Abstract
Clinical trials are governed by an increasingly stringent regulatory framework, which applies to all levels of trial conduct. Study critical immunological endpoints, which define success or failure in early phase clinical immunological trials, require formal pre-trial validation. In this case study, we describe the assay validation process, during which the sensitivity, and precision of immunological endpoint assays were defined. The purpose was the evaluation of two multicentre phase I/II clinical trials from our unit in Southampton, UK, which assess the effects of DNA fusion vaccines on immune responses in HLA-A2+ patients with carcinoembryonic antigen (CEA)-expressing malignancies and prostate cancer. Validated immunomonitoring is being performed using ELISA and IFNγ ELISPOTs to assess humoral and cellular responses to the vaccines over time. The validated primary endpoint assay, a peptide-specific CD8+ IFNγ ELISPOT, was tested in a pre-trial study and found to be suitable for the detection of low frequency naturally occurring CEA- and prostate-derived tumour-antigen-specific T cells in patients with CEA-expressing malignancies and prostate cancer.
Keywords: Immunotherapy, Immunomonitoring, Assay validation, DNA fusion vaccine, CEA, PSMA
Introduction
Measurements of immune responses provide important endpoints for monitoring the efficacy of anti-cancer vaccines in early phase immunotherapeutic trials [1]. The formal validation of these endpoint assays, which define the success or failure of a study, has now become a necessary pre-trial requirement [2–5]. The validation process itself provides an opportunity to characterize and improve the performance quality and consistency of the endpoint assay. As a result validation permits the generation of reliable and interpretable data sets. However, the task of optimizing and characterizing the performance of immunological endpoint assays for the specific conditions of each trial is by no means insignificant or trivial. The substantial impact on resources in terms of time spent, consumables and appropriately trained personnel is particularly difficult for small academic laboratories. As funding for this type of activity is not a priority for granting bodies it must be accommodated within the clinical trial budget. In recent years there has been a welcome growth of interest in the issues surrounding assay validation in the immunotherapy field [6–13]. In particular, the need for comparability between results published by different laboratories worldwide has led to the formation of international working groups and the introduction of large scale initiatives of inter-laboratory testing programmes and proficiency panels, such as the Cancer Vaccine Consortium of the Cancer Research Institute [8] and the Cancer IMmunoTherapy (C-IMT) Monitoring Panel [6]. For individual laboratories and for the immunotherapy community as a whole there are obvious benefits to be gained from these initiatives, ranging from the small-scale optimization of individual protocols to a wider increase in the understanding of and in the credibility of immunomonitoring results.
We are currently undertaking two multicentre phase I/II clinical trials to assess the effect of DNA fusion vaccines on immune responses in HLA-A2+ patients with carcinoembryonic antigen (CEA)-expressing malignancies and prostate cancer. The studies are designed to test anti-tumour DNA fusion vaccines, encoding either the CEA-derived CAP1 peptide or the Prostate-Specific Membrane Antigen peptide, PSMA27, fused to Domain1 (Dom) of Fragment C (FrC) of tetanus toxoid [1, 14]. We are using ELISA to measure vaccine-induced changes in anti-FrC antibody, and ELISPOT for the detection of IFNγ-secreting tumour antigen-specific CD8+ T-cells and FrC-specific CD4+ T-cells. Here we describe our approach to the pre-trial optimization and validation of the ELISA and ELISPOT assays. Our objectives were to show that the assay performance characteristics would meet the requirements of the intended analytical application and to generate reliable data sets with which to assess the immunological outcome of the vaccines. Specifically we intended to assess changes in responses compared to a baseline value. We tested the performance of the CD8+ T-cell IFNγ ELISPOT for a sample of the patient population in a pre-trial study. Additionally, our participation in a series of inter-lab testing panels with C-IMT [6] has provided a hitherto unavailable means of external validation.
Materials and methods
DNA vaccine clinical trials
The anti-CEA DNA Vaccine (ACVA) study, GTAC 076 (www.advisorybodies.doh.gov.uk/genetics/gtac/index.htm), and the anti-PSMA27 DNA Vaccine (PSMA27) study, GTAC 089, are multicentre phase I/II trials of antigen/pDom fusion genes [1]. Peripheral blood mononuclear cells (PBMCs) and serum samples are being collected for immune monitoring over a series of 16 (ACVA) or 19 (PSMA27) timepoints for the study duration of 64 or 72 weeks, respectively.
Sample collection and storage
All human samples were obtained following ethical approval by the national (NREC) and local research ethics committees (LREC), and were taken and used with informed consent of the donors. Time between venesection and processing of fresh blood samples was pre-defined to be shorter than 4 h. Procedures were performed by trained personnel according to formal standard operating procedures (SOPs). The ELISA was validated using human serum samples, donated by healthy volunteers and stored frozen at −80°C. The IFNγ ELISPOT assays were validated using cryopreserved PBMCs from healthy donors. PBMCs were isolated from heparinized blood samples using Lymphoprep (Axis Shield), recovery and viability assessed using a manual haemocytometer and Trypan Blue exclusion. Aliquots of 5 × 106 PBMCs were cryopreserved in RPMI 1640 with 50% human AB serum (Sigma) and 10% dimethylsulphoxide using a Nalgene controlled rate freezing container overnight at −80°C before transfer to LN2 vapour phase for storage until required. For the CD8+ ELISPOT, volunteers were genotyped for the presence of HLA-A0201 by PCR in the transplant immunology laboratory of our centre.
Anti-FrC ELISA and anti DOM ELISA
Our vaccine design incorporates DNA encoding a minimized section (Dom) of FrC, which is a ~50 kDa c-terminal portion of tetanus toxoid [1, 14]. For the anti-FrC ELISA, recombinant FrC protein antigen was generated in house by expression in Escherichia coli. Briefly, 96-well plates were coated with 1.25 μg/ml FrC overnight at 4°C. Plates were blocked at 37°C for 1 h with 1% Bovine serum albumin (Sigma). Triplicates of the reference standard tetanus antitoxin human immunoglobulin, (National Institute of Biological Standards and Control, UK) were double diluted to give seven values, ranging from 0.01 IU/ml—0.000156 IU/ml. Quality control (QC) samples, previously bulk-prepared from pools of human serum and stored frozen as single use aliquots, were used as internal standards to cover high, mid and low acceptance ranges of the standard curve. Test sera were plated in triplicate at appropriate dilutions (initial titrations had been performed to establish the dilution that would fall on the linear part of the curve) and plates incubated at 37°C for 1 h. Detection was by addition of goat anti-human IgG (Fc) antibody conjugated with horseradish peroxidase for 1 h, followed by o-phenylenediamine substrate (both Sigma) Absorbance was read at 490 nm using a BIORad Ultramark 680 ELISA Microplate reader. FrC-specific antibody was quantified in relative antibody units (RAU) against the tetanus antitoxin reference standard. The ELISA was designed to detect relative changes in anti-FrC antibody over time. A positive response was set at ≥twofold compared to baseline. The statistical significance of differences between different timepoints was determined where P < 0.05 using a Student’s t-test.
