We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Strategies to relieve immunosuppression in pancreatic cancer

    Max Schnurr

    Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany

    ,
    Peter Duewell

    Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany

    ,
    Christian Bauer

    Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany

    ,
    Simon Rothenfusser

    Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany

    ,
    Kirsten Lauber

    Department of Radiation Oncology, Klinikum der Universität München, Munich, Germany

    ,
    Stefan Endres

    Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany

    &
    Sebastian Kobold

    Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany

    Published Online:https://doi.org/10.2217/imt.15.9

    Despite continuous progress in the understanding of deregulated pathways in pancreatic cancer cells and development of targeted therapies, therapeutic advances with clinical benefit have been scarce over the last decades. The recent success of immunotherapy for some solid cancers has fueled optimism that this approach might also work for pancreatic cancer. However, a highly immunosuppressive microenvironment mediated by tumor, stromal and immune cells creates a major hurdle for immunotherapy. Mouse models have helped to unravel critical immunosuppressive mechanisms that could serve as novel therapeutic targets. Here we review new promising strategies that alone or in combination with other modalities, such as chemotherapy or irradiation, have the potential to lead to tumor immune control and finally better clinical outcome.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest.

    References

    • 1 Wolfgang CL, Herman JM, Laheru DA et al. Recent progress in pancreatic cancer. CA Cancer J. Clin. 63(5), 318–348 (2013).
    • 2 Mazur PK, Siveke JT. Genetically engineered mouse models of pancreatic cancer: unravelling tumour biology and progressing translational oncology. Gut 61(10), 1488–1500 (2012).
    • 3 Robert C, Thomas L, Bondarenko I et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364(26), 2517–2526 (2011).
    • 4 Topalian SL, Hodi FS, Brahmer JR et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366(26), 2443–2454 (2012).
    • 5 Zheng L, Xue J, Jaffee EM, Habtezion A. Role of immune cells and immune-based therapies in pancreatitis and pancreatic ductal adenocarcinoma. Gastroenterology 144(6), 1230–1240 (2013).
    • 6 Shibuya KC, Goel VK, Xiong W et al. Pancreatic ductal adenocarcinoma contains an effector and regulatory immune cell infiltrate that is altered by multimodal neoadjuvant treatment. PLoS ONE 9(5), e96565 (2014).
    • 7 Wormann SM, Diakopoulos KN, Lesina M, Algul H. The immune network in pancreatic cancer development and progression. Oncogene 33(23), 2956–2967 (2014).
    • 8 Sideras K, Braat H, Kwekkeboom J et al. Role of the immune system in pancreatic cancer progression and immune modulating treatment strategies. Cancer Treat Rev. 40(4), 513–522 (2014).
    • 9 Salman B, Zhou D, Jaffee EM, Edil BH, Zheng L. Vaccine therapy for pancreatic cancer. Oncoimmunology 2(12), e26662 (2013).
    • 10 Bauer C, Dauer M, Saraj S et al. Dendritic cell-based vaccination of patients with advanced pancreatic carcinoma: results of a pilot study. Cancer Immunol. Immunother. 60(8), 1097–1107 (2011).
    • 11 Shindo Y, Hazama S, Maeda Y et al. Adoptive immunotherapy with MUC1-mRNA transfected dendritic cells and cytotoxic lymphocytes plus gemcitabine for unresectable pancreatic cancer. J. Transl. Med. 12, 175 (2014).
    • 12 Weden S, Klemp M, Gladhaug IP et al. Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int. J. Cancer 128(5), 1120–1128 (2011).
    • 13 Inman KS, Francis AA, Murray NR. Complex role for the immune system in initiation and progression of pancreatic cancer. World J. Gastroenterol. 20(32), 11160–11181 (2014).
