Immunological Disorders and Immunotherapy

Immunological Disorders and Immunotherapy
Open Access

ISSN: 2593-8509

+44-77-2385-9429

Mini Review - (2016) Volume 1, Issue 2

Immunotherapy in Pancreatic Cancer; the Road Less Traveled

Galam Leem and Si Young Song*
Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine and Yonsei Institute of Gastroenterology, Seoul, Korea, E-mail: sysong@yuhs.ac
*Corresponding Author: Si Young Song, Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine and Yonsei Institute of Gastroenterology, Seoul, Korea, Tel: +82-2-2228-1957, Fax: +82-2-2227-7900 Email:

Abstract

Pancreatic cancer shows extremely low responses to chemo- and radiation therapies due to its inherent genetic instability, immunosuppressive microenvironment, and the complex peritumoral stroma. They make the pancreatic cancer most lethal malignancy with 5 year survival of less than 6%, so the need of new therapeutic approaches is urgently demanded. Immunotherapies have shown promises in other multiple cancers by augmenting anti-tumor immunity, so it stood out for the alternative care of pancreatic cancer. However, up to now, immunotherapeutic agents lack efficacy in pancreatic cancer. In this review, we give an overview of immune-related therapeutic strategies, clinical trials in pancreatic cancer and current obstacles that we face. We discuss the ways to overcome those current obstacles here, and by getting over them, we hope to get over the poor prognosis of pancreatic cancer.

<

Keywords: Immunosuppressive microenvironment, Malignancy, Peritumoral stroma, Pancreatic cancer

Introduction

Pancreatic cancer is the most lethal malignancy with a 5 year survival of less than 6% [1]. In contrast to the increment in other cancer survival, the advance in pancreatic cancer is very slow and despite surgery, locoregional therapy, chemotherapy and radiotherapy, the overall median survival is still less than 1 year from diagnosis. Surgical resection is the only curative treatment, but even after surgery, 5 year survival does not reach to 20% [2]. The reason for low objective response rate to conventional standard therapy is because of the inherent genetic instability of pancreatic cancer cells, immunosuppressive microenvironment at the tumor site, and the complex peritumoral stroma [3,4]. As probe or inhibitor of histone methyltransferase G9a is able to induce cell senescence or death in pancreatic carcinoma, more attention has been drawn to disruption the function of G9a in the treatment of pancreatic adenocarcinoma [5,6]. However, as apart from being as a histone modifier, G9a is also essential for the maintenance of global and loci specific DNA methylation, inhibition of G9a may cause even more severe genetic instability of pancreatic cancer [7]. Therefore, new strategies, such as immunotherapeutic methods, need to be studied in order to eradicate pancreatic carcinoma. Immunotherapy started since the end of the 19th century first by European physician, William Coley, with the idea of recruiting and activating the host’s T cells to recognize and attack tumor-specific antigens, by themselves [8,9]. In pancreatic cancer, from animal models, it is known that pre-invasive pancreatic lesions contain high density of immune suppressor cells, but nearly absent immune effector cells, suggesting that antitumor immunity including cytotoxic and adaptive immunity may be already defective [10]. Therefore, reactivating the antitumor immunity in pancreatic cancer has been highlighted as a new therapeutic option for the last several decades. Immunotherapies in pancreatic cancer diverse into 4 main categories; checkpoint inhibitors/immune modulators, therapeutic vaccines, adoptive T cell transfer, and monoclonal antibodies.