A post-validation modification of the ELISA was developed using Dom protein as antigen. Recombinant Dom protein was generated in a mammalian expression system as described below.
IFNγ ELISPOT
CD8+ memory responses were measured against 10 μg/ml of a viral peptide pool [15] comprising equal amounts of cytomegalovirus (CMV), CMV pp65 493–499, (NLVPMVAVT); influenza A, Matrix 1 58–66, (GILCFVFTL); Epstein Barr virus (EBV), BMLFI 259–217 (GLCTLVAML), and measles, non-structural C protein 84–92 (KLWESPQEI) [16]. An HIV peptide, IV9 RT 476–484 (ILKEPVHGV) [17] served as negative control. All trial patients were consented to and offered counselling with respect to HIV testing; HIV positivity is a key exclusion criterion to our studies, as we anticipated that HIV infection would compromize the patients’ immunocompetence. Tumour antigen derived peptides for the pre-trial study were: CAP1, CEA 605–613 (YLSGANLNL) [18] two heteroclitic derivatives, CAP1-6D, (YLSGADLNL) [19] and CAP1-8D (own unpublished sequence, YLSGANLDL), IMI, CEA 691–699 (IMIGVLVGV) [20], PSMA27, 27–35, (VLAGGFFLL) [21]. HLA A0201-binding peptides were obtained, with certificates of ≥95% purity, from Peptide Protein Research Ltd (Fareham, UK). CD4+ responses were measured against 20 μg/ml recombinant FrC protein. Recombinant cκappa-tagged protein was generated in house, using a mammalian 293-F expression system (Freestyle, Invitrogen), and purified on a column of polyclonal sheep anti-human free kappa linked to Sepharose 4B beads. Protein quality was confirmed on SDS gels and anti-kappa blots and endotoxin levels were tested before use in ELISPOT. In both assays 5 μg/ml phytohemagglutinin (PHA) was used to confirm that cells were able to produce detectable IFNγ.
In brief, clear Multiscreen 96-well ELISPOT plates (Millipore, MAIPS4510) were pre-coated with 15 μg/ml anti-human IFNγ antibody (mAb 1 D1 K, Mabtech), and left overnight at 4°C. Thawed PBMCs (4 × 105/well) were incubated in triplicate for 20 h (CD8+) or 40 h (CD4+), at 37°C, 5% CO2 with medium only, test antigen (10 μg/ml peptide or 20 μg/ml FrC protein) or 5 μg/ml PHA in RPMI 1640 with 10% human AB serum. IFNγ secreting memory T-cells specific for the antigen were detected as spots using 1 μg/ml biotinylated IFNγ antibody (mAb 7B61 biotin, Mabtech) followed by 1 μg/ml streptavidin alkaline phosphatise (Mabtech) and a BCIP/NBT detection kit (Zymed). Spots were counted using an AID automated ELISPOT plate reader (ELRO2) A positive response to an antigen was defined where mean spot forming cells (SFC)—baseline is greater than 10 SFC/well and also >2σ above medium only wells. The statistical significance of differences between different timepoints was determined where P < 0.05 using a Student’s t-test.
Stability of materials
All commercially sourced reagents were stored according to manufacturers’ instructions; batch numbers were recorded and expiry dates adhered to. The stability of antigenic proteins, peptides and serum samples were assessed over three freeze thaw cycles and at relevant temperatures (4°C, room temperature) for 4, 24 h or 7 days following thawing (results not shown). We determined that these materials should be stored frozen (recombinant protein −20°C, peptides and serum −80°C) and used immediately after thawing. Serum should not undergo more than two freeze-thaw cycles and proteins and peptides should not be subjected to more than three freeze thaw cycles.
The stability of frozen serum (QC samples) was confirmed in the ELISA over a period of 17 months, and ongoing stability checks are performed. The stability of peptides was tested by thin layer chromatography (check for degradants), and had been confirmed over 16 months.
Results
Assay validation
Our approach to assay validation followed guidelines from several sources [5, 22–24]. The overall validation process is outlined in Fig. 1. Endpoint immunoassays defined in the Clinical Trial Protocol were optimized and validated by trained personnel to demonstrate their reliability and suitability as immunological endpoint assays. Sample handling systems, including sample processing, storage, tracking and handling by external study centres, were also validated. Formal SOPs of validated assays and sample handling systems were incorporated into an Analytical Study Plan prior to commencement of the trial. Additional resources for assay validation were obtained through a pre-trial feasibility study and by participation in the C-IMT inter-laboratory monitoring Panel [6]. Each stage was initiated following discussion with a designated qualified person from Cancer Research UK (CR-UK), as the main funding body. We adopted their recommended quality system of Good Clinical Laboratory Practice defined by the British Association for Research into Quality Assurance (BARQA) [24]. In line with this, formally documented Validation Plans and Reports were generated for review and approval by the funding body prior to commencement of the trial. All three of the immunological assays described in this case study had been developed previously in house and were well established as research assays in our laboratory. They therefore required only a limited amount of optimization to transfer the method into the intended clinical setting. The assay validation parameters were specificity, accuracy, sensitivity and precision.
Fig. 1.
Flow chart of the validation process for endpoint immunomonitoring of anti-tumour DNA fusion vaccines in clinical trials. The main validation process (grey background) was performed in conjunction with the funding body, Cancer Research UK (CR-UK) (see main text)
Validation of the anti-FrC ELISA
We used an anti-FrC ELISA with a tetanus antitoxin reference standard to detect humoral responses to the vaccine. The ELISA had originally been developed as a research assay to test the efficacy of vaccine designs in pre-clinical murine models. Some modifications were necessary for use in the human setting. Mice generate high anti-FrC antibody levels in response to vaccination and, unlike many individuals from the human population, do not possess any pre-existing anti-tetanus immunity. As the increase post DNA vaccination was unknown, the ELISA needed to be sensitive enough to measure small changes for clinical trial evaluation.
Reference standard
Quantification of anti-FrC IgG was relative to serial dilutions of the reference standard of tetanus antitoxin human immunoglobulin (see "Materials and methods”). We introduced a series of QC samples to monitor the standard curve, and maintain accuracy. These were made up from pools of human serum with known, independently measured, anti-tetanus titres (high, intermediate and low). The QC samples were also used to test dilutional linearity. This was demonstrated (r 2 > 0.99) within the range of 1:200 and 1:1,600, but not for 1:3,200 or 1:6,400 (not shown). It was decided that patient test samples should undergo an initial titration to establish the dilution that would fall within the linear range of the standard curve.
