of side effects such as neuropathy and bone marrow suppression and due also to chemoresistance, relapse and metastasis even after surgical resection.

TUMOR MICROENVIRONMENT OF PANCREATIC CANCER

Tumor cells alone were initially considered the specific target of chemotherapy, leading to a focus on the cytotoxicity of agents inhibiting DNA repair, tubulin formation and cell proliferation[15]. However, recent research has identified the tumor microenvironment as comprising tumor cells, host immune cells such as T cells, Tregs and MDSCs, and cancer-associated fibroblasts or stromal cells that support or suppress each other[9]. Each of these cellular components contributes to treatment response and patient prognosis, with tumor cells forming a network through direct interactions and cytokines providing important signals to initiate cell invasion into vessels and lymph nodes, leading to distant metastasis. Desmosomes are also one of the specific features of pancreas carcinoma that make drug delivery so difficult and prevent immune cells from infiltrating to tumor nests[16].

These evidences collectively indicate that tumor cells are thought to grow, interacting with the microenvironment, highlighting the need to clarify the specific mechanisms by which each chemotherapeutic agent improves the tumor microenvironment to contribute to treatment efficacy.

The following sections are arranged to describe recent evidence for the effects of chemotherapeutic agents on the cellular components of the tumor microenvironment (Table 1).

Table 1 Targets of chemotherapy to improve tumor microenvironment.
Cellular componentsTarget moleculesChemotherapeutic agentsUnderlying mechanismRef.
TILCD4, CD8 positive T lymphocytes RAS/MAPKGEM, TS-1, MEK inhibitor, PD1/PDL1 immune checkpoint inhibitorsIncrease lymphocyte infiltration[22] [24]
DCGEMProliferation of DC and CTL[26]
TregCD4 and FoxP3 positive T lymphocytesGEM, cyclophosphamideDepletion[21]
MDSCCCL2, CCR2, GMCSFGEM, 5-FUIncrease differentiation Depletion[34,35]
[5]
CAFPalladin positive fibroblastsGEMDepletion[38]
mTOR/4E-BP1 pathwayGEM, PasireotideReduce tumor growth and chemoresistance[39]
INFILTRATING T LYMPHOCYTES

A number of reports have suggested that the accumulation of CD4 and CD8 lymphocytes in solid tumors offers a good prognostic indicator for patient survival[17,18]. In terms of pancreatic cancer, Tewari et al[19] demonstrated a positive correlation between prognosis and the presence of tumor infiltrating T cells. Although the clinical relevance differs among types of cancer, in association with the HLA class I expression level[20], some agents have been reported to induce T-cell infiltration into pancreas cancers[21]. Homma et al[22] showed that CD4+ and CD8+ cells were significantly increased after neoadjuvant chemotherapy comprising gemcitabine and TS-1 followed by radiotherapy (NACRT), and a high accumulation of CD4+ cells offered a good prognostic marker for pancreas carcinoma after NACRT. Teng et al[23] recently classified the types of tumor microenvironment based on the presence or absence of T-cell infiltration and expression of PD1 along with patient prognosis.

Loi et al[24] recently suggested that therapeutic cooperation of MEK and PD-1/PD-L1 immune check point inhibitors could increase tumor-infiltrating lymphocytes through RAS/MAPK pathways in breast cancer. Great expectations are held for increased control of the tumor microenvironment, especially with tumor-infiltrating lymphocytes enabling further improvements in patient prognosis associated with immune check point inhibitors.

DENDRITIC CELLS

Dendritic cells are the most potent antigen presenting cells and play a crucial part in the initiation, programing, and regulation of antitumor immunity, directing cytotoxic T lymphocytes and natural killer cells to become potent antitumor effectors capable of eradicating malignant cells[25,26]. Recently it had been reported that dendritic cells are impaired in number and display maturation defects disable to function as antigen presenting cells in pancreatic cancer due to the inflammation of the disease[27]. Meanwhile, chemotherapy can promote immunogenic cell apoptosis enhancing immunogenicity and mediating efficient phagocytosis by dendritic cell[7]. Moreover, gemcitabine can enhance the cross presentation of tumor associated antigens by dendritic cells and as well as inducing the proliferation of DC and CTL[26]. Those strategies utilizing chemotherapeutic agents might be useful to overcome negative microenvironment.

