Immunomodulation in Pancreatic Cancer
Abstract
:Simple Summary
Abstract
1. Introduction
2. The Tumor Microenvironment
3. Tumor Microbiome
4. Biomarkers of Long-Term Survivors
5. Adjuvant and Neoadjuvant Therapies
6. Combination Strategies
7. T-Cell Activation Strategies for PDAC Treatment
7.1. Chimeric Antigen Receptor (CAR) T-Cells
7.2. Microbiome Modification
7.2.1. Antibiotics
7.2.2. Fecal Microbiota Transplantation
7.2.3. Bacterial Consortia
7.3. Oncolytic Vaccine Combinations
8. Conclusions
Funding
Conflicts of Interest
References
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA. Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saad, A.M.; Turk, T.; Al-Husseini, M.J.; Abdel-Rahman, O. Trends in pancreatic adenocarcinoma incidence and mortality in the United States in the last four decades; A SEER-based study. BMC Cancer 2018, 18, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Orth, M.; Metzger, P.; Gerum, S.; Mayerle, J.; Schneider, G.; Belka, C.; Schnurr, M.; Lauber, K. Pancreatic ductal adenocarcinoma: Biological hallmarks, current status, and future perspectives of combined modality treatment approaches. Radiat. Oncol. 2019, 14, 1–20. [Google Scholar] [CrossRef] [PubMed]
- Kleeff, J.; Korc, M.; Apte, M.; La Vecchia, C.; Johnson, C.D.; Biankin, A.V.; Neale, R.E.; Tempero, M.; Tuveson, D.A.; Hruban, R.H.; et al. Pancreatic cancer. Nat. Rev. Dis. Prim. 2016, 2, 1–23. [Google Scholar] [CrossRef] [PubMed]
- Muñoz Martín, A.J.; Adeva, J.; Martínez-Galán, J.; Reina, J.J.; Hidalgo, M. Pancreatic ductal adenocarcinoma: Metastatic disease. Clin. Transl. Oncol. 2017, 19, 1423–1429. [Google Scholar] [CrossRef]
- Vaccaro, V.; Sperduti, I.; Milella, M. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 365, 768–769. [Google Scholar] [CrossRef]
- 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]
- Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
- He, J.; Ahuja, N.; Makary, M.A.; Cameron, J.L.; Eckhauser, F.E.; Choti, M.A.; Hruban, R.H.; Pawlik, T.M.; Wolfgang, C.L. 2564 resected periampullary adenocarcinomas at a single institution: Trends over three decades. Hpb 2014, 16, 83–90. [Google Scholar] [CrossRef] [Green Version]
- 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]
- 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] [PubMed]
- Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 trial of single agent ipilimumab (Anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef] [PubMed]
- Sohal, D.P.S.; Kennedy, E.B.; Khorana, A.; Copur, M.S.; Crane, C.H.; Garrido-Laguna, I.; Krishnamurthi, S.; Moravek, C.; O’Reilly, E.M.; Philip, P.A.; et al. Metastatic pancreatic cancer: ASCO clinical practice guideline update. J. Clin. Oncol. 2018, 36, 2545–2556. [Google Scholar] [CrossRef] [PubMed]
- Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; de Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/ mismatch repair–deficient cancer: Results from the phase II KEYNOTE-158 study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Watanabe, I.; Hasebe, T.; Sasaki, S.; Konishi, M.; Inoue, K.; Nakagohri, T.; Oda, T.; Mukai, K.; Kinoshita, T. Advanced pancreatic ductal cancer: Fibrotic focus and β-catenin expression correlate with outcome. Pancreas 2003, 26, 326–333. [Google Scholar] [CrossRef] [PubMed]
- Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog Signaling. Science 2011, 324, 1457–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erkan, M.; Kurtoglu, M.; Kleeff, J. The role of hypoxia in pancreatic cancer: A potential therapeutic target? Expert Rev. Gastroenterol. Hepatol. 2016, 10, 301–316. [Google Scholar] [CrossRef] [PubMed]
- Kamphorst, J.J.; Nofal, N.; Commisso, C.; Hackett, S.R.; Lu, W.; Grabocka, E.; Vander Heiden, M.G.; Miller, G.; Drebin, J.A.; Bar-Sagi, D.; et al. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 2015, 75, 544–553. [Google Scholar] [CrossRef] [Green Version]
- Apte, M.V.; Pirola, R.C.; Wilson, J.S. Pancreatic stellate cells: A starring role in normal and diseased pancreas. Front. Physiol. 2012, 3, 344. [Google Scholar] [CrossRef] [Green Version]
- Özdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [Green Version]
- Xu, Z.; Vonlaufen, A.; Phillips, P.A.; Fiala-Beer, E.; Zhang, X.; Yang, L.; Biankin, A.V.; Goldstein, D.; Pirola, R.C.; Wilson, J.S.; et al. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am. J. Pathol. 2010, 177, 2585–2596. [Google Scholar] [CrossRef] [PubMed]
- Mace, T.A.; Bloomston, M.; Lesinski, G.B. Pancreatic cancer-associated stellate cells: A viable target for reducing immunosuppression in the tumor microenvironment. Oncoimmunology 2013, 2, 5–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ene-Obong, A.; Clear, A.J.; Watt, J.; Wang, J.; Fatah, R.; Riches, J.C.; Marshall, J.F.; Chin-Aleong, J.; Chelala, C.; Gribben, J.G.; et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013, 145, 1121–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Orhan, A.; Vogelsang, R.P.; Andersen, M.B.; Madsen, M.T.; Hölmich, E.R.; Raskov, H.; Gögenur, I. The prognostic value of tumour-infiltrating lymphocytes in pancreatic cancer: A systematic review and meta-analysis. Eur. J. Cancer 2020, 132, 71–84. [Google Scholar] [CrossRef]
- Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.B.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [Green Version]
- Bockorny, B.; Semenisty, V.; Macarulla, T.; Borazanci, E.; Wolpin, B.M.; Stemmer, S.M.; Golan, T.; Geva, R.; Borad, M.J.; Pedersen, K.S.; et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: The COMBAT. trial. Nat. Med. 2020, 1–8. [Google Scholar] [CrossRef]
- Lin, J.H.; Huffman, A.P.; Wattenberg, M.M.; Walter, D.M.; Carpenter, E.L.; Feldser, D.M.; Beatty, G.L.; Furth, E.E.; Vonderheide, R.H. Type 1 conventional dendritic cells are systemically dysregulated early in pancreatic carcinogenesis. J. Exp. Med. 2020, 217, e20190673. [Google Scholar] [CrossRef]
- Hegde, S.; Krisnawan, V.E.; Herzog, B.H.; Zuo, C.; Breden, M.A.; Knolhoff, B.L.; Hogg, G.D.; Tang, J.P.; Baer, J.M.; Mpoy, C.; et al. Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 2020, 37, 289–307.e9. [Google Scholar] [CrossRef]
- Karsunky, H.; Merad, M.; Cozzio, A.; Weissman, I.L.; Manz, M.G. Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid-committed progenitors to Flt3+ dendritic cells in vivo. J. Exp. Med. 2003, 198, 305–313. [Google Scholar] [CrossRef] [Green Version]
- Khalil, M.; Vonderheide, R.H. Anti-CD40 agonist antibodies: Preclinical and clinical experience. Update Cancer Ther. 2007, 2, 61–65. [Google Scholar] [CrossRef] [Green Version]
- Leao, I.C.; Ganesan, P.; Armstrong, T.D.; Jaffee, E.M. Effective depletion of regulatory T cells allows the recruitment of mesothelin-specific CD8+T cells to the anti-tumor immune response against a mesothelin-expressing mouse pancreatic adenocarcinoma. Clin. Transl. Sci. 2008, 1, 228–239. [Google Scholar] [CrossRef] [PubMed]
- Keenan, B.P.; Saenger, Y.; Kafrouni, M.I.; Leubner, A.; Lauer, P.; Maitra, A.; Rucki, A.A.; Gunderson, A.J.; Coussens, L.M.; Dirk, G.; et al. 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, 1784–1794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Lazarus, J.; Steele, N.G.; Yan, W.; Lee, H.J.; Nwosu, Z.C.; Halbrook, C.J.; Menjivar, R.E.; Kemp, S.B.; Sirihorachai, V.R.; et al. Regulatory T-cell depletion alters the tumor microenvironment and accelerates pancreatic carcinogenesis. Cancer Discov. 2020, 10, 422–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ugel, S.; De Sanctis, F.; Mandruzzato, S.; Bronte, V. Tumor-induced myeloid deviation: When myeloid-derived suppressor cells meet tumor-Associated macrophages. J. Clin. Investig. 2015, 125, 3365–3376. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Velez-Delgado, A.; Mathew, E.; Li, D.; Mendez, F.M.; Flannagan, K.; Rhim, A.D.; Simeone, D.M.; Beatty, G.L.; Di Magliano, M.P. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017, 66, 124–136. [Google Scholar] [CrossRef] [Green Version]
