Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 May 15.
Published in final edited form as: Clin Cancer Res. 2011 May 10;17(10):3064–3076. doi: 10.1158/1078-0432.CCR-10-2234

Recommendations from the iSBTc-SITC/FDA/NCI Workshop on Immunotherapy Biomarkers

Lisa H Butterfield 1, A Karolina Palucka 2,3, Cedrik M Britten 4,5, Madhav V Dhodapkar 6, Leif Håkansson 7, Sylvia Janetzki 8, Yutaka Kawakami 9, Thomas-Oliver Kleen 10, Peter P Lee 11, Cristina Maccalli 12, Holden T Maecker 13, Vernon C Maino 14, Michele Maio 15, Anatoli Malyguine 16, Giuseppe Masucci 17, Graham Pawelec 18, Douglas M Potter 19, Licia Rivoltini 20, Lupe G Salazar 21, Dolores J Schendel 22, Craig L Slingluff Jr 23, Wenru Song 24, David F Stroncek 25, Hideaki Tahara 26, Magdalena Thurin 27, Giorgio Trinchieri 28, Sjoerd H van Der Burg 29, Theresa L Whiteside 30, Jon M Wigginton 31, Francesco Marincola 32, Samir Khleif 33, Bernard A Fox 34, Mary L Disis 21
PMCID: PMC3096674  NIHMSID: NIHMS271139  PMID: 21558394

Abstract

Purpose

To facilitate development of innovative immunotherapy approaches, especially for treatment concepts exploiting the potential benefits of personalized therapy, there is a need to develop and validate tools to identify patients who can benefit from immunotherapy. Despite substantial effort, we do not yet know which parameters of anti-tumor immunity to measure and which assays are optimal for those measurements.

Experimental Design

The iSBTc-SITC, FDA and NCI partnered to address these issues for immunotherapy of cancer. Here, we review the major challenges, give examples of approaches and solutions and present our recommendations.

Results and Conclusions

While specific immune parameters and assays are not yet validated, we recommend following standardized (accurate, precise and reproducible) protocols and use of functional assays for the primary immunologic readouts of a trial; consideration of central laboratories for immune monitoring of large, multi-institutional trials; and standardized testing of several phenotypic and functional potential potency assays specific to any cellular product. When reporting results, the full QA/QC performed, selected examples of truly representative raw data and assay performance characteristics should be included. Lastly, to promote broader analysis of multiple aspects of immunity, and gather data on variability, we recommend that in addition to cells and serum, that RNA and DNA samples be banked (under standardized conditions) for later testing. We also recommend that sufficient blood be drawn to allow for planned testing of the primary hypothesis being addressed in the trial, and that additional baseline and post-treatment blood is banked for testing novel hypotheses (or generating new hypotheses) that arise in the field.

Keywords: Immunotherapy, Immune Monitoring, Vaccines, iSBTc, SITC

Introduction

Immunotherapy of cancer is becoming increasingly utilized and is becoming a part of cancer management alongside more classical approaches such as chemotherapy or radiotherapy. The US FDA has recently granted full approval for the first therapeutic cancer vaccine “Provenge” (1) based on a randomized phase III trial (2); and other recent trials have reached their primary endpoints according to the investigators (35). However, challenges exist that need to be resolved to facilitate development of innovative approaches, especially for treatment concepts exploiting the potential benefits of personalized therapy. There is a need for development and validation of tools to identify patients who can benefit from a particular form of immunotherapy. For example, only a fraction of patients are eligible for adoptive tumor-infiltrating lymphocyte (TIL) cell transfer (6), only a fraction of patients can achieve durable regressions in response to cell or antigen vaccination (7), or antibody therapies, and we do not know the mechanisms responsible. Despite substantial efforts from many groups, we do not know which parameters of immune responses, and which assays used to assess these parameters, are optimal for efficacy analysis. Indeed, the tumor-specific cellular immune response promoted by immunization often has not correlated with clinical cancer regression despite the induced cytotoxic T cells detected in in vitro assays (811). The major reason is that objective clinical response rates are usually below 10%, preventing meaningful correlations of specific T cell response rates with clinical responses in small sized, early stage trials. Other possible reasons are:

  • A lack of agreement over which immunological parameter to measure, what time point(s) is relevant, and which assay to use to make that measurement.

  • Immunological assays used may not accurately reflect an in vivo immune response, for example cytotoxicity assays with lengthy in vitro stimulation.

  • The analysis of single parameters alone may not provide sufficient insights about complex immune system-tumor interactions.

  • Common immunoassays do not take into account changes: in the differentiation of immune cells, in the antigenic profile of tumors and responding T cells, in T cell homing receptors or the complex analysis of “private” antigen responses.

Several key issues must be addressed to move the field forward

  • How to ensure the quality of sample processing and storage for the future studies?

  • How to standardize the assays and which assays should we standardize?

  • How to link immune and clinical outcomes with cellular product characteristics?

  • How to utilize the existing knowledge and assays to allow for meta-analyses of laboratory and clinical results?

Following discussions between representatives of the FDA leadership, the NCI, the NIH and Industry, as well as experts in the field of immunotherapy, the iSBTc-SITC/NCI/FDA Taskforce on immunotherapy biomarkers was created. This group led a workshop held on October 28, 2009, in conjunction with the Annual Meeting of the International Society for Biological Therapy of Cancer; iSBTc (now known as the Society for Immunotherapy of Cancer, SITC).. This workshop was also a follow up to the 2001 Workshop of the iSBTc-SITC (12) and included participation from 6 partner societies and representation of 20 countries. The results of the discussions of the Taskforce and recommendations from the Workshop participants and from representatives of several international immunotherapy societies are presented here.

Background

The major goal for the field of immunotherapy is to improve the clinical efficacy of immune-based therapies. To do so, we require immunologic biomarkers of efficacy. The NCI Translational Research Working Group (TRWG) has incorporated the need for these biomarkers in their developmental pathways, which are frameworks for bench to bedside development of new therapies and which include a pathway for “Immune Response Modifiers” (13). The FDA pharmacogenomics guidance (14) defines a valid biomarker as “a biomarker that is measured in an analytical test system with well-established performance characteristics and for which there is an established scientific framework or body of evidence that elucidates the physiologic, toxicologic, pharmacologic, or clinical significance of the test results.” This clearly states the need for biomarker assay standardization and also implies the inherent complexity in accomplishing this goal in the field of immunotherapy of cancer.

The “Critical Path” is the FDA's initiative to identify and prioritize the most pressing medical product development problems and the greatest opportunities for rapid improvement in public health benefits (15). Its primary purpose is to ensure that basic scientific discoveries translate more rapidly into new and better medical treatments by creating new tools to find answers about how the safety and effectiveness of new medical products can be demonstrated in faster timeframes with more certainty and at lower costs. The Critical Path has Six Areas of Focus (including: biomarker development and product manufacturing) and the Critical Areas for Biomarker Development are described as: Biospecimens, Analytical Performance, Standardization and Harmonization, Bioinformatics, Collaboration and Data Sharing, Stakeholder Education and Communication, Regulatory Issues and Science Policy. The importance of biomarkers is clear. Good biomarkers offer the prospect for earlier diagnosis, focusing expensive and invasive therapies on the right populations, monitoring disease progression and therapeutic benefits as well as facilitating drug development and discovery; clearly many more roles than just surrogate endpoints. Guidelines for incorporation of biomarker studies in early clinical trials of novel agents have been published from members of the Biomarkers Taskforce of the NCI Investigational Drug Steering Committee (16). The work of the Immunotherapy Biomarkers Taskforce is addressing several challenges specific to immune-based therapies, as follows:

  1. Processing and storage of blood samples to bank PBMC and serum for immunologic studies.

  2. Characterization of cellular products for therapy

  3. Assay standardization and harmonization before testing patient samples

  4. Centralization of immunological monitoring

  5. Standardized (or standardizable) assays which should be used for clinical trial antitumor immune response determination

  6. How assay data should be analyzed for “responder” and “non-responder” identification

  7. Reporting immunological monitoring data in publications

  8. Validation of specific assays and/or analytes as biomarkers of clinical response

  9. Novel assays in development for immunological testing of patients

Challenges, Examples and Recommendations

1. Processing and storage of blood samples to bank PBMC and serum for immunologic studies

Challenge

Multiple variables in drawing blood and obtaining tissue can impact the quality of the cells obtained and their proper testing.