Assay specificity
We confirmed that the tetanus antitoxin reference standard could be used to derive accurate measures of serum antibody titres to the FrC component of tetanus toxin. The relationship between measurements of anti-FrC and anti-tetanus antibody titres were demonstrated in sera from healthy volunteers. In an independent research study healthy volunteers were vaccinated with tetanus toxoid and anti-tetanus IgG responses were assessed pre- and up to 20 weeks post- vaccine by ELISA [25]. The same sera were available for making comparative measurements of anti-FrC IgG. Measurements of FrC-specific antibody were qualitatively comparable, both in amplitude and timing, to responses against the full tetanus toxoid (Fig. 2a, b). Direct comparison of all measurements demonstrated that there was a strong positive correlation between the two independent data sets (r 2 = 0.94). We could therefore justify the quantification of anti-FrC antibody in RAU using the tetanus antitoxin standard.
Fig. 2.
Anti-FrC ELISA specificity: the tetanus antitoxin reference standard was demonstrated to be valid for quantifying anti-FrC antibody; a strong correlation was observed (r 2 = 0.94) between independent ELISA measurements of a anti-tetanus (IU/ml) and b anti-FrC (relative antibody units) antibody in four healthy volunteers following vaccination with tetanus toxoid adapted from Ref. [25]. c Assay selectivity was demonstrated by comparison of FrC-specific antibody in serum (n = 5) with remaining non-specific background after pre-incubation of serum with FrC protein (mean background18 ± 3%)
As our volunteer population had received a tetanus vaccination at some time in the past, it was not possible to obtain a negative control serum sample. The level of non-specific interference from the biological matrix in the absence of anti-FrC antibody was therefore assessed experimentally. Sera (n = 5) were pre-incubated with FrC to deplete anti-FrC antibodies, and the remaining absorbance provided a measure of non-specific background. FrC-specific antibody titre decreased by 82 ± 3% (Fig. 2c), and confirmed that the assay had a good level of selectivity.
Inter- and intra-assay variability
The within-lab precision (inter-assay variability) and repeatability (intra-assay variability) of the ELISA were assessed simultaneously. Two operators performed four separate ELISA assays, 2 on 1 day and two on another. Overall assay variability was quantified as the mean co-efficient of variance (%CV) between plates, assays and operators (determined as percentage variations of triplicates where %CV = [(σ/mean) × 100%]. Intra-assay variability was determined as the overall mean %CV from series’ of four (high responder) or eight (low responder) sets of triplicates. Inter- and intra-assay variability was acceptable, and did not exceed 25% for high or low responses (Table 1). The mean inter-operator CV was greater, with a maximum of 28% observed for the low responder. This confirmed the need for batch testing to minimize assay variability, and we therefore determined that each series of samples (all timepoints from one patient) should be assayed together on the same day by a single operator.
Table 1.
Anti-FrC ELISA: intra- and inter-assay variability
| Parameter | Mean (%CV) High responder (FrC-specific ab 2.3 RAU/ml) | Mean (%CV) Low responder (FrC-specific ab 0.9 RAU/ml) |
|---|---|---|
| Inter-assay variation (same operator) | 12.0 ± 6.8 | 13.1 ± 4.0 |
| Inter-operator variation (different operators) | 11.9 ± 6.2 | 15.6 ± 11.3 |
| Intra-assay variation (well-well) | 6.5 ± 6.4 | 3.7 ± 2.5 |
Overall assay variability was quantified as the mean co-efficient of variance (%CV) between plates, assays and operators (determined as percentage variations of triplicates where %CV = [(σ/mean) × 100%]. Intra-assay variability was determined as the overall mean %CV from series of four (high responder) or eight (low responder) sets of triplicates
Our acceptance criteria for the precision and accuracy of this assay had been set at a value of 25%, and 30% at the lowest QC value [23]. As a secondary endpoint assay, the validation results demonstrated acceptable levels of precision for detecting relative changes in anti-FrC antibody over time.
Post-validation modifications
The ELISA had originally been developed for testing DNA fusion vaccines with FrC specificity. Results from an earlier trial of individual DNA idiotypic scFv-FrC fusion vaccines in patients with follicular lymphoma revealed that positive detection of vaccine-related increases in anti-FrC antibody was dependent on the level of pre-existing immunity (manuscript in preparation [26]). Positive responses (>twofold) were less easily detected in patients with higher initial anti-tetanus immunity. A further parameter of “weak positive” response was therefore introduced where the antibody response was <twofold, with a clear statistically significant increase above baseline over multiple timepoints.
In our current trials, the vaccine designs incorporate a sequence for the minimized Domain 1 portion of FrC [14]. Following successful generation of recombinant Dom protein, we have now developed an anti-Dom ELISA to complement results from the anti-FrC ELISA. Only minor modification was required, within-lab precision has been demonstrated and the assay exhibits comparable performance characteristics (results not shown). Pre-existing titres of anti-Dom antibody appear to be uniformly low in the sera so far tested, compared to the varied pre-existing anti-FrC titres (data not shown, own unpublished observations). Results from the anti-Dom ELISA are therefore able to provide additional confirmation of the “weak positive” anti-FrC responses.
Validation of the anti-CAP1 and anti-PSMA27 IFN γ ELISPOTs
ELISPOT was selected as the most suitable methodology for quantifying both tumour antigen-specific and FrC-specific IFNγ-secreting T-cells in our clinical setting. The assay appeared to fulfill the desired criteria for monitoring T-cell responses to the anti-CEA vaccine [11]. It had a reported sensitivity of 1:10,000–1:50,000 antigen-specific T-cells [11, 27, 28]; it was suitable for batch testing of cryopreserved PBMCs over a series of timepoints [27, 28]; it was cost effective and relatively simple to perform for trained personnel. A further advantage was that both CD8+ and CD4+ assays could be set up simultaneously. This made efficient use of the limited cell numbers, reagent costs and time. Our intention was to use the identification of tumour antigen-specific T cells in the trial population as an indication of where further characterization (e.g. by multimer staining, cytotoxicity assays) should be made.
