Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2071 word(s) 2071 2021-02-25 09:03:27 |
2 Format correct Meta information modification 2071 2021-02-25 11:02:12 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Rho, G. Pancreatic Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/7587 (accessed on 27 July 2024).
Rho G. Pancreatic Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/7587. Accessed July 27, 2024.
Rho, Gyu-Jin. "Pancreatic Cancer" Encyclopedia, https://encyclopedia.pub/entry/7587 (accessed July 27, 2024).
Rho, G. (2021, February 25). Pancreatic Cancer. In Encyclopedia. https://encyclopedia.pub/entry/7587
Rho, Gyu-Jin. "Pancreatic Cancer." Encyclopedia. Web. 25 February, 2021.
Pancreatic Cancer
Edit

Pancreatic cancer arises when cells in the pancreas start to divide uncontrollably and form a mass. There are different types of cancer cells based on their origin, for example, carcinoma (cancer of epithelial cells), sarcoma (cancer of mesenchymal cells in blood vessels, muscles, and other tissues), myeloma/leukemia/lymphoma (blood cell-related cancer), and adenocarcinoma (cancer of mucus-producing glandular cells).

Pancreatic Cancer Immunotherapy

1. Introduction

The pancreas is an organ located in the abdomen having both exocrine and endocrine functions. It plays an essential role in the digestion of food by releasing enzymes from its exocrine part, maintains blood glucose level by producing two major hormones: glucagon and insulin secreted from the endocrinal region of the pancreas. Normal healthy cells become cancerous when a series of changes take place in the DNA sequence, leads the cell to divide uncontrollably and migrate to adjacent cells. Cancer is the second leading cause of death worldwide and was accountable for an estimated 9.6 million deaths in 2018 (World Health Organization (WHO), 2018). It is the major public health issue and the main cause of death in Korea [1], second leading in the United States [2], and one of the leading causes of death in India [3]. One of the leading causes of cancer mortality and the most deadly malignant neoplasm is pancreatic cancer [4]. In 2012, around 338,000 individuals had pancreatic cancer worldwide, making it the eleventh most prevalent cancer. Around 458,918 new pancreatic cancer cases were identified worldwide in 2018, representing 2.5% of all cancers [5]. The American Cancer Society estimated about 57,600 new cases (30,400 male and 27,200 female) of pancreatic cancer and predicted that 47,050 patients (24,640 male and 22,410 female) will die of pancreatic cancer in 2020 [2]. In 2019, pancreatic cancer was the fourth leading cause of cancer deaths. It has been projected to become the second leading cause by 2030 [6][7].

Pancreatic cancer arises when cells in the pancreas start to divide uncontrollably and form a mass. There are different types of cancer cells based on their origin, for example, carcinoma (cancer of epithelial cells), sarcoma (cancer of mesenchymal cells in blood vessels, muscles, and other tissues), myeloma/leukemia/lymphoma (blood cell-related cancer), and adenocarcinoma (cancer of mucus-producing glandular cells). Two main subtypes of pancreatic cancer have been narrowly classified into exocrine and endocrine. Pancreatic ductal adenocarcinoma (PDAC) is an exocrine cell tumor mainly of the ductal cells, more common (>85%) than endocrine cell tumors (<5%) [8]. About 50% of PDACs are detected when the tumor is locally invasive or metastatic. PDAC has a 5-year survival rate of 6% (ranges from 2% to 10%) [6][9]. Exocrine cancer is the most common form of pancreatic cancer, which comprises 95% of all pancreatic cancers [10][11]. Out of all exocrine cancers, the most common and aggressive form is ductal cancer, i.e., PDAC. It is one of the most malignant tumors, characterized by uncontrollable growth [9]. Approximately 85% to 90% of pancreatic cancers are PDAC [11]. Recently, researchers have reviewed the current therapeutic options, dysregulated pathways, tumor microenvironment, and many other factors associated with PDAC [6][12]. Approximately 60% to 70% of cases emerge from the head of the pancreas, which comprises the bile duct; these cases are typically diagnosed earlier than body and tail tumors [13]. Tail and body tumors are linked with a poorer prognosis [14]. In patients with PDAC, the most common symptoms are abdominal pain, weight loss, and jaundice [15], whereas the new onset of type 2 diabetes is a less common symptom [16].