Examples

This area has been addressed in depth by the Immunologic Monitoring Consortium (investigators from the University of Washington, Duke, BD Biosciences and Coulter), and their cryopreservation and thawing recommendations are outlined in Appendix 1A. In addition, recent studies have tested the importance of time from blood draw to PBMC processing, and established that the shorter the time, the superior the viability and functionality of the PBMC (17, 18). The use of CPT versus Ficoll for PBMC separation, and shipping of CPT tubes (BD Vacutainer) versus shipping whole blood has been investigated. There are data to suggest that CPT can perform equivalently to Ficoll (19), but it remains unclear whether shipping spun CPT tubes is superior to shipping heparinized whole blood. In order to allow for multi-institutional (and multi-continent) trials with minimal blood sample function loss, the AIDS Network has established an Immunology Quality Assessment (IQA) Program to evaluate and enhance the comparability of immunological laboratories handling blood samples from patients (seeAppendix 1B and #4, centralized laboratories). This approach could be of great benefit to the cancer immunotherapy community.

An additional critical issue is the volume of blood collected (20). Ideally, banking samples for future analyses using newly-developed techniques would potentially allow a better understanding of the mechanisms of response or exploration of novel prognostic biomarkers. In a study of 416 blood draws each aimed at taking 250 cc of blood, a median of 200cc was actually collected in patients with Stage III or IV breast cancer. The hematocrit of these patients was not significantly decreased during the time of these blood draws, data which may facilitate IRB approvals of larger volume blood draws (20). PBMC samples stored for extended periods are being tested for function (21)Appendix 1C).

Recommendations

We recommend following standardized processing, cryopreservation, storage and thawing protocols already tested by the Immunologic Monitoring Consortium, or testing the same parameters in your own laboratory and stating the extent of standardization in the associated publications. Consider drawing large (200–250cc) volumes or performing pre- and post-treatment aphereses, to allow for broad assessment of multiple immune parameters, including cells and serum and/or plasma.

2. Characterization of cellular products for therapy

Challenge

A wide variety of cellular products are being tested for therapy of cancer, from minimally manipulated autologous blood products, to cultured cell lines, and antigen loaded, matured dendritic cells (DC) (22). These are required to undergo FDA-mandated tests before release and administration (21 CFR 211.65). Some are relatively straightforward (safety, identity, purity) and others are more complex (potency (developed in phase I–II, to be utilized for phase III)), stability (acceptable conditions for both short- and long-term storage), consistency (batch to batch comparability). Products that do not meet the pre-specified release criteria must not be administered. Autologous products can be highly variable between patients and are challenging to characterize and standardize, and such variability, often minimally characterized, can impact immune biomarkers.

Examples

We have included an example of the testing (both exploratory and for product release) performed for a current autologous DC-based vaccine clinical trial (inAppendix 2). The methods for testing safety are well standardized. Measures of identity and purity are necessarily specific to the product, and are generally flow cytometry-based. These might include lineage, activation and differentiation markers. Potency assays remain exploratory to date, and include testing cell surface and intracellular proteins, cytokines and chemokines produced, and activation of target cells (i.e. lymphocyte proliferation stimulated by DC vaccines; killing by NK cells or T cells).

Two examples of candidate potency assays for antigen presenting cells (APC) are CD54 expression (23) and IL-12p70 production (24). CD54 upregulation on the vaccine cells seemed to correlate with overall survival in two phase 3 clinical trials (25). Spontaneous and induced IL-12p70 secretion assays have been standardized (26) and data are now being collected from multiple ongoing DC-based vaccine trials to determine if this functional readout correlates with clinical outcome and could become validated as a potency assay. No potency assays have yet been validated, and even DC, which have been tested in many different clinical trials, are sufficiently heterogeneous that each modification in antigen loading and maturation may result in a different functional profile; hence these assays must still be considered exploratory.

Recommendations

Standardize and utilize multiple phenotypic and functional assay parameters specific to the cellular product. In addition to safety, purity and descriptive identity testing for product release, development of candidate potency assays should begin early in clinical testing. Readers are encouraged to refer to FDA's Draft Guidance for Industry (released in Oct. 2008) Potency Tests for Cellular and Gene Therapy Products (27).

3. Assay standardization and harmonization before testing patient samples

Challenge

Patient blood samples are hard-won resources for understanding the effects of immunotherapy, and are often extremely limited. Therefore, the primary immune response assays performed using them must be robust and standardized before use in the context of a clinical trial.

Examples

Preclinical and clinical development phases of immunologic biomarkers have been outlined to clarify different stages involved (28). Assay standardization is described in CLIA Guidelines (seeAppendix 3; (29)) and The International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH which brings together the regulatory authorities of Europe, Japan and the United States and the pharmaceutical industry to discuss scientific and technical aspects of product registration) (30). The general standardization requirements include quality sample processing and storage (see topic #1, above), an SOP for each sample type which maintains sample integrity, and a QC program for each sample type. SOPs need to be established for each component of the testing process, including sample processing, the immune assay, and analysis. For several common cellular immune biomarker assays (cytokine flow cytometry, MHC tetramers and ELISPOT) a detailed standard procedure is available (31) and suggestions for standardization is included.

Assay standardization and validation also involve technical performance characteristics of the assay (e.g., reproducibility intra-day and inter-day, overall precision and accuracy, specificity, sensitivity, assay range (16)). Criteria for analytical performance of the assay are: the accuracy, precision, reportable range, reference ranges/intervals (normal values), turn-around time and failure rate of the assay as it is to be performed in the trial. Limits of acceptable performance, and the quality control data should be obtained. Data on positive and negative controls, calibrators, and reference standards should be available. If the assay is to be performed at more than one site, inter-laboratory variability in the measurements needs to be assessed; sources of variation should be minimized to maintain performance at all sites within acceptable limits. Prospective evaluation of assay performance is often executed on a reference population. A bank of PBMC, serum or leukapheresis products is helpful as a source of material useful for performing quality assurance testing (32, 33). Commercially available ELISA or Luminex kits are generally well standardized for use. For cellular assays, an example is characterizing CMV-specific T cells responses in PBMC, which allows the identification of low (<0.5%), intermediate (0.5–2%) and high (>2%) level responses which could serve as a surrogate for the range of immunity that might be seen in an immunotherapy study (34).

Establishing clinical utility of the assay involves measuring the relevant response in immunotherapy trials, i.e., the magnitude of the association between the assay result and the immunological and clinical endpoint measured. Scoring procedures need to be established for quantitative, semi-quantitative or qualitative assays (see also topic #8, below).

In the last several years, harmonization efforts have been organized in order to understand and reduce variability in ELISPOT, ICS and MHC tetramer assays between multiple international laboratories. These efforts have involved large proficiency panel programs and include the option for each laboratory to use their own materials, reagents and protocols, to test centrally distributed, pretested PBMC and antigens, with general logistical guidelines to allow for comparison of results obtained from participating labs. These programs allow for the identification of protocol variables which may influence assay outcome. Panel findings have been summarized in harmonization guidelines. An important aspect of these harmonization efforts is that they do not require strict protocol adherence across all laboratories. Recent findings indicate that serum is not required for ex vivo IFN-gamma ELISPOT, according to collaborative studies of different protocols from the European CIMT Immunoguiding Program and the Cancer Immunotherapy Consortium of the Cancer Research Institute (33, 3538), and other international collaborative studies (32, 38).

The AIDS Consortium have been leaders in inter-laboratory proficiency testing and in developing programs which resolve variation between laboratories performing immune assays (seeAppendix 1B) (39). Technician understanding of procedures and training have been hurdles to inter-lab variation and are surmountable. Use of standard methods and reagents across laboratories also increases inter-lab comparability (40). Indeed, standardized training procedures and reagents allowed for highly concordant IFNγ ELISPOT results to be obtained across 7 international labs on 3 continents (41). In another inter-lab comparison study of the IFNγ ELISPOT, 11 assay “novices” were given identical cells, reagents and SOPs, and the results showed that even without prior experience, standardized procedures and reagents can yield reproducible results across labs, countries and continents (32). The tuberculosis community (including the WHO Initiative for Vaccine Research) has also been involved in ELISPOT assay and related cytokine assay standardization. (42, 43).

