NIH Public Access Author Manuscript Cold Spring Harb Protoc. Author manuscript; available in PMC 2014 June 24.

Similar documents
Disclosure Information. Mary L. Disis

Posters and Presentations

Synergistic combinations of targeted immunotherapy to combat cancer

staining and flow cytometry

Why are validated immunogenicity assays important for HIV vaccine development?

Biomarker integration in clinical trials for immunotherapies

NKTR-255: Accessing The Immunotherapeutic Potential Of IL-15 for NK Cell Therapies

Novel RCC Targets from Immuno-Oncology and Antibody-Drug Conjugates

International Society of Breast Pathology. Immune Targeting in Breast Cancer. USCAP 2017 Annual Meeting

BIOMARKER DRIVEN EARLY PHASE ONCOLOGY CLINICAL TRIALS; CHALLENGES IN THE REAL WORLD SID KATUGAMPOLA CENTRE FOR DRUG DEVELOPMENT CANCER RESEARCH UK

Combining ADCs with Immuno-Oncology Agents

Kinetics of tumor-specific T-cell response development after active immunization in patients with HER-2/neu overexpressing cancers

Emerging Tissue and Serum Markers

Supplementary Figure 1. Double-staining immunofluorescence analysis of invasive colon and breast cancers. Specimens from invasive ductal breast

Type 1 Diabetes: Islet expressing GAD65 (green) with DAPI (Blue) Islet expressing Insulin (red) in 3D confocal imaging

Identification of novel immune regulators of tumor growth using highthroughput

Reviewers' comments: Reviewer #1 (Remarks to the Author):

DEVELOPMENT OF CELLULAR IMMUNOLOGY

Cancer Progress. The State of Play in Immuno-Oncology

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity

Manipulating the Tumor Environment

NKTR-255: Accessing IL-15 Therapeutic Potential through Robust and Sustained Engagement of Innate and Adaptive Immunity

Challenges in Development and Validation of an Intracellular Cytokine Staining assay

Emerging Concepts of Cancer Immunotherapy

Daratumumab: Uncovering a novel mechanism of action for an approved antibody therapy

CD25-PE (BD Biosciences) and labeled with anti-pe-microbeads (Miltenyi Biotec) for depletion of CD25 +

Fluorochrome Panel 1 Panel 2 Panel 3 Panel 4 Panel 5 CTLA-4 CTLA-4 CD15 CD3 FITC. Bio) PD-1 (MIH4, BD) ICOS (C398.4A, Biolegend) PD-L1 (MIH1, BD)

Basic Principles of Tumor Immunotherapy and Mechanisms of Tumor Immune Suppression. Bryon Johnson, PhD

Program Summary Breast Immune-Oncology Conference: Emerging Opportunities-Enhancing Awareness of Clinical Trials

NKTR-214 plus NKTR-262, a Scientifically-Guided Rational Combination Approach for Immune Oncology

A Versatile Tool to Study Immune Checkpoint Therapeutics: Syngeneic Tumor Mouse Models in vivo

Glioblastoma and CNS tumors

Tumor Immunity and Immunotherapy. Andrew Lichtman M.D., Ph.D. Brigham and Women s Hospital Harvard Medical School

Enhanced Cancer Vaccine Effectiveness with NKTR-214, a CD122-Biased Cytokine

DISCOVERING ATCC IMMUNOLOGICAL CELLS - MODEL SYSTEMS TO STUDY THE IMMUNE AND CARDIOVASCULAR SYSTEMS

Enhancing the Clinical Activity of HER2/neu Specific T Cells. William Gwin, MD Internal Medicine, Resident University of Washington

ACTR (Antibody Coupled T-cell Receptor): A universal approach to T-cell therapy

A Cellular Antigen Stabilisation Reagent

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Anti-PD-L1 antibody [28-8] ab205921

Immunotherapy: The Newest Treatment Route

Therapeutic PD L1 and LAG 3 blockade rapidly clears established blood stage Plasmodium infection

IDENTIFICATION OF BIOMARKERS FOR EARLY DIAGNOSIS OF BREAST CANCER"

Supplementary Figure 1

Immunotherapie: algemene principes

MHC MULTIMER PROFICIENCY PANEL 2017

Cancer Immunotherapy Survey

Immunotherapy in lung cancer. Saurabh maji

Cancer immunity and immunotherapy. General principles

Do Your Flow Cytometric LDTs. Validation Guidelines? Fiona E. Craig, MD University of Pittsburgh School of Medicine