Additionally, studies have shown that PDAC and diabetes are co-related; at the time of cancer diagnosis, one- to two-thirds of patients with PDAC are diabetic [17]. The key concern is whether the growth of cancer is susceptible to diabetes or the consequence of the tumor is diabetes. The five leading behavioral and dietary risks, such as high body mass index, low consumption of fruit and vegetables, physical inactivity, alcohol, and tobacco, are responsible for about one-third of cancer deaths [4]. About 8% of pancreatic cancers occur in families who carry mutations in tumor suppressor genes, including P16Ink4a/CDKN2A, BRCA2, MLH1, MSH2, STK1, or VHL [18]. In 95% of PDAC cases, activating mutations in the KRAS oncogene are detected, but agents that can successfully target this high prevalence change in PDAC are not yet available. Available traditional strategies: surgery, radiation, and chemotherapy have been widely used, but no significant improvements have been shown. Overall survival remains poor for metastatic cancer, with less than 20% of patients surviving after the end of the first year [19]. For the better treatment of PDAC, alternative treatment approaches are desperately needed. Furthermore, stem cell therapy, which has shown therapeutic efficacy for solid tumors (breast, prostate, and lung carcinomas), can be one of the best options to treat PDAC [20].

2. Immunotherapy for Pancreatic Cancer

Several targeted strategies, including new stromal modulation, immunotherapeutic approaches, and targeting main signaling pathway effectors, are in progress, along with the development of novel cytotoxic therapeutic strategies. The stroma encompasses approximately 90% of the tumor mass, which promotes the progression of fibrosis and immunosuppression [21]. In addition to facilitating tumor development, the PDAC stroma has been shown to attenuate the delivery of antitumor treatments, inactivation of cytotoxic CD8+ T cells, and increasing the number of immunosuppressive cells [22][23]. During the progression of the disease, the number of pancreatic stellate cells and PDAC specific cancer associated fibroblasts increase abundantly [24]. These activated stellate cells promote tumor growth by reducing the migration of CD8+ T cells to juxtatumoral stromal compartments [25]. Stellate cells also stimulate T cell anergy and apoptosis induced by galectin-1, resulting in evasion of immune surveillance by the cancer cells [25][26].

Furthermore, B lymphocytes contribute actively to PDAC fibrogenesis by activation and differentiation of cancer associated fibroblasts [27]. Minici et al. reviewed the immunological mechanisms that promote and inhibit the anti-tumor immunity of B cells. B cells can restrict tumor growth through phagocytosis by macrophages, facilitating tumor killing by NK cells, generating tumor-reactive antibodies, and the priming of CD4+ and CD8+ T cells [28]. B cells can facilitate tumor growth through the production of autoantibodies and tumor growth factors [28]. Further, targeting particular B cell subtypes can be beneficial for the treatment of cancer as the activities of Th1, and CD8+ cytolytic T cells can be directly and indirectly inhibited by regulatory B cells.