Our ELISPOT protocol was closely based on that published by Smith et al. [28]. We performed pre-validation optimization of incubation times and reagent concentrations to generate the best signal/noise ratio. Human AB serum was batch tested to generate the lowest background. Both the CD8+ and CD4+ ELISPOTS were validated to the same standard and both assays demonstrated very similar performance characteristics. Here we present the main results from validation of the primary endpoint assay, anti-CAP1/PSMA27 CD8+ T-cell IFNγ ELISPOT. As reference samples with defined numbers of antigen-specific T cells were not available to us at the time of assay validation, we used a pool of viral peptides as positive control (see "Materials and methods”). For our purposes this did not pose a problem, as for the clinical trials our interpretations of the ELISPOT were based on changes over time rather than absolute numbers.
PBMC quality: cryopreservation and sample handling
As cryopreservation has been shown to affect some cellular cytokine response profiles [12], we investigated the validity of using frozen, rather than fresh PBMCs in our IFNγ ELISPOT protocol. Responses to the viral peptide pool were measured in freshly extracted PBMCs from three healthy volunteers. The remaining cells were cryopreserved in liquid N2. Repeat assay were performed on the thawed cells 1 and 2 weeks later. The cryopreservation process did not lead to loss of response (P > 0.05, Student’s t-test, data not shown), which confirmed the findings of other workers [8, 27–29]. Furthermore, T-cell responses were stable following cryopreservation over the course of 17 months (Fig. 3a). This also agreed with other studies [28], and supported our requirement of batch testing each series of frozen patient samples from the trial. This was especially important for this multicentre trial, where PBMCs would be prepared and cryopreserved at external sites, then shipped to our laboratory in batches for later analysis.
Fig. 3.
CD8+ IFNγ ELISPOT: a PBMC stability: aliquots of 5 × 106 cryopreserved PBMCs from the same donor were thawed at different times over a period of 17 months. Background IFNγ release from unstimulated cells (white columns) was consistently low over time. IFNγ release in response to the viral peptide pool (cells only background subtracted, dark grey columns) was not compromised. b PBMC titration: cell inputs of 2 × 105 and 4 × 105/well gave similar responses to the viral peptide pool when normalized to spots/106 PBMCs. The sum of the individual viral peptides equaled the sum of the pool. This was not observed for an input of 1 × 105, due to a high variability for triplicates (up to 83%) where spot counts <10. c Determination of lowest cut-off value for spot forming cells (SFC)/well was by comparison of mean SFC/well and %CV = [(mean/σ) × 100%] for triplicates. %CV was unacceptably high where SFC <10
It has become increasingly evident that results of cellular assays can be compromised by poor sample quality, through inappropriate handling, inconsistent processing methods, freezing techniques [8, 12, 13]. In a multicentre trial it is absolutely critical to maintain consistency in sample quality. We therefore generated an SOP to standardize the procedures for sample handling at external study centres. The aims were:
To promote consistency in sample handling procedures between participating centres.
To work within GCP guidelines.
To maintain a coherent, auditable sample tracking system.
To obtain consistent interpretable results in endpoint assays for monitoring immune responses.
For geographically close external centres (up to 1 h driving time) blood samples were shipped by courier to arrive in Southampton for processing within 4 h of being drawn. For more distant centres training in sample handling, processing and freezing procedures was undertaken according to our SOPs, and the resultant sample quality tested following shipping to Southampton. Testing included monitoring cell yield and viability and comparing functional responses in IFNγ ELISPOT.
Assay sensitivity
Selections of optimal cell numbers per well were made as part of the validation process. The optimal PBMC input needed to be a balance between the requirements of sensitive detection of low frequency tumour associated antigen-specific cells above background, and the limitations of sample availability from trial patients. Following normalization, PBMC inputs of 2 × 105 and 4 × 105/well generated similar results in response to the viral pool (Fig. 3b). Responses to the viral pool also equaled the sum of responses to the individual component peptides. There was some discrepancy in these values for lower cell input of 1 × 105. The lack of correlation here between responses to the viral pool and the individual peptides was found to be caused by a large variability in the triplicates where spot counts/well were <10. This allowed us to identify the cut-off below which the values became unreliable. By taking all measurements (single peptides, viral pool and PHA) and plotting the %CV against the mean SFC/well of triplicates, it became apparent that there was a clear inverse relationship between the two (Fig. 3c). Similar observations have been made for the enumeration of human papilloma virus by Lathey [30]. We set our cut-off at 10 SFC/well, below which the inter-well variability was unacceptably high (>25%). Cell input number was set at 4 × 105/well to maximize the chance of detecting low frequency antigen specific cells.
Inter-assay and inter-operator variability
Inter-assay and inter-operator variability were assessed simultaneously; two trained operators performed identical ELISPOT assays on samples from donors with low (>10 < 50 SFC/well), mid (>50 < 100 SFC/well) or high (>100 SFC/well) responses [8] to the viral pool, individual viral peptides and PHA. Identical assays were also performed on different days to provide an assessment of within-lab inter-assay variability. Results are shown in Table 2. Inter-assay CVs, at 24% or below, were within the acceptable range (<25%), apart from a low response at 51%. The mean SFC/well for this response was 15 ± 10. Spot counts were above the >10 cut-off, but this could not be called a positive response because the mean SFC <2σ above the medium-only value. This response was therefore excluded from the assessment. Inter-operator variability for low and mid value responses (<100 SFC/well) was considerably higher than observed for intra-or inter-assay variability, and reached or exceeded 25% (Table 2). Reasons for this variability were not identified, as cross-counting and cross-analysis of samples processed by the other operator revealed that there were no differences between the two (data not shown).
Table 2.
CD8+ T-cell IFNγ ELISPOT: intra- and inter-assay variability
| Donor | A | B | ||||
| Antigen | VPP | VPP | ||||
| Mean response ± σ SFC/well | 100 ± 25 | 47 ± 7 | ||||
| Intra-assay mean %CV (triplicates) | 7 | 15 | ||||
| Donor | C | D | E | C | D | E |
| Antigen | VPP | VPP | VPP | PHA | PHA | PHA |
| Mean response ± σ SFC/well | 15 ± 10 | 97 ± 23 | 35 ± 10 | 372 ± 62 | 389 ± 56 | 462 ± 43 |
| Inter-assay mean %CV (same operator) | 51 | 7 | nd | 13 | 17 | nd |
| Inter-operator mean %CV (different operators) | 62 | 24 | 29 | 16 | 9 | 9 |
| Donor | F | F | F | F | F | |
| Antigen | measles | CMV | flu | EBV | VPP | |
| Mean response ± σ SFC/well | 0 ± 2 | 11 ± 2 | 25 ± 4 | 41 ± 10 | 63 ± 9 | |
| Inter-assay mean %CV (same operator) | 388 | 22 | 17 | 24 | 14 |
Variability in responses (donors A–F) is shown as mean %CV = [(σ/mean) × 100%] for intra-assay (well–well, triplicates), inter-plate (plate–plate, same operator) and inter-operator (plate–plate, different operators) validation. Grey shaded boxes show corresponding values for IFNγ release in response to viral peptide pool (VPP), PHA or individual viral antigen peptides
Although there were differences in the absolute spot counts from each operator, a strong positive correlation was observed between the two sets of results (r 2 = 0.97). This showed that either operator would generate an identical assessment of positivity from responses to the viral pool or PHA. Similar observations have previously been made by other workers [9, 10]. Our results confirmed that each series of timepoints from a patient should be batch-tested by a single operator to minimize assay variability.
