Pancreatic Cancer Signaling Pathways, Genetic Alterations, and Tumor Microenvironment: The Barriers Affecting the Method of Treatment
Abstract
:1. Introduction
2. Signaling Pathways
2.1. K-Ras Oncogene
2.2. Tumor Suppressor Genes: TP53, SMAD4/DPC4, and P16/CDKN2A
2.3. Signaling of Growth Factor Receptors
2.3.1. EFGR
2.3.2. IGF
2.3.3. FGFR
2.3.4. VEGF
2.3.5. RAGE
2.4. EMT in PDAC
2.4.1. TGF-β Signaling Pathway in EMT
2.4.2. Wnt/β-Catenin Signaling Pathway
2.4.3. Signaling Pathway of Notch
2.4.4. Snail Transcription Factors
2.4.5. Zeb Transcription Factors
2.4.6. bHLH Transcription Factors
3. The Tumor Microenvironment (TME)
4. Immune Checkpoints
4.1. CTLA-4
4.2. PD-1
4.3. PD-L1
4.4. LAG3
4.5. VISTA
5. T-Cell Mediated Recognition of Somatic Mutations
6. Blockade of Immune Checkpoints in Pancreatic Cancer
7. Neoantigen Formation in Pancreatic Cancer
8. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
Elaborated Phrasese | Abbreviations |
Pancreatic ductal adenocarcinoma | PDAC |
programmed cell death protein-1 | PD-1 |
programmed cell death ligand-1 | PD-L1 |
cytotoxic T-lymphocyte-associated protein 4 | CTLA-4 |
Guanine nucleotide exchange factors | GEFs |
GTPase activating proteins | GAPs |
mitogen-activated protein kinase | MAPK |
extracellular signal-regulated kinase | Erk |
phosphoinositide 3-kinases | PI3Ks |
Phosphoinositide-dependent kinase-1 | PDK-1 |
Ral guanine nucleotide exchange factors | RalGEFs |
epidermal growth factor | EGF |
epidermal growth factor receptor | EGFR |
fibroblast growth factor | FGF |
FGF receptor | FGFR |
insulin-like growth factor | IGF |
insulin-like growth factor receptor | IGFR |
platelet-derived growth factor | PDGF |
vascular endothelial growth factor | VEGF |
transforming growth factor-α | TGF-α |
Hypoxia-inducible factor 1-alpha | HIF1-α |
Snail family of zinc finger transcription factors | Snail 1 and 2 |
zinc-finger-enhancer binding protein | Zeb-1 and 2 |
Casein kinase 1 | CK1 |
lymphoid enhancer factor | LEF |
tumor microenvironment | TME |
antigen-presenting cells | APCs |
interferon-γ | IFN-γ |
Interleukin-2 | IL-2 |
tumor-infiltrating lymphocytes | TILs |
Lymphocyte Activation Gene 3 | LAG3 |
basic helix-loop-helix | bHLH |
Regulatory T cells | Tregs |
plasmacytoid dendritic cells | pDCs |
V-domain Ig suppressor of T cell activation | VISTA |
Signal transducer and activator of transcription 3 | STAT3 |
major histocompatibility complex | MHC |
Nonsmall-cell lung carcinoma | NSCLC |
Renal cell carcinoma | RCC |
Hepatocellular carcinoma | HCC |
High microsatellite instability | MSI-H |
Microsatellite instable | MSI |
carboxy terminal Cys2His2 | C2H2 |
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 70. [Google Scholar] [CrossRef]
- Mathur, P.; Sathishkumar, K.; Chaturvedi, M.; Das, P.; Sudarshan, K.L.; Santhappan, S.; Nallasamy, V.; John, A.; Narasimhan, S.; Roselind, F.S.; et al. Cancer Statistics, 2020: Report from National Cancer Registry Programme, India. JCO Glob. Oncol. 2020, 6, 1063–1075. [Google Scholar] [CrossRef] [PubMed]
- Thomas, R.M.; Kim, J.; Revelo-Penafiel, M.P.; Angel, R.; Dawson, D.W.; Lowy, A.M. The chemokine receptor CXCR4 is expressed in pancreatic intraepithelial neoplasia. Gut 2008, 57, 1555–1560. [Google Scholar] [CrossRef]
- Kim, J.; Yip, M.L.R.; Shen, X.; Li, H.; Hsin, L.-Y.C.; LaBarge, S.; Heinrich, E.L.; Lee, W.; Lu, J.; Vaidehi, N. Identification of Anti-Malarial Compounds as Novel Antagonists to Chemokine Receptor CXCR4 in Pancreatic Cancer Cells. PLoS ONE 2012, 7, e31004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, X.; Artinyan, A.; Jackson, D.; Thomas, R.M.; Lowy, A.M.; Kim, J. Chemokine Receptor CXCR4 Enhances Proliferation in Pancreatic Cancer Cells Through AKT and ERK Dependent Pathways. Pancreas 2010, 39, 81–87. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Heinrich, E.L.; Li, L.; Lu, J.; Choi, A.H.; Levy, R.A.; Wagner, J.E.; Yip, M.R.; Vaidehi, N.; Kim, J. CCR9-mediated signaling through β-catenin and identification of a novel CCR9 antagonist. Mol. Oncol. 2015, 9, 1599–1611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.-L.; Gourgou-Bourgade, S.; De La Fouchardière, C.; et al. Folfirinox versus Gemcitabine for Metastatic Pancreatic Cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [Green Version]
- Lynch, D.V.; Lin, T.-T.; Myers, S.P.; Leibo, S.P.; MacIntyre, R.J.; Pitt, R.E.; Steponkus, P.L. A two-step method for permeabilization of Drosophila eggs. Cryobiology 1989, 26, 445–452. [Google Scholar] [CrossRef]
- Wattenberg, M.M.; Asch, D.; Yu, S.; O’Dwyer, P.J.; Domchek, S.M.; Nathanson, K.L.; Rosen, M.A.; Beatty, G.L.; Siegelman, E.S.; Reiss, K.A. Platinum response characteristics of patients with pancreatic ductal adenocarcinoma and a germline BRCA1, BRCA2 or PALB2 mutation. Br. J. Cancer 2020, 122, 333–339. [Google Scholar] [CrossRef]
- Gnoni, A.; Brunetti, O.; Longo, V.; Calabrese, A.; Argentiero, A.L.; Calbi, R.; Solimando Antonio, G.; Licchetta, A. Immune system and bone microenvironment: Rationale for targeted cancer therapies. Oncotarget 2020, 11, 480–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Waddell, N.; Pajic, M.; Patch, A.-M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, S.; Zhang, X.; Parsons, D.W.; Lin, J.C.-H.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Kamiyama, H.; Jimeno, A.; et al. Core Signaling Pathways in Human Pancreatic Cancers Revealed by Global Genomic Analyses. Science 2008, 321, 1801–1806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [Green Version]