    • 14 Neesse A, Michl P, Frese KK et al. Stromal biology and therapy in pancreatic cancer. Gut 60(6), 861–868 (2011).
    • 15 Rhim AD, Oberstein PE, Thomas DH et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25(6), 735–747 (2014).
    • 16 Ozdemir BC, Pentcheva-Hoang T, Carstens JL et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25(6), 719–734 (2014).
    • 17 Suso EM, Dueland S, Rasmussen AM et al. hTERT mRNA dendritic cell vaccination: complete response in a pancreatic cancer patient associated with response against several hTERT epitopes. Cancer Immunol. Immunother. 60(6), 809–818 (2011).
    • 18 Lutz E, Yeo CJ, Lillemoe KD et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A Phase II trial of safety, efficacy, and immune activation. Ann. Surg. 253(2), 328–335 (2011).
    • 19 Le DT, Lutz E, Uram JN et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J. Immunother. 36(7), 382–389 (2013).
    • 20 Correale P, Aquino A, Giuliani A et al. Treatment of colon and breast carcinoma cells with 5-fluorouracil enhances expression of carcinoembryonic antigen and susceptibility to HLA-A(*)02.01 restricted, CEA-peptide-specific cytotoxic T cells in vitro. Int. J. Cancer. 104(4), 437–445 (2003).
    • 21 Bauer C, Bauernfeind F, Sterzik A et al. Dendritic cell-based vaccination combined with gemcitabine increases survival in a murine pancreatic carcinoma model. Gut 56(9), 1275–1282 (2007).
    • 22 Bauer C, Sterzik A, Bauernfeind F et al. Concomitant gemcitabine therapy negatively affects DC vaccine-induced CD8(+) T-cell and B-cell responses but improves clinical efficacy in a murine pancreatic carcinoma model. Cancer Immunol. Immunother. 63(4), 321–333 (2014).
    • 23 Mundy-Bosse BL, Lesinski GB, Jaime-Ramirez AC et al. Myeloid-derived suppressor cell inhibition of the IFN response in tumor-bearing mice. Cancer Res. 71(15), 5101–5110 (2011).
    • 24 Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 31, 51–72 (2013).•• This review article highlights how chemotherapeutic drugs mediate so far unrecognized immunological effects via induction of immunogenic forms of tumor cell death ultimately leading to an adaptive immune response against the tumor.
    • 25 Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 26(4), 332–342 (2003).
    • 26 Dudley ME, Yang JC, Sherry R et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 26(32), 5233–5239 (2008).
    • 27 Peiper M, Sato T, Streichert T, Eisenberger CF, Knoefel WT, Izbicki JR. Cytotoxic T lymphocyte mediated recognition of human pancreatic cancer cells. Int. J. Cancer. 99(1), 88–92 (2002).
    • 28 Terashima T, Mizukoshi E, Arai K et al. P53, hTERT, WT-1, and VEGFR2 are the most suitable targets for cancer vaccine therapy in HLA-A24 positive pancreatic adenocarcinoma. Cancer Immunol. Immunother. 63(5), 479–489 (2014).
    • 29 Abe Y, Ito T, Baba E et al. Nonmyeloablative allogeneic hematopoietic stem cell transplantation as immunotherapy for pancreatic cancer. Pancreas 38(7), 815–819 (2009).
    • 30 Kanda Y, Komatsu Y, Akahane M et al. Graft-versus-tumor effect against advanced pancreatic cancer after allogeneic reduced-intensity stem cell transplantation. Transplantation 79(7), 821–827 (2005).
    • 31 Kaneko T, Goto S, Kato A et al. Efficacy of immuno-cell therapy in patients with advanced pancreatic cancer. Anticancer Res. 25(6A), 3709–3714 (2005).
    • 32 Mukherjee P, Ginardi AR, Madsen CS et al. Mice with spontaneous pancreatic cancer naturally develop MUC-1-specific CTLs that eradicate tumors when adoptively transferred. J. Immunol. 165(6), 3451–3460 (2000).
    • 33 Sivinski CL, Kohlgraf KG, Vanlith ML, Morikane K, Tempero RM, Hollingsworth MA. Molecular requirements for CD8-mediated rejection of a MUC1-expressing pancreatic carcinoma: implications for tumor vaccines. Cancer Immunol. Immunother. 51(6), 327–340 (2002).
