Reader‑free ELISPOT assay for immuno‑monitoring in peptide‑based cancer vaccine immunotherapy

  • Authors:
    • Sae Hayashi
    • Rin Imanishi
    • Mayuko Adachi
    • Sayaka Ikejima
    • Jun Nakata
    • Soyoko Morimoto
    • Fumihiro Fujiki
    • Sumiyuki Nishida
    • Akihiro Tsuboi
    • Naoki Hosen
    • Hiroko Nakajima
    • Kana Hasegawa
    • Yoshihiro Oka
    • Haruo Sugiyama
    • Yusuke Oji
  • View Affiliations

  • Published online on: March 5, 2020     https://doi.org/10.3892/br.2020.1289
  • Pages: 244-250
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer vaccine immunotherapy is a therapy that induces cellular immune responses against a target molecule to elicit clinical anti‑tumor effects. These cellular immune responses against the target molecule are monitored to evaluate whether the antigen‑specific cellular immune responses are induced and maintained during the vaccination period. Enzyme‑linked immunospot (ELISPOT) assay is widely performed to analyze not only the frequency of immune cells, but also their effector functions as determined by their cytokine production/secretion. The present study aimed to develop a reader‑free ELISPOT assay using a handy membrane‑punching device termed ELI 8. With the assistance of particle analysis by ImageJ software, the results of spot counting were reproducible with high inter‑assay and inter‑examiner concordance. Immune cells that produce and secrete Th1 cytokines without antigen‑peptide stimulation of peripheral blood mononuclear cells (PBMCs) were detected, and their frequencies in patients with cancer were significantly higher compared with those in healthy individuals. These frequencies varied between individuals, as well as between time points during the course of cancer vaccine immunotherapy in each patient. Due to the variability in spontaneous cytokine production/secretion by PBMCs, an antigen‑specific immune response (IR) index is proposed, which is a ratio of the number of spot‑forming cells (SFCs) subjected to antigen‑stimulation to that of SFCs with spontaneous cytokine secretion without antigen‑stimulation. This index may be used as a marker for antigen‑specific cellular immune responses in patients treated with cancer immunotherapy. The IR index successfully detected the induction of Wilms' tumor 1‑specific cellular immune responses in patients with cancer treated with cancer vaccine immunotherapy.

Introduction

Immunotherapy has been established as the fourth mode of cancer treatment with the advent of immune checkpoint inhibitors, which have become new therapeutic targets for various tumors such as malignant melanoma (1), non-small cell lung cancer (2,3), gastric cancer (4,5), malignant mesothelioma (6) and Hodgkin's lymphoma (7). Recent reports have suggested the clinical benefits of immune checkpoint inhibitors in combination with chemotherapy (8,9). Therapeutic cancer vaccine immunotherapy is a therapy that induces cellular immune responses against the target molecule to elicit clinical anti-tumor effects (10). Although cancer vaccine immunotherapy has not been established as a monotherapy, cancer vaccines may be efficiently combined with other modalities, including immune checkpoint inhibitors (10,11). It is essential to monitor cellular immune responses against the target molecule to evaluate the induction and maintenance of antigen-specific cellular immune responses during the vaccination period; analysis of antigen-specific cellular immune responses includes in vivo testing and DTH skin reaction test as well as ex vivo tests such as flow cytometric multimer, proliferation and enzyme-linked immunospot (ELISPOT) assays (12). The ELISPOT assay detects cytokine-producing cells in the antigen-stimulation conditions. Therefore, it is possible to analyze not only the frequency of antigen-specific immune cells, but also the effector functions of immune cells as determined by cytokine production/secretion (12). In addition, the ELISPOT assay is adaptable to human leukocyte antigen (HLA) class II binding helper T lymphocyte epitopes, for which qualified multimers for flow cytometric assay are not currently available to the best of our knowledge. This assay is also capable of multi-sample measurement since the procedures are simple and the assay is commonly performed in 96-well plates. Therefore, the ELISPOT assay is widely used as a monitoring tool for cellular immune response in clinical trials for infectious diseases (13,14) and cancer immunotherapy (15-17).