Checkpoint Inhibitors/Immune Modulators

When the cancer cells or cancer cells expressing proteins are recognized as antigens, tumor specific T cells are activated. Those cells are controlled by inhibitory and stimulatory signals, called immune checkpoints. The blockade of inhibitory immune checkpoints would induce T cell proliferation and prolong the life of activated T cells, consequently enhances antitumor immune responses [11]. Cytotoxic T-lymphocyte associated protein-4 (CTLA-4) and programmed cell death protein-1 (PD1), CD28 inhibitors are major negative costimulator expressed on activated T cells [12], and have become the first clinical landscape in cancer immunotherapy. Despite their success in certain malignancies, in pancreatic cancer, past phase I and II clinical trials with ipilimumab (CTLA-4 inhibitor), pidilizumab (anti- PD1 mAb), CP-870893 (monoclonal antibody of CD40 receptor), and BMS-936559 (PDL1 antibody) did not prove any effectiveness [13-15]. Due to enhancement of general immunity and temporal immunosuppression, nearly 10% of patients were undergone G3-4 adverse effects, called ‘immune-related adverse events (irAEs), including dermatologic, gastrointestinal, hepatic, endocrine, and other less common inflammatory events [16,17]. To overcome those irAEs, dose adjustment and temporal use of systemic corticosteroids are recommended, depends on the severity of events.

Therapeutic Vaccines

Autologous pancreatic cancer cells express several specific antigens, including Wilms’ tumor gene 1 (WT1) (75% of patients), mucin 1 (MUC1) (over 85%), human telomerase reverse transcriptase (hTERT) (88%), mutated K-RAS (73%), and carcinoembryonic antigen (CEA) (over 90%) [18-24]. With the idea of administering these tumor antigens to stimulate the patients’ own immune system to recognize them, tumor vaccinations have been tested. There are two major categories of therapeutic vaccines; whole-cell vaccines and antigenspecific vaccines. Whole-cell vaccines are made by transfecting human tumor cell lines with or without granulocyte-macrophage colonystimulating factors (GM-CSF); GVAX (GM-CSF) and Algenpantucel-L (a-1,3 galactosyl transferase) [25-28]. Despite their promises in postsurgical patients, for metastatic pancreatic cancer patients, they did not show any benefits. Antigen-specific vaccines, peptide vaccines, try to target antigens and other factors expressed by tumor cells; GV1001 (hTERT), Tacemotide (MUC-1), Kras (K-RAS), WT1 (WT1), and KIF-20A (HLA-A24) [29-34]. This approach has not delivered any improvement in overall survival or disease free survival. Nonspecific vaccines like IMM-101 (contains a wide variety of pathogen-associated molecular patterns (PAMPs)), dendritic cells (DC) also tried, but no single agent of vaccines proved its efficacy [35,36].

Adoptive T Cell Therapy

T cells are removed from a patient and genetically engineered to recognize tumor specific proteins, and then re-introduced to the patient aiming to enhance antitumor responses. Chimeric antigen receptors (CARs) are transmembrane proteins comprising an antibody-derived single-chain variable fragment specific for a tumor antigen fused to a hinge region, a spacer, a membrane spanning element and signaling domain [37]. CAR therapy has been an innovative strategy to redirect T cells against tumors and shows promises in leukemia targeting CD 19 antigen. Up to now, due to less identified potential antigens and its expensive and time-consuming process, the progress in pancreatic cancer is very slow [38]. However, if those problems would be overcome, engineered T cells in CAR therapy would seem to prove its clear ability to kill tumor cells significantly.

Monoclonal Antibodies

Monoclonal antibodies (mAbs) bind to targeted antigens, same editopes, expressed on tumor cells and play an inhibitory role by preventing formation of antigen-ligand complex [39]. MUC-1, overexpressed in 90% of pancreatic cancer, has been the first target of mAb in pancreatic cancer; PAM4, clivatuzumab. Starting from MUC-1 mAb, endothelial growth factor receptor (EGFR), EGFR2 (HER2), vascular endothelial growth factor (VEGFR), CD40, MUC5a, etc. have been targeted of mAbs, and started clinical trials; no significant improvement in phase III clinical trials was shown, yet [40-46].(Table 1).