Recommendations

We support the use of standardized assays (following CLIA and ICH guidelines) with full disclosure of methodology specifics for the primary immunologic readouts of a trial. Standard protocols and critical assay parameters for several most commonly used assays (particularly for the ELISPOT) have been published (3242) and should be strongly considered for clinical trial immune response testing. Participation in external proficiency panels and use of pre-screened PBMC and other additional controls can be very important for comparing results between sites and potentially, between trials.

4. Centralization of immunological monitoring

Challenge

Clinical trial blood and tissue sample processing, cGMP cellular product production and immunologic monitoring assay standardization are extremely laborious and costly. To address these concerns, several institutions have invested in centralized laboratories.

Examples

The benefits of these labs include: high quality and reliability with QA/QC programs; state-of-the-art assay development, standardization and validation as well as decreased cost of immune monitoring (due to large discounted purchases, batched assay testing, previously developed assays and well-trained staff). In addition, assay consultation and result interpretation in conjunction with data analysis by biostatisticians can be available; and banks of samples for normal controls exist for comparisons and normal ranges. One drawback is timing for sample handling: shipping from a clinical site to a central lab necessitates a delay of up to 24 hr in sample processing, which may adversely affect PBMC functional responses as well as the expression of some labile markers on cells and in serum (also: (44) (e.g., CD62L). Standardized shipping conditions (i.e., materials, logistics, monitored temperature) can address some of the time-dependent alterations. A different approach is setting up a regional central laboratory network with centralized training and oversight (see AIDS network IQA inAppendix 1B).

The following are four examples of multiple centralized laboratories across the U.S. The Laboratory of Cell Mediated Immunity (LCMI), SAIC-Frederick, Inc. is a centralized contractor lab performing immunological monitoring for the NIH (A. Malyguine, Director). The lab is CLIA certified, and performs many assays, including modified ELISPOT assays including peptide, whole protein and tumor cells. Normal donors with known responses to recall antigens serve as positive controls, and new assay variations undergo optimization studies. Cellular Technology Limited (CTL) operates a GLP compliance and CLIA certified laboratory for specimen processing, storage and immune monitoring with an emphasis on ELISPOT. CTL was awarded multiple IDIQ contracts by the NIH for validation of T cell-based immune monitoring approaches and has served as ELISPOT reference laboratory to the Immune Tolerance Network (ITN), and as a central laboratory to the Cancer Vaccine Consortium (CVC). The ECOG Central Immunology Laboratory which operates at the University of Pittsburgh Cancer Institute (UPCI) (T.L. Whiteside and L.H. Butterfield, Directors), provides ECOG network investigators with processing, storage, and testing of specimens under CAP-inspected, CLIA compliant conditions. Samples are received via protocol-specific specimen shipping kits prepared by the laboratory for clinical sites. In E1696, a phase II multi-epitope peptide vaccine trial for patients with measurable metastatic melanoma, a significant difference was reported in overall survival of patients stratified by immune response status, with responders living longer than non-responders (median overall survival 21.3 vs. 10.8 months, p=0.033) (45). However, in a Cox model AJCC stage at diagnosis was the most significant predictor of overall survival (p=0.002) and immune response status trended to significance (p=0.073). Examples of % CV (coefficient of variation) and assay controls from ECOG 1696 ELISPOTs are listed in Appendix 4. Similarly, in an analysis of immune responses to a 12-peptide vaccine performed in the Human Immune Therapy Center at the University of Virginia (46), there was a significant correlation between ELISPOT reactivity and disease-free survival in a univariate analysis of 48 patients with resected Stage II-IV melanoma who received a multipeptide vaccine in the adjuvant setting. This analysis did not stratify for stage; so other factors could have contributed to this observed correlation. Lastly, in a trial testing a poly-epitope DNA prime/vaccinia boost vaccine in Stage III/IV melanoma patients, in which immune monitoring was centrally performed, a significant correltion was seen between MHC tetramer responses and median survival, and a trend towards correlation between IFN-γ ELISPOT response and median survival was seen (47).

Recommendations

We recommend consideration of central laboratories for immune monitoring, due to their experience in standardized assay conduct and existing infrastructure. In particular, they can be a critical part of larger scale, multi-center clinical trials for minimizing variation. It is recommended that central laboratories establish (1) the historical data on any specific standardized assay for the selected parameter as the reference, and (2) provide a service to conduct a comparability test to validate the data that are generated in other study sites with the reference data as the control if applicable.

5. Standardized (or standardizable) assays which should be used for clinical trial antitumor immune response determination

Challenges

There are many assays potentially capable of measuring aspects of immune function, and limited blood and tissue samples require choosing a limited number of possible assays. The field of immunologic monitoring is also constantly evolving. The small numbers of complete clinical responders, small scale trials and variability in assays chosen and assay conduct make identification of the crucial assay parameters to measure difficult to identify.

Examples

The choice of immune assay will, in large part, depend on the proposed mechanisms of action of the immune intervention. A vaccine designed to generate a specific antibody response, for example, would focus on the assessment of humoral immunity rather than a cellular response. To test specific immune effectors, there are many choices for the methods: ELISPOT, MHC-peptide multimer staining, intracellular cytokine staining, as well as soluble cytokines, NK cells, Th phenotype, etc. Not only the magnitude of the response and frequency of effector cells, but also the antigenic breadth and degree of multi-functionality have all been shown to be critical in model systems and specific clinical trials (48) and broad polyepitopic immune responses have been associated with complete clinical response in a small group of therapeutically vaccinated patients with vulvar intraepithelial neoplasia (49). In contrast, vaccine failure was associated with higher frequencies of disease-specific CD4+CD25+Foxp3-positive T cells and a low production of IFNγ by disease-specific T cells upon first vaccination (50). The need for several assays to explain the success and failure of this therapeutic vaccine illustrates the need for sufficient amounts of blood, taken at different time points.

The ELISPOT, perhaps the most thoroughly standardized assay to date (see #5 above), identifies the number of functional antigen-specific cells, multiple samples may be tested simultaneously, and it can be used for testing more than one analyte or function (51). It has been shown that two measures of cytotoxicity, the Granzyme B ELISPOT assay (52) and standard 51Cr release correlated better with each other than MHC tetramer or IFNg ELISPOT assays, in clinical setting (53). In addition, with automated analysis methods, great reproducibility and accuracy for detecting specific T cells can be achieved. An alternative functional assay is intracellular staining for cytokines or other effector molecules using flow cytometry. Multicolor analysis thus provided can complement ELISPOT, as it can provide additional information regarding multiple cytokines that are produced by specific surface-stained cell subsets. Correlation of tetramer assay results with in vitro cytotoxicity in clinical trial material has been previously observed (54). However, in a multi-center cooperative group trial, MHC tetramer frequencies and differentiation stage did not correlate with clinical outcome, but IFNg ELISPOT response did (Schaefer, submitted’ 10).

Another important issue is the target used in the assay. In two Phase II trials of vaccination with a cocktail of altered HLA-A2 tumor peptides in early melanoma (N=40) and prostate carcinoma (N=20) patients, the ex-vivo IFN-γ ELISPOT and HLA/peptide multimer staining showed a rapid induction of peptide-specific CD8+ T cells of the majority of vaccinated patients. However, clinical efficacy only correlated with significant anti-tumor cell activity in vitro. These data clearly stress the need for including tumor cell recognition when monitoring patients treated with tumor vaccines, especially if based on modified peptides, in order to gain better information about tumor reactivity (55, 56).