Presently, many clinical trials are trying to evaluate the efficiency of immunotherapeutic approaches in PDAC, including cancer vaccination [29], immune checkpoint inhibitors [30], monoclonal antibodies, adoptive cell transfer [31], chemo-radiotherapy or other molecularly focused agents, and combinations with other immunotherapeutic agents or immune modulators, though none of these studies have demonstrated improvements in practice. Activating a patient′s T cells is the key basis of cancer immunotherapy in order to destroy tumor cells. Furthermore, important steps of immunotherapy are defined as follows: reduction in tumor-specific cells presenting antigen, T cell activation, T cells infiltration into tumors, cancer cell recognition by T cells, and cancer cell elimination [32]. Anti-CTLA-4 (Ipilimumab) and anti-PD-1/anti-PDL-1 (Nivolumab/Pembrolizumab) agents have shown promising results in the activation of T cells and offer an efficient tumor immunotherapy strategy [33]. Despite showing Powerful outcomes of some malignancies, most of them in phase I and II clinical studies have not shown any clinical effectiveness in PDAC [34]. The immunosuppressive activity of CTL-4 results in the reduction of T effector cell activation and elevation in the activity of T regulatory cells [35]. The programmed cell death protein 1 (PD-1) is present largely on T cells, tumor cells, and tumor infiltrating lymphocytes [28]. The binding of PD-1 (Programmed death-ligand, PDL-1/PDL-2) leads to a reduction in T cell proliferation and secretion of antitumor cytokines [28].

A varied range of clinical trials (Table 1) on pancreatic cancer based on cytotoxic chemotherapy, vaccine-associated checkpoint inhibitors, immune checkpoint monotherapy, dual checkpoint combination therapy, and using other inhibitory agents have been completed or are presently ongoing. These clinical trials followed several therapeutic techniques:

Table 1. Clinical trials of novel agents for PDAC and other pancreatic cancers.