Therapeutic category Investigator Phase Target Agent Patients/N Outcomes
Checkpoint inhibitors Royal et al. [13] II CTLA-4 Ipilimumab Advanced/27 PR: 3.7%
Anjali et al. [14] Ib CTLA-4 Ipilimumab Advanced/ 13 PR: 15%/
SD: 38%
Brahmer et al. [15] I PDL-1 Nivolumab Advanced/14 No objective responses
Therapeutic Vaccines
Whole cell vaccines Jaffee et al. [47] I GM-CSF vaccine GVAX with chemotherapy Resected/ 14 DFS greater than 10 years
Laheru et al. [48] II GM-CSF vaccine GVAX with cyclophosphamide (Cy) Advanced/ 50 OS benefit with Cy: 2.4 months
Lutz et al. [28] II GM-CSF vaccine GVAX with chemoradiotherapy (5-FU) Resected/ 60 DFS: 17.3 months
OS: 24.8 months
Hardacre et al. [25] II Algenpantucel-L Algenpantucel-L with chemotherapy Resected/ 70 DFS: 62%
OS: 86%
Lutz et al. [49] Pilot GM-CSF vaccine GVAX with cyclophosphamide (Cy) Resected/ 54 Vaccine-induced intratumoral tertiary lymphoid aggregation was observed.
Upregulation of PD-1 and PD-L1 in the tumor was observed after vaccination.
Antigen-specific vaccines Abou-Alfa et al. [29] I K-ras Kras Resected/ 24 DFS: 8.6 months
OS: 20.3 months
Bernhardt et al. [30] I/II hTERT GV1001 Advanced/ 48 OS: 8.6 months
Middleton et al. [31] III hTERT GV1001 with sequential chemotherapy
GV1001 with concurrent chemotherapy
Chemotherapy only
Advanced/ 1052 PFS: 4.5%, OS: 6.9%
PFS: 6.6%, OS: 8.4%
PFS: 6.4%, OS 7.9%
Ramanathan et al. [50] I MUC-1 MUC-1 Advanced/ 16 OS: 12.0 months
Mayanagi et al. [32] I WT1 DC Advanced/ 32 PFS: 4.2 months
OS: 8.1 months
Asahara et al. [34] I/II HLA-A24 KIF20A Advanced/ 31 PFS: 2.0 months
OS: 4.7 months
Adoptive T cell therapy Beatty et al. [38] I Mesothelin CARTmeso Advanced/ 2 Acceptable safety in both IV and intra-tumoral injection
Monoclonal antibodies Gulec et al. [40] I MUC-1 PAM4 Advanced/ 20 PR: 15%, SD: 20%
Ocean et al. [41] I MUC-1 PAM4 with gemcitabine Advanced/ 38 PR: 15%, SD: 27%
Philip et al. [42] III EGFR Cetuximab with gemcitabine
Gemcitabine
Advanced/ 745 PR: 8%, SD: 37%
PR: 7%, SD: 30%
Assenat et al. [43] I/II EGFR/HER2 Cetuximab/Trastuzumab Advanced, Previously treated/ 38 PFS: 1.8 months
OS: 4.6 months
Kindler et al. (2010) [44] III VEGFR Bevacizumab with gemcitabine
Gemcitabine
Advanced/ 535 CR/PR: 13%, SD: 42%, PFS: 3.8 months, OS: 5.8 months
CR/PR: 10%, SD: 34%, PFS: 2.9 months, OS: 5.9 months
Beatty et al. (2013) [45] I CD40 CP 807,893 with gemcitabine Advanced/ 21 PR: 19%, SD: 52%
PFS: 5.6 months, OS: 7.4 months
PR; partial response, SD; stable disease, CR; complete response, PFS; progression free survival, DFS; disease free survival, OS; overall survival

Table 1: Summary of main clinical trials of immunotherapy in pancreatic cancers.

Challenges in Immunotherapy

Up to now, any single agent of immunotherapies in pancreatic cancer did not prove its efficacy significantly. There are some reasons for that, but mainly it seems to be thought that pancreatic cancer is such a heterogeneous disease, requiring polyvalent approaches [51]. It might be not sufficient to remove the inhibitory immune regulators or to active innate and cytotoxic immunity, alone. In order to bypass the immunosuppressive environments and boost immune responses in pancreatic cancer, an orchestrated approach would be required. Many preclinical studies support evidences for combination of vaccines with immune checkpoint inhibitors, and clinical trials begin to show promises in the treatment of pancreatic cancer; especially clinical trials for the combination of PD-1 inhibitor and GVAX do [52,53]. To make further steps possible, we still face two simple questions; 1) whom to treat, and 2) how to deliver drugs through microenvironment.