- Kanda, M.; Matthaei, H.; Wu, J.; Hong, S.; Yu, J.; Borges, M.; Hruban, R.H.; Maitra, A.; Kinzler, K.; Vogelstein, B.; et al. Presence of Somatic Mutations in Most Early-Stage Pancreatic Intraepithelial Neoplasia. Gastroenterology 2012, 142, 730–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Leone, P.; Di Lernia, G.; Solimando, A.G.; Cicco, S.; Saltarella, I.; Lamanuzzi, A.; Ria, R.; Frassanito, M.A.; Ponzoni, M.; Ditonno, P.; et al. Bone marrow endothelial cells sustain a tumor-specific CD8+ T cell subset with suppressive function in myeloma patients. Oncoimmunology 2018, 8, e1486949. [Google Scholar] [CrossRef] [Green Version]
- Collins, M.A.; Bednar, F.; Zhang, Y.; Brisset, J.-C.; Galbán, S.; Galbán, C.J.; Rakshit, S.; Flannagan, K.S.; Adsay, N.V.; Di Magliano, M.P. Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. J. Clin. Investig. 2012, 122, 639–653. [Google Scholar] [CrossRef] [Green Version]
- Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer 2003, 3, 11–22. [Google Scholar] [CrossRef]
- Eser, S.; Reiff, N.; Messer, M.; Seidler, B.; Gottschalk, K.; Dobler, M.; Hieber, M.; Arbeiter, A.; Klein, S.; Kong, B.; et al. Selective Requirement of PI3K/PDK1 Signaling for Kras Oncogene-Driven Pancreatic Cell Plasticity and Cancer. Cancer Cell 2013, 23, 406–420. [Google Scholar] [CrossRef] [Green Version]
- Maitra, A.; Kern, S.E.; Hruban, R.H. Molecular pathogenesis of pancreatic cancer. Best Pract. Res. Clin. Gastroenterol. 2006, 20, 211–226. [Google Scholar] [CrossRef] [PubMed]
- Aguirre, A.J.; Bardeesy, N.; Sinha, M.; Lopez, L.; Tuveson, D.A.; Horner, J.; Redston, M.S.; Depinho, R.A. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes Dev. 2003, 17, 3112–3126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Redston, M.S.; Caldas, C.; Seymour, A.B.; Hruban, R.H.; Da Costa, L.; Yeo, C.J.E.; Kern, S. p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res. 1994, 54, 3025–3033. [Google Scholar] [PubMed]
- Siegel, P.M.; Massagué, J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nat. Rev. Cancer 2003, 3, 807–820. [Google Scholar] [CrossRef]
- Tascilar, M.; Skinner, H.G.; Rosty, C.; Sohn, T.E.; Wilentz, R.; Offerhaus, G.J.; Adsay, V.A.; Abrams, R.; Cameron, J.L.E.; Kern, S.; et al. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2001, 7, 4115–4121. [Google Scholar]
- Baláz, P.; Friess, H.; Büchler, M.W. Growth factors in pancreatic health and disease. Pancreatology 2001, 1, 343–355. [Google Scholar] [CrossRef] [PubMed]
- Troiani, T.; Martinelli, E.; Capasso, A.; Morgillo, F.; Orditura, M.; De Vita, F.; Ciardiello, F. Targeting EGFR in pancreatic cancer treatment. Curr. Drug Targets 2012, 13, 802–810. [Google Scholar] [CrossRef] [PubMed]
- Tobita, K.; Kijima, H.; Dowaki, S.; Kashiwagi, H.; Ohtani, Y.; Oida, Y.; Yamazaki, H.; Nakamura, M.; Ueyama, Y.; Tanaka, M.; et al. Epidermal growth factor receptor expression in human pancreatic cancer: Significance for liver metastasis. Int. J. Mol. Med. 2003, 11, 305–309. [Google Scholar] [CrossRef]
- Moschos, S.J.; Mantzoros, C.S. The Role of the IGF System in Cancer: From Basic to Clinical Studies and Clinical Applications. Oncology 2002, 63, 317–332. [Google Scholar] [CrossRef]
- Valsecchi, M.E.; McDonald, M.; Brody, J.R.; Hyslop, T.; Freydin, B.; Yeo, C.J.; Solomides, C.; Peiper, S.C.; Witkiewicz, A.K. Epidermal growth factor receptor and insulin-like growth factor 1 receptor expression predict poor survival in pancreatic ductal adenocarcinoma. Cancer 2012, 118, 3484–3493. [Google Scholar] [CrossRef]
- Rieder, S.W.; Michalski, C.; Friess, H. Insulin-like growth factor signaling as a therapeutic target in pancreatic cancer. Anti-Cancer Agents in Medicinal Chemistry. Anti-Cancer Agents Med. Chem. 2011, 11, 427–433. [Google Scholar] [CrossRef]
- Zhang, H.; Hylander, B.L.; LeVea, C.A.; Repasky, E.; Straubinger, R.M.A.; Adjei, A.; Ma, W.W. Enhanced FGFR signalling predisposes pancreatic cancer to the effect of a potent FGFR inhibitor in preclinical models. Br. J. Cancer 2013, 110, 320–329. [Google Scholar] [CrossRef] [Green Version]
- Polireddy, K.; Chen, Q. Cancer of the Pancreas: Molecular Pathways and Current Advancement in Treatment. J. Cancer 2016, 7, 1497–1514. [Google Scholar] [CrossRef] [Green Version]
- Seo, Y.; Baba, H.; Fukuda, T.; Takashima, M.; Sugimachi, K. High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer 2000, 88, 2239–2245. [Google Scholar] [CrossRef]
- Hotz, H.G.A.; Reber, H.; Hotz, B.; Sanghavi, P.C.; Yu, T.; Foitzik, T.; Buhr, H.J.; Hines, O.J. Angiogenesis inhibitor TNP-470 reduces human pancreatic cancer growth. J. Gastrointest. Surg. 2001, 5, 131–138. [Google Scholar] [CrossRef]
- Solorzano, C.C.; Baker, C.H.; Bruns, C.J.; Killion, J.J.; Ellis, L.M.; Wood, J.M.; Fidler, I.J. Inhibition of Growth and Metastasis of Human Pancreatic Cancer Growing in Nude Mice by PTK 787/ZK222584, an Inhibitor of the Vascular Endothelial Growth Factor Receptor Tyrosine Kinases. Cancer Biother. Radiopharm. 2001, 16, 359–370. [Google Scholar] [CrossRef]
- Kang, R.; Tang, D.E.; Schapiro, N.; Loux, T.; Livesey, K.M.; Billiar, T.R.; Wang, H.; Van Houten, B.; Lotze, M.T.; Zeh, H.J. The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics. Oncogene 2014, 33, 567–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H.; et al. EMT and Dissemination Precede Pancreatic Tumor Formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef] [Green Version]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zeisberg, M.; Neilson, E.G. Biomarkers for epithelial-mesenchymal transitions. J. Clin. Investig. 2009, 119, 1429–1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thiery, J.P.; Sleeman, J.P. Complex networks orchestrate epithelial–mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 2006, 7, 131–142. [Google Scholar] [CrossRef] [PubMed]
