IJGII Inernational Journal of Gastrointestinal Intervention

pISSN 2636-0004 eISSN 2636-0012
ESCI
scopus

Article

home All Articles View

Review Article

Gastrointestinal Intervention 2014; 3(1): 24-26

Published online June 30, 2014 https://doi.org/10.1016/j.gii.2014.03.002

Copyright © International Journal of Gastrointestinal Intervention.

Endoscopic ultrasound-guided immunotherapy

Atsushi Irisawa1,*, Goro Shibukawa1, Tadayuki Takagi2, Yoko Abe1, Akiko Saito1, Koh Imbe1, Koki Hoshi1, Akane Yamabe1, and Ryo Igarashi1

1Department of Gastroenterology, Fukushima Medical University, Aizu Medical Center, Kawahigashi, Aizuwakamatsu, Japan, 2Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Kawahigashi, Aizuwakamatsu, Japan

Correspondence to:*Corresponding author. Department of Gastroenterology, Fukushima Medical University, Aizu Medical Center, 21-2, Maeda, Tanisawa, Kawahigashi, Aizuwakamatsu 969-3492, Japan., E-mail address:irisawa@fmu.ac.jp (A. Irisawa).

Received: February 11, 2014; Accepted: March 27, 2014

Anti-tumoral endoscopic ultrasound-guided fine-needle injection (EUS-FNI), with its minimally invasive access for anti-tumoral agent delivery, is the most exciting field of intervention EUS. Pancreatic cancer is regarded as a systemic disease even if imaging modalities reveal no visible metastasis. From that perspective, immunological therapy is performed. To date, several reports have described immunotherapy under EUS-guidance. The first report of EUS-FNI intended for immunotherapy for advanced pancreatic cancer was published in 2000. In that study, an allogeneic mixed-lymphocyte culture was injected into tumors of eight patients with unresectable local pancreatic adenocarcinoma. The study of dendritic cells (DCs) for cancer has continued to develop in recent years. Actually, DCs are potent antigen-presenting cells for the induction of primary T-cell dependent immune response. When injected intratumorally, DCs acquire and process tumor antigens in situ, migrate to regional lymphoid organs, and initiate a strong tumor-specific immune response. To date, three reports have described EUS-FNI of DCs into pancreatic cancer: two for unresectable and one for pre-surgical operations. Every study has indicated the feasibility and safety. Furthermore, these reports showed that EUS-guided DCs injection might be an important option for treating advanced pancreatic cancer. EUS-guided immunotherapy is a very exciting field in interventional EUS for obstinate cancers.

Keywords: Dendritic cell, EUS-FNA, EUS-FNI, Endoscopic ultrasound, Immunotherapy

Pancreatic cancer is among the most lethal cancers because of its attendant difficulty in early diagnosis and its resistance to standard treatments such as surgical resection, radiation, and chemotherapy. Pancreatic cancer, which entails high mortality in the United States and European countries, is the fifth leading cause of cancer deaths among men and sixth among women in Japan. With 5-year survival of less than 10%, it is the major leading cause of cancer-related death in the world.1 Therefore, new therapeutic methods are required as an urgent issue.

Interventional endoscopic ultrasound (EUS) is widely performed not only for tissue diagnosis but also for treatment in patients with abnormalities of various organs. Treatment using EUS-guided fine-needle injection (EUS-FNI) has expanded the clinical utility of EUS.27 Antitumoral EUS-FNI, with its minimally invasive access for antitumoral agent delivery, is the most exciting field of interventional EUS today. Several applications of EUS-FNI for anti-cancer efforts have included drug delivery into the tumors, such as ablation using ethanol, chemotherapy, gene therapy, and cytoim-plantation. These procedures are divisible onto three categories based on the associated therapeutic mechanisms: physicochemical therapy, molecular biological therapy, and immunological therapy.2 Since Chang et al8 first reported EUS-FNI for advanced pancreatic cancer in 2000, various antitumoral agents have been injected directly into tumors.714 Recently, several immunotherapies using EUS have been performed using the benefits of EUS guidance: minimally invasive, direct access, and high resolution. As described in this paper, we reviewed EUS-guided oncologic immunotherapy for pancreatic cancer.