Pathological Condition Enrolled Patients Intervention National Clinical Trial Number Outcome Measures Phase Status Result
Neoplasms, Pancreas 40 Cancer stem cell vaccine NCT02074046 Determine the safety of immunization Phase 1/2 Completed CTLs harvested from CSC-vaccinated hosts were capable of killing CSCs in vitro
Metastatic pancreatic cancer 98 Gemcitabine, Nab-Paclitaxel, GDC-0449 NCT01088815 Progression free survival, safety of combination therapy Phase 2 Completed Median progression-free survival and overall survival were 5.42 months and 9.79 months, respectively
Metastatic pancreatic adenocarcinoma 139 BBI608 either in combination with Gemcitabine and nab-Paclitaxel, mFOLFIRINOX, FOLFIRI, or MM-398 with 5-FU and Leucovorin NCT02231723 Safety, Adverse effects Phase 1 Completed Inhibit cancer stemness pathways, including Nanog, by targeting stemness kinases.
Metastatic Pancreatic Ductal Adenocarcinoma 65 MEDI4736 Monotherapy, Tremelimumab + MEDI4736 NCT02558894 Response Rate, Overall survival, progression free survival, Phase 2 Completed Monotherapy reflected a population of patients with mPDAC who had poor prognoses and rapidly progressing disease
PDAC, Pancreatic Cancer 21 Ipilimumab, Gemcitabine hydrochloride NCT01473940 Overall survival, progression free survival, recovery of tumor immune surveillance Phase 1 Completed Median progression-free and overall survival were 2.78 months and 6.90 months, respectively.
Second-line, third-line and Greater Metastatic Pancreatic Cancer 303 GVAX Pancreas Vaccine, CRS-207, Chemotherapy, Cyclophosphamide NCT02004262 Overall survival and adverse effects Phase 2 Completed Median overall survival in the primary cohort was 3.7, 5.4, and 4.6 months in arms A, B, and C, respectively (*)
Pancreatic Neoplasm 22 Monoclonal antibody, chemotherapy NCT00711191 Overall survival, progression free survival, and time to Progression Phase 1 Completed Well tolerated and associated with antitumor activity in patients with PDAC and improved overall survival
Pancreatic
Adenocarcinoma metastatic
10 Melphalan, BCNU, Vitamin B12, Vitamin C, and autologous hematopoietic stem cell NCT04150042 Response rate in metastatic lesions, overall survival, progression free survival Phase 1 Ongoing NA
Resectable pancreatic adenocarcinoma 42 HIPEC-Gemcitabine NCT03251365 Morbidity, survival Phase 2/3 Ongoing NA
PDAC, pancreatic cancer, metastasis 36 Ascorbic acid, Paclitaxel, Cisplatin, Gemcitabine NCT03410030 Determination of preliminary efficacy Phase 1/2 Ongoing NA
Pancreatic Cancer 81 Pembrolizumab, Gemcitabine, Docetaxel, Nab-paclitaxel, Vinorelbine, Irinotecan, Liposomal Doxorubicin NCT02331251 Determine the recommended phase 2 dose Phase 1/2 Terminated The median progression-free survival and overall survival was 9.1 and 15.0 months, respectively
Pancreatic Cancer 15 Fludarabine, Anti-mesothelin chimeric T cell receptor (CAR) transduced peripheral blood lymphocytes (PBL), Cyclophosphamide, Aldesleukin NCT01583686 Tumor regression response and adverse effects Phase 1/2 Terminated MORAb-009 (chimeric monoclonal antibody) is well tolerated
Pancreatic adenocarcinoma 10 Allogeneic hematopoietic stem cell transplantation NCT02207985 Disease free survival Phase 1/2 Unknown Patients are tumor-free for 9 years after diagnosis
* CTLs: cytotoxic T lymphocytes; Cy/GVAX + CRS-207 (arm A), CRS-207 (arm B), or physician′s choice of single-agent chemotherapy (arm C); HIPEC: Hyperthermic Intraperitoneal Chemotherapy; NA: Not available.
  • Monotherapy includes the administration of several PD-1(MEDI4736, MPDL3280A, and pembrolizumab,) and CTL-4 (tremelimumab and ipilimumab) inhibitors and inhibition of double checkpoints: either by a combination of the above mentioned inhibitors or with other agents, such as anti-CCR-5 (mogamulizumab) [36].
  • Combination of chemotherapeutic agents and immune checkpoint inhibitors: PD-1/CTL4 inhibitors leads to the activation of T cell that is efficient for immunotherapy. When PD-1/CTL4 inhibitors combined with commonly used chemotherapeutic agents such as Nab-paclitaxel, gemcitabine, carboplatin, and FOLFOX improved overall survival [37]. Remarkably, therapeutic procedures using a combination of immune checkpoint inhibitors with radiotherapy or chemotherapy have shown significant outcomes [38][39].
  • Vaccination therapy is founded on the basis of the distribution of tumor antigens to antigen presenting cells (APCs), followed by induction of an organized immune response. Cancer specific DNA mutations produce new antigens, which, in turn, results in a unique sequence of the peptide. Variety of vaccines for pancreatic cancer treatment includes whole-cell vaccines, dendritic-cell based vaccines, peptide and DNA vaccines, telomerase peptide vaccines, Ras peptide vaccines, and survivin-targeted vaccines [40]; however, regardless of the enhanced immune system, showed poor clinical results. GVAX is an allogeneic irradiated whole-cell tumor vaccine genetically modified for the secretion of granulocyte macrophage colony stimulating factor and promotes cytolytic action against tumors, the most widely studied vaccine for pancreatic cancer [41]. Furthermore, the clinical studies when GVAX is applied in combination with 5-Fluorouracil/cyclophosphamide based chemotherapy have shown the same results regarding disease-free and median survival as that of GVAX applied alone [42]. On the other hand, when the above mentioned ipilimumab (immune checkpoint inhibitor) is applied in combination with GVAX, it leads to better survival [43].
  • Adoptive T cell immunotherapy is based on the modification of autologous T cells, which stimulates the immune response against the tumor. The patients receiving mesothelin-targeting chimeric antigen receptor-T (CAR) cells have shown overexpression of a membrane antigen in pancreatic cancer, exhibited adequate patience but unsuccessful in showing good results [44]. Along with mesothelin, other cancer-associated antigens are being studied alone or in combination with chemotherapy as potential targets of CAR-T cells based therapy [44].
  • Immune modulating agents that target the microenvironment of the pancreas can also exert extensive antitumor activity. Anti-CD40 agonistic antibodies used in combination with gemcitabine in PDAC patients showed significant results [45]. PDAC patients treated with a CCR2 inhibitor (PF-04136309) exhibited fractional response and constant tumor when used in combination with FOLFIRINOX [46]. Several chemokine receptor molecules are under examination in clinical trials against PDAC.