Overall, we concluded that a positive response to an antigen must be >10 spots/well (background removed); for the detection of antigen-specific responses over time in vaccinated patients mean SFC—baseline must be >10/well and also >2σ above baseline. Additionally we mandate a statistically significant increase over two or more timepoints for a result to be interpretable as a response.
Application of the CD8+ IFNγ ELISPOT to the patient population in a pre-trial feasibility study
We were able to examine the performance of PBMCs from the patient population in a pre-trial feasibility study with separate LREC approval. The study was designed to assess whether patients with solid tumours are immunocompetent and able to mount T-cell responses, and whether they possess pre-existing cellular immunity to CEA-derived antigens or PSMA27. Consenting HLA-A2+ patients with CEA-expressing cancer (CEA group, n = 17, serum CEA levels ranging from 1.5 to 956 μg/ml, mean age 68 ± 10 year) or prostate cancer (PrCa group n = 13, serum PSA levels ranging from 0.06 to 180 μg/ml, mean age 75 ± 7 year) were recruited, together with a group of healthy volunteers (n = 6, mean age 34 ± 4 year). We used the validated CD8+ T-cell IFNγ ELISPOT, to measure responses to the CEA derived peptide, CAP1, and peptides from the transmembrane domain of CEA (IMI) and PSMA27.(see Sect. ”Materials and methods” for details of peptides). CAP1 and two heteroclitic peptide analogues of CAP1, CAP1-6D and CAP1-8D (containing substitutions of aspartic acid at positions 6 or 8, respectively) were also evaluated individually, in order to observe whether the epitopes gave differential responses [19].
We had previously calculated that a minimum PBMC yield of 6 × 106/10 ml blood would be required at each timepoint of the DNA vaccine studies to provide sufficient for the endpoint analyses. We were able to confirm that this would be feasible, with respective yields for patients in CEA and PrCa groups of 7.9 ± 3.6 (range 1.5–14.3) and 6.4 ± 3.8 (range 1.3–18.9) × 106/10 ml blood. Mean recovery after cryopreservation was >80% and viability >90%, similar to values obtained for healthy control samples. To ensure sufficient yields for the DNA vaccine trials we also introduced an initial screen to test PBMC yields for candidate patients with borderline lymphopenia (1.0–1.5 × 109/l).
The extent of disease in the study population was varied (Table 3). Widespread disease was present in 16/17 of patients with CEA-expressing malignancies and 5/13 of the PrCa group. ELISPOT results are given in Table 4. All patients exhibited a response to PHA. Positive responses (34/36) to the control viral peptide pool confirmed that T-cell recall responses were intact even in patients with metastatic malignancies. No responses were seen against the HIV negative control peptide. For both patient groups cellular responses to the viral pool were greater than for healthy controls, irrespective of disease extent. Age-associated clonal expansion of CMV-specific CD8+ T-cells has been observed previously in an elderly population (mean age 92) [31], and was associated with phenotypic changes and decreased proportions of CMV specific-IFNγ releasing cells. However, the absolute numbers of IFNγ-secreting PBMCs were similar to those observed in our own study. We are currently generating data for an age-matched control population.
Table 3.
Patient characteristics for pre-trial study to assess pre-existing immunity to tumour-derived antigenic peptides using CD8+ T-cell IFNγ ELISPOT
| Patient group | Disease | Most recent treatment | Response to treatment |
|---|---|---|---|
| CEA group (n = 17) patients with CEA expressing malignancies | 11 Colorectal carcinomas | Chemotherapy 17/17 | Progressed 4/17 |
| 2 Non-small cell lung cancers | Stable 4/17 | ||
| 2 Oesophageal carcinomas | Partial response 9/17 | ||
| 1 Hepatocellular carcinoma | |||
| 1 Duodenal adenocarcinoma | |||
| PrCa group (n = 13) patients with prostate cancer | 8 With localized disease | Radical radiotherapy 4/8 | Progressed 3/8 |
| Radical prostatectomy 1/8 | |||
| Hormonal therapy 2/8 | Partial response 5/8 | ||
| No therapy 1/8 | |||
| 5 With widespread disease | Hormonal therapy 5/5 | Progressed 4/5 | |
| Stable 1/5 |
Table 4.
CD8+ T-cell IFNγ ELISPOT: table of results of pre-trial pre-existing immunity study
| Study group | PHA | Viral pool | IMI | CAP-1/deriv | PSMA 27 | HIV | ||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| + | Mean SFC/106 | σ | + | Mean SFC/106 | σ | + | Mean SFC/106 | σ | + | Mean SFC/106 | σ | + | Mean SFC/106 | σ | + | |
| CEA | 17 | 925 | 889 | 15 | 675 | 503 | 1 | 110 | 0 | 0 | 0 | 1 | 33 | 0 | ||
| PrCa | 13 | 1085 | 896 | 13 | 637 | 670 | 5 | 69 | 61 | 5 | 42 | 12 | 2 | 41 | 5 | 0 |
| Control | 6 | 1,625 | 1,553 | 6 | 102 | 75 | 4 | 89 | 50 | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Responses to PHA and antigenic peptides are given for each study group showing number of responders (+), mean spot forming cells (SFC)/106 PBMCs and standard deviation (σ). Responses to, CAP1, CAP1-6D and CAP1-8D were evaluated for each individual peptide but are displayed summarized
Cellular responses against the self-antigen, CEA trans-membrane peptide (IMI), were detected in 1/17 of the group of patients with CEA expressing cancers (CEA group), 5/13 of the PrCa group and 4/6 of the healthy controls (Table 4). Of these, only the patient in the CEA group had metastatic disease. Low level cellular immune responses to CAP-1 peptides (representative of CAP1, CAP1-6D and CAP1-8D) and PSMA27 were observed mainly in the PrCa group. However, although cellular responses were generally absent in patients with high serum levels of the tumour antigens CEA or PSA, there was no simple correlation between serum levels of CEA and the CAP1 peptide responses, or serum levels of PSA and the PSMA27 peptide responses.