Recent flow cytometric measures of cell-mediated immunity can evaluate both the target cell death and effector cell function simultaneously, allowing for more efficient acquisition of both tumor target cell cytolysis and CTL activation. A flow cytometry-based cytotoxicity assay has been developed to simultaneously measure NK cell cytotoxicity and NK cell phenotype (57). Another cytotoxicity assay that has been optimized to utilize low numbers of antigen-specific T cells has been described (58) in which peptide/MHC multimer-positive CD8+ T-cells were purified, cloned, expanded and tested for CD107a cell surface expression and their cytotoxicity evaluated based on the frequency of dead cells in CMTMR-labeled target cell populations. This assay proved to be more sensitive than the 51Cr-release assay. Similarly, combining the measure of CD107a by CD8+ effector cells with the apoptosis marker Annexin V binding to target cells has been used (34, 59, 60).

There is increasing enthusiasm for poly-functional flow cytometry methods, which are already standardized in the HIV community. As discussed at the 2009 workshop, CMV, HIV, and cancer can all induce endogenous T cell responses of varying magnitudes; but only CMV responses tend to be protective. The T cell response signatures for CMV, HIV, and cancer may be very different: CMV elicits a relatively high proportion of IFNg+IL-2+ cells with heterogeneous phenotypes including many effectors. HIV elicits few CD8+IL-2+ T cells and intermediate phenotypes. Cancer patients may show low magnitude, IL-2+ but not IFNg+ T cells and central memory phenotype. The mechanisms leading to these different signatures need to be further elucidated (61).

It is essential to develop well-established quality control standards that can be made available among different laboratories for assay standardization to ensure assay consistency between sites. Minimally, a description of internal (e.g., positive antigen or peptide, negative controls, mitogens) and external controls need to be provided for each assay (see #8 below). The Taskforce members have been in discussions with the BRB of the DTP, NCI (Frederick, MD) about creation of a repository for assay standards. This may be feasible once sources for the agreed-upon standards are identified. Alternatively, commercial sources are currently available (SeraCare, Cellular Technology Limited, others). Another important aspect of standardization is testing the extent to which an assay can be run from batched cryopreserved samples, or whether only fresh samples yield reliable results. Lastly, how to balance standardized assays for immune responses (which allow the field to move forward by having some ability to compare trials) with research questions (which drive innovation and may identify novel biomarkers with greater specificity for clinical outcomes)? Larger volumes of blood drawn (without negative impact on the patient (#1, above)) may allow for this balance with institutional IRB approval. Alternatively, performing pre- and post-treatment aphereses provides cells for monitoring as well as a resource for research questions and assay development (62, 63).

Recommendations

We support choosing a standardized, functional assay as the primary readout of the immune response, which addresses the specific hypothesis being tested and proposed mechanism of action of the intervention. There are now strong data that testing multiple functional parameters (multiple cytokines, recognition of not only peptides, but also tumor cells) can yield important information.

6. How assay data should be analyzed for “responder” and “non-responder” identification

Challenge

While many assays yield data on changes in anti-tumor immunity, distinguishing assay variation from normal human variation and treatment-induced responses is not trivial.

Examples

All binary response definitions for individual patients are effectively “seat of pants”; statistical properties are unknown and in practice, unknowable—i.e., false positive and false negative rates are unknown. Use of a single standardized definition could allow the results of different studies to be compared (but will not solve this problem). At the UPCI, the Biostatistics Facility (W. Gooding) developed a definition for individual IFNγ ELISPOT response that has achieved a measure of success insofar as it has been demonstrated to correlate with clinical response (45). The definition assumes that there are 3 wells each for identical test samples (tst) and control samples (ctrl). The variable y is set equal to mean (tst)-mean (ctrl). If the number’s of cells is different for tst and ctrl, the tst counts are scaled by ratio of the number of tst cells to the number of ctrl cells, and y is set to 0 if the number of responding tst cells in any well is less than the number of responding control cells in any well. An individual’s response to treatment is then determined by the following criteria: y (post-treatment) divided by y (pre-treatment) must be greater than 2; and y (post-treatment) must be greater than 10. This definition is essentially based on a factor of 2-fold increase in post- over pre-treatment background-corrected ELISPOT counts, albeit with considerable protection against false-positives due to small counts. This definition has also been employed in other multicenter trials (64), and an adjustment of it for use with stimulated ELISPOT assays has been shown to have low false positive results, and to correlate with clinical outcome (46). It must be kept in mind that both true immune response and clinical response are continuous, not binary, variables. Different definitions of binary response for individual patients are arbitrary and will correlate differently with clinical outcome. Thus, the relationship of degree of immune response with the degree of clinical outcome might be used to refine binary response criteria. Moodie et al (65) have recently compared response definitions for ELISPOT assays. Although they define response in terms of a comparison of test and control samples, the methods that they describe could generally be adapted to a comparison of pre- and post-treatment samples.

Recommendations

Obtain multiple pre-therapy samples (at different times) for analysis; these can be used to assess pre- therapy variability of the biomarker level. Tighten response criteria: require positive responses at two consecutive post-therapy time points; this is useful for limiting post-therapy variability. Consider using clinical response to refine the definition of immune response. When immune response is the primary outcome of interest in a trial, use non-parametric techniques (such as the Wilcoxon signed-ranks test) to assess response of the entire sample of patents as a group (49). This avoids the problems associated with attempting to define individual responses. To date, no one knows how big an absolute or relative increase in the frequency of antigen-specific T cells between two time points should be to be considered a biologically relevant response ((66) for empirical rules).

7. Reporting immunological monitoring data in publications

Challenge

Evaluation of immunological monitoring studies is not possible without disclosure of the details of the methods that are known to influence test outcome variability, the assay conduct and its interpretation.

Examples

Reporting of complex data sets is a challenge, as different styles exist that limit comparability. The concept of “minimal information” projects and structured reporting of data sets was pioneered by A. Brazama (67). The concept has been adapted for T cell assays (68) and multiple minimal information (MI) projects with overlap now exist. The REMARK criteria have become the standard for publication of prognostic tumor marker studies (69). The REMARK recommendations clearly outline what should be reported to interpret patient selection, sample storage, assay performance, and critical statistical analysis. These recommendations are accepted by most major journals including those published by ASCO and AACR, yet few immune-based studies adhere to them (69). In the recent FDA draft guidance for therapeutic cancer vaccines (Sept. 2009), recommendations include consideration of performing at least two assays and that all assay parameters and controls should be clearly described (7078). It is recommended that investigators make themselves familiar with these different projects in order to choose the appropriate guidelines for the assays chosen.

Recommendations

We suggest that the following study aspects should always be included when reporting results, independent of other applicable guidelines: the QA/QC performed, reference populations included, all testing of reagents and controls, at least some selected examples of truly representative raw data and the assay performance characteristics. These parameters will allow appropriate reviewer and reader evaluation of the quality and potential impact of the data.

8. Validation of specific assays and/or analytes as biomarkers of clinical response

Challenge

When data from basic and translational research settings (and exploratory testing in Phase I trials) suggest that a specific immune assay or other biomarker correlates with clinical outcome, standardization and validation is needed to substantiate those data and to allow possible comparisons between treatments and trials.

Examples

GLP guidelines from the FDA for general lab conduct for assay performance are available (79). For the ELISPOT assay, validation of the assay has been addressed (80). A recent report (81) has described many aspects of standardization of MHC tetramer, IFNg ELISPOT and IFNγ real-time PCR, and the authors also attempted to validate the assays for determining the absolute frequency of antigen-specific T cells. They concluded that this could not be accomplished without a “gold standard” measure for such cells.

Quantification of polyfunctional cytokine-producing T cells (often IFNγ, IL-2 and TNF) by multi-color flow cytometry is being used in infectious disease models and HIV patients for correlation with clinical outcome (60, 82). This is an immunological readout which may also serve as a biomarker for clinical response, and there is already standardized shared software for analysis of the flow cytometry data (SPICE, M. Roederer, VRC, NIAID, NIH). Peptide pools are commonly paired as antigen sources for this assay, and they are evaluated by another shared software package (Deconvolute This) (83).

Conventional response criteria may not adequately assess the activity of immunotherapeutic agents. Therefore, systemic criteria, immune-related response criteria (irRC), have been defined to capture relevant clinical response patterns observed in melanoma patients undergoing immune therapy. Use of irRC may allow for improved comprehensive evaluation of immunotherapeutic agents in clinical trials and potentially offer guidance in clinical care, as well as being a more appropriate comparator for correlation with in vitro measures of antitumor immunity (84). Consideration of the time required for evolution of immune responses may require collection of patient blood samples over a longer time period.