- Heldin, C.-H.; Vanlandewijck, M.; Moustakas, A. Regulation of EMT by TGFβ in cancer. FEBS Lett. 2012, 586, 1959–1970. [Google Scholar] [CrossRef] [Green Version]
- Derynck, R.; Zhang, Y.E. Smad-dependent and Smad-independent pathways in TGF-β family signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef] [PubMed]
- Levy, L.; Hill, C.S. Smad4 Dependency Defines Two Classes of Transforming Growth Factor β (TGF-β) Target Genes and Distinguishes TGF-β-Induced Epithelial-Mesenchymal Transition from its Antiproliferative and Migratory Responses. Mol. Cell. Biol. 2005, 25, 8108–8125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ellenrieder, V.; Hendler, S.F.; Boeck, W.; Seufferlein, T.; Menke, A.; Ruhland, C.; Adler, G.; Gress, T.M. Transforming growth factor β1 treatment leads to an epithelial-mesenchymal transdifferentiation of pancreatic cancer cells requiring extracellular signal-regulated kinase 2 activation. Cancer Res. 2001, 61, 4222–4228. [Google Scholar] [PubMed]
- Clevers, H. Wnt/β-Catenin Signaling in Development and Disease. Cell 2006, 127, 469–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, W.; Wang, Z.; Zhang, W.; Qian, K.; Li, H.; Kong, D.; Li, Y.; Tang, Y. Mutated K-ras activates CDK8 to stimulate the epithelial-to-mesenchymal transition in pancreatic cancer in part via the Wnt/β-catenin signaling pathway. Cancer Lett. 2015, 356, 613–627. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.-H.; Luo, Y.; Jiang, Y.-G.; He, D.-L.; Wu, C.-T. Knockdown of β-Catenin through shRNA cause a reversal of EMT and metastatic phenotypes induced by HIF-1α. Cancer Investig. 2011, 29, 377–382. [Google Scholar] [CrossRef]
- Guruharsha, K.G.; Kankel, M.W.; Artavanis-Tsakonas, S. The Notch signalling system: Recent insights into the complexity of a conserved pathway. Nat. Rev. Genet. 2012, 13, 654–666. [Google Scholar] [CrossRef] [Green Version]
- Krebs, M.; Solimando, A.G.; Kalogirou, C.; Marquardt, A.; Frank, T.; Sokolakis, I.; Hatzichristodoulou, G.; Kneitz, S.; Bargou, R.; Kübler, H.; et al. miR-221-3p Regulates VEGFR2 Expression in High-Risk Prostate Cancer and Represents an Escape Mechanism from Sunitinib In Vitro. J. Clin. Med. 2020, 9, 670. [Google Scholar] [CrossRef] [Green Version]
- Brunetti, O.; Gnoni, A.; Licchetta, A.; Longo, V.; Calabrese, A.; Argentiero, A.; Delcuratolo, S.; Solimando, A.G.; Casadei-Gardini, A.; Silvestris, N. Predictive and Prognostic Factors in HCC Patients Treated with Sorafenib. Medicina 2019, 55, 707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamanuzzi, A.; Saltarella, I.; DeSantis, V.; Frassanito, M.A.; Leone, P.; Racanelli, V.; Nico, B.; Ribatti, D.; Ditonno, P.; Prete, M.; et al. Inhibition of mTOR complex 2 restrains tumor angiogenesis in multiple myeloma. Oncotarget 2018, 9, 20563–20577. [Google Scholar] [CrossRef] [Green Version]
- Güngör, C.; Zander, H.; Effenberger, K.E.; Vashist, Y.K.; Kalinina, T.; Izbicki, J.R.; Yekebas, E.; Bockhorn, M. Notch signaling activated by replication stress-induced expression of midkine drives epithelial-mesenchymal transition and chemo-resistance in pancreatic cancer. Cancer Res. 2011, 71, 5009–5019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nieto, M.A. The snail superfamily of zinc-finger transcription factors. Nat. Rev. Mol. Cell Biol. 2002, 3, 155–166. [Google Scholar] [CrossRef] [PubMed]
- Hotz, B.; Arndt, M.; Dullat, S.; Bhargava, S.; Buhr, H.-J.; Hotz, H.G. Epithelial to Mesenchymal Transition: Expression of the Regulators Snail, Slug and Twist in Pancreatic Cancer. Clin. Cancer Res. 2007, 13, 4769–4776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nishioka, R.; Itoh, S.; Gui, T.; Gai, Z.; Oikawa, K.; Kawai, M.; Tani, M.; Yamaue, H.; Muragaki, Y. SNAIL induces epithelial-to-mesenchymal transition in a human pancreatic cancer cell line (BxPC3) and promotes distant metastasis and invasiveness in vivo. Exp. Mol. Pathol. 2010, 89, 149–157. [Google Scholar] [CrossRef]
- Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.-C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemo-resistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
- Peinado, H.; Ballestar, E.; Esteller, M.; Cano, A. Snail Mediates E-Cadherin Repression by the Recruitment of the Sin3A/Histone Deacetylase 1 (HDAC1)/HDAC2 Complex. Mol. Cell. Biol. 2004, 24, 306–319. [Google Scholar] [CrossRef] [Green Version]
- Bronsert, P.; Kohler, I.; Timme, S.; Kiefer, S.; Werner, M.; Schilling, O.; Vashist, Y.; Makowiec, F.; Brabletz, T.; Hopt, U.T.; et al. Prognostic significance of Zinc finger E-box binding homeobox 1 (ZEB1) expression in cancer cells and cancer-associated fibroblasts in pancreatic head cancer. Surgery 2014, 156, 97–108. [Google Scholar] [CrossRef]
- Aghdassi, A.; Sendler, M.; Guenther, A.; Mayerle, J.; Behn, C.-O.; Heidecke, C.-D.; Friess, H.; Büchler, M.; Evert, M.; Lerch, M.M.; et al. Recruitment of histone deacetylases HDAC1 and HDAC2 by the transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancer. Gut 2012, 61, 439–448. [Google Scholar] [CrossRef] [PubMed]
- Wellner, U.F.; Schubert, J.; Burk, U.C.; Schmalhofer, O.; Zhu, F.; Sonntag, A.; Waldvogel, B.; Vannier, C.; Darling, D.; Zur Hausen, A.; et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat. Cell Biol. 2009, 11, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
- Teng, Y.; Li, X. The roles of HLH transcription factors in epithelial mesenchymal transition and multiple molecular mechanisms. Clin. Exp. Metastasis 2013, 31, 367–377. [Google Scholar] [CrossRef]
- Pérez-Moreno, M.A.; Locascio, A.; Rodrigo, I.; Dhondt, G.; Portillo, F.; Nieto, M.A.; Cano, A. A new role for E12/E47 in the repression ofe-cadherin expression and epithelial-mesenchymal transitions. J. Biol. Chem. 2001, 276, 27424–27431. [Google Scholar] [CrossRef] [Green Version]