Discussion
ELISPOT and ELISA are widely used tools for monitoring immune responses to anti-cancer vaccines. The assays are relatively simple to perform and facilitate the batch testing of sequential samples. However, current legislation surrounding the use of human material in clinical trials of new investigational agents [2–5] dictates that these endpoint assays should be subject to validation to demonstrate that they are “fit for purpose”. Thus for each new trial the assay validation parameters are tailored to the particular requirements as endpoints. This may range from a definitive quantification of a biomarker to a qualitative indication of a positive or negative response [32]. For immunomonitoring of our DNA vaccine studies, assays were designed to measure relative vaccine-induced changes of FrC-specific antibody, FrC-specific T-cells and tumour-specific CD8+ T-cells over time. Our choice of IFNγ ELISPOT as the primary immunological endpoint was influenced by several factors: the assay had a long history of use with wide acceptance in the community [8, 33–39]; we had used ELISPOT in our pre-clinical models, and possessed the experience and technical capabilities to fulfil the regulatory requirements, and those of the sponsor. For immunomonitoring of our clinical trials, the CD8+ IFNγ ELISPOT is intended to be used in conjunction with other assays, such as tetramer, intracellular staining and cytotoxicity assays, together with phenotypic evaluation by FACS. These assays however presented more technical difficulties with regard to the extent of validation that was possible, and are being undertaken as research assays to consolidate the immunological information generated. The relative merits of ELISPOT and other tests in this context have been subject to a careful review by Coulie and van der Bruggen [40]. More recently, important developments in FACS based multi-parametric analyses have been described [41]; these enable the simultaneous characterization of multiple T-cell functions and hold great promise for the elucidation of vaccine-induced T-cell responses.
For the purpose of this case study, we have focused on the ELISA and ELISPOT assays as conceptual examples of validating immunological endpoints for a specific purpose. The assay validations were performed by trained operators according to a formal validation plan, and SOPs of the validated methods were generated. However, the overall validation process was not confined to the assays themselves, and considerable effort was applied to the implementation of quality systems surrounding sample collection, processing and tracking to ensure consistency between participating centres (Fig. 1).
In addition to the assessments of precision and sensitivity, which had been performed using PBMCs from healthy donors, we were also able to confirm that the primary endpoint assay, the CD8+ IFNγ ELISPOT, was applicable to the relevant patient population. We used the validated ELISPOT to measure pre-existing CD8+ T-cell responses to the viral peptide pool and a panel of tumour antigens in patients with CEA-expressing cancer or prostate cancer, compared to a healthy control group. The yield and quality of PBMCs was not compromised in the patient population, and T-cell responses to recall viral antigens were intact. We concluded that even in patients with advanced disease, T-cell responses to recall antigens are present and can be readily enumerated using our validated CD8+ IFNγ ELISPOT protocol, and that memory T-cell responses to viral peptides do not appear to be negatively affected by disease status.
Tumour antigen-specific T-cell responses were detected in all groups at frequencies of up to 110/106, which confirmed that the ELISPOT was an appropriate tool to use for this purpose. Numerous reports have been made of naturally occurring T-cell immunity to a range of tumour associated antigens (reviewed in [42]), including CAP1 [43]. It is difficult to interpret the biological significance of these observations, as they appear to be dependent on multiple factors; the type of tumour, the extent of disease, the nature of the antigen and whether patients had undergone chemotherapy have all been thought to influence the existence of tumour antigen-specific T-cells in peripheral blood [42]. In our own study, responses to one or more of the test antigens were observed in 1/17 (8%) of the CEA group, 10/13 (77%) of the PrCa group and in 4/6 (67%) of the control group (all IMI-specific). To our knowledge the IMI-specific T cell responses in a healthy population is a new finding, which, although beyond the scope of this paper, warrants further investigation in follow up studies using larger numbers of volunteers. With regard to the patients, our observations suggested that naturally occurring cellular immunity to these tumour antigens may be compromised in the presence of high antigen levels or advanced disease. However, the presence of a measurable T-cell repertoire against tumour antigens such as CEA and PSMA in patients with low disease burden suggests that this clinical setting can be exploited for vaccination against tumour antigens, although the testing of anti-tumour vaccination may best be performed in the adjuvant setting.
During 2005 and 2006 our laboratory participated in an international inter-laboratory testing project to compare protocols for enumeration of antigen-specific T-cell responses, including tetramer staining and IFNγ ELISPOT [6]. In Phase I the major protocol variables influencing sensitivity were identified from the results of 12 European centres. Incorporation of new guidelines into the Phase II panel led to successful increases in sensitivity and reductions in inter-centre variability. From our own perspective we were able to benefit from the combined professional expertise of the other participating centres; we received performance feedback showing that our cell recoveries, low background and detection of positive responses at a frequency of 12 SFC/well compared favourably with other experienced laboratories, and providing confirmation of the validity of our own protocol choices. This form of external validation proved to be invaluable for testing the validity of our method within a wider sphere. Consequently the concept of “fit for purpose” can now be extended from intra-laboratory validation to the purpose of generating comparable and acceptable results across the wider scientific community.
Acknowledgments
Serum samples from healthy volunteers vaccinated with tetanus toxoid were kindly provided by Dr. Gianfranco Di Genova. We would like to thank Dr. Richard Sugar (QA Manager, Drug Development Office, Cancer Research UK) for his valuable support and assistance in the planning and documentation of the assay validations. Our thanks to Dr. J.P.Kerr for collation of clinical data for the study of pre-existing cellular immunity. AM and CO are supported by Cancer Research UK (CR-UK), FC is supported by Experimental Cancer Medicine Centre (ECMC) grant funding (joint CR–UK and the Department of Health of England, Scotland, Wales and Northern Ireland).
Conflict of interest statement
None declared.
Footnotes
This paper is a Focussed Research Review based on a presentation given at the Sixth Annual Meeting of the Association for Immunotherapy of Cancer (CIMT), held in Mainz, Germany, 15–16 May 2008.