REGULATORY T CELLS

Tregs are defined as T cells expressing both CD4 and forkhead box P3 (FoxP3), and are usually associated with poor prognosis and immunosuppression in various cancers. Transcriptional FoxP3 is a crucial intracellular marker and also a key developmental and functional factor for CD4+FoxP3+ Tregs[28]. In terms of pancreatic cancer, multimodal chemotherapy including GEM, cyclophosphamide, and taxane has been demonstrated to decrease Tregs in the tumor microenvironment[5]. Low Treg percentage in circulation at 1 year after PC resection had been correlated with improved survival[29]. We have also previously shown that neoadjuvant treatment of pancreatic ductal adenocarcinoma with chemotherapy and chemoradiotherapy can alter the local Treg balance in favor of antitumor immunity in resected human sections[21]. Another paper by Keenan et al[30] showed that immunization of mice with Listeria Monocytegenes engineered to express k-ras along with depletion of Treg cells reduced progression of early stages PanINs. Also, Shibuya et al[31] recently reported that CD8 effector T cells show marked accumulation in the tumor microenvironment, but are suppressed by Tregs and PD-L1 expressed on T cells. These findings have therefore led to expectations for novel strategies of multimodal chemotherapy in combination with immune checkpoint inhibitors reducing Tregs.

TUMOR-ASSOCIATED MACROPHAGES

Tumor-associated macrophages (TAMs) are derived from CCR2+ monocytes in the spleen and peripheral blood, infiltrating into the tumor and developing into macrophages on stimulation by the releasing hormone CCL2 and colony-stimulating factor 1 (CSF-1)[32,33]. TAMs have recently been reported to limit the effects of chemotherapy and promote tumor chemoresistance[34]. Michem et al. reported that targeting TAMs by inhibiting CSF1R or C-C chemokine receptor 2 (CCR2) could decrease the number of pancreatic tumor-initiating cells and improve chemotherapeutic efficacy in vivo. The Denargo group reported that the combination of cytotoxic chemotherapy and blockade of CSF1R, which is prominently expressed by monocytes, Mo-MDSC and macrophages, resulted in improved anti-tumor T-cell responses[32]. Furthermore, Sanford et al[33] reported that the CCL2/CCR2 chemokine axis plays a crucial role in the recruitment of inflammatory monocytes from bone marrow to peripheral sites of inflammation and an increased ratio of inflammatory monocytes in blood compared to bone marrow offers a novel predictor of decreased patient survival following tumor resection. These lines of evidence clearly show that chemotherapy combined with chemokine blockade might reduce the chemoresistance associated with the exclusion of TAMs.

MYELOID DERIVE SUPPRESSOR CELLS

MDSCs are heterogeneous populations of immune cells derived from progenitor cells in bone marrow. MDSCs with a phenotype of CD33+HLA-DR-/low that are lineage-negative (CD14-, CD15-) are well described as immunosuppressive in cancer patients contributing to tumor progression by damping T-cell immunity and promoting angiogenesis[35,36]. Many chemotherapeutic drugs have long been thought to exclude MDSCs from various cancers. Zheng et al[5] showed that GEM and 5-FU have a direct killing effect on MDSCs. In contrast, Takeuchi et al[36] reported that GEM could increase MDSC numbers through increases in GM-CSF levels, converting M2 macrophages into suppressive MDSCs. Therefore MDSC in peripheral blood might be a possible predictive biomarker of chemotherapy failure in PC patients[37]. Also, GEM and 5-FU have been reported to activate NLRP3 inflammasomes in MDSCs, leading to interleukin-1β release, which restrains their antitumor efficacy[38]. More recently, Hu et al[39] reported TNFB2R as important for its suppressive function. Uniquely, Sanford et al[40] recently reported the clinical utility of zoledronic acid. This agent is usually utilized to improve calcium imbalances in patients osteoporosis, but also prevents tumor-mediated myelopoiesis associated with the generation of MDSC. Further studies are warranted to adjust the balance between direct reduction of MDSCs and indirect promotion of MDSCs by chemotherapy in combination with the multimodal strategies described above.

CANCER-ASSOCIATED FIBROBLASTS

Fibrous stroma associated with cancer in the tumor micro environment has increasingly been recognized as involving cancer-associated fibroblasts (CAFs). These cells are reported to contribute to poorer survival in various tumors, including pancreatic ductal adenocarcinoma, which has been reported to contain large numbers of CAFs[41]. The characteristically dense desmosome in pancreatic cancer acts as a barrier to drug delivery, thus contributing to chemoresistance[4]. Among the many markers of CAFs, Sato et al[41] reported that palladin, a CAF marker, could represent an independent marker of poor prognosis and a biomarker to predict the efficiency of chemotherapy or even disease recurrence. Duluc et al[42] recently revealed one of the underlying mechanisms abrogating pancreatic cancer chemoresistance through the mTOR/4E-BP1 pathway, allowing GEM-based chemotherapy combined with sst1 receptor-activating pasireotide to reduce tumor growth and chemoresistance. This kind of anti-stromal targeted therapy could be expected in addition to host immune cell-targeted therapy, as an adjunct to direct killing of cancer cells.