Darwinian selection and Newtonian physics wrapped up in systems biology

Chapter 7 Conclusions

B Cells Promote Pancreatic Tumorigenesis

Role of microenvironment and tumor interactions in melanoma progression with special regard to the prognostic significance of immune cell infiltrate

Therapeutic efficacy of MUC1- specific CTL and CD137 costimulation. mammary cancer model. Pinku Mukherjee & Sandra Gendler

Exploiting tumour infiltrating lymphocytes (TILs) as a therapeutic strategy in epithelial ovarian cancer

Combination Immunotherapy Approaches Chemotherapy, Radiation Therapy, and Dual Checkpoint Therapy

Optimizing Intracellular Flow Cytometry:

Tim-3 as a target for tumor immunotherapy

Tumor Associated Macrophages as a Novel Target for Cancer Therapy

Checkpoint Regulators Cancer Immunotherapy takes centre stage. Dr Oliver Klein Department of Medical Oncology 02 May 2015

Toxoplasma gondii IgM (Toxo IgM)

Therapeutic Cancer Vaccines

COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16

ONCO-HU TM MICE FOR IMMUNOTHERAPEUTIC DRUG DISCOVERY. Brian W. Soper, Ph.D. Senior Technical Information Scientist

Durham NC PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland Distribution Unlimited

Toxoplasma gondii IgM ELISA Kit

Letter to Editor Tissue micro arrays for immunohistochemical detection of inflammatory infiltrates in renal cell carcinoma

Glioblastoma and CNS tumors

Experimental Design for Immunologists

MHC Tetramers and Monomers for Immuno-Oncology and Autoimmunity Drug Discovery

The Role of Immunotherapy in Prostate Cancer: What s Trending?

Immune Checkpoints. PD Dr med. Alessandra Curioni-Fontecedro Department of Hematology and Oncology Cancer Center Zurich University Hospital Zurich

FcγRIIIA (CD16)-expressing monocytes mediate the depletion of tumor-infiltrating Tregs via ipilimumab-dependent ADCC in melanoma patients

Biomarcatori per la immunoterapia: cosa e come cercare Paolo Graziano

Triple-Negative Breast Cancer Time to Slice and Dice? Carsten Denkert, MD Charité University Hospital Berlin, Germany

VENTANA PD-L1 (SP142) Assay Guiding immunotherapy in NSCLC

THE FUTURE OF IMMUNOTHERAPY IN COLORECTAL CANCER. Prof. Dr. Hans Prenen, MD, PhD Oncology Department University Hospital Antwerp, Belgium

Herpes Simplex Virus 2 IgM HSV 2 IgM

Index. neurosurgery.theclinics.com. Note: Page numbers of article titles are in boldface type.

Exploring Immunotherapies: Beyond Checkpoint Inhibitors

Copyright. Tocagen Inc. Lead Product Candidate: Toca 511 & Toca FC Preclinical Overview

IMMUNOTHERAPY FOR CANCER A NEW HORIZON. Ekaterini Boleti MD, PhD, FRCP Consultant in Medical Oncology Royal Free London NHS Foundation Trust

In Vitro and Ex Vivo Immuno- Oncology Drug Discovery

Toxoplasma gondii IgM ELISA Kit

Update on Colorectal Cancer: Integrating the Host Immune Response 3 rd Emirates Surgical Pathology Conference 15 December 2017 Dubai, UAE

CONTRACTING ORGANIZATION: Johns Hopkins University School of Medicine Baltimore, MD 21205

Releasing the Brakes on Tumor Immunity: Immune Checkpoint Blockade Strategies

What to Measure, How to Measure It

Douglas Jolly Executive VP R&D Tocagen Inc.