- Ansieau, S.; Morel, A.-P.; Hinkal, G.; Bastid, J.; Puisieux, A. Twisting an embryonic transcription factor into an oncoprotein. Oncogene 2010, 29, 3173–3184. [Google Scholar] [CrossRef] [Green Version]
- Büchler, P.; Reber, H.A.; Lavey, R.S.; Tomlinson, J.; Büchler, M.W.; Friess, H.; Hines, O.J. Tumor hypoxia correlates with metastatic tumor growth of pancreatic cancer in an orthotopic murine model1. J. Surg. Res. 2004, 120, 295–303. [Google Scholar] [CrossRef] [PubMed]
- Yang, F.; Sun, L.; Li, Q.; Han, X.; Lei, L.; Zhang, H.; Shang, Y. SET8 promotes epithelial-mesenchymal transition and confers TWIST dual transcriptional activities. EMBO J. 2011, 31, 110–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, J.; Qin, L.; He, T.; Qin, J.; Hong, J.; Wong, J.; Liao, L.; Xu, J. The TWIST/Mi2/NuRD protein complex and its essential role in cancer metastasis. Cell Res. 2011, 21, 275–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoseini, S.S.; Cheung, N.-K.V. Immunotherapy of hepatocellular carcinoma using chimeric antigen receptors and bispecific antibodies. Cancer Lett. 2017, 399, 44–52. [Google Scholar] [CrossRef] [PubMed]
- Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef] [PubMed]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.-J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and Activity of Anti-PD-L1 Antibody in Patients with Advanced Cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
- Puls, T.J.; Tan, X.; Whittington, C.F.; Voytik-Harbin, S.L. 3D collagen fibrillar microstructure guides pancreatic cancer cell phenotype and serves as a critical design parameter for phenotypic models of EMT. PLoS ONE 2017, 12, e0188870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuczek, D.E.; Larsen, A.M.H.; Thorseth, M.-L.; Carretta, M.; Kalvisa, A.; Siersbæk, M.S.; Simões, A.M.C.; Roslind, A.; Engelholm, L.H.; Noessner, E.; et al. Collagen density regulates the activity of tumor-infiltrating T cells. J. Immunother. Cancer 2019, 7, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weniger, M.; Honselmann, K.C.; Liss, A.S. The Extracellular Matrix and Pancreatic Cancer: A Complex Relationship. Cancers 2018, 10, 316. [Google Scholar] [CrossRef] [Green Version]
- Sato, N.; Kohi, S.; Hirata, K.; Goggins, M. Role of hyaluronan in pancreatic cancer biology and therapy: Once again in the spotlight. Cancer Sci. 2016, 107, 569–575. [Google Scholar] [CrossRef]
- Russano, M.; Napolitano, A.; Ribelli, G.; Iuliani, M.; Simonetti, S.; Citarella, F.; Pantano, F.; Dell’Aquila, E.; Anesi, C.; Silvestris, N.; et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: The potentiality of blood samples. J. Exp. Clin. Cancer Res. 2020, 39, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Yu, P.; Fu, Y.-X. Tumor-infiltrating T lymphocytes: Friends or foes? Lab. Investig. 2006, 86, 231–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Q.; Liao, Q.; Zhao, Y. Chemotherapy and tumor microenvironment of pancreatic cancer. Cancer Cell Int. 2017, 17, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Brown, K.E.; Freeman, G.J.; Wherry, E.J.; Sharpe, A.H. Role of PD-1 in regulating acute infections. Curr. Opin. Immunol. 2010, 22, 397–401. [Google Scholar] [CrossRef] [Green Version]
- Goldberg, M.V.; Drake, C.G. LAG-3 in cancer immunotherapy. In Cancer Immunology and Immunotherapy; Springer: Berlin/Heidelberg, Germany, 2010; pp. 269–278. [Google Scholar]
- Gravitz, L. Cancer immunotherapy. Nature 2013, 504. [Google Scholar] [CrossRef]
- Couzin-Frankel, J. Cancer Immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef] [Green Version]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, Activity and Immune Correlates of Anti-PD-1 Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
- Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Ann. Rev. Immunol. 2001, 19, 565–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Egen, J.G.; Kuhns, M.S.; Allison, J.P. CTLA-4: New insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002, 3, 611–618. [Google Scholar] [CrossRef] [PubMed]
- Krummel, M.F.; Allison, J.P. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Masteller, E.L.; Chuang, E.; Mullen, A.C.; Reiner, S.L.; Thompson, C.B. Structural analysis of CTLA-4 function in vivo. J. Immunol. 2000, 164, 5319–5327. [Google Scholar] [CrossRef] [Green Version]
- Schneider, H.; Downey, J.; Smith, A.; Zinselmeyer, B.H.; Rush, C.; Brewer, J.M.; Wei, B.; Hogg, N.; Garside, P.; Rudd, C.E. Reversal of the TCR stop signal by CTLA-4. Science 2006, 313, 1972–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Linsley, P.S.; Bradshaw, J.; Greene, J.; Peach, R.; Bennett, K.L.; Mittler, R.S. Intracellular Trafficking of CTLA-4 and Focal Localization Towards Sites of TCR Engagement. Immunology 1996, 4, 535–543. [Google Scholar] [CrossRef] [Green Version]
- Wing, K.; Onishi, Y.; Prieto-Martin, P.; Yamaguchi, T.; Miyara, M.; Fehervari, Z.; Nomura, T.; Sakaguchi, S. CTLA-4 Control over Foxp3+ Regulatory T Cell Function. Science 2008, 322, 271–275. [Google Scholar] [CrossRef]
- Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 pathways: Similarities, differences and implications of their inhibition. Am. J. Clin. Oncol. 2016, 39, 98. [Google Scholar] [CrossRef] [Green Version]
- Fife, B.T.; Bluestone, J.A. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 2008, 224, 166–182. [Google Scholar] [CrossRef]
- Chen, D.S.; Irving, B.A.; Hodi, F.S. Molecular Pathways: Next-Generation Immunotherapy—Inhibiting Programmed Death-Ligand 1 and Programmed Death-1. Clin. Cancer Res. 2012, 18, 6580–6587. [Google Scholar] [CrossRef] [Green Version]
- Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