Whom to treat

Recent data support that overall response rate to immunotherapy is around 20% [54-60]. That emphasizes the importance to find the right person for immunotherapy who would show good response. There could be two possible approaches; select patients with biomarkers or with cancer status. Eventually, immunotherapy should extend the indications to all stages of pancreatic cancer, but it would be more promising to start with patients of possible minimal residual disease after resection to prevent recurrence with minimal toxicities. Past studies were focused on finding serum biomarkers, but yet, no biomarkers show any benefit. For the GV1001, telomerase peptide vaccine, high eotaxin levels at baseline were once reported to correlate with a longer overall survival, but following studies did not find any correlation between eotaxin levels and overall survival. This distress in finding serum biomarkers may be due to complex peritumoral stroma of pancreatic cancer which separates tumor burden from systemic circulation. If we can stabilize tumor cells obtained by endoscopic or percutaneous biopsy from patients, and analyze the specific antigens for each patient, we can specify the immunotherapy that compatible to each patient; ‘half-individualized immunotherapy’.

How to deliver

Like other failures of chemotherapy, immunotherapy also has the same issue; how to deliver immune regulating drugs or activated T lymphocytes to the cancer. There would be three available approaches to improve; 1) combination with chemotherapy, 2) combination with radiotherapy, and 3) in situ immunization. It had been assumed that chemotherapy could not be used in combination with immunotherapy because of its immunosuppressive activities for a long time. However, in the aspects of reducing immune suppressive cells, increasing antigen presenting cells by cytotoxic activities, and alternative mechanisms of immunogenicity, chemotherapy is now recognized as providing additional benefit to immunotherapy. Many preclinical studies support that theme [61-68], and clinical trials begin to show positive results [69-77]. Radiotherapy also was known to have potential to be a good combination therapy with immunotherapy. As we can imagine easily from its ‘abscopal effect’, radiation can convert the irradiat`ed tumor into an ‘immunogenic hub’ [78]. Once, tumor have damaged with radiation exposure, cancer cells dye of apoptosis and necrosis, and those dead cancer cells promote uptake by dendritic cells, presentation of tumor-derived antigens to T cells, activation of cytotoxic T cells, exposure of specific proteins on tumor cell surface, and release of ATP and high mobility group protein B1 (HMGB1) [79]. Thus, radiation seems to play an important role in modifying systemic immune system into more suitable conditions for immunotherapy. Concepts of ‘in situ immunization’ were started from the idea of how to invade cancer cells directly. This use of immunotherapy show significant antitumor effect in bladder cancer, melanoma, and head and neck cancer [80-82]. For the pancreatic cancer, it is not that difficult to inject drugs into cancer cells with techniques of endoscopic ultrasonography. In situ immunization can provide promises to immunotherapy in pancreatic cancer.

Conclusion

In spite of advances in anticancer treatment, pancreatic cancer remains a lethal malignancy. Surgery, locoregional therapies, chemotherapy and radiotherapy could not prove the survival more than 1 year. Regarding to its immune surveillance characteristics, immunotherapy has potentials to treat pancreatic cancer more efficiently. Since 19th century, many advances and clinical trials have done in the field of immunotherapy, but in pancreatic cancer, it is still evolving and unexplored landscape. So far, no significant efficacy was proven in clinical trials of pancreatic cancer and people begin to face obstacles; whom to treat and how to deliver. If we can get the tumor tissue from patients efficiently, it might be possible to choose individualized appropriate target antigens. And combination with chemotherapy or radiotherapy, or in situ immunization can improve the efficacy of immunotherapies. There are far ways to go, and many obstacles to overcome, but as we overcome one obstacle, one patient can live one month longer.