Basic Principles of Tumor Immunotherapy. Ryan J. Sullivan, M.D. Massachusetts General Hospital Cancer Center Boston, MA

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland Approved for public release; distribution unlimited

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Immunology Lecture 4. Clinical Relevance of the Immune System

EXPERIMENTAL MODELS OF CEREBRAL MALARIA

Potential Rebalancing of the Immune System by Anti-CD52 Therapy

PD-L1 Expression and Signaling by Tumors and Macrophages: Comparative Studies in Mice and Dogs

Simultaneous blockade of PD-1 and VEGFR2 induces synergistic. Short title: Synergistic antitumour effect by dual blockade of PD-1 and VEGFR2

Naive, memory and regulatory T lymphocytes populations analysis

Exploring the PD-L1 Pathway

Transcription:

NIH Public Access Author Manuscript Published in final edited form as: Cold Spring Harb Protoc. ; 2014(3): 231 234. doi:10.1101/pdb.top078816. Evaluation of Cancer Immunity in Mice Mary L. Disis 1,3 and Karolina Palucka 2,3 1 Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington 98109 2 Baylor Institute for Immunology Research, Dallas, Texas 75204 and Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York 10029 Abstract There have been significant advances in the development and application of novel therapeutic approaches and improved diagnostics for cancer in the past decade. Manipulation and/or assessment of cancer-specific immunity have been central to these advances. Murine models are a standard for the preclinical development of cancer immunotherapeutics. However, critical advances in our understanding of the role of the immune microenvironment and the assessment of cancer-specific immunity have not been fully applied to rodent models. Methods to preserve the function of immune cells after cryopreservation and standard approaches to quantitative immune assays have not been developed. Furthermore, a detailed evaluation of the immune tumor environment, which can impact a clinical response to different agents, is rarely undertaken as models are being contemplated. Rapid translation of immunoncology agents to the clinic will require standardization of immunologic assay methods and a more detailed immunologic characterization of common mouse models. Outlined here are the critical elements in assessing immunity in cancer mouse models and suggestions concerning the standardization of approaches when using these models for the study of immunoncology. Introduction Innovative immunotherapeutics have entered the clinic largely based on the recognition that immune cells and their mediators, when chronically engaged, may both hinder and foster tumor development. Along with stimulating specific immune responses, modulating immune-related receptors and proteins in the tumor microenvironment has become an area of intense clinical investigation. Indeed, the immune microenvironment has been shown to be an important determinant of response to standard therapies in cancer patients (Denkert et al. 2010; DeNardo et al. 2011). Murine models have long played a major role in the evaluation of immune-based therapies for cancer. Standardized methods for cryopreservation of murine lymphocytes, measuring adaptive or innate immunity and characterizing tumor immune infiltrates, have not been fully developed across multiple mouse models as such assays have been for humans. 2013 Cold Spring Harbor Laboratory Press 3 Correspondence: ndisis@uw.edu; karolinp@baylorhealth.edu.

Disis and Palucka Page 2 Developmental work is needed in several areas to improve the utility of mouse models as predictors of optimized cancer immmunotherapeutics. Cryopreservation of Murine Lymphocytes and Immune Cells Studies with human T and B cells have showed that there are several components in the cryopreservation process that can adversely impact the function of lymphocytes once they are thawed (Disis et al. 2006). These components include the additive in which the cells are frozen, the temperature at the time of thawing, and the length of time between thawing and use of the cells in assays. Extensive analysis of these parameters for cryopreservation of human lymphocytes has resulted in the adoption of standard operating procedures (SOPs) for the freezing and thawing of human lymphocytes to maintain basic immune functions such as antigen-specific proliferation and cytokine production and, thus, assay validity. Detailed evaluations and procedures for the standardization of cryopreservation for murine lymphocytes to retain immune function have not been published. In a recent review of procedures collecting, processing, and storing T-regulatory cells (T reg ), a subtype of CD4 + T-lymphocytes, authors catalogued numerous methods of processing both murine and human T reg (Daniele et al. 2011); however, no SOP or consensus protocol was presented. Cryopreservation of murine lymphocytes to retain immune functionality is important for several reasons: (1) to facilitate sample sharing for the development of new immune-based technologies for the assessment of adaptive and innate cancer associated immunity, (2) to allow replicate assay performance adding to overall data quality, and (3) to allow batch analysis of similar experimental groups potentially decreasing assay variability. In general, cancer-specific immunity is of a lower functional avidity, different Th phenotype, and, in most cases, of a much lower magnitude than immunity to infectious agents. For these reasons, protocols developed in infectious disease research may not be directly applicable to the analysis of cancer-specific immune responses. Good viability of lymphocytes (>70%) after cryopreservation and thawing has been shown to be associated with retained immunologic function. Preliminary studies with murine lymphocytes showed that viability is preserved when the lymphocytes are thawed at 37 C and the freezing media has been screened for the ability to maintain both viability and recovery of cells (E Gad, University of Washington, pers. comm.). Indeed, in a screening study of five commercially available media suitable for cryopreservation, two of the five products resulted in <70% viability after thawing in more than 20 specimens tested. Studies are ongoing to detail the elements of a protocol for cryopreservation of murine lymphocytes for immunoncology research. At minimum, investigators should record and report the median and range of viability of murine lymphocytes, when thawed, as a potential source of assay variability. Standardization and Standard Operating Procedures for Immune Assays Over the last decade, there has been an explosion in the development of quantitative immune assays to both characterize and enumerate tumor-specific T- and B-cell responses that are suitable for use with both human and murine cells. Immune competent murine models of common cancers could allow a greater understanding and/or delineation of useful immune