The Wilms' tumor 1 (WT1) gene was originally isolated as a gene responsible for pediatric kidney neoplasm and had been regarded as a tumor suppressor gene (18). However, a number of researchers consider the WT1 gene to serve an oncogenic role in leukemia (19-22) and a wide variety of solid tumors (23-25) based on the results reported by our group and other groups (26-28). The WT1 protein is highly immunogenic (29). Immunotherapies targeting WT1 have been developed in a number of countries as novel, promising therapeutic strategies for various types of cancer such as leukemia, glioblastoma and pancreatic cancer (30-39).

The aim of the present study was to test a simple Reader-free ELISPOT assay method for reproducibility and apply it to the analysis of cytokine production/secretion of PBMCs in healthy volunteers and patients with cancer, including those who were treated with WT1 peptide-based vaccine immunotherapy.

Materials and methods

Peripheral blood mononuclear cells (PBMCs)

PBMCs were obtained with written informed consent from 17 patients with cancer (12 male and 5 female; median age, 45 years; age range, 21-72 years) and six healthy individuals (2 male and 4 female; median age, 24 years; age range, 23-52 years). The types of cancer included seven cases of glioblastoma, seven cases of anaplastic glioma, one case of lung cancer, one case of salivary gland cancer and one case of rhabdomyosarcoma. Of the 17 patients, one patient with lung cancer and two patients with salivary gland cancer and glioblastoma were enrolled in clinical trials of WT1 peptide vaccine cancer immunotherapy registered as UMIN#000002001 and UMIN#000023579, respectively. In the clinical trials, WT1 peptide vaccine was administered weekly (40) or biweekly for three months. Peripheral blood was collected before and one, two, and three months after the initiation of the treatment. PBMCs were isolated from heparinized whole blood using the Ficoll-Paque method (GE Healthcare) according to the manufacturer's instructions and cryopreserved in liquid nitrogen until use. The present study was performed under the approval of the Ethical Review Board of the Faculty of Medicine, Osaka University (Suita, Japan).

Peptide synthesis

Peptides for the ELISPOT assay were synthesized by PH Japan. The amino acid sequences were as follows: WT1-235 peptide, CMTWNQMNL; WT1-126 peptide, RMFPNAPYL.

ELISPOT assay

Following hydrophilization treatment with 35% ethanol for 1 min and three washes with PBS, a membrane of each well in a 96-well filtration plate (Merck KGaA) was incubated with capture antibodies, anti-human interferon-γ (IFN-γ) monoclonal antibody (cat. no. 3420-3-250; Mabtech AB; final concentration, 15 µg/ml in PBS) and anti-human tumor necrosis factor-α (TNF-α) monoclonal antibody (cat. no. 3510-3-250; Mabtech AB; final concentration, 7.5 µg/ml in PBS) at 4˚C overnight. Following four washes with PBS, the membrane was incubated with 200 µl 1X Blocking one (cat. no. 03953-95; Nacalai Tesque, Inc.) for 2 h and washed three times with PBS. Thawed PBMCs were suspended in FBS-free RPMI-1640 medium (Nacalai Tesque, Inc.) and 5x104 cells per 100 µl were seeded in each well in triplicate and incubated with 5% CO2 in a humidified atmosphere at 37˚C for 48 h. To stimulate PBMCs, an antigen peptide was added to each well at a final concentration of 10 µg/ml. Following removal of the cell suspension, each membrane was washed with 200 µl PBS containing 0.05% Tween-20 for 10 min and treated with 100 µl ACCUMAX™ (Sigma-Aldrich; Merck KGaA) at room temperature for 15 min with gentle agitation. After three washes with PBS containing 0.05% Tween-20, each membrane was incubated at 4˚C overnight with the corresponding detection antibodies in PBS containing 1% BSA and 0.05% Tween 20: Biotinylated anti-human IFN-γ monoclonal antibody (cat. no. 3420-6-250; Mabtech AB; final concentration, 3 µg/ml) and biotinylated-anti-human TNF-α monoclonal antibody (cat. no. 3510-6-250; Mabtech AB; final concentration, 1.5 µg/ml). Following four washes with PBS, each membrane was incubated with alkaline phosphatase-conjugated streptavidin (cat. no. 3310-8; Mabtech AB; diluted 1:500 with 0.05% Tween-20 in phosphate buffered saline without magnesium and calcium [PBS (-)]) at room temperature for 1 h. After washing both sides of the membranes with deionized water for 3 min, the spots were stained with BCIP/NBT solution (Nacalai Tesque, Inc.) for 3 min followed by washing with deionized water. Following drying at 4˚C overnight, a strip of clear adhesive tape was attached to the back of the membranes of 8 wells in a single row. The membranes were punched out with an acrylic device ELI 8 (Create Ltd.). The membranes were subsequently sandwiched a second strip of adhesive tape and scanned at the resolution of 1,200 dpi. The generated digital images were analyzed by spot counting using particle analysis by ImageJ 1.45 software (National Institutes of Health) (Fig. 1).