References

  1. Siegel RL, Miller KD, Jemal A (2015) Cancer statistics. CA Cancer J Clin 65: 5-29.
  2. Seufferlein T, Bachet JB, van Cutsem E, Rougier P (2012) Pancreatic adenocarcinoma: ESMO-ESDO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 23 Suppl 7: vii33-40.
  3. Rucki AA, Zheng L (2014) Pancreatic cancer stroma: understanding biology leads to new therapeutic strategies. World J Gastroenterol 20: 2237-2246.
  4. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, et al. (2012) Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366: 883-892.
  5. Yuan Y, Wang Q, Paulk J, Kubicek S, Kemp MM, et al. (2012) A small-molecule probe of the histone methyltransferase G9a induces cellular senescence in pancreatic adenocarcinoma. ACS Chem Biol 7: 1152-1157.
  6. Yuan Y, Tang AJ, Castoreno AB, Kuo SY, Wang Q, et al. (2013) Gossypol and an HMT G9a inhibitor act in synergy to induce cell death in pancreatic cancer cells. Cell Death Dis 4: e690.
  7. Zhang T, Termanis A, Özkan B, Bao XX, Culley J, et al. (2016) G9a/GLP Complex Maintains Imprinted DNA Methylation in Embryonic Stem Cells. Cell Rep 15: 77-85.
  8. Coley WB (1893) Hawkins on Tubercular Peritonitis. Ann Surg 17: 462-464.
  9. Laheru D, Jaffee EM (2005) Immunotherapy for pancreatic cancer - science driving clinical progress. Nat Rev Cancer 5: 459-467.
  10. Clark CE, Beatty GL, Vonderheide RH (2009) Immunosurveillance of pancreatic adenocarcinoma: insights from genetically engineered mouse models of cancer. Cancer Lett 279: 1-7.
  11. Rosenberg SA, Yang JC, Restifo NP (2004) Cancer immunotherapy: moving beyond current vaccines. Nat Med 10: 909–915.
  12. Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12: 252-264.
  13. Royal RE, Levy C, Turner K, Mathur A, Hughes M, et al. (2010) Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 33: 828-833.
  14. Anjali N, Sheetal M, Kircher M, Nimeiri HS, Benson AB, et al. (2015) Results of the phase Ib study of ipilimumab and gemcitabine for advanced pancreas cancer. J Clin Oncol 33.
  15. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366: 2455-2465.
  16. Naidoo J, Page DB, Li BT, Connell LC, Schindler K, et al. (2015) Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol 26: 2375.
  17. Champiat S, Lambotte O, Barreau E, Belkhir R, Berdelou A, et al. (2016) Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol 27: 559-574.
  18. Dodson LF, Hawkins WG, Goedegebuure P (2011) Potential targets for pancreatic cancer immunotherapeutics. Immunotherapy 3: 517-537.
  19. Wenandy L, Sorensen RB, Svane IM, Thor Straten P, Andersen MH (2008) RhoC a new target for therapeutic vaccination against metastatic cancer. Cancer Immunol Immunother 57: 1871-1878.
  20. Oji Y, Nakamori S, Fujikawa M, Nakatsuka S, Yokota A, et al. (2004) Overexpression of the Wilms' tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci 95: 583-587.
  21. Ueda M, Miura Y, Kunihiro O, Ishikawa T, Ichikawa Y, (2005) MUC1 overexpression is the most reliable marker of invasive carcinoma in intraductal papillary-mucinous tumor (IPMT). Hepato-gastroenterology 52: 398-403.
  22. Seki K, Suda T, Aoyagi Y, Sugawara S, Natsui M, et al. (2001) Diagnosis of pancreatic adenocarcinoma by detection of human telomerase reverse transcriptase messenger RNA in pancreatic juice with sample qualification. Clin Cancer Res 7: 1976-1981.
  23. Gjertsen MK, Bakka A, Breivik J, Saeterdal I, Solheim BG, (1995) Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet 346: 1399-1400.
  24. Yamaguchi K, Enjoji M, Tsuneyoshi M (1991) Pancreatoduodenal carcinoma: a clinicopathologic study of 304 patients and immunohistochemical observation for CEA and CA19-9. J Surg Oncol 47: 148-154.
  25. Hardacre JM, Mulcahy M, Small W, Talamonti M, Obel J, et al. (2013) Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: a phase 2 study. J Gastrointest Surg 17: 94-100.