DISCUSSION

With our developing understanding of the cell components in the tumor microenvironment and the effects of chemotherapy in improving this environment, chemotherapeutic agents have gradually been revealed to represent a two-edged sword with effects that both promote and suppress immunity[5]. Such therapies deplete one factor of immune suppression while at the same time inducing another mechanism to inhibit host immune responses. Some experimental data in this paper show that these problems might be overcome by multimodal combination chemoimmunotherapy in addition to standard chemotherapy, blocking antibodies for cytokine release or utilizing immune checkpoint inhibitors. Beaty et al[43] reported combining chemotherapy with the agonist CD40, as a member of the TNF receptor superfamily, for surgically incurable PDA and observed tumor regression in some patients. Takeuchi et al[36] likewise reported that anti-GM-CSF antibody blocking could accelerate the formation of immunosuppressive myeloid cells in the tissue microenvironment of human pancreatic cancer.

Chemotherapeutic protocols including timing and dose might also be further explored and modified based on both reductions in tumor size and the induction of anti-tumor-specific immunity. Metronomic chemotherapy or low-dose chemotherapy has been reported to induce anti-tumor T-cell immunity in vivo[44]. One of the underlying mechanisms might be that such low-toxicity doses of cytotoxic agents induce minimal suppression of tumor cells while concomitantly inducing minimal suppression of immune-promoting cells based on altered immune balance.

Lastly, future evidence should be accumulated regarding these balances in the tumor microenvironment during multimodal chemotherapy by measuring biomarkers locally and systematically. Biopsy specimens provide information of infiltrating T lymphocyte levels in the tumor microenvironment, offering possible predictors of beneficial response to chemotherapy in breast and pancreas cancers. SPARC expression levels in the stroma could represent a target for nab-paclitaxel. Although data must continue to be accumulated, miRNA might reflect changes in immune balances and predict the efficacy of chemoimmunotherapy[45]. Taking all these lines of evidences together in combination with the properties of emerging agents, current problems seem likely to be overcome, at least in part, and the prognosis of pancreas cancer can be expected to continue improving in the coming decades.

CONCLUSION

The chemotherapeutic agents have both immune promoting and suppressing effects in the tumor microenvironment of pancreatic cancer. Based on the better understanding of tumor microenvironment following chemotherapy, the treatment protocol could be modified as personalized medicine and the prognosis of pancreas cancer would be more improved utilizing multimodal chemotherapy.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: Japan

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P- Reviewer: Chen YC, Gonzalez-Perez RR, Seuningen IV S- Editor: Gong ZM L- Editor: A E- Editor: Wu HL