- Chen, L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat. Rev. Immunol. 2004, 4, 336–347. [Google Scholar] [CrossRef] [PubMed]
- Hino, R.; Kabashima, K.; Kato, Y.; Yagi, H.; Nakamura, M.; Honjo, T.; Okazaki, T.; Tokura, Y. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 2010, 116, 1757–1766. [Google Scholar] [CrossRef] [PubMed]
- Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti-PD-1 Therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, W.; Chen, L. Inhibitory B7-family molecules in the tumour microenvironment. Nat. Rev. Immunol. 2008, 8, 467–477. [Google Scholar] [CrossRef]
- Wang, L.; Pino-Lagos, K.; De Vries, V.C.; Guleria, I.; Sayegh, M.H.; Noelle, R.J. Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells. Proc. Natl. Acad. Sci. USA 2008, 105, 9331–9336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Francisco, L.M.; Salinas, V.H.; Brown, K.E.; Vanguri, V.K.; Freeman, G.J.; Kuchroo, V.K.; Sharpe, A.H. PD-L1 regulates the development, maintenance and function of induced regulatory T cells. J. Exp. Med. 2009, 206, 3015–3029. [Google Scholar] [CrossRef]
- Akbari, O.; Stock, P.; Singh, A.; Lombardi, V.; Lee, W.; Freeman, G.; Sharpe, A.; Umetsu, D.; Dekruyff, R. PD-L1 and PD-L2 modulate airway inflammation and iNKT-cell-dependent airway hyperreactivity in opposing directions. Mucosal Immunol. 2010, 3, 81–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huber, S.; Hoffmann, R.; Muskens, F.; Voehringer, D. Alternatively activated macrophages inhibit T-cell proliferation by Stat6-dependent expression of PD-L2. Blood 2010, 116, 3311–3320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Butte, M.J.; Keir, M.E.; Phamduy, T.B.; Sharpe, A.H.; Freeman, G.J. Programmed Death-1 Ligand 1 Interacts Specifically with the B7-1 Costimulatory Molecule to Inhibit T Cell Responses. Immunity 2007, 27, 111–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruffo, E.; Wu, R.C.; Bruno, T.C.; Workman, C.J.; Vignali, D.A. Lymphocyte-activation gene 3 (LAG3): The next immune checkpoint receptor. Semin. Immunol. 2019, 42, 101305. [Google Scholar] [CrossRef] [PubMed]
- Workman, C.J.; Dugger, K.J.; Vignali, D.A.A. Cutting Edge: Molecular Analysis of the Negative Regulatory Function of Lymphocyte Activation Gene-3. J. Immunol. 2002, 169, 5392–5395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Workman, C.J.; Vignali, D.A.A. Negative Regulation of T Cell Homeostasis by Lymphocyte Activation Gene-3 (CD223). J. Immunol. 2005, 174, 688–695. [Google Scholar] [CrossRef] [PubMed]
- Andrews, L.P.; Marciscano, A.E.; Drake, C.G.; Vignali, D.A.A. LAG3 (CD223) as a cancer immunotherapy target. Immunol. Rev. 2017, 276, 80–96. [Google Scholar] [CrossRef]
- Zhang, Q.; Chikina, M.; Szymczak-Workman, A.L.; Horne, W.; Kolls, J.K.; Vignali, K.M.; Normolle, D.; Bettini, M.; Workman, C.J.; Vignali, D.A.A. LAG3 limits regulatory T cell proliferation and function in autoimmune diabetes. Sci. Immunol. 2017, 2, eaah4569. [Google Scholar] [CrossRef] [Green Version]
- Huard, B.; Prigent, P.; Tournier, M.; Bruniquel, D.; Triebel, F. CD4/major histocompatibility complex class II interaction analyzed with CD4-and lymphocyte activation gene-3 (LAG-3) Ig fusion proteins. Eur. J. Immunol. 1995, 25, 2718–2721. [Google Scholar] [CrossRef]
- Flies, D.B.; Wang, S.; Xu, H.; Chen, L. Cutting Edge: A Monoclonal Antibody Specific for the Programmed Death-1 Homolog Prevents Graft-versus-Host Disease in Mouse Models. J. Immunol. 2011, 187, 1537–1541. [Google Scholar] [CrossRef]
- Liu, J.; Xie, X.; Xuan, C.; Li, T.; Wang, L.; Teng, L.; Liu, J. High-Density Infiltration of V-domain Immunoglobulin Suppressor of T-cell Activation Up-regulated Immune Cells in Human Pancreatic Cancer. Pancreas 2018, 47, 725–731. [Google Scholar] [CrossRef]
- Wang, L.; Rubinstein, R.; Lines, J.L.; Wasiuk, A.; Ahonen, C.; Guo, Y.; Lu, L.-F.; Gondek, D.; Wang, Y.; Fava, R.A.; et al. Vista, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 2011, 208, 577–592. [Google Scholar] [CrossRef] [PubMed]
- Lines, J.L.; Pantazi, E.; Mak, J.; Sempere, L.F.; Wang, L.; O’Connell, S.; Ceeraz, S.; Suriawinata, A.A.; Yan, S.; Ernstoff, M.S.; et al. Vista is an Immune Checkpoint Molecule for Human T Cells. Cancer Res. 2014, 74, 1924–1932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flies, D.B.; Han, X.; Higuchi, T.; Zheng, L.; Sun, J.; Ye, J.J.; Chen, L. Coinhibitory receptor PD-1H preferentially suppresses CD4+ T cell-mediated immunity. J. Clin. Investig. 2014, 124, 1966–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blando, J.; Sharma, A.; Higa, M.G.; Zhao, H.; Vence, L.; Yadav, S.S.; Kim, J.; Sepulveda, A.M.; Sharp, M.; Maitra, A.; et al. Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 1692–1697. [Google Scholar] [CrossRef] [Green Version]
- Huang, X.; Zhang, X.; Li, E.; Zhang, G.; Wang, X.; Tang, T.; Bai, X.; Liang, T. Vista: An immune regulatory protein checking tumor and immune cells in cancer immunotherapy. J. Hematol. Oncol. 2020, 13, 1–13. [Google Scholar] [CrossRef]
- Coulie, P.G.; Van den Eynde, B.J.; Van Der Bruggen, P.; Boon-Falleur, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146. [Google Scholar] [CrossRef]
- Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 2020, 11, 1–3. [Google Scholar] [CrossRef]
- Henriksen, A.; Dyhl-Polk, A.; Chen, I.; Nielsen, D. Checkpoint inhibitors in pancreatic cancer. Cancer Treat. Rev. 2019, 78, 17–30. [Google Scholar] [CrossRef]
- Eso, Y.; Shimizu, T.; Takeda, H.; Takai, A.; Marusawa, H. Microsatellite instability and immune checkpoint inhibitors: Toward precision medicine against gastrointestinal and hepatobiliary cancers. J. Gastroenterol. 2020, 55, 15–26. [Google Scholar] [CrossRef] [Green Version]
