New Pharmacological Approaches for Pancreatic Cancer Therapy
A Light at the End of the Tunnel?
Vineet K. Gupta
Sylvester Comprehensive Cancer Center and DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, USA
Search for more papers by this authorSulagna Banerjee
Sylvester Comprehensive Cancer Center and DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, USA
Search for more papers by this authorAshok K. Saluja
Sylvester Comprehensive Cancer Center and DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, USA
Search for more papers by this authorVineet K. Gupta
Sylvester Comprehensive Cancer Center and DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, USA
Search for more papers by this authorSulagna Banerjee
Sylvester Comprehensive Cancer Center and DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, USA
Search for more papers by this authorAshok K. Saluja
Sylvester Comprehensive Cancer Center and DeWitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, USA
Search for more papers by this authorJ. Enrique Domínguez-Muñoz MD, PhD
Director
Department of Gastroenterology and Hepatology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
Search for more papers by this authorSummary
Pancreatic cancer is one of the most aggressive malignancies worldwide, with a five-year survival rate of only 9%. Treatment of pancreatic cancer involves a multidisciplinary approach using chemotherapy, surgery, radiation therapy, and palliative care, which are selected based on stage of the disease. One of the main challenges when faced with this deadly disease is the heterogeneity among patients with regard to symptoms, genetic mutation, tumor evolution, sensitivity to treatments, and predisposition to early metastasis. Desmoplasia, one of the key features of pancreatic tumors, is responsible for the low survival rate in patients with this cancer. Surgical resection of tumor remains the best curative treatment available to pancreatic cancer patients. Understanding the pathogenesis of pancreatic cancer has revealed various potential targets for the development of effective therapy. Metabolic inhibitors or antimetabolites have been used in cancer therapy for more than 50 years.
References
- Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69(1): 7–34.
- Saluja A, Maitra A. Pancreatitis and pancreatic cancer. Gastroenterology 2019; 156(7): 1937–1940.
- Burris HA III, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 1997; 15(6): 2403–2413.
- Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011; 364(19): 1817–1825.
- Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013; 369(18): 1691–1703.
- Collisson EA, Bailey P, Chang DK, Biankin AV. Molecular subtypes of pancreatic cancer. Nat Rev Gastroenterol Hepatol 2019; 16(4): 207–220.
- Singh S, Hasselluhn MC, Neesse A. A tangled tale of molecular subtypes in pancreatic cancer. Gut 2019; 68(6): 953–954.
- Kloppel G, Luttges J. WHO classification 2000: exocrine pancreatic tumors. Verh Dtsch Ges Pathol 2001; 85: 219–228.
- Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016; 531(7592): 47–52.
- Bertucci F, Birnbaum DJ, Finetti P, et al. Prognostic value of molecular subtypes in pancreatic cancer. Pancreas 2017; 46(4): e29–e31.
- Puleo F, Nicolle R, Blum Y, et al. Stratification of pancreatic ductal adenocarcinomas based on tumor and microenvironment features. Gastroenterology 2018; 155(6): 1999–2013.e3.
- Apte MV, Wilson JS, Lugea A, Pandol SJ. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology 2013; 144(6): 1210–1219.
- Apte MV, Park S, Phillips PA, et al. Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas 2004; 29(3): 179–187.
- Bachem MG, Schunemann M, Ramadani M, et al. Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology 2005; 128(4): 907–921.
- Lohr M, Schmidt C, Ringel J, et al. Transforming growth factor-beta1 induces desmoplasia in an experimental model of human pancreatic carcinoma. Cancer Res 2001; 61(2): 550–555.
- Apte MV, Haber PS, Darby SJ, et al. Pancreatic stellate cells are activated by proinflammatory cytokines: implications for pancreatic fibrogenesis. Gut 1999; 44(4): 534–541.
- Heldin CH, Rubin K, Pietras K, Ostman A. High interstitial fluid pressure: an obstacle in cancer therapy. Nat Rev Cancer 2004; 4(10): 806–813.
- Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009; 324(5933): 1457–1461.
- Rosty C, Goggins M. Early detection of pancreatic carcinoma. Hematol Oncol Clin North Am 2002; 16(1): 37–52.
- Aggarwal G, Rabe KG, Petersen GM, Chari ST. New-onset diabetes in pancreatic cancer: a study in the primary care setting. Pancreatology 2012; 12(2): 156–161.
- Gullo L, Tomassetti P, Migliori M, et al. Do early symptoms of pancreatic cancer exist that can allow an earlier diagnosis? Pancreas 2001; 22(2): 210–213.