Disis and Palucka Page 3 biomarker candidates for validation in human clinical trials, but there has been little effort to develop the standardization needed to intensely mine rodent models for immune biomarkers that could be surrogates of clinical response after immune therapy. The two major quantitative assays used in translational immunoncology in both humans and rodents are enzyme-linked immunosorbent spot assay or ELISPOT and flow cytometry. ELISPOT is a more labor-intensive assay than flow cytometry and requires the stimulation of lymphocytes with antigen- and protein-based assessment of cytokine secretion. There are many sources of variability in ELISPOT. First is the length of time the T cells are allowed to produce cytokine after antigen stimulation and whether two or more stimulations are needed to obtain signal. The longer the incubation the greater the potential for cytokine production from unstimulated cells that will increase the background of the assay and lower sensitivity. As stated above, cancer-specific responses are often of low magnitude and the number of lymphocytes plated in each well may impact signal. The greater the number of lymphocytes, the more likely low signal will be detected. However, sensitivity may also be decreased by an increase in the background due to nonspecific secretion by resting cells. Finally, appropriate cryopreservation methods that preserve lymphocyte viability will impact assay validity (as discussed above). Studies of immunomodulation in mouse models should include basic assay development to ensure high antigen-specific signal with low background responses. Flow cytometric methods are less labor intensive but are highly dependent on quality reagents. Monoclonal antibodies used to stain surface receptors or intracellular cytokines can vary greatly in their avidity for target and their utility in a variety of assays. If a specific antibody works well for immunohistochemical staining, that same antibody may not be as effective in flow cytometry. Flow cytometric analyses should use positive controls to titer antibody concentrations to optimize signal. Groups such as the National Cancer Institute supported Mouse Models of Human Cancer Consortium are creating reference lists that provide usage data on common immune-related antibodies along with performance comments and suggested starting concentrations (see further description below). A final consideration in assessing the validity of immune response data in any murine model is the genetic background of the model itself. As inbred strains, it is well known that T cells in mice can be skewed to a predominant Type I versus Type II response. Specific strains are commonly used in studies of infectious disease to evaluate lack of response to infectious agents, but a catalogue of these differences has not been made for cancer models. For example, FVB, a common strain for transgenic mice construction, have a Th2 bias in cellular immunity as compared to a strain such as BALB/c (Kim et al. 2012). The phenotype bias of a particular strain may greatly impact immunologic monitoring as well as ability to respond to immune-based cancer therapies. There are no existing protocols to fully assess Th1/Th2 skewing before the use of a particular strain. A thorough evaluation of the literature should be the first step when contemplating a particular model. The use of two or more strains in testing cancer immunotherapeutics is an alternative. Finally, some information may be gleaned by stimulating T cells with nonspecific mitogens and assessing whether the immune response elicited is predominantly Th1 or Th2 (Christie et al. 1999).