In concordance analysis, two different examiners with the experience of ELISPOT assay of >6 months performed spot counting with the assistance of ImageJ software. Scanned images of colored membranes from six wells of ELISPOT assay with variable numbers of spots were used. Examiner-1 performed spot counting of six images on two different days for analysis of inter-assay concordance.

Statistical analysis

Difference in IFN-γ and TNF-α secretion by PBMCs between patients with cancer and healthy individuals was analyzed by Welch's t-test using Statcel 3 software (OMS Publisher). Individual values are presented. P<0.05 was considered to indicate a statistically significant difference.

Results

Inter-assay and inter-examiner concordance of the ImageJ software-assisted spot counting step in the ELISPOT assay

First, a reader-free ELIPSOT assay was developed (Fig. 1). For the membrane preparation step, the present study developed a handy acrylic punching device ELI 8. With this device, the membranes of eight wells were easily punched out at once in an array on a strip of adhesive tape. Since the counting reproducibility of spot numbers is an important factor in the ELISPOT assay, the inter-assay and inter-examiner concordances in the spot-counting step of the ELISPOT assay were examined using six sample membranes with a variable number of spots. The number of spots detected on each sample membrane was scored as follows: -, no spot; 1+, 1-9 spots; 2+, 10-29 spots; 3+, 30-89 spots; 4+, ≥90 spots. Concordance was defined as follows: i) Scores for one sample judged by one examiner on different days or by two or more examiners are identical; ii) scores for one sample are different, but are in a range across the border number between two score categories. For example, 5-15 (10±5) spots, 1+ and 2+; 20-40 (30±10) spots, 2+ and 3+; 75-105 (90±15) spots, 3+ and 4+.

First, inter-assay concordance was examined by Examiner 1. As presented in Table I, the scores judged on two different days were identical in all six examined samples. Subsequently, inter-examiner concordance was examined. The scores judged by Examiner 2 were also identical in all six examined samples (Table I). These results indicated that ImageJ software-assisted spot counting was reproducible with good inter-assay and inter-examiner concordance.

Table I

Inter-assay and inter-examiner concordance of spot counting.

Table I

Inter-assay and inter-examiner concordance of spot counting.

 Score (spot count)
Sample no.Examiner 1 Day 1Examiner 1 Day 8Examiner 2 Day 1
11+ (9)1+ (8)1+ (8)
22+ (12)2+ (17)2+ (13)
32+ (29)2+ (27)2+ (16)
43+ (36)3+ (37)3+ (33)
53+ (50)3+ (50)3+ (40)
64+ (176)4+ (143)4+ (152)
Increased spontaneous production/secretion of Th1 type cytokines by PBMCs in patients with cancer

Counts of spot-forming cells (SFCs) are defined as the number of SFCs in the respective antigen-stimulated test conditions minus the number of SFCs in antigen-free control conditions. Thus, spontaneous production and secretion of Th1 type cytokines IFN-γ and TNF-α by PBMCs in the absence of antigen peptides was analyzed by the ELISPOT assay in 17 patients with cancer and six healthy subjects. The numbers of cells that spontaneously produced IFN-γ and TNF-α were between 8 and 548 (median, 103) and between 23 and 756 (median, 100), respectively, per 1.5x105 PBMCs in 17 patients. By contrast, the numbers of IFN-γ and TNF-α producing cells in healthy subjects were between 5 and 28 (median, 7) and between 5 and 26 (median, 11.5), respectively, per 1.5x105 PBMCs (Fig. 2A). To investigate the spontaneous cytokine secretion by immune cells in cancer vaccine-treated patients, spontaneous production of IFN-γ by PBMCs was analyzed in patients with cancer treated with WT1 peptide vaccine cancer immunotherapy at different time points during three months of treatment. Pt-01 was a patient with lung cancer treated with the WT1-235 peptide vaccine 12 times. Pt-02 and Pt-03 were patients with salivary gland cancer and glioblastoma, respectively, who were treated with WT1 Trio peptide vaccine composed of three WT1 peptides including WT1-126 and WT1-235 seven times. WT1-126 and WT1-235 are HLA class I-binding CTL peptides specific for HLA-A*02:01 and HLA-A* 24:02, respectively. SFCs with spontaneous secretion of IFN-γ increased 40.7- and 4.1-fold in two patients, but decreased 0.2-fold in one patient after three months of WT1 peptide vaccine cancer immunotherapy (Fig. 2B).