References
- 1.Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev. 2008;8:108–120. doi: 10.1038/nrc2326. [DOI] [PubMed] [Google Scholar]
- 2.Fontaine N, Rosengren B. Directive 2001/20/EC implementation of good clinical practice in the conduct of clinical trials on medicinal products for human use. Official Journal of the European Communities. 2001;L121:34. [Google Scholar]
- 3.HMSO . UK Statutory Instrument No. 1031. The medicines for human use (clinical trials) regulations. London: HMSO; 2004. [Google Scholar]
- 4.HMSO (2006) UK Statutory Instrument No. 1928. The medicines for human use (clinical trials) amendment regulations. HMSO
- 5.ICH harmonised tripartite guidelines for good clinical practice. CPMP/ICH/135/95/1997. http://www.emea.europa.eu/pdfs/human/ich/013595en.pdf
- 6.Britten CM, Gouttefangeas C, Welters MJ, Pawelec G, Koch S, Ottensmeier C, Mander A, Walter S, Paschen A, Muller-Berghaus J, Haas I, Mackensen A, Kollgaard T, Thor Straten P, Schmitt M, Giannopoulos K, Maier R, Veelken H, Bertinetti C, Konur A, Huber C, Stevanovic S, Wolfel T, van der Burg SH. The CIMT-monitoring panel: a two-step approach to harmonize the enumeration of antigen-specific CD8+ T lymphocytes by structural and functional assays. Cancer Immunol Immunother. 2008;57:289–302. doi: 10.1007/s00262-007-0378-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Britten CM, Janetzki S, van der Burg SH, Gouttefangeas C, Hoos A. Toward the harmonization of immune monitoring in clinical trials: quo vadis? Cancer Immunol Immunother. 2008;57:285–288. doi: 10.1007/s00262-007-0379-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Janetzki S, Panageas KS, Ben-Porat L, Boyer J, Britten CM, Clay TM, Kalos M, Maecker HT, Romero P, Yuan J, Kast WM, Hoos A. Results and harmonization guidelines from two large-scale international Elispot proficiency panels conducted by the Cancer Vaccine Consortium (CVC/SVI) Cancer Immunol Immunother. 2008;57:303–315. doi: 10.1007/s00262-007-0380-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Maecker HT, Hassler J, Payne JK, Summers A, Comatas K, Ghanayem M, Morse MA, Clay TM, Lyerly HK, Bhatia S, Ghanekar SA, Maino VC, Delarosa C, Disis ML. Precision and linearity targets for validation of an IFNgamma ELISPOT, cytokine flow cytometry, and tetramer assay using CMV peptides. BMC Immunol. 2008;9:9. doi: 10.1186/1471-2172-9-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Malyguine A, Strobl SL, Shafer-Weaver KA, Ulderich T, Troke A, Baseler M, Kwak LW, Neelapu SS. A modified human ELISPOT assay to detect specific responses to primary tumor cell targets. J Transl Med. 2004;2:9. doi: 10.1186/1479-5876-2-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Scheibenbogen C, Romero P, Rivoltini L, Herr W, Schmittel A, Cerottini JC, Woelfel T, Eggermont AM, 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. 2000;244:81–89. doi: 10.1016/S0022-1759(00)00257-X. [DOI] [PubMed] [Google Scholar]
- 12.Shreffler WG, Visness CM, Burger M, Cruikshank WW, Lederman HM, de la Morena M, Grindle K, Calatroni A, Sampson HA, Gern JE. Standardization and performance evaluation of mononuclear cell cytokine secretion assays in a multicenter study. BMC Immunol. 2006;7:29. doi: 10.1186/1471-2172-7-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Smith JG, Joseph HR, Green T, Field JA, Wooters M, Kaufhold RM, Antonello J, Caulfield MJ. Establishing acceptance criteria for cell-mediated-immunity assays using frozen peripheral blood mononuclear cells stored under optimal and suboptimal conditions. Clin Vaccine Immunol. 2007;14:527–537. doi: 10.1128/CVI.00435-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Rice J, Dunn S, Piper K, Buchan SL, Moss PA, Stevenson FK. DNA fusion vaccines induce epitope-specific cytotoxic CD8(+) T cells against human leukemia-associated minor histocompatibility antigens. Cancer Res. 2006;66:5436–5442. doi: 10.1158/0008-5472.CAN-05-3130. [DOI] [PubMed] [Google Scholar]
- 15.Currier JR, Kuta EG, Turk E, Earhart LB, Loomis-Price L, Janetzki S, Ferrari G, Birx DL, Cox JH. A panel of MHC class I restricted viral peptides for use as a quality control for vaccine trial ELISPOT assays. J Immunol Methods. 2002;260:157–172. doi: 10.1016/S0022-1759(01)00535-X. [DOI] [PubMed] [Google Scholar]
- 16.van Els CA, Herberts CA, van der Heeft E, Poelen MC, den Brink JA, van der Kooi A, Hoogerhout P, Jan ten Hove G, Meiring HD, de Jong AP. A single naturally processed measles virus peptide fully dominates the HLA-A*0201-associated peptide display and is mutated at its anchor position in persistent viral strains. Eur J Immunol. 2000;30:1172–1181. doi: 10.1002/(SICI)1521-4141(200004)30:4<1172::AID-IMMU1172>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 17.Tsomides TJ, Aldovini A, Johnson RP, Walker BD, Young RA, Eisen HN. Naturally processed viral peptides recognized by cytotoxic T lymphocytes on cells chronically infected by human immunodeficiency virus type 1. J Exp Med. 1994;180:1283–1293. doi: 10.1084/jem.180.4.1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton JM, Schlom J. Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine. J Natl Cancer Inst. 1995;87:982–990. doi: 10.1093/jnci/87.13.982. [DOI] [PubMed] [Google Scholar]
- 19.Zaremba S, Barzaga E, Zhu M, Soares N, Tsang KY, Schlom J. Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen. Cancer Res. 1997;57:4570–4577. [PubMed] [Google Scholar]
- 20.Kawashima I, Hudson SJ, Tsai V, Southwood S, Takesako K, Appella E, Sette A, Celis E. The multi-epitope approach for immunotherapy for cancer: identification of several CTL epitopes from various tumor-associated antigens expressed on solid epithelial tumors. Hum Immunol. 1998;59:1–14. doi: 10.1016/S0198-8859(97)00255-3. [DOI] [PubMed] [Google Scholar]
- 21.Lu J, Celis E. Recognition of prostate tumor cells by cytotoxic T lymphocytes specific for prostate-specific membrane antigen. Cancer Res. 2002;62:5807–5812. [PubMed] [Google Scholar]
- 22.FDA . Guidance for industry: bioanalytical method validation. New York: Marcel Dekker; 2001. [Google Scholar]