Recommendations

The immunotherapy field continues to produce novel data from immune assessments in patients which correlate to clinical outcome in different diseases and treatment settings. These candidate biomarkers should first be standardized (85) and then validated by other investigators. The evolution of anti-tumor immunity may necessitate longer term immunologic monitoring.

9. Novel assays in development for immunological testing of patients

Challenge

In order to move towards more sensitive, high throughput evaluations, there must first be quality sample acquisition for analysis and hypothesis testing. Also, it is not only immunotherapy trials which must be evaluated. Most biologic agents used singly or in combination with conventional drugs for cancer therapy engage the immune system. Other biologic agents that specifically target growth factor receptors, blood vessels/endothelial cells, tumor cells, or tumor-associated antigens often involve immunologic mechanisms.

Examples

Directly assaying the tumor environment, performing expression arrays (from the tumor), testing for determinant/epitope spreading, and testing genetic aspects of the host (SNPs, GWAS, HLA) are not yet commonly performed. One example of a large scale initiative to analyze patient tumors for individual gene expression patterns is “M2Gen,” a research collaboration between H. Lee Moffitt Cancer Center and Merck & Co. Researchers are collecting tumor tissues from patients to identify the biological markers unique to each tumor (86).

Immune profiling can include: high throughput molecular profiling platforms to study the human immune system, polychromatic flow cytometry, RNA profiling (mRNA, miRNA, RNAseq), SNP arrays (soon genome sequence), multiplex serum chemokines, cytokines profiles, protein arrays and peptide arrays (87) for serologic responses, mapping antigenic repertoire and semi-quantitative immunohistochemistry of the tumor (88). In addition to the positive effects of antitumor effector activation, critical aspects of tumor immune suppression should be investigated: the frequency and function of MDSC and Treg, functional defects in TIL and in circulating immune cells, cytokine imbalance (Th2 vs. Th1), failure to generate central memory T cells, persistent activation of T cells, spontaneous apoptosis of T cells, T cell senescence, and presence of soluble factors in serum that induce death in immune cells. All of these immune deficits have been reported, but are not yet part of the regular immune monitoring repertoire. This is crucial for understanding why some approaches are not successful, and for personalized selection of available anti-cancer therapies in the future.

An important aspect of these broad assessments of immunity, particularly with newer, high throughput approaches, is data management (89). Currently data are often stored in multiple clinical and laboratory databases requiring manual data entry and coordination. Informatics must address: the ability to integrate data from multiple technology platforms, the ability to integrate clinical and biomarker data from multiple projects, and include an emphasis on data dissemination/high availability (to allow for downstream analyses by biostat/bioinformatics teams; for access/query by investigators; and ultimately to promote insight, sharing data with study participants, collaborators, consortia members, scientific community and streamline data export to public repositories). A goal inherent to this is defining a universal data element set to accompany all high-quality biospecimens.

Recommendations

We recommend that both RNA and DNA samples as well as sera and plasma be banked under standardized conditions for later testing in multiplex, molecular assays (from blood and the tumor, and to study the microenvironment). Improved collection of tumor and TIL are crucial for understanding the impact of different therapeutic approaches. We also reiterate that sufficient blood be drawn to allow for the planned testing of the primary hypothesis being investigated in the trial, such that additional baseline and post-treatment blood is banked for testing novel hypotheses (or generating new hypotheses) that arise in the field during the time required for trial design, approvals, enrollment and conclusion.

Discussion

Immunotherapy clinical trials can only benefit from careful study of the effects on patient immune responses and the state of immune function (and dysfunction). Because of the large variation between patients, elimination of as much variation as possible in procedures used for handling blood and tumor specimens, and in procedures for assays, is essential. Equally essential is the thorough reporting of the level of standardization and the specific methods used for specimens, assays and analysis. We are also recommending an increased level of banking of diverse biologic specimens for unspecified future research. We recognize that implementation of this goal will require discussion and cooperation between human subjects’ protection committees and patient advocate representatives along with researchers and clinicians.

As a service to the community, in addition to the present report, we propose the following next steps:

  1. Specific SOP recommendations (many of which have been standardized and some of which are published) with links to be posted on the iSBTc-SITC website for easy access (and also to other society web sites).

  2. Promoting greater focus on standardization and validation assay guidelines (CLSI, CLIA, ICH)

  3. Improved data reporting: immune biomarker studies should adhere to published guidelines, especially those that are advocated by the journals to which the papers are submitted such as REMARK. Many efforts are ongoing to refine these recommendations: MIFlowCyt, MIACA, MIATA, MIBBI, that will help to select the “MI” projects most applicable to a specific scientific question or setting.

  4. Further discussion at the follow-up workshop: “Symposium on Immuno-Oncology Biomarkers, 2010 and Beyond: Perspectives from the iSBTc-SITC Biomarker Task Force,” Masur Auditorium on the NIH campus, Building 10, Clinical Center, Bethesda, MD, September 30th, 2010 (90).

  5. Strengthening of inter-immunotherapy society communication and collaborations (M.L. Disis, B.A. Fox, manuscript in preparation, 2010)

While not directly in the hands of individual investigators and smaller teams, we also recommend greater funding levels to support the acquisition of blood and tumor samples for embedded correlative studies as well as unspecified banking for future analysis. It is only with resources of tumor, serum/plasma, PBMC, DNA & RNA that we will be able to learn as much as possible about the state of immunity in cancer patients, the positive effects of our interventions and the inhibitory effects of tumor progression that we have yet to overcome. These freshly tested and banked samples, collected and assayed under standardized conditions, will also be crucial in allowing us to better understand patient-to-patient variability and take steps towards more effective and personalized approaches.

Statement of Translational Relevance.

Progress in the field of immunotherapy has been slow, but recent clinical successes have given strong support to the potential of this approach as a treatment modality in cancer. In order to define biomarkers to identify patients who will have clinical benefit, and to ultimately identify appropriate patient groups to enroll, clinical trials testing immune-based interventions should perform more thorough and standardized immunologic assays to fully study clinical responders and non-responders, and report data and its analysis methodology in greater detail. An increased focus on immune assessments in these patients will allow us to learn more about the mechanisms of action of the tested interventions and the positive and negative immune responses in treated patients.

Acknowledgements

We thank Dr. Raj Puri, (Director, Division of Cellular and Gene Therapies, Office of Cellular, Tissue and Gene Therapies, FDA/Center for Biologics Evaluation and Research), the FDA liaison to iSBTc-SITC, for providing critical critique of the manuscript. We also thank the staff of iSBTc-SITC for production and technical assistance in the manuscript preparation: Tara Withington, CAE, Executive Director; Angela Kilbert, Director of Administration; Chloe Surinak, Project Manager; Roseann Marotz, Meetings Manager; and Jimmy Balwit, Scientific Communications.

Grant Support:

NIH grants P50 CA121973 and RO1 CA104524 to LH Butterfield; NIH grants RO1 CA119123 and R21 CA123864 to BA Fox; research grant (“Standardization of Immune Monitoring”) to CM Britten and SH van der Burg from the Wallace Coulter Foundation, Florida USA.

Appendix 1A

Sample Collection, cryopreservation and thawing (Lymphocytes/PBMC)

Factors That Did Not Matter

  • Shipping on dry ice, 24, 48, 72 hours vs. liquid nitrogen (p>0.05)

  • Large (100cc) vs. small volume (25cc) dilution for thaw (p>0.05)

  • Time (5–10 min.) and speed of spin at wash, 1200–1500 rpm, (p>0.05)

  • # cells/vial (1, 2, 3×10e7) (p>0.05)

Factors that did matter: the thawing method. Additives like human serum albumin, dextran and FBS were superior to human AB serum; washing thawed cells in medium pre-warmed to 25°C to 37°C was superior to chilled (4°C) medium (91).

Open Access Protocols are available from the Immunologic Monitoring Consortium (91, 92)

Appendix 1B

Examples from the AIDS Network

Immunology Quality Assessment (IQA) Program: The IQA is a resource designed to help immunologists evaluate and enhance the integrity and comparability of immunological laboratory determinations performed on patients enrolled in multi-site HIV/AIDS investigations.