These results indicated that the changes in the numbers of spontaneous cytokine-producing immune cells should be taken into consideration in the monitoring of antigen-specific cellular immune responses.

Antigen-specific immune response (IR) index as a marker for antigen-specific cellular immune response

Secretion of IFN-γ by PBMCs was analyzed by ELISPOT assay in three patients with cancer at the indicated time points during three months of WT1 peptide vaccine cancer immunotherapy. WT1-235 and WT1-126 peptides were used for antigen-stimulation of HLA-A*24:02 patients (Pt-01 and Pt-03) and HLA-A*02:01 patient (Pt-02), respectively. First, WT1 antigen-specific IFN-γ secretion by PBMCs was described as antigen-specific spot number: (Number of SFCs in antigen-stimulated test conditions)-(number of SFCs in antigen-free control conditions) (Fig. 3). As presented in Fig. 3A, masked by the number of spontaneous cytokine-producing cells, antigen-specific IFN-γ-secreting spot numbers became negative at multiple time points. In addition, the number of SFCs provided no information about the frequency of antigen-specific cytokine-secreting cells in the total pool of cytokine-producing cells. Therefore, when the number of antigen-specific SFCs increased after vaccine immunotherapy, it remained unclear whether this increase was due to antigen-non-specific effects or the induction of antigen-specific cellular immune responses. Therefore, WT1 antigen-specific IFN-γ secretion by PBMCs was also described as antigen-specific IR index: (Number of SFCs in antigen-stimulated test conditions)/(number of SFCs in antigen-free, control conditions). With this index, the direction of the cellular immune responses to the targeted antigen WT1 was successfully detected (Fig. 3B).

Discussion

In the present study, a reader-free ELISPOT assay was developed using a membrane-punching device ELI 8. Using particle analysis by ImageJ, the results of spot counting were reproducible with good inter-assay and inter-examiner concordance. ELISPOT analysis demonstrated that immune cells that produced and secreted Th1 cytokines without antigen-peptide stimulation were present in PBMCs, and that their frequencies in patients with cancer were significantly higher compared with those in healthy individuals. These frequencies varied between individuals or time points during the course of cancer vaccine immunotherapy. Due to the variability in spontaneous cytokine production/secretion by PBMCs, the present study proposed an antigen-specific IR index rather than the number of spot-forming cells as a marker for the cellular immune responses in patients treated with cancer vaccine immunotherapy. This index successfully detected the induction of WT1-specific cellular immune responses in patients with cancer treated with WT1 peptide vaccine immunotherapy.

The ELISPOT assay is performed for various immuno-monitoring purposes including clinical trials for infectious diseases (13,14) and cancer immunotherapy (15-17). For reader-free ELISPOT assay, the preparation of ELISPOT membrane for spot counting can be a time-consuming process. A single-well punch kit, ELIPUNCH (EMD Millipore) is not currently available. To the best of our knowledge, Eli.Punch (A.EL.VIS GmbH) is the only available punching tool for the ELISPOT assay. In addition, since Eli.Punch is a device specifically designed for 96-well punching, it does not allow flexibility in well numbers. In the present study, a handy acrylic punching device ELI 8 was developed. With this device, membranes of eight wells may be easily punched out at once in an array on a strip of adhesive tape, allowing increased flexibility in the scale of the assay compared with commercially available methods. In addition, punching with ELI 8 is economical due to minimal requirements such as adhesive tape.

In the present study, digital images of scanned membranes were converted to binary images and analyzed using free ImageJ particle analysis software provided by the National Institutes of Health. In addition to saving labor by semi-automation, analysis using ImageJ demonstrated that spot counting in the ELISPOT assay achieved high inter-assay and inter-examiner concordance. In the spot counting process, the setting of the threshold for determining a spot is a crucial step; as an examiner typically sets the threshold value, recording this value makes the analysis process traceable. Therefore, the threshold setting data may be useful for education purposes to match the criteria of threshold setting among multiple examiners, including beginners.