- 23.Miller KJ, Bowsher RR, Celniker A, Gibbons J, Gupta S, Lee JW, Swanson SJ, Smith WC, Weiner RS. Workshop on bioanalytical methods validation for macromolecules: summary report. Pharm Res. 2001;18:1373–1383. doi: 10.1023/A:1013062600566. [DOI] [PubMed] [Google Scholar]
- 24.Stiles T, Grant V, Mawbey N. Good clinical laboratory practice. Cambridge: British Association for Research into Quality Assurance (BARQA); 2001. [Google Scholar]
- 25.Di Genova G, Roddick J, McNicholl F, Stevenson FK. Vaccination of human subjects expands both specific and bystander memory T cells but antibody production remains vaccine specific. Blood. 2006;107:2806–2813. doi: 10.1182/blood-2005-08-3255. [DOI] [PubMed] [Google Scholar]
- 26.Mander A, McCann K, Di Genova G, Roddick J, Low L, Johnson PW, Hamblin T, Stevenson FK, Ottensmeier C (2007) DNA fusion vaccination in follicular lymphoma: induction of anti-idiotypic immune responses in patients with follicular lymphoma. National Cancer Research Institute (NCRI) Conference 2007:Abstract LB65
- 27.Kreher CR, Dittrich MT, Guerkov R, Boehm BO, Tary-Lehmann M. CD4+ and CD8+ cells in cryopreserved human PBMC maintain full functionality in cytokine ELISPOT assays. J Immunol Methods. 2003;278:79–93. doi: 10.1016/S0022-1759(03)00226-6. [DOI] [PubMed] [Google Scholar]
- 28.Smith JG, Liu X, Kaufhold RM, Clair J, Caulfield MJ. Development and validation of a gamma interferon ELISPOT assay for quantitation of cellular immune responses to varicella-zoster virus. Clin Diagn Lab Immunol. 2001;8:871–879. doi: 10.1128/CDLI.8.5.871-879.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Maecker HT, Moon J, Bhatia S, Ghanekar SA, Maino VC, Payne JK, Kuus-Reichel K, Chang JC, Summers A, Clay TM, Morse MA, Lyerly HK, DeLaRosa C, Ankerst DP, Disis ML. Impact of cryopreservation on tetramer, cytokine flow cytometry, and ELISPOT. BMC Immunol. 2005;6:17. doi: 10.1186/1471-2172-6-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lathey JL (2003) Preliminary steps toward validating a clinical bioassay: a case study of the ELIspot assay. Biopharm Int
- 31.Ouyang Q, Wagner WM, Voehringer D, Wikby A, Klatt T, Walter S, Muller CA, Pircher H, Pawelec G. Age-associated accumulation of CMV-specific CD8+ T cells expressing the inhibitory killer cell lectin-like receptor G1 (KLRG1) Exp Gerontol. 2003;38:911–920. doi: 10.1016/S0531-5565(03)00134-7. [DOI] [PubMed] [Google Scholar]
- 32.Cummings J, Ward TH, Greystoke A, Ranson M, Dive C. Biomarker method validation in anticancer drug development. Br J Pharmacol. 2008;153:646–656. doi: 10.1038/sj.bjp.0707441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Keilholz U, Weber J, Finke JH, Gabrilovich DI, Kast WM, Disis ML, Kirkwood JM, Scheibenbogen C, Schlom J, Maino VC, Lyerly HK, Lee PP, Storkus W, Marincola F, Worobec A, Atkins MB. Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the Society for Biological Therapy. J Immunother. 2002;25:97–138. doi: 10.1097/00002371-200203000-00001. [DOI] [PubMed] [Google Scholar]
- 34.Diefenbach CS, Gnjatic S, Sabbatini P, Aghajanian C, Hensley ML, Spriggs DR, Iasonos A, Lee H, Dupont B, Pezzulli S, Jungbluth AA, Old LJ, Dupont J. Safety and immunogenicity study of NY-ESO-1b peptide and montanide ISA-51 vaccination of patients with epithelial ovarian cancer in high-risk first remission. Clin Cancer Res. 2008;14:2740–2748. doi: 10.1158/1078-0432.CCR-07-4619. [DOI] [PubMed] [Google Scholar]
- 35.Butterfield LH, Comin-Anduix B, Vujanovic L, Lee Y, Dissette VB, Yang JQ, Vu HT, Seja E, Oseguera DK, Potter DM, Glaspy JA, Economou JS, Ribas A. Adenovirus MART-1-engineered autologous dendritic cell vaccine for metastatic melanoma. J Immunother. 2008;31:294–309. doi: 10.1097/CJI.0b013e31816a8910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Welters MJ, Kenter GG, Piersma SJ, Vloon AP, Lowik MJ, Berends-van der Meer DM, Drijfhout JW, Valentijn AR, Wafelman AR, Oostendorp J, Fleuren GJ, Offringa R, Melief CJ, van der Burg SH. Induction of tumor-specific CD4+ and CD8+ T-cell immunity in cervical cancer patients by a human papillomavirus type 16 E6 and E7 long peptides vaccine. Clin Cancer Res. 2008;14:178–187. doi: 10.1158/1078-0432.CCR-07-1880. [DOI] [PubMed] [Google Scholar]
- 37.Harrop R, Drury N, Shingler W, Chikoti P, Redchenko I, Carroll MW, Kingsman SM, Naylor S, Griffiths R, Steven N, Hawkins RE. Vaccination of colorectal cancer patients with TroVax given alongside chemotherapy (5-fluorouracil, leukovorin and irinotecan) is safe and induces potent immune responses. Cancer Immunol Immunother. 2008;57:977–986. doi: 10.1007/s00262-007-0428-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Miller AM, Ozenci V, Kiessling R, Pisa P. Immune monitoring in a phase 1 trial of a PSA DNA vaccine in patients with hormone-refractory prostate cancer. J Immunother. 2005;28:389–395. doi: 10.1097/01.cji.0000165353.19171.41. [DOI] [PubMed] [Google Scholar]
- 39.Waeckerle-Men Y, Uetz-von Allmen E, Fopp M, von Moos R, Bohme C, Schmid HP, Ackermann D, Cerny T, Ludewig B, Groettrup M, Gillessen S. Dendritic cell-based multi-epitope immunotherapy of hormone-refractory prostate carcinoma. Cancer Immunol Immunother. 2006;55:1524–1533. doi: 10.1007/s00262-006-0157-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Coulie PG, van der Bruggen P. T-cell responses of vaccinated cancer patients. Curr Opin Immunol. 2003;15:131–137. doi: 10.1016/S0952-7915(03)00009-8. [DOI] [PubMed] [Google Scholar]
- 41.Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–258. doi: 10.1038/nri2274. [DOI] [PubMed] [Google Scholar]
- 42.Nagorsen D, Scheibenbogen C, Marincola FM, Letsch A, Keilholz U. Natural T cell immunity against cancer. Clin Cancer Res. 2003;9:4296–4303. [PubMed] [Google Scholar]
- 43.Nagorsen D, Scheibenbogen C, Schaller G, Leigh B, Schmittel A, Letsch A, Thiel E, Keilholz U. Differences in T-cell immunity toward tumor-associated antigens in colorectal cancer and breast cancer patients. Int J Cancer. 2003;105:221–225. doi: 10.1002/ijc.11052. [DOI] [PubMed] [Google Scholar]