  26. Le DT, Lutz E, Uram JN, Sugar EA, Onners B, et al. (2013) Evaluation of ipilimumab in combination with allogenic pancreatic tumor cells transfected with GM-CSF gene in previously treated pancreatic cancer. Immunotherapy 36: 382–389.
  27. Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, (2015) Safety and survival with GVAX pancreas prime and Listeria Monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 33: 132–153.
  28. Lutz E, Yeo CJ, Lillemoe KD, Biedrzycki B, Kobrin B, et al. (2011) A lethally irradiated allogenic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma: a phase II trial of safety, efficacy, and immune activation. Ann Surg 253: 328–335.
  29. Abou-Alfa GK, Chapman PB, Feilchenfeldt J, Brennan MF, Capanu M, et al. (2011) Targeting mutated K-ras in pancreatic adenocarcinoma using an adjuvant vaccine. Am J Clin Oncol 34: 321–325.
  30. Bernhardt SL, Gjertsen MK, Trachsel S, Møller M, Eriksen JA, et al. (2006) Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: A dose escalating phase I/II study. Br J Cancer 95: 1474-1482.
  31. Middleton G, Silcocks P, Cox T, Valle J, Wadsley J, et al. (2014) Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): an open-label, randomised, phase 3 trial. Lancet Oncol 15: 829-840.
  32. Mayanagi S, Kitago M, Sakurai T, Matsuda T, Fujita T, et al. (2015) Phase I pilot study of Wilms tumor gene 1 peptide-pulsed dendritic cell vaccination combined with gemcitabine in pancreatic cancer. Cancer Sci 106: 397-406.
  33. Rong Y, Qin X, Jin D, Lou W, Wu L, et al. (2012) A phase I pilot trial of MUC1-peptide-pulsed dendritic cells in the treatment of advanced pancreatic cancer. Clin Exp Med 12: 173-180.
  34. Asahara S, Takeda K, Yamao K, Maguchi H, Yamaue H (2013) Phase I/II clinical trial using HLA-A24-restricted peptide vaccine derived from KIF20A for patients with advanced pancreatic cancer. J Transl Med 11: 291.
  35. Dalgleish AG (2015) The IMAGE I Trial Investigators. A multicenter randomized, open-label, proof of concept, phase II trial comparing gemcitabine with and without IMM-101 in advanced pancreatic cancer. J Clin Oncol 33.
  36. Kimura Y, Tsukada J, Tomoda T, Takahashi H, Imai K, et al. (2012) Clinical and immunologic evaluation of dendritic cell-based immunotherapy in combination with gemcitabine and/or S-1 in patients with advanced pancreatic carcinoma. Pancreas 41: 195-205.
  37. Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, et al. (2009) Control of large, established tumor xenografts with genetically retarted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci USA 106: 3360-3365.
  38. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, et al. (2014) Mesothelin-specific chimeric antigen receptor mRNA engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res 2: 112-120.
  39. Jiang XR, Song A, Bergelson S, Arroll T, Parekh B, et al. (2011) Advances in the assessment and control of the effector functions of therapeutic antibodies. Nat Rev Drug Discov 10: 101-111.
  40. Gulec SA, Cohen SJ, Pennington KL, Zuckier LS, Hauke RJ, et al. (2011) Treatment of advanced pancreatic carcinoma with 90Y-Clivatuzumab Tetraxetan: a phase I single-dose escalation trial. Clin Cancer Res 17: 4091-4100.
  41. Ocean AJ, Pennington KL, Guarino MJ, Sheikh A, Bekaii-Saab T, et al. (2012) Fractionated radioimmunotherapy with (90) Y-clivatuzumab tetraxetan and low-dose gemcitabine is active in advanced pancreatic cancer: a phase 1 trial. Cancer 118: 5487–5506.
  42. Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, et al. (2010) Phase III study in comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: southwest oncology group-directed intergroup trial S0205. J Clin Oncol 28: 3605-3610.