References
1.  Ansari D, Chen BC, Dong L, Zhou MT, Andersson R. Pancreatic cancer: translational research aspects and clinical implications. World J Gastroenterol. 2012;18:1417-1424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
2.  Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12135]  [Cited by in F6Publishing: 12698]  [Article Influence: 1587.3]  [Reference Citation Analysis (2)]
3.  Nakao A, Takeda S, Inoue S, Nomoto S, Kanazumi N, Sugimoto H, Fujii T. Indications and techniques of extended resection for pancreatic cancer. World J Surg. 2006;30:976-982; discussion 983-984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 124]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
4.  Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Ther Adv Med Oncol. 2015;7:68-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 109]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
5.  Zheng Y, Dou Y, Duan L, Cong C, Gao A, Lai Q, Sun Y. Using chemo-drugs or irradiation to break immune tolerance and facilitate immunotherapy in solid cancer. Cell Immunol. 2015;294:54-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 66]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
6.  Zitvogel L, Kepp O, Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol. 2011;8:151-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 474]  [Cited by in F6Publishing: 498]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
7.  Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1986]  [Cited by in F6Publishing: 2201]  [Article Influence: 122.3]  [Reference Citation Analysis (0)]
8.  Yamamura Y, Tsuchikawa T, Miyauchi K, Takeuchi S, Wada M, Kuwatani T, Kyogoku N, Kuroda A, Maki T, Shichinohe T. The key role of calreticulin in immunomodulation induced by chemotherapeutic agents. Int J Clin Oncol. 2015;20:386-394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
9.  Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4060]  [Cited by in F6Publishing: 4976]  [Article Influence: 497.6]  [Reference Citation Analysis (0)]
10.  Gingis-Velitski S, Loven D, Benayoun L, Munster M, Bril R, Voloshin T, Alishekevitz D, Bertolini F, Shaked Y. Host response to short-term, single-agent chemotherapy induces matrix metalloproteinase-9 expression and accelerates metastasis in mice. Cancer Res. 2011;71:6986-6996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 95]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
11.  Blanquicett C, Saif MW, Buchsbaum DJ, Eloubeidi M, Vickers SM, Chhieng DC, Carpenter MD, Sellers JC, Russo S, Diasio RB. Antitumor efficacy of capecitabine and celecoxib in irradiated and lead-shielded, contralateral human BxPC-3 pancreatic cancer xenografts: clinical implications of abscopal effects. Clin Cancer Res. 2005;11:8773-8781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 55]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
12.  Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403-2413.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4838]  [Cited by in F6Publishing: 5099]  [Article Influence: 392.2]  [Reference Citation Analysis (1)]
14.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4035]  [Cited by in F6Publishing: 4330]  [Article Influence: 393.6]  [Reference Citation Analysis (0)]
15.  Curtin NJ. DNA repair dysregulation from cancer driver to therapeutic target. Nat Rev Cancer. 2012;12:801-817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 715]  [Cited by in F6Publishing: 720]  [Article Influence: 60.0]  [Reference Citation Analysis (0)]
16.  Li X, Ma Q, Xu Q, Duan W, Lei J, Wu E. Targeting the cancer-stroma interaction: a potential approach for pancreatic cancer treatment. Curr Pharm Des. 2012;18:2404-2415.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, Ohtani H. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58:3491-3494.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Tsuchikawa T, Ikeda H, Cho Y, Miyamoto M, Shichinohe T, Hirano S, Kondo S. Association of CD8+ T cell infiltration in oesophageal carcinoma lesions with human leucocyte antigen (HLA) class I antigen expression and survival. Clin Exp Immunol. 2011;164:50-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
19.  Tewari N, Zaitoun AM, Arora A, Madhusudan S, Ilyas M, Lobo DN. The presence of tumour-associated lymphocytes confers a good prognosis in pancreatic ductal adenocarcinoma: an immunohistochemical study of tissue microarrays. BMC Cancer. 2013;13:436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
20.  Tanaka K, Tsuchikawa T, Miyamoto M, Maki T, Ichinokawa M, Kubota KC, Shichinohe T, Hirano S, Ferrone S, Dosaka-Akita H. Down-regulation of Human Leukocyte Antigen class I heavy chain in tumors is associated with a poor prognosis in advanced esophageal cancer patients. Int J Oncol. 2012;40:965-974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
21.  Tsuchikawa T, Hirano S, Tanaka E, Matsumoto J, Kato K, Nakamura T, Ebihara Y, Shichinohe T. Novel aspects of preoperative chemoradiation therapy improving anti-tumor immunity in pancreatic cancer. Cancer Sci. 2013;104:531-535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
22.  Homma Y, Taniguchi K, Murakami T, Nakagawa K, Nakazawa M, Matsuyama R, Mori R, Takeda K, Ueda M, Ichikawa Y. Immunological impact of neoadjuvant chemoradiotherapy in patients with borderline resectable pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2014;21:670-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
23.  Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying Cancers Based on T-cell Infiltration and PD-L1. Cancer Res. 2015;75:2139-2145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 879]  [Cited by in F6Publishing: 1039]  [Article Influence: 115.4]  [Reference Citation Analysis (0)]
24.  Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, Combs S, Rimm DL, Giltnane JM, Estrada MV. RAS/MAPK Activation Is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation Between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin Cancer Res. 2016;22:1499-1509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in F6Publishing: 380]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