  • ~83 Participating Laboratories

  • 6 test shipments per year

  • The viability of PBMC and the viable yield before freezing and after thawing is tested

  • IQA reviews statistical report and identifies sites that are having difficulty performing assays

  • Poor performers are contacted to discuss specific problems

  • Laboratories fax histogram results to the IQA for central review

Laboratories who serve both the Aids Clinical Trials Group (ACTG) and IMPAACT trials must conform to the stringent criteria of the ACTG (80% viability and 80% viable cell recovery of PBMC) (93) (94)

Appendix 1C

Large volume blood samples from cancer patients (median volume collected: 200cc; median yield: 0.8 × 10e6 PBMC/ml) (21)

Validation in Stored Samples

Ongoing Performance

Analysis of 80 samples

Sample age range: 30–2500 days, av. 600

Median recovery: 70%

Mean recovery: 70%

Range: 22–130%

Appendix 2

Cellular Product minimal safety, purity and identity tests

The following is an example of the specific release tests which are required by the FDA for early phase trials involving autologous, in vitro manipulated cellular products (in this example, DC). This example also shows the identity/purity testing chosen for this type of product, and the candidate potency test being performed.

Viability

The cells are counted by microscopic observation on a hemacytometer, and a differential count (DC vs. lymphocytes) is obtained using trypan blue dye. Minimum 70% viability.

Purity

The DC must express MHC class II and CD86 by flow cytometry in a minimum of 70% of the cells. Additional phenotyping (MHC class I, CD80, CD83, CCR7, others) is performed to fully characterize the DC, and is for research proposes.

Sterility

DC are tested by bacterial (aerobic and anaerobic) and fungal cultures at the Clinical Microbiology Laboratory. Final results of the microbial cultures are available in 14 days. Prior to release of the DC for vaccine use, a standard gram stain is performed and must be negative for the presence of microorganisms.

Mycoplasma testing of cell suspensions (not supernatants) is performed using a rapid detection system, based on nucleic acid hybridization or by PCR. The cell preparation must be negative for mycoplasma.

Endotoxin testing is performed on the cell culture at the time of harvest and prior to release of the final product. The acceptable endotoxin level is <5 EU/kg of body weight per dose.

Potency

To define a measure of potency for the DC, we determine their ability to produce IL-12p70 and IL-10 by Luminex assay (26). This test is performed batched, with and without activation by CD40L and/or LPS, and is available several weeks after vaccine injection. Data will be correlated with measures of DC phenotype and clinical outcome.

Additionally, a 0.5 ml sample of the final DC preparation from each vaccination time is cryopreserved for possible ancillary testing in the future. These samples are stored a minimum of one year after vaccine administration.

Appendix 3

CLIA (Clinical Laboratory Improvements Amendments) (29)

Test Accuracy (close agreement to the true value),

Precision (agreement of independent results: same day, different day),

Reproducibility (intra-assay and inter-assay)

Reportable range (limits of detection)

Normal ranges (pools of healthy donors, accumulated patient samples: test at least 20, include a banked healthy donor control in patient assays),

Personnel competency testing (minimally, annually)

Equipment validation, monitoring

Reagent tracking

Appendix 4

Preliminary examples from the ECOG Central Immunology Laboratory ELISPOT assays of large scale, multi-institution clinical trials (n = 20 from each trial, from >70 (E1696) and >200 (E4697) patient ELISPOT assays):

E4697 (2008–2009)
spontaneous PMA/I (+)/OKT3
Healthy control ave.: 4.9 (54%CV) 304 (19.2%CV intra-assay)
(48% CV inter-assay)
Patient ave.: 0.7 (35%CV) 81 (38.7 %CV)
E1696 (2002–2003)
spontaneous PMA/I (+)/PHA
Healthy control ave.: 5.4 (56%CV) 284 (15.5%CV intra-assay)
(51% CV inter-assay)
Patient ave.: 19 (40%CV) 171 (18.8 %CV)