In the ELISPOT assay, antigen-specific cytokine secretion of immune cells is often reported as the number of SFCs in the respective antigen-stimulated test conditions minus the number of SFCs in antigen-free control conditions (41,42). The results of the present clearly demonstrated that there is a statistically significant difference between spontaneous cytokine production/secretion in patients with cancer and healthy individuals, and that spontaneous cytokine production/secretion changed over time in the three patients treated with WT1 peptide vaccine. Despite a small sample size, these results demonstrated that spontaneous production/secretion of cytokines in immune cells varied between individuals and over time during the course of cancer vaccine immunotherapy. Since the number of antigen-specific SFCs does not provide information about the frequency of antigen-specific cytokine-secreting cells in the total pool of cytokine-producing cells, changes in the number of spontaneous cytokine-producing immune cells should be taken into consideration in monitoring antigen-specific cellular immune responses. In addition, detection of antigen-specific cytokine secretion by immune cells in an ELISPOT assay may be better reported not only in terms of antigen-specific SFC numbers, but additionally with regard to a supplementary antigen-specific IR index (Fig. 4). One advantage of the antigen-specific IR index is its robustness in measuring error as an indicator of antigen-specific cytokine secretion. The number of spontaneous IFN-γ-secreting PBMCs changes widely even within the same patient. As demonstrated by the concordance analysis in the present study, as the number of spots increases, it is expected that the measurement error also increases. Assuming that the true numbers of SFCs for antigen-stimulation and antigen-free control conditions are 160 and 150, respectively, with a measurement error of 10%, SFCs for the two conditions would be counted as 154-176 and 135-165, respectively. Thus, antigen-specific cytokine secretion would be reported as between -11 and 41 with respect to antigen-specific SFC number, which is a wide range of variation; however, it would be reported as between 0.93 and 1.303 with respect to antigen-specific IR index. This simulation indicates that the antigen-specific IR index may be more resistant to measurement error compared with the antigen-specific SFC number as an indicator of antigen-specific cytokine secretion.

Acknowledgements

Not applicable.

Funding

This work was supported in part by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan (grant no. KAKENHI#15K09476).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

YuO, SH, SM, FF and HS contributed to the study conception and design. SH, RI, MA and SI performed the ELISPOT analysis. YuO, JN, SN, AT, NH, HN, KH and YoO contributed to the acquisition of the patient samples and conception of the study. YuO and SH drafted the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Blood samples were obtained with written informed consent. The present study was approved by the Ethics Committee of Osaka University Hospital (approval nos. 13110 and 11293).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369:134–144. 2013.PubMed/NCBI View Article : Google Scholar

2 

Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, et al: Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 373:123–135. 2015.PubMed/NCBI View Article : Google Scholar

3 

Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van den Heuvel MM, Ciuleanu TE, Badin F, et al: First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 376:2415–2426. 2017.PubMed/NCBI View Article : Google Scholar

4 

Togasaki K, Sukawa Y, Kanai T and Takaishi H: Clinical efficacy of immune checkpoint inhibitors in the treatment of unresectable advanced or recurrent gastric cancer: An evidence-based review of therapies. Onco Targets Ther. 11:8239–8250. 2018.PubMed/NCBI View Article : Google Scholar

5 

Kelly RJ: Immunotherapy for esophageal and gastric cancer. Am Soc Clin Oncol Educ Book. 37:292–300. 2017.PubMed/NCBI View Article : Google Scholar

6 

Forde PM, Scherpereel A and Tsao AS: Use of immune checkpoint inhibitors in mesothelioma. Curr Treat Options Oncol. 20(18)2019.PubMed/NCBI View Article : Google Scholar

7 

Matsuki E and Younes A: Checkpoint inhibitors and other immune therapies for hodgkin and non-hodgkin lymphoma. Curr Treat Options Oncol. 17(31)2016.PubMed/NCBI View Article : Google Scholar

8 

Addeo A, Banna GL, Metro G and Di Maio M: Chemotherapy in combination with immune checkpoint inhibitors for the first-line treatment of patients with advanced non-small cell lung cancer: A systematic review and literature-based meta-analysis. Front Oncol. 9(264)2019.PubMed/NCBI View Article : Google Scholar