25.  Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 2014;15:e257-e267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 503]  [Article Influence: 50.3]  [Reference Citation Analysis (0)]
26.  Kajihara M, Takakura K, Kanai T, Ito Z, Matsumoto Y, Shimodaira S, Okamoto M, Ohkusa T, Koido S. Advances in inducing adaptive immunity using cell-based cancer vaccines: Clinical applications in pancreatic cancer. World J Gastroenterol. 2016;22:4446-4458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
27.  Tjomsland V, Spångeus A, Sandström P, Borch K, Messmer D, Larsson M. Semi mature blood dendritic cells exist in patients with ductal pancreatic adenocarcinoma owing to inflammatory factors released from the tumor. PLoS One. 2010;5:e13441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
28.  Aida K, Miyakawa R, Suzuki K, Narumi K, Udagawa T, Yamamoto Y, Chikaraishi T, Yoshida T, Aoki K. Suppression of Tregs by anti-glucocorticoid induced TNF receptor antibody enhances the antitumor immunity of interferon-α gene therapy for pancreatic cancer. Cancer Sci. 2014;105:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
29.  Yamamoto T, Yanagimoto H, Satoi S, Toyokawa H, Hirooka S, Yamaki S, Yui R, Yamao J, Kim S, Kwon AH. Circulating CD4+CD25+ regulatory T cells in patients with pancreatic cancer. Pancreas. 2012;41:409-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
30.  Keenan BP, Saenger Y, Kafrouni MI, Leubner A, Lauer P, Maitra A, Rucki AA, Gunderson AJ, Coussens LM, Brockstedt DG. A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice. Gastroenterology. 2014;146:1784-94.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 99]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
31.  Shibuya KC, Goel VK, Xiong W, Sham JG, Pollack SM, Leahy AM, Whiting SH, Yeh MM, Yee C, Riddell SR. Pancreatic ductal adenocarcinoma contains an effector and regulatory immune cell infiltrate that is altered by multimodal neoadjuvant treatment. PLoS One. 2014;9:e96565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
32.  Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73:1128-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 639]  [Cited by in F6Publishing: 709]  [Article Influence: 59.1]  [Reference Citation Analysis (0)]
33.  Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, Mitchem JB, Plambeck-Suess SM, Worley LA, Goetz BD. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res. 2013;19:3404-3415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 410]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
34.  De Palma M, Lewis CE. Cancer: Macrophages limit chemotherapy. Nature. 2011;472:303-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 100]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
35.  Di Mitri D, Toso A, Alimonti A. Tumor-infiltrating myeloid cells drive senescence evasion and chemoresistance in tumors. Oncoimmunology. 2015;4:e988473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
36.  Takeuchi S, Baghdadi M, Tsuchikawa T, Wada H, Nakamura T, Abe H, Nakanishi S, Usui Y, Higuchi K, Takahashi M. Chemotherapy-Derived Inflammatory Responses Accelerate the Formation of Immunosuppressive Myeloid Cells in the Tissue Microenvironment of Human Pancreatic Cancer. Cancer Res. 2015;75:2629-2640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 96]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
37.  Markowitz J, Brooks TR, Duggan MC, Paul BK, Pan X, Wei L, Abrams Z, Luedke E, Lesinski GB, Mundy-Bosse B. Patients with pancreatic adenocarcinoma exhibit elevated levels of myeloid-derived suppressor cells upon progression of disease. Cancer Immunol Immunother. 2015;64:149-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 81]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
38.  Bruchard M, Mignot G, Derangère V, Chalmin F, Chevriaux A, Végran F, Boireau W, Simon B, Ryffel B, Connat JL. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19:57-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 494]  [Cited by in F6Publishing: 566]  [Article Influence: 47.2]  [Reference Citation Analysis (0)]
39.  Hu X, Li B, Li X, Zhao X, Wan L, Lin G, Yu M, Wang J, Jiang X, Feng W. Transmembrane TNF-α promotes suppressive activities of myeloid-derived suppressor cells via TNFR2. J Immunol. 2014;192:1320-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 129]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
40.  Sanford DE, Porembka MR, Panni RZ, Mitchem JB, Belt BA, Plambeck-Suess SM, Lin G, Denardo DG, Fields RC, Hawkins WG. A Study of Zoledronic Acid as Neo-Adjuvant, Perioperative Therapy in Patients with Resectable Pancreatic Ductal Adenocarcinoma. J Cancer Ther. 2013;4:797-803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
41.  Sato D, Tsuchikawa T, Mitsuhashi T, Hatanaka Y, Marukawa K, Morooka A, Nakamura T, Shichinohe T, Matsuno Y, Hirano S. Stromal Palladin Expression Is an Independent Prognostic Factor in Pancreatic Ductal Adenocarcinoma. PLoS One. 2016;11:e0152523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
42.  Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, Gayral M, Cordelier P, Delisle MB, Bousquet-Dubouch MP. Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Mol Med. 2015;7:735-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 148]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
43.  Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612-1616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1152]  [Cited by in F6Publishing: 1204]  [Article Influence: 92.6]  [Reference Citation Analysis (0)]
44.  Tongu M, Harashima N, Monma H, Inao T, Yamada T, Kawauchi H, Harada M. Metronomic chemotherapy with low-dose cyclophosphamide plus gemcitabine can induce anti-tumor T cell immunity in vivo. Cancer Immunol Immunother. 2013;62:383-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
45.  Giovannetti E, Funel N, Peters GJ, Del Chiaro M, Erozenci LA, Vasile E, Leon LG, Pollina LE, Groen A, Falcone A. MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010;70:4528-4538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 347]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]