- Hunter, K.A.; Socinski, M.A.; Villaruz, L.C. PD-L1 Testing in Guiding Patient Selection for PD-1/PD-L1 Inhibitor Therapy in Lung Cancer. Mol. Diagn. Ther. 2018, 22, 1–10. [Google Scholar] [CrossRef]
- Mehnert, J.M.; Panda, A.; Zhong, H.; Hirshfield, K.; Damare, S.; Lane, K.; Sokol, L.; Stein, M.N.; Rodriguez-Rodriquez, L.; Kaufman, H.L.; et al. Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer. J. Clin. Investig. 2016, 126, 2334–2340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johanns, T.M.; Miller, C.A.; Dorward, I.G.; Tsien, C.; Chang, E.; Perry, A.; Uppaluri, R.; Ferguson, C.; Schmidt, R.E.; Dahiya, S.; et al. Immunogenomics of Hypermutated Glioblastoma: A Patient with Germline POLE Deficiency Treated with Checkpoint Blockade Immunotherapy. Cancer Discov. 2016, 6, 1230–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahn, S.-M.; Ansari, A.A.; Kim, J.; Kim, D.; Chun, S.-M.; Kim, J.; Kim, T.W.; Park, I.; Yu, C.-S.; Jang, S.J. The somatic POLE P286R mutation defines a unique subclass of colorectal cancer featuring hypermutation, representing a potential genomic biomarker for immunotherapy. Oncotarget 2016, 7, 68638–68649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, B.; Märkl, B. Immunologic Biomarkers and Biomarkers for Immunotherapies in Gastrointestinal Cancer. Visc. Med. 2019, 35, 3–10. [Google Scholar] [CrossRef]
- Fessler, J.; Matson, V.; Gajewski, T.F. Exploring the emerging role of the microbiome in cancer immunotherapy. J. Immunother. Cancer 2019, 7, 108. [Google Scholar] [CrossRef] [PubMed]
- Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; Lucas, A.S.; et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2019, 178, 795–806. [Google Scholar] [CrossRef] [PubMed]
- Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef] [Green Version]
- Gettinger, S.; Rizvi, N.A.; Chow, L.Q.; Borghaei, H.; Brahmer, J.; Ready, N.; Gerber, D.E.; Shepherd, F.A.; Antonia, S.; Goldman, J.W.; et al. Nivolumab Monotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2016, 34, 2980–2987. [Google Scholar] [CrossRef]
- Ibarrondo, F.J.; Comin-Anduix, B.; Escuin-Ordinas, H. Tremelimumab: Research and clinical development. OncoTargets Ther. 2016, 9, 1767–1776. [Google Scholar] [CrossRef] [Green Version]
- O’Reilly, E.M.; Oh, D.-Y.; Dhani, N.; Renouf, D.J.; Lee, M.A.; Sun, W.; Fisher, G.A.; Hezel, A.F.; Chang, S.-C.; Vlahovic, G.; et al. A randomized phase 2 study of durvalumab monotherapy and in combination with tremelimumab in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC): ALPS study. J. Clin. Oncol. 2018, 36, 217. [Google Scholar] [CrossRef]
- Duffy, A.G.; Makarova-Rusher, O.V.E.; Kleiner, D.; Alewine, C.; Figg, W.D.; Steinberg, S.M.; Levy, E.B.; Krishnasamy, V.P.; Wood, B.J.; Citrin, D.E.; et al. A pilot study of immune checkpoint inhibition in combination with radiation therapy in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2017, 35, e15786. [Google Scholar] [CrossRef]
- Renouf, D.J.; Dhani, N.C.; Kavan, P.; Jonker, D.J.; Wei, A.C.-C.; Hsu, T.; Tang, P.A.; Graham, B.; Gallinaro, L.; Hasan, T.; et al. The Canadian Cancer Trials Group PA.7 trial: Results from the safety run in of a randomized phase II study of gemcitabine (GEM) and nab-paclitaxel (Nab-P) versus GEM, nab-P, durvalumab (D) and tremelimumab (T) as first-line therapy in metastatic pancreatic ductal adenocarcinoma (mPDAC). J. Clin. Oncol. 2018, 36, 349. [Google Scholar] [CrossRef]
- Katz, M.H.G.; Varadhachary, G.R.; Bauer, T.W.; Acquavella, N.; Merchant, N.B.; Le, T.M.; Javle, M.M.; Shroff, R.T.; Overman, M.J.; Fogelman, D.R.; et al. Preliminary safety data from a randomized multicenter phase Ib/II study of neoadjuvant chemoradiation therapy (CRT) alone or in combination with pembrolizumab in patients with resectable or borderline resectable pancreatic cancer. J. Clin. Oncol. 2017, 35, 4125. [Google Scholar] [CrossRef]
- Aglietta, M.; Barone, C.; Sawyer, M.; Moore, M.; Miller, W., Jr.; Bagalà, C.; Colombi, F.; Cagnazzo, C.; Gioeni, L.; Wang, E. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann. Oncol. 2014, 25, 1750–1755. [Google Scholar] [CrossRef] [PubMed]
- Kalyan, A.; Kircher, S.M.; Mohindra, N.A.; Nimeiri, H.S.; Maurer, V.; Rademaker, A.; Benson, A.B.; Mulcahy, M.F. Ipilimumab and gemcitabine for advanced pancreas cancer: A phase Ib study. J. Clin. Oncol. 2016, 34, e15747. [Google Scholar] [CrossRef]
- Weiss, G.J.; Blaydorn, L.; Beck, J.; Bornemann-Kolatzki, K.; Urnovitz, H.; Schütz, E.; Khemka, V. Phase Ib/II study of gemcitabine, nab-paclitaxel and pembrolizumab in metastatic pancreatic adenocarcinoma. Investig. New Drugs 2018, 36, 96–102. [Google Scholar] [CrossRef]
- Mahalingam, D.; Fountzilas, C.; Moseley, J.L.; Noronha, N.; Cheetham, K.; Dzugalo, A.; Nuovo, G.; Gutierrez, A.; Arora, S.P. A study of REOLYSIN in combination with pembrolizumab and chemotherapy in patients (pts) with relapsed metastatic adenocarcinoma of the pancreas (MAP). J. Clin. Oncol. 2017, 35, e15753. [Google Scholar] [CrossRef]
- Wainberg, Z.A.; Hochster, H.S.; George, B.; Gutierrez, M.; Johns, M.E.; Chiorean, E.G.; Kwak, E.L.; Kalyan, A.; Manax, V.; Ye, M.; et al. Phase I study of nivolumab (nivo) + nab-paclitaxel (nab-P) ± gemcitabine (Gem) in solid tumors: Interim results from the pancreatic cancer (PC) cohorts. J. Clin. Oncol. 2017, 35, 412. [Google Scholar] [CrossRef]
- Borazanci, E.H.; Jameson, G.S.; Borad, M.J.; Ramanathan, R.K.; Korn, R.L.; Caldwell, L.; Ansaldo, K.; Hendrickson, K.; Marceau, K.; Von Hoff, D.D. A phase II pilot trial of nivolumab (N) + albumin bound paclitaxel (AP) + paricalcitol (P) + cisplatin (C) + gemcitabine (G) (NAPPCG) in patients with previously untreated metastatic pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2018, 36, 358. [Google Scholar] [CrossRef]