  43. Assenat E, Azria D, Mollevi C, Guimbaud R, Tubiana-Mathieu N, et al. (2015) Dual targeting of HER1/EGFR and HER2 with cetuximab and trastuzumab in patients with metastatic pancreatic cancer after gemcitabine failure: results of the “THERAPY” phase 1-2 trial. Oncotarget 20: 12796–12808.
  44. Kindler HL, Niedzwiecki D, Hollis D, Sutherland S, Schrag D, et al. (2010) Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol 28: 3617–3622.
  45. Beatty GL, Torigian DA, Chiorean EG, Saboury B, Brothers A, et al. (2013) 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 19: 6286-6295.
  46. Morse M, Diaz LA, Azad NS, Laheru D, Haley S, et al. (2012) A phase I/II safety study of NPC-1C: a novel, therapeutic antibody to treat pancreas and colorectal cancers. J Clin Oncol 30.
  47. Jaffee EM, Hruban RH, Biedrzycki B, Laheru D, Schepers K, et al. (2001) Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J Clin Oncol 19: 145-156.
  48. Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, et al. (2008) Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res 14: 1455-1463.
  49. Ramanathan RK, Lee KM, McKolanis J, Hiltbold E, Schraut W, et al. (2005) Phase I study of a MUC1 vaccine composed of different doses of MUC1 with SB-AS2 adjuvant in resected locally advanced pancreatic cancer. Cancer Immunol Immunother 54: 254-264.
  50. Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, et al. (2012) Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491: 399-405.
  51. Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G (2014) Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation, Cancer Immunol. Res 2: 616-631.
  52. Le DT, Lutz E, Uram JN, Sugar EA, Onners B (2013) Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J Immunother 36: 382-389.
  53. Soares KC, Rucki AA, Wu AA, Olino K, Xiao Q, et al. (2015) PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J Immunother 38: 1-11.
  54. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, et al. (2015) Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 373: 123-135.
  55. Paz-Ares L, Horn L, Borghaei H, Spigel DR, Steins M (2015) Phase III, randomized trial (CheckMate 057) of nivolumab (NIVO) versus docetaxel (DOC) in advanced non-squamous cell (non-SQ) non-small cell lung cancer (NSCLC). J Clin Oncol 33.
  56. Muro K, Bang YJ, Shankaran V, Geva R, Catenacci DVT (2015) Relationship between PD-L1 expression and clinical outcomes in patients (pts) with advanced gastric cancer treated with the anti-PD-1 monoclonal antibody pembrolizumab (Pembro; MK-3475) in KEYNOTE-012. J Clin Oncol 33.
  57. Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, et al. (2015) Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. J Clin Oncol 33: 1430-1437.
  58. McDermott DF, Drake CG, Sznol M, Choueiri TK, Powderly JD (2015) Survival, durable response, and long-term safety in patients with previously treated advanced renal cell carcinoma receiving nivolumab. J Clin Oncol 33: 2013–2020
  59. Seiwert TY, Haddad RI, Gupta S, Mehra R, Tahara M (2015) Antitumor activity and safety of pembrolizumab in patients (pts) with advanced squamous cell carcinoma of the head and neck (SCCHN): preliminary results from KEYNOTE-012 expansion cohort. J Clin Oncol 33.
  60. El-Khoueiry AB, Melero I, Crocenzi TS, Welling TH, Cheung TY (2015) Phase I/II safety and antitumor activity of nivolumab in patients with advanced heaptocellular carcinoma (HCC): CA209-040. J Clin Oncol 33.
  61. Zitvogel L, Apetoh L, Ghiringhelli F, André F, Tesniere A, et al. (2008) The anticancer immune response: indispensable for therapeutic success? J Clin Invest 118: 1991-2001.
  62. Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, et al. (2005) Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med 202: 1691-1701.
  63. Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, et al. (2014) Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 3: e955691.