Pancreatic cancer is regarded as a systemic disease even if imaging modalities reveal no visible metastasis. From this perspective, immunological therapy is performed. To date, four reports in the relevant literature have described immunotherapy under EUS guidance. In particular, EUS-FNI using dendritic cells (DCs) is a promising procedure as immunotherapy for cancers (Table 1).

Allogenic mixed lymphocyte culture

Chang et al8 injected cytoimplants directly into pancreatic cancer and evaluated its technical feasibility and safety in 2000. They used allogeneic mixed lymphocyte cultures (cytoimplants). Cytoimplants were generated by coincubation of host and allogeneic donor peripheral blood mononuclear cells, causing the release of cytokines and the activation of immune effector cells. In this clinical trial, eight patients with unresectable pancreatic cancer received EUS-guided injections of cytoimplant. The first two patients received 3 billion cells, the next three patients received 6 billion cells, and the last three patients received 9 billion cells. Results showed two partial responses and one minor response, with a median survival of 13.2 months. No procedure-related complication occurred. The study demonstrated the possibilities of EUS-FNI based on immunotherapy for unresectable pancreatic cancer. Unfortunately, no active clinical protocol has existed to evaluate cytoimplants for pancreatic cancer for more than 10 years.

Immature DCs for unresectable pancreatic cancer

Immunotherapy is a novel approach that has been investigated using different types of tumors including pancreatic cancer. During the past 2 decades, adoptive immunotherapy based on tumor-infiltrating lymphocytes or lymphokine-activated killer (LAK) cells has been used in clinical trials.15,16 These early results provided the first evidence that the manipulation of the immune system represents a promising tool in cancer immunotherapy. Furthermore, Steinman et al17 discovered the crucial role of DCs for the induction of primary T cell-dependent immune responses. DCs are now regarded as the best adjuvant for antitumor immunity.

The first results for an animal model of pancreatic cancer were reported in 2002 by Akiyama et al.18 They demonstrated that tumor growth was significantly inhibited by 82% in hamsters treated (subcutaneous injection) using DCs pulsed with tumor lysate and N[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methylsulfate, and suggested that DC-based immunotherapy might be a useful approach for the treatment of pancreatic cancers. Techniques of interventional EUS permit direct intratumoral injection of DCs. Intratumorally injected DCs would acquire and process tumor antigens in situ, migrate to regional lymphoid organs via lymphoid vessels, and initiate a significant tumor specific immune response. In addition, DCs have antigen-capturing abilities and processing abilities as well as trafficking abilities only during their immature phase.19

Based on this background, the author performed EUS-FNI of immature DCs into unresectable pancreatic cancer in seven patients with prior chemotherapy (e.g., gemcitabine) failure.12 Immature DCs were generated by the patients’ peripheral blood mononuclear cells after stimulation with granulocyte–macrophage colony-stimulating factor and interleukin 4. The patients received intratumoral injections of 10 billion or more immature DCs at two to three sites using EUS-FNI. Five of the seven patients underwent irradiation prior to the initial DC injection to facilitate tumor-associated antigens for DC cross-presentation. DC was administered on Day 1, Day 8, and Day 15. The cycles were repeated every 28 days to the greatest extent possible. All DC injections were tolerated, with no clinical toxicity. No complication associated with the procedure was noted. The median patient survival was 9.9 months in spite of being refractory to gemcitabine. Results show that intratumoral injection of unpulsed DCs into the pancreatic cancer using EUS-FNI is safe, and that it can induce some clinical responses in patients with advanced diseases. Later, Hirooka et al20 reported the results of a combination therapy of gemcitabine with immunotherapy for patients with inoperable, locally advanced pancreatic cancer. Five patients received the treatment course, which consisted of an intravenous gemcitabine administration at 1000 mg/m (Day 1) and the EUS-FNI of OK432-pulsed DCs into a tumor, with subsequent intravenous infusion of LAK cells stimulated with anti-CD3 monoclonal antibody (CD3-LAKs; Day 4), at 2-week intervals. Results showed no severe treatment-related adverse events. Moreover, three of the five patients demonstrated effective responses to this clinical trial: one patient had partial remission; and two patients had long stable disease lasting longer than 6 months. In the patient with partial remission, results demonstrated that DC-based vaccination combined with gemcitabine administration induces tumor antigen-specific cytotoxic T lymphocytes. Consequently, they concluded that the combined therapy is to be regarded as synergistically effective and that it might have a role in the therapy of pancreatic cancer for inducing tumor antigen-specific cytotoxic T lymphocytes.