- O’Hara, M.H.; O’Reilly, E.M.; Rosemarie, M.; Varadhachary, G.; Wainberg, Z.A.; Ko, A.; Fisher, G.A.; Rahma, O.; Lyman, J.P.; Cabanski, C.R. Abstract CT004: A Phase Ib Study of CD40 Agonistic Monoclonal Antibody APX005M Together with Gemcitabine (Gem) and Nab-Paclitaxel (NP) With or Without Nivolumab (Nivo) in Untreated Metastatic Ductal Pancreatic Adenocarcinoma (PDAC) Patients; AACR: Philadelphia, PA, USA, 2019. [Google Scholar]
- Wang-Gillam, A.; Plambeck-Suess, S.; Goedegebuure, P.; Simon, P.O.; Mitchem, J.B.; Hornick, J.R.; Sorscher, S.; Picus, J.; Suresh, R.; Lockhart, A.C.; et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Investig. New Drugs 2013, 31, 707–713. [Google Scholar] [CrossRef] [Green Version]
- Le, D.T.; Lutz, E.; Uram, J.N.; Sugar, E.A.; Onners, B.; Solt, S.; Zheng, L.; Diaz, L.A., Jr.; Donehower, R.C.; Jaffee, E.M. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J. Immunother. 2013, 36, 382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Overman, M.; Javle, M.E.; Davis, R.; Vats, P.; Kumar-Sinha, C.; Xiao, L.; Mettu, N.B.; Parra, E.R.; Benson, A.B.; Lopez, C.D.; et al. Randomized phase II study of the Bruton tyrosine kinase inhibitor acalabrutinib, alone or with pembrolizumab in patients with advanced pancreatic cancer. J. Immunother. Cancer 2020, 8, e000587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nesselhut, J.; Marx, D.; Lange, H.; Regalo, G.; Cillien, N.; Chang, R.Y.; Nesselhut, T. Systemic treatment with anti-PD-1 antibody nivolumab in combination with vaccine therapy in advanced pancreatic cancer. J. Clin. Oncol. 2016, 34, 3092. [Google Scholar] [CrossRef]
- Doi, T.; Muro, K.; Ishii, H.; Kato, T.; Tsushima, T.; Takenoyama, M.; Oizumi, S.; Gemmoto, K.; Suna, H.; Enokitani, K.; et al. A Phase I Study of the Anti-CC Chemokine Receptor 4 Antibody, Mogamulizumab, in Combination with Nivolumab in Patients with Advanced or Metastatic Solid Tumors. Clin. Cancer Res. 2019, 25, 6614–6622. [Google Scholar] [CrossRef] [Green Version]
- Wainberg, Z.; Piha-Paul, S.; Luke, J.; Kim, E.; Thompson, J.; Britten, C.; Johnson, J.; Pfanzelter, N.; Gordon, M.; Rasco, D. First-in-human phase 1 dose escalation and expansion of a novel combination, anti-CSF-1 receptor (cabiralizumab) plus anti-PD-1 (nivolumab), in patients with advanced solid tumors. J. Immunother. Cancer 2017, 5, 101186. [Google Scholar]
- Overman, M.J.; Lorusso, P.; Strickler, J.H.; Patel, S.P.; Clarke, S.J.; Noonan, A.M.; Prasanna, T.; Amin, M.A.; Nemunaitis, J.J.; Desai, J.; et al. Safety, efficacy and pharmacodynamics (PD) of MEDI9447 (oleclumab) alone or in combination with durvalumab in advanced colorectal cancer (CRC) or pancreatic cancer (panc). J. Clin. Oncol. 2018, 36, 4123. [Google Scholar] [CrossRef]
- Naing, A.; Powderly, J.D.; Falchook, G.; Creelan, B.; Nemunaitis, J.; Lutzky, J.; Diab, A.; Wang, J.S.; Laing, N.; Niewood, M.; et al. Abstract CT177: Epacadostat plus durvalumab in patients with advanced solid tumors: Preliminary results of the ongoing, open-label, phase I/II ECHO-203 study. Clin. Trials 2018, 78, CT177. [Google Scholar] [CrossRef]
- Cavalieri, C.C. Pembrolizumab in gastrointestinal (GI) malignancies with defective DNA mismatch repair (dMMR): A single institution experience. J. Clin. Oncol. 2017, 35, e23081. [Google Scholar] [CrossRef]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
- Hu, Z.I.; Shia, J.; Stadler, Z.K.; Varghese, A.M.; Capanu, M.; Salo-Mullen, E.; Lowery, M.A.; Diaz, L.A.; Mandelker, D.; Yu, K.H.; et al. Evaluating Mismatch Repair Deficiency in Pancreatic Adenocarcinoma: Challenges and Recommendations. Clin. Cancer Res. 2018, 24, 1326–1336. [Google Scholar] [CrossRef] [Green Version]
- Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.-L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Makohon-Moore, A.P.; Matsukuma, K.; Zhang, M.; Reiter, J.G.; Gerold, J.M.; Jiao, Y.; Sikkema, L.; Attiyeh, M.A.; Yachida, S.; Sandone, C.; et al. Precancerous neoplastic cells can move through the pancreatic ductal system. Nature 2018, 561, 201–205. [Google Scholar] [CrossRef] [PubMed]
- Balachandran, V.P.; Łuksza, M.; Zhao, J.N.; Makarov, V.; Moral, J.A.; Remark, R.; Herbst, B.; Askan, G.; Bhanot, U.; Senbabaoglu, Y.; et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017, 551, 512–516. [Google Scholar] [CrossRef] [PubMed]
- Argentiero, A.; De Summa, S.; Di Fonte, R.; Iacobazzi, R.M.; Porcelli, L.; Da Vià, M.; Brunetti, O.; Azzariti, A.; Silvestris, N.; Solimando, A.G. Gene Expression Comparison between the Lymph Node-Positive and -Negative Reveals a Peculiar Immune Microenvironment Signature and a Theranostic Role for WNT Targeting in Pancreatic Ductal Adenocarcinoma: A Pilot Study. Cancers 2019, 11, 942. [Google Scholar] [CrossRef] [Green Version]
- Porcelli, L.; Iacobazzi, R.M.; Di Fonte, R.; Serratì, S.; Intini, A.; Solimando, A.G.; Brunetti, O.; Calabrese, A.; Leonetti, F.; Azzariti, A.; et al. CAFs and TGF-β Signaling Activation by Mast Cells Contribute to Resistance to Gemcitabine/Nabpaclitaxel in Pancreatic Cancer. Cancers 2019, 11, 330. [Google Scholar] [CrossRef] [Green Version]
- Lowery, M.A.; Jordan, E.J.; Basturk, O.; Ptashkin, R.N.; Zehir, A.; Berger, M.F.; Leach, T.; Herbst, B.; Askan, G.; Maynard, H.; et al. Real-Time Genomic Profiling of Pancreatic Ductal Adenocarcinoma: Potential Actionability and Correlation with Clinical Phenotype. Clin. Cancer Res. 2017, 23, 6094–6100. [Google Scholar] [CrossRef] [Green Version]
- Biankin, A.V.; Piantadosi, S.; Hollingsworth, S.J. Patient-centric trials for therapeutic development in precision oncology. Nature 2015, 526, 361–370. [Google Scholar] [CrossRef] [Green Version]