References

  • 1.Dendreon. www.dendreon.com. [cited; Available from: http://investor.dendreon.com/phoenix.zhtml?c=120739&p=irol-newsArticle&ID=1398598&highlight=
  • 2.Kantoff PWHC, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Drecier R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF. Sepuleucel-T immunotherapy for castration-resistant prostate cancer. New England Journal of Medicine. 2010;363:411–481. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 3. http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=65&abstractID=32035.
  • 4.Schwartzentruber DJ, Lawson D, Richards J, Conry RM, Miller D, Triesman J, Gailani F, Riley LB, Vena D, Hwu P. A phase III multi-institutional randomized study of immunization with the gp100:209-217 (210M) peptide followed by high-dose IL-2 compared with high-dose IL-2 alone in patients with metastatic melanoma. J Clin Oncol. 2009;27:18s. [Google Scholar]
  • 5.Schuster SJ, Neelapu SS, Gause BL, Muggia FM, Gockerman JP, Sotomayor EM, Winter JN, Flowers CR, Stergiou AM, Kwak LW. Idiotype vaccine therapy (BiovaxID) in follicular lymphoma in first complete remission: Phase III clinical trial results. ASCO Meeting Abstract. 2009 [Google Scholar]
  • 6.Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–129. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8:299–308. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lee KH, Wang E, Nielsen MB, et al. Increased vaccine-specific T cell frequency after peptide-based vaccination correlates with increased susceptibility to in vitro stimulation but does not lead to tumor regression. J Immunol. 1999;163:6292–6300. [PubMed] [Google Scholar]
  • 9.Butterfield LH, Comin-Anduix B, Vujanovic L, et al. 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]
  • 10.Butterfield LH, Ribas A, Dissette VB, et al. Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res. 2003;9:998–1008. [PubMed] [Google Scholar]
  • 11.Ribas A, Glaspy JA, Lee Y, et al. Role of dendritic cell phenotype, determinant spreading, and negative costimulatory blockade in dendritic cell-based melanoma immunotherapy. J Immunother. 2004;27:354–367. doi: 10.1097/00002371-200409000-00004. [DOI] [PubMed] [Google Scholar]
  • 12.Keilholz U, Weber J, Finke JH, et al. 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]
  • 13. ( http://www.cancer.gov/images/trwg/immune_oct08.pdf.
  • 14. ( http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm079855.pdf).
  • 15. http://www.fda.gov/ScienceResearch/SpecialTopics/CriticalPathInitiative/default.htm.
  • 16.Dancey JE, Dobbin KK, Groshen S, et al. Guidelines for the development and incorporation of biomarker studies in early clinical trials of novel agents. Clin Cancer Res. 16:1745–1755. doi: 10.1158/1078-0432.CCR-09-2167. [DOI] [PubMed] [Google Scholar]
  • 17.Bull M, Lee D, Stucky J, et al. Defining blood processing parameters for optimal detection of cryopreserved antigen-specific responses for HIV vaccine trials. J Immunol Methods. 2007;322:57–69. doi: 10.1016/j.jim.2007.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kierstead LS, Dubey S, Meyer B, et al. Enhanced rates and magnitude of immune responses detected against an HIV vaccine: effect of using an optimized process for isolating PBMC. AIDS Res Hum Retroviruses. 2007;23:86–92. doi: 10.1089/aid.2006.0129. [DOI] [PubMed] [Google Scholar]
  • 19.Ruitenberg JJ, Mulder CB, Maino VC, Landay AL, Ghanekar SA. VACUTAINER CPT and Ficoll density gradient separation perform equivalently in maintaining the quality and function of PBMC from HIV seropositive blood samples. BMC Immunol. 2006;7:11. doi: 10.1186/1471-2172-7-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. http://www.irb.pitt.edu/irbmailings/listbot6-20-05.htm.
  • 21.Disis ML, Dang Y, Slota MS, Childs J, Higgins D, Bates N, Lu H, O'Byrne B, Jackson E, Salazar LG, Clay T, Maino V, Ghanekar S, Maecker H. Members of the Immunologic Monitoring Consortium. 2009 http://www.isbtc.org/meetings/access.php?content=workshop09pres&loc=/meetings/am09/workshop09/presentations/Disis_915am.pdf.
  • 22.Nicolette CA, Healey D, Tcherepanova I, et al. Dendritic cells for active immunotherapy: optimizing design and manufacture in order to develop commercially and clinically viable products. Vaccine. 2007;25 Suppl 2:B47–B60. doi: 10.1016/j.vaccine.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 23.Sheikh NA, Jones LA. CD54 is a surrogate marker of antigen presenting cell activation. Cancer Immunol Immunother. 2008;57:1381–1390. doi: 10.1007/s00262-008-0474-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mailliard RB, Wankowicz-Kalinska A, Cai Q, et al. alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64:5934–5937. doi: 10.1158/0008-5472.CAN-04-1261. [DOI] [PubMed] [Google Scholar]
  • 25.Higano CS, Schellhammer PF, Small EJ, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115:3670–3679. doi: 10.1002/cncr.24429. [DOI] [PubMed] [Google Scholar]
  • 26.Butterfield LH, Gooding W, Whiteside TL. Development of a potency assay for human dendritic cells: IL-12p70 production. J Immunother. 2008;31:89–100. doi: 10.1097/CJI.0b013e318158fce0. [DOI] [PubMed] [Google Scholar]
  • 27.Draft Guidance for Industry-Potency Tests for Cellular and Gene Therapy Products. 2008 http://www.fda.gov/cber/gdlns/testcellgene.htm.
  • 28.Dang Y, Disis ML. Identification of immunologic biomarkers associated with clinical response after immune-based therapy for cancer. Ann N Y Acad Sci. 2009;1174:81–87. doi: 10.1111/j.1749-6632.2009.04937.x. [DOI] [PubMed] [Google Scholar]
  • 29. http://www.cms.hhs.gov/CLIA/05_CLIA_Brochures.asp.
  • 30.ICH. [cited; Available from: http://www.ich.org/LOB/media/MEDIA417.pdf.
  • 31.NCCLS. Performance of Single Cell Immune Response Assays: Proposed Guidelines. Number 25. Volume 23. 940 West Valley Road, Suite 1400, Wayne, PA 19087-1898: NCCLS document I/LA26-P [ISBN 1-56238-507-0] NCCLS; 2003. [Google Scholar]
  • 32.Zhang W, Caspell R, Karulin AY, et al. ELISPOT assays provide reproducible results among different laboratories for T-cell immune monitoring--even in hands of ELISPOT-inexperienced investigators. J Immunotoxicol. 2009;6:227–234. doi: 10.3109/15476910903317546. [DOI] [PubMed] [Google Scholar]
  • 33.Janetzki S, Panageas KS, Ben-Porat L, et al. 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]
  • 34.Maecker HT, Hassler J, Payne JK, et al. 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]
  • 35.Britten CM, Gouttefangeas C, Welters MJ, et al. 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]
  • 36.Britten CM, Janetzki S, Ben-Porat L, et al. Harmonization guidelines for HLA-peptide multimer assays derived from results of a large scale international proficiency panel of the Cancer Vaccine Consortium. Cancer Immunol Immunother. 2009;58:1701–1713. doi: 10.1007/s00262-009-0681-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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]
  • 38.Mander A, Gouttefangeas C, Ottensmeier C, et al. Serum is not required for ex vivo IFN-gamma ELISPOT: a collaborative study of different protocols from the European CIMT Immunoguiding Program. Cancer Immunol Immunother. 59:619–627. doi: 10.1007/s00262-009-0814-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Fahey JL, Aziz N, Spritzler J, et al. Need for an external proficiency testing program for cytokines, chemokines, and plasma markers of immune activation. Clin Diagn Lab Immunol. 2000;7:540–548. doi: 10.1128/cdli.7.4.540-548.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Denny TN, Gelman R, Bergeron M, et al. A North American multilaboratory study of CD4 counts using flow cytometric panLeukogating (PLG): a NIAID-DAIDS Immunology Quality Assessment Program Study. Cytometry B Clin Cytom. 2008;74 Suppl 1:S52–S64. doi: 10.1002/cyto.b.20417. [DOI] [PubMed] [Google Scholar]
  • 41.Boaz MJ, Hayes P, Tarragona T, et al. Concordant proficiency in measurement of T-cell immunity in human immunodeficiency virus vaccine clinical trials by peripheral blood mononuclear cell and enzyme-linked immunospot assays in laboratories from three continents. Clin Vaccine Immunol. 2009;16:147–155. doi: 10.1128/CVI.00326-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Smith SG, Joosten SA, Verscheure V, et al. Identification of major factors influencing ELISpot-based monitoring of cellular responses to antigens from Mycobacterium tuberculosis. PLoS One. 2009;4:e7972. doi: 10.1371/journal.pone.0007972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hanekom WA, Dockrell HM, Ottenhoff TH, et al. Immunological outcomes of new tuberculosis vaccine trials: WHO panel recommendations. PLoS Med. 2008;5:e145. doi: 10.1371/journal.pmed.0050145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Avache S, Panelli M, Marincola FM, Stroncek DF. Effects of storage time and exogenous protease inhibitors on plasma protein levels. Am J Clin Pathol. 2006;126:174–184. doi: 10.1309/3WM7-XJ7R-D8BC-LNKX. [DOI] [PubMed] [Google Scholar]
  • 45.Kirkwood JM, Lee S, Moschos SJ, et al. Immunogenicity and antitumor effects of vaccination with peptide vaccine+/−granulocyte-monocyte colony-stimulating factor and/or IFN-alpha2b in advanced metastatic melanoma: Eastern Cooperative Oncology Group Phase II Trial E1696. Clin Cancer Res. 2009;15:1443–1451. doi: 10.1158/1078-0432.CCR-08-1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Slingluff CL, Jr, Petroni GR, Chianese-Bullock KA, et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res. 2007;13:6386–6395. doi: 10.1158/1078-0432.CCR-07-0486. [DOI] [PubMed] [Google Scholar]