Immature DCs for resectable pancreatic cancer as neoadjuvant therapy

Recently, Endo et al21 reported the feasibility and safety of EUS-FNI using immature DCs with OK-432 into resectable pancreatic cancer prior to the surgical operation as neoadjuvant therapy. Nine patients enrolled in this trial (DC group) were compared with 15 patients who had undergone the operation without immature DC injection (non-DC group). The authors evaluated not only the clinical utility but also the histological changes within the tumor and lymph nodes by immunohistochemical examination of infiltrating inflammatory cells (CD4+, CD8+, Foxp3+, and CD83+). The authors reported that two patients, one of whom was stage IV with distant lymph node metastasis, had remarkably survived more than 5 years without requiring adjuvant therapy. Additionally, they showed that CD83+ cells had accumulated significantly in the regional lymph nodes of the DCs injection group as well as Foxp3+ cells in the regional and distant lymph nodes in comparison with the no DCs injection group. EUS-FNI of immature DCs as neo-adjuvant therapy might be useful for improved prognosis in patients with resectable pancreatic cancer.

EUS-guided intervention has opened new and exciting clinical applications for the management of malignancies. In particular, EUS-guided immunotherapy is an exciting field in interventional EUS for obstinate cancers. For greater development in this field, it is anticipated that endosonographers, basic scientists, and engineers will collaborate fruitfully with much greater mutual effort. Once an effective therapy becomes available, it is likely to become important for cancer treatment. This effort remains a work in progress.