References

  1. Hong, S.; Won, Y.J.; Park, Y.R.; Jung, K.W.; Kong, H.J.; Lee, E.S. Cancer Statistics in Korea: Incidence, Mortality, Survival, and Prevalence in 2017. Cancer Res. Treat. Off. J. Kor. Cancer Assoc. 2020, 52, 335–350.
  2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30.
  3. Mathur, P.; Sathishkumar, K.; Chaturvedi, M.; Das, P.; Sudarshan, K.L.; Santhappan, S.; Nallasamy, V.; John, A.; Narasimhan, S.; Roselind, F.S. ICMR-NCDIR-NCRP Investigator Group. Cancer Statistics, 2020: Report From National Cancer Registry Programme, India. JCO Glob. Oncol. 2020, 6, 1063–1075.
  4. Ilic, M.; Ilic, I. Epidemiology of pancreatic cancer. World J. Gastroenterol. 2016, 22, 9694–9705.
  5. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424.
  6. Sarantis, P.; Koustas, E.; Papadimitropoulou, A.; Papavassiliou, A.G.; Karamouzis, M.V. Pancreatic ductal adenocarcinoma: Treatment hurdles, tumor microenvironment and immunotherapy. World J. Gastrointest. Oncol. 2020, 12, 173–181.
  7. Ying, H.; Dey, P.; Yao, W.; Kimmelman, A.C.; Draetta, G.F.; Maitra, A.; DePinho, R.A. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2016, 30, 355–385.
  8. Hidalgo, M.; Cascinu, S.; Kleeff, J.; Labianca, R.; Löhr, J.M.; Neoptolemos, J.; Real, F.X.; Van Laethem, J.L.; Heinemann, V. Addressing the challenges of pancreatic cancer: Future directions for improving outcomes. Pancreatology 2015, 15, 8–18.
  9. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2017. CA Cancer J. Clin. 2017, 67, 7–30.
  10. Zhou, J.; Enewold, L.; Stojadinovic, A.; Clifton, G.T.; Potter, J.F.; Peoples, G.E.; Zhu, K. Incidence rates of exocrine and endocrine pancreatic cancers in the United States. Cancer Causes Control. 2010, 21, 853–861.
  11. Bond-Smith, G.; Banga, N.; Hammond, T.M.; Imber, C.J. Pancreatic adenocarcinoma. BMJ 2012, 344, e2476.
  12. 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.
  13. Corbo, V.; Tortora, G.; Scarpa, A. Molecular pathology of pancreatic cancer: From bench-to-bedside translation. Curr. Drug Targ. 2012, 13, 744–752.
  14. Ghaneh, P.; Costello, E.; Neoptolemos, J.P. Biology and management of pancreatic cancer. Postgrad. Med. J. 2008, 84, 478–497.
  15. Porta, M.; Fabregat, X.; Malats, N.; Guarner, L.; Carrato, A.; de Miguel, A.; Ruiz, L.; Jariod, M.; Costafreda, S.; Coll, S.; et al. Exocrine pancreatic cancer: Symptoms at presentation and their relation to tumour site and stage. Clin. Transl. Oncol. 2005, 7, 189–197.
  16. De Souza, A.; Khawaja, K.I.; Masud, F.; Saif, M.W. Metformin and pancreatic cancer: Is there a role? Cancer Chemother. Pharmacol. 2016, 77, 235–242.
  17. Ben, Q.; Xu, M.; Ning, X.; Liu, J.; Hong, S.; Huang, W.; Zhang, H.; Li, Z. Diabetes mellitus and risk of pancreatic cancer: A meta-analysis of cohort studies. Eur. J. Cancer 2011, 47, 1928–1937.
  18. Wormann, S.M.; Algul, H. Risk factors and therapeutic targets in pancreatic cancer. Front. Oncol. 2013, 3, 282.