Disis and Palucka Page 4 Characterization of the Tumor Immune Environment There are many rodent models available for the translational evaluation of immune-based cancer therapeutics. Rarely are tumors developing in those models fully characterized at baseline for infiltrating immune elements or other factors that might influence data quality. Given the functional relevance of immune cellular networking in tumors, it is imperative to incorporate immunometrics such as the immunoscore (a score including a composite of immune cellular and protein biomarkers assessed in tumor tissue) (Fridman et al. 2011) into traditional experimental schemes. These predictors, if present in rodent models, could greatly assist with defining a hierarchical prioritization among future newly developed immune therapeutic approaches. Tumor infiltrating CD8 + T cells, memory T cells, and a lack of infiltrating immune regulatory cells have been shown to be associated with clinical outcomes across several human cancer types. In melanoma and colon cancer such infiltrates provide prognostic information that adds power to already existing staging systems (Bindea et al. 2010). As stated in the introduction, immune infiltrates in tumors may contribute significantly to clinical response to common cancer therapies. An immune score has not been developed for rodent tumors nor have immune infiltrates been assessed extensively in common rodent models used for the study of cancer immune therapies. Our lack of understanding of the baseline immune microenvironment in therapeutic models may be a source of variability in both the preclinical and correlative science assessment of new agents and a major reason for a lack of correlation of rodent models with disease outcomes in human clinical trials. An evaluation of a basic panel of markers should be the standard for the minimal characterization of the murine tumor immune environment. Such a panel would include analysis of infiltrating cells similar to the human immune score (T cells infiltrating tumor), regulatory elements; both cell- and receptor-based (T reg and myeloid-derived suppressor cells [MDSCs]), and markers that would further phenotype the immune microenvironment (e.g., PD-1, PDL-1, Tbet, GATA3, CTLA-4) (W Fridman, R Schreiber, E Engleman, et al., pers. comm.). Work is ongoing to determine appropriate antibodies and methods for the development of a murine immune score. SOPs must be created for immune assessment with the expectation that baseline data will be provided as an essential component of overall data interpretation. One key component in determining the extent of immune infiltrates in a tumor is the use of monoclonal and polyclonal antibodies in the context of flow cytometry or immunohistochemical staining. Commercial antibodies vary greatly in quality for specific procedures. Members of the Mouse Models of Human Cancer Consortium have instituted a glossary of validated reagents and protocols that are useful for functional phenotyping of the immune system and immune environment in murine cancer models: http:// emice.nci.nih.gov/characterizing-models/mice/antibody/?searchterm=antibodies. The glossary includes basic information on reagent performance, concentrations needed for signal detection, and clone numbers for easy identification.

Disis and Palucka Page 5 Conclusions References Detailed immune characterization of common rodent models and development and standardization of immune-based assays, methods, and operating procedures are needed to better use mouse models for the rapid clinical translation of immune-based cancer therapy. Full characterization of the tumor immune microenvironment and an ongoing quality assurance plan for immune monitoring should be put in place to maintain data reproducibility during the period of model use. These data are also key for understanding response, or lack of response, when evaluating immune-based cancer therapeutics. Bindea G, Mlecnik B, Fridman WH, Pagès F, Galon J. Natural immunity to cancer in humans. Curr Opin Immunol. 2010; 22:215 222. [PubMed: 20207124] Christe M, Rutti B, Brossard M. Influence of the genetic background and parasite load of mice on the immune response developed against nymphs of Ixodes ricinus. Parasitol Res. 1999; 85:557 561. [PubMed: 10382605] Daniele N, Scerpa MC, Landi F, Caniglia M, Miele MJ, Locatelli F, Isacchi G, Zinno F. T reg cells: Collection, processing, storage and clinical use. Pathol Res Pract. 2011; 207:209 215. [PubMed: 21397410] DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011; 1:54 67. [PubMed: 22039576] Denkert C, Loibl S, Noske A, Roller M, Müller BM, Komor M, Budczies J, Darb-Esfahani S, Kronenwett R, Hanusch C, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010; 28:105 113. [PubMed: 19917869] Disis ML, dela Rosa C, Goodell V, Kuan LY, Chang JC, Kuus-Reichel K, Clay TM, Kim Lyerly H, Bhatia S, Ghanekar SA. Maximizing the retention of antigen specific lymphocyte function after cryopreservation. J Immunol Methods. 2006; 308:13 18. [PubMed: 16337957] Fridman WH, Galon J, Pagès F, Tartour E, Sautès-Fridman C, Kroemer G. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res. 2011; 71:5601 5605. [PubMed: 21846822] Kim EM, Bae YM, Choi MH, Hong ST. Cyst formation, increased anti-inflammatory cytokines and expression of chemokines support for Clonorchis sinensis infection in FVB mice. Parasitol Int. 2012; 61:124 129. [PubMed: 21820080]