9 

Gamerith G, Kocher F, Rudzki J and Pircher A: ASCO. 2018 NSCLC highlights-combination therapy is key. Memo. 11:266–271. 2018.PubMed/NCBI View Article : Google Scholar

10 

Tran T, Blanc C, Granier C, Saldmann A, Tanchot C and Tartour E: Therapeutic cancer vaccine: Building the future from lessons of the past. Semin Immunopathol. 41:69–85. 2019.PubMed/NCBI View Article : Google Scholar

11 

Hu Z, Ott PA and Wu CJ: Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 18:168–182. 2018.PubMed/NCBI View Article : Google Scholar

12 

Lamano JB, Ampie L, Choy W, Kesavabhotla K, DiDomenico JD, Oyon DE, Parsa AT and Bloch O: Immunomonitoring in glioma immunotherapy: Current status and future perspectives. J Neurooncol. 127:1–13. 2016.PubMed/NCBI View Article : Google Scholar

13 

Ogunjimi B, Smits E, Hens N, Hens A, Lenders K, Ieven M, Van Tendeloo V, Van Damme P and Beutels P: Exploring the impact of exposure to primary varicella in children on varicella-zoster virus immunity of parents. Viral Immunol. 24:151–157. 2011.PubMed/NCBI View Article : Google Scholar

14 

Blume J, Kostler J and Weissert R: Benefit of ELISpot in early diagnosis of tuberculous meningoencephalitis: Case report and literature review. eNeurologicalSci. 1:51–53. 2015.PubMed/NCBI View Article : Google Scholar

15 

Butterfield LH: The society for immunotherapy of cancer biomarkers task force recommendations review. Semin Cancer Biol. 52:12–15. 2018.PubMed/NCBI View Article : Google Scholar

16 

Smith SG, Harris SA, Satti I, Bryan D, Walker KB, Dockrell HM, McShane H and Ho MM: Assay optimisation and technology transfer for multi-site immuno-monitoring in vaccine trials. PLoS One. 12(e0184391)2017.PubMed/NCBI View Article : Google Scholar

17 

Scheibenbogen C, Romero P, Rivoltini L, Herr W, Schmittel A, Cerottini JC, Woelfel T, Eggermont AM and Keilholz U: Quantitation of antigen-reactive T cells in peripheral blood by IFNgamma-ELISPOT assay and chromium-release assay: A four-centre comparative trial. J Immunol Methods. 244:81–89. 2000.PubMed/NCBI View Article : Google Scholar

18 

Call KM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Haber DA, Rose EA, Kral A, Yeger H and Lewis WH: Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms' tumor locus. Cell. 60:509–520. 1990.PubMed/NCBI View Article : Google Scholar

19 

Inoue K, Sugiyama H, Ogawa H, Nakagawa M, Yamagami T, Miwa H, Kita K, Hiraoka A, Masaoka T and Nasu K: WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood. 84:3071–3079. 1994.PubMed/NCBI

20 

Inoue K, Tamaki H, Ogawa H, Oka Y, Soma T, Tatekawa T, Oji Y, Tsuboi A, Kim EH, Kawakami M, et al: Wilms' tumor gene (WT1) competes with differentiation-inducing signal in hematopoietic progenitor cells. Blood. 91:2969–2976. 1998.PubMed/NCBI

21 

Ito K, Oji Y, Tatsumi N, Shimizu S, Kanai Y, Nakazawa T, Asada M, Jomgeow T, Aoyagi S, Nakano Y, et al: Antiapoptotic function of 17AA(+)WT1 (Wilms' tumor gene) isoforms on the intrinsic apoptosis pathway. Oncogene. 25:4217–4229. 2006.PubMed/NCBI View Article : Google Scholar

22 

Algar EM, Khromykh T, Smith SI, Blackburn DM, Bryson GJ and Smith PJ: A WT1 antisense oligonucleotide inhibits proliferation and induces apoptosis in myeloid leukaemia cell lines. Oncogene. 12:1005–1014. 1996.PubMed/NCBI

23 

Oji Y, Miyoshi S, Maeda H, Hayashi S, Tamaki H, Nakatsuka S, Yao M, Takahashi E, Nakano Y, Hirabayashi H, et al: Overexpression of the Wilms' tumor gene WT1 in de novo lung cancers. Int J Cancer. 100:297–303. 2002.PubMed/NCBI View Article : Google Scholar