  19. Mayo, S.C.; Nathan, H.; Cameron, J.L.; Olino, K.; Edil, B.H.; Herman, J.M.; Hirose, K.; Schulick, R.D.; Choti, M.A.; Wolfgang, C.L.; et al. Conditional survival in patients with pancreatic ductal adenocarcinoma resected with curative intent. Cancer 2012, 118, 2674–2681.
  20. Kanojia, D.; Balyasnikova, I.V.; Morshed, R.A.; Frank, R.T.; Yu, D.; Zhang, L.; Spencer, D.A.; Kim, J.W.; Han, Y.; Yu, D.; et al. Neural stem cells secreting anti-her2 antibody improve survival in a preclinical model of her2 overexpressing breast cancer brain metastases. Stem Cells 2015, 33, 2985–2994.
  21. Neesse, A.; Michl, P.; Frese, K.K.; Feig, C.; Cook, N.; Jacobetz, M.A.; Lolkema, M.P.; Buchholz, M.; Olive, K.P.; Gress, T.M.; et al. Stromal biology and therapy in pancreatic cancer. Gut 2011, 60, 861–868.
  22. Lonardo, E.; Frias-Aldeguer, J.; Hermann, P.C.; Heeschen, C. Pancreatic stellate cells form a niche for cancer stem cells and promote their self-renewal and invasiveness. Cell Cycle 2012, 11, 1282–1290.
  23. Wörmann, S.M.; Diakopoulos, K.N.; Lesina, M.; Algül, H. The immune network in pancreatic cancer development and progression. Oncogene 2014, 33, 2956–2967.
  24. Shi, C.; Washington, M.K.; Chaturvedi, R.; Drosos, Y.; Revetta, F.L.; Weaver, C.J.; Buzhardt, E.; Yull, F.E.; Blackwell, T.S.; Sosa-Pineda, B.; et al. Fibrogenesis in pancreatic cancer is a dynamic process regulated by macrophage–stellate cell interaction. Lab. Investig. 2014, 94, 409–421.
  25. 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.
  26. Tang, D.; Gao, J.; Wang, S.; Yuan, Z.; Ye, N.; Chong, Y.; Xu, C.; Jiang, X.; Li, B.; Yin, W.; et al. Apoptosis and anergy of T cell induced by pancreatic stellate cells-derived galectin-1 in pancreatic cancer. Tumor Biol. 2015, 36, 5617–5626.
  27. Minici, C.; Rigamonti, E.; Lanzillotta, M.; Monno, A.; Rovati, L.; Maehara, T.; Kaneko, N.; Deshpande, V.; Protti, M.P.; De Monte, L.; et al. B lymphocytes contribute to stromal reaction in pancreatic ductal adenocarcinoma. Oncoimmunology 2020, 9, 1794359.
  28. Yuen, G.J.; Demissie, E.; Pillai, S. B lymphocytes and cancer: A love–hate relationship. Trends Cancer 2016, 2, 747–757.
  29. Le, D.T.; Jaffee, E.M. Next-generation cancer vaccine approaches: Integrating lessons learned from current successes with promising biotechnologic advances. J. Natl. Compr. Cancer Network 2013, 11, 766–772.
  30. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264.
  31. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, 542–551.
  32. Zhang, J.; Wolfgang, C.L.; Zheng, L. Precision Immuno-Oncology: Prospects of Individualized Immunotherapy for Pancreatic Cancer. Cancers 2018, 10, 39.
  33. Iwai, Y.; Ishida, M.; Tanaka, Y.; Okazaki, T.; Honjo, T.; Minato, N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297.
  34. AstraZeneca. Study of Tremelimumab in Patients with Advanced Solid Tumors. 2015. Available online: https://clinicaltrials.gov/show/NCT02527434 (accessed on 20 December 2020).
  35. Blank, C.U.; Enk, A. Therapeutic use of anti-CTLA-4 antibodies. Int. Immunol. 2015, 27, 3–10.