    • 34 Schmitz-Winnenthal FH, Volk C, Z'graggen K et al. High frequencies of functional tumor-reactive T cells in bone marrow and blood of pancreatic cancer patients. Cancer Res. 65(21), 10079–10087 (2005).
    • 35 Urba WJ. At the bench: adoptive cell therapy for melanoma. J. Leukoc. Biol. 95(6), 867–874 (2014).
    • 36 Urba WJ, Longo DL. Redirecting T cells. N. Engl. J. Med. 365(8), 754–757 (2011).
    • 37 Abate-Daga D, Lagisetty KH, Tran E et al. A Novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. Human Gene Ther. 25(12), 1003–1012 (2014).
    • 38 Chmielewski M, Hahn O, Rappl G et al. T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice. Gastroenterology 143(4), 1095–1107 e1092 (2012).
    • 39 Hekele A, Dall P, Moritz D et al. Growth retardation of tumors by adoptive transfer of cytotoxic T lymphocytes reprogrammed by CD44v6-specific scFv:zeta-chimera. Int. J. Cancer. 68(2), 232–238 (1996).
    • 40 Groth A, Salnikov AV, Ottinger S et al. New gene-immunotherapy combining TRAIL-lymphocytes and EpCAMxCD3 Bispecific antibody for tumor targeting. Clin. Cancer Res. 18(4), 1028–1038 (2012).
    • 41 Chen Y, Ayaru L, Mathew S, Morris E, Pereira SP, Behboudi S. Expansion of anti-mesothelin specific CD4+ and CD8+ T cell responses in patients with pancreatic carcinoma. PLoS ONE 9(2), e88133 (2014).
    • 42 Mccarty TM, Liu X, Sun JY, Peralta EA, Diamond DJ, Ellenhorn JD. Targeting p53 for adoptive T-cell immunotherapy. Cancer Res. 58(12), 2601–2605 (1998).
    • 43 Schmidt J, Ryschich E, Sievers E, Schmidt-Wolf IG, Buchler MW, Marten A. Telomerase-specific T-cells kill pancreatic tumor cells in vitro and in vivo. Cancer 106(4), 759–764 (2006).
    • 44 Klug F, Prakash H, Huber PE et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24(5), 589–602 (2013).• Demonstrates that local tumor irradiation significantly alters the functional profile of infiltrating tumor-associated macrophages thereby releasing the brake of T-cell inhibition in tumors.
    • 45 Ene-Obong A, Clear AJ, Watt J et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 145(5), 1121–1132 (2013).
    • 46 Feig C, Jones JO, Kraman M et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl Acad. Sci. USA 110(50), 20212–20217 (2013).• Provides evidence that effectivity of checkpoint inhibition strongly depends on factors in the tumor stroma and that rational combination therapies can overcome this hurdle.
    • 47 Moon EK, Carpenito C, Sun J et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 17(14), 4719–4730 (2011).
    • 48 Olive KP, Jacobetz MA, Davidson CJ et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324(5933), 1457–1461 (2009).
    • 49 Ribas A. Tumor immunotherapy directed at PD-1. N. Engl. J. Med. 366(26), 2517–2519 (2012).
    • 50 Perez-Gracia JL, Labiano S, Rodriguez-Ruiz ME, Sanmamed MF, Melero I. Orchestrating immune check-point blockade for cancer immunotherapy in combinations. Curr. Opin. Immunol. 27, 89–97 (2014).
    • 51 Hamid O, Robert C, Daud A et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 369(2), 134–144 (2013).
    • 52 Loos M, Giese NA, Kleeff J et al. Clinical significance and regulation of the costimulatory molecule B7-H1 in pancreatic cancer. Cancer Lett. 268(1), 98–109 (2008).
    • 53 Yamato I, Sho M, Nomi T et al. Clinical importance of B7-H3 expression in human pancreatic cancer. Br. J. Cancer 101(10), 1709–1716 (2009).
    • 54 Yang M, Sun T, Zhou Y et al. The functional cytotoxic T lymphocyte-associated Protein 4 49G-to-A genetic variant and risk of pancreatic cancer. Cancer 118(19), 4681–4686 (2012).
    • 55 Sandin LC, Eriksson F, Ellmark P, Loskog AS, Totterman TH, Mangsbo SM. Local CTLA4 blockade effectively restrains experimental pancreatic adenocarcinoma growth in vivo. Oncoimmunology 3(1), e27614 (2014).