  64. Kepp O, Menger L, Vacchelli E, Locher C, Adjemian S, et al. (2013) Crosstalk between ER stress and immunogenic cell death. Cytokine Growth Factor Rev 24: 311-318.
  65. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, et al. (2007) Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 13: 54-61.
  66. van der Sluis TC, van Duikeren S, Huppelschoten S, Jordanova ES, Beyranvand Nejad E, et al. (2015) Vaccine-induced tumor necrosis factor-producing T cells synergize with cisplatin to promote tumor cell death. Clin Cancer Res 21: 781-794.
  67. Ma Y, Adjemian S, Mattarollo SR, Yamazaki T, Aymeric L, et al. (2013) Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity 38: 729-741.
  68. Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, et al. (2014) Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ 21: 79-91.
  69. Garcia-Martinez E, Gil GL, Benito AC, Gonzalez-Billalabeitia E, Conesa MA, et al. (2014) Tumor-infiltrating immune cell profiles and their change after neoadjuvant chemotherapy predict response and prognosis of breast cancer. Breast Cancer Res 16:488.
  70. Denkert C, von Minckwitz G, Brase JC, Sinn BV2, Gade S, et al. (2015) Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol 33: 983-991.
  71. Passero FC Jr, Saif MW (2015) Advancements in the Management of Pancreatic Cancer: 2015 ASCO Gastrointestinal Cancers Symposium (San Francisco, CA, USA. January 15-17, 2015). JOP 16: 99-103.
  72. Antonia SJ, Brahmer JR, Gettinger S, Chow LQ, Juergens R, et al. (2014) Nivolumab (Anti-PD-1; BMS-936558, ONO-4538) in combination with platinum-based doublet chemotherapy (PT-DC) in advanced non-small cell lung cancer (NSCLC). Int J Radiat Oncol Biol Phys 90: S2.
  73. Rizvi NA, Shepherd FA, Antonia SJ, Brahmer JR, Chow LQ, et al. (2014) First-line monotherapy with nivolumab (Anti-PD-1; BMS- 936558, ONO-4538) in advanced non-small cell lung cancer (NSCLC): safety, efficacy, and correlation of outcomes with PD-L1 status. Int J Radiat Oncol Biol Phys 90: S31.
  74. Reck M, Bondarenko I, Luft A, Serwatowski P, Barlesi F, et al. (2013) Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol 24: 75-83.
  75. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, et al. (2014) Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563-567.
  76. Lesterhuis WJ, Punt CJ, Hato SV, Eleveld-Trancikova D, Jansen BJ, et al. (2011) Platinum-based drugs disrupt STAT6- mediated suppression of immune responses against cancer in humans and mice. J Clin Invest 121: 3100-3108.
  77. Zhang P, Su DM, Liang M, Fu J (2008) Chemopreventive agents induce programmed death-1-ligand 1 (PD-L1) surface expression in breast cancer cells and promote PD-L1-mediated T cell apoptosis. Mol Immunol 45: 1470-1476.
  78. Bindea G, Mlecnik B, Angell HK, Galon J (2014) The immune landscape of human tumors: Implications for cancer immunotherapy. Oncoimmunology 3: e27456.
  79. Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, et al. (2014) The role of chemoattractant receptors in shaping the tumor microenvironment. Biomed Res Int 2014: 751392.
  80. Cohen MH, Jessup JM, Felix EL, Weese JL, Herberman RB (1978) Intralesional treatment of recurrent metastatic cutaneous malignant melanoma: a randomized prospective study of intralesional Bacillus Calmette-Guerin versus intralesional dinitrochlorobenzene. Cancer 41: 2456-2463.
  81. Morton DL, Eilber FR, Holmes EC, Hunt JS, Ketcham AS, et al. (1974) BCG immunotherapy of malignant melanoma: summary of a seven-year experience. Ann Surg 180: 635-643.
  82. Bier J, Kleinschuster S, Bier H, Rapp H (1982) Intratumor immunotherapy with BCG cell wall preparations: development of a new therapy approach for head-neck tumors. Arch Otorhinolaryngol 236: 245-255.
Citation: Leem G, Song SY (2016) Immunotherapy in Pancreatic Cancer; the Road Less Traveled. Immunol Disord Immunother 1: 106.

Copyright: © 2016 Leem G, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top