24 

Oji Y, Suzuki T, Nakano Y, Maruno M, Nakatsuka S, Jomgeow T, Abeno S, Tatsumi N, Yokota A, Aoyagi S, et al: Overexpression of the Wilms' tumor gene W T1 in primary astrocytic tumors. Cancer Sci. 95:822–827. 2004.PubMed/NCBI View Article : Google Scholar

25 

Kanai T, Ito Z, Oji Y, Suka M, Nishida S, Takakura K, Kajihara M, Saruta M, Fujioka S, Misawa T, et al: Prognostic significance of Wilms' tumor 1 expression in patients with pancreatic ductal adenocarcinoma. Oncol Lett. 16:2682–2692. 2018.PubMed/NCBI View Article : Google Scholar

26 

Jomgeow T, Oji Y, Tsuji N, Ikeda Y, Ito K, Tsuda A, Nakazawa T, Tatsumi N, Sakaguchi N, Takashima S, et al: Wilms' tumor gene WT1 17AA(-)/KTS(-) isoform induces morphological changes and promotes cell migration and invasion in vitro. Cancer Sci. 97:259–270. 2006.PubMed/NCBI View Article : Google Scholar

27 

Tatsumi N, Oji Y, Tsuji N, Tsuda A, Higashio M, Aoyagi S, Fukuda I, Ito K, Nakamura J, Takashima S, et al: Wilms' tumor gene WT1-shRNA as a potent apoptosis-inducing agent for solid tumors. Int J Oncol. 32:701–711. 2008.PubMed/NCBI

28 

Wagner KD, Wagner N, Wellmann S, Schley G, Bondke A, Theres H and Scholz H: Oxygen-regulated expression of the Wilms' tumor suppressor Wt1 involves hypoxia-inducible factor-1 (HIF-1). FASEB J. 17:1364–1366. 2003.PubMed/NCBI View Article : Google Scholar

29 

Oka Y, Udaka K, Tsuboi A, Elisseeva OA, Ogawa H, Aozasa K, Kishimoto T and Sugiyama H: Cancer Immunotherapy targeting Wilms' tumor gene WT1 product. J Immunol. 164:1873–1880. 2000.PubMed/NCBI View Article : Google Scholar

30 

Gao L, Bellantuono I, Elsässer A, Marley SB, Gordon MY, Goldman JM and Stauss HJ: Selective elimination of leukemic CD34(+) progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood. 95:2198–2203. 2000.PubMed/NCBI

31 

Oka Y, Tsuboi A, Taguchi T, Osaki T, Kyo T, Nakajima H, Elisseeva OA, Oji Y, Kawakami M, Ikegame K, et al: Induction of WT1 (Wilms' tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc Natl Acad Sci USA. 101:13885–13890. 2004.PubMed/NCBI View Article : Google Scholar

32 

Tawara I, Kageyama S, Miyahara Y, Fujiwara H, Nishida T, Akatsuka Y, Ikeda H, Tanimoto K, Terakura S, Murata M, et al: Safety and persistence of WT1-specific T-cell receptor gene-transduced lymphocytes in patients with AML and MDS. Blood. 130:1985–1994. 2017.PubMed/NCBI View Article : Google Scholar

33 

Keilholz U, Letsch A, Busse A, Asemissen AM, Bauer S, Blau IW, Hofmann WK, Uharek L, Thiel E and Scheibenbogen C: A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood. 113:6541–6548. 2009.PubMed/NCBI View Article : Google Scholar

34 

Anguille S, Van de Velde AL, Smits EL, Van Tendeloo VF, Juliusson G, Cools N, Nijs G, Stein B, Lion E, Van Driessche A, et al: Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood. 130:1713–1721. 2017.PubMed/NCBI View Article : Google Scholar

35 

Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, Gostick E, Vermeulen K, Pieters K, Nijs G, et al: Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci USA. 107:13824–13829. 2010.PubMed/NCBI View Article : Google Scholar

36 

Oji Y, Oka Y, Nishida S, Tsuboi A, Kawakami M, Shirakata T, Takahashi K, Murao A, Nakajima H, Narita M, et al: WT1 peptide vaccine induces reduction in minimal residual disease in an imatinib-treated CML patient. Eur J Haematol. 85:358–360. 2010.PubMed/NCBI View Article : Google Scholar