Published Data on EUS-guided Immunotherapy using Dendritic Cells

AuthorNo. of patientsDiseaseIndicationCombination therapySuccess rate of EUS-FNIEfficacy/prognosisComplications
Irisawa et al 2007127Unresectable pancreatic cancer with metastasisRefractory to intravenous gemcitabineNone100%2 mixed response, 2 stable disease >6 mo. Median survival was 9.9 mo.No severe event
Hirooka et al 2009205Unresectable locally advanced pancreatic causerFirst line treatmentIntravenous gemcitabina100%1 partial remission, 2 stable disease >6 mo.No severe event
Endo et al 2012219Unresectable pancreatic cancerPrior to surgerySurgery100%1 patient (stage IV) with distant lymph node metastasis survived >6 mo.No severe event
  1. Bardou, M, and Le Ray, I (2013). Treatment of pancreatic cancer: a narrative review of cost-effectiveness studies. Best Pract Res Clin Gastroenterol. 27, 881-92.
    Pubmed CrossRef
  2. Irisawa, A, Hikichi, T, Shibukawa, G, Takagi, T, Imamura, H, and Ohira, H (2009). Interventional endoscopic ultrasonography: applications for cancer related treatment. Dig Endosc. 21, S57-60.
    Pubmed CrossRef
  3. Yan, BM, and Van Dam, J (2008). Endoscopic ultrasound-guided intratumoral therapy for pancreatic cancer. Can J Gastroenterol. 22, 405-10.
    Pubmed KoreaMed
  4. Chang, KJ, and Irisawa, A (2009). EUS 2008 Working Group document: evaluation of EUS-guided injection therapy for tumors. Gastrointest Endosc. 69, S54-8.
    Pubmed CrossRef
  5. Suzuki, R, Irisawa, A, and Bhutani, MS (2013). Endoscopic ultrasound-guided oncologic therapy for pancreatic cancer. Diagn Ther Endosc. 2013, 157581.
    Pubmed KoreaMed CrossRef
  6. Seo, DW (2010). EUS-guided antitumor therapy for pancreatic tumors. Gut Liver. 4, S76-81.
    Pubmed KoreaMed CrossRef
  7. Ponnudurai, R, Sachithanandan, S, and George, A (2011). Endoscopic ultrasound-guided injection therapy for hepatobiliary disease. J Hepatobiliary Pancreat Sci. 18, 311-8.
    Pubmed CrossRef
  8. Chang, KJ, Nguyen, PT, Thompson, JA, Kurosaki, TT, Casey, LR, and Leung, EC (2000). Phase I clinical trial of allogeneic mixed lymphocyte culture (cytoimplant) delivered by endoscopic ultrasound-guided fine-needle injection in patients with advanced pancreatic carcinoma. Cancer. 88, 1325-35.
    Pubmed CrossRef
  9. Hecht, JR, Bedford, R, Abbruzzese, JL, Lahoti, S, Reid, TR, and Soetikno, RM (2003). A phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin Cancer Res. 9, 555-61.
    Pubmed
  10. Jin, Z, Du, Y, Li, Z, Jiang, Y, Chen, J, and Liu, Y (2008). Endoscopic ultrasonography-guided interstitial implantation of iodine 125-seeds combined with chemotherapy in the treatment of unresectable pancreatic carcinoma: a prospective pilot study. Endoscopy. 40, 314-20.
    Pubmed CrossRef
  11. Chang, KJ, Lee, JG, Holcombe, RF, Kuo, J, Muthusamy, R, and Wu, ML (2008). Endoscopic ultrasound delivery of an antitumor agent to treat a case of pancreatic cancer. Nat Clin Pract Gastroenterol Hepatol. 5, 107-11.
    Pubmed CrossRef
  12. Irisawa, A, Takagi, T, Kanazawa, M, Ogata, T, Sato, Y, and Takenoshita, S (2007). Endoscopic ultrasound-guided fine-needle injection of immature dendritic cells into advanced pancreatic cancer refractory to gemcitabine: a pilot study. Pancreas. 35, 189-90.
    Pubmed CrossRef
  13. Oh, HC, Seo, DW, Song, TJ, Moon, SH, Park do, H, and Soo Lee, S (2011). Endoscopic ultrasonography-guided ethanol lavage with paclitaxel injection treats patients with pancreatic cysts. Gastroenterology. 140, 172-9.
    CrossRef
  14. Herman, JM, Wild, AT, Wang, H, Tran, PT, Chang, KJ, and Taylor, GE (2013). Randomized phase III multi-institutional study of TNFerade biologic with fluorouracil and radiotherapy for locally advanced pancreatic cancer: final results. J Clin Oncol. 31, 886-94.
    Pubmed CrossRef
  15. Kondo, H, Hazama, S, Kawaoka, T, Yoshino, S, Yoshida, S, and Tokuno, K (2008). Adoptive immunotherapy for pancreatic cancer using MUC1 peptide-pulsed dendritic cells and activated T lymphocytes. Anticancer Res. 28, 379-87.
    Pubmed
  16. Kobari, M, Egawa, S, Shibuya, K, Sunamura, M, Saitoh, K, and Matsuno, S (2000). Effect of intraportal adoptive immunotherapy on liver metastases after resection of pancreatic cancer. Br J Surg. 87, 43-8.
    CrossRef
  17. Steinman, RM, and Witmer, MD (1978). Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci USA. 75, 5132-6.
    Pubmed KoreaMed CrossRef
  18. Akiyama, Y, Maruyama, K, Nara, N, Hojo, T, Cheng, JY, and Mori, T (2002). Antitumor effects induced by dendritic cell-based immunotherapy against established pancreatic cancer in hamsters. Cancer Lett. 184, 37-47.
    Pubmed CrossRef
  19. Steinman, RM, Hawiger, D, Liu, K, Bonifaz, L, Bonnyay, D, and Mahnke, K (2003). Dendritic cell function in vivo during the steady state: a role in peripheral tolerance. Ann N Y Acad Sci. 987, 15-25.
    Pubmed CrossRef
  20. Hirooka, Y, Itoh, A, Kawashima, H, Hara, K, Nonogaki, K, and Kasugai, T (2009). A combination therapy of gemcitabine with immunotherapy for patients with inoperable locally advanced pancreatic cancer. Pancreas. 38, e69-74.
    Pubmed CrossRef
  21. Endo, H, Saito, T, Kenjo, A, Hoshino, M, Terashima, M, and Sato, T (2012). Phase I trial of preoperative intratumoral injection of immature dendritic cells and OK-432 for resectable pancreatic cancer patients. J Hepatobil Pancreat Sci. 19, 465-75.
    CrossRef