  • 47.Dangoor A, Lorigan P, Keilholz U, et al. Clinical and immunological responses in metastatic melanoma patients vaccinated with a high-dose poly-epitope vaccine. Cancer Immunol Immunother. 59:863–873. doi: 10.1007/s00262-009-0811-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Welters MJ, Kenter GG, Piersma SJ, et al. 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]
  • 49.Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838–1847. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]
  • 50.Welters MJ, Kenter GG, de Vos van Steenwijk PJ, et al. Success or failure of vaccination for HPV16-positive vulvar lesions correlates with kinetics and phenotype of induced T-cell responses. Proc Natl Acad Sci U S A. 107:11895–11899. doi: 10.1073/pnas.1006500107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Quast S, Zhang W, Shive C, et al. IL-2 absorption affects IFN-gamma and IL-5, but not IL-4 producing memory T cells in double color cytokine ELISPOT assays. Cell Immunol. 2005;237:28–36. doi: 10.1016/j.cellimm.2005.09.002. [DOI] [PubMed] [Google Scholar]
  • 52.Rininsland FH, Helms T, Asaad RJ, Boehm BO, Tary-Lehmann M. Granzyme B ELISPOT assay for ex vivo measurements of T cell immunity. J Immunol Methods. 2000;240:143–155. doi: 10.1016/s0022-1759(00)00191-5. [DOI] [PubMed] [Google Scholar]
  • 53.Shafer-Weaver K, Rosenberg S, Strobl S, Gregory Alvord W, Baseler M, Malyguine A. Application of the granzyme B ELISPOT assay for monitoring cancer vaccine trials. J Immunother. 2006;29:328–335. doi: 10.1097/01.cji.0000203079.35612.c8. [DOI] [PubMed] [Google Scholar]
  • 54.Dhodapkar MV, Steinman RM, Sapp M, et al. Rapid generation of broad T-cell immunity in humans after a single injection of mature dendritic cells. J Clin Invest. 1999;104:173–180. doi: 10.1172/JCI6909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Iero M, Filipazzi P, Castelli C, et al. Modified peptides in anti-cancer vaccines: are we eventually improving anti-tumour immunity? Cancer Immunol Immunother. 2009;58:1159–1167. doi: 10.1007/s00262-008-0610-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Malyguine A, Strobl SL, Shafer-Weaver KA, et al. 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]
  • 57.Kim GG, Donnenberg VS, Donnenberg AD, Gooding W, Whiteside TL. A novel multiparametric flow cytometry-based cytotoxicity assay simultaneously immunophenotypes effector cells: comparisons to a 4 h 51Cr-release assay. J Immunol Methods. 2007;325:51–66. doi: 10.1016/j.jim.2007.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Devevre E, Romero P, Mahnke YD. LiveCount Assay: concomitant measurement of cytolytic activity and phenotypic characterisation of CD8(+) T-cells by flow cytometry. J Immunol Methods. 2006;311:31–46. doi: 10.1016/j.jim.2006.01.011. [DOI] [PubMed] [Google Scholar]
  • 59.Zaritskaya L, Shafer-Weaver KA, Gregory MK, Strobl SL, Baseler M, Malyguine A. Application of a flow cytometric cytotoxicity assay for monitoring cancer vaccine trials. J Immunother. 2009;32:186–194. doi: 10.1097/CJI.0b013e318197b1b2. [DOI] [PubMed] [Google Scholar]
  • 60.Maecker HT, Dunn HS, Suni MA, et al. Use of overlapping peptide mixtures as antigens for cytokine flow cytometry. J Immunol Methods. 2001;255:27–40. doi: 10.1016/s0022-1759(01)00416-1. [DOI] [PubMed] [Google Scholar]
  • 61.Nomura L, Maino VC, Maecker HT. Standardization and optimization of multiparameter intracellular cytokine staining. Cytometry A. 2008;73:984–991. doi: 10.1002/cyto.a.20602. [DOI] [PubMed] [Google Scholar]
  • 62.Dols A, Smith JW, 2nd, Meijer SL, et al. Vaccination of women with metastatic breast cancer, using a costimulatory gene (CD80)-modified, HLA-A2-matched, allogeneic, breast cancer cell line: clinical and immunological results. Hum Gene Ther. 2003;14:1117–1123. doi: 10.1089/104303403322124828. [DOI] [PubMed] [Google Scholar]
  • 63.Walker EB, Haley D, Petrausch U, et al. Phenotype and functional characterization of long-term gp100-specific memory CD8+ T cells in disease-free melanoma patients before and after boosting immunization. Clin Cancer Res. 2008;14:5270–5283. doi: 10.1158/1078-0432.CCR-08-0022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Slingluff CL, Jr, Petroni GR, Olson WC, Smolkin ME, Ross MI, Haas NB, Grosh WW, Boisvert ME, Kirkwood JM, Chianese-Bullock KA. Effect of GM-CSF on circulating CD8+ and CD4+ T cell responses to a multipeptide melanoma vaccine: Outcome of a multicenter randomized trial. Clin Cancer Res. 2009;15:7036–7044. doi: 10.1158/1078-0432.CCR-09-1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Moodie Z, Price L, Gouttefangeas C, et al. Response definition criteria for ELISPOT assays revisited. Cancer Immunol Immunother. doi: 10.1007/s00262-010-0875-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dubey S, Clair J, Fu TM, et al. Detection of HIV vaccine-induced cell-mediated immunity in HIV-seronegative clinical trial participants using an optimized and validated enzyme-linked immunospot assay. J Acquir Immune Defic Syndr. 2007;45:20–27. doi: 10.1097/QAI.0b013e3180377b5b. [DOI] [PubMed] [Google Scholar]
  • 67.Brazma A, Hingamp P, Quackenbush J, et al. Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet. 2001;29:365–371. doi: 10.1038/ng1201-365. [DOI] [PubMed] [Google Scholar]
  • 68. http://www.miataproject.org.
  • 69.McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Reporting recommendations for tumor marker prognostic studies (REMARK) J Natl Cancer Inst. 2005;97:1180–1184. doi: 10.1093/jnci/dji237. [DOI] [PubMed] [Google Scholar]
  • 70.Alvarez DF, Helm K, Degregori J, Roederer M, Majka S. Publishing flow cytometry data. Am J Physiol Lung Cell Mol Physiol. 298:L127–L130. doi: 10.1152/ajplung.00313.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hoos A, Parmiani G, Hege K, et al. A clinical development paradigm for cancer vaccines and related biologics. J Immunother. 2007;30:1–15. doi: 10.1097/01.cji.0000211341.88835.ae. [DOI] [PubMed] [Google Scholar]
  • 72. http://mibbi.org/index.php/MIBBI_portal.
  • 73. http://www.mibbi.org.
  • 74.Janetzki S, Britten CM, Kalos M, et al. "MIATA"-minimal information about T cell assays. Immunity. 2009;31:527–528. doi: 10.1016/j.immuni.2009.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lee JA, Spidlen J, Boyce K, et al. MIFlowCyt: the minimum information about a Flow Cytometry Experiment. Cytometry A. 2008;73:926–930. doi: 10.1002/cyto.a.20623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Spidlen J, Moore W, Parks D, et al. Data File Standard for Flow Cytometry, version FCS 3.1. Cytometry A. 77:97–100. doi: 10.1002/cyto.a.20825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Taylor CF, Field D, Sansone SA, et al. Promoting coherent minimum reporting guidelines for biological and biomedical investigations: the MIBBI project. Nat Biotechnol. 2008;26:889–896. doi: 10.1038/nbt.1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.2009 www.fda.gov/biologicsbloodvaccines/guidancecomplianceregulatoryinformation/default.htm.
  • 79. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfCFR/CFRSearch.cfm.
  • 80.Cox JH, Ferrari G, Janetzki S. Measurement of cytokine release at the single cell level using the ELISPOT assay. Methods. 2006;38:274–282. doi: 10.1016/j.ymeth.2005.11.006. [DOI] [PubMed] [Google Scholar]
  • 81.Xu Y, Theobald V, Sung C, et al. Validation of a HLA-A2 tetramer flow cytometric method, IFNgamma real time RT-PCR, and IFNgamma ELISPOT for detection of immunologic response to gp100 and MelanA/MART-1 in melanoma patients. J Transl Med. 2008;6:61. doi: 10.1186/1479-5876-6-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Koup RA, Roederer M, Lamoreaux L, et al. Priming immunization with DNA augments immunogenicity of recombinant adenoviral vectors for both HIV-1 specific antibody and T-cell responses. PLoS One. 5:e9015. doi: 10.1371/journal.pone.0009015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Roederer M, Koup RA. Optimized determination of T cell epitope responses. J Immunol Methods. 2003;274:221–228. doi: 10.1016/s0022-1759(02)00423-4. [DOI] [PubMed] [Google Scholar]
  • 84.Wolchok JD, Hoos A, O'Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412–7420. doi: 10.1158/1078-0432.CCR-09-1624. [DOI] [PubMed] [Google Scholar]
  • 85.Kalos M. An integrative paradigm to impart quality to correlative science. J Transl Med. 8:26. doi: 10.1186/1479-5876-8-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. http://www.moffitt.org/Site.aspx?spid=C54AF116F69244D49BACE202F69BC2A6.
  • 87.Gaseitsiwe S, Valentini D, Mahdavifar S, et al. Pattern recognition in pulmonary tuberculosis defined by high content peptide microarray chip analysis representing 61 proteins from M. tuberculosis. PLoS One. 2008;3:e3840. doi: 10.1371/journal.pone.0003840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
  • 89.Chaussabel D, Ueno H, Banchereau J, Quinn C. Data management: it starts at the bench. Nat Immunol. 2009;10:1225–1227. doi: 10.1038/ni1209-1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. www.isbtc.org/meetings.
  • 91.Disis ML, dela Rosa C, Goodell V, et al. Maximizing the retention of antigen specific lymphocyte function after cryopreservation. J Immunol Methods. 2006;308:13–18. doi: 10.1016/j.jim.2005.09.011. [DOI] [PubMed] [Google Scholar]
  • 92.Maecker HT, Moon J, Bhatia S, et al. 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]
  • 93. http://aactg.org/about-actg.
  • 94. http://iqa.center.duke.edu/modules/dhvi_iqa/index.php?id=1.

RESOURCES