- Wong D, Ko AH, Hwang J, et al. Serum CA19-9 decline compared to radiographic response as a surrogate for clinical outcomes in patients with metastatic pancreatic cancer receiving chemotherapy. Pancreas 2008; 37(3): 269–274.
- Chari ST, Kelly K, Hollingsworth MA, et al. Early detection of sporadic pancreatic cancer: summative review. Pancreas 2015; 44(5): 693–712.
- Huang Z, Liu F. Diagnostic value of serum carbohydrate antigen 19-9 in pancreatic cancer: a meta-analysis. Tumour Biol 2014; 35(8): 7459–7465.
- Hasan S, Jacob R, Manne U, Paluri R. Advances in pancreatic cancer biomarkers. Oncol Rev 2019; 13(1): 410.
- Koopmann J, Rosenzweig CN, Zhang Z, et al. Serum markers in patients with resectable pancreatic adenocarcinoma: macrophage inhibitory cytokine 1 versus CA19-9. Clin Cancer Res 2006; 12(2): 442–446.
- Liu D, Chang CH, Gold DV, Goldenberg DM. Identification of PAM4 (clivatuzumab)-reactive epitope on MUC5AC: a promising biomarker and therapeutic target for pancreatic cancer. Oncotarget 2015; 6(6): 4274–4285.
- Melo SA, Luecke LB, Kahlert C, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015; 523(7559): 177–182.
- Buanes TA. Role of surgery in pancreatic cancer. World J Gastroenterol 2017; 23(21): 3765–3770.
- Kalser MH, Ellenberg SS. Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 1985; 120(8): 899–903.
- Neoptolemos JP, Dunn JA, Stocken DD, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet 2001; 358(9293): 1576–1585.
- Uegaki K, Nio Y, Inoue Y, et al. Clinicopathological significance of epidermal growth factor and its receptor in human pancreatic cancer. Anticancer Res 1997; 17(5B): 3841–3847.
- Lee CC, Shiao HY, Wang WC, Hsieh HP. Small-molecule EGFR tyrosine kinase inhibitors for the treatment of cancer. Expert Opin Investig Drugs 2014; 23(10): 1333–1348.
- Bloomston M, Bhardwaj A, Ellison EC, Frankel WL. Epidermal growth factor receptor expression in pancreatic carcinoma using tissue microarray technique. Dig Surg 2006; 23(1–2): 74–79.
- Einama T, Ueda S, Tsuda H, et al. Membranous and cytoplasmic expression of epidermal growth factor receptor in metastatic pancreatic ductal adenocarcinoma. Exp Ther Med 2012; 3(6): 931–936.
- Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2007; 25(15): 1960–1966.
- Philip PA, Benedetti J, Corless CL, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol 2010; 28(22): 3605–3610.
- Halfdanarson TR, Foster NR, Kim GP, et al. A phase II randomized trial of panitumumab, erlotinib, and gemcitabine versus erlotinib and gemcitabine in patients with untreated, metastatic pancreatic adenocarcinoma: North Central Cancer Treatment Group Trial N064B (Alliance). Oncologist 2019; 24(5): 589–e160.
- Aghdassi A, Phillips P, Dudeja V, et al. Heat shock protein 70 increases tumorigenicity and inhibits apoptosis in pancreatic adenocarcinoma. Cancer Res 2007; 67(2): 616–625.
- Phillips PA, Dudeja V, McCarroll JA, et al. Triptolide induces pancreatic cancer cell death via inhibition of heat shock protein 70. Cancer Res 2007; 67(19): 9407–9416.
-
Chugh R, Sangwan V, Patil SP, et al. A preclinical evaluation of Minnelide as a therapeutic agent against pancreatic cancer. Sci Transl Med
2012; 4(156): 156ra39.
10.1126/scitranslmed.3004334 Google Scholar
- Banerjee S, Sangwan V, McGinn O, et al. Triptolide-induced cell death in pancreatic cancer is mediated by O-GlcNAc modification of transcription factor Sp1. J Biol Chem 2013; 288(47): 33927–33938.
- Modi S, Kir D, Giri B, et al. Minnelide overcomes oxaliplatin resistance by downregulating the DNA repair pathway in pancreatic cancer. J Gastrointest Surg 2016; 20(1): 13–24.
- Sherman MH, Yu RT, Engle DD, et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 2014; 159(1): 80–93.
- Dauer P, Zhao X, Gupta VK, et al. Inactivation of cancer-associated-fibroblasts disrupts oncogenic signaling in pancreatic cancer cells and promotes its regression. Cancer Res 2018; 78(5): 1321–1333.