- Longo, V.; Brunetti, O.; Gnoni, A.; Licchetta, A.; Delcuratolo, S.; Memeo, R.; Solimando, A.G.; Argentiero, A. Emerging Role of Immune Checkpoint Inhibitors in Hepatocellular Carcinoma. Medicina 2019, 55, 698. [Google Scholar] [CrossRef] [Green Version]
- Foley, K.; Kim, V.; Jaffee, E.M.; Zheng, L. Current progress in immunotherapy for pancreatic cancer. Cancer Lett. 2016, 381, 244–251. [Google Scholar] [CrossRef] [Green Version]
Tumor Type | Immune Checkpoint Inhibitor | Overall Response Rate % |
---|---|---|
NSCLC | Nivolumab | 20 |
RCC | Nivolumab | 25 |
Melanoma | Nivolumab | 32 |
Melanoma | Nivolumab + Ipilimumab | 61 |
Melanoma | Pembrolizumab | 33 |
Gastric/GEJ | Pembrolizumab | 22 |
HCC | Nivolumab | 20 |
MSI-H CRC | Nivolumab + Ipilimumab | 81 |
MSS CRC | Pembrolizumab | 0 |
MSI-H CRC | Pembrolizumab | 40 |
Pancreatic | Nivolumab + Ipilimumab | 1–2 |
Phase of Trial | Therapy | No of Patients | Setting | Median PFS/OS (Months) | Response | Reference |
---|---|---|---|---|---|---|
II | Ipilimumab | 27 | Locally advanced/metastatic 74% pretreated | OS: 4.5 | 0% RR 1 delayed response | [130] |
I | BMS-936559 | 14 | Advanced Pre-treated | NR | 0% RR | [70] |
II randomized | Durvalumab vs. Durvalumab + tremelimumab | 33 32 | Metastatic 2nd-line | OS: 3.6 OS: 3.1 | 6% DCR 9% DCR 3% PR | [131] |
Pilot study | Durvalumab Durvalumab + tremelimumab + SBRT | 24 | Metastatic Pre-treated | NR | 21% SD | [132] |
II (safety profile) | Durvalumab + tremelimumab + gemcitabine + nab-paclitaxel | 11 | Metastatic No prior treatment | PFS:7.9 | 73% PR 100% DCR | [133] |
Ib/II randomized | Pembrolizumab + capecitabine + RT vs. Capecitabine + RT | 14 8 | Neoadjuvant: 50% resectable/50% borderline resectable | NR | 71% underwent surgery 50% underwent surgery | [134] |
Ib dose escalation | Gemcitabine-> tremelimumab (different doses) | 34 | Metastatic No prior treatment | OS: 7.4 | 21% SD 6% PR | [135] |
Ib | Ipilimumab + gemcitabine | 16 | Advanced No prior gemcitabine in advanced setting | PFS: 2.5 OS: 8.5 | 13% PR 31% SD | [136] |
Ib/II | Pembrolizumab + gemcitabine + nab-paclitaxel | 12 5 | Metastatic No prior treatment Pre-treated | PFS: 9.1 OS: 15.0 NR | 25% PR 67% SD 40% SD | [137] |
II | Pembrolizumab + reolysin + 5-FU Pemrolizumab + reolysin + gemcitabine Pembrolizumab + reolysin + irinotecan | 11 | Metastatic 2nd line | NR | 9% PR 18% SD | [138] |
I | Nivolumab + nab-paclitaxel + gemcitabine | 50 | Locally advanced/metastatic No prior treatment PD-L1 expression ≥ 1%: 24% PD-L1 expression ≥: 12% | PFS: 5.5 OS: 9.9 | 2% CR 16% PR 46% SD | [139] |
II, pilot | Nivolumab + nab-paclitaxel + gemcitabine + paricalcitol | 10 | Metastatic 1st line | PFS: 8.2 | 80% PR 100% DCR | [140] |
Ib | Gemcitabine + nab-paclitaxel + APX005M (anti-CD40 antibody) ± nivolumab | 30 | Metastatic No prior treatment | NR | 47% PR 27% SD | [141] |
I Dose escalation | Gemcitabine Gemcitabine + 0.5 mg IMP321 Gemcitabine + 2.0 mg IMP321 | 6 6 5 | Advanced 1st line | OS: 16.7 TTP: 10.2 OS: 5.6 TTP: 2.0 OS: 6.4 TTP: 5.3 | 83% SD 33% SD 60% SD | [142] |
Ib randomized | Ipilimumab vs. Ipilimumab + vaccine | 15 15 | Advanced/metastatic Pre-treated | OS: 3.6 OS: 5.7 | 13% SD 0% SD | [143] |
II randomized | Acalabrutinib vs. Pembrolizumab + acalabrutinib | 26 32 | Metastatic Pre-treated | NR | 15% SD 9% PR 16% SD | [144] |
Pilot study | Nivolumab + dendritic cells | 7 | Metastatic | NR | 29% PR | [145] |
I | Nivolumab + magamulizumab (anti-CC-chemokine receptor 4 antibody) | 15 | Advanced/metastatic | NR | 7%PR 33% SD | [146] |
I Dose escalation | Nivolumab + cabiralizumab (antibody directed against CSF-1 receptor) | 31 evaluable | Advanced Pre-treated | NR | 10% PR 3% SD | [147] |
I/II | Oleclumab (antibody targeting CD73) ± durvalumab | 20 | Advanced Pre-treated | NR | 10% PR 15% SD | [148] |
I/II | Durvalumab + epacadostat | 15 | Advanced Pre-treated | NR | 27% SD | [149] |
Retrospective | pembrolizumab | 2 | dMMR | NR | 50% PR 50% SD | [150] |
II | pembrolizumab | 8 | Advanced Pre-treated dMMR/MSI positive | NR | 25% CR 37% PR 12% SD | [151] |
Retrospective | PD-L1 inhibitor + IDO1 (amino acid degrading enzyme) inhibitor or PD-1 inhibitor | 7 | Advanced Pre-treated dMMR | NR | 14% CR 29% PR 14% SD | [152] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Javadrashid, D.; Baghbanzadeh, A.; Derakhshani, A.; Leone, P.; Silvestris, N.; Racanelli, V.; Solimando, A.G.; Baradaran, B. Pancreatic Cancer Signaling Pathways, Genetic Alterations, and Tumor Microenvironment: The Barriers Affecting the Method of Treatment. Biomedicines 2021, 9, 373. https://doi.org/10.3390/biomedicines9040373
Javadrashid D, Baghbanzadeh A, Derakhshani A, Leone P, Silvestris N, Racanelli V, Solimando AG, Baradaran B. Pancreatic Cancer Signaling Pathways, Genetic Alterations, and Tumor Microenvironment: The Barriers Affecting the Method of Treatment. Biomedicines. 2021; 9(4):373. https://doi.org/10.3390/biomedicines9040373
Chicago/Turabian StyleJavadrashid, Darya, Amir Baghbanzadeh, Afshin Derakhshani, Patrizia Leone, Nicola Silvestris, Vito Racanelli, Antonio Giovanni Solimando, and Behzad Baradaran. 2021. "Pancreatic Cancer Signaling Pathways, Genetic Alterations, and Tumor Microenvironment: The Barriers Affecting the Method of Treatment" Biomedicines 9, no. 4: 373. https://doi.org/10.3390/biomedicines9040373
APA StyleJavadrashid, D., Baghbanzadeh, A., Derakhshani, A., Leone, P., Silvestris, N., Racanelli, V., Solimando, A. G., & Baradaran, B. (2021). Pancreatic Cancer Signaling Pathways, Genetic Alterations, and Tumor Microenvironment: The Barriers Affecting the Method of Treatment. Biomedicines, 9(4), 373. https://doi.org/10.3390/biomedicines9040373