- Dominguez, G.A.; Condamine, T.; Mony, S.; Hashimoto, A.; Wang, F.; Liu, Q.; Forero, A.; Bendell, J.; Witt, R.; Hockstein, N.; et al. Selective targeting of myeloid-derived suppressor cells in cancer patients using DS-8273a, an agonistic TRAIL-R2 antibody. Clin. Cancer Res. 2017, 23, 2942–2950. [Google Scholar] [CrossRef] [Green Version]
- Noy, R.; Pollard, J.W. Tumor-Associated Macrophages: From Mechanisms to Therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [Green Version]
- Knudsen, E.S.; Vail, P.; Balaji, U.; Ngo, H.; Botros, I.W.; Makarov, V.; Riaz, N.; Balachandran, V.; Leach, S.; Thompson, D.M.; et al. Stratification of pancreatic ductal adenocarcinoma: Combinatorial genetic, stromal, and immunologic markers. Clin. Cancer Res. 2017, 23, 4429–4440. [Google Scholar] [CrossRef] [Green Version]
- Lankadasari, M.B.; Mukhopadhyay, P.; Mohammed, S.; Harikumar, K.B. TAMing pancreatic cancer: Combat with a double edged sword. Mol. Cancer 2019, 18, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Candido, J.B.; Morton, J.P.; Bailey, P.; Campbell, A.D.; Karim, S.A.; Jamieson, T.; Lapienyte, L.; Gopinathan, A.; Clark, W.; McGhee, E.J.; et al. CSF1R+ Macrophages Sustain Pancreatic Tumor Growth through T Cell Suppression and Maintenance of Key Gene Programs that Define the Squamous Subtype. Cell Rep. 2018, 23, 1448–1460. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; De Nardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014, 74, 5057–5069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.; Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 2020, 368, 973–980. [Google Scholar] [CrossRef] [PubMed]
- Geller, L.T.; Barzily-rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 1160, 1156–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
- Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef] [Green Version]
- 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, S12–S16. [Google Scholar] [CrossRef]
- Moral, J.A.; Leung, J.; Rojas, L.A.; Ruan, J.; Zhao, J.; Sethna, Z.; Ramnarain, A.; Gasmi, B.; Gururajan, M.; Redmond, D.; et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature 2020, 579, 130–135. [Google Scholar] [CrossRef]
- Li, D.; Xie, K.; Wolff, R.; Abbruzzese, J.L. Pancreatic cancer. Lancet 2004, 363, 1049–1057. [Google Scholar] [CrossRef]
- Seufferlein, T.; Ettrich, T.J. Treatment of pancreatic cancer—neoadjuvant treatment in resectable pancreatic cancer (PDAC). Transl. Gastroenterol. Hepatol. 2019, 4. [Google Scholar] [CrossRef] [PubMed]
- Neoptolemos, J.P.; Palmer, D.H.; Ghaneh, P.; Psarelli, E.E.; Valle, J.W.; Halloran, C.M.; Faluyi, O.; O’Reilly, D.A.; Cunningham, D.; Wadsley, J.; et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): A multicentre, open-label, randomised, phase 3 trial. Lancet 2017, 389, 1011–1024. [Google Scholar] [CrossRef]
- O’Donnell, J.S.; Hoefsmit, E.P.; Smyth, M.J.; Blank, C.U.; Teng, M.W.L. The promise of neoadjuvant immunotherapy and surgery for cancer treatment. Clin. Cancer Res. 2019, 25, 5743–5751. [Google Scholar] [CrossRef] [PubMed]
- Amin, S.; Baine, M.; Meza, J.; Lin, C. The impact of neoadjuvant and adjuvant immunotherapy on the survival of pancreatic cancer patients: A retrospective analysis. BMC Cancer 2020, 20, 1–10. [Google Scholar] [CrossRef]
- Tran, T.B.; Maker, V.K.; Maker, A.V. Impact of Immunotherapy after Resection of Pancreatic Cancer. J. Am. Coll. Surg. 2019, 229, 19–27.e1. [Google Scholar] [CrossRef]
- Von Hoff, D.D.; Ramanathan, R.K.; Borad, M.J.; Laheru, D.A.; Smith, L.S.; Wood, T.E.; Korn, R.L.; Desai, N.; Trieu, V.; Iglesias, J.L.; et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: A phase I/II trial. J. Clin. Oncol. 2011, 29, 4548–4554. [Google Scholar] [CrossRef] [Green Version]