    • 56 Royal RE, Levy C, Turner K et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 33(8), 828–833 (2010).
    • 57 Brahmer JR, Tykodi SS, Chow LQ et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366(26), 2455–2465 (2012).
    • 58 Von Hoff DD, Ramanathan RK, Borad MJ et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a Phase I/II trial. J. Clin. Oncol. 29(34), 4548–4554 (2011).
    • 59 Stromnes IM, Brockenbrough JS, Izeradjene K et al. Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut 63(11), 1769–1781 (2014).
    • 60 Rakoff-Nahoum S, Medzhitov R. Toll-like receptors and cancer. Nat. Rev. Cancer 9(1), 57–63 (2009).
    • 61 Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 140(6), 805–820 (2010).
    • 62 Vaz J, Andersson R. Intervention on toll-like receptors in pancreatic cancer. World J. Gastroenterol. 20(19), 5808–5817 (2014).
    • 63 Ochi A, Nguyen AH, Bedrosian AS et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J. Exp. Med. 209(9), 1671–1687 (2012).
    • 64 Ochi A, Graffeo CS, Zambirinis CP et al. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J. Clin. Invest. 122(11), 4118–4129 (2012).• Challenges the view that TLR ligands mediate antitumor immunity via induction of innate and adaptive immune responses, but may rather promote carcinogenesis via driving proinflammatory responses.
    • 65 Hamzah J, Altin JG, Herringson T et al. Targeted liposomal delivery of TLR9 ligands activates spontaneous antitumor immunity in an autochthonous cancer model. J. Immunol. 183(2), 1091–1098 (2009).
    • 66 Jacobs C, Duewell P, Heckelsmiller K et al. An ISCOM vaccine combined with a TLR9 agonist breaks immune evasion mediated by regulatory T cells in an orthotopic model of pancreatic carcinoma. Int. J. Cancer. 128(4), 897–907 (2011).
    • 67 Pratesi G, Petrangolini G, Tortoreto M et al. Therapeutic synergism of gemcitabine and CpG-oligodeoxynucleotides in an orthotopic human pancreatic carcinoma xenograft. Cancer Res. 65(14), 6388–6393 (2005).
    • 68 Van Den Boorn JG, Hartmann G. Turning tumors into vaccines: co-opting the innate immune system. Immunity 39(1), 27–37 (2013).
    • 69 Schmidt A, Rothenfusser S, Hopfner KP. Sensing of viral nucleic acids by RIG-I: from translocation to translation. Eur. J. Cell Biol. 91(1), 78–85 (2012).
    • 70 Tormo D, Checinska A, Alonso-Curbelo D et al. Targeted activation of innate immunity for therapeutic induction of autophagy and apoptosis in melanoma cells. Cancer Cell 16(2), 103–114 (2009).
    • 71 Besch R, Poeck H, Hohenauer T et al. Proapoptotic signaling induced by RIG-I and MDA-5 results in type I interferon-independent apoptosis in human melanoma cells. J. Clin. Invest. 119(8), 2399–2411 (2009).
    • 72 Bhoopathi P, Quinn BA, Gui Q et al. Pancreatic cancer-specific cell death induced in vivo by cytoplasmic-delivered polyinosine-polycytidylic acid. Cancer Res. 74(21), 6224–6235 (2014).
    • 73 Ellermeier J, Wei J, Duewell P et al. Therapeutic efficacy of bifunctional siRNA combining TGF-beta1 silencing with RIG-I activation in pancreatic cancer. Cancer Res. 73(6), 1709–1720 (2013).• Provides proof-of-concept that RLR ligands can be exploited for immunotherapy of pancreatic cancer and a rationale for the development of bifunctional siRNAs that combine RIG-I activation with RNAi-mediated silencing of immunosuppressive factors.
    • 74 Duewell P, Steger A, Lohr H et al. RIG-I-like helicases induce immunogenic cell death of pancreatic cancer cells and sensitize tumors toward killing by CD8(+) T cells. Cell Death Differ. 21(12), 1825–1837 (2014).
    • 75 Goldeck M, Schlee M, Hartmann G, Hornung V. Enzymatic synthesis and purification of a defined RIG-I ligand. Methods Mol. Biol. 1169, 15–25 (2014).