- Banerjee S, Modi S, McGinn O, et al. Impaired synthesis of stromal components in response to minnelide improves vascular function, drug delivery, and survival in pancreatic cancer. Clin Cancer Res 2016; 22(2): 415–425.
- Jaster R, Hilgendorf I, Fitzner B, et al. Regulation of pancreatic stellate cell function in vitro: biological and molecular effects of all-trans retinoic acid. Biochem Pharmacol 2003; 66(4): 633–641.
- Bailey JM, Swanson BJ, Hamada T, et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin Cancer Res 2008; 14(19): 5995–6004.
- Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013; 62(1): 112–120.
- Hingorani SR, Zheng L, Bullock AJ, et al. HALO 202: Randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol 2018; 36(4): 359–366.
- Kozono S, Ohuchida K, Eguchi D, et al. Pirfenidone inhibits pancreatic cancer desmoplasia by regulating stellate cells. Cancer Res 2013; 73(7): 2345–2356.
- Liyanage UK, Moore TT, Joo HG, et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol 2002; 169(5): 2756–2761.
- Zhao F, Obermann S, von Wasielewski R, et al. Increase in frequency of myeloid-derived suppressor cells in mice with spontaneous pancreatic carcinoma. Immunology 2009; 128(1): 141–149.
- Amedei A, Niccolai E, Benagiano M, et al. Ex vivo analysis of pancreatic cancer-infiltrating T lymphocytes reveals that ENO-specific Tregs accumulate in tumor tissue and inhibit Th1/Th17 effector cell functions. Cancer Immunol Immunother 2013; 62(7): 1249–1260.
- Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013; 13(4): 227–242.
- Nomi T, Sho M, Akahori T, et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res 2007; 13(7): 2151–2157.
- Geng L, Huang D, Liu J, et al. B7-H1 up-regulated expression in human pancreatic carcinoma tissue associates with tumor progression. J Cancer Res Clin Oncol 2008; 134(9): 1021–1027.
- 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 2012; 366(26): 2455–2465.
- Winograd R, Byrne KT, Evans RA, et al. Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol Res 2015; 3(4): 399–411.
- 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 2010; 33(8): 828–833.
- 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 2011; 253(2): 328–335.
- Lutz ER, Wu AA, Bigelow E, et al. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res 2014; 2(7): 616–631.
- 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 2013; 36(7): 382–389.
- Park B, Yee C, Lee KM. The effect of radiation on the immune response to cancers. Int J Mol Sci 2014; 15(1): 927–943.
- Seifert L, Werba G, Tiwari S, et al. Radiation therapy induces macrophages to suppress T-cell responses against pancreatic tumors in mice. Gastroenterology 2016; 150(7): 1659–1672.e5.
- Ruffell B, Affara NI, Coussens LM. Differential macrophage programming in the tumor microenvironment. Trends Immunol 2012; 33(3): 119–126.
- Sanford DE, Belt BA, Panni RZ, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res 2013; 19(13): 3404–3415.
- Zhu Y, Knolhoff BL, Meyer MA, et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res 2014; 74(18): 5057–5069.
- Luengo A, Gui DY, Vander Heiden MG. Targeting metabolism for cancer therapy. Cell Chem Biol 2017; 24(9): 1161–1180.
- Rajeshkumar NV, Dutta P, Yabuuchi S, et al. Therapeutic targeting of the Warburg effect in pancreatic cancer relies on an absence of p53 function. Cancer Res 2015; 75(16): 3355–3364.
- Kim DJ, Park YS, Kang MG, et al. Pyruvate kinase isoenzyme M2 is a therapeutic target of gemcitabine-resistant pancreatic cancer cells. Exp Cell Res 2015; 336(1): 119–129.
- Rajeshkumar NV, Yabuuchi S, Pai SG, et al. Treatment of pancreatic cancer patient-derived xenograft panel with metabolic inhibitors reveals efficacy of phenformin. Clin Cancer Res 2017; 23(18): 5639–5647.
- Viale A, Pettazzoni P, Lyssiotis CA, et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014; 514(7524): 628–632.
- Li D, Fu Z, Chen R, et al. Inhibition of glutamine metabolism counteracts pancreatic cancer stem cell features and sensitizes cells to radiotherapy. Oncotarget 2015; 6(31): 31151–31163.
- Gupta VK, Sharma NS, Kesh K, et al. Metastasis and chemoresistance in CD133 expressing pancreatic cancer cells are dependent on their lipid raft integrity. Cancer Lett 2018; 439: 101–112.
- Lee HS, Lee SH, Lee HJ, et al. Statin use and its impact on survival in pancreatic cancer patients. Medicine (Baltimore) 2016; 95(19): e3607.