37 

Nishida S, Ishikawa T, Egawa S, Koido S, Yanagimoto H, Ishii J, Kanno Y, Kokura S, Yasuda H, Oba MS, et al: Combination gemcitabine and WT1 peptide vaccination improves progression-free survival in advanced pancreatic ductal adenocarcinoma: A phase II randomized study. Cancer Immunol Res. 6:320–331. 2018.PubMed/NCBI View Article : Google Scholar

38 

Oji Y, Inoue M, Takeda Y, Hosen N, Shintani Y, Kawakami M, Harada T, Murakami Y, Iwai M, Fukuda M, et al: WT1 peptide-based immunotherapy for advanced thymic epithelial malignancies. Int J Cancer. 142:2375–2382. 2018.PubMed/NCBI View Article : Google Scholar

39 

Zauderer MG, Tsao AS, Dao T, Panageas K, Lai WV, Rimner A, Rusch VW, Adusumilli PS, Ginsberg MS, Gomez D, et al: A randomized phase ii trial of adjuvant galinpepimut-S, WT-1 analogue peptide vaccine, after multimodality therapy for patients with malignant pleural mesothelioma. Clin Cancer Res. 23:7483–7489. 2017.PubMed/NCBI View Article : Google Scholar

40 

Oji Y, Hashimoto N, Tsuboi A, Murakami Y, Iwai M, Kagawa N, Chiba Y, Izumoto S, Elisseeva O, Ichinohasama R, et al: Association of WT1 IgG antibody against WT1 peptide with prolonged survival in glioblastoma multiforme patients vaccinated with WT1 peptide. Int J Cancer. 139:1391–1401. 2016.PubMed/NCBI View Article : Google Scholar

41 

Cox MC, Castiello L, Mattei M, Santodonato L, D'Agostino G, Muraro E, Martorelli D, Lapenta C, Di Napoli A, Di Landro F, et al: Clinical and antitumor immune responses in relapsed/refractory follicular lymphoma patients after intranodal injections of IFNα-dendritic cells and rituximab: A phase I clinical tril. Clin Cancer Res. 25:5231–5241. 2019.PubMed/NCBI View Article : Google Scholar

42 

Kalli KR, Block MS, Kasi PM, Erskine CL, Hobday TJ, Dietz A, Padley D, Gustafson MP, Shreeder B, Puglisi-Knutson D, et al: Folate receptor alpha peptide vaccine generates immunity in breast and ovarian cancer patients. Clin Cancer Res. 24:3014–3025. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2020
Volume 12 Issue 5

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hayashi S, Imanishi R, Adachi M, Ikejima S, Nakata J, Morimoto S, Fujiki F, Nishida S, Tsuboi A, Hosen N, Hosen N, et al: Reader‑free ELISPOT assay for immuno‑monitoring in peptide‑based cancer vaccine immunotherapy. Biomed Rep 12: 244-250, 2020
APA
Hayashi, S., Imanishi, R., Adachi, M., Ikejima, S., Nakata, J., Morimoto, S. ... Oji, Y. (2020). Reader‑free ELISPOT assay for immuno‑monitoring in peptide‑based cancer vaccine immunotherapy. Biomedical Reports, 12, 244-250. https://doi.org/10.3892/br.2020.1289
MLA
Hayashi, S., Imanishi, R., Adachi, M., Ikejima, S., Nakata, J., Morimoto, S., Fujiki, F., Nishida, S., Tsuboi, A., Hosen, N., Nakajima, H., Hasegawa, K., Oka, Y., Sugiyama, H., Oji, Y."Reader‑free ELISPOT assay for immuno‑monitoring in peptide‑based cancer vaccine immunotherapy". Biomedical Reports 12.5 (2020): 244-250.
Chicago
Hayashi, S., Imanishi, R., Adachi, M., Ikejima, S., Nakata, J., Morimoto, S., Fujiki, F., Nishida, S., Tsuboi, A., Hosen, N., Nakajima, H., Hasegawa, K., Oka, Y., Sugiyama, H., Oji, Y."Reader‑free ELISPOT assay for immuno‑monitoring in peptide‑based cancer vaccine immunotherapy". Biomedical Reports 12, no. 5 (2020): 244-250. https://doi.org/10.3892/br.2020.1289