  36. O’Reilly, E.M.; Oh, D.Y.; Dhani, N.; Renouf, D.J.; Lee, M.A.; Sun, W.; Fisher, G.; Hezel, A.; Chang, S.C.; Vlahovic, G.; et al. Durvalumab With or Without Tremelimumab for Patients With Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019, 5, 1431–1438.
  37. Cong, J.; Wang, Y.; Zhang, X.; Zhang, N.; Liu, L.; Soukup, K.; Michelakos, T.; Hong, T.; DeLeo, A.; Cai, L.; et al. A novel chemoradiation targeting stem and nonstem pancreatic cancer cells by repurposing disulfiram. Cancer Lett. 2017, 409, 9–19.
  38. Kamath, S.D.; Kalyan, A.; Kircher, S.; Nimeiri, H.; Fought, A.J.; Benson, A.; Mulcahy, M. Ipilimumab and Gemcitabine for Advanced Pancreatic Cancer: A Phase Ib Study. Oncologist 2020, 25, 808–815.
  39. Weiss, G.J.; Blaydorn, L.; Beck, J.; Bornemann-Kolatzki, K.; Urnovitz, H.; Schutz, E.; Khemka, V. Correction to: Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Investig. New Drugs 2019, 37, 797.
  40. Schizas, D.; Charalampakis, N.; Kole, C.; Economopoulou, P.; Koustas, E.; Gkotsis, E.; Ziogas, D.; Psyrri, A.; Karamouzis, M.V. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat. Rev. 2020, 86, 102016.
  41. Thomas, A.M.; Santarsiero, L.M.; Lutz, E.R.; Armstrong, T.D.; Chen, Y.C.; Huang, L.Q.; Laheru, D.A.; Goggins, M.; Hruban, R.H.; Jaffee, E.M. Mesothelin-specific CD8(+) T cell responses provide evidence of in vivo cross-priming by antigen-presenting cells in vaccinated pancreatic cancer patients. J. Exp. Med. 2004, 200, 297–306.
  42. Le, D.T.; Picozzi, V.J.; Ko, A.H.; Wainberg, Z.A.; Kindler, H.; Wang-Gillam, A.; Oberstein, P.; Morse, M.A.; Zeh, H.J.; Weekes, C.; et al. Results from a Phase IIb, Randomized, Multicenter Study of GVAX Pancreas and CRS-207 Compared with Chemotherapy in Adults with Previously Treated Metastatic Pancreatic Adenocarcinoma (ECLIPSE Study). Clin. Cancer Res. 2019, 25, 5493–5502.
  43. Le, D.T.; Lutz, E.; Uram, J.N.; Sugar, E.A.; Onners, B.; Solt, S.; Zheng, L.; Diaz, L.A.; Donehower, R.C.; Jaffee, E.M.; 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, 382–389.
  44. Thind, K.; Padrnos, L.J.; Ramanathan, R.K.; Borad, M.J. Immunotherapy in pancreatic cancer treatment: A new frontier. Ther. Adv. Gastroenterol. 2017, 10, 168–194.
  45. Beatty, G.L.; Torigian, D.A.; Chiorean, E.G.; Saboury, B.; Brothers, A.; Alavi, A.; Troxel, A.B.; Sun, W.; Teitelbaum, U.R.; Vonderheide, R.H.; et al. A phase I study of an agonist CD40 monoclonal antibody (CP- 870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2013, 19, 6286–6295.
  46. Nywening, T.M.; Wang-Gillam, A.; Sanford, D.E.; Belt, B.A.; Panni, R.Z.; Cusworth, B.M.; Toriola, A.T.; Nieman, R.K.; Worley, L.A.; Yano, M.; et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: A single-centre, open-label, dose-finding, nonrandomised, phase 1b trial. Lancet Oncol. 2016, 17, 651–662.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 490
Revisions: 2 times (View History)
Update Date: 25 Feb 2021
1000/1000
Video Production Service