- Homma, Y.; Taniguchi, K.; Murakami, T.; Nakagawa, K.; Nakazawa, M.; Matsuyama, R.; Mori, R.; Takeda, K.; Ueda, M.; Ichikawa, Y.; et al. Immunological impact of neoadjuvant chemoradiotherapy in patients with borderline resectable pancreatic ductal adenocarcinoma. Ann. Surg. Oncol. 2014, 21, 670–676. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Renouf, D.J.; Dhani, N.C.; Kavan, P.; Jonker, D.J.; Wei, A.C.; Tang, P.A.; Graham, B.; Gallinaro, L.; Hasan, T.; Li, W.; 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 pancreat. J. Clin. Oncol. 2018, 36, 349. [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.; et al. 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 2019, 79, CT004. [Google Scholar] [CrossRef]
- Date, V.; Nair, S. Emerging vistas in CAR T-cell therapy: Challenges and Opportunities in Solid Tumors. Expert Opin. Biol. Ther. 2020, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Beatty, G.L.; O’Hara, M.H.; Lacey, S.F.; Torigian, D.A.; Nazimuddin, F.; Chen, F.; Kulikovskaya, I.M.; Soulen, M.C.; McGarvey, M.; Nelson, A.M.; et al. Activity of Mesothelin-Specific Chimeric Antigen Receptor T Cells Against Pancreatic Carcinoma Metastases in a Phase 1 Trial. Gastroenterology 2018, 155, 29–32. [Google Scholar] [CrossRef] [PubMed]
- Raj, D.; Yang, M.H.; Rodgers, D.; Hampton, E.N.; Begum, J.; Mustafa, A.; Lorizio, D.; Garces, I.; Propper, D.; Kench, J.G.; et al. Switchable CAR-T cells mediate remission in metastatic pancreatic ductal adenocarcinoma. Gut 2019, 68, 1052–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stromnes, I.M.; Schmitt, T.M.; Hulbert, A.; Brockenbrough, J.S.; Nguyen, H.; Cuevas, C.; Dotson, A.M.; Tan, X.; Hotes, J.L.; Greenberg, P.D.; et al. T cells Engineered against a native antigen can surmount immunologic and physical barriers to treat pancreatic ductal adenocarcinoma. Physiol. Behav. 2016, 176, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stromnes, I.M.; Burrack, A.L.; Hulbert, A.; Bonson, P.; Brockenbrough, J.S.; Raynor, J.F.; Spartz, E.J.; Robert, H.; Greenberg, P.D.; Hingorani, S.R.; et al. Differential effects of depleting versus programming tumor-associated macrophages on engineered T cells in pancreatic ductal adenocarcinoma. Cancer Immunol. Res. 2020, 7, 977–989. [Google Scholar] [CrossRef] [PubMed]
- Oda, S.K.; Anderson, K.G.; Ravikumar, P.; Bonson, P.; Garcia, N.M.; Jenkins, C.M.; Zhuang, S.; Daman, A.W.; Chiu, E.Y.; Bates, B.M.; et al. A Fas-4-1BB fusion protein converts a death to a pro-survival signal and enhances T cell therapy. J. Exp. Med. 2020, 217, e20191166. [Google Scholar] [CrossRef]
- Chanyi, R.M.; Craven, L.; Harvey, B.; Reid, G.; Silverman, M.J.; Burton, J.P. Faecal microbiota transplantation: Where did it start? What have studies taught us? Where is it going? SAGE Open Med. 2017, 5, 205031211770871. [Google Scholar] [CrossRef]
- Tanoue, T.; Morita, S.; Plichta, D.R.; Skelly, A.N.; Suda, W.; Sugiura, Y.; Narushima, S.; Vlamakis, H.; Motoo, I.; Sugita, K.; et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019, 565, 600–605. [Google Scholar] [CrossRef]
- Beatty, G.L.; Eghbali, S.; Kim, R. Deploying Immunotherapy in Pancreatic Cancer: Defining Mechanisms of Response and Resistance. Am. Soc. Clin. Oncol. Educ. Book 2017, 37, 267–278. [Google Scholar] [CrossRef]
- Lutz, E.R.; Wu, A.A.; Bigelow, E.; Sharma, R.; Mo, G.; Soares, K.; Solt, S.; Dorman, A.; Wamwea, A.; Yager, A.; et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol. Res. 2014, 2, 616–631. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Krishnamoorthy, M.; Lenehan, J.G.; Burton, J.P.; Maleki Vareki, S. Immunomodulation in Pancreatic Cancer. Cancers 2020, 12, 3340. https://doi.org/10.3390/cancers12113340
Krishnamoorthy M, Lenehan JG, Burton JP, Maleki Vareki S. Immunomodulation in Pancreatic Cancer. Cancers. 2020; 12(11):3340. https://doi.org/10.3390/cancers12113340
Chicago/Turabian StyleKrishnamoorthy, Mithunah, John G. Lenehan, Jeremy P. Burton, and Saman Maleki Vareki. 2020. "Immunomodulation in Pancreatic Cancer" Cancers 12, no. 11: 3340. https://doi.org/10.3390/cancers12113340
APA StyleKrishnamoorthy, M., Lenehan, J. G., Burton, J. P., & Maleki Vareki, S. (2020). Immunomodulation in Pancreatic Cancer. Cancers, 12(11), 3340. https://doi.org/10.3390/cancers12113340