    • 76 Kurahara H, Shinchi H, Mataki Y et al. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. J. Surg. Res. 167(2), e211–e219 (2011).
    • 77 Ohki S, Shibata M, Gonda K et al. Circulating myeloid-derived suppressor cells are increased and correlate to immune suppression, inflammation and hypoproteinemia in patients with cancer. Oncol. Rep. 28(2), 453–458 (2012).
    • 78 Ruffell B, Affara NI, Coussens LM. Differential macrophage programming in the tumor microenvironment. Trends Immunol. 33(3), 119–126 (2012).
    • 79 Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 12(4), 253–268 (2012).
    • 80 Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 21(6), 836–847 (2012).
    • 81 Bayne LJ, Beatty GL, Jhala N et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 21(6), 822–835 (2012).•• Elegant study that shows how pancreatic cancers induce an immunosuppressive microenvironment via recruitment of myeloid cells to the tumor, thereby keeping T-effector cells dysfunctional.
    • 82 Kraman M, Bambrough PJ, Arnold JN et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 330(6005), 827–830 (2010).
    • 83 Beatty GL, Chiorean EG, Fishman MP et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331(6024), 1612–1616 (2011).•• Very good example of how unexpected results from clinical trials can stimulate fascinating preclincial work (‘from clinic to benchside’) to address novel mechanism of action.
    • 84 Kuhnemuth B, Muhlberg L, Schipper M et al. CUX1 modulates polarization of tumor-associated macrophages by antagonizing NF-kappaB signaling. Oncogene 34(2), 177–187 (2013).
    • 85 Ries CH, Cannarile MA, Hoves S et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 25(6), 846–859 (2014).
    • 86 Munn DH, Mellor AL. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 34(3), 137–143 (2013).
    • 87 Koblish HK, Hansbury MJ, Bowman KJ et al. Hydroxyamidine inhibitors of indoleamine-2,3-dioxygenase potently suppress systemic tryptophan catabolism and the growth of IDO-expressing tumors. Mol Cancer Ther. 9(2), 489–498 (2010).
    • 88 Lauber K, Ernst A, Orth M, Herrmann M, Belka C. Dying cell clearance and its impact on the outcome of tumor radiotherapy. Front. Oncol. 2, 116 (2012).
    • 89 Burnette BC, Liang H, Lee Y et al. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 71(7), 2488–2496 (2011).
    • 90 Hennel R, Brix N, Seidl K et al. Release of monocyte migration signals by breast cancer cell lines after ablative and fractionated gamma-irradiation. Radiat. Oncol. 9(1), 85 (2014).
    • 91 Formenti SC, Demaria S. Radiation therapy to convert the tumor into an in situ vaccine. Int. J. Radiat. Oncol. Biol. Phys. 84(4), 879–880 (2012).
    • 92 Shibamoto Y, Nishimura U, Abe M. Intraoperative radiotherapy and hyperthermia for unresectable pancreatic cancer. Hepatogastroenterology 43(8), 326–332 (1996).
    • 93 Garcia-Carbonero R, Carnero A, Paz-Ares L. Inhibition of HSP90 molecular chaperones: moving into the clinic. Lancet Oncol. 14(9), e358–369 (2013).
    • 94 Milanovic D, Firat E, Grosu AL, Niedermann G. Increased radiosensitivity and radiothermosensitivity of human pancreatic MIA PaCa-2 and U251 glioblastoma cell lines treated with the novel Hsp90 inhibitor NVP-HSP990. Radiat. Oncol. 8, 42 (2013).
    • 95 Lauber K, Munoz LE, Berens C, Jendrossek V, Belka C, Herrmann M. Apoptosis induction and tumor cell repopulation: the yin and yang of radiotherapy. Radiat. Oncol. 6, 176 (2011).
    • 96 Hardacre JM, Mulcahy M, Small W et al. Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: a Phase 2 study. J. Gastrointest. Surg. 17(1), 94–100; discussion 100–101 (2013).
    • 97 Gunturu KS, Rossi GR, Saif MW. Immunotherapy updates in pancreatic cancer: are we there yet? Adv. Med. Oncol. 5(1), 81–89 (2013).
    • 98 Deng L, Liang H, Burnette B et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J. Clin. Invest. 124(2), 687–695 (2014).